Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 1128-1128
    Abstract: Advances in risk-adapted cytotoxic chemotherapy, hematopoietic stem cell transplantation and supportive care have contributed to significant improvements in the survival of patients with acute lymphoid leukemia (ALL) and acute myeloid leukemia (AML) over the past few decades. However, despite such progress, a significant percentage of both adult and pediatric leukemia patients become refractory to therapy or relapse and eventually die of disease. Hence, there remains an urgent need for the development of effective and targeted therapies for acute leukemia. Recent genetic profiling of solid and hematologic malignancies has identified epigenetic factors as a critical group of genes recurrently mutated in cancer. Additionally, epigenetic dysregulation has been shown to play an important role in the development, progression and maintenance of leukemia. Therefore, pharmacological inhibition of epigenetic factors represents a potential avenue for the development of novel epigenetic-targeted therapies. In order to identify epigenetic vulnerabilities in leukemia, we developed an epigenetic-focused shRNA screen to search for novel therapeutic targets in human leukemia cell lines both in vitro and in vivo. Specifically, T- and B-ALL cell lines were transduced with a library of shRNAs targeting 449 genes including epigenetic readers, writers and erasers and other chromatin-related factors. Selected cells were subsequently cultured in vitro and concurrently injected into mice. Engraftment of inoculated cells and disease progression were monitored through bioluminescence imaging. Amongst the universe of epigenetic regulatory proteins, the arginine methyl transferase, PRMT5, emerged as the most significantly depleted factor in both in vitro and in vivo screenings. Chemical inhibition of PRMT5 enzymatic activity effectively reduced protein symmetric dimethyl arginine methylation, altered splicing, inhibited cell growth and promoted apoptosis of both ALL and AML cell lines in vitro. In addition, inhibition of PRMT5 in vivo using patient-derived xenograft (PDX) T-ALL mouse models demonstrated diminished tumor growth and prolonged survival. Notably, quantification of peripheral blood cell numbers showed that pharmacologic PRMT5 inhibition was well tolerated and did not affect normal hematopoiesis in mice suggesting that a therapeutic window exists for anticancer drugs targeting PRMT5 in acute leukemia. Overall, our data indicates that pre-mRNA processing and in particular RNA splicing modulation may represent novel therapeutic targets in leukemia. Note: This abstract was not presented at the meeting. Citation Format: Yunyue Wang, Hui Huang, Daniel Diolaiti, Marta Sanchez Martin, Beata Modzelewski, Lianna J. Marks, Allison R. Rainey, Ervin S. Gaviria, Maria L. Sulis, Filemon S. Dela Cruz, Adolfo A. Ferrando, Andrew L. Kung. Identification of arginine methyltransferase PRMT5 as a novel therapeutic target in T-cell acute lymphoblastic leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1128. doi:10.1158/1538-7445.AM2017-1128
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    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 ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 16 ( 2017-08-15), p. 4365-4377
    Abstract: ETV6-RUNX1 is associated with the most common subtype of childhood leukemia. As few ETV6-RUNX1 carriers develop precursor B-cell acute lymphocytic leukemia (pB-ALL), the underlying genetic basis for development of full-blown leukemia remains to be identified, but the appearance of leukemia cases in time-space clusters keeps infection as a potential causal factor. Here, we present in vivo genetic evidence mechanistically connecting preleukemic ETV6-RUNX1 expression in hematopoetic stem cells/precursor cells (HSC/PC) and postnatal infections for human-like pB-ALL. In our model, ETV6-RUNX1 conferred a low risk of developing pB-ALL after exposure to common pathogens, corroborating the low incidence observed in humans. Murine preleukemic ETV6-RUNX1 pro/preB cells showed high Rag1/2 expression, known for human ETV6-RUNX1 pB-ALL. Murine and human ETV6-RUNX1 pB-ALL revealed recurrent genomic alterations, with a relevant proportion affecting genes of the lysine demethylase (KDM) family. KDM5C loss of function resulted in increased levels of H3K4me3, which coprecipitated with RAG2 in a human cell line model, laying the molecular basis for recombination activity. We conclude that alterations of KDM family members represent a disease-driving mechanism and an explanation for RAG off-target cleavage observed in humans. Our results explain the genetic basis for clonal evolution of an ETV6-RUNX1 preleukemic clone to pB-ALL after infection exposure and offer the possibility of novel therapeutic approaches. Cancer Res; 77(16); 4365–77. ©2017 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 4479-4479
