Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
Type of Medium
Language
Subjects(RVK)
  • 1
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 2737-2737
    Abstract: TH17 cells are T helper cells generated from naïve CD4 population in the presence of TGF-beta and IL-6 or IL-1-beta; and expanded in presence of IL-23. TH17 cells predominantly produce IL-17 and/or IL-22 and participate in both development of immunity and autoimmunity. Since TH17 cells are involved in modulation of immune response and we haveobserved both B and T cell immune dysfunction in multiple myeloma (MM), we have further evaluated the role of TH17 cells and associated cytokines in tumor cell growth, as well as, the imbalance of immune homeostasis in myeloma. We first observed that the baseline frequency of TH17 cells was significantly higher in mononuclear cells from MM patient peripheral blood (n=11) (2 folds) and bone marrow (n=4) (50%) compared to normal donor samples. Furthermore, under polarizing conditions with cytokine cocktail, significantly higher number of TH17 cells were induced in presence of TGF-beta in mononuclear cells from MM patients in both blood (4 fold) and bone-marrow (9 fold) compared to normal donor samples. These results indicate that the cytokines present in the BM microenvironment favor the development of TH17 cell subset. We next evaluated the serum levels of TH17-associated cytokines in sera from patients with MM, as well as, normal donors. We observed significant increase in levels of IL-17 (2 fold), IL-21 (5 fold), IL-22 (8 fold) and IL-23 (6 fold) in myeloma (n=17) compared with normal donor (n=6). Importantly, IL-23 levels were 10 fold higher in myeloma BM samples compared with matching blood samples. Next, we evaluated the effect of TH17-associated pro-inflammatory cytokines on MM cell growth and survival. We observed induction of proliferation of 6 MM cell lines, as measured by thymidine incorporation, by IL-17 (30%), IL-21 (35%) and IL-22 (25%). Additionally, IL-17 significantly induced growth of MM cells in clonogenic assay. These growth promoting effects are not observed in the presence of anti-IL-17 antibody. In an effort to evaluate the cellular basis for this growth promoting effect of these cytokines, we investigated production of MM growth promoting cytokines in the BM milieu. We observed induction of IL-6 production by IL-17 from BM stromal cells alone and in the presence of MM cells. Production of IL-6 induced by IL-17 was abrogated (40–50%) by transduction of siRNA targeting IRF-4 into co-culture components (stromal and myeloma cells). The inhibition of IL-17-induced IL-6 production in stromal cells was also observed with JAK-2 (70%) and MEK (76%) inhibitors suggesting role of multiple signalling pathways in induction of IL-6. Finally, we have also observed negative effects of TH17- related cytokines, especially IL-17 and IL-22, on development of IFN-gamma producing TH1 cells suggesting immunosuppressive effect. Taken together these results suggest significant role of IL-17 and related cytokines in myeloma cell growth, as well as, immune dysfunction observed in these patients. This data provides the preclinical rationale to target IL-17 in myeloma to suppress growth as well as improve immune responsiveness.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 5370-5370
    Abstract: Background: Lenalidomide 25 mg once daily, 3 weeks on and 1 week off, with weekly dexamethasone is considered standard treatment for patients with multiple myeloma, although patients with comorbidities treated with this regimen frequently require dose reductions due to toxicity. We investigated the use of a lower initial lenalidomide dose in an effort to assess dose-associated toxicity as well as efficacy. Methods: This multi-center, open-label randomized trial included relapsed multiple myeloma patients randomized in a 1:1 ratio to receive lenalidomide 25 mg (Arm A) or lenalidomide 15 mg (Arm B) orally once daily for 21 days of each 28 day cycle. Both groups also received dexamethasone 40 mg once weekly. Patients continued the treatment until disease progression, with dose reductions or cessation of lenalidomide or dexamethasone as needed for toxicity. The primary objective of the trial was to monitor dose reductions and toxicities and compare response rates based on IMWG criteria and event-free survival in each arm. Results: A total