Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 1510-1510
    Abstract: Pediatric AML has a relapse rate approaching 40%, indicating that resistance to chemotherapy remains a critical problem. We are focused on identifying and overcoming environment-mediated mechanisms of resistance. We previously reported that the sensitivity of G-CSF- and IL-6-induced STAT3 signaling was significantly associated with outcome in pediatric AML patients. In this follow-up study, we used conditioned medium (CM) from HS5 stromal cells as a more physiological stimulus to evaluate the capacity of primary AML cells to activate signaling pathways. We studied 111 diagnostic bone marrow samples from patients who enrolled on Children's Oncology Group (COG) AML treatment studies, AAML03P1 or AAML0531, and provided informed consent to bank bone marrow. The same chemotherapy backbone was used in both trials. All samples had at least 70% viability after thawing. Thawed cells were divided into aliquots for stimulation with 50% CM, 10 ng/ml G-CSF, or 5 ng/ml IL-6. Unstimulated cells were used for isotype controls and for determination of basal signaling levels. Following 15 min stimulation, cells were fixed and processed for FACS analysis of CD45, pY-STAT3, pY-STAT5, pERK1/2, and pAKT. Responses were expressed as the fold change in mean fluorescence intensity for stimulated cells over unstimulated cells (ΔMFI). The ΔMFIs for pY-STAT3 and pY-STAT5 varied between 0.1 and 31.8. The ΔMFIs for pERK1/2 and pAKT rarely exceeded 2. Samples with a robust response to one stimulus generally responded robustly to the others. The Pearson correlation coefficient (r) for CM-induced pY-STAT3 v. G-CSF-induced pY-STAT3 was 0.8511 (p 〈 0.00001). The CM-induced pY-STAT5 and G-CSF-induced pY-STAT5 responses were significantly but less strongly correlated (r=0.2765; p=0.0043). Cut point analyses identified response thresholds that distinguished patients with higher EFS from those with lower EFS. We found that higher CM-induced pY-STAT5 was significantly associated with an inferior EFS (Figure 1), with HR 2.06 for those with ΔMFI 〉 1.96 (p=0.034). Previously we found that higher inducible pY-STAT3 was associated with significantly better EFS (Redell, et al, Blood, 2013; Long, et al, Oncotarget, 2017). In this study, no cut point for CM-induced pY-STAT3 ΔMFI distinguished patients with good or poor EFS. Cytogenetics and FLT3/ITD were not significantly different for groups above and below the ΔMFI cut points. The finding that robust STAT5 signaling is associated with inferior EFS suggests that a factor that signals primarily through STAT5 and not STAT3 (e.g. GM-CSF, IL-3, FL) could contribute to treatment resistance and relapse. Interestingly, most of the samples that failed to activate STAT3/5 pathways in response to G-CSF also failed to respond to the cocktail of factors in CM, suggesting a generalized signaling dysfunction. To investigate intrinsic gene expression differences, we leveraged existing RNA-seq data for the samples for which we generated signaling responses. We obtained RNA-seq data for 27 samples from the NCI Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database. RNA-seq data for an additional 45 samples were provided through collaboration with the University of Washington. Of these 72 samples, 29 samples were resistant to CM-induced pY-STAT3 (ΔMFI 〈 2, CM-R) and 43 samples were sensitive (ΔMFI 〉 2, CM-S). Raw counts were normalized and analyzed by EdgeR with GLM algorithm and blocking to control for batch effects from the two datasets. We found that 219 features were significantly differentially expressed (DE; FDR 〈 0.05), with 192 being expressed more highly in the CM-R group. Gene set enrichment analysis identified the "matrisome" gene set, including genes encoding growth factors and matrix proteins, as being significantly enriched in the CM-R group. For example, genes encoding G-CSF, GM-CSF, WNT7B, and integrin B3 were upregulated in CM-R samples. Additionally, a number of non-HOX homeobox genes, including DLX2, DLX3 and MSX2, were increased in CM-R samples. There was no difference in the mean %blasts for the samples in the CM-R and CM-S groups, arguing against the increased expression of matrix-related genes being due to a higher proportion of non-blast cells in the CM-R samples. Our integration of gene expression with inducible STAT3/5 responses will yield novel insights into extrinsic survival signaling and mechanisms of dysfunction. 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: 2018