    Abstract: Chromatin remodeling complexes are crucial for the viability of the cells due to their role in regulating interactions between DNA and histones and, therefore, modifying the accessibility of the genetic information to the transcriptional machinery. This relevance can also been seen in the SWI/SNF complex that has been associated with cancer in the last deep-sequencing efforts on tumoral genomes. BRG1 is the helicase/ATPase catalytic subunit of the SWI/SNF complex and it is frequently lost in NSCLC cell lines with a high mutation rate. In primary tumors, the loss of expression of BRG1 is also frequent, however it cannot be explained by mutations or by promoter hypermethylation. In this study we have focused on the regulation of the expression of BRG1 by microRNAs. These are small RNA molecules that do not code for any protein but they have key roles in gene expression regulation by binding to the 3’UTR of the transcripts. In addition, their levels have been found altered in several diseases including cancer. Our hypothesis is that the deregulation observed in BRG1 levels in primary tumors can be associated with changes in the microRNAs that control its expression. We have studied the 3’UTR of BRG1 by performing a Rapid Amplification of 3’ cDNA Ends (3’ RACE) and we have cloned the resulting 3’UTRs after a luciferase coding sequence of a plasmid in other to use the luminescence signal as a method to analyze the binding of the microRNAs tested to the 3’UTR of BRG1. Previous bioinformatic analyses were performed to choose the best microRNAs that could potentially regulate BRG1. Our preliminary results indicate that some microRNAs are able to bind to the 3’UTRs and successfully repress the expression of catalytic subunit of SWI/SNF. Importantly, one of these microRNAs is well known for developing oncogenic activities in lung cancer and it has been related with poor prognosis. In conclusion, the activity of the SWI/SNF complex can be regulated by microRNAs and this regulation may be relevant during lung carcinogenesis. From the other perspective, SWI/SNF complex could also impact in lung carcinogenesis regulating microRNAs expression, an aspect that we are currently evaluating. Citation Format: Paola Peinado Fernández, Isabel Fernández Coira, Eva E. Rufino Palomares, Octavio A. Romero, Chanatip Metheetrairut, Laura Boyero Corral, Julián Carretero, Esther Farez Vidal, Marta Cuadros Celorrio, Fernando Reyes Zurita, Victoria Sánchez Martín, Carlos Baliñas Gavira, Jose A. Lupiáñez Cara, Montse Sánchez Céspedes, Frank Slack, Pedro P. Medina. Unveiling the relationship between the SWI/SNF chromatin remodeling complex and noncoding RNAs. [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 4479.
    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 ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 3 ( 2011-02-01), p. 832-841
    Abstract: In bladder cancer, increased caveolin-1 (Cav-1) expression and decreased Src expression and kinase activity correlate with tumor aggressiveness. Here, we investigate the clinical and functional significance, if any, of this reciprocal expression in bladder cancer metastasis. We evaluated the ability of tumor Cav-1 and Src RNA and protein expression to predict outcome following cystectomy in 257 patients enrolled in two independent clinical studies. In both, high Cav-1 and low Src levels were associated with metastasis development. We overexpressed or depleted Cav-1 and Src protein levels in UMUC-3 and RT4 human bladder cancer cells and evaluated the effect of this on actin stress fibers, migration using Transwells, and lung metastasis following tail vein inoculation. Cav-1 depletion or expression of active Src in metastatic UMUC-3 cells decreases actin stress fibers, cell migration, and metastasis, while Cav-1 overexpression or Src depletion increased the migration of nonmetastatic RT4 cells. Biochemical studies indicated that Cav-1 mediates these effects via its phosphorylated form (pY14), whereas Src effects are mediated through phosphorylation of p190RhoGAP and these pathways converge to reduce activity of RhoA, RhoC, and Rho effector ROCK1. Treatment with a ROCK inhibitor reduced UMUC-3 lung metastasis in vivo, phenocopying the effect of Cav-1 depletion or expression of active Src. Src suppresses whereas Cav-1 promotes metastasis of bladder cancer through a pharmacologically tractable common downstream signaling pathway. Clinical evaluation of personalized therapy to suppress metastasis development based on Cav-1 and Src profiles seems warranted. Cancer Res; 71(3); 832–41. ©2010 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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 ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 965-965
    Abstract: Activating mutations of NOTCH1 are common in T-cell acute lymphoblastic leukemia (T-ALL), making NOTCH signaling a promising target for drugs such as γ-secretase inhibitors (GSIs), which block a proteolytic cleavage required for NOTCH activation. Aberrant activation of the PI3K-AKT pathway due to mutational loss of PTEN is found in 20% of human T-ALLs and has been linked with in vitro resistance to GSIs. Still, the specific mechanisms driving the antileukemic effects of NOTCH inhibition and the role of PTEN loss in resistance to anti-NOTCH therapies in T-ALL remain poorly understood. Here we used a mouse model of NOTCH1-induced leukemia built into conditional-inducible Pten knockout mice to demonstrate that genetic loss of Pten results in GSI resistance, accelerated mortality and progression under therapy in vivo. Importantly, gene expression