of 33 relapsed multiple myeloma patients were treated on this clinical trial at 6 different Veteran's Administration Hospitals in the United States, with 16 patients randomized to the lenalidomide 25 mg group (Arm A) and 17 patients to lenalidomide 15 mg group (Arm B). All patients were male, with a median age of 70.5 years (range 53 to 87) and an average of 6 comorbid conditions; including, but not limited to, 64% with cardiovascular disease, 55% with urologic/renal dysfunction (33% with renal dysfunction), 36% with metabolic disorders and 33% with respiratory disorders. Patients were taking an average of 12 medications with significant risk of polypharmacy. We observed significantly higher frequency of dose reductions with the lenalidomide 25 mg dose compared to 15 mg dose (10 vs 4; p= 0.03). Of those requiring lenalidomide dose reductions the median number of comorbid conditions was 6.5 (mean 7.5). The majority of lenalidomide dose reductions were due to Grade III serious adverse events, such as infection, fatigue, rash and cytopenias. Four patients developed a venous thromboembolism. Five patients in each arm required dose reductions in dexamethasone. With a median follow-up of 10 months, 4 patients remain on study. The remainder of patients withdrew from the trial, most frequently due to disease progression, adverse events, or worsening of comorbid conditions. Only one death occurred, which was unrelated to the patient's multiple myeloma. Twenty-five patients (76%) achieved at least a partial response with treatment, including 12 patients in Arm A and 13 patients in Arm B. Overall there were 3 complete responses (2 in Arm A and 1 in Arm B), which all occurred at a lenalidomide dose of 15 mg. The event-free survival was similar between both groups (at 18 months: 62% in Arm A and 59% in arm B, p= 0.82). Conclusions: In older individuals with multiple comorbidities, lenalidomide 15 mg is associated with less frequent toxicities and dose reductions without significantly affecting response rates or event-free survival. These results provide the rationale for a lower initial lenalidomide dose for relapsed multiple myeloma patients with multiple comorbidities. Disclosures No relevant conflicts of interest to declare.
    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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 446-446
    Abstract: Abstract 446 Waldenstrom's macroglobulinemia (WM), similar to multiple myeloma (MM), is associated with immune dysfunction. Both T and B cell dysfunctions are reported with suppressed uninvolved immunoglobulin, and inadequate vaccine and T cell responses. Although some mechanisms mediating immune dysregulation in WM have been studied, its molecular and cellular basis remains ill defined. Similarly, number of inflammatory cytokines and chemokines has been implicated in this process, but their effect on WM cell growth and immune function has not been well characterized. Recently, TH17 cells, a new CD4 cell population, has been identified by the presence of IL-17. TH17 cells play an important role in auto-immunity and in the development of anti-tumor immunity. As TH17 cells support MM cell growth and induce immune dysfunction in MM, we have evaluated the the role of TH17 cells and associated pro-inflammatory cytokines in WM. We first analyzed T helper cell subsets (TH1, TH2, and TH17) in freshly isolated PBMC from WM, and observed that all three cell types were decreased in WM compared with normal donors. Particularly, the IFN-γ producing TH1 cells from patients with WM were significantly reduced compared to normal donors (11±2% vs 30±3% respectively, P 〈 0.01). However, unlike MM, IL-17 producing TH17 cell numbers were reduced in PBMC from WM patients (n=8) compared to PBMC from normal donors (n=8) and patients with MM (n=11), (1.5±0.5 vs 2.5±0.5% vs 4.50±0.8% respectively; p 〈 0.05). Furthermore, when we polarized isolated naïve CD4 cells from WM patients using TH17 polarizing cocktail consisting of IL-6, IL-1β, IL-23 and TGF-β to induce TH17 cells differentiation, WM patients, unlike MM patients, showed significantly lower induction of TH17 cells in CD4 population compared to normal donor TH17 cells (0.3±0.1% WM; 11.9±2 % MM and 3.6±0.7% ND). Next, we evaluated the serum levels of cytokines and chemokines in sera from patients with WM in comparison with normal donors. The sera from WM patients