    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: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1744-1744
    Abstract: Chronic inflammation of the colon increases cancer development risk. Ulcerative colitis, characterized by excessive inflammation initiated by innate immune cells and exacerbated by a dysregulation in adaptive immunity, can give rise to colitis-associated colon cancer (CAC). Whereas overactivity of effector T cells and loss of immunosuppressive cells are hallmarks of ulcerative colitis, the opposite is true for CAC, with CAC tumors exhibiting a lack of effector T cell infiltration and a preponderence of immunosuppressive Treg cells and myeloid-derived suppressor cells (MDSCs). Recently, hypoxia has been identified as a potential driver in the pathogenesis of ulcerative colitis, with hypoxia persisting upon progression to CAC tumor formation. We have previously demonstrated that (i) hypoxia generates an immunosuppressive tumor microenvironment that limits effector T cell infiltration and activation, (ii) OMX, a first-in-class anti-cancer therapy designed to reverse tumor hypoxia to enhance immunotherapeutic efficacy, accumulates in preclinical rodent and spontaneous canine tumors and reduces tumor hypoxia, and (iii) OMX promotes effector T cell infiltration, reduces Treg cells, and enhances checkpoint inhibitor efficacy, resulting in greater tumor control. Given that CAC tumors are hypoxic and immunosuppressed, we hypothesized that hypoxia drives CAC tumor immunosuppression, and accordingly, that reversal of hypoxia with OMX may restore immunosensitivity and elicit an anti-tumor response. Here, using a chemically induced mouse model of CAC generated by administering azoxymethane (AOM) followed by repeated cycles of dextran sulfate sodium (DSS) exposure, we show that OMX treatment exhibits anti-tumor efficacy in advanced CAC tumors. We characterized CAC tumor progression from 8 to 12 weeks post-tumor induction, and confirmed previous reports that advanced CAC tumors are indeed hypoxic, and that immunosuppressive Treg cells and MDSCs are more abundant in CAC tumors relative to adjacent normal mucosa or control non-AOM/DSS-treated colons. Moreover, we observed a negative correlation between hypoxia and CD8+ T cell infiltration into CAC tumors. OMX single agent treatment reduced both CAC tumor number and total CAC tumor burden. Of note, OMX treatment reversed colon length shortening that was characteristic of tumor-bearing mice, indicative of a restoration of colon crypt regeneration and hence normal colon biology. Investigations into the immunological mechanism(s) responsible for OMX anti-tumor efficacy are currently underway. Taken together, our data suggest that OMX, by delivering oxygen to hypoxic CAC tumor regions, may be sufficient to induce an immunological change in the CAC tumor microenvironment from an immunosuppressive to an immunopermissive state, leading to tumor responses and a restoration of normal physiology. Citation Format: Kevin G. Leong, Yuqiong Pan, Changan Guo, Padmini Narayanan, Jonathan A. Winger, Stephen P. Cary, Natacha Le Moan, Ana Krtolica. Reversal of advanced colitis-associated colon cancer by OMX, a novel oxygen carrier that immunosensitizes the hypoxic tumor microenvironment [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1744.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Cambridge University Press (CUP) ; 1985
    In:  International Astronomical Union Colloquium Vol. 87 ( 1985), p. 397-403
    In: International Astronomical Union Colloquium, Cambridge University Press (CUP), Vol. 87 ( 1985), p. 397-403
    Abstract: We report fast photometric observations on AM CANUM VENATICORUM (AM CVn) the ultra short period, hydrogen deficient variable. We have detected on 24th February, 1985 an intense flare of (Δm)peak≈0.34 in white light lasting over 200s. Following this flare we observe an enhanced double humped structure lasting for 1051s which is the dominant periodicity exhibited by AM CVn. We have also detected the 525s and 1051s periods. In addition, we report flickerings, lasting typically 1-2 minutes, that are characteristic of cataclysmic variables.
