183 results on '"Socci, Nd"'
Search Results
2. An autoimmune stem-like CD8 T cell population drives type 1 diabetes
- Author
-
Gearty SV, Dundar F, Zumbo P, Espinosa-Carrasco G, Shakiba M, Sanchez-Rivera FJ, Socci ND, Trivedi P, Lowe SW, Lauer P, Mohibullah N, Viale A, DiLorenzo TP, Betel D, and Schietinger A
- Subjects
General Economics, Econometrics and Finance - Published
- 2022
3. Akt phosphorylation of La regulates specific mRNA translation in glial progenitors
- Author
-
Brenet, F, Socci, ND, Sonenberg, N, and Holland, EC
- Published
- 2009
4. Human PGBD5 DNA transposase promotes site-specific oncogenic mutations in rhabdoid tumors
- Author
-
Henssen, AG, additional, Koche, R, additional, Zhuang, J, additional, Jiang, E, additional, Reed, C, additional, Eisenberg, A, additional, Still, E, additional, Rodríguez-Fos, E, additional, Gonzalez, S, additional, Puiggròs, M, additional, Blackford, AN, additional, Mason, CE, additional, de Stanchina, E, additional, Gönen, M, additional, Emde, AK, additional, Shah, M, additional, Arora, K, additional, Reeves, C, additional, Socci, ND, additional, Perlman, E, additional, Antonescu, CR, additional, Roberts, CWM, additional, Steen, H, additional, Mullen, E, additional, Jackson, SP, additional, Torrents, D, additional, Weng, Z, additional, Armstrong, SA, additional, and Kentsis, A, additional
- Published
- 2017
- Full Text
- View/download PDF
5. Integrated genomic analyses of ovarian carcinoma
- Author
-
Bell, D, Berchuck, A, Birrer, M, Chien, J, Cramer, DW, Dao, F, Dhir, R, DiSaia, P, Gabra, H, Glenn, P, Godwin, AK, Triche, T, Berman, BP, Van den Berg, DJ, Buckley, J, Baylin, SB, Zhang, J, Spellman, PT, Purdom, E, Iacocca, M, Shelton, T, Voet, D, Neuvial, P, Bengtsson, H, Jakkula, LR, Durinck, S, Han, J, Dorton, S, Marr, H, Zhang, H, Choi, YG, Wang, V, Wilkinson, J, Nguyen, H, Wang, NJ, Imielinski, M, Ngai, J, Conboy, JG, Parvin, B, Feiler, HS, Speed, TP, Gray, JW, Wu, CJ, Bloom, T, Levine, DA, Li, L, Socci, ND, Liang, Y, Taylor, BS, Kalloger, S, Schultz, N, Borsu, L, Lash, AE, Brennan, C, Ardlie, K, Viale, A, Shukla, S, Grimm, D, Sander, C, Ladanyi, M, Hoadley, KA, Meng, S, Du, Y, Karlan, BY, Shi, Y, Fennell, T, Cibulskis, K, Lawrence, MS, Meyerson, M, Hatfield, M, Mills, GB, Sivachenko, A, Jing, R, Park, RW, Liu, Y, Park, PJ, Ramos, AH, Noble, M, Chin, L, Carter, H, Kim, D, Morris, S, Winckler, W, Karchin, R, Morrison, C, Baldwin, J, Korkola, JE, Yena, P, Heiser, LM, Getz, G, Cho, RJ, Hu, Z, Gabriel, S, Mutch, D, Cerami, E, Rhodes, P, Olshen, A, Verhaak, RGW, Lander, ES, Reva, B, Antipin, Y, Shen, R, Olvera, N, Mankoo, P, Sheridan, R, Ciriello, G, Sherman, M, Chang, WK, Bernanke, JA, Hayes, DN, Carter, SL, Haussler, D, Orsulic, S, Benz, CC, Paulauskis, J, Stuart, JM, Zhang, N, Benz, SC, Sanborn, JZ, Vaske, CJ, Mermel, CH, Zhu, J, Szeto, C, Scott, GK, Yau, C, Rabeno, B, Ding, L, Park, K, Balu, S, Perou, CM, Saksena, G, Wilkerson, MD, Millis, S, Kahn, A, Turman, YJ, Fulton, RS, Onofrio, RC, Greene, JM, Sfeir, R, Jensen, MA, Chen, J, Whitmore, J, Alonso, S, Jordan, J, Chu, A, Rader, JS, Koboldt, DC, Zang, D, Gross, J, Barker, A, Compton, C, Eley, G, Ferguson, M, Fielding, P, Gerhard, DS, Myles, R, McLellan, MD, Schaefer, C, Helms, EB, Shaw, KRM, Sikic, BI, Vaught, J, Vockley, JB, Good, PJ, Guyer, MS, Ozenberger, B, Wylie, T, Peterson, J, Thomson, E, Smith-McCune, K, Sood, AK, Bowtell, D, Hubbard, D, Penny, R, Testa, JR, Chang, K, Walker, J, Dinh, HH, Drummond, JA, Fowler, G, Zhou, X, Gunaratne, P, Hawes, AC, Kovar, CL, Lewis, LR, Gupta, S, Morgan, MB, O'Laughlin, M, Newsham, IF, Santibanez, J, Reid, JG, Trevino, LR, Wu, J, Wu, Y-Q, Wang, M, Muzny, DM, Wheeler, DA, Gibbs, RA, Crenshaw, A, Sivachenko, AY, Topal, MD, Sougnez, C, Dooling, DJ, Fulton, L, Akbani, R, Abbott, R, Dees, ND, Zhang, Q, Kandoth, C, Wendl, M, Schierding, W, Shen, D, Harris, CC, Baggerly, KA, Schmidt, H, Wilson, RK, Kalicki, J, Delehaunty, KD, Fronick, CC, Demeter, R, Cook, L, Wallis, JW, Lin, L, Magrini, VJ, Yung, WK, Hodges, JS, Protopopov, A, Eldred, JM, Smith, SM, Pohl, CS, Vandin, F, Raphael, BJ, Weinstock, GM, Mardis, R, Kim, TM, Hartmann, L, Perna, I, Xiao, Y, Ren, G, Sathiamoorthy, N, Petrelli, N, Lee, E, Kucherlapati, R, Absher, DM, Huang, M, Waite, L, Sherlock, G, Brooks, JD, Li, JZ, Weinstein, JN, Xu, J, Myers, RM, Laird, PW, Cope, L, Herman, JG, Shen, H, Huntsman, DG, Weisenberger, DJ, Noushmehr, H, Pan, F, Massachusetts Institute of Technology. Department of Biology, Lander, Eric S., and Meyerson, Matthew L.
