GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (15)
  • 1
    In: Cancer Discovery, American Association for Cancer Research (AACR), ( 2022-11-11), p. OF1-OF26
    Abstract: Diffuse midline gliomas are uniformly fatal pediatric central nervous system cancers that are refractory to standard-of-care therapeutic modalities. The primary genetic drivers are a set of recurrent amino acid substitutions in genes encoding histone H3 (H3K27M), which are currently undruggable. These H3K27M oncohistones perturb normal chromatin architecture, resulting in an aberrant epigenetic landscape. To interrogate for epigenetic dependencies, we performed a CRISPR screen and show that patient-derived H3K27M-glioma neurospheres are dependent on core components of the mammalian BAF (SWI/SNF) chromatin remodeling complex. The BAF complex maintains glioma stem cells in a cycling, oligodendrocyte precursor cell–like state, in which genetic perturbation of the BAF catalytic subunit SMARCA4 (BRG1), as well as pharmacologic suppression, opposes proliferation, promotes progression of differentiation along the astrocytic lineage, and improves overall survival of patient-derived xenograft models. In summary, we demonstrate that therapeutic inhibition of the BAF complex has translational potential for children with H3K27M gliomas. Significance: Epigenetic dysregulation is at the core of H3K27M-glioma tumorigenesis. Here, we identify the BRG1–BAF complex as a critical regulator of enhancer and transcription factor landscapes, which maintain H3K27M glioma in their progenitor state, precluding glial differentiation, and establish pharmacologic targeting of the BAF complex as a novel treatment strategy for pediatric H3K27M glioma.
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 4030-4030
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 4030-4030
    Abstract: Cytoplasmic dislocation and loss of nucleolar localization of the chaperone protein NPM1c occurs in 30% of AML following a frame shift C-terminal mutation in exon 12 of NPM1 (creating NPM1c), which contributes to differentiation arrest, growth and survival of AML stem progenitor cells. In present studies, we determined that knock out (KO) of mutant (mt) NPM1 (NPM1c) in the AML OCI-AML3, utilizing targeted gRNA (compared to control) and CRISPR-Cas9, induced growth arrest, morphologic differentiation and loss of viability of the KO cells over 5 to 9 days. This was associated with depletion of protein levels of NPM1c, c-Myc, and MEIS1, but upregulation of PU.1, RUNX1, CD11b, p21 and NOXA levels. ChIP-Seq analysis with H3K27Ac antibody 5d after KO demonstrated reduced peak density at the super-enhancers of MYC, MEIS1, and HOXA9. RNA-Seq analysis showed negative enrichment of HALLMARK gene sets of mRNA expression of MEIS1/HOXA9, MYC targets, ribosome/translation and cell cycle. Compared to OCI-AML3 with NPM1c, OCI-AML3 with NPM1 KO exhibited abrogation of the Menin inhibitor SNDX-50469-induced differentiation and apoptosis. Additionally, NPM1 KO also significantly reduced sensitivity of OCI-AML3 cells to apoptosis induced by targeted agents previously shown to demonstrate efficacy against AML cells with NPM1c, including KPT-330 (XPO1 inhibitor), homoharringtonine (protein translation inhibitor), and anti-AML chemotherapeutic agents, including daunorubicin, cytarabine and etoposide. However, NPM1 KO sensitized OCI-AML3 cells to ATRA (all trans retinoic acid)-induced differentiation and loss of viability, which was associated with marked induction of p21 and CD11b. Utilizing RNA-Seq signature of NPM1c KO and interrogating the LINCS1000-CMap data set of gene expression signatures, we determined that among the top expression mimickers were several pan-HDAC inhibitors and a WEE1 kinase inhibitor. Treatment with adavosertib (WEE1 inhibitor, MK-1775) or romidepsin (HDAC inhibitor) dose-dependently induced apoptosis in OCI-AML3 cells, as well as in 5 samples of patient-derived (PD) AML cells with NPM1c. Moreover, co-treatment with adavosertib and romidepsin synergistically induced lethality in OCI-AML3 and PD AML cells with NPM1c. This was associated with marked reduction in protein expressions of c-Myc, c-Myb, MEIS1 and CDK4/6, but upregulation of TP53 and p21 levels. These findings highlight that presence of NPM1c mechanistically regulates the sensitivity of AML cells to Menin inhibitor, ATRA and anti-AML chemotherapeutic agents. They also identify the novel non-chemotherapy combination of pan-HDAC inhibitor and WEE1 inhibitor for its potential synergistic activity against AML with NPM1c. Citation Format: Christopher P. Mill, Warren C. Fiskus, John A. Davis, Courtney D. DiNardo, Christine Birdwell, Qi Jin, Koichi Takahashi, Joseph D. Khoury, Kapil N. Bhalla. Efficacy of novel combination therapy against AML cells with mutant NPM1 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 4030.