545 results on '"Leonidas C. Platanias"'
Search Results
2. A novel approach to overcome drug resistance in acute myeloid leukemia
- Author
-
Candice Mazewski and Leonidas C. Platanias
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
3. Discovery of a signaling feedback circuit that defines interferon responses in myeloproliferative neoplasms
- Author
-
Diana Saleiro, Jeremy Q. Wen, Ewa M. Kosciuczuk, Frank Eckerdt, Elspeth M. Beauchamp, Chidera V. Oku, Gavin T. Blyth, Mariafausta Fischietti, Liliana Ilut, Marco Colamonici, William Palivos, Paula A. Atsaves, Dean Tan, Masha Kocherginsky, Rona Singer Weinberg, Eleanor N. Fish, John D. Crispino, Ronald Hoffman, and Leonidas C. Platanias
- Subjects
Science - Abstract
Interferon alpha (IFNalpha) therapy is showing promising results to treat myeloproliferative neoplasms (MPNs). Here, the authors show that IFNalpha response requires ULK1 phosphorylation to induce p38-MAPK signalling but it is counteracted by ROCK1-2 activation suggesting combination therapy of IFNalpha-ROCK1-2 inhibition may improve MPNs treatment.
- Published
- 2022
- Full Text
- View/download PDF
4. Central nervous system immune interactome is a function of cancer lineage, tumor microenvironment, and STAT3 expression
- Author
-
Hinda Najem, Martina Ott, Cynthia Kassab, Arvind Rao, Ganesh Rao, Anantha Marisetty, Adam M. Sonabend, Craig Horbinski, Roel Verhaak, Anand Shankar, Santhoshi N. Krishnan, Frederick S. Varn, Víctor A. Arrieta, Pravesh Gupta, Sherise D. Ferguson, Jason T. Huse, Gregory N. Fuller, James P. Long, Daniel E. Winkowski, Ben A. Freiberg, Charles David James, Leonidas C. Platanias, Maciej S. Lesniak, Jared K. Burks, and Amy B. Heimberger
- Subjects
Immunology ,Oncology ,Medicine - Abstract
BACKGROUND Immune cell profiling of primary and metastatic CNS tumors has been focused on the tumor, not the tumor microenvironment (TME), or has been analyzed via biopsies.METHODS En bloc resections of gliomas (n = 10) and lung metastases (n = 10) were analyzed via tissue segmentation and high-dimension Opal 7-color multiplex imaging. Single-cell RNA analyses were used to infer immune cell functionality.RESULTS Within gliomas, T cells were localized in the infiltrating edge and perivascular space of tumors, while residing mostly in the stroma of metastatic tumors. CD163+ macrophages were evident throughout the TME of metastatic tumors, whereas in gliomas, CD68+, CD11c+CD68+, and CD11c+CD68+CD163+ cell subtypes were commonly observed. In lung metastases, T cells interacted with CD163+ macrophages as dyads and clusters at the brain-tumor interface and within the tumor itself and as clusters within the necrotic core. In contrast, gliomas typically lacked dyad and cluster interactions, except for T cell CD68+ cell dyads within the tumor. Analysis of transcriptomic data in glioblastomas revealed that innate immune cells expressed both proinflammatory and immunosuppressive gene signatures.CONCLUSION Our results show that immunosuppressive macrophages are abundant within the TME and that the immune cell interactome between cancer lineages is distinct. Further, these data provide information for evaluating the role of different immune cell populations in brain tumor growth and therapeutic responses.FUNDING This study was supported by the NIH (NS120547), a Developmental research project award (P50CA221747), ReMission Alliance, institutional funding from Northwestern University and the Lurie Comprehensive Cancer Center, and gifts from the Mosky family and Perry McKay. Performed in the Flow Cytometry & Cellular Imaging Core Facility at MD Anderson Cancer Center, this study received support in part from the NIH (CA016672) and the National Cancer Institute (NCI) Research Specialist award 1 (R50 CA243707). Additional support was provided by CCSG Bioinformatics Shared Resource 5 (P30 CA046592), a gift from Agilent Technologies, a Research Scholar Grant from the American Cancer Society (RSG-16-005-01), a Precision Health Investigator Award from University of Michigan (U-M) Precision Health, the NCI (R37-CA214955), startup institutional research funds from U-M, and a Biomedical Informatics & Data Science Training Grant (T32GM141746).
- Published
- 2022
- Full Text
- View/download PDF
5. Combined PI3Kα-mTOR Targeting of Glioma Stem Cells
- Author
-
Frank D. Eckerdt, Jonathan B. Bell, Christopher Gonzalez, Michael S. Oh, Ricardo E. Perez, Candice Mazewski, Mariafausta Fischietti, Stewart Goldman, Ichiro Nakano, and Leonidas C. Platanias
- Subjects
Medicine ,Science - Abstract
Abstract Glioblastoma (GBM) is the most common and lethal primary intrinsic tumour of the adult brain and evidence indicates disease progression is driven by glioma stem cells (GSCs). Extensive advances in the molecular characterization of GBM allowed classification into proneural, mesenchymal and classical subtypes, and have raised expectations these insights may predict response to targeted therapies. We utilized GBM neurospheres that display GSC characteristics and found activation of the PI3K/AKT pathway in sphere-forming cells. The PI3Kα selective inhibitor alpelisib blocked PI3K/AKT activation and inhibited spheroid growth, suggesting an essential role for the PI3Kα catalytic isoform. p110α expression was highest in the proneural subtype and this was associated with increased phosphorylation of AKT. Further, employing the GBM BioDP, we found co-expression of PIK3CA with the neuronal stem/progenitor marker NES was associated with poor prognosis in PN GBM patients, indicating a unique role for PI3Kα in PN GSCs. Alpelisib inhibited GSC neurosphere growth and these effects were more pronounced in GSCs of the PN subtype. The antineoplastic effects of alpelisib were substantially enhanced when combined with pharmacologic mTOR inhibition. These findings identify the alpha catalytic PI3K isoform as a unique therapeutic target in proneural GBM and suggest that pharmacological mTOR inhibition may sensitize GSCs to selective PI3Kα inhibition.
- Published
- 2020
- Full Text
- View/download PDF
6. Association of a novel circulating tumor DNA next-generating sequencing platform with circulating tumor cells (CTCs) and CTC clusters in metastatic breast cancer
- Author
-
Andrew A. Davis, Qiang Zhang, Lorenzo Gerratana, Ami N. Shah, Youbin Zhan, Wenan Qiang, Brian S. Finkelman, Lisa Flaum, Amir Behdad, William J. Gradishar, Leonidas C. Platanias, and Massimo Cristofanilli
- Subjects
ctDNA ,NGS ,CTCs ,CTC clusters ,MBC ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Purpose Liquid biopsies, including circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs), can be used to understand disease prognosis, tumor heterogeneity, and dynamic response to treatment in metastatic breast cancer (MBC). We explored a novel, 180-gene ctDNA panel and the association of this platform with CTCs and CTC clusters. Methods A total of 40 samples from 22 patients with MBC were included in the study. For the primary analysis, all patients had ctDNA sequencing using the PredicinePLUS™ platform. CTCs and CTC clusters were examined using the CellSearch™ System. Clinical and pathological variables were reported using descriptive analyses. Associations between CTC count and specific genomic alterations were tested using the Mann-Whitney U test. Results Of 43 sequenced patients, 40 (93%) had at least one detectable genomic alteration with a median of 6 (range 1–22). Fifty-seven different genes were altered, and the landscape of genomic alterations was representative of MBC, including the commonly encountered alterations TP53, PTEN, PIK3CA, ATM, BRCA1, CCND1, ESR1, and MYC. In patients with predominantly hormone-receptor-positive MBC, the number of CTCs was significantly associated with alterations in ESR1 (P
- Published
- 2019
- Full Text
- View/download PDF
7. Type I Interferon (IFN)-Regulated Activation of Canonical and Non-Canonical Signaling Pathways
- Author
-
Candice Mazewski, Ricardo E. Perez, Eleanor N. Fish, and Leonidas C. Platanias
- Subjects
interferon ,signaling ,MAP kinase signaling ,signal transducer and activator of transcription ,mammalian target of rapamycin ,mRNA translation ,Immunologic diseases. Allergy ,RC581-607 - Abstract
For several decades there has been accumulating evidence implicating type I interferons (IFNs) as key elements of the immune response. Therapeutic approaches incorporating different recombinant type I IFN proteins have been successfully employed to treat a diverse group of diseases with significant and positive outcomes. The biological activities of type I IFNs are consequences of signaling events occurring in the cytoplasm and nucleus of cells. Biochemical events involving JAK/STAT proteins that control transcriptional activation of IFN-stimulated genes (ISGs) were the first to be identified and are referred to as “canonical” signaling. Subsequent identification of JAK/STAT-independent signaling pathways, critical for ISG transcription and/or mRNA translation, are denoted as “non-canonical” or “non-classical” pathways. In this review, we summarize these signaling cascades and discuss recent developments in the field, specifically as they relate to the biological and clinical implications of engagement of both canonical and non-canonical pathways.