showed significantly elevated levels of IL-2 (5 folds), IL-15 (2 folds) and GM-CSF (2 folds) among 19 cytokines, compared with sera from normal donors. When we evaluated TH17 cell-associated cytokines, both IL-1-beta (3 folds) and IL-17 (2 folds) were significantly elevated in sera from WM patients compared with sera from normal donors. In addition, we observed modulation of chemokines including, MCP-1, MIP-1, Eotaxin and RANTES in sera from WM patients. Finally, when we cultured WM cell-line in the presence or absence of IL-17 with or without stromal cells, we observed significant induction of WM cell proliferation by IL-17 and its inhibition by anti-IL17 antibody. These data shows that although similar to MM, there is immune dysfunction in WM, the mechanisms driving these effects especially cytokine milieu, and TH17 cell population are different between MM and WM. Disclosures: Treon: Millennium Pharmaceuticals, Genentech BiOncology, Biogen IDEC, Celgene, Novartis, Cephalon: Consultancy, Honoraria, Research Funding; Celgene Corporation: Research Funding; Novartis Corporation: Research Funding; Genentech: Consultancy, Research Funding. Munshi:Millennium Pharmaceuticals: Honoraria, Speakers Bureau.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 115, No. 26 ( 2010-07-01), p. 5385-5392
    Abstract: Elevated cytokines in bone marrow (BM) micro-environment (interleukin-6 [IL-6], transforming growth factor-beta [TGF-β] , and IL-1β) may play an important role in observed immune dysfunction in multiple myeloma (MM). As IL-6 and TGF-β are important for the generation of T-helper 17 (TH17) cells, we evaluated and observed a significantly elevated baseline and induced frequency of Th17 cells in peripheral blood mononuclear cells (PBMCs) and BM mononuclear cells (BMMCs) from MM patients compared with healthy donors. We observed significant increase in levels of serum IL-17, IL-21, IL-22, and IL-23 in blood and BM in MM compared with healthy donors. We also observed that myeloma PBMCs after TH17 polarization significantly induced IL-1α, IL-13, IL-17, and IL-23 production compared with healthy donor PBMCs. We next observed that IL-17 promotes myeloma cell growth and colony formation via IL-17 receptor, adhesion to bone marrow stromal cells (BMSCs) as well as increased growth in vivo in murine xenograft model of human MM. Additionally, we have observed that combination of IL-17 and IL-22 significantly inhibited the production of TH1-mediated cytokines, including interferon-γ (IFN-γ), by healthy donor PBMCs. In conclusion, IL-17–producing Th17 cells play an important role in MM pathobiology and may be an important therapeutic target for anti-MM activity and to improve immune function.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 3464-3464
    Abstract: Hsp90 inhibitor has shown promising anti-tumor activity through the destabilization and eventual degradation of Hsp90 client proteins critical for cell survival. In this study, we examined the in vitro effects of Hsp90 inhibitor on the phenotype and function of human T lymphocytes and NK cells. We observed no significant effects of Hsp90 inhibitor treatment on cell survivals. However, Hsp90 inhibitor treatment for 24 hours led to irreversible down-regulation of expression of critical T-cell surface antigens including CD3, CD4, CD8, CD28, CD154 (CD40L) and TCRab. Among the antigens evaluated, expression of CD4 antigen was most significantly downregulated (untrt vs. trt = 326 vs. 88 in Mean Fluorescence Intensity) following Hsp90 inhibitor treatment. Decreased CD3+ T lymphocytes proliferation (untrt vs. trt = 222839 cpm vs. 111102 cpm, 3[H]-thymidine incorporation) and reduced IFN-g secretion (untrt vs. trt = 77 vs. 48 pg/ml) was observed upon stimulation with allogeneic dendritic cells following 24 hrs treatments of T cells with Hsp90 inhibitor. Furthermore, CD3+ T-cell proliferation in response to mitogen stimulation, as measured by flow cytometry using CFSE was decreased following Hsp90 inhibitor treatment (untrt vs. trt = 41% vs. 3%, CFSE). Specifically, the CD4+CD28+ (untrt vs. trt = 32% vs. 1%) and CD8+CD28+ (untrt vs. trt = 27% vs. 17%) activated T-cell subpopulations displayed a significant decrease in proliferation in response to mitogen. Similarly, NK cells displayed decreased activation receptor expression including CD2, CD11a, CD94, NKp30, NKp44, NKp46, and KARp50.3 and reduced cytotoxic activity against multiple myeloma cells (untrt vs. trt = 49% vs. 11% against MM1S cells, 65% vs. 8% against ARP cells) following Hsp90 inhibitor treatment. These studies demonstrate that Hsp90 inhibitor treatment significantly affects phenotype and fun ction of human T-lymphocytes as well as NK cells, and suggest the need to monitor immune functions in patients being treated with Hsp90 inhibitor in our future studies.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 3986-3986