profiling of tumor cells upon NOTCH inhibition showed that while NOTCH direct target genes are downregulated in both Pten-positive and Pten-deleted tumors, Pten loss results in global reversal of much of the transcriptional effects of NOTCH inhibition. Thus, Pten loss rescued the downregulation of genes involved in anabolic pathways and the upregulation of genes involved in catabolic pathways and autophagy induced by GSI treatment in Pten wild type leukemias. Consistently, electron microscopy and LC3 analysis demonstrated increased autophagy in NOTCH1 induced tumors upon GSI treatment, which was reversed upon Pten deletion. Moreover, global metabolomics analyses demonstrated that NOTCH inactivation induces a global anabolic shutdown in T-ALL with a marked block of glycolysis and glutaminolysis in Pten wild type tumor cells. Of note, flux studies using 13C-labeled glucose and 13C-labeled glutamine pointed to decreased metabolism with a specific block in glutaminolysis as critical effectors of the antileukemic effects of GSIs in T-ALL. Notably, each of the metabolic effects associated with NOTCH inhibition was effectively reversed upon genetic loss of Pten, supporting that metabolic rescue induced by Pten loss may mediate resistance to GSI therapy. Consistently, treatment with methyl pyruvate, a membrane soluble metabolite, effectively rescued the antileukemic effects of NOTCH inhibition, while, inhibition of glutaminolysis with a glutaminase inhibitor (BPTES) strongly and synergistically enhanced the antileukemic effects of NOTCH inhibition in T-ALL. Overall, these results formally demonstrate that loss of PTEN induces in vivo resistance to NOTCH inhibition in T-ALL; highlight the fundamental role of NOTCH1 in the control of tumor cell metabolism; implicates sustained carbon metabolism induced by Pten loss as mechanism of resistance to GSI therapy; and provide the framework for the development of novel therapies targeting glutaminolysis and cell metabolism in T-ALL. Citation Format: Daniel Herranz, Alberto Ambesi-Impiombato1, Jessica Sudderth, Marta Sánchez-Martín, Valeria Tosello, Luyao Xu, Mireia Castillo, Carlos Cordon-Cardo, Andrew L. Kung, Ralph J. DeBerardinis, Adolfo Ferrando. An oncogenic metabolic switch mediates resistance to NOTCH1 inhibition in T-ALL. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 965. doi:10.1158/1538-7445.AM2014-965
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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 ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 4131-4131
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 4131-4131
    Abstract: Checkpoint inhibitors (CPIs) have become the cornerstone of immune-based oncology therapy; still many cancer patients do not benefit from these agents. Resistance to checkpoint inhibitors is due in part to the presence of immunosuppressive molecules which prevent immune activation despite T cell checkpoint inhibition. Aryl Hydrocarbon Receptor (AHR) is a transcription factor that mediates the immune response in multiple innate and adaptive immune cells subsequent to binding of a diverse set of endogenous and exogenous ligands. One such AHR agonist is kynurenine. Kynurenine, metabolized from tryptophan by indoleamine-pyrrole 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase 2 (TDO2), binds to AHR and leads to a net immunosuppressive tumor microenvironment. Given that kynurenine can be synthesized by both IDO1 and TDO2 and that AHR is activated by endogenous ligands other than kynurenine, AHR inhibition provides a novel approach to reverse immunosuppression in a broad range of tumor types. We demonstrated that AHR antagonism affects multiple immune cell types and can lead to pro-inflammatory phenotypes in human T cells and myeloid cells in vitro and in murine tumor models. AHR antagonism inhibits growth in the B16-IDO, CT26 and MC38 models and reverses the immunosuppressive microenvironment as indicated by changes in T cell and myeloid cell populations. Immune signature changes characterized via Nanostring in multiple tumor models treated with AHR antagonists were also assessed. Oral dosing of AHR antagonists led to tumor growth inhibition as a single agent and increased anti-tumor activity when combined with checkpoint inhibitors. In addition, beneficial anti-tumor activity occurred with AHR antagonists combined with chemotherapy or with radiation therapy in syngeneic mouse tumor models. Our data indicate that reversing AHR-mediated immune suppression in the tumor microenvironment drives anti-tumor activity alone and in combination with other therapeutic modalities. Overall, our data demonstrates that AHR is an attractive target for reversing immune suppression in tumors. Therefore, we are developing AHR antagonists and translational insights to treat patients most likely to benefit from this approach. Citation Format: Jeremy Tchaicha, Silvia Coma, Meghan Walsh, Jill Cavanaugh, Marta Sanchez-Martin, X. Michelle Zhang, Alfredo Castro, Jeff Ecsedy, Mark Manfredi, Karen McGovern. Overcoming aryl hydrocarbon receptor mediated tumor immunosuppression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4131.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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