- Published
- 2020
- Full Text
- View/download PDF
8. Landscape of circulating tumour DNA in metastatic breast cancer
- Author
-
Andrew A. Davis, Saya Jacob, Lorenzo Gerratana, Ami N. Shah, Firas Wehbe, Neelima Katam, Qiang Zhang, Lisa Flaum, Kalliopi P. Siziopikou, Leonidas C. Platanias, William J. Gradishar, Amir Behdad, and Massimo Cristofanilli
- Subjects
Circulating tumour DNA ,cell-free DNA ,Next-generation sequencing ,Genomics ,Metastatic breast cancer ,Medicine ,Medicine (General) ,R5-920 - Abstract
Background: We describe the genomic landscape of circulating tumour DNA (ctDNA) across pathological subtypes of metastatic breast cancer. Methods: 255 clinically annotated patients with ctDNA testing by Guardant360 were stratified into HR+, HER2+, and TNBC cohorts. Frequency and heterogeneity of alterations were reported. Paired ctDNA and tissue sequencing were compared for a subset of patients. The association of ctDNA and metastatic sites of disease on imaging was also assessed. Findings: 89% of patients had at least one ctDNA alteration detected. The most common single nucleotide variants (SNVs) for HR+ patients were PIK3CA, ESR1, and TP53. For HER2+, these were TP53, PIK3CA, and ERBB2 with ERBB2 as the most frequent copy number variant (CNV). For TNBC, the most common SNVs were TP53 and PIK3CA, and the most frequent CNVs were MYC, CCNE1, and PIK3CA. TNBC patients had a significantly higher mutant allele frequency (MAF) of the highest variant compared to HR+ or HER2+ patients (P
- Published
- 2020
- Full Text
- View/download PDF
9. Innate Immune Mechanisms and Immunotherapy of Myeloid Malignancies
- Author
-
Sara Small, Yazan Numan, and Leonidas C. Platanias
- Subjects
immunotherapy ,AML ,interferon ,antibodies ,myeloid malignancies ,CAR-T ,Biology (General) ,QH301-705.5 - Abstract
Similar to other cancers, myeloid malignancies are thought to subvert the immune system during their development. This subversion occurs via both malignant cell-autonomous and non-autonomous mechanisms and involves manipulation of the innate and adaptive immune systems. Multiple strategies are being studied to rejuvenate, redirect, or re-enforce the immune system in order to fight off myeloid malignancies. So far, the most successful strategies include interferon treatment and antibody-based therapies, though chimeric antigen receptor (CAR) cells and immune checkpoint inhibitors are also promising therapies. In this review, we discuss the inherent immune mechanisms of defense against myeloid malignancies, currently-approved agents, and agents under investigation. Overall, we evaluate the efficacy and potential of immuno-oncology in the treatment of myeloid malignancies.
- Published
- 2021
- Full Text
- View/download PDF
10. A simple, low-cost staining method for rapid-throughput analysis of tumor spheroids
- Author
-
Frank Eckerdt, Angel Alvarez, Jonathan Bell, Constadina Arvanitis, Asneha Iqbal, Ahmet D. Arslan, Bo Hu, Shi-Yuan Cheng, Stewart Goldman, and Leonidas C. Platanias
- Subjects
neurospheres ,tumor spheroids ,cancer stem cell ,glioblastoma ,acridine orange ,microscopy ,Biology (General) ,QH301-705.5 - Abstract
Tumor spheroids are becoming an important tool for the investigation of cancer stem cell (CSC) function in tumors; thus, low-cost and high-throughput methods for drug screening of tumor spheroids are needed. Using neurospheres as non-adherent three-dimensional (3-D) cultures, we developed a simple, low-cost acridine orange (AO)–based method that allows for rapid analysis of live neurospheres by fluorescence microscopy in a 96-well format. This assay measures the cross-section area of a spheroid, which corresponds to cell viability. Our novel method allows rapid screening of a panel of anti-proliferative drugs to assess inhibitory effects on the growth of cancer stem cells in 3-D cultures.
- Published
- 2016
- Full Text
- View/download PDF
11. Evolving Therapeutic Strategies for the Classic Philadelphia-Negative Myeloproliferative Neoplasms
- Author
-
Jason B. Kaplan, Brady L. Stein, Brandon McMahon, Francis J. Giles, and Leonidas C. Platanias
- Subjects
Myeloproliferative neoplasm ,Myelofibrosis ,Therapy ,Targeted ,Novel ,Combination ,Medicine ,Medicine (General) ,R5-920 - Abstract
Despite the emergence of JAK inhibitors, there is a need for disease-modifying treatments for Philadelphia-negative myeloproliferative neoplasms (MPNs). JAK inhibitors ameliorate symptoms and address splenomegaly, but because of the heterogeneous contributors to the disease process, JAK inhibitor monotherapy incompletely addresses the burden of disease. The ever-growing understanding of MPN pathogenesis has provided the rationale for testing novel and targeted therapeutic agents, as monotherapies or in combination, in preclinical and clinical settings. A number of intriguing options have emerged, and it is hoped that further progress will lead to significant changes in the natural history of MPNs.
- Published
- 2016
- Full Text
- View/download PDF
12. Central Role of ULK1 in Type I Interferon Signaling
- Author
-
Diana Saleiro, Swarna Mehrotra, Barbara Kroczynska, Elspeth M. Beauchamp, Pawel Lisowski, Beata Majchrzak-Kita, Tushar D. Bhagat, Brady L. Stein, Brandon McMahon, Jessica K. Altman, Ewa M. Kosciuczuk, Darren P. Baker, Chunfa Jie, Nadereh Jafari, Craig B. Thompson, Ross L. Levine, Eleanor N. Fish, Amit K. Verma, and Leonidas C. Platanias
- Subjects
Biology (General) ,QH301-705.5 - Abstract
We provide evidence that the Unc-51-like kinase 1 (ULK1) is activated during engagement of the type I interferon (IFN) receptor (IFNR). Our studies demonstrate that the function of ULK1 is required for gene transcription mediated via IFN-stimulated response elements (ISRE) and IFNγ activation site (GAS) elements and controls expression of key IFN-stimulated genes (ISGs). We identify ULK1 as an upstream regulator of p38α mitogen-activated protein kinase (MAPK) and establish that the regulatory effects of ULK1 on ISG expression are mediated possibly by engagement of the p38 MAPK pathway. Importantly, we demonstrate that ULK1 is essential for antiproliferative responses and type I IFN-induced antineoplastic effects against malignant erythroid precursors from patients with myeloproliferative neoplasms. Together, these data reveal a role for ULK1 as a key mediator of type I IFNR-generated signals that control gene transcription and induction of antineoplastic responses.
- Published
- 2015
- Full Text
- View/download PDF
13. Circulating microRNAs: promising biomarkers in aplastic anemia
- Author
-
Jonathan B. Bell, Sameem Abedin, and Leonidas C. Platanias
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2017
- Full Text
- View/download PDF
14. Deregulation of Interferon Signaling in Malignant Cells
- Author
-
Leonidas C. Platanias, Surinder Kaur, and Efstratios Katsoulidis
- Subjects
interferon ,signaling pathways ,cancer ,Medicine ,Pharmacy and materia medica ,RS1-441 - Abstract
Interferons (IFNs) are a family of cytokines with potent antiproliferative, antiviral, and immunomodulatory properties. Much has been learned about IFNs and IFN-activated signaling cascades over the last 50 years. Due to their potent antitumor effects in vitro and in vivo, recombinant IFNs have been used extensively over the years, alone or in combination with other drugs, for the treatment of various malignancies. This review summarizes the current knowledge on IFN signaling components and pathways that are deregulated in human malignancies. The relevance of deregulation of IFN signaling pathways in defective innate immune surveillance and tumorigenesis are discussed.
