24 results on '"Papillon-Cavanagh, Simon"'
Search Results
2. STK11 and KEAP1 mutations as prognostic biomarkers in an observational real-world lung adenocarcinoma cohort
- Author
-
Papillon-Cavanagh, Simon, Doshi, Parul, Dobrin, Radu, Szustakowski, Joseph, and Walsh, Alice M.
- Published
- 2020
- Full Text
- View/download PDF
3. Pan-cancer analysis of the effect of biopsy site on tumor mutational burden observations
- Author
-
Papillon-Cavanagh, Simon, Hopkins, Julia F., Ramkissoon, Shakti H., Albacker, Lee A., and Walsh, Alice M.
- Published
- 2021
- Full Text
- View/download PDF
4. The molecular landscape of ETMR at diagnosis and relapse
- Author
-
Lambo, Sander, Gröbner, Susanne N., Rausch, Tobias, Waszak, Sebastian M., Schmidt, Christin, Gorthi, Aparna, Romero, July Carolina, Mauermann, Monika, Brabetz, Sebastian, Krausert, Sonja, Buchhalter, Ivo, Koster, Jan, Zwijnenburg, Danny A., Sill, Martin, Hübner, Jens-Martin, Mack, Norman, Schwalm, Benjamin, Ryzhova, Marina, Hovestadt, Volker, Papillon-Cavanagh, Simon, Chan, Jennifer A., Landgraf, Pablo, Ho, Ben, Milde, Till, Witt, Olaf, Ecker, Jonas, Sahm, Felix, Sumerauer, David, Ellison, David W., Orr, Brent A., Darabi, Anna, Haberler, Christine, Figarella-Branger, Dominique, Wesseling, Pieter, Schittenhelm, Jens, Remke, Marc, Taylor, Michael D., Gil-da-Costa, Maria J., Łastowska, Maria, Grajkowska, Wiesława, Hasselblatt, Martin, Hauser, Peter, Pietsch, Torsten, Uro-Coste, Emmanuelle, Bourdeaut, Franck, Masliah-Planchon, Julien, Rigau, Valérie, Alexandrescu, Sanda, Wolf, Stephan, Li, Xiao-Nan, Schüller, Ulrich, Snuderl, Matija, Karajannis, Matthias A., Giangaspero, Felice, Jabado, Nada, von Deimling, Andreas, Jones, David T. W., Korbel, Jan O., von Hoff, Katja, Lichter, Peter, Huang, Annie, Bishop, Alexander J. R., Pfister, Stefan M., Korshunov, Andrey, and Kool, Marcel
- Published
- 2019
- Full Text
- View/download PDF
5. The histone mark H3K36me2 recruits DNMT3A and shapes the intergenic DNA methylation landscape
- Author
-
Weinberg, Daniel N., Papillon-Cavanagh, Simon, Chen, Haifen, Yue, Yuan, Chen, Xiao, Rajagopalan, Kartik N., and Horth, Cynthia
- Subjects
Post-translational modification -- Analysis ,DNA -- Analysis ,Methylation -- Analysis ,Histones -- Analysis ,Methyltransferases -- Analysis ,Environmental issues ,Science and technology ,Zoology and wildlife conservation - Abstract
Enzymes that catalyse CpG methylation in DNA, including the DNA methyltransferases 1 (DNMT1), 3A (DNMT3A) and 3B (DNMT3B), are indispensable for mammalian tissue development and homeostasis.sup.1-4. They are also implicated in human developmental disorders and cancers.sup.5-8, supporting the critical role of DNA methylation in the specification and maintenance of cell fate. Previous studies have suggested that post-translational modifications of histones are involved in specifying patterns of DNA methyltransferase localization and DNA methylation at promoters and actively transcribed gene bodies.sup.9-11. However, the mechanisms that control the establishment and maintenance of intergenic DNA methylation remain poorly understood. Tatton-Brown-Rahman syndrome (TBRS) is a childhood overgrowth disorder that is defined by germline mutations in DNMT3A. TBRS shares clinical features with Sotos syndrome (which is caused by haploinsufficiency of NSD1, a histone methyltransferase that catalyses the dimethylation of histone H3 at K36 (H3K36me2).sup.8,12,13), which suggests that there is a mechanistic link between these two diseases. Here we report that NSD1-mediated H3K36me2 is required for the recruitment of DNMT3A and maintenance of DNA methylation at intergenic regions. Genome-wide analysis shows that the binding and activity of DNMT3A colocalize with H3K36me2 at non-coding regions of euchromatin. Genetic ablation of Nsd1 and its paralogue Nsd2 in mouse cells results in a redistribution of DNMT3A to H3K36me3-modified gene bodies and a reduction in the methylation of intergenic DNA. Blood samples from patients with Sotos syndrome and NSD1-mutant tumours also exhibit hypomethylation of intergenic DNA. The PWWP domain of DNMT3A shows dual recognition of H3K36me2 and H3K36me3 in vitro, with a higher binding affinity towards H3K36me2 that is abrogated by TBRS-derived missense mutations. Together, our study reveals a trans-chromatin regulatory pathway that connects aberrant intergenic CpG methylation to human neoplastic and developmental overgrowth. H3K36me2 targets DNMT3A to intergenic regions and this process, together with H3K36me3-mediated recruitment of DNMT3B, has a key role in establishing and maintaining genomic DNA methylation landscapes., Author(s): Daniel N. Weinberg [sup.1] , Simon Papillon-Cavanagh [sup.2] , Haifen Chen [sup.2] , Yuan Yue [sup.3] , Xiao Chen [sup.4] [sup.5] , Kartik N. Rajagopalan [sup.6] , Cynthia Horth [...]