    Type of Medium: Online Resource
    ISSN: 0252-9211
    Language: English
    Publisher: Cambridge University Press (CUP)
    Publication Date: 1985
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood Advances, American Society of Hematology, Vol. 3, No. 24 ( 2019-12-23), p. 4215-4227
    Abstract: Atovaquone induces AML blast apoptosis and prolongs survival in AML xenografts. Atovaquone induces proapoptotic signaling and inhibits the mTOR pathway through upregulation of ATF4 and also suppresses OXPHOS.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 2876449-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Society of Hematology ; 2019
    In:  Blood Vol. 134, No. Supplement_1 ( 2019-11-13), p. 3731-3731
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 3731-3731
    Abstract: Despite aggressive chemotherapy, relapse occurs in almost half of children with acute myeloid leukemia (AML), with very dismal survival. Novel and mechanism-driven therapies are desperately needed to conquer chemotherapy resistance and leukemic stemness in pediatric AML. Within the bone marrow niche, stromal cells protect leukemia cells from chemotherapy, maintain leukemic stemness, and eventually lead to disease recurrence. We developed an in vitro AML cell-stromal cell co-culture model to mimic bone marrow microenvironment. Stroma-leukemia cell interaction leads to activation of various signaling molecules in AML cells that allow them to evade apoptosis. One such example is extracellular signal-regulated kinases 1/2 (ERK1/2), important pro-survival proteins. ERK1/2 are activated by the Ras/Raf/ mitogen-activated protein kinase kinase (MAPK/ERK kinase or MEK) pathway downstream of signals from the stroma. We recently showed that stromal co-culture activates ERK1/2 in pediatric AML samples, contributing to chemotherapy resistance (Long, et al, 2017, Oncotarget, 8:90037). To identify genes that are regulated in AML cells by ERK1/2 activation, 4 pediatric AML samples were cultured alone, or co-cultured with mOrange-expressing stroma for 24 hours, in the presence or absence of a selective MEK inhibitor, selumetinib (1 μM). Thereafter, cells were flow sorted to exclude mOrange+ stroma and CD45high/SSClow lymphocytes. Sorted AML cells underwent total RNA extraction for nCounter® PanCancer Pathways Panel (Nanostring Technologies) gene expression profiling study. We focused on the genes that were either up- or down-regulated by co-culture with stroma, and changed in the reverse direction by the addition of selumetinib. We chose a few genes among the list (BMP2, BNIP3, H2AFX, DUSP2, FZD3, BCL2L1, CHEK2) that are reported to be involved in oncogenesis, chemotherapy resistance, cell growth and survival. Using qRT-PCR, we confirmed bone morphogenic protein 2 (BMP2) to be upregulated in AML cells by stroma, and the effect of stroma was reversed by selumetinib. Further, we confirmed the same change of BMP2 at the protein level by FACS. Smad1, 5 and 8 are transcriptional factors immediately downstream from BMP receptors and play a central role in BMP signal transduction. Using FACS we discovered stroma-induced activation of Smad 1/8 in pediatric AML patient samples, which was partially alleviated by selumetinib and a selective BMP inhibitor, K02288 (10 μM). BMPs are growth factors that belong to the transforming growth factor beta (TGF-beta) superfamily and are thought to be involved in stem cell properties such as self-renewal. To determine if the BMP-Smad pathway plays a role in chemotherapy resistance, 4 pediatric AML patient samples were cultured on or off stromal cells, and treated with cytarabine (10 μM) with or without K02288 (10 μM) for 24h. Cells were analyzed for cytarabine-induced apoptosis with Annexin V staining by FACS, excluding stromal cells and lymphocytes. K02288 treatment did not alter cytarabine-induced apoptosis. We next tested the potential role of BMP-Smad pathway in leukemic self-renewal in pediatric AML samples. Pediatric AML patient samples were plated at 50,000/ml or 100,000/ml in Methocult medium (H-4535) to quantify stem and progenitor cells. Samples were treated with vehicle or K02288 (10 μM). Colonies and