1,159 results on '"Tamayo, Pablo"'
Search Results
2. Clinical and genomic characterization of chemoradiation-resistant HPV-positive oropharyngeal squamous cell carcinoma.
- Author
-
Guo, Theresa, Zamuner, Fernando, Ting, Stephanie, Chen, Liam, Rooper, Lisa, Tamayo, Pablo, Fakhry, Carole, Gaykalova, Daria, and Mehra, Ranee
- Subjects
HPV ,genomics ,oropharyngeal squamous cell carcinoma ,persistent disease ,platinum resistance ,treatment resistance - Abstract
INTRODUCTION: Most patients with HPV-positive oropharyngeal squamous cell carcinoma (OPSCC) have an excellent response to chemoradiation, and trials are now investigating de-escalated treatment. However, up to 25% of patients with HPV-positive OPSCC will experience recurrence, and up to 5% will even progress through primary treatment. Currently, there are no molecular markers to identify patients with poor prognosis who would be harmed by de-escalation. Herein we report the clinical and genomic characteristics of persistent HPV-positive OPSCC after definitive platinum-based chemoradiation therapy. METHODS: Patients with HPV-positive OPSCC treated with curative intent platinum-based chemoradiation between 2007 and 2017 at two institutions and with a persistent locoregional disease were included. We evaluated clinical characteristics, including smoking status, age, stage, treatment, and overall survival. A subset of five patients had tissue available for targeted exome DNA sequencing and RNA sequencing. Genomic analysis was compared to a previously published cohort of 47 treatment-responsive HPV+ OPSCC tumors after batch correction. Mutational landscape, pathway activation, and OncoGPS tumor states were employed to characterize these tumors. RESULTS: Ten patients met the inclusion criteria. The tumor and nodal stages ranged from T1 to T4 and N1 to N2 by AJCC 8th edition staging. All patients were p16-positive by immunohistochemistry, and eight with available in situ hybridization were confirmed to be HPV-positive. The 1-year overall survival from the time of diagnosis was 57%, and the 2-year overall survival was 17%. TP53 mutations were present in three of five (60%) persistent tumors compared to 2% (one of 47) of treatment-responsive HPV-positive tumors (p = 0.008). Other genes with recurrent mutations in persistent HPV-positive OPSCC tumors were NF1, KMT2D, PIK3C2B, and TFGBR2. Compared to treatment-responsive HPV-positive tumors, persistent tumors demonstrated activation of DNA Repair and p53, EMT, MYC, SRC, and TGF-beta signaling pathways, with post-treatment samples demonstrating significant activation of the PI3K-EMT-Stem pathways compared to pretreatment samples. CONCLUSION: Chemoradiation-resistant HPV-positive OPSCC occurs infrequently but portends a poor prognosis. These tumors demonstrate higher rates of p53 mutation and activation of MYC, SRC, and TGF-beta pathways. A comparison of tumors before and after treatment demonstrates PI3K-EMT-Stem pathways post-treatment in HPV-positive tumors with persistent disease after platinum-based chemoradiation.
- Published
- 2024
3. Gαs is dispensable for β-arrestin coupling but dictates GRK selectivity and is predominant for gene expression regulation by β2-adrenergic receptor
- Author
-
Burghi, Valeria, Paradis, Justine S, Officer, Adam, Adame-Garcia, Sendi Rafael, Wu, Xingyu, Matthees, Edda SF, Barsi-Rhyne, Benjamin, Ramms, Dana J, Clubb, Lauren, Acosta, Monica, Tamayo, Pablo, Bouvier, Michel, Inoue, Asuka, von Zastrow, Mark, Hoffmann, Carsten, and Gutkind, J Silvio
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,Biotechnology ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Underpinning research ,Aetiology ,Generic health relevance ,Humans ,beta-Arrestin 1 ,beta-Arrestin 2 ,beta-Arrestins ,Cyclic AMP-Dependent Protein Kinases ,Gene Expression Regulation ,GTP-Binding Proteins ,Mitogen-Activated Protein Kinases ,Phosphorylation ,Receptors ,Adrenergic ,beta-2 ,HEK293 Cells ,GTP-Binding Protein alpha Subunits ,Protein Structure ,Tertiary ,Protein Isoforms ,Enzyme Activation ,G protein ,G protein–coupled receptor ,gene expression ,signaling ,β-arrestin ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
β-arrestins play a key role in G protein-coupled receptor (GPCR) internalization, trafficking, and signaling. Whether β-arrestins act independently of G protein-mediated signaling has not been fully elucidated. Studies using genome-editing approaches revealed that whereas G proteins are essential for mitogen-activated protein kinase activation by GPCRs., β-arrestins play a more prominent role in signal compartmentalization. However, in the absence of G proteins, GPCRs may not activate β-arrestins, thereby limiting the ability to distinguish G protein from β-arrestin-mediated signaling events. We used β2-adrenergic receptor (β2AR) and its β2AR-C tail mutant expressed in human embryonic kidney 293 cells wildtype or CRISPR-Cas9 gene edited for Gαs, β-arrestin1/2, or GPCR kinases 2/3/5/6 in combination with arrestin conformational sensors to elucidate the interplay between Gαs and β-arrestins in controlling gene expression. We found that Gαs is not required for β2AR and β-arrestin conformational changes, β-arrestin recruitment, and receptor internalization, but that Gαs dictates the GPCR kinase isoforms involved in β-arrestin recruitment. By RNA-Seq analysis, we found that protein kinase A and mitogen-activated protein kinase gene signatures were activated by stimulation of β2AR in wildtype and β-arrestin1/2-KO cells but absent in Gαs-KO cells. These results were validated by re-expressing Gαs in the corresponding KO cells and silencing β-arrestins in wildtype cells. These findings were extended to cellular systems expressing endogenous levels of β2AR. Overall, our results support that Gs is essential for β2AR-promoted protein kinase A and mitogen-activated protein kinase gene expression signatures, whereas β-arrestins initiate signaling events modulating Gαs-driven nuclear transcriptional activity.
- Published
- 2023
4. Syk inhibition reprograms tumor-associated macrophages and overcomes gemcitabine-induced immunosuppression in pancreatic ductal adenocarcinoma
- Author
-
Rohila, Deepak, Park, In Hwan, Pham, Timothy V, Weitz, Jonathan, de Mendoza, Tatiana Hurtado, Madheswaran, Suresh, Ishfaq, Mehreen, Beaman, Cooper, Tapia, Elisabette, Sun, Siming, Patel, Jay, Tamayo, Pablo, Lowy, Andrew M, and Joshi, Shweta
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Rare Diseases ,Pancreatic Cancer ,Digestive Diseases ,Cancer ,Development of treatments and therapeutic interventions ,2.1 Biological and endogenous factors ,Aetiology ,5.2 Cellular and gene therapies ,Mice ,Animals ,Humans ,Gemcitabine ,Tumor-Associated Macrophages ,Carcinoma ,Pancreatic Ductal ,Pancreatic Neoplasms ,Immune Tolerance ,Immunosuppression Therapy ,Tumor Microenvironment ,Cell Line ,Tumor ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an insidious disease with a low 5-year survival rate. PDAC is characterized by infiltration of abundant tumor-associated macrophages (TAM), which promote immune tolerance and immunotherapeutic resistance. Here we report that macrophage spleen tyrosine kinase (Syk) promotes PDAC growth and metastasis. In orthotopic PDAC mouse models, genetic deletion of myeloid Syk reprogrammed macrophages into immunostimulatory phenotype, increased the infiltration, proliferation, and cytotoxicity of CD8+ T cells, and repressed PDAC growth and metastasis. Furthermore, gemcitabine (Gem) treatment induced an immunosuppressive microenvironment in PDAC by promoting protumorigenic polarization of macrophages. In contrast, treatment with the FDA-approved Syk inhibitor R788 (fostamatinib) remodeled the tumor immune microenvironment, "re-educated" protumorigenic macrophages towards an immunostimulatory phenotype and boosted CD8+ T-cell responses in Gem-treated PDAC in orthotopic mouse models and an ex vivo human pancreatic slice culture model. These findings illustrate the potential of Syk inhibition for enhancing the antitumor immune responses in PDAC and support the clinical evaluation of R788 either alone or together with Gem as a potential treatment strategy for PDAC.SignificanceSyk blockade induces macrophage polarization to an immunostimulatory phenotype, which enhances CD8+ T-cell responses and improves gemcitabine efficacy in pancreatic ductal adenocarcinoma, a clinically challenging malignancy.
- Published
- 2023
5. Multimodal perturbation analyses of cyclin-dependent kinases reveal a network of synthetic lethalities associated with cell-cycle regulation and transcriptional regulation.
- Author
-
Ford, Kyle, Munson, Brenton P, Fong, Samson H, Panwala, Rebecca, Chu, Wai Keung, Rainaldi, Joseph, Plongthongkum, Nongluk, Arunachalam, Vinayagam, Kostrowicki, Jarek, Meluzzi, Dario, Kreisberg, Jason F, Jensen-Pergakes, Kristen, VanArsdale, Todd, Paul, Thomas, Tamayo, Pablo, Zhang, Kun, Bienkowska, Jadwiga, Mali, Prashant, and Ideker, Trey
- Subjects
Humans ,Neoplasms ,Cell Cycle ,Protein-Arginine N-Methyltransferases ,Cell Cycle Checkpoints ,Rare Diseases ,Breast Cancer ,Orphan Drug ,Genetics ,Cancer ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Generic health relevance - Abstract
Cell-cycle control is accomplished by cyclin-dependent kinases (CDKs), motivating extensive research into CDK targeting small-molecule drugs as cancer therapeutics. Here we use combinatorial CRISPR/Cas9 perturbations to uncover an extensive network of functional interdependencies among CDKs and related factors, identifying 43 synthetic-lethal and 12 synergistic interactions. We dissect CDK perturbations using single-cell RNAseq, for which we develop a novel computational framework to precisely quantify cell-cycle effects and diverse cell states orchestrated by specific CDKs. While pairwise disruption of CDK4/6 is synthetic-lethal, only CDK6 is required for normal cell-cycle progression and transcriptional activation. Multiple CDKs (CDK1/7/9/12) are synthetic-lethal in combination with PRMT5, independent of cell-cycle control. In-depth analysis of mRNA expression and splicing patterns provides multiple lines of evidence that the CDK-PRMT5 dependency is due to aberrant transcriptional regulation resulting in premature termination. These inter-dependencies translate to drug-drug synergies, with therapeutic implications in cancer and other diseases.
- Published
- 2023
6. Targeting macrophage Syk enhances responses to immune checkpoint blockade and radiotherapy in high-risk neuroblastoma
- Author
-
Rohila, Deepak, Park, In Hwan, Pham, Timothy V, Jones, Riley, Tapia, Elisabette, Liu, Kevin X, Tamayo, Pablo, Yu, Alice, Sharabi, Andrew B, and Joshi, Shweta
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Neuroblastoma ,Cancer ,Pediatric ,Neurosciences ,Immunotherapy ,Orphan Drug ,Rare Diseases ,Pediatric Cancer ,Animals ,Mice ,Immune Checkpoint Inhibitors ,N-Myc Proto-Oncogene Protein ,Macrophages ,Tumor Microenvironment ,macrophage ,neuroblastoma ,immune suppression ,Syk ,T cells ,Medical Microbiology ,Biochemistry and cell biology ,Genetics - Abstract
BackgroundNeuroblastoma (NB) is considered an immunologically cold tumor and is usually less responsive to immune checkpoint blockade (ICB). Tumor-associated macrophages (TAMs) are highly infiltrated in NB tumors and promote immune escape and resistance to ICB. Hence therapeutic strategies targeting immunosuppressive TAMs can improve responses to ICB in NB. We recently discovered that spleen tyrosine kinase (Syk) reprograms TAMs toward an immunostimulatory phenotype and enhances T-cell responses in the lung adenocarcinoma model. Here we investigated if Syk is an immune-oncology target in NB and tested whether a novel immunotherapeutic approach utilizing Syk inhibitor together with radiation and ICB could provide a durable anti-tumor immune response in an MYCN amplified murine model of NB.MethodsMyeloid Syk KO mice and syngeneic MYCN-amplified cell lines were used to elucidate the effect of myeloid Syk on the NB tumor microenvironment (TME). In addition, the effect of Syk inhibitor, R788, on anti-tumor immunity alone or in combination with anti-PDL1 mAb and radiation was also determined in murine NB models. The underlying mechanism of action of this novel therapeutic combination was also investigated.ResultsHerein, we report that Syk is a marker of NB-associated macrophages and plays a crucial role in promoting immunosuppression in the NB TME. We found that the blockade of Syk in NB-bearing mice markedly impairs tumor growth. This effect is facilitated by macrophages that become immunogenic in the absence of Syk, skewing the suppressive TME towards immunostimulation and activating anti-tumor immune responses. Moreover, combining FDA-approved Syk inhibitor, R788 (fostamatinib) along with anti-PDL1 mAb provides a synergistic effect leading to complete tumor regression and durable anti-tumor immunity in mice bearing small tumors (50 mm3) but not larger tumors (250 mm3). However, combining radiation to R788 and anti-PDL1 mAb prolongs the survival of mice bearing large NB9464 tumors.ConclusionCollectively, our findings demonstrate the central role of macrophage Syk in NB progression and demonstrate that Syk blockade can "reeducate" TAMs towards immunostimulatory phenotype, leading to enhanced T cell responses. These findings further support the clinical evaluation of fostamatinib alone or with radiation and ICB, as a novel therapeutic intervention in neuroblastoma.