- Published
- 2019
- Full Text
- View/download PDF
6. Histone H3K36 mutations promote sarcomagenesis through altered histone methylation landscape
- Author
-
Lu, Chao, Jain, Siddhant U., Hoelper, Dominik, Bechet, Denise, Molden, Rosalynn C., Ran, Leili, Murphy, Devan, Venneti, Sriram, Hameed, Meera, Pawel, Bruce R., Wunder, Jay S., Dickson, Brendan C., Lundgren, Stefan M., Jani, Krupa S., De Jay, Nicolas, Papillon-Cavanagh, Simon, Andrulis, Irene L., Sawyer, Sarah L., Grynspan, David, Turcotte, Robert E., Nadaf, Javad, Fahiminiyah, Somayyeh, Muir, Tom W., Majewski, Jacek, Thompson, Craig B., Chi, Ping, Garcia, Benjamin A., Allis, C. David, Jabado, Nada, and Lewis, Peter W.
- Published
- 2016
7. H3K27M induces defective chromatin spread of PRC2-mediated repressive H3K27me2/me3 and is essential for glioma tumorigenesis
- Author
-
Harutyunyan, Ashot S., Krug, Brian, Chen, Haifen, Papillon-Cavanagh, Simon, Zeinieh, Michele, De Jay, Nicolas, Deshmukh, Shriya, Chen, Carol C. L., Belle, Jad, Mikael, Leonie G., Marchione, Dylan M., Li, Rui, Nikbakht, Hamid, Hu, Bo, Cagnone, Gael, Cheung, Warren A., Mohammadnia, Abdulshakour, Bechet, Denise, Faury, Damien, McConechy, Melissa K, Pathania, Manav, Jain, Siddhant U., Ellezam, Benjamin, Weil, Alexander G., Montpetit, Alexandre, Salomoni, Paolo, Pastinen, Tomi, Lu, Chao, Lewis, Peter W., Garcia, Benjamin A., Kleinman, Claudia L., Jabado, Nada, and Majewski, Jacek
- Published
- 2019
- Full Text
- View/download PDF
8. Epigenetic dysregulation: a novel pathway of oncogenesis in pediatric brain tumors
- Author
-
Fontebasso, Adam M., Gayden, Tenzin, Nikbakht, Hamid, Neirinck, Michael, Papillon-Cavanagh, Simon, Majewski, Jacek, and Jabado, Nada
- Published
- 2014
- Full Text
- View/download PDF
9. A multi‐omic single cell sequencing approach to develop a CD8 T cell specific gene signature for anti‐PD1 response in solid tumors.
- Author
-
Kumar, Namit, Papillon‐Cavanagh, Simon, Tang, Hao, Wang, Shiliang, Stromko, Caitlyn, Ho, Ching‐Ping, Soni‐Sheth, Sonal, Vasquez‐Grinnell, Steven, Broz, Miranda L., Tenney, Daniel J., Wichroski, Michael J., Walsh, Alice M., Hu, Yanhua, and Benci, Joseph L.
- Subjects
T cells ,T cell receptors ,TUMOR-infiltrating immune cells ,CLINICAL trials ,IMMUNE checkpoint proteins ,TUMOR suppressor genes ,BRAF genes - Abstract
Immune checkpoint blockade (ICB) has led to durable clinical responses in multiple cancer types. However, biomarkers that identify which patients are most likely to respond to ICB are not well defined. Many putative biomarkers developed from a small number of samples often fail to maintain their predictive status in larger validation cohorts. We show across multiple human malignancies and syngeneic murine tumor models that neither pretreatment T cell receptor (TCR) clonality nor changes in clonality after ICB correlate with response. Dissection of tumor infiltrating lymphocytes pre‐ and post‐ICB by paired single‐cell RNA sequencing and single‐cell TCR sequencing reveals conserved and distinct transcriptomic features in expanded TCR clonotypes between anti‐PD1 responder and nonresponder murine tumor models. Overall, our results indicate a productive anti‐tumor response is agnostic of TCR clonal expansion. Further, we used single‐cell transcriptomics to develop a CD8+ T cell specific gene signature for a productive anti‐tumor response and show the response signature to be associated with overall survival (OS) on nivolumab monotherapy in CheckMate‐067, a phase 3 clinical trial in metastatic melanoma. These results highlight the value of leveraging single‐cell assays to dissect heterogeneous tumor and immune subsets and define cell‐type specific transcriptomic biomarkers of ICB response. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
10. Mutation in The Nuclear-Encoded Mitochondrial Isoleucyl–tRNA Synthetase IARS2 in Patients with Cataracts, Growth Hormone Deficiency with Short Stature, Partial Sensorineural Deafness, and Peripheral Neuropathy or with Leigh Syndrome
- Author
-
Schwartzentruber, Jeremy, Buhas, Daniela, Majewski, Jacek, Sasarman, Florin, Papillon-Cavanagh, Simon, Thiffaut, Isabelle, Sheldon, Katherine M., Massicotte, Christine, Patry, Lysanne, Simon, Mariella, Zare, Amir S., McKernan, Kevin J., Consortium, FORGE Canada, Michaud, Jacques, Boles, Richard G., Deal, Cheri L., Desilets, Valerie, Shoubridge, Eric A., and Samuels, Mark E.