viable cells were counted and normalized to control 7-14 days after plating. Harvested cells were stained for CD34, CD14 and CD11b to evaluate differentiation by FACS. Remaining cells were replated at the same density for 2 more rounds. We found that K02288 reduced colony counts (e.g., 100±0% in vehicle control, v. 25±10% in K02288, n=5, p 〈 0.002, post 2nd plating) and decreased the number of viable cells at 2nd and 3rd plating (Figure 1). The BMP inhibitor also promoted differentiation of leukemic stem cell in pediatric AML samples, as evidenced by increased CD14 and CD11b expression. In contrast, for normal pediatric bone marrow samples, K02288 did not change colony counts or the number of viable cells, and it did not promote differentiation. Those data suggested BMP-Smad is likely to contribute to leukemic stemness in pediatric AML without disrupting normal hematopoietic stem cells. Therefore, BMP-Smad pathway may be a promising therapeutic target to reduce leukemia burden and improve survival for children with AML. 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: 2019
    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: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1723-1723
    Abstract: Despite improvements in treatment modalities, pancreatic cancer remains the fourth leading cause of cancer mortality in the US. The 5-year survival rate is less than 6% because of the extent of metastases at the time of diagnosis. The treatment of pancreatic cancer remains challenging however there is much optimism with targeted strategies and trial designs. The interrogation of pathway centric targets that regulate the regenerative and metastatic activities in the tumor, adequate molecular-hypothesis driven trial designs with robust prognostic and predictive markers of response are necessary for the development of effective therapeutic strategies for long term anti-tumor benefit. Here, we examined the efficacy of CEP1430 and CEP1507 to target cancer stem cells (CSC) and circulating tumor cells (CTC) in human pancreatic cancer. The pancreatic CSCs expressed the following markers(CD133,CD44,CXCR4,SSEA3/4,Oct4,ALDH,Telomerase, & Nestin). In vitro CEP1430 inhibited the growth of CSCs whereas CEP1507 inhibited CTCs that expressed the following markers [CK19, CEA, CA19-9,CK8 /18,h-TERT,C-MET,CK20,EpCAM, Vimentin, E- Cadherin, MUC1, CEA CAM5,CD45 neg]. Gemcitabine on the other hand suppressed the viability of non-CSCs (differentiated tumor Cells) by 70%. In accord, in vivo studies showed that CEP1507 when combined with gemcitabine eliminated the engraftment of human pancreatic cancer CTCs by 85% (n = 20,p & lt;0.001). In contrast, CEP1430 in combination with gemcitabine selectively inhibited CSCs by 80% (n = 20,p & lt;0.001) and was more effective than the individual agents. This data demonstrates that CEP1430 and CEP1507 that target CSCs and CTCs respectively may change the treatment paradigm and enhance the efficacy of gemcitabine in advanced pancreatic cancer. Further studies will evaluate the full potential of this approach to target the CSCs - and metastasis-initiating cells (CTCs) as combination therapy against locally advanced and metastatic pancreatic cancer. Citation Format: Cristian Sharma, Padmini Narayanan, Michael Sharma, Robert Rodriguez, Miriam Navel, Donna Stanton, Natalee Amezcua, Shaleekha Sharma, Mandana Amiri, Jitesh Jani, Sherven Sharma, Jay P. Sharma. Novel drug candidates CEP1430 and CEP1507 against cancer stem cells and circulating tumor cells in advanced stage pancreatic cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1723.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Electrochimica Acta, Elsevier BV, Vol. 236 ( 2017-05), p. 359-370
    Type of Medium: Online Resource
    ISSN: 0013-4686
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2017
    detail.hit.zdb_id: 1483548-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Clinical and Translational Science, Wiley, Vol. 1, No. 3 ( 2008-12), p. 209-214
    Type of Medium: Online Resource
    ISSN: 1752-8054 , 1752-8062
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2008
    detail.hit.zdb_id: 2433157-0