- Published
- 2010
- Full Text
- View/download PDF
15. Targeting CHAF1B Enhances IFN Activity against Myeloproliferative Neoplasm Cells
- Author
-
Diana Saleiro, Ewa M. Kosciuczuk, Mariafausta Fischietti, Ricardo E. Perez, G. Sohae Yang, Frank Eckerdt, Elspeth M. Beauchamp, Ye Hou, Qixuan Wang, Rona Singer Weinberg, Eleanor N. Fish, Feng Yue, Ronald Hoffman, and Leonidas C. Platanias
- Abstract
Interferons (IFNs) are cytokines with potent antineoplastic and antiviral properties. IFNα has significant clinical activity in the treatment of myeloproliferative neoplasms (MPN), but the precise mechanisms by which it acts are not well understood. Here, we demonstrate that chromatin assembly factor 1 subunit B (CHAF1B), an Unc-51-like kinase 1 (ULK1)-interactive protein in the nuclear compartment of malignant cells, is overexpressed in patients with MPN. Remarkably, targeted silencing of CHAF1B enhances transcription of IFNα-stimulated genes and promotes IFNα-dependent antineoplastic responses in primary MPN progenitor cells. Taken together, our findings indicate that CHAF1B is a promising newly identified therapeutic target in MPN and that CHAF1B inhibition in combination with IFNα therapy might offer a novel strategy for treating patients with MPN. Significance: Our findings raise the potential for clinical development of drugs targeting CHAF1B to enhance IFN antitumor responses in the treatment of patients with MPN and should have important clinical translational implications for the treatment of MPN and possibly in other malignancies.
- Published
- 2023
- Full Text
- View/download PDF
16. MNK Proteins as Therapeutic Targets in Leukemia
- Author
-
Candice Mazewski and Leonidas C Platanias
- Subjects
Oncology ,Pharmacology (medical) - Published
- 2023
- Full Text
- View/download PDF
17. PRL2 phosphatase enhances oncogenic FLT3 signaling via dephosphorylation of the E3 ubiquitin ligase CBL at tyrosine 371
- Author
-
Hongxia Chen, Yunpeng Bai, Michihiro Kobayashi, Shiyu Xiao, Wenjie Cai, Sergio Barajas, Sisi Chen, Jinmin Miao, Frederick Nguele Meke, Sasidhar Vemula, James P. Ropa, James M. Croop, H. Scott Boswell, Jun Wan, Yuzhi Jia, Huiping Liu, Loretta S. Li, Jessica K. Altman, Elizabeth A. Eklund, Peng Ji, Wei Tong, Hamid Band, Danny T. Huang, Leonidas C. Platanias, Zhong-Yin Zhang, and Yan Liu
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Acute myeloid leukemia (AML) is an aggressive blood cancer with poor prognosis. FMS-like tyrosine kinase receptor-3 (FLT3) is one of the major oncogenic receptor tyrosine kinases aberrantly activated in AML. Although protein tyrosine phosphatase PRL2 is highly expressed in some subtypes of AML compared with normal human hematopoietic stem and progenitor cells, the mechanisms by which PRL2 promotes leukemogenesis are largely unknown. We discovered that genetic and pharmacological inhibition of PRL2 significantly reduce the burden of FLT3-internal tandem duplications–driven leukemia and extend the survival of leukemic mice. Furthermore, we found that PRL2 enhances oncogenic FLT3 signaling in leukemia cells, promoting their proliferation and survival. Mechanistically, PRL2 dephosphorylates the E3 ubiquitin ligase CBL at tyrosine 371 and attenuates CBL-mediated ubiquitination and degradation of FLT3, leading to enhanced FLT3 signaling in leukemia cells. Thus, our study reveals that PRL2 enhances oncogenic FLT3 signaling in leukemia cells through dephosphorylation of CBL and will likely establish PRL2 as a novel druggable target for AML.
- Published
- 2023
- Full Text
- View/download PDF
18. Targeting ULK1 Decreases IFNγ-Mediated Resistance to Immune Checkpoint Inhibitors
- Author
-
Sarah E. Fenton, Markella Zannikou, Liliana Ilut, Mariafausta Fischietti, Chunni Ji, Chidera V. Oku, Curt M. Horvath, I. Caroline Le Poole, Marcus Bosenberg, Elizabeth T. Bartom, Masha Kocherginsky, Leonidas C. Platanias, and Diana Saleiro
- Subjects
Cancer Research ,Oncology ,Molecular Biology - Abstract
Immune checkpoint inhibitors (ICI) have transformed the treatment of melanoma. However, the majority of patients have primary or acquired resistance to ICIs, limiting durable responses and patient survival. IFNγ signaling and the expression of IFNγ-stimulated genes correlate with either response or resistance to ICIs, in a context-dependent manner. While IFNγ-inducible immunostimulatory genes are required for response to ICIs, chronic IFNγ signaling induces the expression of immunosuppressive genes, promoting resistance to these therapies. Here, we show that high levels of Unc-51 like kinase 1 (ULK1) correlate with poor survival in patients with melanoma and overexpression of ULK1 in melanoma cells enhances IFNγ-induced expression of immunosuppressive genes, with minimal effects on the expression of immunostimulatory genes. In contrast, genetic or pharmacologic inhibition of ULK1 reduces expression of IFNγ-induced immunosuppressive genes. ULK1 binds IRF1 in the nuclear compartment of melanoma cells, controlling its binding to the programmed death-ligand 1 promoter region. In addition, pharmacologic inhibition of ULK1 in combination with anti-programmed cell death protein 1 therapy further reduces melanoma tumor growth in vivo. Our data suggest that targeting ULK1 represses IFNγ-dependent immunosuppression. These findings support the combination of ULK1 drug-targeted inhibition with ICIs for the treatment of patients with melanoma to improve response rates and patient outcomes. Implications: This study identifies ULK1, activated downstream of IFNγ signaling, as a druggable target to overcome resistance mechanisms to ICI therapy in metastatic melanoma.
- Published
- 2022
- Full Text
- View/download PDF
19. Subtype-specific 3D genome alteration in acute myeloid leukaemia
- Author
-
Jie Xu, Fan Song, Huijue Lyu, Mikoto Kobayashi, Baozhen Zhang, Ziyu Zhao, Ye Hou, Xiaotao Wang, Yu Luan, Bei Jia, Lena Stasiak, Josiah Hiu-yuen Wong, Qixuan Wang, Qi Jin, Qiushi Jin, Yihao Fu, Hongbo Yang, Ross C. Hardison, Sinisa Dovat, Leonidas C. Platanias, Yarui Diao, Yue Yang, Tomoko Yamada, Aaron D. Viny, Ross L. Levine, David Claxton, James. R. Broach, Hong Zheng, and Feng Yue
- Subjects
Multidisciplinary ,Genome, Human ,Gene Expression Regulation, Leukemic ,Reproducibility of Results ,DNA Methylation ,Article ,Chromatin ,Leukemia, Myeloid, Acute ,Enhancer Elements, Genetic ,Humans ,Gene Silencing ,DNA (Cytosine-5-)-Methyltransferases ,CRISPR-Cas Systems ,Promoter Regions, Genetic ,Sequence Analysis - Abstract
Acute myeloid leukaemia (AML) represents a set of heterogeneous myeloid malignancies, and hallmarks include mutations in epigenetic modifiers, transcription factors and kinasessup1-5/sup. The extent to which mutations in AML drive alterations in chromatin 3D structure and contribute to myeloid transformation is unclear. Here we use Hi-C and whole-genome sequencing to analyse 25 samples from patients with AML and 7 samples from healthy donors. Recurrent and subtype-specific alterations in A/B compartments, topologically associating domains and chromatin loops were identified. RNA sequencing, ATAC with sequencing and CUTamp;Tag for CTCF, H3K27ac and H3K27me3 in the same AML samples also revealed extensive and recurrent AML-specific promoter-enhancer and promoter-silencer loops. We validated the role of repressive loops on their target genes by CRISPR deletion and interference. Structural variation-induced enhancer-hijacking and silencer-hijacking events were further identified in AML samples. Hijacked enhancers play a part in AML cell growth, as demonstrated by CRISPR screening, whereas hijacked silencers have a downregulating role, as evidenced by CRISPR-interference-mediated de-repression. Finally, whole-genome bisulfite sequencing of 20 AML and normal samples revealed the delicate relationship between DNA methylation, CTCF binding and 3D genome structure. Treatment of AML cells with a DNA hypomethylating agent and triple knockdown of DNMT1, DNMT3A and DNMT3B enabled the manipulation of DNA methylation to revert 3D genome organization and gene expression. Overall, this study provides a resource for leukaemia studies and highlights the role of repressive loops and hijacked cis elements in human diseases.