- Published
- 2014
- Full Text
- View/download PDF
11. mRMRe: an R package for parallelized mRMR ensemble feature selection
- Author
-
De Jay, Nicolas, Papillon-Cavanagh, Simon, Olsen, Catharina, El-Hachem, Nehme, Bontempi, Gianluca, and Haibe-Kains, Benjamin
- Published
- 2013
- Full Text
- View/download PDF
12. Comparison and validation of genomic predictors for anticancer drug sensitivity
- Author
-
Papillon-Cavanagh, Simon, De Jay, Nicolas, Hachem, Nehme, Olsen, Catharina, Bontempi, Gianluca, Aerts, Hugo J W L, Quackenbush, John, and Haibe-Kains, Benjamin
- Published
- 2013
- Full Text
- View/download PDF
13. Mutation in The Nuclear-Encoded Mitochondrial Isoleucyl-t RNA Synthetase IARS2 in Patients with Cataracts, Growth Hormone Deficiency with Short Stature, Partial Sensorineural Deafness, and Peripheral Neuropathy or with Leigh Syndrome.
- Author
-
Schwartzentruber, Jeremy, Buhas, Daniela, Majewski, Jacek, Sasarman, Florin, Papillon‐Cavanagh, Simon, Thiffaut, Isabelle, Sheldon, Katherine M., Massicotte, Christine, Patry, Lysanne, Simon, Mariella, Zare, Amir S., McKernan, Kevin J., Michaud, Jacques, Boles, Richard G., Deal, Cheri L., Desilets, Valerie, Shoubridge, Eric A., and Samuels, Mark E.
- Abstract
ABSTRACT Mutations in the nuclear-encoded mitochondrial aminoacyl-t RNA synthetases are associated with a range of clinical phenotypes. Here, we report a novel disorder in three adult patients with a phenotype including cataracts, short-stature secondary to growth hormone deficiency, sensorineural hearing deficit, peripheral sensory neuropathy, and skeletal dysplasia. Using SNP genotyping and whole-exome sequencing, we identified a single likely causal variant, a missense mutation in a conserved residue of the nuclear gene IARS2, encoding mitochondrial isoleucyl-t RNA synthetase. The mutation is homozygous in the affected patients, heterozygous in carriers, and absent in control chromosomes. IARS2 protein level was reduced in skin cells cultured from one of the patients, consistent with a pathogenic effect of the mutation. Compound heterozygous mutations in IARS2 were independently identified in a previously unreported patient with a more severe mitochondrial phenotype diagnosed as Leigh syndrome. This is the first report of clinical findings associated with IARS2 mutations. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
14. Recurrent somatic mutations in ACVR1 in pediatric midline high-grade astrocytoma.
- Author
-
Fontebasso, Adam M, Papillon-Cavanagh, Simon, Schwartzentruber, Jeremy, Nikbakht, Hamid, Gerges, Noha, Fiset, Pierre-Olivier, Bechet, Denise, Faury, Damien, De Jay, Nicolas, Ramkissoon, Lori A, Corcoran, Aoife, Jones, David T W, Sturm, Dominik, Johann, Pascal, Tomita, Tadanori, Goldman, Stewart, Nagib, Mahmoud, Bendel, Anne, Goumnerova, Liliana, and Bowers, Daniel C
- Subjects
- *
ASTROCYTOMAS , *HISTONES , *GENETIC mutation , *BONE morphogenetic proteins , *METHIONINE , *PONS Varolii - Abstract
Pediatric midline high-grade astrocytomas (mHGAs) are incurable with few treatment targets identified. Most tumors harbor mutations encoding p.Lys27Met in histone H3 variants. In 40 treatment-naive mHGAs, 39 analyzed by whole-exome sequencing, we find additional somatic mutations specific to tumor location. Gain-of-function mutations in ACVR1 occur in tumors of the pons in conjunction with histone H3.1 p.Lys27Met substitution, whereas FGFR1 mutations or fusions occur in thalamic tumors associated with histone H3.3 p.Lys27Met substitution. Hyperactivation of the bone morphogenetic protein (BMP)-ACVR1 developmental pathway in mHGAs harboring ACVR1 mutations led to increased levels of phosphorylated SMAD1, SMAD5 and SMAD8 and upregulation of BMP downstream early-response genes in tumor cells. Global DNA methylation profiles were significantly associated with the p.Lys27Met alteration, regardless of the mutant histone H3 variant and irrespective of tumor location, supporting the role of this substitution in driving the epigenetic phenotype. This work considerably expands the number of potential treatment targets and further justifies pretreatment biopsy in pediatric mHGA as a means to orient therapeutic efforts in this disease. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
15. Fusion of TTYH1 with the C19MC microRNA cluster drives expression of a brain-specific DNMT3B isoform in the embryonal brain tumor ETMR.