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2013
    In:  The Journal of Immunology Vol. 191, No. 10 ( 2013-11-15), p. 5196-5203
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 191, No. 10 ( 2013-11-15), p. 5196-5203
    Abstract: LPS activates platelets through TLR4, aiding productive sepsis, with stimulated splicing and translation of stored heteronuclear pro–IL-1β RNA. Although the IL-1R type 1 (IL-1R1) receptor for IL-1 shares downstream components with the TLR4 receptor, platelets are not known to express IL-1R1, nor are they known to respond to this cytokine. We show by flow cytometry and Western blotting that platelets express IL-1R1, and that IL-1β and IL-1α stimulate heteronuclear I-1β splicing and translation of the newly made mRNA in platelets. Platelets also respond to the IL-1β they make, which is exclusively associated with shed microparticles. Specific blockade of IL-1R1 with IL-1R antagonist suppressed platelet stimulation by IL-1, so IL-1β stimulates its own synthesis in an autocrine signaling loop. Strikingly, IL-1R antagonist inhibition, pharmacologic or genetic suppression of pro–IL-1β processing to active cytokine by caspase-1, or blockade of de novo protein synthesis also blocked LPS-induced IL-1β mRNA production. Robust stimulation of platelets by LPS therefore also required IL-1β amplification. Activated platelets made IL-1β in vivo as IL-1β rapidly accumulated in occluded murine carotid arteries by posttranscriptional RNA splicing unique to platelets. We conclude that IL-1β is a platelet agonist, that IL-1β acts through an autocrine stimulatory loop, that an IL-1β autocrine loop is required to amplify platelet activation by LPS, and that platelets immobilized in occlusive thrombi are activated over time to produce IL-1β. IL-1 is a new platelet agonist that promotes its own synthesis, connecting thrombosis with immunity.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2013
    detail.hit.zdb_id: 1475085-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 4121-4121
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 4121-4121
    Abstract: Preclinical studies have shown that GR activation leads to decreased response to antimetabolites, taxanes, and platinum agents, while GR inhibition enhances therapeutic efficacy. The novel, selective GR antagonist ORIC-101 is under clinical evaluation in combination with anticancer therapies. In this study we set out to assess which cancer types and chemotherapeutics are responsive to combination with ORIC-101. Specifically, we employed caspase assays, in vitro and in vivo tumor growth inhibition studies, and transcriptional profiling in a panel of preclinical models spanning triple-negative breast, ovarian, non-small cell lung cancers, pancreatic ductal adenocarcinoma, hepatocellular carcinoma, sarcoma, and renal cell carcinoma. We found that ORIC-101 reverses GR-mediated antiapoptosis effects towards diverse chemotherapeutics, including paclitaxel, gemcitabine, and cisplatin, across the seven cancer types. Furthermore, ORIC-101 completely overcomes GR-driven chemoprotection and tumor growth in colony formation assays in vitro and in xenograft studies in vivo. At the molecular level, transcriptional profiling and pathway enrichment analysis showed that ORIC-101 fully reverses GR-activated pathways that are directly involved in drug resistance, such as epithelial-to-mesenchymal transition and antiapoptosis, supporting a pan-cancer role of GR as a mediator of therapy resistance. Altogether, ORIC-101, a selective and potent GR antagonist, overcomes resistance to common chemotherapeutics across multiple cancer models. Clinical evaluation of ORIC-101 in combination with nab-paclitaxel is currently ongoing in advanced solid tumors. Citation Format: Haiying Zhou, Shravani Barkund, Aleksandr Pankov, Ganapati Hegde, Wayne Kong, Padmini Narayanan, Jessica D. Sun, Omar Kabbarah, Lori S. Friedman, Anneleen Daemen. ORIC-101 overcomes resistance to diverse chemotherapeutics across cancer types [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4121.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    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