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 24_Supplement ( 2010-12-15), p. P6-14-06-P6-14-06
    Abstract: Background: Several gene signature-based predictors of response to targeted drugs have been proposed in the literature but none has been prospectively tested as patient selection tools in the clinic. The goal of this trial is to assess the positive predictive value of 3 conceptually different multi-gene signatures as predictors of response to the multitargeted kinase inhibitor dasatinib. Methods: This clinical trial requires biopsy of a metastatic lesion for gene expression profiling and employs a parallel, multi-arm, two-step, phase II design. Three markers are assessed including a (i) cell-line derived dasatinib-sensitivity signature, (ii) a src-pathway activity signature and (iii) a dasatinib target index calculated as the weighted average expression of all known dasatinib targets. Only markerpositive patients are treated with dasatinib 100 mg po daily and each marker arm is considered as a separate study with early stopping rules for futility (minimum sample size 9, maximum sample size 40/marker arm). A predictor is considered worthy of further study if the clinical benefit rate (i.e. positive predictive value) is ≥25%. Results: Forty seven patients were accrued from July 2009 through June, 2010, 49 biopsies were performed (soft tissues n=31, liver n=8, bone n=3, lung n=1, adrenal gland n=1), 6 samples had poor cellularity and 3 failed array QC. There was no patient recall, hospitalization or emergency room visit due to biopsy procedure. The median time from biopsy to genomic prediction result was 5 days (range 3-7). Twenty three (57%) patients had positive result for at least 1 predictor (5 were positive for 2) and 20 are receiving therapy (3 withdraw or progressed before therapy began). Responses as of June 2010; Target index arm (n=9): 5 PD (progressive disease), 4 SD (3 stable disease at 8 weeks 1 SD at 16 weeks); SRC Pathway arm (n=5): 3 PD, 2 SD at 8 weeks; Cell line predictor arm (n=6): 2 PD, 1 SD at 8 weeks, 3 not yet reached response evaluation. None of the 3 predictive marker arms have met early stopping yet and accrual is ongoing. Conclusion: Gene-expression signature based patient selection for targeted therapy is feasible and FNA biopsies of metastatic lesions for genomic testing are safe. Updated efficacy results will be reported. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr P6-14-06.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 16, No. 21 ( 2010-11-01), p. 5351-5361
    Abstract: Purpose: We examined in a prospective, randomized, international clinical trial the performance of a previously defined 30-gene predictor (DLDA-30) of pathologic complete response (pCR) to preoperative weekly paclitaxel and fluorouracil, doxorubicin, and cyclophosphamide (T/FAC) chemotherapy, and assessed if DLDA-30 also predicts increased sensitivity to FAC-only chemotherapy. We compared the pCR rates after T/FAC versus FACx6 preoperative chemotherapy. We also did an exploratory analysis to identify novel candidate genes that differentially predict response in the two treatment arms. Experimental Design: Two hundred and seventy-three patients were randomly assigned to receive either weekly paclitaxel × 12 followed by FAC × 4 (T/FAC, n = 138), or FAC × 6 (n = 135) neoadjuvant chemotherapy. All patients underwent a pretreatment fine-needle aspiration biopsy of the tumor for gene expression profiling and treatment response prediction. Results: The pCR rates were 19% and 9% in the T/FAC and FAC arms, respectively (P & lt; 0.05). In the T/FAC arm, the positive predictive value (PPV) of the genomic predictor was 38% [95% confidence interval (95% CI), 21-56%], the negative predictive value was 