    Abstract: Abstract 3986 B cell-malignancies exhibit considerable immune dysfunction particularly in multiple myeloma (MM). We have previously demonstrated that in T cell-compartment, regulatory T helper cells are dysfunctional in multiple myeloma (MM) while Th17 cells are significantly elevated and IL-17 produced by them is associated with MM cell growth and survival as well as suppressed immune responses and bone disease. We have here investigated the B cell-subsets and their ability to re-program anti-tumor immunity in MM. We have first characterised four different B cell-subsets (B1a, B1b, B2 and regulatory B cells) using 10-color flow cytometric analysis in both peripheral blood and bone-marrow (BM) samples from MM patients compared with normal healthy donors. We observe that CD5+ B1a-B cells are significantly elevated in peripheral blood of MM patients (N=7) compared to healthy donor (N=15) (42±8% vs 13±3%, respectively, p 〈 0.05); while normal B cells (B2 cells) are significantly reduced in peripheral blood (29.8±6.5, p 〈 0.05) and in the BM samples (11±4.8, N=4, p 〈 0.05) of MM patients compared to healthy donors (59±3, and 60.2±2, N=10, respectively). We also observed that both B1b (47.9±18 vs. 22.8±4) and regulatory B cells (7.1±4.5 vs. 1.54±0.3) are elevated in BM samples of MM compared to healthy donors, however there were no differences in B1b and regulatory B cells in the peripheral blood of MM compared to healthy donor samples. Interestingly, in myeloma we observe higher levels of activated B cell subsets but lower levels of memory B cell subsets compared to healthy donors. These results, particularly very low levels of normal B cells in MM patients, may explain the decreased levels of uninvolved immunoglobulin in MM. As removal of B cell population has been shown to re-program T helper cell populations, we next investigated impact of B cell population on T cell activation. We activated normal PBMC via the anti-CD3 antibody, in the presence or absence of B or CD25+ cells and measured intra-cellular IFN-γ levels in CD69+ cells. We found that the absence of B cells significantly inhibited interferon-producing T cells compared to PBMC (by 43%; p 〈 0.05). Importantly, following removal of CD25+ cells, which consists of both Tregs and activated memory T cells, with or without B cells, we did not observe any difference in the inhibition of IFN-γ, indicating that B cells are influencing memory T cells rather than naïve T cells for the production of IFN-γ. This prompted us to identify the phenotypic signature of regulatory T cell populations when purified memory T cells are polarized with the regulatory T cell cocktail in presence or absence of B cells. We observed that B cells reduce FOXP3 expression by 18 %(N=5) and establish cognitive interactions with T cells. This occurred by increasing the expression of GITR (154%) and CTLA4 (54%); while reducing PD1 (−24%) and OX40 (−21%) expression on T cells without affecting HLA expression. We have also observed these improvements by B cell modulation on T cells in MM. Our results indicate that targeting these re-programmable capabilities of B cells to modulate T helper cell populations may enable us to improve T cell function in MM; and may improve immune function in MM and also allow us to enhance responses to vaccinations. Disclosures: Ghobrial: Millennium: Advisory Board Other; Novartis: Advisory Board, Advisory Board Other. Richardson:Novartis: 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; Johnson & Johnson: Membership on an entity's Board of Directors or advisory committees. Treon:Onyx: Research Funding; Celgene: Research Funding; Pharmacyclics: Research Funding; Cephalon: Consultancy; Avila: Consultancy. Anderson:Celgene, Millennium, BMS, Onyx: Membership on an entity's Board of Directors or advisory committees; Acetylon, Oncopep: Scientific Founder, Scientific Founder Other.