- Published
- 2023
7. Peace polysemy in business and peace agendas
- Author
-
Uribe-Jaramillo, Maria Teresa and Zapata-Tamayo, Pablo
- Published
- 2024
- Full Text
- View/download PDF
8. Inferring cellular and molecular processes in single-cell data with non-negative matrix factorization using Python, R and GenePattern Notebook implementations of CoGAPS
- Author
-
Johnson, Jeanette A. I., Tsang, Ashley P., Mitchell, Jacob T., Zhou, David L., Bowden, Julia, Davis-Marcisak, Emily, Sherman, Thomas, Liefeld, Ted, Loth, Melanie, Goff, Loyal A., Zimmerman, Jacquelyn W., Kinny-Köster, Ben, Jaffee, Elizabeth M., Tamayo, Pablo, Mesirov, Jill P., Reich, Michael, Fertig, Elana J., and Stein-O’Brien, Genevieve L.
- Published
- 2023
- Full Text
- View/download PDF
9. Analysis of CDK12 alterations in a pan‐cancer database
- Author
-
Pan, Elizabeth, Cabal, Angelo, Javier‐DesLoges, Juan, Patel, Devin, Panian, Justine, Lee, Suzanna, Shaya, Justin, Nonato, Taylor, Xu, Xiaojun, Stewart, Tyler, Rose, Brent, Shabaik, Ahmed, Cohen, Ezra, Kurzrock, Razelle, Tamayo, Pablo, and McKay, Rana R
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Clinical Research ,Colo-Rectal Cancer ,Cancer ,Human Genome ,Genetics ,Digestive Diseases ,Rare Diseases ,2.1 Biological and endogenous factors ,Aetiology ,Good Health and Well Being ,Cyclin-Dependent Kinases ,Data Management ,Humans ,Male ,Neoplasms ,Prostate ,Retrospective Studies ,biomarkers ,cancer genetics ,clinical cancer research ,genomics ,Biochemistry and Cell Biology ,Oncology and carcinogenesis - Abstract
BackgroundCDK12 inactivation leading to increased neoantigen burden has been hypothesized to sensitize tumors to immune checkpoint inhibition. Pan-cancer data regarding the frequency of CDK12 alterations are limited. We aimed to characterize CDK12 alterations across all cancer types through real-world clinical-grade sequencing.MethodsThis was a single-center retrospective analysis of 4994 cancer patients who underwent tissue or blood genomic profiling, including CDK12 assessment, conducted as part of routine care from December 2012 to January 2020. Prevalence, clinical characteristics, and treatment outcomes of patients with tumors with pathogenic CDK12 alterations were described.ResultsIn all, 39 (0.78%, n = 39/4994) patients had pathogenic CDK12 alterations. Among CDK12-altered tumors, the most common organ site was prostate (n = 9, 23.1%) followed by colorectal (n = 5, 12.8%). Adenocarcinoma was the most common histology (n = 26, 66.7%). Median follow-up from time of diagnosis was 4.02 years. Median overall survival from time of metastasis was 4.43 years (95% CI: 3.11-5.74). Ten patients with CDK12-altered tumors received at least one immune checkpoint inhibitor-containing regimen. The majority of patients (n = 6/10, 60%) experienced an objective response. Progression-free survival for patients who had metastatic disease and received a checkpoint inhibitor-containing regimen was 1.16 years (95% CI: 0.32-2.00).ConclusionCDK12 alterations are rare events across hematologic and solid tumor malignancies. They represent a clinically distinct molecular cancer subtype which may have increased responsiveness to checkpoint inhibition. Prospective studies are warranted to investigate checkpoint inhibition in CDK12-altered tumors.
- Published
- 2022
10. Differential regulation of TNFα and IL-6 expression contributes to immune evasion in prostate cancer
- Author
-
Deichaite, Ida, Sears, Timothy J, Sutton, Leisa, Rebibo, Daniel, Morgan, Kylie, Nelson, Tyler, Rose, Brent, Tamayo, Pablo, Ferrara, Napoleone, Asimakopoulos, Fotis, and Carter, Hannah
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Cancer ,Prostate Cancer ,Urologic Diseases ,Aging ,2.1 Biological and endogenous factors ,Aetiology ,Inflammatory and immune system ,Male ,Humans ,Immune Evasion ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,Tumor Necrosis Factor Inhibitors ,Prostatic Neoplasms ,Inflammation ,Tumor Microenvironment ,Prostate cancer ,TNF ,IL-6 ,ADAMTS-4 ,AP-1 ,FOSB ,Bevacizumab ,SELE ,Macrophage polarization ,Immune remodeling ,prostate cancer ,bevacizumab ,macrophage polarization ,immune remodeling ,Medical and Health Sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
BackgroundThe role of the inflammatory milieu in prostate cancer progression is not well understood. Differences in inflammatory signaling between localized and metastatic disease may point to opportunities for early intervention.MethodsWe modeled PCa disease progression by analyzing RNA-seq of localized vs. metastatic patient samples, followed by CIBERSORTx to assess their immune cell populations. The VHA CDW registry of PCa patients was analyzed for anti-TNF clinical outcomes.ResultsWe observed statistically significant opposing patterns of IL-6 and TNFα expression between localized and metastatic disease. IL-6 was robustly expressed in localized disease and downregulated in metastatic disease. The reverse was observed with TNFα expression. Metastatic disease was also characterized by downregulation of adhesion molecule E-selectin, matrix metalloproteinase ADAMTS-4 and a shift to M2 macrophages whereas localized disease demonstrated a preponderance of M1 macrophages. Treatment with anti-TNF agents was associated with earlier stage disease at diagnosis.ConclusionsOur data points to clearly different inflammatory contexts between localized and metastatic prostate cancer. Primary localized disease demonstrates local inflammation and adaptive immunity, whereas metastases are characterized by immune cold microenvironments and a shift towards resolution of inflammation and tissue repair. Therapies that interfere with these inflammatory networks may offer opportunities for early intervention in monotherapy or in combination with immunotherapies and anti-angiogenic approaches.
- Published
- 2022
11. CHMP2A regulates tumor sensitivity to natural killer cell-mediated cytotoxicity
- Author
-
Bernareggi, Davide, Xie, Qi, Prager, Briana C, Yun, Jiyoung, Cruz, Luisjesus S, Pham, Timothy V, Kim, William, Lee, Xiqing, Coffey, Michael, Zalfa, Cristina, Azmoon, Pardis, Zhu, Huang, Tamayo, Pablo, Rich, Jeremy N, and Kaufman, Dan S
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Immunology ,Oncology and Carcinogenesis ,Dental/Oral and Craniofacial Disease ,Rare Diseases ,Cancer ,Stem Cell Research ,Vaccine Related ,2.1 Biological and endogenous factors ,Aetiology ,Animals ,Apoptosis ,Cell Line ,Tumor ,Cytotoxicity ,Immunologic ,Endosomal Sorting Complexes Required for Transport ,Head and Neck Neoplasms ,Humans ,Immunotherapy ,Killer Cells ,Natural ,Mice ,Squamous Cell Carcinoma of Head and Neck - Abstract
Natural killer (NK) cells are known to mediate killing of various cancer types, but tumor cells can develop resistance mechanisms to escape NK cell-mediated killing. Here, we use a "two cell type" whole genome CRISPR-Cas9 screening system to discover key regulators of tumor sensitivity and resistance to NK cell-mediated cytotoxicity in human glioblastoma stem cells (GSC). We identify CHMP2A as a regulator of GSC resistance to NK cell-mediated cytotoxicity and we confirm these findings in a head and neck squamous cells carcinoma (HNSCC) model. We show that deletion of CHMP2A activates NF-κB in tumor cells to mediate increased chemokine secretion that promotes NK cell migration towards tumor cells. In the HNSCC model we demonstrate that CHMP2A mediates tumor resistance to NK cells via secretion of extracellular vesicles (EVs) that express MICA/B and TRAIL. These secreted ligands induce apoptosis of NK cells to inhibit their antitumor activity. To confirm these in vitro studies, we demonstrate that deletion of CHMP2A in CAL27 HNSCC cells leads to increased NK cell-mediated killing in a xenograft immunodeficient mouse model. These findings illustrate a mechanism of tumor immune escape through EVs secretion and identify inhibition of CHMP2A and related targets as opportunities to improve NK cell-mediated immunotherapy.
- Published
- 2022
12. Human Induced Pluripotent Stem Cell-Derived Macrophages Ameliorate Liver Fibrosis
- Author
-
Pouyanfard, Somayeh, Meshgin, Nairika, Cruz, Luisjesus S, Diggle, Karin, Hashemi, Hamidreza, Pham, Timothy V, Fierro, Manuel, Tamayo, Pablo, Fanjul, Andrea, Kisseleva, Tatiana, and Kaufman, Dan S
- Subjects
Medical Biotechnology ,Biomedical and Clinical Sciences ,Regenerative Medicine ,Liver Disease ,Digestive Diseases ,Chronic Liver Disease and Cirrhosis ,Stem Cell Research ,Rare Diseases ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,2.1 Biological and endogenous factors ,Development of treatments and therapeutic interventions ,5.2 Cellular and gene therapies ,Aetiology ,Oral and gastrointestinal ,Inflammatory and immune system ,Good Health and Well Being ,Animals ,Cell Differentiation ,Cytokines ,Humans ,Induced Pluripotent Stem Cells ,Liver Cirrhosis ,Macrophages ,Mice ,cellular therapy ,induced pluripotent stem cells ,liver ,liver regeneration ,Biological Sciences ,Technology ,Medical and Health Sciences ,Immunology ,Biological sciences ,Biomedical and clinical sciences - Abstract
With an increasing number of patients with degenerative hepatic diseases, such as liver fibrosis, and a limited supply of donor organs, there is an unmet need for therapies that can repair or regenerate damaged liver tissue. Treatment with macrophages that are capable of phagocytosis and anti-inflammatory activities such as secretion of matrix metalloproteinases (MMPs) provide an attractive cellular therapy approach. Human induced pluripotent stem cells (iPSCs) are capable of efficiently generating a large-scale, homogenous population of human macrophages using fully defined feeder- and serum-free differentiation protocol. Human iPSC-macrophages exhibit classical surface cell markers and phagocytic activity similar to peripheral blood-derived macrophages. Moreover, gene and cytokine expression analysis reveal that these macrophages can be efficiently polarized to pro-inflammatory M1 or anti-inflammatory M2 phenotypes in presence of LPS + IFN-γ and IL-4 + IL-13, respectively. M1 macrophages express high level of CD80, TNF-α, and IL-6 while M2 macrophages show elevated expression of CD206, CCL17, and CCL22. Here, we demonstrate that treatment of liver fibrosis with both human iPSC-derived macrophage populations and especially M2 subtype significantly reduces fibrogenic gene expression and disease associated histological markers including Sirius Red, αSMA and desmin in immunodeficient Rag2-/- γc-/- mice model, making this approach a promising cell-based avenue to ameliorate fibrosis.
- Published
- 2021
13. An expanded universe of cancer targets.
- Author
-
Hahn, William C, Bader, Joel S, Braun, Theodore P, Califano, Andrea, Clemons, Paul A, Druker, Brian J, Ewald, Andrew J, Fu, Haian, Jagu, Subhashini, Kemp, Christopher J, Kim, William, Kuo, Calvin J, McManus, Michael, B Mills, Gordon, Mo, Xiulei, Sahni, Nidhi, Schreiber, Stuart L, Talamas, Jessica A, Tamayo, Pablo, Tyner, Jeffrey W, Wagner, Bridget K, Weiss, William A, Gerhard, Daniela S, and Cancer Target Discovery and Development Network
- Subjects
Cancer Target Discovery and Development Network ,Animals ,Humans ,Neoplasms ,Disease Models ,Animal ,Drug Delivery Systems ,Genomics ,Tumor Escape ,Clinical Trials as Topic ,Tumor Microenvironment ,Biotechnology ,Rare Diseases ,Cancer ,Orphan Drug ,Genetics ,2.1 Biological and endogenous factors ,Developmental Biology ,Biological Sciences ,Medical and Health Sciences - Abstract
The characterization of cancer genomes has provided insight into somatically altered genes across tumors, transformed our understanding of cancer biology, and enabled tailoring of therapeutic strategies. However, the function of most cancer alleles remains mysterious, and many cancer features transcend their genomes. Consequently, tumor genomic characterization does not influence therapy for most patients. Approaches to understand the function and circuitry of cancer genes provide complementary approaches to elucidate both oncogene and non-oncogene dependencies. Emerging work indicates that the diversity of therapeutic targets engendered by non-oncogene dependencies is much larger than the list of recurrently mutated genes. Here we describe a framework for this expanded list of cancer targets, providing novel opportunities for clinical translation.