88% (95% CI, 77-95%), and the area under the receiver operating characteristic curve (AUC) was 0.711. In the FAC arm, the PPV was 9% (95% CI, 1-29%) and the AUC was 0.584. This suggests that the genomic predictor may have regimen specificity. Its performance was similar to a clinical variable–based predictor nomogram. Conclusions: Gene expression profiling for prospective response prediction was feasible in this international trial. The 30-gene predictor can identify patients with greater than average sensitivity to T/FAC chemotherapy. However, it captured molecular equivalents of clinical phenotype. Next-generation predictive markers will need to be developed separately for different molecular subsets of breast cancers. Clin Cancer Res; 16(21); 5351–61. ©2010 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 19, No. 9 ( 2013-05-01), p. 2460-2472
    Abstract: Purpose: We aim to identify tumor-specific alternative splicing events having potential applications in the early detection, diagnosis, prognosis, and therapy for cancers. Experimental Design: We analyzed RNA-seq data on 470 clear cell renal cell carcinomas (ccRCC) and 68 kidney tissues to identify tumor-specific alternative splicing events. We further focused on the fibroblast growth factor receptor 2 (FGFR2) isoform switch and characterized ccRCCs expressing different FGFR2 isoforms by integrated analyses using genomic data from multiple platforms and tumor types. Results: We identified 113 top candidate alternatively spliced genes in ccRCC. Prominently, the FGFR2 gene transcript switched from the normal IIIb isoform (“epithelial”) to IIIc isoform (“mesenchymal”) in nearly 90% of ccRCCs. This switch is kidney specific as it was rarely observed in other cancers. The FGFR2-IIIb ccRCCs show a transcriptome and methylome resembling those from normal kidney, whereas FGFR2-IIIc ccRCCs possess elevated hypoxic and mesenchymal expression signatures. Clinically, FGFR2-IIIb ccRCCs are smaller in size, of lower tumor grade, and associated with longer patient survival. Gene set enrichment and DNA copy number analyses indicated that FGFR2-IIIb ccRCCs are closely associated with renal oncocytomas and chromophobe RCCs (chRCC). A reexamination of tumor histology by pathologists identified FGFR2-IIIb tumors as chRCCs and clear cell papillary RCCs (ccpRCC). Conclusions: FGFR2 IIIb RCCs represent misdiagnosed ccRCC cases, suggesting FGFR2 isoform testing can be used in the diagnosis of RCC subtypes. The finding of a prevalent isoform switch of FGFR2 in a tissue-specific manner holds promise for the future development of FGFR2-IIIc as a distinct early detection biomarker and therapeutic target for ccRCC. Clin Cancer Res; 19(9); 2460–72. ©2013 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 2648-2648
    Abstract: The EVI1 gene is located on 3q26.2 and encodes a zinc fingers-containing transcription factor. Nuclear β-catenin-TBL1/R1-TCF7L2 axis activity and EVI1 expression characterize leukemia stem-progenitor cells, supporting their self-renewal and blocking differentiation. EVI1 is overexpressed in AML with chromosome translocation t(3;3) or inv(3) at 3q26, where the distal GATA2 hematopoietic enhancer is repositioned to induce EVI1 overexpression while repressing GATA2. EVI1 overexpression confers poor response to therapy and inferior relapse-free and overall survival in AML. Tegavivint (BC-2059, Iterion) targets TBL1/R1 and disrupts its binding to β-catenin and TCF7L2, repressing MYC, cyclin D1 and Survivin, leading to apoptosis of AML cells. Here, we determined that treatment with TV (10-100 nM) dose-dependently induced apoptosis in AML cell lines (UCSD-AML1, MUTZ3 and OCI-AML20) and patient-derived (PD) AML cells (AML191 and AML194) with 3q26.2 lesions with/without monosomy 7. This was associated with attenuation of protein levels of EVI1, TCF7L2, c-Myc, c-Myb, RUNX1, CEBPα, c-KIT, BCL2, Bcl-xL and MCL1, but upregulation of CD11b, BIM and cleaved PARP levels. Additionally, by reducing BRD4 occupancy at GATA2 enhancer and repressing EVI1, the pan-BET protein inhibitor OTX015 (100-1000 nM) also dose-dependently induced apoptosis of AML cell lines and PD AML cells with t(3:3)/inv(3). RNA-Seq and gene set enrichment analysis in AML cells showed that TV treatment caused log2 fold-enrichment of gene sets of inflammatory