- Published
- 2022
- Full Text
- View/download PDF
20. Advances in the pharmacological management of acute myeloid leukemia in adults
- Author
-
Yazan Numan, Yasmin Abaza, Jessica K Altman, and Leonidas C Platanias
- Subjects
Pharmacology ,Adult ,Leukemia, Myeloid, Acute ,Proto-Oncogene Proteins c-bcl-2 ,Mutation ,Humans ,Pharmacology (medical) ,CD47 Antigen ,General Medicine ,Protein-Tyrosine Kinases ,Protein Kinase Inhibitors - Abstract
With advances in molecular medicine and precision approaches, there has been significant improvement in the treatment of acute myeloid leukemia (AML) in recent years. This reflects better understanding of molecular and metabolic pathways in leukemia cells, including BCL2 upregulation that prevents apoptosis, FLT3 tyrosine kinase activating mutations that allow uncontrolled proliferation, and IDH mutations that result in differentiation block.We performed a compressive review of important pre-clinical studies in AML that involve major molecular and metabolic pathways in AML, and we discussed standard therapeutic modalities and ongoing clinical trials for patients with AML, as well as an overall update of recent efforts in this area.Targeting these pathways has resulted in improvement in the overall survival of some groups of AML patients. Secondary AML and TP53 mutated AML remain challenging subtypes of AML with limited treatment options and represent areas of unmet research need. Ongoing work with menin inhibitors in MLL rearranged leukemia, which comprise a large portion of secondary AML cases, the development of CAR T cell products and targeting the CD47 receptor on macrophages in myeloid neoplasms including in TP53 mutated AML have provided hope for these challenging subtypes of AML.
- Published
- 2023
21. FIGURE 4 from Targeting CHAF1B Enhances IFN Activity against Myeloproliferative Neoplasm Cells
- Author
-
Leonidas C. Platanias, Ronald Hoffman, Feng Yue, Eleanor N. Fish, Rona Singer Weinberg, Qixuan Wang, Ye Hou, Elspeth M. Beauchamp, Frank Eckerdt, G. Sohae Yang, Ricardo E. Perez, Mariafausta Fischietti, Ewa M. Kosciuczuk, and Diana Saleiro
- Abstract
Inhibition of CHAF1B increases IFNα-induced anticlonogenic effects in MPN. HEL cells (A) and SET-2 cells (B) were transfected with either control (Ctrl) siRNA or CHAF1B siRNA, and leukemic CFU-L colony formation was assessed in clonogenic assays in the presence or absence of IFNα, as indicated. Data are expressed as percentage of colony formation over control siRNA-transfected untreated cells (control), and the scatter dot plots represent means ± SEM of four independent experiments for each cell line. C, Clonogenic capability of primary malignant erythroid progenitors isolated from PV patients transfected with either control (Ctrl) siRNA or CHAF1B siRNA and either left untreated or treated with IFNα, as indicated. Data are expressed as percentage of colony formation over control siRNA-transfected untreated cells (control) and shown are means ± SEM of six independent experiments using cells from six different patients with PV. Percentages for the same patient are represented by the same symbol and different patients have different symbols. A–C, Statistical analyses were performed using one-way ANOVA followed by Tukey multiple comparisons test. *, P < 0.05; **, P < 0.01; ****, P < 0.0001.
- Published
- 2023
- Full Text
- View/download PDF
22. FIGURE 2 from Targeting CHAF1B Enhances IFN Activity against Myeloproliferative Neoplasm Cells
- Author
-
Leonidas C. Platanias, Ronald Hoffman, Feng Yue, Eleanor N. Fish, Rona Singer Weinberg, Qixuan Wang, Ye Hou, Elspeth M. Beauchamp, Frank Eckerdt, G. Sohae Yang, Ricardo E. Perez, Mariafausta Fischietti, Ewa M. Kosciuczuk, and Diana Saleiro
- Abstract
CHAF1B interacts with ULK1 in the nuclear compartment of JAK2V617F-positive cells and is overexpressed in patients with MPN. A and B, Left, ULK1–protein complexes were co-IP using an anti-ULK1 antibody from cytosolic and nuclear protein fractions isolated from untreated or IFNα-treated (10 or 240 minutes) HEL (A) or SET-2 (B) cells and then resolved by SDS-PAGE. As control, cytosolic and nuclear lysates isolated from cells treated with IFNα for 240 minutes were incubated with normal rabbit IgG (RIgG) antibody. Interaction between ULK1 and CHAF1B was assessed by immunoblotting with anti-ULK1 and anti-CHAF1B antibodies. Note: ǂ, unspecific band. A and B, Right, Equal amounts of cytosolic and nuclear protein lysates isolated from untreated and IFNα-treated HEL (A) and SET-2 (B) cells used for co-IPs were resolved by SDS-PAGE, transferred to PVDF membranes and then immunoblotted with antibodies against ULK1, CHAF1B, α-tubulin (cytosolic marker), and lamin A/C (nuclear marker), as indicated. A and B, Blots are representative of three independent experiments. C, Scatter dot plot of log2CHAF1B mRNA expression in neutrophils from healthy individuals (normal, n = 11) and patients with ET (n = 47), PMF (n = 18), and PV (n = 28). Data were extracted from NCBI GEO: GSE54646 study (21) and analyzed using GraphPad Prim 8. Shown are means ± SEM. Statistical analysis was performed using one-way ANOVA followed by Dunnett multiple comparisons test to assess P values between patients with MPN and healthy individuals. *, P < 0.05; ***, P < 0.001; ****, P < 0.0001. Scatter dot plots of log2CHAF1B mRNA expression in neutrophils from patients with ET (green triangles), PMF (purple diamonds), and PV (red squares) carrying wild-type (WT) or mutant (MUT) JAK2 (D), CALR (E), and TET2 (F) genes (JAK2 WT: ET n = 20, PMF n = 10, PV n = 5; JAK2 MUT: ET n = 26, PMF n = 8, PV n = 23; CALR WT: ET n = 27, PMF n = 11, PV n = 26; CALR MUT: ET n = 15, PMF n = 5, PV n = 1; TET2 WT: ET n = 45, PMF n = 17, PV n = 24; TET2 MUT: ET n = 2, PMF n = 1, PV n = 4). Data were extracted from NCBI GEO: GSE54646 study (21) and analyzed using GraphPad Prim 8. Patients for which no information was available for the mutational status for the JAK2, CALR, or TET2 genes were excluded from the analysis. Shown are means ± SEM. Statistical analyses were performed using two-sample two-tailed t test: *, P < 0.05.