- Subjects
endocrine system diseases ,Serous carcinoma ,Messenger ,Gene Dosage ,Cancer Genome Atlas Research Network ,GYNECOLOGIC-ONCOLOGY-GROUP ,GRADE SEROUS CARCINOMA ,0302 clinical medicine ,Ovarian carcinoma ,Aged ,Carcinoma ,DNA Methylation ,Female ,Gene Expression Profiling ,Gene Expression Regulation, Neoplastic ,Humans ,MicroRNAs ,Middle Aged ,Mutation ,Ovarian Neoplasms ,RNA, Messenger ,Genomics ,Multidisciplinary ,Genetics ,HYBRID SELECTION ,0303 health sciences ,female genital diseases and pregnancy complications ,3. Good health ,Multidisciplinary Sciences ,Serous fluid ,BRCA MUTATION CARRIERS ,030220 oncology & carcinogenesis ,DNA methylation ,PARP inhibitor ,Science & Technology - Other Topics ,General Science & Technology ,Biology ,Article ,03 medical and health sciences ,CLEAR-CELL CARCINOMA ,MD Multidisciplinary ,microRNA ,HIGH-THROUGHPUT ANNOTATION ,medicine ,DRIVER MUTATIONS ,Gene ,030304 developmental biology ,Neoplastic ,Science & Technology ,MUTANT-CELLS ,SOMATIC MUTATIONS ,medicine.disease ,CANCER STATISTICS ,Gene Expression Regulation ,Cancer research ,RNA ,Ovarian cancer - Abstract
A catalogue of molecular aberrations that cause ovarian cancer is critical for developing and deploying therapies that will improve patients’ lives. The Cancer Genome Atlas project has analysed messenger RNA expression, microRNA expression, promoter methylation and DNA copy number in 489 high-grade serous ovarian adenocarcinomas and the DNA sequences of exons from coding genes in 316 of these tumours. Here we report that high-grade serous ovarian cancer is characterized by TP53 mutations in almost all tumours (96%); low prevalence but statistically recurrent somatic mutations in nine further genes including NF1, BRCA1, BRCA2, RB1 and CDK12; 113 significant focal DNA copy number aberrations; and promoter methylation events involving 168 genes. Analyses delineated four ovarian cancer transcriptional subtypes, three microRNA subtypes, four promoter methylation subtypes and a transcriptional signature associated with survival duration, and shed new light on the impact that tumours with BRCA1/2 (BRCA1 or BRCA2) and CCNE1 aberrations have on survival. Pathway analyses suggested that homologous recombination is defective in about half of the tumours analysed, and that NOTCH and FOXM1 signalling are involved in serous ovarian cancer pathophysiology., National Institutes of Health (U.S.) (Grant U54HG003067), National Institutes of Health (U.S.) (Grant U54HG003273), National Institutes of Health (U.S.) (Grant U54HG003079), National Institutes of Health (U.S.) (Grant U24CA126543), National Institutes of Health (U.S.) (Grant U24CA126544), National Institutes of Health (U.S.) (Grant U24CA126546), National Institutes of Health (U.S.) (Grant U24CA126551), National Institutes of Health (U.S.) (Grant U24CA126554), National Institutes of Health (U.S.) (Grant U24CA126561), National Institutes of Health (U.S.) (Grant U24CA126563), National Institutes of Health (U.S.) (Grant U24CA143882), National Institutes of Health (U.S.) (Grant U24CA143731), National Institutes of Health (U.S.) (Grant U24CA143835), National Institutes of Health (U.S.) (Grant U24CA143845), National Institutes of Health (U.S.) (Grant U24CA143858), National Institutes of Health (U.S.) (Grant U24CA144025), National Institutes of Health (U.S.) (Grant U24CA143866), National Institutes of Health (U.S.) (Grant U24CA143867), National Institutes of Health (U.S.) (Grant U24CA143848), National Institutes of Health (U.S.) (Grant U24CA143843), National Institutes of Health (U.S.) (Grant R21CA135877)
- Published
- 2010
6. A recurrent germline PAX5 mutation confers susceptibility to pre-B cell acute lymphoblastic leukemia
- Author
-