response, TNFα and interferon signaling, TGFβ and apoptosis signaling, but negative enrichment of gene sets of c-Myc, E2F, DNA replication/repair, as well as showed reduction in expression of WNT targets and 17-gene stemness score. Confocal microscopy showed that TV treatment disrupted co-localization EVI1 and β-catenin with TBL1, also confirmed by Proximity Ligation Assay. Mass cytometry (CyTOF) analysis confirmed that TV treatment attenuated EVI1, c-Myc, RUNX1, β-catenin, TBL1/R1, Bcl-xL, BCL2, MCL1 and Ki67 but augmented protein levels of APC and cleaved PARP in phenotypically characterized AML stem cells harboring t(3:3)/inv(3) and EVI1 overexpression (with high expression of CLEC12A, CD123, CD244, CD99). In vitro co-treatment with TV and BCL2 inhibitor venetoclax or OTX015 synergistically induced apoptosis (determined by SynergyFinder) of AML cell lines and the PD AML cells. In the tail-vein infused and engrafted PD AML191 cell model in NSG mice, treatment with TV and/or venetoclax or OTX015 for 6 weeks significantly reduced AML burden and improved overall survival of the NSG mice more than treatment with each drug alone or vehicle control, without any toxicity. These findings highlight that targeted inhibition of TBL1/R1-nuclear β-catenin-TCF7L2 axis combined with BET protein or BCL2 inhibition is effective therapy against AML models harboring 3q26 lesions and EVI1 overexpression. Citation Format: Christine Birdwell, Warren C. Fiskus, Christopher P. Mill, John A. Davis, Qi Jin, Courtney D. DiNardo, Koichi Takahashi, Stephen Horrigan, Tapan M. Kadia, Naval Daver, Kapil N. Bhalla. Novel combination therapies against AML with 3q26 lesions and EVI1 overexpression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2648.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1779-1779
    Abstract: Cholangiocarcinoma (CCA) is a lethal malignancy of the biliary epithelium that can arise in any part of the biliary tree. Surgery is the only curative treatment for CCA, but only ∼30% of patients present with resectable disease. The remaining 70% of patients present with advanced or metastatic disease and, if eligible, undergo systemic chemotherapy with the first-line combination of gemcitabine and cisplatin. This combination was shown in a phase II clinical trial to significantly increase median survival from 8.1 to 11.7 months, compared to gemcitabine alone. In order to improve on current treatment, pre-clinical evaluation of novel therapeutics is essential to improving outcome. Unfortunately, the paucity of data describing characteristics common to CCA make development of targeted therapy difficult. However, as is true for other types of solid tumors, c-Myc expression likely contributes to CCA phenotype: c-Myc expression has been observed in 95% of CCA tumors, and experimental down-regulation of c-Myc decreases the invasive potential of CCA cells in vitro. Recently it has become possible to inhibit expression of c-Myc using BET inhibitors. Therefore, we evaluated the efficacy of the bromodomain (BET) inhibitor JQ1 using in vivo models of CCA. The five patient-derived xenograft (PDX) models of CCA that we developed are the first such models to be reported. These models retain the heterogeneity, architecture and specific genetic characteristics of the primary tumors from which they were derived. We used three of these models to examine whether the BET inhibitor JQ1 inhibited CCA tumor growth and generated expression profiles of vehicle- and drug-treated tumors. We administered 50 mg/kg of JQ1 i.p. daily for 20 days and monitored tumor growth. This treatment regimen was well tolerated by tumor-bearing mice, without apparent toxicity. Our data demonstrate that JQ1 suppressed tumor growth in two of the three models, compared to vehicle control treated mice. The data also showed that JQ1-treated tumors had lower levels of c-Myc RNA (↓5-fold) and protein and of RNA encoding multiple transcriptional targets downstream of this oncogenic transcription factor. We conclude that BET inhibitors such as JQ1 warrant further investigation as potentially effective drugs for the treatment of CCA. Citation Format: Patrick L. Garcia, Aubrey L. Miller, Kelly Kreitzburg, Tracy L. Gamblin, Leona N. Council, John D. Christein, Pablo Arnoletti, Marty Heslin, Sushanth Reddy, Joseph H. Richardson, Eddy S. Yang, Jun Qi, James E. Bradner, Karina J. Yoon. Bromodomain inhibitor JQ1 inhibits cholangiocarcinoma tumor growth in patient-derived xenograft models. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1779. doi:10.1158/1538-7445.AM2015-1779