- Author
-
Kleinman, Claudia L, Gerges, Noha, Papillon-Cavanagh, Simon, Sin-Chan, Patrick, Pramatarova, Albena, Quang, Dong-Anh Khuong, Adoue, Véronique, Busche, Stephan, Caron, Maxime, Djambazian, Haig, Bemmo, Amandine, Fontebasso, Adam M, Spence, Tara, Schwartzentruber, Jeremy, Albrecht, Steffen, Hauser, Peter, Garami, Miklos, Klekner, Almos, Bognar, Laszlo, and Montes, Jose-Luis
- Subjects
MICRORNA genetics ,GENE expression ,EMBRYONAL tumors ,BRAIN tumors ,EPIGENETICS ,IMMUNOHISTOCHEMISTRY - Abstract
Embryonal tumors with multilayered rosettes (ETMRs) are rare, deadly pediatric brain tumors characterized by high-level amplification of the microRNA cluster C19MC. We performed integrated genetic and epigenetic analyses of 12 ETMR samples and identified, in all cases, C19MC fusions to TTYH1 driving expression of the microRNAs. ETMR tumors, cell lines and xenografts showed a specific DNA methylation pattern distinct from those of other tumors and normal tissues. We detected extreme overexpression of a previously uncharacterized isoform of DNMT3B originating at an alternative promoter that is active only in the first weeks of neural tube development. Transcriptional and immunohistochemical analyses suggest that C19MC-dependent DNMT3B deregulation is mediated by RBL2, a known repressor of DNMT3B. Transfection with individual C19MC microRNAs resulted in DNMT3B upregulation and RBL2 downregulation in cultured cells. Our data suggest a potential oncogenic re-engagement of an early developmental program in ETMR via epigenetic alteration mediated by an embryonic, brain-specific DNMT3B isoform. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
16. Spatial and temporal homogeneity of driver mutations in diffuse intrinsic pontine glioma.
- Author
-
Nikbakht, Hamid, Panditharatna, Eshini, Mikael, Leonie G., Li, Rui, Gayden, Tenzin, Osmond, Matthew, Ho, Cheng-Ying, Kambhampati, Madhuri, Hwang, Eugene I., Faury, Damien, Siu, Alan, Papillon-Cavanagh, Simon, Bechet, Denise, Ligon, Keith L., Ellezam, Benjamin, Ingram, Wendy J., Stinson, Caedyn, Moore, Andrew S., Warren, Katherine E., and Karamchandani, Jason
- Published
- 2016
- Full Text
- View/download PDF
17. Mutation in The Nuclear-Encoded Mitochondrial Isoleucyl-tRNA Synthetase IARS2 in Patients with Cataracts, Growth Hormone Deficiency with Short Stature, Partial Sensorineural Deafness, and Peripheral Neuropathy or with Leigh Syndrome.
- Author
-
Schwartzentruber, Jeremy, Buhas, Daniela, Majewski, Jacek, Sasarman, Florin, Papillon‐Cavanagh, Simon, Thiffault, Isabelle, Sheldon, Katherine M., Massicotte, Christine, Patry, Lysanne, Simon, Mariella, Zare, Amir S., McKernan, Kevin J., Consortium, FORGE Canada, Michaud, Jacques, Boles, Richard G., Deal, Cheri L., Desilets, Valerie, Shoubridge, Eric A., and Samuels, Mark E.
- Published
- 2015
- Full Text
- View/download PDF
18. Histone H3K36 mutations promote sarcomagenesis through altered histone methylation landscape.
- Author
-
Chao Lu, Jain, Siddhant U., Hoelper, Dominik, Bechet, Denise, Molden, Rosalynn C., Ran, Leili, Murphy, Devan, Venneti, Sriram, Hameed, Meera, Pawel, Bruce R., Wunder, Jay S., Dickson, Brendan C., Lundgren, Stefan M., Jani, Krupa S., De Jay, Nicolas, Papillon-Cavanagh, Simon, Andrulis, Irene L., Sawyer, Sarah L., Grynspan, David, and Turcotte, Robert E.