- Published
- 2021
14. BTK Inhibition Reverses MDSC-Mediated Immunosuppression and Enhances Response to Anti-PDL1 Therapy in Neuroblastoma.
- Author
-
Ishfaq, Mehreen, Pham, Timothy, Beaman, Cooper, Tamayo, Pablo, Yu, Alice L, and Joshi, Shweta
- Subjects
Bruton tyrosine kinase ,immunosuppression ,myeloid derived suppressor cells ,neuroblastoma ,Oncology and Carcinogenesis - Abstract
MDSCs are immune cells of myeloid lineage that plays a key role in promoting tumor growth. The expansion of MDSCs in tumor-bearing hosts reduces the efficacy of checkpoint inhibitors and CAR-T therapies, and hence strategies that deplete or block the recruitment of MDSCs have shown benefit in improving responses to immunotherapy in various cancers, including NB. Ibrutinib, an irreversible molecular inhibitor of BTK, has been widely studied in B cell malignancies, and recently, this drug is repurposed for the treatment of solid tumors. Herein we report that BTK is highly expressed in both granulocytic and monocytic murine MDSCs isolated from mice bearing NB tumors, and its increased expression correlates with a poor relapse-free survival probability of NB patients. Moreover, in vitro treatment of murine MDSCs with ibrutinib altered NO production, decreased mRNA expression of Ido, Arg, Tgfβ, and displayed defects in T-cell suppression. Consistent with these findings, in vivo inhibition of BTK with ibrutinib resulted in reduced MDSC-mediated immune suppression, increased CD8+ T cell infiltration, decreased tumor growth, and improved response to anti-PDL1 checkpoint inhibitor therapy in a murine model of NB. These results demonstrate that ibrutinib modulates immunosuppressive functions of MDSC and can be used either alone or in combination with immunotherapy for augmenting antitumor immune responses in NB.
- Published
- 2021
15. SPT6 promotes epidermal differentiation and blockade of an intestinal-like phenotype through control of transcriptional elongation.
- Author
-
Li, Jingting, Xu, Xiaojun, Tiwari, Manisha, Chen, Yifang, Fuller, Mackenzie, Bansal, Varun, Tamayo, Pablo, Das, Soumita, Ghosh, Pradipta, and Sen, George L
- Subjects
Epidermis ,Cells ,Cultured ,Keratinocytes ,Humans ,RNA Polymerase II ,Tumor Suppressor Proteins ,Transcription Factors ,RNA ,Small Interfering ,Tissue Culture Techniques ,Cell Differentiation ,Infant ,Newborn ,Male ,Adult Stem Cells ,Cell Transdifferentiation ,Promoter Regions ,Genetic ,Gene Knockdown Techniques ,Primary Cell Culture ,Transcription Elongation ,Genetic ,Chromatin Immunoprecipitation Sequencing ,RNA-Seq - Abstract
In adult tissue, stem and progenitor cells must tightly regulate the balance between proliferation and differentiation to sustain homeostasis. How this exquisite balance is achieved is an area of active investigation. Here, we show that epidermal genes, including ~30% of induced differentiation genes already contain stalled Pol II at the promoters in epidermal stem and progenitor cells which is then released into productive transcription elongation upon differentiation. Central to this process are SPT6 and PAF1 which are necessary for the elongation of these differentiation genes. Upon SPT6 or PAF1 depletion there is a loss of human skin differentiation and stratification. Unexpectedly, loss of SPT6 also causes the spontaneous transdifferentiation of epidermal cells into an intestinal-like phenotype due to the stalled transcription of the master regulator of epidermal fate P63. Our findings suggest that control of transcription elongation through SPT6 plays a prominent role in adult somatic tissue differentiation and the inhibition of alternative cell fate choices.
- Published
- 2021
16. Functional Precision Medicine Identifies New Therapeutic Candidates for Medulloblastoma
- Author
-
Rusert, Jessica M, Juarez, Edwin F, Brabetz, Sebastian, Jensen, James, Garancher, Alexandra, Chau, Lianne Q, Tacheva-Grigorova, Silvia K, Wahab, Sameerah, Udaka, Yoko T, Finlay, Darren, Seker-Cin, Huriye, Reardon, Brendan, Gröbner, Susanne, Serrano, Jonathan, Ecker, Jonas, Qi, Lin, Kogiso, Mari, Du, Yuchen, Baxter, Patricia A, Henderson, Jacob J, Berens, Michael E, Vuori, Kristiina, Milde, Till, Cho, Yoon-Jae, Li, Xiao-Nan, Olson, James M, Reyes, Iris, Snuderl, Matija, Wong, Terence C, Dimmock, David P, Nahas, Shareef A, Malicki, Denise, Crawford, John R, Levy, Michael L, Van Allen, Eliezer M, Pfister, Stefan M, Tamayo, Pablo, Kool, Marcel, Mesirov, Jill P, and Wechsler-Reya, Robert J
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Clinical Sciences ,Oncology and Carcinogenesis ,Brain Disorders ,Neurosciences ,Pediatric ,Orphan Drug ,Biotechnology ,Genetic Testing ,Cancer ,Human Genome ,Pediatric Research Initiative ,Rare Diseases ,Brain Cancer ,Pediatric Cancer ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Generic health relevance ,Good Health and Well Being ,Animals ,Antineoplastic Agents ,Cell Line ,Tumor ,Cerebellar Neoplasms ,Child ,Dactinomycin ,Gene Expression Regulation ,Neoplastic ,High-Throughput Screening Assays ,Humans ,Male ,Medulloblastoma ,Mice ,Inbred NOD ,Mutation ,Polymorphism ,Single Nucleotide ,Precision Medicine ,Exome Sequencing ,Xenograft Model Antitumor Assays ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Medulloblastoma is among the most common malignant brain tumors in children. Recent studies have identified at least four subgroups of the disease that differ in terms of molecular characteristics and patient outcomes. Despite this heterogeneity, most patients with medulloblastoma receive similar therapies, including surgery, radiation, and intensive chemotherapy. Although these treatments prolong survival, many patients still die from the disease and survivors suffer severe long-term side effects from therapy. We hypothesize that each patient with medulloblastoma is sensitive to different therapies and that tailoring therapy based on the molecular and cellular characteristics of patients' tumors will improve outcomes. To test this, we assembled a panel of orthotopic patient-derived xenografts (PDX) and subjected them to DNA sequencing, gene expression profiling, and high-throughput drug screening. Analysis of DNA sequencing revealed that most medulloblastomas do not have actionable mutations that point to effective therapies. In contrast, gene expression and drug response data provided valuable information about potential therapies for every tumor. For example, drug screening demonstrated that actinomycin D, which is used for treatment of sarcoma but rarely for medulloblastoma, was active against PDXs representing Group 3 medulloblastoma, the most aggressive form of the disease. Functional analysis of tumor cells was successfully used in a clinical setting to identify more treatment options than sequencing alone. These studies suggest that it should be possible to move away from a one-size-fits-all approach and begin to treat each patient with therapies that are effective against their specific tumor. SIGNIFICANCE: These findings show that high-throughput drug screening identifies therapies for medulloblastoma that cannot be predicted by genomic or transcriptomic analysis.
- Published
- 2020
17. HPV E2, E4, E5 drive alternative carcinogenic pathways in HPV positive cancers
- Author
-
Ren, Shuling, Gaykalova, Daria A, Guo, Theresa, Favorov, Alexander V, Fertig, Elana J, Tamayo, Pablo, Callejas-Valera, Juan Luis, Allevato, Mike, Gilardi, Mara, Santos, Jessica, Fukusumi, Takahito, Sakai, Akihiro, Ando, Mizuo, Sadat, Sayed, Liu, Chao, Xu, Guorong, Fisch, Kathleen M, Wang, Zhiyong, Molinolo, Alfredo A, Gutkind, J Silvio, Ideker, Trey, Koch, Wayne M, and Califano, Joseph A
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Cervical Cancer ,Infectious Diseases ,Genetics ,Human Genome ,Sexually Transmitted Infections ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,Antineoplastic Combined Chemotherapy Protocols ,Carcinogenesis ,Cell Line ,Tumor ,Cell Proliferation ,Datasets as Topic ,Disease Models ,Animal ,Disease-Free Survival ,Female ,Gene Expression Regulation ,Neoplastic ,Gene Expression Regulation ,Viral ,Host-Pathogen Interactions ,Human papillomavirus 16 ,Humans ,Mice ,Mice ,Transgenic ,Oncogene Proteins ,Viral ,Papillomavirus Infections ,Pharyngeal Neoplasms ,Primary Cell Culture ,Receptors ,Fibroblast Growth Factor ,Signal Transduction ,Squamous Cell Carcinoma of Head and Neck ,TOR Serine-Threonine Kinases ,Tumor Suppressor Protein p53 ,Uterine Cervical Neoplasms ,Clinical Sciences ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
The dominant paradigm for HPV carcinogenesis includes integration into the host genome followed by expression of E6 and E7 (E6/E7). We explored an alternative carcinogenic pathway characterized by episomal E2, E4, and E5 (E2/E4/E5) expression. Half of HPV positive cervical and pharyngeal cancers comprised a subtype with increase in expression of E2/E4/E5, as well as association with lack of integration into the host genome. Models of the E2/E4/E5 carcinogenesis show p53 dependent enhanced proliferation in vitro, as well as increased susceptibility to induction of cancer in vivo. Whole genomic expression analysis of the E2/E4/E5 pharyngeal cancer subtype is defined by activation of the fibroblast growth factor receptor (FGFR) pathway and this subtype is susceptible to combination FGFR and mTOR inhibition, with implications for targeted therapy.
- Published
- 2020
18. Mesenchymal and MAPK Expression Signatures Associate with Telomerase Promoter Mutations in Multiple Cancers
- Author
-
Stern, Josh Lewis, Hibshman, Grace, Hu, Kevin, Ferrara, Sarah E, Costello, James C, Kim, William, Tamayo, Pablo, Cech, Thomas R, and Huang, Franklin W
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Cancer ,Genetics ,2.1 Biological and endogenous factors ,Aetiology ,Generic health relevance ,Cell Line ,Tumor ,Chromatin Immunoprecipitation ,Epithelial-Mesenchymal Transition ,Extracellular Signal-Regulated MAP Kinases ,Gene Expression Profiling ,Gene Expression Regulation ,Neoplastic ,Gene Regulatory Networks ,Humans ,Mutation ,Neoplasms ,Promoter Regions ,Genetic ,Sequence Analysis ,RNA ,Small Molecule Libraries ,Telomerase ,Tumor Microenvironment ,Oncology and Carcinogenesis ,Developmental Biology ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
In a substantial fraction of cancers TERT promoter (TERTp) mutations drive expression of the catalytic subunit of telomerase, contributing to their proliferative immortality. We conducted a pan-cancer analysis of cell lines and find a TERTp mutation expression signature dominated by epithelial-to-mesenchymal transition and MAPK signaling. These data indicate that TERTp mutants are likely to generate distinctive tumor microenvironments and intercellular interactions. Analysis of high-throughput screening tests of 546 small molecules on cell line growth indicated that TERTp mutants displayed heightened sensitivity to specific drugs, including RAS pathway inhibitors, and we found that inhibition of MEK1 and 2, key RAS/MAPK pathway effectors, inhibited TERT mRNA expression. Consistent with an enrichment of mesenchymal states in TERTp mutants, cell lines and some patient tumors displayed low expression of the central adherens junction protein E-cadherin, and we provide evidence that its expression in these cells is regulated by MEK1/2. Several mesenchymal transcription factors displayed elevated expression in TERTp mutants including ZEB1 and 2, TWIST1 and 2, and SNAI1. Of note, the developmental transcription factor SNAI2/SLUG was conspicuously elevated in a significant majority of TERTp-mutant cell lines, and knock-down experiments suggest that it promotes TERT expression. IMPLICATIONS: Cancers harboring TERT promoter mutations are often more lethal, but the basis for this higher mortality remains unknown. Our study identifies that TERTp mutants, as a class, associate with a distinct gene and protein expression signature likely to impact their biological and clinical behavior and provide new directions for investigating treatment approaches for these cancers.