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 14_Supplement ( 2020-07-15), p. PR11-PR11
    Abstract: High-risk neuroblastoma (NB) is an aggressive tumor of the peripheral sympathetic nervous system. Patients with NB have poor overall survival despite increases in intensity of anti-NB therapy, and survivors are typically left with long-term treatment-related morbidities. Thus, there is a need to develop novel targeted therapies that kill tumor cells without toxicity to normal tissues. We recently demonstrated that NB relies on a set of genes for survival, termed “dependencies.” One NB dependency that regulates numerous other dependencies is the histone acetyltransferase (HAT) enzyme, EP300. EP300 catalyzes the acetylation of histone H3, lysine-27 (H3K27ac) that defines active enhancer and promoter elements. This mark can also be catalyzed by the paralogous protein CBP; however, CBP is not required for NB survival despite generally being expressed in NB. Thus, selective inhibition of EP300 may result in anti-NB effects with minimal toxicity to normal tissues where CBP compensates. Here, we demonstrate that EP300, but not CBP, controls NB cell survival through regulation of the oncogenic enhancer landscape of NB. Conventional small-molecule inhibition of EP300/CBP or CRISPR-cas9-mediated knockout of EP300, but not CBP, results in neuroblastic differentiation associated with loss of the NB lineage-defining and oncogenic core transcriptional regulatory circuitry. All agents targeting EP300 equivalently target CREBBP due to their extensive protein homology. Thus, to pharmacologically eliminate EP300 and spare CBP, we designed a novel proteolysis-targeting chimera (PROTAC) agent (“JQAD1”). JQAD1 is a cereblon-dependent, selective degrader of EP300 with minimal off-target effects on CBP in NB cell lines, low passage primary cells, and in xenografts in vivo. JQAD1 is exceedingly stable and well tolerated in vivo. JQAD1 treatment results in loss of the transcriptional circuitry driving NB, transcriptional collapse. and irreversible commitment of NB cells to apoptosis in vitro and in vivo. This study defines the mechanism by which EP300 centrally regulates NB cell fate through epigenetic regulation of the transcriptional state and provides the first EP300-selective pharmacologic agent for evaluation in a myriad of other EP300-dependent malignancies. This abstract is also being presented as Poster B11. Citation Format: Adam D. Durbin, Virangika Wimalasena, Mark W. Zimmerman, Li Deyao, Elizabeth S. Frank, Paul Park, Ken Morita, Neekesh V. Dharia, Ken N. Ross, Ernst Schonbrunn, Richard A. Young, Brian J. Abraham, Kimberly Stegmaier, A. Thomas Look, Jun Qi. EP300 controls the oncogenic enhancer landscape of high-risk neuroblastoma [abstract]. In: Proceedings of the AACR Special Conference on the Advances in Pediatric Cancer Research; 2019 Sep 17-20; Montreal, QC, Canada. Philadelphia (PA): AACR; Cancer Res 2020;80(14 Suppl):Abstract nr PR11.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 4028-4028
    Abstract: In MLL1 rearranged (MLL1r) AML (~10%), N-terminus of MLL1 gene is fused to the C-terminus of a fusion partner, e.g. AF9, AF4, ENL and ELL, creating MLL1 fusion protein (MLL-FP), which increases expression of leukemogenic HOXA9 and its co-factor MEIS1. In AML with mutant (mt) NPM1 (NPM1c), MLL1 is the main driver of HOXA9, MEIS1 and FLT3, promoting self-renewal and growth of AML stem-progenitor cells (LSCs). Treatment with Menin inhibitor (MI), SNDX-50469 or SNDX-5613, disrupts binding of Menin to its binding pocket in MLL1/2 and MLL1-FP, reducing expressions of their targets and inducing differentiation and apoptosis. In the phase I/II AUGMENT-101 clinical trial, SNDX-5613 monotherapy was well tolerated and achieved objective remissions in patients with previously treated relapsed/refractory AML harboring MLL1r or NPM1c. However, majority of patients either fail to respond or eventually relapse. A minority of MLL1r AML (~9%) also exhibit a co-mutation in TP53, which is known to confer therapy resistance