- Published
- 2023
- Full Text
- View/download PDF
23. FIGURE 3 from Targeting CHAF1B Enhances IFN Activity against Myeloproliferative Neoplasm Cells
- Author
-
Leonidas C. Platanias, Ronald Hoffman, Feng Yue, Eleanor N. Fish, Rona Singer Weinberg, Qixuan Wang, Ye Hou, Elspeth M. Beauchamp, Frank Eckerdt, G. Sohae Yang, Ricardo E. Perez, Mariafausta Fischietti, Ewa M. Kosciuczuk, and Diana Saleiro
- Abstract
Gene-targeted inhibition of CHAF1B increases IFNα-inducible mRNA expression of ISGs in JAK2V617F-positive leukemic cells. qRT-PCR analyses of the indicated genes in control (Ctrl) siRNA or CHAF1B siRNA-transfected JAK2V617F-positive HEL (A) and SET-2 (B) cells, either left untreated or treated with IFNα for 6 hours. GAPDH mRNA expression was used for normalization. Data are expressed as fold change over control siRNA-transfected untreated cells (control) and represent means ± SEM of three independent experiments for each cell line. Statistical analyses were performed using one-way ANOVA followed by Tukey multiple comparisons test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. C, ChIP assay was performed in untreated and IFNα-treated (for 6 hours at 5,000 IU/mL) HEL cells at the ISG15 promoter and the IFIT1 promoter for CHAF1B binding using an anti-CHAF1B antibody. IgG antibody was used for each promoter region as negative control. Scatter dot plots show data as percent enrichment relative to input ± SEM for three independent experiments. Statistical analyses were performed using one-way ANOVA followed by Tukey multiple comparisons test. ns, not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001. D, Immunoblot analysis of ULK1 expression in control siRNA (Ctrlsi) and ULK1 siRNA (ULK1si) transfected HEL cells either left untreated or treated with IFNα (for 6 hours at 5,000 IU/mL), as indicated. GAPDH levels were used as loading control. Blots are representative of three independent experiments used to perform ChIP assays shown in E. E, ChIP assay was performed in untreated (UT) and IFNα-treated control (Ctrl) siRNA- and ULK1 siRNA-transfected HEL cells at the ISG15 promoter and the IFIT1 promoter for CHAF1B binding using an anti-CHAF1B antibody. F, HEL cells were pretreated for 1 hour with either DMSO (Ctrl and IFNα groups) or SBI-0206965 (SBI; 10 μmol/L; SBI and SBI+IFNα groups) followed by 6 hours of treatment with either DMSO (Ctrl), SBI (10 μmol/L), IFNα (5,000 IU/mL) or SBI+IFNα, as indicated. ChIP assay was performed in HEL cells at the ISG15 promoter and the IFIT1 promoter for CHAF1B binding using an anti-CHAF1B antibody. E and F, IgG antibody was used for each promoter region as negative control. Scatter dot plots show data as percent enrichment relative to input ± SEM for three independent experiments. Statistical analyses were performed using two-way ANOVA followed by Tukey multiple comparisons test: *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. Relevant statistical differences are shown for the binding of CHAF1B to ISG15 and IFIT1 promoter regions between experimental conditions.
- Published
- 2023
- Full Text
- View/download PDF
24. Figure S1 from Targeting CHAF1B Enhances IFN Activity against Myeloproliferative Neoplasm Cells
- Author
-
Leonidas C. Platanias, Ronald Hoffman, Feng Yue, Eleanor N. Fish, Rona Singer Weinberg, Qixuan Wang, Ye Hou, Elspeth M. Beauchamp, Frank Eckerdt, G. Sohae Yang, Ricardo E. Perez, Mariafausta Fischietti, Ewa M. Kosciuczuk, and Diana Saleiro
- Abstract
Biological processes in which putative ULK1-protein complexes are involved in the cytosol.
- Published
- 2023
- Full Text
- View/download PDF
25. Data from Targeting CHAF1B Enhances IFN Activity against Myeloproliferative Neoplasm Cells
- Author
-
Leonidas C. Platanias, Ronald Hoffman, Feng Yue, Eleanor N. Fish, Rona Singer Weinberg, Qixuan Wang, Ye Hou, Elspeth M. Beauchamp, Frank Eckerdt, G. Sohae Yang, Ricardo E. Perez, Mariafausta Fischietti, Ewa M. Kosciuczuk, and Diana Saleiro
- Abstract
Interferons (IFNs) are cytokines with potent antineoplastic and antiviral properties. IFNα has significant clinical activity in the treatment of myeloproliferative neoplasms (MPN), but the precise mechanisms by which it acts are not well understood. Here, we demonstrate that chromatin assembly factor 1 subunit B (CHAF1B), an Unc-51-like kinase 1 (ULK1)-interactive protein in the nuclear compartment of malignant cells, is overexpressed in patients with MPN. Remarkably, targeted silencing of CHAF1B enhances transcription of IFNα-stimulated genes and promotes IFNα-dependent antineoplastic responses in primary MPN progenitor cells. Taken together, our findings indicate that CHAF1B is a promising newly identified therapeutic target in MPN and that CHAF1B inhibition in combination with IFNα therapy might offer a novel strategy for treating patients with MPN.Significance:Our findings raise the potential for clinical development of drugs targeting CHAF1B to enhance IFN antitumor responses in the treatment of patients with MPN and should have important clinical translational implications for the treatment of MPN and possibly in other malignancies.
- Published
- 2023
- Full Text
- View/download PDF
26. Computational ranking-assisted identification of Plexin-B2 in homotypic and heterotypic clustering of circulating tumor cells in breast cancer metastasis
- Author
-
Emma Schuster, Nurmaa Dashzeveg, Yuzhi Jia, Kibria Golam, Tong Zhang, Andrew Hoffman, Youbin Zhang, Chunlei Zheng, Erika Ramos, Rokana Taftaf, Lamiaa El- Shennawy, David Scholten, Reta B. Kitata, Valery Adorno-Cruz, Carolina Reduzzi, Sabina Spahija, Rong Xu, Kalliopi P. Siziopikou, Leonidas C. Platanias, Ami Shah, William J. Gradishar, Massimo Cristofanilli, Chia-Feng Tsai, Tujin Shi, and Huiping Liu
- Subjects
Article - Abstract
Metastasis is the cause of over 90% of all deaths associated with breast cancer, yet the strategies to predict cancer spreading based on primary tumor profiles and therefore prevent metastasis are egregiously limited. As rare precursor cells to metastasis, circulating tumor cells (CTCs) in multicellular clusters in the blood are 20-50 times more likely to produce viable metastasis than single CTCs. However, the molecular mechanisms underlying various CTC clusters, such as homotypic tumor cell clusters and heterotypic tumor-immune cell clusters, are yet to be fully elucidated. Combining machine learning-assisted computational ranking with experimental demonstration to assess cell adhesion candidates, we identified a transmembrane protein Plexin- B2 (PB2) as a new therapeutic target that drives the formation of both homotypic and heterotypic CTC clusters. High PB2 expression in human primary tumors predicts an unfavorable distant metastasis-free survival and is enriched in CTC clusters compared to single CTCs in advanced breast cancers. Loss of PB2 reduces formation of homotypic tumor cell clusters as well as heterotypic tumor-myeloid cell clusters in triple-negative breast cancer. Interactions between PB2 and its ligand Sema4C on tumor cells promote homotypic cluster formation, and PB2 binding with Sema4A on myeloid cells (monocytes) drives heterotypic CTC cluster formation, suggesting that metastasizing tumor cells hijack the PB2/Sema family axis to promote lung metastasis in breast cancer. Additionally, using a global proteomic analysis, we identified novel downstream effectors of the PB2 pathway associated with cancer stemness, cell cycling, and tumor cell clustering in breast cancer. Thus, PB2 is a novel therapeutic target for preventing new metastasis.
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Table S4 from SLFN11 Negatively Regulates Noncanonical NFκB Signaling to Promote Glioblastoma Progression
- Author
-
Leonidas C. Platanias, C. David James, Feng Yue, Aneta H. Baran, Amy B. Heimberger, Elspeth M. Beauchamp, Lukas D. Streich, Christopher Gonzalez, Sang Ho, Candice Mazewski, Jamie N. Guillen Magaña, Ricardo E. Perez, Frank Eckerdt, and Mariafausta Fischietti
- Abstract
Supplementary Table S4 lists proteins that were found to bind SLFN11 after irradiation.