- Published
- 2020
19. Cannabinoids Promote Progression of HPV-Positive Head and Neck Squamous Cell Carcinoma via p38 MAPK Activation
- Author
-
Liu, Chao, Sadat, Sayed H, Ebisumoto, Koji, Sakai, Akihiro, Panuganti, Bharat A, Ren, Shuling, Goto, Yusuke, Haft, Sunny, Fukusumi, Takahito, Ando, Mizuo, Saito, Yuki, Guo, Theresa, Tamayo, Pablo, Yeerna, Huwate, Kim, William, Hubbard, Jacqueline, Sharabi, Andrew B, Gutkind, J Silvio, and Califano, Joseph A
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Drug Abuse (NIDA only) ,Sexually Transmitted Infections ,Infectious Diseases ,Substance Misuse ,Genetics ,Dental/Oral and Craniofacial Disease ,Rare Diseases ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,Apoptosis ,Cannabinoids ,Cell Movement ,Cell Proliferation ,Female ,Head and Neck Neoplasms ,Humans ,Mice ,Mice ,Nude ,Papillomaviridae ,Papillomavirus Infections ,Prognosis ,Receptors ,Cannabinoid ,Squamous Cell Carcinoma of Head and Neck ,Tumor Cells ,Cultured ,Xenograft Model Antitumor Assays ,p38 Mitogen-Activated Protein Kinases ,Oncology & Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis - Abstract
PurposeHuman papillomavirus (HPV)-related head and neck squamous cell carcinoma (HNSCC) is associated with daily marijuana use and is also increasing in parallel with increased marijuana use in the United States. Our study is designed to define the interaction between cannabinoids and HPV-positive HNSCC.Experimental designThe expression of cannabinoid receptors CNR1 and CNR2 was analyzed using The Cancer Genome Atlas (TCGA) HNSCC data. We used agonists, antagonists, siRNAs, or shRNA-based models to explore the roles of CNR1 and CNR2 in HPV-positive HNSCC cell lines and animal models. Cannabinoid downstream pathways involved were determined by Western blotting and analyzed in a primary HPV HNSCC cohort with single-sample gene set enrichment analysis (ssGSEA) and the OncoGenome Positioning System (Onco-GPS).ResultsIn TCGA cohort, the expression of CNR1 and CNR2 was elevated in HPV-positive HNSCC compared with HPV-negative HNSCC, and knockdown of CNR1/CNR2 expression inhibited proliferation in HPV-positive HNSCC cell lines. Specific CNR1 and CNR2 activation as well as nonselective cannabinoid receptor activation in cell lines and animal models promoted cell growth, migration, and inhibited apoptosis through p38 MAPK pathway activation. CNR1/CNR2 antagonists suppressed cell proliferation and migration and induced apoptosis. Using whole-genome expression analysis in a primary HPV HNSCC cohort, we identified specific p38 MAPK pathway activation signature in tumors from HPV HNSCC patients with objective measurement of concurrent cannabinoid exposure.ConclusionsCannabinoids can promote progression of HPV-positive HNSCC through p38 MAPK pathway activation.
- Published
- 2020
20. Macrophage Syk–PI3Kγ Inhibits Antitumor Immunity: SRX3207, a Novel Dual Syk–PI3K Inhibitory Chemotype Relieves Tumor Immunosuppression
- Author
-
Joshi, Shweta, Liu, Kevin X, Zulcic, Muamera, Singh, Alok R, Skola, Dylan, Glass, Christopher K, Sanders, P Dominick, Sharabi, Andrew B, Pham, Timothy V, Tamayo, Pablo, Shiang, Daniel, Dinh, Huy Q, Hedrick, Catherine C, Morales, Guillermo A, Garlich, Joseph R, and Durden, Donald L
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Genetics ,Cancer ,Animals ,Apoptosis ,Carcinoma ,Lewis Lung ,Cell Proliferation ,Class Ib Phosphatidylinositol 3-Kinase ,Colonic Neoplasms ,Cytokines ,Humans ,Immune Tolerance ,Immunosuppression Therapy ,Macrophages ,Melanoma ,Experimental ,Mice ,Mice ,Inbred C57BL ,NF-kappa B ,Phosphoinositide-3 Kinase Inhibitors ,Signal Transduction ,Syk Kinase ,Tumor Cells ,Cultured ,Xenograft Model Antitumor Assays ,Pharmacology and Pharmaceutical Sciences ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Macrophages (MΦ) play a critical role in tumor growth, immunosuppression, and inhibition of adaptive immune responses in cancer. Hence, targeting signaling pathways in MΦs that promote tumor immunosuppression will provide therapeutic benefit. PI3Kγ has been recently established by our group and others as a novel immuno-oncology target. Herein, we report that an MΦ Syk-PI3K axis drives polarization of immunosuppressive MΦs that establish an immunosuppressive tumor microenvironment in in vivo syngeneic tumor models. Genetic or pharmacologic inhibition of Syk and/or PI3Kγ in MΦs promotes a proinflammatory MΦ phenotype, restores CD8+ T-cell activity, destabilizes HIF under hypoxia, and stimulates an antitumor immune response. Assay for transposase-accessible Chromatin using Sequencing (ATAC-seq) analyses on the bone marrow-derived macrophages (BMDM) show that inhibition of Syk kinase promotes activation and binding of NF-κB motif in SykMC-KO BMDMs, thus stimulating immunostimulatory transcriptional programming in MΦs to suppress tumor growth. Finally, we have developed in silico the "first-in-class" dual Syk/PI3K inhibitor, SRX3207, for the combinatorial inhibition of Syk and PI3K in one small molecule. This chemotype demonstrates efficacy in multiple tumor models and represents a novel combinatorial approach to activate antitumor immunity.
- Published
- 2020
21. The multikinase inhibitor RXDX-105 is effective against neuroblastoma in vitro and in vivo.
- Author
-
Flynn, Sean M, Lesperance, Jacqueline, Macias, Andrew, Phanhthilath, Nikki, Paul, Megan Rose, Kim, Jong Wook, Tamayo, Pablo, and Zage, Peter E
- Subjects
BRAF ,CEP-32496 ,RET ,RXDX-105 ,neuroblastoma ,Oncology and Carcinogenesis - Abstract
Neuroblastoma is the most common extracranial solid tumor of childhood and accounts for 15% of all pediatric cancer-related deaths. New therapies are needed to improve outcomes for children with high-risk and relapsed tumors. Inhibitors of the RET kinase and the RAS-MAPK pathway have previously been shown to be effective against neuroblastoma, suggesting that combined inhibition may have increased efficacy. RXDX-105 is a small molecule inhibitor of multiple kinases, including the RET and BRAF kinases. We found that treatment of neuroblastoma cells with RXDX-105 resulted in a significant decrease in cell viability and proliferation in vitro and in tumor growth and tumor vascularity in vivo. Treatment with RXDX-105 inhibited RET phosphorylation and phosphorylation of the MEK and ERK kinases in neuroblastoma cells and xenograft tumors, and RXDX-105 treatment induced both apoptosis and cell cycle arrest. RXDX-105 also showed enhanced efficacy in combination with 13-cis-retinoic acid, which is currently a component of maintenance therapy for children with high-risk neuroblastoma. Our results demonstrate that RXDX-105 shows promise as a novel therapeutic agent for children with high-risk and relapsed neuroblastoma.
- Published
- 2019
22. DNA methylation identifies genetically and prognostically distinct subtypes of myelodysplastic syndromes
- Author
-
Reilly, Brian, Tanaka, Tiffany N, Diep, Dinh, Yeerna, Huwate, Tamayo, Pablo, Zhang, Kun, and Bejar, Rafael
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Rare Diseases ,Genetics ,Human Genome ,Hematology ,Aged ,DNA Methylation ,Humans ,Myelodysplastic Syndromes ,Prognosis ,Cardiovascular medicine and haematology - Abstract
Recurrent mutations implicate several epigenetic regulators in the early molecular pathobiology of myelodysplastic syndromes (MDS). We hypothesized that MDS subtypes defined by DNA methylation (DNAm) patterns could enhance our understanding of MDS disease biology and identify patients with convergent epigenetic profiles. Bisulfite padlock probe sequencing was used to measure DNAm of ∼500 000 unique cytosine guanine dinucleotides covering 140 749 nonoverlapping regulatory regions across the genome in bone marrow DNA samples from 141 patients with MDS. Application of a nonnegative matrix factorization (NMF)-based decomposition of DNAm profiles identified 5 consensus clusters described by 5 NMF components as the most stable grouping solution. Each of the 5 NMF components identified by this approach correlated with specific genetic abnormalities and categorized patients into 5 distinct methylation clusters, each largely defined by a single NMF component. Methylation clusters displayed unique differentially methylated regulatory loci enriched for active and bivalent promoters and enhancers. Two clusters were enriched for samples with complex karyotypes, although only one had an increased number of TP53 mutations. Each of the 3 most frequently mutated splicing factors, SF3B1, U2AF1, and SRSF2, was enriched in different clusters. Mutations of ASXL1, EZH2, and RUNX1 were coenriched in the SRSF2-containing cluster. In multivariate analysis, methylation cluster membership remained independently associated with overall survival. Targeted DNAm profiles identify clinically relevant subtypes of MDS not otherwise distinguished by mutations or clinical features. Patients with diverse genetic lesions can converge on common DNAm states with shared pathogenic mechanisms and clinical outcomes.
- Published
- 2019
23. Metformin Inhibits Progression of Head and Neck Squamous Cell Carcinoma by Acting Directly on Carcinoma-Initiating Cells
- Author
-
Wu, Xingyu, Yeerna, Huwate, Goto, Yusuke, Ando, Toshinori, Wu, Victoria H, Zhang, Xuefeng, Wang, Zhiyong, Amornphimoltham, Panomwat, Murphy, Anne N, Tamayo, Pablo, Chen, Qianming, Lippman, Scott M, and Gutkind, J Silvio
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Biological Sciences ,Dental/Oral and Craniofacial Disease ,Clinical Research ,Genetics ,Digestive Diseases ,Sexually Transmitted Infections ,Rare Diseases ,Infectious Diseases ,Orphan Drug ,Cancer ,Prevention ,Aetiology ,2.1 Biological and endogenous factors ,Good Health and Well Being ,AMP-Activated Protein Kinases ,Animals ,Antineoplastic Agents ,Cell Line ,Tumor ,Cell Survival ,Electron Transport Complex I ,Epithelial-Mesenchymal Transition ,Female ,Gene Expression Regulation ,Neoplastic ,Head and Neck Neoplasms ,Humans ,Metformin ,Mice ,Nude ,Pyruvates ,Saccharomyces cerevisiae Proteins ,Squamous Cell Carcinoma of Head and Neck ,TOR Serine-Threonine Kinases ,Xenograft Model Antitumor Assays ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Metformin may reduce the progression of head and neck squamous cell carcinoma (HNSCC); however, whether metformin acts by altering the host metabolism or targets cancer-initiating cells remains poorly understood. This gap in knowledge has prevented the stratification of patient populations who are most likely to benefit from metformin treatment. Here, we explored whether metformin acts directly on HNSCC cells to inhibit aberrant cell growth. To investigate the tumor cell autonomous effects of metformin, we engineered representative HPV- and HPV+ HNSCC cells harboring typical genetic alternations to express the yeast mitochondrial NADH dehydrogenase (NDI1) protein, which is insensitive to metformin. NDI1 expression rescued the inhibitory effects of metformin on mitochondrial complex I, abolished the ability of metformin to activate AMP-activated protein kinase, and inhibited mTOR signaling both in vitro and in vivo, and was sufficient to render metformin ineffective to prevent HNSCC tumor growth. This experimental system provided an opportunity to identify metformin-regulated transcriptional programs linked to cancer cell growth inhibition in the tumor microenvironment. Remarkably, computational analysis of the metformin-induced transcriptome revealed that metformin downregulated gene expression signatures associated with cancer stemness and epithelial-mesenchymal transition, concomitant with increased expression of squamous differentiation genes. These findings support that metformin may act directly on cancer-initiating cells to prevent their progression to HNSCC, which may inform the selection of patients at risk of developing HNSCC in future early-stage clinical trials. SIGNIFICANCE: Metformin's ability to directly target HNSCC-initiating cells instead of exerting cancer preventive activity based solely on its systemic effects may inform the selection of patients in future precision prevention trials.
- Published
- 2019
24. Modelling bistable tumour population dynamics to design effective treatment strategies
- Author
-
Akhmetzhanov, Andrei R, Kim, Jong Wook, Sullivan, Ryan, Beckman, Robert A, Tamayo, Pablo, and Yeang, Chen-Hsiang
- Subjects
Biological Sciences ,Mathematical Sciences ,Cancer ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Drug Resistance ,Neoplasm ,Humans ,Melanoma ,Models ,Biological ,Mutation ,Protein Kinase Inhibitors ,Proto-Oncogene Proteins B-raf ,Tumour management ,Targeted treatments ,Drug resistance ,State transitions ,Pathway regulation ,Oncogenic states ,Information and Computing Sciences ,Evolutionary Biology ,Biological sciences ,Mathematical sciences - Abstract
Despite recent advances in targeted drugs and immunotherapy, cancer remains "the emperor of all maladies" due to almost inevitable emergence of resistance. Drug resistance is thought to be driven by genetic alterations and/or dynamic plasticity that deregulate pathway activities and regulatory programs of a highly heterogeneous tumour. In this study, we propose a modelling framework to simulate population dynamics of heterogeneous tumour cells with reversible drug resistance. Drug sensitivity of a tumour cell is determined by its internal states, which are demarcated by coordinated activities of multiple interconnected oncogenic pathways. Transitions between cellular states depend on the effects of targeted drugs and regulatory relations between the pathways. Under this framework, we build a simple model to capture drug resistance characteristics of BRAF-mutant melanoma, where two cell states are determined by two mutually inhibitory - main and alternative - pathways. We assume that cells with an activated main pathway are proliferative yet sensitive to the BRAF inhibitor, and cells with an activated alternative pathway are quiescent but resistant to the drug. We describe a dynamical process of tumour growth under various drug regimens using the explicit solutions of mean-field equations. Based on these solutions, we compare efficacy of three treatment strategies from simulated data: static treatments with continuous and constant dosages, periodic treatments with regular intermittent active phases and drug holidays, and treatments derived from optimal control theory (OCT). Periodic treatments outperform static treatments with a considerable margin, while treatments based on OCT outperform the best periodic treatment. Our results provide insights regarding optimal cancer treatment modalities for heterogeneous tumours, and may guide the development of optimal therapeutic strategies to circumvent plastic drug resistance. They can also be used to evaluate the efficacy of suboptimal treatments that may account for side effects of the treatment and the cost of its application.