    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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 3113-3113
    Abstract: We have previously demonstrated that Th17 cells, which produce IL-17A, are significantly elevated in peripheral blood and bone marrow (BM) of patients with Multiple Myeloma (MM) and IL-17A promotes MM cell growth and survival, both in vitro and in vivo via IL-17A receptor. We have recently evaluated and observed that anti-IL-17A monoclonal antibody (mAb) significantly inhibited MM cell growth in vitro, while IL-17A induced proliferation of MM cells compared to control. We have also observed significant down-regulation of IL-6 production by anti-IL-17A mAb in MM-BMSC co-culture. Importantly, the administration of anti-IL-17A mAb weekly for 4 weeks in the SCIDhu model of human myeloma, where MM cells grow within the human microenvironment in mice led to a significant inhibition of tumor growth compared to the control mice. This remarkable activity of anti-IL17 mAb raised the question of whether the myeloma cells themselves are a possible source of IL-17. In this study, we used transcriptome sequencing (RNA-Seq) data to evaluate the expression of IL-17A in primary CD138+ myeloma cells (N=17) compared to normal plasma cells (NPC) (N=5). Whereas none of the NPCs expressed IL-17A, it was significantly over-expressed in majority of MM cells. In addition, these data also showed that the expression of other IL-17 family members (IL-7B, C, D, E & F) and Th17-associated pro-inflammatory cytokines (IL-21, IL-22 & IL-23) were not significantly elevated in primary myeloma cells compared to normal donor plasma cells. We further validated these observations by IL-17 immunoblot showing IL17 expression in all MM cell lines and 10 out of 14 primary patient MM cells; confirmed IL-17 expression in MM cells by quantitative RT-PCR, and flow cytometry and by immuno-histochemistry and confocal microscopy. We observed that IL-17 knock down by IL-17-specific siRNA inhibited MM cell growth as well as their ability to induce IL-6 production in co-cultures with BMSC. Finally, expression profile data from 172 uniformly treated patients showed that patients with lower IL-17A expression had superior overall survival compared to those with higher expression. These data confirms that MM cells express IL-17 and targeting it with a mAb will abrogate the autocrine loop making it an attractive therapeutic target. Disclosures: No relevant conflicts of interest to declare.
    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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 456-456
    Abstract: Abstract 456 We have previously demonstrated that IL-17 producing TH17cells, a new subset of T helper cells, are significantly elevated in peripheral blood and bone marrow (BM) from patients with multiple myeloma (MM) and IL-17 produced by these cells promotes MM cell growth and survival, suppresses immune responses and induces osteoclast differentiation both in vitro and in vivo. Based on these observations we have investigated the effects of human anti-IL-17A monoclonal antibody (mAb), AIN-457, in MM. We observed that whereas IL-17A induced proliferation of MM cells (+30.7+2.7%) compared to control; anti-IL-17A mAb AIN-457 significantly inhibited MM cell growth both in presence and absence of BM stromal cells, as measured by thymidine incorporation (−18.7+1.5% and −22.7+2.6% respectively). We have further confirmed these inhibitory effects of anti-IL-17A antibody using MM cell colony forming assay with MethoCult agar plates. While presence of IL-17A increased the colony number from 80 in control plates to 188, presence of AIN-457 reduced the colonies to 〈 40 per unit area (p 〈 0.01). Evaluating the mechanism of action, IL-17A induced IL-6 production (+289.6+38%; p 〈 0.01); while AIN457 significantly down-regulated IL-6 production (−25+7%; p 〈 0.05) in MM-BMSC co-culture. We also observed that AIN-457 significantly reduced adhesion of MM cells to stromal cells (27%, p=0.011). AIN457 significantly inhibited IL-6 production in human fetal bone chips in the presence of MM cells within 24 hours of ex-vivo culture (control − 487+39 pg/ml; IL-17 990+27 pg/ml; p 〈 0.01 and AIN-457 − 326+7 pg/ml; p 〈 0.01). Since