- Subjects
- *
HISTONES , *CHILDHOOD cancer , *GENETIC mutation , *CHROMATIN , *METHYLTRANSFERASES - Abstract
Several types of pediatric cancers reportedly contain high-frequency missense mutations in histone H3, yet the underlying oncogenic mechanism remains poorly characterized. Here we report that the H3 lysine 36-to-methionine (H3K36M) mutation impairs the differentiation of mesenchymal progenitor cells and generates undifferentiated sarcoma in vivo. H3K36M mutant nucleosomes inhibit the enzymatic activities of several H3K36 methyltransferases. Depleting H3K36 methyltransferases, or expressing an H3K36I mutant that similarly inhibits H3K36 methylation, is sufficient to phenocopy the H3K36M mutation. After the loss of H3K36 methylation, a genome-wide gain in H3K27 methylation leads to a redistribution of polycomb repressive complex 1 and de-repression of its target genes known to block mesenchymal differentiation. Our findings are mirrored in human undifferentiated sarcomas in which novel K36M/I mutations in H3.1 are identified. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
19. Mutations in C5ORF42 Cause Joubert Syndrome in the French Canadian Population
- Author
-
Srour, Myriam, Schwartzentruber, Jeremy, Hamdan, Fadi F., Ospina, Luis H., Patry, Lysanne, Labuda, Damian, Massicotte, Christine, Dobrzeniecka, Sylvia, Capo-Chichi, José-Mario, Papillon-Cavanagh, Simon, Samuels, Mark E., Boycott, Kym M., Shevell, Michael I., Laframboise, Rachel, Désilets, Valérie, Maranda, Bruno, Rouleau, Guy A., Majewski, Jacek, and Michaud, Jacques L.
- Subjects
- *
GENETIC mutation , *CEREBELLUM diseases , *FRENCH people , *CANADIANS , *RHOMBENCEPHALON , *DEVELOPMENTAL delay , *OCULAR hypotony , *APRAXIA , *GENETICS , *DISEASES - Abstract
Joubert syndrome (JBTS) is an autosomal-recessive disorder characterized by a distinctive mid-hindbrain malformation, developmental delay with hypotonia, ocular-motor apraxia, and breathing abnormalities. Although JBTS was first described more than 40 years ago in French Canadian siblings, the causal mutations have not yet been identified in this family nor in most French Canadian individuals subsequently described. We ascertained a cluster of 16 JBTS-affected individuals from 11 families living in the Lower St. Lawrence region. SNP genotyping excluded the presence of a common homozygous mutation that would explain the clustering of these individuals. Exome sequencing performed on 15 subjects showed that nine affected individuals from seven families (including the original JBTS family) carried rare compound-heterozygous mutations in C5ORF42. Two missense variants (c.4006C>T [p.Arg1336Trp] and c.4690G>A [p.Ala1564Thr]) and a splicing mutation (c.7400+1G>A), which causes exon skipping, were found in multiple subjects that were not known to be related, whereas three other truncating mutations (c.6407del [p.Pro2136Hisfs∗31], c.4804C>T [p.Arg1602∗], and c.7477C>T [p.Arg2493∗]) were identified in single individuals. None of the unaffected first-degree relatives were compound heterozygous for these mutations. Moreover, none of the six putative mutations were detected among 477 French Canadian controls. Our data suggest that mutations in C5ORF42 explain a large portion of French Canadian individuals with JBTS. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
20. Biomarker analysis from CheckMate 214: nivolumab plus ipilimumab versus sunitinib in renal cell carcinoma.
- Author
-
Motzer RJ, Choueiri TK, McDermott DF, Powles T, Vano YA, Gupta S, Yao J, Han C, Ammar R, Papillon-Cavanagh S, Saggi SS, McHenry MB, Ross-Macdonald P, and Wind-Rotolo M
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Clinical Trials, Phase III as Topic, Humans, Ipilimumab pharmacology, Ipilimumab therapeutic use, Nivolumab pharmacology, Nivolumab therapeutic use, Sunitinib therapeutic use, Tumor Microenvironment, Carcinoma, Renal Cell pathology, Kidney Neoplasms pathology
- Abstract
Background: The phase 3 CheckMate 214 trial demonstrated higher response rates and improved overall survival with nivolumab plus ipilimumab versus sunitinib in first-line therapy for advanced clear-cell renal cell carcinoma (RCC). An unmet need exists to identify patients with RCC who are most likely to benefit from treatment with nivolumab plus ipilimumab., Methods: In exploratory analyses, pretreatment levels of programmed death ligand 1 were assessed by immunohistochemistry. Genomic and transcriptomic biomarkers (including tumor mutational burden and gene expression signatures) were also investigated., Results: Biomarkers previously associated with benefit from immune checkpoint inhibitor-containing regimens in RCC were not predictive for survival in patients with RCC treated with nivolumab plus ipilimumab. Analysis of gene expression identified an association between an inflammatory response and progression-free survival with nivolumab plus ipilimumab., Conclusions: The exploratory analyses reveal relationships between molecular biomarkers and provide supportive data on how the inflammation status of the tumor microenvironment