- Published
- 2019
25. Illuminating the Onco-GPCRome: Novel G protein–coupled receptor-driven oncocrine networks and targets for cancer immunotherapy
- Author
-
Wu, Victoria, Yeerna, Huwate, Nohata, Nijiro, Chiou, Joshua, Harismendy, Olivier, Raimondi, Francesco, Inoue, Asuka, Russell, Robert B, Tamayo, Pablo, and Gutkind, J Silvio
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Immunology ,Oncology and Carcinogenesis ,Cancer ,Aetiology ,Underpinning research ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Animals ,DNA Copy Number Variations ,Humans ,Immunotherapy ,Mutation ,Neoplasms ,Receptors ,G-Protein-Coupled ,Signal Transduction ,G protein-coupled receptor ,G protein ,signal transduction ,cancer ,immunotherapy ,drug repurposing ,oncocrine signaling ,precision therapies ,G protein–coupled receptor ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
G protein-coupled receptors (GPCRs) are the largest gene family of cell membrane-associated molecules mediating signal transmission, and their involvement in key physiological functions is well-established. The ability of GPCRs to regulate a vast array of fundamental biological processes, such as cardiovascular functions, immune responses, hormone and enzyme release from endocrine and exocrine glands, neurotransmission, and sensory perception (e.g. vision, odor, and taste), is largely due to the diversity of these receptors and the layers of their downstream signaling circuits. Dysregulated expression and aberrant functions of GPCRs have been linked to some of the most prevalent human diseases, which renders GPCRs one of the top targets for pharmaceutical drug development. However, the study of the role of GPCRs in tumor biology has only just begun to make headway. Recent studies have shown that GPCRs can contribute to the many facets of tumorigenesis, including proliferation, survival, angiogenesis, invasion, metastasis, therapy resistance, and immune evasion. Indeed, GPCRs are widely dysregulated in cancer and yet are underexploited in oncology. We present here a comprehensive analysis of GPCR gene expression, copy number variation, and mutational signatures in 33 cancer types. We also highlight the emerging role of GPCRs as part of oncocrine networks promoting tumor growth, dissemination, and immune evasion, and we stress the potential benefits of targeting GPCRs and their signaling circuits in the new era of precision medicine and cancer immunotherapies.
- Published
- 2019
26. Publisher Correction: Chromatin dysregulation and DNA methylation at transcription start sites associated with transcriptional repression in cancers.
- Author
-
Ando, Mizuo, Saito, Yuki, Xu, Guorong, Bui, Nam Q, Medetgul-Ernar, Kate, Pu, Minya, Fisch, Kathleen, Ren, Shuling, Sakai, Akihiro, Fukusumi, Takahito, Liu, Chao, Haft, Sunny, Pang, John, Mark, Adam, Gaykalova, Daria A, Guo, Theresa, Favorov, Alexander V, Yegnasubramanian, Srinivasan, Fertig, Elana J, Ha, Patrick, Tamayo, Pablo, Yamasoba, Tatsuya, Ideker, Trey, Messer, Karen, and Califano, Joseph A
- Subjects
Genetics ,Cancer - Abstract
The original version of this Article contained an error in the author affiliations. Trey Ideker was incorrectly associated with 'Department of Medicine (Oncology), Stanford University School of Medicine, 875 Blake Wilbur Dr, Palo Alto, CA 94304, USA.' This has now been corrected in both the PDF and HTML versions of the Article.
- Published
- 2019
27. Chromatin dysregulation and DNA methylation at transcription start sites associated with transcriptional repression in cancers.
- Author
-
Ando, Mizuo, Saito, Yuki, Xu, Guorong, Bui, Nam Q, Medetgul-Ernar, Kate, Pu, Minya, Fisch, Kathleen, Ren, Shuling, Sakai, Akihiro, Fukusumi, Takahito, Liu, Chao, Haft, Sunny, Pang, John, Mark, Adam, Gaykalova, Daria A, Guo, Theresa, Favorov, Alexander V, Yegnasubramanian, Srinivasan, Fertig, Elana J, Ha, Patrick, Tamayo, Pablo, Yamasoba, Tatsuya, Ideker, Trey, Messer, Karen, and Califano, Joseph A
- Subjects
Cell Line ,Tumor ,Chromatin ,Humans ,Neoplasms ,Proto-Oncogene Proteins c-myc ,Histones ,Signal Transduction ,DNA Methylation ,Gene Expression Regulation ,Neoplastic ,Gene Silencing ,Mutation ,Transcription Initiation Site ,E1A-Associated p300 Protein ,CREB-Binding Protein ,Datasets as Topic ,Human Genome ,Cancer ,Genetics ,Rare Diseases ,2.1 Biological and endogenous factors - Abstract
Although promoter-associated CpG islands have been established as targets of DNA methylation changes in cancer, previous studies suggest that epigenetic dysregulation outside the promoter region may be more closely associated with transcriptional changes. Here we examine DNA methylation, chromatin marks, and transcriptional alterations to define the relationship between transcriptional modulation and spatial changes in chromatin structure. Using human papillomavirus-related oropharyngeal carcinoma as a model, we show aberrant enrichment of repressive H3K9me3 at the transcriptional start site (TSS) with methylation-associated, tumor-specific gene silencing. Further analysis identifies a hypermethylated subtype which shows a functional convergence on MYC targets and association with CREBBP/EP300 mutation. The tumor-specific shift to transcriptional repression associated with DNA methylation at TSSs was confirmed in multiple tumor types. Our data may show a common underlying epigenetic dysregulation in cancer associated with broad enrichment of repressive chromatin marks and aberrant DNA hypermethylation at TSSs in combination with MYC network activation.
- Published
- 2019
28. APOBEC-related mutagenesis and neo-peptide hydrophobicity: implications for response to immunotherapy
- Author
-
Boichard, Amélie, Pham, Timothy V, Yeerna, Huwate, Goodman, Aaron, Tamayo, Pablo, Lippman, Scott, Frampton, Garrett M, Tsigelny, Igor F, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Clinical Research ,Vaccine Related ,Immunization ,Cancer ,Genetics ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Aetiology ,2.1 Biological and endogenous factors ,Mutagenesis ,APOBEC ,neo-epitopes ,immunotherapy ,cancer ,Oncology and carcinogenesis - Abstract
Tumor-associated neo-antigens are mutated peptides that allow the immune system to recognize the affected cell as foreign. Cells carrying excessive mutation load often develop mechanisms of tolerance. PD-L1/PD-1 checkpoint immunotherapy is a highly promising approach to overcome these protective signals and induce tumor shrinkage. Yet, the nature of the neo-antigens driving those beneficial responses remains unclear. Here, we show that APOBEC-related mutagenesis - a mechanism at the crossroads between anti-viral immunity and endogenous nucleic acid editing - increases neo-peptide hydrophobicity (a feature of immunogenicity), as demonstrated by in silico computation and in the TCGA pan-cancer cohort, where APOBEC-related mutagenesis was also strongly associated with immune marker expression. Moreover, APOBEC-related mutagenesis correlated with immunotherapy response in a cohort of 99 patients with diverse cancers, and this correlation was independent of the tumor mutation burden (TMB). Combining APOBEC-related mutagenesis estimate and TMB resulted in greater predictive ability than either parameter alone. Based on these results, further investigation of APOBEC-related mutagenesis as a marker of response to anti-cancer checkpoint blockade is warranted.
- Published
- 2019
29. A Platform of Synthetic Lethal Gene Interaction Networks Reveals that the GNAQ Uveal Melanoma Oncogene Controls the Hippo Pathway through FAK
- Author
-
Feng, Xiaodong, Arang, Nadia, Rigiracciolo, Damiano Cosimo, Lee, Joo Sang, Yeerna, Huwate, Wang, Zhiyong, Lubrano, Simone, Kishore, Ayush, Pachter, Jonathan A, König, Gabriele M, Maggiolini, Marcello, Kostenis, Evi, Schlaepfer, David D, Tamayo, Pablo, Chen, Qianming, Ruppin, Eytan, and Gutkind, J Silvio
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Biotechnology ,Stem Cell Research ,Cancer ,Rare Diseases ,Stem Cell Research - Nonembryonic - Non-Human ,Genetics ,Eye Disease and Disorders of Vision ,2.1 Biological and endogenous factors ,Aetiology ,Animals ,Cell Line ,Tumor ,Cell Proliferation ,Computational Biology ,Focal Adhesion Kinase 1 ,GTP-Binding Protein alpha Subunits ,Gq-G11 ,Genes ,Lethal ,Hippo Signaling Pathway ,Humans ,Melanoma ,Mice ,Neoplasm Transplantation ,Phosphorylation ,Prognosis ,Protein Serine-Threonine Kinases ,Signal Transduction ,Survival Analysis ,Uveal Neoplasms ,FAK ,G protein ,GNA11 ,GNAQ ,Hippo ,MOB1 ,PTK2 ,YAP ,signal transduction ,uveal melanoma ,Protein-Serine-Threonine Kinases ,Neurosciences ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Activating mutations in GNAQ/GNA11, encoding Gαq G proteins, are initiating oncogenic events in uveal melanoma (UM). However, there are no effective therapies for UM. Using an integrated bioinformatics pipeline, we found that PTK2, encoding focal adhesion kinase (FAK), represents a candidate synthetic lethal gene with GNAQ activation. We show that Gαq activates FAK through TRIO-RhoA non-canonical Gαq-signaling, and genetic ablation or pharmacological inhibition of FAK inhibits UM growth. Analysis of the FAK-regulated transcriptome demonstrated that GNAQ stimulates YAP through FAK. Dissection of the underlying mechanism revealed that FAK regulates YAP by tyrosine phosphorylation of MOB1, inhibiting core Hippo signaling. Our findings establish FAK as a potential therapeutic target for UM and other Gαq-driven pathophysiologies that involve unrestrained YAP function.
- Published
- 2019
30. Comprehensive Genomic Profiling Reveals Diverse but Actionable Molecular Portfolios across Hematologic Malignancies: Implications for Next Generation Clinical Trials
- Author
-
Galanina, Natalie, Bejar, Rafael, Choi, Michael, Goodman, Aaron, Wieduwilt, Matthew, Mulroney, Carolyn, Kim, Lisa, Yeerna, Huwate, Tamayo, Pablo, Vergilio, Jo-Anne, Mughal, Tariq I, Miller, Vincent, Jamieson, Catriona, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Pediatric Cancer ,Cancer ,Hematology ,Human Genome ,Lymphoma ,Rare Diseases ,Pediatric ,Genetics ,Biotechnology ,Good Health and Well Being ,next generation sequencing ,lymphoid malignancies ,myeloid malignancies ,precision medicine ,Oncology and carcinogenesis - Abstract
Background: The translation of genomic discoveries to the clinic is the cornerstone of precision medicine. However, incorporating next generation sequencing (NGS) of hematologic malignancies into clinical management remains limited. Methods: We describe 235 patients who underwent integrated NGS profiling (406 genes) and analyze the alterations and their potential actionability. Results: Overall, 227 patients (96.5%) had adequate tissue. Most common diagnoses included myelodysplastic syndrome (22.9%), chronic lymphocytic leukemia (17.2%), non-Hodgkin lymphoma (13.2%), acute myeloid leukemia (11%), myeloproliferative neoplasm (9.2%), acute lymphoblastic leukemia (8.8%), and multiple myeloma (7.5%). Most patients (N = 197/227 (87%)) harbored ≥1 genomic alteration(s); 170/227 (75%), ≥1 potentially actionable alteration(s) targetable by an FDA-approved (mostly off-label) or an investigational agent. Altogether, 546 distinct alterations were seen, most commonly involving TP53 (10.8%), TET2 (4.6%), and DNMT3A (4.2%). The median tumor mutational burden (TMB) was low (1.7 alterations/megabase); 12% of patients had intermediate or high TMB (higher TMB correlates with favorable response to anti-PD1/PDL1 inhibition in solid tumors). In conclusion, 96.5% of patients with hematologic malignancies have adequate tissue for comprehensive genomic profiling. Most patients had unique molecular signatures, and 75% had alterations that may be pharmacologically tractable with gene- or immune-targeted agents.