- Published
- 2023
- Full Text
- View/download PDF
28. Supplementary Figures from Myeloid-Derived Suppressive Cells Promote B cell–Mediated Immunosuppression via Transfer of PD-L1 in Glioblastoma
- Author
-
Maciej S. Lesniak, Irina V. Balyasnikova, Yu Han, Aurora Lopez-Rosas, Leonidas C. Platanias, Craig Horbinski, Christina L. Appin, Seong Jae Kang, Mariafausta Fischietti, Ting Xiao, David Hou, Katarzyna C. Pituch, Peng Zhang, Jason Miska, Aida Rashidi, and Catalina Lee-Chang
- Abstract
Supplementary figures
- Published
- 2023
- Full Text
- View/download PDF
29. Data from SLFN11 Negatively Regulates Noncanonical NFκB Signaling to Promote Glioblastoma Progression
- Author
-
Leonidas C. Platanias, C. David James, Feng Yue, Aneta H. Baran, Amy B. Heimberger, Elspeth M. Beauchamp, Lukas D. Streich, Christopher Gonzalez, Sang Ho, Candice Mazewski, Jamie N. Guillen Magaña, Ricardo E. Perez, Frank Eckerdt, and Mariafausta Fischietti
- Abstract
Glioblastoma (GBM) is an aggressive and incurable brain tumor in nearly all instances, whose disease progression is driven in part by the glioma stem cell (GSC) subpopulation. Here, we explored the effects of Schlafen family member 11 (SLFN11) in the molecular, cellular, and tumor biology of GBM. CRISPR/Cas9-mediated knockout of SLFN11 inhibited GBM cell proliferation and neurosphere growth and was associated with reduced expression of progenitor/stem cell marker genes, such as NES, SOX2, and CD44. Loss of SLFN11 stimulated expression of NFκB target genes, consistent with a negative regulatory role for SLFN11 on the NFκB pathway. Furthermore, our studies identify p21 as a direct transcriptional target of NFκB2 in GBM whose expression was stimulated by loss of SLFN11. Genetic disruption of SLFN11 blocked GBM growth and significantly extended survival in an orthotopic patient-derived xenograft model. Together, our results identify SLFN11 as a novel component of signaling pathways that contribute to GBM and GSC with implications for future diagnostic and therapeutic strategies.Significance:We identify a negative regulatory role for SLFN11 in noncanonical NFκB signaling that results in suppression of the cell-cycle inhibitor p21. We provide evidence that SLFN11 contributes to regulation of stem cell markers in GBM, promoting the malignant phenotype. In addition, SLFN11 targeting triggers p21 expression and antitumor responses. Our studies define a highly novel function for SLFN11 and identify it as a potential therapeutic target for GBM.
- Published
- 2023
- Full Text
- View/download PDF
30. Supplementary Figures S1-S3, Table S1 from SLFN11 Negatively Regulates Noncanonical NFκB Signaling to Promote Glioblastoma Progression
- Author
-
Leonidas C. Platanias, C. David James, Feng Yue, Aneta H. Baran, Amy B. Heimberger, Elspeth M. Beauchamp, Lukas D. Streich, Christopher Gonzalez, Sang Ho, Candice Mazewski, Jamie N. Guillen Magaña, Ricardo E. Perez, Frank Eckerdt, and Mariafausta Fischietti
- Abstract
Fig. S1: Expression of SLFN family members after SLFN11 knockout. Fig. S2: Reduced 3-D invasion after SLFN11 knockout. Fig. S3: Expression of stem/progenitor markers after SLFN11 add-back. Table S1: Key resources table.
- Published
- 2023
- Full Text
- View/download PDF
31. Data from Myeloid-Derived Suppressive Cells Promote B cell–Mediated Immunosuppression via Transfer of PD-L1 in Glioblastoma
- Author
-
Maciej S. Lesniak, Irina V. Balyasnikova, Yu Han, Aurora Lopez-Rosas, Leonidas C. Platanias, Craig Horbinski, Christina L. Appin, Seong Jae Kang, Mariafausta Fischietti, Ting Xiao, David Hou, Katarzyna C. Pituch, Peng Zhang, Jason Miska, Aida Rashidi, and Catalina Lee-Chang
- Abstract
The potent immunosuppression induced by glioblastoma (GBM) is one of the primary obstacles to finding effective immunotherapies. One hallmark of the GBM-associated immunosuppressive landscape is the massive infiltration of myeloid-derived suppressor cells (MDSC) and, to a lesser extent, regulatory T cells (Treg) within the tumor microenvironment. Here, we showed that regulatory B cells (Breg) are a prominent feature of the GBM microenvironment in both preclinical models and clinical samples. Forty percent of GBM patients (n = 60) scored positive for B-cell tumor infiltration. Human and mouse GBM-associated Bregs were characterized by immunosuppressive activity toward activated CD8+ T cells, the overexpression of inhibitory molecules PD-L1 and CD155, and production of immunosuppressive cytokines TGFβ and IL10. Local delivery of B cell–depleting anti-CD20 immunotherapy improved overall survival of animals (IgG vs. anti-CD20 mean survival: 18.5 vs. 33 days, P = 0.0001), suggesting a potential role of Bregs in GBM progression. We unveiled that GBM-associated MDSCs promoted regulatory B-cell function by delivering microvesicles transporting membrane-bound PD-L1, able to be up-taken by tumoral B cells. The transfer of functional PD-L1 via microvesicles conferred Bregs the potential to suppress CD8+ T-cell activation and acquisition of an effector phenotype. This work uncovered the role of B cells in GBM physiopathology and provides a mechanism by which the GBM microenvironment controls B cell–mediated immunosuppression.See related Spotlight on p. 1902
- Published
- 2023
- Full Text
- View/download PDF
32. Supplementary Tables and Figure Legends from Myeloid-Derived Suppressive Cells Promote B cell–Mediated Immunosuppression via Transfer of PD-L1 in Glioblastoma
- Author
-
Maciej S. Lesniak, Irina V. Balyasnikova, Yu Han, Aurora Lopez-Rosas, Leonidas C. Platanias, Craig Horbinski, Christina L. Appin, Seong Jae Kang, Mariafausta Fischietti, Ting Xiao, David Hou, Katarzyna C. Pituch, Peng Zhang, Jason Miska, Aida Rashidi, and Catalina Lee-Chang
- Abstract
Supplementary Tables and Supplementary Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
33. Supplementary Figures and Supplementary Materials and Methods with References from Targeting ULK1 Decreases IFNγ-Mediated Resistance to Immune Checkpoint Inhibitors
- Author
-
Diana Saleiro, Leonidas C. Platanias, Masha Kocherginsky, Elizabeth T. Bartom, Marcus Bosenberg, I. Caroline Le Poole, Curt M. Horvath, Chidera V. Oku, Chunni Ji, Mariafausta Fischietti, Liliana Ilut, Markella Zannikou, and Sarah E. Fenton
- Abstract
S1. Overexpression of ULK1 in melanoma cells has minor effects on transcription of IFNγ-induced immunostimulatory genes.S2. Effects of gene targeted inhibition of ULK1 in transcription of IFNγ-induced immunostimulatory genes in melanoma cells.S3. Effects of drug-targeted inhibition of ULK1 in transcription of IFNγ-induced immunostimulatory genes in melanoma cells.S4. Effects of siRNA-mediated knockdown of ULK1 on IFNγ-inducedSTAT1 phosphorylation.S5. Identification of putative cytoplasmic and nuclear ULK1 binding partners in melanoma cells upon IFNγ stimulation by mass spectrometry analysis.S6. Pharmacological inhibition of ULK1 enhances the anti-tumor effects of anti-PD-1 therapy in YUMMER1.7 mouse melanoma in vivo model.S7. Tumor infiltrating CD8+ T cells and TAMCs in vehicle-isotype-treated mice.S8. Tumor infiltrating CD8+ T cells and TAMCs in ULK inhibitor-treated mice.S9. Tumor infiltrating CD8+ T cells and TAMCs in anti-PD-1-treated mice.S10. Tumor infiltrating CD8+ T cells and TAMCs in ULKi plus anti-PD-1-treated mice.S11. Tumor infiltrating CD4+ T cells and Tregs in vehicle-isotype-treated mice.S12. Tumor infiltrating CD4+ T cells and Tregs in ULK inhibitor-treated mice.S13. Tumor infiltrating CD4+ T cells and Tregs in anti-PD-1treated mice.S14. Tumor infiltrating CD4+ T cells and Tregs in ULKi plus anti-PD-1-treated mice.S15. Tumor infiltrating CD8+ T cells and TAMCs in control, ULKi and/or anti-PD-1-treated mice.S16. Tumor infiltrating CD4+ T cells and Tregs in control, ULKi and/or anti-PD-1-treated mice.S17. Gating strategy for flow cytometric immunophenotyping of mouse melanoma tumors.
- Published
- 2023
- Full Text
- View/download PDF
34. Supplementary Figure 1 from MNK Inhibition Disrupts Mesenchymal Glioma Stem Cells and Prolongs Survival in a Mouse Model of Glioblastoma
- Author
-
Leonidas C. Platanias, C. David James, Ramana V. Davuluri, Craig Horbinski, Ichiro Nakano, Shi-Yuan Cheng, Stewart Goldman, Angel A. Alvarez, Jessica Clymer, Ahmet Dirim Arslan, Yingtao Bi, Hridi Hussain, Lisa P. Magnusson, Kristen Alley, Frank D. Eckerdt, and Jonathan B. Bell
- Abstract
Supplementary materials and methods for ALDEFLUOR assay and CD44 staining and data showing that patient-derived GSCs are enriched for ALDH+ cells and express CD44.