Shah, S, Schrader, KA, Vijai, J, Miething, C, Hayes, J, Klein, RJ, Gao, X, Kitzing, T, Lowe, SW, Offit, K, Littman, J, Offit, L, Rau-Murthy, R, Fleischut, MH, Corines, M, Manschreck, C, Waanders, E, Wei, L, Ma, J, Chen, S-C, Song, G, Cheng, J, Raimondi, SC, Roberts, KG, Downing, JR, Mullighan, CG, Kuiper, RP, Simons, A, Timms, AE, Wechsler, J, Horwitz, MS, Yang, J, Zhang, J, Wu, G, Rusch, M, Nagahawatte, P, Meyers, P, Bhojwani, D, Sandlund, JT, Jhanwar, S, Murali, R, Maslak, P, Fleisher, M, Murty, VV, Schiffman, JD, Onel, K, Plon, SE, Wheeler, DA, Ritter, D, Ziegler, DS, Sutton, R, Tucker, K, Chenevix-Trench, G, Li, J, Huntsman, DG, Hansford, S, Senz, J, Walsh, T, Lee, M, King, M-C, Hahn, CN, Scott, HS, Lo, SM, Levine, RL, Viale, A, Socci, ND, Nathanson, KL, Daly, M, Lipkin, SM, Altshuler, D, Shah, S, Schrader, KA, Vijai, J, Miething, C, Hayes, J, Klein, RJ, Gao, X, Kitzing, T, Lowe, SW, Offit, K, Littman, J, Offit, L, Rau-Murthy, R, Fleischut, MH, Corines, M, Manschreck, C, Waanders, E, Wei, L, Ma, J, Chen, S-C, Song, G, Cheng, J, Raimondi, SC, Roberts, KG, Downing, JR, Mullighan, CG, Kuiper, RP, Simons, A, Timms, AE, Wechsler, J, Horwitz, MS, Yang, J, Zhang, J, Wu, G, Rusch, M, Nagahawatte, P, Meyers, P, Bhojwani, D, Sandlund, JT, Jhanwar, S, Murali, R, Maslak, P, Fleisher, M, Murty, VV, Schiffman, JD, Onel, K, Plon, SE, Wheeler, DA, Ritter, D, Ziegler, DS, Sutton, R, Tucker, K, Chenevix-Trench, G, Li, J, Huntsman, DG, Hansford, S, Senz, J, Walsh, T, Lee, M, King, M-C, Hahn, CN, Scott, HS, Lo, SM, Levine, RL, Viale, A, Socci, ND, Nathanson, KL, Daly, M, Lipkin, SM, and Altshuler, D
- Abstract
Somatic alterations of the lymphoid transcription factor gene PAX5 (also known as BSAP) are a hallmark of B cell precursor acute lymphoblastic leukemia (B-ALL), but inherited mutations of PAX5 have not previously been described. Here we report a new heterozygous germline variant, c.547G>A (p.Gly183Ser), affecting the octapeptide domain of PAX5 that was found to segregate with disease in two unrelated kindreds with autosomal dominant B-ALL. Leukemic cells from all affected individuals in both families exhibited 9p deletion, with loss of heterozygosity and retention of the mutant PAX5 allele at 9p13. Two additional sporadic ALL cases with 9p loss harbored somatic PAX5 substitutions affecting Gly183. Functional and gene expression analysis of the PAX5 mutation demonstrated that it had significantly reduced transcriptional activity. These data extend the role of PAX5 alterations in the pathogenesis of pre-B cell ALL and implicate PAX5 in a new syndrome of susceptibility to pre-B cell neoplasia. © 2013 Nature America, Inc. All rights reserved.
- Published
- 2013
7. Identification of angiogenesis/metastases genes predicting chemoradiotherapy response in patients with laryngopharyngeal carcinoma.
- Author
-
Ganly I, Talbot S, Carlson D, Viale A, Maghami E, Osman I, Sherman E, Pfister D, Chuai S, Shaha AR, Kraus D, Shah JP, Socci ND, and Singh B
- Published
- 2007
8. PGBD5 promotes site-specific oncogenic mutations in human tumors
- Author
-
Henssen, AG, Koche, R, Zhuang, J, Jiang, E, Reed, C, Eisenberg, A, Still, E, MacArthur, IC, Rodríguez-Fos, E, Gonzalez, S, Puiggròs, M, Blackford, AN, Mason, CE, De Stanchina, E, Gönen, M, Emde, A-K, Shah, M, Arora, K, Reeves, C, Socci, ND, Perlman, E, Antonescu, CR, Roberts, CWM, Steen, H, Mullen, E, Jackson, SP, Torrents, D, Weng, Z, Armstrong, SA, and Kentsis, A
- Subjects
embryonal neoplasms ,mutagenesis ,3. Good health - Abstract
Genomic rearrangements are a hallmark of human cancers. Here, we identify the piggyBac transposable element derived 5 (PGBD5) gene as encoding an active DNA transposase expressed in the majority of childhood solid tumors, including lethal rhabdoid tumors. Using assembly-based whole-genome DNA sequencing, we found previously undefined genomic rearrangements in human rhabdoid tumors. These rearrangements involved PGBD5-specific signal (PSS) sequences at their breakpoints and recurrently inactivated tumor-suppressor genes. PGBD5 was physically associated with genomic PSS sequences that were also sufficient to mediate PGBD5-induced DNA rearrangements in rhabdoid tumor cells. Ectopic expression of PGBD5 in primary immortalized human cells was sufficient to promote cell transformation in vivo. This activity required specific catalytic residues in the PGBD5 transposase domain as well as end-joining DNA repair and induced structural rearrangements with PSS breakpoints. These results define PGBD5 as an oncogenic mutator and provide a plausible mechanism for site-specific DNA rearrangements in childhood and adult solid tumors.