- Published
- 2019
31. The Prognostic Significance of Homologous Recombination Repair Pathway Alterations in Metastatic Hormone Sensitive Prostate Cancer
- Author
-
Lee, Aaron M., Saidian, Ava, Shaya, Justin, Nonato, Taylor, Cabal, Angelo, Randall, J. Michael, Millard, Frederick, Stewart, Tyler, Rose, Brent, Tamayo, Pablo, and McKay, Rana R.
- Published
- 2022
- Full Text
- View/download PDF
32. Comprehensive Genomic Profiling Reveals Diverse but Actionable Molecular Portfolios across Hematologic Malignancies: Implications for Next Generation Clinical Trials.
- Author
-
Galanina, Natalie, Bejar, Rafael, Choi, Michael, Goodman, Aaron, Wieduwilt, Matthew, Mulroney, Carolyn, Kim, Lisa, Yeerna, Huwate, Tamayo, Pablo, Vergilio, Jo-Anne, Mughal, Tariq I, Miller, Vincent, Jamieson, Catriona, and Kurzrock, Razelle
- Subjects
lymphoid malignancies ,myeloid malignancies ,next generation sequencing ,precision medicine ,Rare Diseases ,Human Genome ,Pediatric ,Biotechnology ,Genetics ,Hematology ,Cancer ,Pediatric Cancer ,Lymphoma ,Oncology and Carcinogenesis - Abstract
Background: The translation of genomic discoveries to the clinic is the cornerstone of precision medicine. However, incorporating next generation sequencing (NGS) of hematologic malignancies into clinical management remains limited. Methods: We describe 235 patients who underwent integrated NGS profiling (406 genes) and analyze the alterations and their potential actionability. Results: Overall, 227 patients (96.5%) had adequate tissue. Most common diagnoses included myelodysplastic syndrome (22.9%), chronic lymphocytic leukemia (17.2%), non-Hodgkin lymphoma (13.2%), acute myeloid leukemia (11%), myeloproliferative neoplasm (9.2%), acute lymphoblastic leukemia (8.8%), and multiple myeloma (7.5%). Most patients (N = 197/227 (87%)) harbored ≥1 genomic alteration(s); 170/227 (75%), ≥1 potentially actionable alteration(s) targetable by an FDA-approved (mostly off-label) or an investigational agent. Altogether, 546 distinct alterations were seen, most commonly involving TP53 (10.8%), TET2 (4.6%), and DNMT3A (4.2%). The median tumor mutational burden (TMB) was low (1.7 alterations/megabase); 12% of patients had intermediate or high TMB (higher TMB correlates with favorable response to anti-PD1/PDL1 inhibition in solid tumors). In conclusion, 96.5% of patients with hematologic malignancies have adequate tissue for comprehensive genomic profiling. Most patients had unique molecular signatures, and 75% had alterations that may be pharmacologically tractable with gene- or immune-targeted agents.
- Published
- 2018
33. Visualizing and Interpreting Single-Cell Gene Expression Datasets with Similarity Weighted Nonnegative Embedding.
- Author
-
Wu, Yan, Tamayo, Pablo, and Zhang, Kun
- Subjects
Humans ,Cluster Analysis ,Gene Expression Profiling ,Genomics ,Gene Expression ,Algorithms ,Computer Graphics ,Databases ,Nucleic Acid ,Single-Cell Analysis ,Transcriptome ,ontology embedding ,single-cell analysis ,transcriptome ,visualization ,Genetics ,1.1 Normal biological development and functioning ,Biochemistry and Cell Biology - Abstract
High-throughput single-cell gene expression profiling has enabled the definition of new cell types and developmental trajectories. Visualizing these datasets is crucial to biological interpretation, and a popular method is t-stochastic neighbor embedding (t-SNE), which visualizes local patterns well but distorts global structure, such as distances between clusters. We developed similarity weighted nonnegative embedding (SWNE), which enhances interpretation of datasets by embedding the genes and factors that separate cell states on the visualization alongside the cells and maintains fidelity when visualizing local and global structure for both developmental trajectories and discrete cell types. SWNE uses nonnegative matrix factorization to decompose the gene expression matrix into biologically relevant factors; embeds the cells, genes, and factors in a 2D visualization; and uses a similarity matrix to smooth the embeddings. We demonstrate SWNE on single-cell RNA-seq data from hematopoietic progenitors and human brain cells. SWNE is available as an R package at github.com/yanwu2014/swne.
- Published
- 2018
34. Overcoming Resistance to Dual Innate Immune and MEK Inhibition Downstream of KRAS
- Author
-
Kitajima, Shunsuke, Asahina, Hajime, Chen, Ting, Guo, Sujuan, Quiceno, Laura Gutierrez, Cavanaugh, Jillian D, Merlino, Ashley A, Tange, Shoichiro, Terai, Hideki, Kim, Jong Wook, Wang, Xiaoen, Zhou, Shan, Xu, Man, Wang, Stephen, Zhu, Zehua, Thai, Tran C, Takahashi, Chiaki, Wang, Yujin, Neve, Richard, Stinson, Susanna, Tamayo, Pablo, Watanabe, Hideo, Kirschmeier, Paul T, Wong, Kwok-Kin, and Barbie, David A
- Subjects
Cancer ,Lung ,Lung Cancer ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,AMP-Activated Protein Kinase Kinases ,AMP-Activated Protein Kinases ,Adaptor Proteins ,Signal Transducing ,Animals ,Antineoplastic Agents ,Immunological ,Carcinoma ,Non-Small-Cell Lung ,Cell Line ,Tumor ,Disease Models ,Animal ,Drug Resistance ,Neoplasm ,HEK293 Cells ,Humans ,Immunity ,Innate ,Insulin-Like Growth Factor I ,Lung Neoplasms ,Mice ,Mice ,Transgenic ,Mitogen-Activated Protein Kinase Kinases ,Phosphoproteins ,Protein Kinase Inhibitors ,Protein Serine-Threonine Kinases ,Proto-Oncogene Proteins p21(ras) ,Transcription Factors ,YAP-Signaling Proteins ,BET inhibitor ,IL-6 ,KRAS ,MEK inhibitor ,STK11/LKB1 ,TBK1 inhibitor ,TP53 ,YAP1 signaling ,innate immune signaling ,non-small-cell lung cancer ,Neurosciences ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
Despite extensive efforts, oncogenic KRAS remains resistant to targeted therapy. Combined downstream RAL-TBK1 and MEK inhibition induces only transient lung tumor shrinkage in KRAS-driven genetically engineered mouse models (GEMMs). Using the sensitive KRAS;LKB1 (KL) mutant background, we identify YAP1 upregulation and a therapy-induced secretome as mediators of acquired resistance. This program is reversible, associated with H3K27 promoter acetylation, and suppressed by BET inhibition, resensitizing resistant KL cells to TBK1/MEK inhibition. Constitutive YAP1 signaling promotes intrinsic resistance in KRAS;TP53 (KP) mutant lung cancer. Intermittent treatment with the BET inhibitor JQ1 thus overcomes resistance to combined pathway inhibition in KL and KP GEMMs. Using potent and selective TBK1 and BET inhibitors we further develop an effective therapeutic strategy with potential translatability to the clinic.
- Published
- 2018
35. Proteomics, Post-translational Modifications, and Integrative Analyses Reveal Molecular Heterogeneity within Medulloblastoma Subgroups
- Author
-
Archer, Tenley C, Ehrenberger, Tobias, Mundt, Filip, Gold, Maxwell P, Krug, Karsten, Mah, Clarence K, Mahoney, Elizabeth L, Daniel, Colin J, LeNail, Alexander, Ramamoorthy, Divya, Mertins, Philipp, Mani, DR, Zhang, Hailei, Gillette, Michael A, Clauser, Karl, Noble, Michael, Tang, Lauren C, Pierre-François, Jessica, Silterra, Jacob, Jensen, James, Tamayo, Pablo, Korshunov, Andrey, Pfister, Stefan M, Kool, Marcel, Northcott, Paul A, Sears, Rosalie C, Lipton, Jonathan O, Carr, Steven A, Mesirov, Jill P, Pomeroy, Scott L, and Fraenkel, Ernest
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Neurosciences ,Pediatric ,Pediatric Research Initiative ,Cancer ,Pediatric Cancer ,Genetics ,Rare Diseases ,Brain Cancer ,Brain Disorders ,Biotechnology ,Adolescent ,Adult ,Biomarkers ,Tumor ,Brain Neoplasms ,Cell Line ,Tumor ,Child ,Child ,Preschool ,DNA Methylation ,DNA-Activated Protein Kinase ,Female ,Gene Expression Profiling ,Hedgehog Proteins ,Humans ,Infant ,Male ,Medulloblastoma ,Nuclear Proteins ,Protein Processing ,Post-Translational ,Proteome ,Proteomics ,Proto-Oncogene Proteins c-myc ,Sequence Analysis ,RNA ,Young Adult ,MYC ,NU-7441 ,SHH ,mass spectrometry ,medulloblastoma ,multi-omics ,network integration ,phospho-proteomics ,proteo-genomics ,radio sensitization ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
There is a pressing need to identify therapeutic targets in tumors with low mutation rates such as the malignant pediatric brain tumor medulloblastoma. To address this challenge, we quantitatively profiled global proteomes and phospho-proteomes of 45 medulloblastoma samples. Integrated analyses revealed that tumors with similar RNA expression vary extensively at the post-transcriptional and post-translational levels. We identified distinct pathways associated with two subsets of SHH tumors, and found post-translational modifications of MYC that are associated with poor outcomes in group 3 tumors. We found kinases associated with subtypes and showed that inhibiting PRKDC sensitizes MYC-driven cells to radiation. Our study shows that proteomics enables a more comprehensive, functional readout, providing a foundation for future therapeutic strategies.
- Published
- 2018
36. Tumor innate immunity primed by specific interferon-stimulated endogenous retroviruses
- Author
-
Cañadas, Israel, Thummalapalli, Rohit, Kim, Jong Wook, Kitajima, Shunsuke, Jenkins, Russell William, Christensen, Camilla Laulund, Campisi, Marco, Kuang, Yanan, Zhang, Yanxi, Gjini, Evisa, Zhang, Gao, Tian, Tian, Sen, Debattama Rai, Miao, Diana, Imamura, Yu, Thai, Tran, Piel, Brandon, Terai, Hideki, Aref, Amir Reza, Hagan, Timothy, Koyama, Shohei, Watanabe, Masayuki, Baba, Hideo, Adeni, Anika Elise, Lydon, Christine Anne, Tamayo, Pablo, Wei, Zhi, Herlyn, Meenhard, Barbie, Thanh Uyen, Uppaluri, Ravindra, Sholl, Lynnette Marie, Sicinska, Ewa, Sands, Jacob, Rodig, Scott, Wong, Kwok Kin, Paweletz, Cloud Peter, Watanabe, Hideo, and Barbie, David Allen
- Subjects
Genetics ,Cancer ,Vaccine Related ,Immunization ,2.1 Biological and endogenous factors ,Aetiology ,Inflammatory and immune system ,Animals ,Cell Line ,Tumor ,Endogenous Retroviruses ,Gene Expression Regulation ,Neoplastic ,Humans ,Immunity ,Innate ,Interferons ,Mice ,Nude ,Neoplasms ,RNA ,Antisense ,Medical and Health Sciences ,Immunology - Abstract
Mesenchymal tumor subpopulations secrete pro-tumorigenic cytokines and promote treatment resistance1-4. This phenomenon has been implicated in chemorefractory small cell lung cancer and resistance to targeted therapies5-8, but remains incompletely defined. Here, we identify a subclass of endogenous retroviruses (ERVs) that engages innate immune signaling in these cells. Stimulated 3 prime antisense retroviral coding sequences (SPARCS) are oriented inversely in 3' untranslated regions of specific genes enriched for regulation by STAT1 and EZH2. Derepression of these loci results in double-stranded RNA generation following IFN-γ exposure due to bi-directional transcription from the STAT1-activated gene promoter and the 5' long terminal repeat of the antisense ERV. Engagement of MAVS and STING activates downstream TBK1, IRF3, and STAT1 signaling, sustaining a positive feedback loop. SPARCS induction in human tumors is tightly associated with major histocompatibility complex class 1 expression, mesenchymal markers, and downregulation of chromatin modifying enzymes, including EZH2. Analysis of cell lines with high inducible SPARCS expression reveals strong association with an AXL/MET-positive mesenchymal cell state. While SPARCS-high tumors are immune infiltrated, they also exhibit multiple features of an immune-suppressed microenviroment. Together, these data unveil a subclass of ERVs whose derepression triggers pathologic innate immune signaling in cancer, with important implications for cancer immunotherapy.