- Published
- 2023
- Full Text
- View/download PDF
35. Data from Potent Antineoplastic Effects of Combined PI3Kα–MNK Inhibition in Medulloblastoma
- Author
-
Leonidas C. Platanias, Rintaro Hashizume, Hidayatullah G. Munshi, Stewart Goldman, Craig Horbinski, David Z. Chen, Quanhong Ma, Ewa M. Kosciuczuk, Gavin T. Blyth, Jessica Clymer, Elspeth M. Beauchamp, Jonathan B. Bell, and Frank Eckerdt
- Abstract
Medulloblastoma is a highly malignant pediatric brain tumor associated with poor outcome. Developing treatments that target the cancer stem cell (CSC) population in medulloblastoma are important to prevent tumor relapse and induce long-lasting clinical responses. We utilized medulloblastoma neurospheres that display CSC characteristics and found activation of the PI3K/AKT pathway in sphere-forming cells. Of all class IA PI3Ks, only the PI3Kα isoform was required for sphere formation by medulloblastoma cells. Knockdown of p110α, but not p110β or p110δ, significantly disrupted cancer stem cell frequencies as determined by extreme limiting dilution analysis (ELDA), indicating an essential role for the PI3Kα catalytic isoform in medulloblastoma CSCs. Importantly, pharmacologic inhibition of the MAPK-interacting kinase (MNK) enhanced the antineoplastic effects of targeted PI3Kα inhibition in medulloblastoma. This indicates that MNK signaling promotes survival in medulloblastoma, suggesting dual PI3Kα and MNK inhibition may provide a novel approach to target and eliminate medulloblastoma CSCs. We also observed a significant reduction in tumor formation in subcutaneous and intracranial mouse xenograft models, which further suggests that this combinatorial approach may represent an efficient therapeutic strategy for medulloblastoma.Implications:These findings raise the possibility of a unique therapeutic approach for medulloblastoma, involving MNK targeting to sensitize medulloblastoma CSCs to PI3Kα inhibition.
- Published
- 2023
- Full Text
- View/download PDF
36. Supplementary Table S1 from Potent Antineoplastic Effects of Combined PI3Kα–MNK Inhibition in Medulloblastoma
- Author
-
Leonidas C. Platanias, Rintaro Hashizume, Hidayatullah G. Munshi, Stewart Goldman, Craig Horbinski, David Z. Chen, Quanhong Ma, Ewa M. Kosciuczuk, Gavin T. Blyth, Jessica Clymer, Elspeth M. Beauchamp, Jonathan B. Bell, and Frank Eckerdt
- Abstract
Pairwise tests for differences in stem cell frequencies.
- Published
- 2023
- Full Text
- View/download PDF
37. Supplementary Figures S1 - S2 from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
These figures show the PN and MES specific drugs identified by the follow-up cell viability screen. Also shown are the enrichment of translation gene sets in MES GSCs. Relates to Figure 1.
- Published
- 2023
- Full Text
- View/download PDF
38. Supplemental Data from Differential Regulation of ZEB1 and EMT by MAPK-Interacting Protein Kinases (MNK) and eIF4E in Pancreatic Cancer
- Author
-
Hidayatullah G. Munshi, Leonidas C. Platanias, Frank D. Eckerdt, David J. Bentrem, Benjamin Kwok, Ahmet D. Arslan, Kazumi Ebine, Christina R. Chow, and Krishan Kumar
- Abstract
Supplemental data related to Figs. 2 and 5 S1. Collagen-dependent MNK phosphorylation in chemoresistant PDAC cells. S2. Targeting eIF4E increases ZEB1 levels in AsPC1 and Panc1 cells. S3. eIF4E and MNK1/2 do not regulate expression of primary transcript of miR-200 (pri-miR-200) microRNAs A and B. S4. miR-141 and miR-200c regulate ZEB1 levels in CD18-CR cells A. S5. Targeting the MNK effector hnRNPA1 increases ZEB1 protein without increasing ZEB1 mRNA levels.
- Published
- 2023
- Full Text
- View/download PDF
39. Data from Targeting ULK1 Decreases IFNγ-Mediated Resistance to Immune Checkpoint Inhibitors
- Author
-
Diana Saleiro, Leonidas C. Platanias, Masha Kocherginsky, Elizabeth T. Bartom, Marcus Bosenberg, I. Caroline Le Poole, Curt M. Horvath, Chidera V. Oku, Chunni Ji, Mariafausta Fischietti, Liliana Ilut, Markella Zannikou, and Sarah E. Fenton
- Abstract
Immune checkpoint inhibitors (ICI) have transformed the treatment of melanoma. However, the majority of patients have primary or acquired resistance to ICIs, limiting durable responses and patient survival. IFNγ signaling and the expression of IFNγ-stimulated genes correlate with either response or resistance to ICIs, in a context-dependent manner. While IFNγ-inducible immunostimulatory genes are required for response to ICIs, chronic IFNγ signaling induces the expression of immunosuppressive genes, promoting resistance to these therapies. Here, we show that high levels of Unc-51 like kinase 1 (ULK1) correlate with poor survival in patients with melanoma and overexpression of ULK1 in melanoma cells enhances IFNγ-induced expression of immunosuppressive genes, with minimal effects on the expression of immunostimulatory genes. In contrast, genetic or pharmacologic inhibition of ULK1 reduces expression of IFNγ-induced immunosuppressive genes. ULK1 binds IRF1 in the nuclear compartment of melanoma cells, controlling its binding to the programmed death-ligand 1 promoter region. In addition, pharmacologic inhibition of ULK1 in combination with anti-programmed cell death protein 1 therapy further reduces melanoma tumor growth in vivo. Our data suggest that targeting ULK1 represses IFNγ-dependent immunosuppression. These findings support the combination of ULK1 drug-targeted inhibition with ICIs for the treatment of patients with melanoma to improve response rates and patient outcomes.Implications:This study identifies ULK1, activated downstream of IFNγ signaling, as a druggable target to overcome resistance mechanisms to ICI therapy in metastatic melanoma.
- Published
- 2023
- Full Text
- View/download PDF
40. Supplementary Table S1 from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
This table shows the initial cell viability screen in GBM cells. Relates to Figure 1C.