9. Tissue-specific features of the T cell repertoire after allogeneic hematopoietic cell transplantation in human and mouse
- Author
-
DeWolf, S, primary, Elhanati, Y, additional, Nichols, K, additional, Waters, NR, additional, Nguyen, CL, additional, Slingerland, JB, additional, Rodriguez, N, additional, Lyudovyk, O, additional, Giardina, PA, additional, Kousa, AI, additional, Andrlová, H, additional, Ceglia, N, additional, Fe, T, additional, Kappagantual, R, additional, Li, Y, additional, Aleynick, N, additional, Baez, P, additional, Murali, R, additional, Hayashi, A, additional, Lee, N, additional, Gipson, B, additional, Rangesa, M, additional, Katsamakis, Z, additional, Dai, A, additional, Blouin, AG, additional, Arcila, M, additional, Masilionis, I, additional, Chaligne, R, additional, Ponce, DM, additional, Landau, HJ, additional, Politikos, I, additional, Tamari, R, additional, Hanash, AM, additional, Jenq, RR, additional, Giralt, SA, additional, Markey, KA, additional, Zhang, Y, additional, Perales, M, additional, Socci, ND, additional, Greenbaum, BD, additional, Iacobuzio-Donahue, CA, additional, Hollmann, TJ, additional, van den Brink, MRM, additional, and Peled, U, additional
- Full Text
- View/download PDF
10. Genome-Wide Profiling of Papillary Thyroid Cancer Identifies MUC1 as an Independent Prognostic Marker
- Author
-
Ashok R. Shaha, Geoffrey Childs, Michael Hezel, Bhuvanesh Singh, Jatin P. Shah, Dennis H. Kraus, Pulivarthi H. Rao, Snehal G. Patel, Ronald Ghossein, Kepal N. Patel, Elizabeth M. Sieczka, Nicholas D. Socci, Giovanni Tallini, Volkert B. Wreesmann, Thomas J. Belbin, Michael B. Prystowsky, Wreesmann VB, Sieczka EM, Socci ND, Hezel M, Belbin TJ, Childs G, Patel SG, Patel KN, Tallini G, Prystowsky M, Shaha AR, Kraus D, Shah JP, Rao PH, Ghossein R, and Singh B
- Subjects
Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Microarray ,Gene Dosage ,Biology ,Polymerase Chain Reaction ,Gene dosage ,law.invention ,Papillary thyroid cancer ,law ,Gene duplication ,medicine ,Humans ,Thyroid Neoplasms ,Thyroid cancer ,Gene ,Polymerase chain reaction ,Gene Expression Profiling ,Mucin-1 ,Gene Amplification ,Reproducibility of Results ,Middle Aged ,Prognosis ,medicine.disease ,Immunohistochemistry ,Carcinoma, Papillary ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Gene expression profiling ,Oncology ,Cancer research ,Female - Abstract
Clinicopathological variables used at present for prognostication and treatment selection for papillary thyroid carcinomas (PTCs) do not uniformly predict tumor behavior, necessitating identification of novel prognostic markers. Complicating the assessment is the long natural history of PTC and our rudimentary knowledge of its genetic composition. In this study we took advantage of differences in clinical behavior of two distinct variants of PTC, the aggressive tall-cell variant (TCV) and indolent conventional PTC (cPTC), to identify molecular prognosticators of outcome using complementary genome wide analyses. Comparative genome hybridization (CGH) and cDNA microarray (17,840 genes) analyses were used to detect changes in DNA copy number and gene expression in pathological cPTC and TCV. The findings from CGH and cDNA microarray analyses were correlated and validated by real-time PCR and immunohistochemical analyses on a series of 100 cases of cPTC and TCV. Genes identified by this approach were evaluated as prognostic markers in cPTC by immunohistochemistry on tissue arrays. CGH identified significant differences in the presence (76 versus 27%; P = 0.001) and type of DNA copy number aberrations in TCV compared with cPTC. Recurrent gains of 1p34–36, 1q21, 6p21–22, 9q34, 11q13, 17q25, 19, and 22 and losses of 2q21–31, 4, 5p14-q21, 6q11–22, 8q11–22, 9q11–32, and 13q21–31 were unique to TCV. Hierarchical clustering of gene expression profiles revealed significant overlap between TCV and cPTC, but further analysis identified 82 dysregulated genes differentially expressed among the PTC variants. Of these, MUC1 was of particular interest because amplification of 1q by CGH correlated with MUC1 amplification by real-time PCR analysis and protein overexpression by immunohistochemistry in TCV (P = 0.005). Multivariate analysis revealed a significant association between MUC1 overexpression and treatment outcome, independent of histopathological categorization (P = 0.03). Analysis of a validation series containing a matched group of aggressive and indolent cPTCs confirmed the association between MUC1 overexpression and survival (relative risk, 2.3; 95% confidence interval, 1.1–5.5; P = 0.03). Our data suggest that MUC1 dysregulation is associated with aggressive behavior of PTC and may serve as a prognostic marker and potential therapeutic target in this disease.