may be important for identifying predictive biomarkers of response and survival with combination immunotherapy in patients with RCC. Further validation may help to provide biomarker-driven precision treatment for patients with RCC., Competing Interests: Competing interests: RJM reports consulting fees from Aveo, Calithera, Eisai, Eli Lilly, EMD Serono, Genentech, Merck, Novartis AG, Pfizer, and Roche, and contracted research to employer MSKCC for Bristol Myers Squibb, Eisai, Exelixis, Genentech, Merck, Pfizer, and Roche. TC reports personal and institutional research undertaken for Alexion, Analysis Group, AstraZeneca, Aveo, Bayer, Bristol Myers Squibb/ER Squibb & Sons LLC, Calithera, Cerulean, Corvus, Eisai, Exelixis, F. Hoffmann-La Roche, Foundation Medicine, Genentech, GlaxoSmithKline, Ipsen, Lilly, Merck, Novartis, Peloton, Pfizer, Prometheus Labs, Roche, Roche Products Limited, Sanofi/Aventis, Takeda, and Tracon; consulting/honoraria or advisory roles from Alexion, Analysis Group, AstraZeneca, Aveo, Bayer, Bristol Myers Squibb/ER Squibb & Sons LLC, Cerulean, Corvus, Eisai, EMD Serono, Exelixis, Foundation Medicine Inc, Genentech, GlaxoSmithKline, Heron Therapeutics, Infinity Pharma, Ipsen, Jansen Oncology, IQVIA, Lilly, Merck, NCCN, NiKang, Novartis, Peloton, Pfizer, Pionyr, Prometheus Labs, Roche, Sanofi/Aventis, Surface Oncology, Tempest, and Up-to-Date; travel, accommodations, expenses, and medical writing in relation to consulting, advisory roles, or honoraria; participation in CME-related events by OncLive, PVI, MJH Life Sciences, and in the NCI GU steering committee; owning Pionyr and Tempest stock; and patents filed, royalties, and other intellectual properties related to biomarkers of immune checkpoint inhibitors and ctDNA. TC is also supported in part by the Dana-Farber/Harvard Cancer Center Kidney SPORE and Program, the Kohlberg Chair at Harvard Medical School and the Trust Family, Michael Brigham, and Loker Pinard Funds for Kidney Cancer Research at DFCI. DM reports honoraria from Alkermes, Bristol Myers Squibb, Calithera Biosciences, Eisai, Eli Lilly, EMD Serono, Iovance, Merck, Pfizer, and Werewolf Therapeutics; and research support from Alkermes Inc, Bristol Myers Squibb, Exelixis, Genentech, Merck, Pfizer, and X4 Pharma. TP reports honoraria for advisory/research boards from Astellas, AstraZeneca, Bristol Myers Squibb, Eisai, Exelixis, Incyte, Ipsen, Johnson & Johnson, Merck, Merck Serono, MSD, Novartis, Pfizer, Roche, and Seattle Genetics; institutional grants and funding from Astellas, AstraZeneca, Bristol Myers Squibb, Eisai, Exelixis, Ipsen, Johnson & Johnson, Merck, Merck Serono, MSD, Novartis, Pfizer, Roche, and Seattle Genetics; and fees for travel and accommodation expenses from AstraZeneca, Ipsen, MSD, Roche, and Pfizer. YV reports no conflicts of interest. SG is employed by, and owns stock in, Bristol Myers Squibb. JY is employed by, and owns stock in, Bristol Myers Squibb. CH is employed by Bristol Myers Squibb. RA is employed by, and owns stock in, Bristol Myers Squibb. SP-C is employed by, and a shareholder of, Bristol Myers Squibb. SSS is employed by, and owns stock in, Bristol Myers Squibb. MBM is currently employed by BeiGene and owns stock in Bristol Myers Squibb. MW-R is currently employed by Agios Pharmaceuticals and owns stock in Agios Pharmaceuticals and Bristol Myers Squibb. MBM, MW-R, and PR-M were employees of Bristol Myers Squibb at the time this work was conducted., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
21. Molecular correlates of response to nivolumab at baseline and on treatment in patients with RCC.
- Author
-
Ross-Macdonald P, Walsh AM, Chasalow SD, Ammar R, Papillon-Cavanagh S, Szabo PM, Choueiri TK, Sznol M, and Wind-Rotolo M
- Subjects
- B7-H1 Antigen genetics, CD4 Antigens genetics, CD8 Antigens genetics, Carcinoma, Renal Cell blood, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell immunology, Cytokines blood, Humans, Immune Checkpoint Inhibitors adverse effects, Kidney Neoplasms blood, Kidney Neoplasms genetics, Kidney Neoplasms immunology, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Mutation, Nivolumab adverse effects, Receptors, Antigen, T-Cell genetics, T-Lymphocytes immunology, T-Lymphocytes metabolism, Time Factors, Treatment Outcome, Biomarkers, Tumor blood, Biomarkers, Tumor genetics, Carcinoma, Renal Cell drug therapy, Drug Resistance, Neoplasm genetics, Immune Checkpoint Inhibitors therapeutic use, Kidney Neoplasms drug therapy, Lymphocytes, Tumor-Infiltrating drug effects, Nivolumab therapeutic use, Programmed Cell Death 1 Receptor antagonists & inhibitors, T-Lymphocytes drug effects
- Abstract