- Published
- 2023
- Full Text
- View/download PDF
41. Data from Differential Regulation of ZEB1 and EMT by MAPK-Interacting Protein Kinases (MNK) and eIF4E in Pancreatic Cancer
- Author
-
Hidayatullah G. Munshi, Leonidas C. Platanias, Frank D. Eckerdt, David J. Bentrem, Benjamin Kwok, Ahmet D. Arslan, Kazumi Ebine, Christina R. Chow, and Krishan Kumar
- Abstract
Human pancreatic ductal adenocarcinoma (PDAC) tumors are associated with dysregulation of mRNA translation. In this report, it is demonstrated that PDAC cells grown in collagen exhibit increased activation of the MAPK-interacting protein kinases (MNK) that mediate eIF4E phosphorylation. Pharmacologic and genetic targeting of MNKs reverse epithelial–mesenchymal transition (EMT), decrease cell migration, and reduce protein expression of the EMT-regulator ZEB1 without affecting ZEB1 mRNA levels. Paradoxically, targeting eIF4E, the best-characterized effector of MNKs, increases ZEB1 mRNA expression through repression of ZEB1-targeting miRNAs, miR-200c and miR-141. In contrast, targeting the MNK effector hnRNPA1, which can function as a translational repressor, increases ZEB1 protein without increasing ZEB1 mRNA levels. Importantly, treatment with MNK inhibitors blocks growth of chemoresistant PDAC cells in collagen and decreases the number of aldehyde dehydrogenase activity–positive (Aldefluor+) cells. Significantly, MNK inhibitors increase E-cadherin mRNA levels and decrease vimentin mRNA levels in human PDAC organoids without affecting ZEB1 mRNA levels. Importantly, MNK inhibitors also decrease growth of human PDAC organoids.Implications: These results demonstrate differential regulation of ZEB1 and EMT by MNKs and eIF4E, and identify MNKs as potential targets in pancreatic cancer. Mol Cancer Res; 14(2); 216–27. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
42. Supplementary Figure Legends and Supplementary Figures 1 and 2 from Direct Binding of Arsenic Trioxide to AMPK and Generation of Inhibitory Effects on Acute Myeloid Leukemia Precursors
- Author
-
Leonidas C. Platanias, Jessica K. Altman, Thomas V. O'Halloran, Benoit Viollet, Elden P. Swindell, Dhaval Nanavati, Ruth Serrano, Ewa M. Kosciuczuk, and Elspeth M. Beauchamp
- Abstract
Supplementary Figure Legends. Supplementary Figure 1: Targeted disruption of AMPKα1/2 does not affect arsenicinducible phosphorylation of AKT at Ser473. Supplementary Figure 2: As- Biotin binds to AMPKα as measured by mass spectroscopy
- Published
- 2023
- Full Text
- View/download PDF
43. Supplementary Table 1 from Targeting ULK1 Decreases IFNγ-Mediated Resistance to Immune Checkpoint Inhibitors
- Author
-
Diana Saleiro, Leonidas C. Platanias, Masha Kocherginsky, Elizabeth T. Bartom, Marcus Bosenberg, I. Caroline Le Poole, Curt M. Horvath, Chidera V. Oku, Chunni Ji, Mariafausta Fischietti, Liliana Ilut, Markella Zannikou, and Sarah E. Fenton
- Abstract
List of the putative ULK1 interactor proteins identified by mass spectrometry analysis in A375 cells
- Published
- 2023
- Full Text
- View/download PDF
44. Data from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
Mesenchymal (MES) and proneural (PN) are two distinct glioma stem cell (GSC) populations that drive therapeutic resistance in glioblastoma (GBM). We screened a panel of 650 small molecules against patient-derived GBM cells to discover compounds targeting specific GBM subtypes. Arsenic trioxide (ATO), an FDA-approved drug that crosses the blood–brain barrier, was identified as a potent PN-specific compound in the initial screen and follow-up validation studies. Furthermore, MES and PN GSCs exhibited differential sensitivity to ATO. As ATO has been shown to activate the MAPK-interacting kinase 1 (MNK1)-eukaryotic translation initiation factor 4E (eIF4E) pathway and subsequent mRNA translation in a negative regulatory feedback manner, the mechanistic role of ATO resistance in MES GBM was explored. In GBM cells, ATO-activated translation initiation cellular events via the MNK1–eIF4E signaling axis. Furthermore, resistance to ATO in intracranial PDX tumors correlated with high eIF4E phosphorylation. Polysomal fractionation and microarray analysis of GBM cells were performed to identify ATO's effect on mRNA translation and enrichment of anti-apoptotic mRNAs in the ATO-induced translatome was found. Additionally, it was determined that MNK inhibition sensitized MES GSCs to ATO in neurosphere and apoptosis assays. Finally, examination of the effect of ATO on patients from a phase I/II clinical trial of ATO revealed that PN GBM patients responded better to ATO than other subtypes as demonstrated by longer overall and progression-free survival.Implications: These findings raise the possibility of a unique therapeutic approach for GBM, involving MNK1 targeting to sensitize MES GSCs to drugs like arsenic trioxide. Mol Cancer Res; 16(1); 32–46. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
45. Supplementary Data from Concordance of Genomic Alterations by Next-Generation Sequencing in Tumor Tissue versus Circulating Tumor DNA in Breast Cancer
- Author
-
Massimo Cristofanilli, Francis J. Giles, William Gradishar, Leonidas C. Platanias, Nike Beaubier, Lisa Flaum, Cesar Santa-Maria, Sarika Jain, Andrew A. Davis, and Young Kwang Chae
- Abstract
Supplemental Table 1. Genes and rearrangements studied; Supplemental Table 2: Genes added over time to Guardant360 assay; Supplemental Table 3. Defining concordance and partial concordance for a particular gene; Supplemental Table 4. Concordance and partial concordance among only genes with genomic alterations in either assay; Supplemental Table 5. Sensitivity, specificity, and diagnostic accuracy across 5 genes, including variants of unknown significance (VUS)
- Published
- 2023
- Full Text
- View/download PDF
46. Supplementary Figure S1 from Potent Antineoplastic Effects of Combined PI3Kα–MNK Inhibition in Medulloblastoma
- Author
-
Leonidas C. Platanias, Rintaro Hashizume, Hidayatullah G. Munshi, Stewart Goldman, Craig Horbinski, David Z. Chen, Quanhong Ma, Ewa M. Kosciuczuk, Gavin T. Blyth, Jessica Clymer, Elspeth M. Beauchamp, Jonathan B. Bell, and Frank Eckerdt
- Abstract
Body weight of mice from flank tumor xenograft experiment
- Published
- 2023
- Full Text
- View/download PDF
47. Supplementary Methods from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
Relates to materials & methods.
- Published
- 2023
- Full Text
- View/download PDF
48. Supplementary Figure 2 from Advanced Age Increases Immunosuppression in the Brain and Decreases Immunotherapeutic Efficacy in Subjects with Glioblastoma
- Author
-
Derek A. Wainwright, David H. Munn, George C. Prendergast, Ursula Grohmann, Judith Campisi, Graham Pawelec, Changyou Zhou, Min Wei, Craig Horbinski, Rimas V. Lukas, Karan Dixit, Jeffrey J. Raizer, Priya Kumthekar, Robert M. Prins, Aaron Y. Mochizuki, Leonidas C. Platanias, Joseph R. Podojil, Jennifer D. Wu, Bin Zhang, Masha Kocherginsky, Jiahui Xu, April Bell, Kristen L. Lauing, Lijie Zhai, and Erik Ladomersky
- Abstract
Supplemental Figure 2. Glioblastoma patient overall survival.
- Published
- 2023
- Full Text
- View/download PDF
49. Supp Fig 2 from Pexmetinib: A Novel Dual Inhibitor of Tie2 and p38 MAPK with Efficacy in Preclinical Models of Myelodysplastic Syndromes and Acute Myeloid Leukemia
- Author
-
Amit Verma, Ulrich Steidl, Kith Pradhan, Laurence E. Burgess, Leonidas C. Platanias, Yiyu Zou, David Chantry, Stefan Gross, Mareli Rodriguez, Mark Munson, Dale Wright, Grant Hogeland, Lance Wollenberg, Suzy Brown, Jennifer Garrus, James Rizzi, Chandan Guha, Yiting Yu, Carolina Schinke, Jacqueline Boultwood, Andrea Pellagatti, Shanisha Gordon-Mitchell, Aditi Shastri, Amer Assal, Sanchari Bhattacharyya, Tushar D. Bhagat, Samira Shahnaz, George Nwankwo, Vineeth Sukrithan, Orsi Giricz, Nandini Ramachandra, Matthias Bartenstein, Ioannis Mantzaris, Shannon L. Winski, Kerry Morrone, and Lohith Bachegowda
- Abstract
Knockdown of Angpt1 does not lead to inhibition of leukemic cell viability
- Published
- 2023
- Full Text
- View/download PDF
50. Supp Table 2 from Pexmetinib: A Novel Dual Inhibitor of Tie2 and p38 MAPK with Efficacy in Preclinical Models of Myelodysplastic Syndromes and Acute Myeloid Leukemia
- Author
-
Amit Verma, Ulrich Steidl, Kith Pradhan, Laurence E. Burgess, Leonidas C. Platanias, Yiyu Zou, David Chantry, Stefan Gross, Mareli Rodriguez, Mark Munson, Dale Wright, Grant Hogeland, Lance Wollenberg, Suzy Brown, Jennifer Garrus, James Rizzi, Chandan Guha, Yiting Yu, Carolina Schinke, Jacqueline Boultwood, Andrea Pellagatti, Shanisha Gordon-Mitchell, Aditi Shastri, Amer Assal, Sanchari Bhattacharyya, Tushar D. Bhagat, Samira Shahnaz, George Nwankwo, Vineeth Sukrithan, Orsi Giricz, Nandini Ramachandra, Matthias Bartenstein, Ioannis Mantzaris, Shannon L. Winski, Kerry Morrone, and Lohith Bachegowda
- Abstract
MDS Patient Characteristics
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.