IL-17A plays a critical role in bone damage, we further evaluated the effect of this mAb on the generation of osteoclasts. When normal BM cells were cultured for three weeks in osteoclast supporting medium, presence of AIN-457 significantly inhibited TRAP+ multinucleated osteoclast cell numbers by 〉 60%. We next evaluated the efficacy of AIN-457 in vivo in the murine models of human myeloma; in the subcutaneous MM xenograft model, we observed significant reduction in tumor volumes by pre-treatment with AIN457 compared to control (142+77 mm versus 355+56 mm, p=0.019) while IL-17A significantly increased MM cell growth (727+135 mm, p=0.01). More importantly in the SCIDhu model of human myeloma where MM cells grow within the human microenvironment in the mice, administration of AIN-457 weekly for 4 weeks after the first detection of tumor in mice led to a significant inhibition of tumor growth as measured by human sIL-6 receptor compared to control mice (5.9±2.2 ng/ml versus 23.2±6.3 ng/ml; n=7; P 〈 0.01). These pre-clinical in vitro and in vivo observations confirm the role of IL-17A produced by TH17 cells in MM and provide the rationale for clinical evaluation of AIN 457 for both anti-myeloma effects as well as to improve bone disease in myeloma. Disclosures: No relevant conflicts of interest to declare.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Echocardiography, Wiley, Vol. 15, No. 4 ( 1998-05), p. 377-379
    Type of Medium: Online Resource
    ISSN: 0742-2822 , 1540-8175
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 1998
    detail.hit.zdb_id: 2041033-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. 11 ( 2021-11), p. e002917-
    Abstract: Immune checkpoint blockade (ICB) induces durable response in approximately 20% of patients with advanced bladder urothelial cancer (aUC). Over 50% of aUCs harbor genomic alterations along the phosphoinositide 3-kinase (PI3K) pathway. The goal of this project was to determine the synergistic effects and mechanisms of action of PI3K inhibition and ICB combination in aUC. Methods Alterations affecting the PI3K pathway were examined in The Cancer Genome Atlas (TCGA) and the Cancer Dependency Map databases. Human and mouse cells with Pten deletion were used for in vitro studies. C57BL/6 mice carrying syngeneic tumors were used to determine in vivo activity, mechanisms of action and secondary resistance of pan-PI3K inhibition, ICB and combination. Results Alterations along the PI3K pathway occurred in 57% of aUCs in TCGA. CRISPR (clustered regularly interspaced short palindromic repeats) knockout of PIK3CA induced pronounced inhibition of cell proliferation (p=0.0046). PI3K inhibition suppressed cancer cell growth, migration and colony formation in vitro. Pan-PI3K inhibition, antiprogrammed death 1 (aPD1) therapy and combination improved the overall survival (OS) of syngeneic mice with PTEN-deleted tumors from 27 days of the control to 48, 37, and 65 days, respectively. In mice with tumors not containing a PI3K pathway alteration, OS was prolonged by the combination but not single treatments. Pan-PI3K inhibition significantly upregulated CD80, CD86, MHC-I, and MHC-II in dendritic cells, and downregulated the transforming growth factor beta pathway with a false discovery rate-adjusted q value of 0.001. Interferon alpha response was significantly upregulated with aPD1 therapy (q value: 〈 0.001) and combination (q value: 0.027). Compared with the control, combination treatment increased CD8 + T-cell infiltration (p=0.005), decreased T reg -cell infiltration (p=0.036), and upregulated the expression of multiple immunostimulatory cytokines and granzyme B (p 〈 0.01). Secondary resistance was associated with upregulation of the mammalian target of rapamycin (mTOR) pathway and multiple Sprr family genes. Conclusions The combination Pan-PI3K inhibition and ICB has significant antitumor effects in aUC with or without activated PI3K pathway and warrants further clinical investigation. This combination creates an immunostimulatory tumor milieu. Secondary resistance is associated with upregulation of the mTOR pathway and Sprr family genes.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    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