- Published
- 2004
11. A Mammalian microRNA Expression Atlas Based on Small RNA Library Sequencing
- Author
-
Astrid Novosel, Markus Landthaler, Julia Schliwka, Sabina Chiaretti, Miklós Palkovits, Roberto Di Lauro, Nicola Iovino, Mirabela Rusu, Roman-Ulrich Müller, Giuseppe Macino, James W. Nagle, Andreas Bosio, Mihaela Zavolan, Peter Wernet, Alice O. Kamphorst, Charles E. Rogler, Chris Sander, Veit Hornung, James J. Russo, Wayne Tam, Thomas Benzing, Alexei A. Aravin, Uta Fuchs, Thomas Tuschl, Pablo Landgraf, Sébastien Pfeffer, Alain Sewer, Hans Ingo Trompeter, Jason M. Inman, Ruchi Choksi, Amanda J. Rice, Arndt Borkhardt, Robert L. Sheridan, Valerio Fulci, Carolina Lin, Leandro C. Hermida, Michael J. Brownstein, Ute Bissels, Bernhard Schermer, Daniela Frezzetti, Minchen Chien, Grace Teng, Jingyue Ju, Quang Phan, Gunther Hartmann, F. Nina Papavasiliou, Robin Foà, Nicholas D. Socci, David B. Weir, Gabriella De Vita, Peter Lichter, Landgraf, P, Rusu, M, Sheridan, R, Sewer, A, Iovino, N, Aravin, A, Pfeffer, S, Rice, A, Kamphorst, Ao, Landthaler, M, Lin, C, Socci, Nd, Hermida, L, Fulci, V, Chiaretti, S, Foa, R, Schliwka, J, Fuchs, U, Novosel, A, Muller, Ru, Schermer, B, Bissels, U, Inman, J, Phan, Q, Chien, M, Weir, Db, Choksi, R, DE VITA, Gabriella, Frezzetti, D, Trompeter, Hi, Hornung, V, Teng, G, Hartmann, G, Palkovits, M, DI LAURO, Roberto, Wernet, P, Macino, G, Rogler, Ce, Nagle, Jw, Ju, J, Papavasiliou, Fn, Benzing, T, Lichter, P, Tam, W, Brownstein, Mj, Bosio, A, Borkhardt, A, Russo, Jj, Sander, C, Zavolan, M, Tuschl, T., Institut de biologie moléculaire des plantes (IBMP), and Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)
- Subjects
Small RNA ,moleneuro ,molimmuno ,rna ,Molecular Sequence Data ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Genome ,General Biochemistry, Genetics and Molecular Biology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,IsomiR ,Mirtron ,Sequence Homology, Nucleic Acid ,microRNA ,Animals ,Humans ,Cell Lineage ,RNA, Messenger ,MOLIMMUNO ,Conserved Sequence ,Phylogeny ,MOLENEURO ,Gene Library ,030304 developmental biology ,Genetics ,Regulation of gene expression ,0303 health sciences ,Base Sequence ,Biochemistry, Genetics and Molecular Biology(all) ,Gene Expression Profiling ,RNA ,Hematopoietic Stem Cells ,Rats ,Gene expression profiling ,MicroRNAs ,Gene Expression Regulation ,Hematologic Neoplasms ,030220 oncology & carcinogenesis - Abstract
SummaryMicroRNAs (miRNAs) are small noncoding regulatory RNAs that reduce stability and/or translation of fully or partially sequence-complementary target mRNAs. In order to identify miRNAs and to assess their expression patterns, we sequenced over 250 small RNA libraries from 26 different organ systems and cell types of human and rodents that were enriched in neuronal as well as normal and malignant hematopoietic cells and tissues. We present expression profiles derived from clone count data and provide computational tools for their analysis. Unexpectedly, a relatively small set of miRNAs, many of which are ubiquitously expressed, account for most of the differences in miRNA profiles between cell lineages and tissues. This broad survey also provides detailed and accurate information about mature sequences, precursors, genome locations, maturation processes, inferred transcriptional units, and conservation patterns. We also propose a subclassification scheme for miRNAs for assisting future experimental and computational functional analyses.
- Published
- 2007
12. Author Correction: Cancer-associated Histone H3 N-terminal arginine mutations disrupt PRC2 activity and impair differentiation.
- Author
-
Nacev BA, Dabas Y, Paul MR, Pacheco C, Mitchener M, Perez Y, Fang Y, Soshnev AA, Barrows D, Carroll T, Socci ND, St Jean SC, Tiwari S, Gruss MJ, Monette S, Tap WD, Garcia BA, Muir T, and Allis CD
- Published
- 2024
- Full Text
- View/download PDF
13. The Evolutionary Forest of Pancreatic Cancer.
- Author
-
Mullen KM, Hong J, Attiyeh MA, Hayashi A, Sakamoto H, Kohutek ZA, McIntyre CA, Zhang H, Makohon-Moore AP, Zucker A, Wood LD, Myers MA, Arnold BJ, Zaccaria S, Chou JF, Capanu M, Socci ND, Raphael BJ, and Iacobuzio-Donahue CA
- Abstract
The genomic features of pancreatic ductal adenocarcinoma (PDAC) have been well described, yet the evolutionary contexts within which those features occur remains unexplored. We studied the genome landscapes, phylogenies and clonal compositions of 91 PDACs in relation to clinicopathologic features. There was no difference in the number of driver mutations or the evolutionary timing that each mutation occurred. High truncal density, a metric of the accumulation of somatic mutations in the lineage that gave rise to each PDAC, was significantly associated with worse overall survival. Polyclonal, monoclonal or mixed polyclonal/monoclonal metastases were identified across the cohort highlighting multiple forms of inter-tumoral heterogeneity. Advanced stage and treated PDACs had higher odds of being polyclonal, whereas oligometastatic PDACs had fewer driver alterations, a lower fractional allelic loss and increased likelihood of being monoclonal. In sum, our findings reveal novel insights into the dynamic nature of the PDAC genome beyond established genetic paradigms.