- Published
- 2018
37. Assembly and activation of the Hippo signalome by FAT1 tumor suppressor.
- Author
-
Martin, Daniel, Degese, Maria S, Vitale-Cross, Lynn, Iglesias-Bartolome, Ramiro, Valera, Juan Luis Callejas, Wang, Zhiyong, Feng, Xiaodong, Yeerna, Huwate, Vadmal, Vachan, Moroishi, Toshiro, Thorne, Rick F, Zaida, Moraima, Siegele, Bradford, Cheong, Sok C, Molinolo, Alfredo A, Samuels, Yardena, Tamayo, Pablo, Guan, Kun Liang, Lippman, Scott M, Lyons, J Guy, and Gutkind, J Silvio
- Subjects
Humans ,Head and Neck Neoplasms ,Protein-Serine-Threonine Kinases ,Hepatocyte Growth Factor ,Adaptor Proteins ,Signal Transducing ,Cadherins ,Proto-Oncogene Proteins ,Phosphoproteins ,Transcription Factors ,Signal Transduction ,Gene Knockdown Techniques ,HEK293 Cells ,Squamous Cell Carcinoma of Head and Neck ,Cancer ,Genetics ,Biotechnology ,Dental/Oral and Craniofacial Disease ,Rare Diseases ,2.1 Biological and endogenous factors - Abstract
Dysregulation of the Hippo signaling pathway and the consequent YAP1 activation is a frequent event in human malignancies, yet the underlying molecular mechanisms are still poorly understood. A pancancer analysis of core Hippo kinases and their candidate regulating molecules revealed few alterations in the canonical Hippo pathway, but very frequent genetic alterations in the FAT family of atypical cadherins. By focusing on head and neck squamous cell carcinoma (HNSCC), which displays frequent FAT1 alterations (29.8%), we provide evidence that FAT1 functional loss results in YAP1 activation. Mechanistically, we found that FAT1 assembles a multimeric Hippo signaling complex (signalome), resulting in activation of core Hippo kinases by TAOKs and consequent YAP1 inactivation. We also show that unrestrained YAP1 acts as an oncogenic driver in HNSCC, and that targeting YAP1 may represent an attractive precision therapeutic option for cancers harboring genomic alterations in the FAT1 tumor suppressor genes.
- Published
- 2018
38. Genome-scale analysis identifies paralog lethality as a vulnerability of chromosome 1p loss in cancer
- Author
-
Viswanathan, Srinivas R, Nogueira, Marina F, Buss, Colin G, Krill-Burger, John M, Wawer, Mathias J, Malolepsza, Edyta, Berger, Ashton C, Choi, Peter S, Shih, Juliann, Taylor, Alison M, Tanenbaum, Benjamin, Pedamallu, Chandra Sekhar, Cherniack, Andrew D, Tamayo, Pablo, Strathdee, Craig A, Lage, Kasper, Carr, Steven A, Schenone, Monica, Bhatia, Sangeeta N, Vazquez, Francisca, Tsherniak, Aviad, Hahn, William C, and Meyerson, Matthew
- Subjects
Cancer ,Pediatric Research Initiative ,Lung Cancer ,Human Genome ,Lung ,Biotechnology ,Genetics ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,Cell Line ,Tumor ,Cell Nucleus ,Chromosomes ,Human ,Pair 1 ,Exons ,Female ,Gene Deletion ,HEK293 Cells ,Humans ,Karyopherins ,Mice ,Mice ,Nude ,Neoplasms ,Nuclear Proteins ,RNA Splicing ,RNA ,Small Interfering ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Functional redundancy shared by paralog genes may afford protection against genetic perturbations, but it can also result in genetic vulnerabilities due to mutual interdependency1-5. Here, we surveyed genome-scale short hairpin RNA and CRISPR screening data on hundreds of cancer cell lines and identified MAGOH and MAGOHB, core members of the splicing-dependent exon junction complex, as top-ranked paralog dependencies6-8. MAGOHB is the top gene dependency in cells with hemizygous MAGOH deletion, a pervasive genetic event that frequently occurs due to chromosome 1p loss. Inhibition of MAGOHB in a MAGOH-deleted context compromises viability by globally perturbing alternative splicing and RNA surveillance. Dependency on IPO13, an importin-β receptor that mediates nuclear import of the MAGOH/B-Y14 heterodimer9, is highly correlated with dependency on both MAGOH and MAGOHB. Both MAGOHB and IPO13 represent dependencies in murine xenografts with hemizygous MAGOH deletion. Our results identify MAGOH and MAGOHB as reciprocal paralog dependencies across cancer types and suggest a rationale for targeting the MAGOHB-IPO13 axis in cancers with chromosome 1p deletion.
- Published
- 2018
39. Systematic Evaluation of Molecular Networks for Discovery of Disease Genes
- Author
-
Huang, Justin K, Carlin, Daniel E, Yu, Michael Ku, Zhang, Wei, Kreisberg, Jason F, Tamayo, Pablo, and Ideker, Trey
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Human Genome ,Biotechnology ,Algorithms ,Computational Biology ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Genome ,Human ,Humans ,genome-wide disease association ,molecular networks ,network analysis ,network propagation ,Biochemistry and Cell Biology ,Biochemistry and cell biology - Abstract
Gene networks are rapidly growing in size and number, raising the question of which networks are most appropriate for particular applications. Here, we evaluate 21 human genome-wide interaction networks for their ability to recover 446 disease gene sets identified through literature curation, gene expression profiling, or genome-wide association studies. While all networks have some ability to recover disease genes, we observe a wide range of performance with STRING, ConsensusPathDB, and GIANT networks having the best performance overall. A general tendency is that performance scales with network size, suggesting that new interaction discovery currently outweighs the detrimental effects of false positives. Correcting for size, we find that the DIP network provides the highest efficiency (value per interaction). Based on these results, we create a parsimonious composite network with both high efficiency and performance. This work provides a benchmark for selection of molecular networks in human disease research.
- Published
- 2018
40. Profiling ranked list enrichment scoring in sparse data elucidates algorithmic tradeoffs
- Author
-
Wenzel, Alexander T., primary, Jun, John, additional, Tamayo, Pablo, additional, and Mesirov, Jill P., additional
- Published
- 2024
- Full Text
- View/download PDF
41. Bending behavior of ecological fiber reinforced concrete
- Author
-
Gonzalez, Laura, primary, Sainz-Aja, Jose, additional, Tamayo, Pablo, additional, and Thomas, Carlos, additional
- Published
- 2022
- Full Text
- View/download PDF
42. List of contributors
- Author
-
Ademola, Daniel, primary, Adeniyi, Adedayo, additional, Adesina, Adeyemi, additional, Aghajanian, Ali, additional, Agrela, Francisco, additional, Aluko, Oluwatobi G., additional, Awoyera, Paul O., additional, Babalola, Olusola E., additional, Bamigboye, Gideon Olukunle, additional, Cabrera, M., additional, Carrizo, Leandro, additional, Cimentada, A., additional, de Azevedo, Afonso R.G., additional, de Brito, J., additional, Del Angel, Gilberto Garcia, additional, Díaz-López, J.L., additional, Frías, M., additional, García-González, Julia, additional, Gonzalez, Laura, additional, Guerra-Romero, M. Ignacio, additional, Juan-Valdés, Andrés, additional, Kareem, Mutiu, additional, Kim, Jeonghyun, additional, Kolawole, John Temitope, additional, Kırgız, Mehmet Serkan, additional, Lopez-Alonso, M., additional, Mansouri, Iman, additional, Martínez-García, Rebeca, additional, Marvila, Markssuel T., additional, Medina, G., additional, Medina Martinez, Cesar, additional, Morán-del Pozo, Julia Ma, additional, Odetoyan, Abimbola, additional, Ohenoja, Katja, additional, Olalusi, Oladimeji B., additional, Orogbade, Blessing, additional, Perumal, Priyadharshini, additional, Rosales, J., additional, Sadowski, Łukasz, additional, Sáez del Bosque, I.F., additional, Sainz-Aja, Jose, additional, Sánchez de Rojas, M.I., additional, Silva, P.R., additional, Silva, R.V., additional, Sosa, Eva, additional, Tamayo, Pablo, additional, Tezcan, Jale, additional, Thomas, Carlos, additional, Ugwu, E.I., additional, Villagrán-Zaccardi, Yury, additional, Xu, Jinjun, additional, and Zega, Claudio, additional
- Published
- 2022
- Full Text
- View/download PDF
43. TORC1/2 kinase inhibition depletes glutathione and synergizes with carboplatin to suppress the growth of MYC-driven medulloblastoma
- Author
-
Maynard, Rachael E., Poore, Brad, Hanaford, Allison R., Pham, Khoa, James, Madison, Alt, Jesse, Park, Youngran, Slusher, Barbara S., Tamayo, Pablo, Mesirov, Jill, Archer, Tenley C., Pomeroy, Scott L., Eberhart, Charles G., and Raabe, Eric H.
- Published
- 2021
- Full Text
- View/download PDF
44. Identification of novel prostate cancer drivers using RegNetDriver: a framework for integration of genetic and epigenetic alterations with tissue-specific regulatory network
- Author
-
Dhingra, Priyanka, Martinez-Fundichely, Alexander, Berger, Adeline, Huang, Franklin W, Forbes, Andre Neil, Liu, Eric Minwei, Liu, Deli, Sboner, Andrea, Tamayo, Pablo, Rickman, David S, Rubin, Mark A, and Khurana, Ekta
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Prostate Cancer ,Urologic Diseases ,Human Genome ,Cancer ,Biotechnology ,2.1 Biological and endogenous factors ,Aetiology ,Algorithms ,Binding Sites ,Carcinogenesis ,Chromosome Mapping ,Cyclic AMP Response Element-Binding Protein ,DNA Methylation ,Deoxyribonuclease I ,Epigenesis ,Genetic ,Gene Expression Regulation ,Neoplastic ,Gene Regulatory Networks ,Humans ,Male ,Nerve Tissue Proteins ,Octamer Transcription Factor-2 ,Organ Specificity ,Polymorphism ,Single Nucleotide ,Promoter Regions ,Genetic ,Prostatic Neoplasms ,Protein Binding ,Protein Interaction Mapping ,Repressor Proteins ,Tissue-specific regulatory network ,Cancer drivers ,Single nucleotide variants ,Structural variants ,DNA methylation ,Prostate cancer ,Environmental Sciences ,Information and Computing Sciences ,Bioinformatics - Abstract
We report a novel computational method, RegNetDriver, to identify tumorigenic drivers using the combined effects of coding and non-coding single nucleotide variants, structural variants, and DNA methylation changes in the DNase I hypersensitivity based regulatory network. Integration of multi-omics data from 521 prostate tumor samples indicated a stronger regulatory impact of structural variants, as they affect more transcription factor hubs in the tissue-specific network. Moreover, crosstalk between transcription factor hub expression modulated by structural variants and methylation levels likely leads to the differential expression of target genes. We report known prostate tumor regulatory drivers and nominate novel transcription factors (ERF, CREB3L1, and POU2F2), which are supported by functional validation.
- Published
- 2017
45. Exome Sequencing of African-American Prostate Cancer Reveals Loss-of-Function ERF Mutations
- Author
-
Huang, Franklin W, Mosquera, Juan Miguel, Garofalo, Andrea, Oh, Coyin, Baco, Maria, Amin-Mansour, Ali, Rabasha, Bokang, Bahl, Samira, Mullane, Stephanie A, Robinson, Brian D, Aldubayan, Saud, Khani, Francesca, Karir, Beerinder, Kim, Eejung, Chimene-Weiss, Jeremy, Hofree, Matan, Romanel, Alessandro, Osborne, Joseph R, Kim, Jong Wook, Azabdaftari, Gissou, Woloszynska-Read, Anna, Sfanos, Karen, De Marzo, Angelo M, Demichelis, Francesca, Gabriel, Stacey, Van Allen, Eliezer M, Mesirov, Jill, Tamayo, Pablo, Rubin, Mark A, Powell, Isaac J, and Garraway, Levi A
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Prostate Cancer ,Biotechnology ,Aging ,Cancer ,Genetics ,Human Genome ,Urologic Diseases ,Aetiology ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Black or African American ,Animals ,Cell Line ,Tumor ,Class I Phosphatidylinositol 3-Kinases ,Exome ,Humans ,Male ,Mice ,Mutation ,PTEN Phosphohydrolase ,Prostatic Neoplasms ,Repressor Proteins ,Exome Sequencing ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
African-American men have the highest incidence of and mortality from prostate cancer. Whether a biological basis exists for this disparity remains unclear. Exome sequencing (n = 102) and targeted validation (n = 90) of localized primary hormone-naïve prostate cancer in African-American men identified several gene mutations not previously observed in this context, including recurrent loss-of-function mutations in ERF, an ETS transcriptional repressor, in 5% of cases. Analysis of existing prostate cancer cohorts revealed ERF deletions in 3% of primary prostate cancers and mutations or deletions in ERF in 3% to 5% of lethal castration-resistant prostate cancers. Knockdown of ERF confers increased anchorage-independent growth and generates a gene expression signature associated with oncogenic ETS activation and androgen signaling. Together, these results suggest that ERF is a prostate cancer tumor-suppressor gene. More generally, our findings support the application of systematic cancer genomic characterization in settings of broader ancestral diversity to enhance discovery and, eventually, therapeutic applications.Significance: Systematic genomic sequencing of prostate cancer in African-American men revealed new insights into prostate cancer, including the identification of ERF as a prostate cancer gene; somatic copy-number alteration differences; and uncommon PIK3CA and PTEN alterations. This study highlights the importance of inclusion of underrepresented minorities in cancer sequencing studies. Cancer Discov; 7(9); 973-83. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 920.