Background: Nivolumab is an immune checkpoint inhibitor targeting the programmed death-1 receptor that improves survival in a subset of patients with clear cell renal cell carcinoma (ccRCC). In contrast to other tumor types that respond to immunotherapy, factors such as programmed death ligand-1 (PD-L1) status and tumor mutational burden show limited predictive utility in ccRCC. To address this gap, we report here the first molecular characterization of nivolumab response using paired index lesions, before and during treatment of metastatic ccRCC., Methods: We analyzed gene expression and T-cell receptor (TCR) clonality using lesion-paired biopsies provided in the CheckMate 009 trial and integrated the results with their PD-L1/CD4/CD8 status, genomic mutation status and serum cytokine assays. Statistical tests included linear mixed models, logistic regression models, Fisher's exact test, and Kruskal-Wallis rank-sum test., Results: We identified transcripts related to response, both at baseline and on therapy, including several that are amenable to peripheral bioassays or to therapeutic intervention. At both timepoints, response was positively associated with T-cell infiltration but not associated with TCR clonality, and some non-Responders were highly infiltrated. Lower baseline T-cell infiltration correlated with elevated transcription of Wnt/β-catenin signaling components and hypoxia-regulated genes, including the Treg chemoattractant CCL28. On treatment, analysis of the non-responding patients whose tumors were highly T-cell infiltrated suggests association of the RIG-I-MDA5 pathway in their nivolumab resistance. We also analyzed our data using previous transcriptional classifications of ccRCC and found they concordantly identified a molecular subtype that has enhanced nivolumab response but is sunitinib-resistant., Conclusion: Our study describes molecular characteristics of response and resistance to nivolumab in patients with metastatic ccRCC, potentially impacting patient selection and first-line treatment decisions., Trial Registration Number: NCT01358721., Competing Interests: Competing interests: PR-M, SDC, SP-C, PMS, RA, AMW, and MW-R were employees of Bristol Myers Squibb at the time of their contribution. TKC has served as a consultant/advisor for Pfizer, GlaxoSmithKline, Novartis, Merck, Bristol Myers Squibb, Bayer, Eisai, Roche, and Prometheus Labs, Inc, and has received institutional research funding from Pfizer, Novartis, GlaxoSmithKline, Bristol Myers Squibb, Merck, Exelixis, Roche, AstraZeneca, Peloton, and Tracon. MS has served as a consultant/advisor for Genentech-Roche, Bristol Myers Squibb, AstraZeneca/MedImmune, Pfizer, Novartis, Kyowa-Kirin, Amgen, Merus, Seattle Genetics, Immune Design, Prometheus, Anaeropharma, Astellas-Agensys, Immunova, Nektar, Neostem, Pierre-Fabre, Eli Lilly, Symphogen, Lion Biotechnologies, Amphivena, and Adaptive Biotechnologies., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
22. Spatial heterogeneity in medulloblastoma.
- Author
-
Morrissy AS, Cavalli FMG, Remke M, Ramaswamy V, Shih DJH, Holgado BL, Farooq H, Donovan LK, Garzia L, Agnihotri S, Kiehna EN, Mercier E, Mayoh C, Papillon-Cavanagh S, Nikbakht H, Gayden T, Torchia J, Picard D, Merino DM, Vladoiu M, Luu B, Wu X, Daniels C, Horswell S, Thompson YY, Hovestadt V, Northcott PA, Jones DTW, Peacock J, Wang X, Mack SC, Reimand J, Albrecht S, Fontebasso AM, Thiessen N, Li Y, Schein JE, Lee D, Carlsen R, Mayo M, Tse K, Tam A, Dhalla N, Ally A, Chuah E, Cheng Y, Plettner P, Li HI, Corbett RD, Wong T, Long W, Loukides J, Buczkowicz P, Hawkins CE, Tabori U, Rood BR, Myseros JS, Packer RJ, Korshunov A, Lichter P, Kool M, Pfister SM, Schüller U, Dirks P, Huang A, Bouffet E, Rutka JT, Bader GD, Swanton C, Ma Y, Moore RA, Mungall AJ, Majewski J, Jones SJM, Das S, Malkin D, Jabado N, Marra MA, and Taylor MD
- Subjects
- Adult, Aged, Aged, 80 and over, Cerebellar Neoplasms pathology, Child, Child, Preschool, Cluster Analysis, DNA Copy Number Variations, Female, Gene Expression Profiling methods, Genetic Heterogeneity, Genome-Wide Association Study, Humans, INDEL Mutation, Male, Medulloblastoma pathology, Middle Aged, Mutation, Polymorphism, Single Nucleotide, Principal Component Analysis, Reverse Transcriptase Polymerase Chain Reaction, Cerebellar Neoplasms genetics, Gene Expression Regulation, Neoplastic, Medulloblastoma genetics, Transcriptome
- Abstract
Spatial heterogeneity of transcriptional and genetic markers between physically isolated biopsies of a single tumor poses major barriers to the identification of biomarkers and the development of targeted therapies that will be effective against the entire tumor. We analyzed the spatial heterogeneity of multiregional biopsies from 35 patients, using a combination of transcriptomic and genomic profiles. Medulloblastomas (MBs), but not high-grade gliomas (HGGs), demonstrated spatially homogeneous transcriptomes, which allowed for accurate subgrouping of tumors from a single biopsy. Conversely, somatic mutations that affect genes suitable for targeted therapeutics demonstrated high levels of spatial heterogeneity in MB, malignant glioma, and renal cell carcinoma (RCC). Actionable targets found in a single MB biopsy were seldom clonal across the entire tumor, which brings the efficacy of monotherapies against a single target into question. Clinical trials of targeted therapies for MB should first ensure the spatially ubiquitous nature of the target mutation.