- Published
- 2024
- Full Text
- View/download PDF
14. Intratumoral Escherichia Is Associated With Improved Survival to Single-Agent Immune Checkpoint Inhibition in Patients With Advanced Non-Small-Cell Lung Cancer.
- Author
-
Elkrief A, Montesion M, Sivakumar S, Hale C, Bowman AS, Begüm Bektaş A, Bradic M, Kang W, Chan E, Gogia P, Manova-Todorova K, Mata DA, Egger JV, Rizvi H, Socci ND, Kelly DW, Rosiek E, Meng F, Tam G, Fan N, Drilon A, Yu HA, Riely GJ, Rekhtman N, Quintanal Villalonga Á, Dogan S, Bhanot U, Gönen M, Loomis B, Hellmann MD, Schoenfeld AJ, Ladanyi M, Rudin CM, and Vanderbilt CM
- Subjects
- Humans, Female, Male, Aged, Middle Aged, Tumor Microenvironment immunology, Aged, 80 and over, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung mortality, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms mortality, Lung Neoplasms pathology, Lung Neoplasms immunology, Lung Neoplasms microbiology, Immune Checkpoint Inhibitors therapeutic use
- Abstract
PURPOSEThe impact of the intratumoral microbiome on immune checkpoint inhibitor (ICI) efficacy in patients with non-small-cell lung cancer (NSCLC) is unknown. Preclinically, intratumoral Escherichia is associated with a proinflammatory tumor microenvironment and decreased metastases. We sought to determine whether intratumoral Escherichia is associated with outcome to ICI in patients with NSCLC.PATIENTS AND METHODSWe examined the intratumoral microbiome in 958 patients with advanced NSCLC treated with ICI by querying unmapped next-generation sequencing reads against a bacterial genome database. Putative environmental contaminants were filtered using no-template controls (n = 2,378). The impact of intratumoral Escherichia detection on overall survival (OS) was assessed using univariable and multivariable analyses. The findings were further validated in an external independent cohort of 772 patients. Escherichia fluorescence in situ hybridization (FISH) and transcriptomic profiling were performed.RESULTSIn the discovery cohort, read mapping to intratumoral Escherichia was associated with significantly longer OS (16 v 11 months; hazard ratio, 0.73 [95% CI, 0.59 to 0.92]; P = .0065) in patients treated with single-agent ICI, but not combination chemoimmunotherapy. The association with OS in the single-agent ICI cohort remained statistically significant in multivariable analysis adjusting for prognostic features including PD-L1 expression ( P = .023). Analysis of an external validation cohort confirmed the association with improved OS in univariable and multivariable analyses of patients treated with single-agent ICI, and not in patients treated with chemoimmunotherapy. Escherichia localization within tumor cells was supported by coregistration of FISH staining and serial hematoxylin and eosin sections. Transcriptomic analysis correlated Escherichia-positive samples with expression signatures of immune cell infiltration.CONCLUSIONRead mapping to potential intratumoral Escherichia was associated with survival to single-agent ICI in two independent cohorts of patients with NSCLC.
- Published
- 2024
- Full Text
- View/download PDF
15. Multiplexed Spatial Profiling of Hodgkin Reed-Sternberg Cell Neighborhoods in Classic Hodgkin Lymphoma.
- Author
-
Pourmaleki M, Jones CJ, Mellinghoff SD, Greenstein BD, Kumar P, Foronda M, Navarrete DA, Campos C, Roshal M, Schultz N, Shah SP, Schietinger A, Socci ND, Hollmann TJ, Dogan A, and Mellinghoff IK
- Subjects
- Humans, Herpesvirus 4, Human isolation & purification, Female, Male, Gene Expression Profiling, Adult, Epstein-Barr Virus Infections complications, Epstein-Barr Virus Infections virology, Middle Aged, CD8-Positive T-Lymphocytes immunology, Aged, Transcriptome, Hodgkin Disease pathology, Hodgkin Disease immunology, Hodgkin Disease virology, Reed-Sternberg Cells pathology, Tumor Microenvironment immunology
- Abstract
Purpose: Classic Hodgkin lymphoma (cHL) is a B-cell lymphoma that occurs primarily in young adults and, less frequently, in elderly individuals. A hallmark of cHL is the exceptional scarcity (1%-5%) of the malignant Hodgkin Reed-Sternberg (HRS) cells within a network of nonmalignant immune cells. Molecular determinants governing the relationship between HRS cells and their proximal microenvironment remain largely unknown., Experimental Design: We performed spatially resolved multiplexed protein imaging and transcriptomic sequencing to characterize HRS cell states, cellular neighborhoods, and gene expression signatures of 23.6 million cells from 36 newly diagnosed Epstein-Barr virus (EBV)-positive and EBV-negative cHL tumors., Results: We show that MHC-I expression on HRS cells is associated with immune-inflamed neighborhoods containing CD8+ T cells, MHC-II+ macrophages, and immune checkpoint expression (i.e., PD1 and VISTA). We identified spatial clustering of HRS cells, consistent with the syncytial variant of cHL, and its association with T-cell-excluded neighborhoods in a subset of EBV-negative tumors. Finally, a subset of both EBV-positive and EBV-negative tumors contained regulatory T-cell-high neighborhoods harboring HRS cells with augmented proliferative capacity., Conclusions: Our study links HRS cell properties with distinct immunophenotypes and potential immune escape mechanisms in cHL., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