- Published
- 2017
46. Decomposing Oncogenic Transcriptional Signatures to Generate Maps of Divergent Cellular States
- Author
-
Kim, Jong Wook, Abudayyeh, Omar O, Yeerna, Huwate, Yeang, Chen-Hsiang, Stewart, Michelle, Jenkins, Russell W, Kitajima, Shunsuke, Konieczkowski, David J, Medetgul-Ernar, Kate, Cavazos, Taylor, Mah, Clarence, Ting, Stephanie, Van Allen, Eliezer M, Cohen, Ofir, Mcdermott, John, Damato, Emily, Aguirre, Andrew J, Liang, Jonathan, Liberzon, Arthur, Alexe, Gabriella, Doench, John, Ghandi, Mahmoud, Vazquez, Francisca, Weir, Barbara A, Tsherniak, Aviad, Subramanian, Aravind, Meneses-Cime, Karina, Park, Jason, Clemons, Paul, Garraway, Levi A, Thomas, David, Boehm, Jesse S, Barbie, David A, Hahn, William C, Mesirov, Jill P, and Tamayo, Pablo
- Subjects
Biological Sciences ,Genetics ,Biotechnology ,Human Genome ,Cancer ,2.1 Biological and endogenous factors ,Aetiology ,Good Health and Well Being ,Biomarkers ,Tumor ,Cell Line ,Tumor ,Gene Expression Profiling ,Gene Expression Regulation ,Neoplastic ,Genes ,ras ,Genome ,Humans ,MAP Kinase Signaling System ,Neoplasms ,Precision Medicine ,Bayesian nomogram ,cellular states ,drug sensitivity ,genetic dependency ,inferential model ,matrix factorization ,oncoGPS ,oncogenic pathway ,precision medicine ,transcriptional signatures ,Biochemistry and Cell Biology ,Biochemistry and cell biology - Abstract
The systematic sequencing of the cancer genome has led to the identification of numerous genetic alterations in cancer. However, a deeper understanding of the functional consequences of these alterations is necessary to guide appropriate therapeutic strategies. Here, we describe Onco-GPS (OncoGenic Positioning System), a data-driven analysis framework to organize individual tumor samples with shared oncogenic alterations onto a reference map defined by their underlying cellular states. We applied the methodology to the RAS pathway and identified nine distinct components that reflect transcriptional activities downstream of RAS and defined several functional states associated with patterns of transcriptional component activation that associates with genomic hallmarks and response to genetic and pharmacological perturbations. These results show that the Onco-GPS is an effective approach to explore the complex landscape of oncogenic cellular states across cancers, and an analytic framework to summarize knowledge, establish relationships, and generate more effective disease models for research or as part of individualized precision medicine paradigms.
- Published
- 2017
47. The GenePattern Notebook Environment
- Author
-
Reich, Michael, Tabor, Thorin, Liefeld, Ted, Thorvaldsdóttir, Helga, Hill, Barbara, Tamayo, Pablo, and Mesirov, Jill P
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Biotechnology ,Human Genome ,Genetics ,Computational Biology ,Gene Expression Profiling ,Genomics ,Software ,User-Computer Interface ,bioinformatics ,integrative genomics ,open science ,reproducible research ,Biochemistry and Cell Biology ,Biochemistry and cell biology - Abstract
Interactive analysis notebook environments promise to streamline genomics research through interleaving text, multimedia, and executable code into unified, sharable, reproducible "research narratives." However, current notebook systems require programming knowledge, limiting their wider adoption by the research community. We have developed the GenePattern Notebook environment (http://www.genepattern-notebook.org), to our knowledge the first system to integrate the dynamic capabilities of notebook systems with an investigator-focused, easy-to-use interface that provides access to hundreds of genomic tools without the need to write code.
- Published
- 2017
48. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway
- Author
-
Viswanathan, Vasanthi S, Ryan, Matthew J, Dhruv, Harshil D, Gill, Shubhroz, Eichhoff, Ossia M, Seashore-Ludlow, Brinton, Kaffenberger, Samuel D, Eaton, John K, Shimada, Kenichi, Aguirre, Andrew J, Viswanathan, Srinivas R, Chattopadhyay, Shrikanta, Tamayo, Pablo, Yang, Wan Seok, Rees, Matthew G, Chen, Sixun, Boskovic, Zarko V, Javaid, Sarah, Huang, Cherrie, Wu, Xiaoyun, Tseng, Yuen-Yi, Roider, Elisabeth M, Gao, Dong, Cleary, James M, Wolpin, Brian M, Mesirov, Jill P, Haber, Daniel A, Engelman, Jeffrey A, Boehm, Jesse S, Kotz, Joanne D, Hon, Cindy S, Chen, Yu, Hahn, William C, Levesque, Mitchell P, Doench, John G, Berens, Michael E, Shamji, Alykhan F, Clemons, Paul A, Stockwell, Brent R, and Schreiber, Stuart L
- Subjects
Cancer ,Cadherins ,Cell Death ,Cell Line ,Tumor ,Cell Lineage ,Cell Transdifferentiation ,Drug Resistance ,Neoplasm ,Epithelial-Mesenchymal Transition ,Glutathione Peroxidase ,Humans ,Iron ,Lipid Peroxidation ,Lipid Peroxides ,Male ,Melanoma ,Mesoderm ,Neoplasms ,Phospholipid Hydroperoxide Glutathione Peroxidase ,Prostatic Neoplasms ,Proteomics ,Proto-Oncogene Proteins B-raf ,Reproducibility of Results ,Zinc Finger E-box-Binding Homeobox 1 ,General Science & Technology - Abstract
Plasticity of the cell state has been proposed to drive resistance to multiple classes of cancer therapies, thereby limiting their effectiveness. A high-mesenchymal cell state observed in human tumours and cancer cell lines has been associated with resistance to multiple treatment modalities across diverse cancer lineages, but the mechanistic underpinning for this state has remained incompletely understood. Here we molecularly characterize this therapy-resistant high-mesenchymal cell state in human cancer cell lines and organoids and show that it depends on a druggable lipid-peroxidase pathway that protects against ferroptosis, a non-apoptotic form of cell death induced by the build-up of toxic lipid peroxides. We show that this cell state is characterized by activity of enzymes that promote the synthesis of polyunsaturated lipids. These lipids are the substrates for lipid peroxidation by lipoxygenase enzymes. This lipid metabolism creates a dependency on pathways converging on the phospholipid glutathione peroxidase (GPX4), a selenocysteine-containing enzyme that dissipates lipid peroxides and thereby prevents the iron-mediated reactions of peroxides that induce ferroptotic cell death. Dependency on GPX4 was found to exist across diverse therapy-resistant states characterized by high expression of ZEB1, including epithelial-mesenchymal transition in epithelial-derived carcinomas, TGFβ-mediated therapy-resistance in melanoma, treatment-induced neuroendocrine transdifferentiation in prostate cancer, and sarcomas, which are fixed in a mesenchymal state owing to their cells of origin. We identify vulnerability to ferroptic cell death induced by inhibition of a lipid peroxidase pathway as a feature of therapy-resistant cancer cells across diverse mesenchymal cell-state contexts.
- Published
- 2017
49. OTX2 Activity at Distal Regulatory Elements Shapes the Chromatin Landscape of Group 3 Medulloblastoma
- Author
-
Boulay, Gaylor, Awad, Mary E, Riggi, Nicolo, Archer, Tenley C, Iyer, Sowmya, Boonseng, Wannaporn E, Rossetti, Nikki E, Naigles, Beverly, Rengarajan, Shruthi, Volorio, Angela, Kim, James C, Mesirov, Jill P, Tamayo, Pablo, Pomeroy, Scott L, Aryee, Martin J, and Rivera, Miguel N
- Subjects
Rare Diseases ,Pediatric Cancer ,Genetics ,Neurosciences ,Biotechnology ,Brain Disorders ,Brain Cancer ,Pediatric ,Cancer ,Human Genome ,Basic Helix-Loop-Helix Transcription Factors ,Cell Line ,Tumor ,Cell Survival ,Cerebellar Neoplasms ,Chromatin ,Enhancer Elements ,Genetic ,Gene Expression Regulation ,Neoplastic ,Humans ,Medulloblastoma ,Mesenchymal Stem Cells ,NIMA-Related Kinases ,Otx Transcription Factors ,Oncology and Carcinogenesis - Abstract
Medulloblastoma is the most frequent malignant pediatric brain tumor and is divided into at least four subgroups known as WNT, SHH, Group 3, and Group 4. Here, we characterized gene regulation mechanisms in the most aggressive subtype, Group 3 tumors, through genome-wide chromatin and expression profiling. Our results show that most active distal sites in these tumors are occupied by the transcription factor OTX2. Highly active OTX2-bound enhancers are often arranged as clusters of adjacent peaks and are also bound by the transcription factor NEUROD1. These sites are responsive to OTX2 and NEUROD1 knockdown and could also be generated de novo upon ectopic OTX2 expression in primary cells, showing that OTX2 cooperates with NEUROD1 and plays a major role in maintaining and possibly establishing regulatory elements as a pioneer factor. Among OTX2 target genes, we identified the kinase NEK2, whose knockdown and pharmacologic inhibition decreased cell viability. Our studies thus show that OTX2 controls the regulatory landscape of Group 3 medulloblastoma through cooperative activity at enhancer elements and contributes to the expression of critical target genes.Significance: The gene regulation mechanisms that drive medulloblastoma are not well understood. Using chromatin profiling, we find that the transcription factor OTX2 acts as a pioneer factor and, in cooperation with NEUROD1, controls the Group 3 medulloblastoma active enhancer landscape. OTX2 itself or its target genes, including the mitotic kinase NEK2, represent attractive targets for future therapies. Cancer Discov; 7(3); 288-301. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 235.
- Published
- 2017
50. An expanded universe of cancer targets
- Author
-
Dancik, Vlado, Gill, Shubhroz, Hua, Bruce, Sharifnia, Tanaz, Viswanathan, Vasanthi, Zou, Yilong, Dela Cruz, Filemon, Kung, Andrew, Stockwell, Brent, Boehm, Jesse, Dempster, Josh, Manguso, Robert, Vazquez, Francisca, Cooper, Lee A.D., Du, Yuhong, Ivanov, Andrey, Lonial, Sagar, Moreno, Carlos S., Niu, Qiankun, Owonikoko, Taofeek, Ramalingam, Suresh, Reyna, Matthew, Zhou, Wei, Grandori, Carla, Shmulevich, Ilya, Swisher, Elizabeth, Cai, Jitong, Chan, Issac S., Dunworth, Matthew, Ge, Yuchen, Georgess, Dan, Grasset, Eloïse M., Henriet, Elodie, Knútsdóttir, Hildur, Lerner, Michael G., Padmanaban, Veena, Perrone, Matthew C., Suhail, Yasir, Tsehay, Yohannes, Warrier, Manisha, Morrow, Quin, Nechiporuk, Tamilla, Long, Nicola, Saultz, Jennifer, Kaempf, Andy, Minnier, Jessica, Tognon, Cristina E., Kurtz, Stephen E., Agarwal, Anupriya, Brown, Jordana, Watanabe-Smith, Kevin, Vu, Tania Q., Jacob, Thomas, Yan, Yunqi, Robinson, Bridget, Lind, Evan F., Kosaka, Yoko, Demir, Emek, Estabrook, Joseph, Grzadkowski, Michael, Nikolova, Olga, Chen, Ken, Deneen, Ben, Liang, Han, Bassik, Michael C., Bhattacharya, Asmita, Brennan, Kevin, Curtis, Christina, Gevaert, Olivier, Ji, Hanlee P., Karlsson, Kasper A.J., Karagyozova, Kremena, Lo, Yuan-Hung, Liu, Katherine, Nakano, Michitaka, Sathe, Anuja, Smith, Amber R., Spees, Kaitlyn, Wong, Wing Hing, Yuki, Kanako, Hangauer, Matt, Kaufman, Dan S., Balmain, Allan, Bollam, Saumya R., Chen, Wei-Ching, Fan, QiWen, Kersten, Kelly, Krummel, Matthew, Li, Yun Rose, Menard, Marie, Nasholm, Nicole, Schmidt, Christin, Serwas, Nina K., Yoda, Hiroyuki, Ashworth, Alan, Bandyopadhyay, Sourav, Bivona, Trevor, Eades, Gabriel, Oberlin, Stefan, Tay, Neil, Wang, Yuhao, Weissman, Jonathan, Hahn, William C., Bader, Joel S., Braun, Theodore P., Califano, Andrea, Clemons, Paul A., Druker, Brian J., Ewald, Andrew J., Fu, Haian, Jagu, Subhashini, Kemp, Christopher J., Kim, William, Kuo, Calvin J., McManus, Michael T., B. Mills, Gordon, Mo, Xiulei, Sahni, Nidhi, Schreiber, Stuart L., Talamas, Jessica A., Tamayo, Pablo, Tyner, Jeffrey W., Wagner, Bridget K., Weiss, William A., and Gerhard, Daniela S.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.