and poor outcome in AML. This creates the need to develop MI-based synergistic combinations with superior efficacy against patient-derived (PD) AML cells harboring MLL-FP or NPM1c. Present studies found that, following MI treatment, CyTOF analysis of PD MLL1-r and NPM1c AML cells showed decline in protein levels of Menin, MEIS1, MEF2C, PBX3, FLT3, CDK6 and BCL2 in phenotypically characterized AML LSCs expressing CLEC12A, CD123, CD244 and CD99. Notably, in vitro co-treatment with SNDX-50469 in combination with venetoclax, OTX015 (pan-BET inhibitor) or abemaciclib (CDK6 inhibitor) induced synergistic (determined by SynergyFinder V2) loss of viability in AML cell lines (MV4-11, MOLM13 and OCI-AML3), as well as in MOLM13 cells with CRISPR-Cas9-mediated knock-in of mutant or allelic loss of TP53 and in PD AML cells with MLL-r or NPM1c, but not in normal CD34+ progenitor cells or AML cells lacking MLL-FP or NPM1c. A CRISPR screen with a validated, domain-specific gRNA library against chromatin regulators revealed BRD4, p300, MOZ and KDM1A as druggable co-dependencies with treatment with MI. Consistent with this, co-treatment with SNDX-50469 and OTX015, or the p300/CBP inhibitor GNE049 was synergistically lethal in vitro in AML cells with MLL1r or NPM1c, either sensitive to MI or tolerant-resistant (induced in vitro) to MI (MITR cells). Co-treatment with SNDX-5613 and venetoclax or OTX015 compared to each drug or vehicle control, administered orally for 3 to 4 weeks to NSG mice engrafted with either MOLM13-GFP/Luciferase xenograft or with PD NPM1c and mtFLT3 AML xenograft, caused significantly greater reduction in AML burden and increased overall survival without weight loss or other toxicities (p & lt; 0.005). These preclinical findings highlight novel MI-based combinations exhibiting superior in vitro and in vivo anti-AML efficacy against AML cells harboring MLL1-FP or NPM1c that are sensitive to MI or the MITR cells. Citation Format: Warren C. Fiskus, Christopher P. Mill, Christine Birdwell, John A. Davis, Qi Jin, Tapan M. Kadia, Courtney D. DiNardo, Koichi Takahashi, Gerard M. McGeehan, Naval Daver, Kapil N. Bhalla. Menin inhibitor-based combinations to improve efficacy and overcome resistance in AML [abstract] . In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 4028.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 10, No. 5 ( 2022-05-03), p. 597-611
    Abstract: Immune checkpoint therapy (ICT) using antibody blockade of programmed cell death protein 1 (PD-1) or cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) can provoke T cell–dependent antitumor activity that generates durable clinical responses in some patients. The epigenetic and transcriptional features that T cells require for efficacious ICT remain to be fully elucidated. Herein, we report that anti–PD-1 and anti–CTLA-4 ICT induce upregulation of the transcription factor BHLHE40 in tumor antigen–specific CD8+ and CD4+ T cells and that T cells require BHLHE40 for effective ICT in mice bearing immune-edited tumors. Single-cell RNA sequencing of intratumoral immune cells in BHLHE40-deficient mice revealed differential ICT-induced immune cell remodeling. The BHLHE40-dependent gene expression changes indicated dysregulated metabolism, NF-κB signaling, and IFNγ response within certain subpopulations of CD4+ and CD8+ T cells. Intratumoral CD4+ and CD8+ T cells from BHLHE40-deficient mice exhibited higher expression of the inhibitory receptor gene Tigit and displayed alterations in expression of genes encoding chemokines/chemokine receptors and granzyme family members. Mice lacking BHLHE40 had reduced ICT-driven IFNγ production by CD4+ and CD8+ T cells and defects in ICT-induced remodeling of macrophages from a CX3CR1+CD206+ subpopulation to an iNOS+ subpopulation that is typically observed during effective ICT. Although both anti–PD-1 and anti–CTLA-4 ICT in BHLHE40-deficient mice led to the same outcome—tumor outgrowth—several BHLHE40-dependent alterations were specific to the ICT that was used. Our results reveal a crucial role for BHLHE40 in effective ICT and suggest that BHLHE40 may be a predictive or prognostic biomarker for ICT efficacy and a potential therapeutic target.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2732517-9
    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...