16. A transposase-derived gene required for human brain development.
- Author
-
Zapater LJ, Lewis SA, Gutierrez RL, Yamada M, Rodriguez-Fos E, Planas-Felix M, Cameron D, Demarest P, Nabila A, Mueller H, Zhao J, Bergin P, Reed C, Chwat-Edelstein T, Pagnozzi A, Nava C, Bourel-Ponchel E, Cornejo P, Dursun A, Özgül RK, Akar HT, Maroofian R, Houlden H, Cheema HA, Anjum MN, Zifarelli G, Essid M, Ben Hafsa M, Benrhouma H, Montoya CIG, Proekt A, Zhao X, Socci ND, Hayes M, Bigot Y, Rabadan R, Torrents D, Kleinmann CL, Kruer MC, Toth M, and Kentsis A
- Abstract
DNA transposable elements and transposase-derived genes are present in most living organisms, including vertebrates, but their function is largely unknown. PiggyBac Transposable Element Derived 5 (PGBD5) is an evolutionarily conserved vertebrate DNA transposase-derived gene with retained nuclease activity in human cells. Vertebrate brain development is known to be associated with prominent neuronal cell death and DNA breaks, but their causes and functions are not well understood. Here, we show that PGBD5 contributes to normal brain development in mice and humans, where its deficiency causes disorder of intellectual disability, movement, and seizures. In mice, Pgbd5 is required for the developmental induction of post-mitotic DNA breaks and recurrent somatic genome rearrangements. In the brain cortex, loss of Pgbd5 leads to aberrant differentiation and gene expression of distinct neuronal populations, including specific types of glutamatergic neurons, which explains the features of PGBD5 deficiency in humans. Thus, PGBD5 might be a transposase-derived enzyme required for brain development in mammals., Competing Interests: Competing interests: Authors declare that they have no competing interests. AK is a consultant for Novartis, Rgenta, Blueprint, and Syndax. RR is a founder and a member of the SAB of Genotwin, and a member of the SAB of Diatech Pharmacogenetics. None of these activities are related to the work described in this manuscript.
- Published
- 2024
- Full Text
- View/download PDF
17. A microglia clonal inflammatory disorder in Alzheimer's Disease.
- Author
-
Vicario R, Fragkogianni S, Weber L, Lazarov T, Hu Y, Hayashi SY, Craddock BP, Socci ND, Alberdi A, Baako A, Ay O, Ogishi M, Lopez-Rodrigo E, Kappagantula R, Viale A, Iacobuzio-Donahue CA, Zhou T, Ransohoff RM, Chesworth R, Abdel-Wahab O, Boisson B, Elemento O, Casanova JL, Miller WT, and Geissmann F
- Abstract
Somatic genetic heterogeneity resulting from post-zygotic DNA mutations is widespread in human tissues and can cause diseases, however few studies have investigated its role in neurodegenerative processes such as Alzheimer's Disease (AD). Here we report the selective enrichment of microglia clones carrying pathogenic variants, that are not present in neuronal, glia/stromal cells, or blood, from patients with AD in comparison to age-matched controls. Notably, microglia-specific AD-associated variants preferentially target the MAPK pathway, including recurrent CBL ring-domain mutations. These variants activate ERK and drive a microglia transcriptional program characterized by a strong neuro-inflammatory response, both in vitro and in patients. Although the natural history of AD-associated microglial clones is difficult to establish in human, microglial expression of a MAPK pathway activating variant was previously shown to cause neurodegeneration in mice, suggesting that AD-associated neuroinflammatory microglial clones may contribute to the neurodegenerative process in patients., Competing Interests: Competing interests. FG has been a paid consultant (no equity) to Third Rock Ventures from 2018 to 2020. Sequencing costs and analysis in this study were covered in part by a SRA between Third Rock venture and MSKCC. This work led to patents PCT/US2022/037893/WO2023004054A1 ‘Methods and compositions for the treatment of alzheimer’s disease’ by MSKCC and PCT/US2018/047964 ‘Kinase mutation-associated neurodegenerative disorders by MSKCC’.
- Published
- 2024
- Full Text
- View/download PDF
18. Mechanism of neurodegeneration mediated by clonal inflammatory microglia.
- Author
-
Vicario R, Fragkogianni S, Pokrovskii M, Mayer C, Lopez-Rodrigo E, Hu Y, Ogishi M, Alberdi A, Baako A, Ay O, Plu I, Sazdovitch V, Heritier S, Cohen-Aubart F, Shor N, Miyara M, Nguyen-Khac F, Viale A, Idbaih A, Amoura Z, Rosenblum MK, Zhang H, Karnoub ER, Sashittal P, Jakatdar A, Iacobuzio-Donahue CA, Abdel-Wahab O, Tabar V, Socci ND, Elemento O, Diamond EL, Boisson B, Casanova JL, Seilhean D, Haroche J, Donadieu J, and Geissmann F
- Abstract
Langerhans cell Histiocytosis (LCH) and Erdheim-Chester disease (ECD) are clonal myeloid disorders, associated with MAP-Kinase activating mutations and an increased risk of neurodegeneration. Surprisingly, we found pervasive PU.1
+ microglia mutant clones across the brain of LCH and ECD patients with and without neurological symptoms, associated with microgliosis, reactive astrocytosis, and neuronal loss. The disease predominated in the grey nuclei of the rhombencephalon, a topography attributable to a local proliferative advantage of mutant microglia. Presence of clinical symptoms was associated with a longer evolution of the disease and a larger size of PU.1+ clones (p= 0.0003). Genetic lineage tracing of PU.1+ clones suggest a resident macrophage lineage or a bone marrow precursor origin depending on patients. Finally, a CSF1R-inhibitor depleted mutant microglia and limited neuronal loss in mice suggesting an alternative to MAPK inhibitors. These studies characterize a progressive neurodegenerative disease, caused by clonal proliferation of inflammatory microglia (CPIM), with a decade(s)-long preclinical stage of incipient disease that represent a therapeutic window for prevention of neuronal death., Competing Interests: Conflict of Interest. FG has performed consulting for Third Rock venture in the past. Targeted Sequencing was funded in part by a grant from Third Rock venture. FG and RV are inventors in MSKCC’s United States application or PCT international application number PCT/US2018/047964 filed on 8/24/2018 (KINASE MUTATION-ASSOCIATED NEURODEGENERATIVE DISORDERS)- Published