- Published
- 2017
- Full Text
- View/download PDF
23. Impaired H3K36 methylation defines a subset of head and neck squamous cell carcinomas.
- Author
-
Papillon-Cavanagh S, Lu C, Gayden T, Mikael LG, Bechet D, Karamboulas C, Ailles L, Karamchandani J, Marchione DM, Garcia BA, Weinreb I, Goldstein D, Lewis PW, Dancu OM, Dhaliwal S, Stecho W, Howlett CJ, Mymryk JS, Barrett JW, Nichols AC, Allis CD, Majewski J, and Jabado N
- Subjects
- Carcinogenesis genetics, Cell Differentiation genetics, Epigenesis, Genetic genetics, Histone Methyltransferases, Histone-Lysine N-Methyltransferase, Humans, Intracellular Signaling Peptides and Proteins genetics, Mutation genetics, Nuclear Proteins genetics, Papillomaviridae pathogenicity, Papillomavirus Infections genetics, Squamous Cell Carcinoma of Head and Neck, Carcinoma, Squamous Cell genetics, DNA Methylation genetics, Head and Neck Neoplasms genetics, Histones genetics
- Abstract
Human papillomavirus (HPV)-negative head and neck squamous cell carcinomas (HNSCCs) are deadly and common cancers. Recent genomic studies implicate multiple genetic pathways, including cell signaling, cell cycle and immune evasion, in their development. Here we analyze public data sets and uncover a previously unappreciated role of epigenome deregulation in the genesis of 13% of HPV-negative HNSCCs. Specifically, we identify novel recurrent mutations encoding p.Lys36Met (K36M) alterations in multiple H3 histone genes. histones. We further validate the presence of these alterations in multiple independent HNSCC data sets and show that, along with previously described NSD1 mutations, they correspond to a specific DNA methylation cluster. The K36M substitution and NSD1 defects converge on altering methylation of histone H3 at K36 (H3K36), subsequently blocking cellular differentiation and promoting oncogenesis. Our data further indicate limited redundancy for NSD family members in HPV-negative HNSCCs and suggest a potential role for impaired H3K36 methylation in their development. Further investigation of drugs targeting chromatin regulators is warranted in HPV-negative HNSCCs driven by aberrant H3K36 methylation.
- Published
- 2017
- Full Text
- View/download PDF
24. Mutations in a novel serine protease PRSS56 in families with nanophthalmos.
- Author
-
Orr A, Dubé MP, Zenteno JC, Jiang H, Asselin G, Evans SC, Caqueret A, Lakosha H, Letourneau L, Marcadier J, Matsuoka M, Macgillivray C, Nightingale M, Papillon-Cavanagh S, Perry S, Provost S, Ludman M, Guernsey DL, and Samuels ME
- Subjects
- Animals, Base Sequence, Canada, Cloning, Molecular, Cohort Studies, DNA Mutational Analysis, Exons, Eye pathology, Genetic Linkage, Genotype, Genotyping Techniques, Heterozygote, Homozygote, Humans, Hyperopia etiology, Hyperopia pathology, Lod Score, Membrane Proteins genetics, Mexico, Mice, Microphthalmos complications, Microphthalmos pathology, Molecular Sequence Data, Mutation, Pedigree, Eye physiopathology, Hyperopia genetics, Microphthalmos genetics, Serine Proteases genetics
- Abstract
Purpose: Nanophthalmos is a rare genetic ocular disorder in which the eyes of affected individuals are abnormally small. Patients suffer from severe hyperopia as a result of their markedly reduced axial lengths, but otherwise are capable of seeing well unlike other more general forms of microphthalmia. To date one gene for nanophthalmos has been identified, encoding the membrane-type frizzled related protein MFRP. Identification of additional genes for nanophthalmos will improve our understanding of normal developmental regulation of eye growth., Methods: We ascertained a cohort of families from eastern Canada and Mexico with familial nanophthalmos. We performed high density microsatellite and high density single nucleotide polymorphism (SNP) genotyping to identify potential chromosomal regions of linkage. We sequenced coding regions of genes in the linked interval by traditional PCR-based Sanger capillary electrophoresis methods. We cloned and sequenced a novel cDNA from a putative causal gene to verify gene structure., Results: We identified a linked locus on chromosome 2q37 with a peak logarithm (base 10) of odds (LOD) score of 4.7. Sequencing of coding exons of all genes in the region identified multiple segregating variants in one gene, recently annotated as serine protease gene (PRSS56), coding for a predicted trypsin serine protease-like protein. One of our families was homozygous for a predicted pathogenic missense mutation, one family was compound heterozygous for two predicted pathogenic missense mutations, and one family was compound heterozygous for a predicted pathogenic missense mutation plus a frameshift leading to obligatory truncation of the predicted protein. The PRSS56 gene structure in public databases is based on a virtual transcript assembled from overlapping incomplete cDNA clones; we have now validated the structure of a full-length transcript from embryonic mouse brain RNA., Conclusions: PRSS56 is a good candidate for the causal gene for nanophthalmos in our families.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.