- 2024
- Full Text
- View/download PDF
19. CDC7 inhibition impairs neuroendocrine transformation in lung and prostate tumors through MYC degradation.
- Author
-
Quintanal-Villalonga A, Kawasaki K, Redin E, Uddin F, Rakhade S, Durani V, Sabet A, Shafer M, Karthaus WR, Zaidi S, Zhan YA, Manoj P, Sridhar H, Kinyua D, Zhong H, Mello BP, Ciampricotti M, Bhanot UK, Linkov I, Qiu J, Patel RA, Morrissey C, Mehta S, Barnes J, Haffner MC, Socci ND, Koche RP, de Stanchina E, Molina-Pinelo S, Salehi S, Yu HA, Chan JM, and Rudin CM
- Subjects
- Humans, Male, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Cell Line, Tumor, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Protein Serine-Threonine Kinases antagonists & inhibitors, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Mice, Animals, Neuroendocrine Tumors genetics, Neuroendocrine Tumors pathology, Neuroendocrine Tumors metabolism, Neuroendocrine Tumors drug therapy, Proteolysis drug effects, Retinoblastoma Binding Proteins genetics, Retinoblastoma Binding Proteins metabolism, Ubiquitin-Protein Ligases, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Prostatic Neoplasms metabolism, Prostatic Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms pathology, Lung Neoplasms metabolism, Lung Neoplasms drug therapy, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism
- Abstract
Neuroendocrine (NE) transformation is a mechanism of resistance to targeted therapy in lung and prostate adenocarcinomas leading to poor prognosis. Up to date, even if patients at high risk of transformation can be identified by the occurrence of Tumor Protein P53 (TP53) and Retinoblastoma Transcriptional Corepressor 1 (RB1) mutations in their tumors, no therapeutic strategies are available to prevent or delay histological transformation. Upregulation of the cell cycle kinase Cell Division Cycle 7 (CDC7) occurred in tumors during the initial steps of NE transformation, already after TP53/RB1 co-inactivation, leading to induced sensitivity to the CDC7 inhibitor simurosertib. CDC7 inhibition suppressed NE transdifferentiation and extended response to targeted therapy in in vivo models of NE transformation by inducing the proteasome-mediated degradation of the MYC Proto-Oncogen (MYC), implicated in stemness and histological transformation. Ectopic overexpression of a degradation-resistant MYC isoform reestablished the NE transformation phenotype observed on targeted therapy, even in the presence of simurosertib. CDC7 inhibition also markedly extended response to standard cytotoxics (cisplatin, irinotecan) in lung and prostate small cell carcinoma models. These results nominate CDC7 inhibition as a therapeutic strategy to constrain lineage plasticity, as well as to effectively treat NE tumors de novo or after transformation. As simurosertib clinical efficacy trials are ongoing, this concept could be readily translated for patients at risk of transformation., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
20. Antigen escape as a shared mechanism of resistance to BCMA-directed therapies in multiple myeloma.
- Author
-
Firestone RS, Socci ND, Shekarkhand T, Zhu M, Qin WG, Hultcrantz M, Mailankody S, Tan CR, Korde N, Lesokhin AM, Hassoun H, Shah U, Maclachlan KH, Rajeeve S, Landau HJ, Scordo M, Shah GL, Lahoud OB, Giralt S, Murata K, Usmani SZ, and Chung DJ
- Subjects
- Humans, Male, Female, Middle Aged, Antibodies, Bispecific therapeutic use, Aged, Antibodies, Monoclonal, Humanized, Multiple Myeloma immunology, Multiple Myeloma drug therapy, Multiple Myeloma genetics, Multiple Myeloma pathology, Multiple Myeloma therapy, B-Cell Maturation Antigen genetics, B-Cell Maturation Antigen immunology, Drug Resistance, Neoplasm
- Abstract
Abstract: B-cell maturation antigen (BCMA)-targeting therapeutics have dramatically improved outcomes in relapsed/refractory multiple myeloma (RRMM). However, whether the mechanisms of resistance between these therapies are shared and how the identification of such mechanisms before therapy initiation could refine clinical decision-making remains undefined. We analyzed outcomes for 72 RRMM patients treated with teclistamab, a CD3 × BCMA bispecific antibody, 42% (30/72) of whom had prior BCMA-directed therapy exposure. Malignant plasma cell BCMA expression was present in all BCMA therapy-naïve patients. Prior therapy-mediated loss of plasma cell BCMA expression before teclistamab treatment, measured by immunohistochemistry, was observed in 3 patients, none of whom responded to teclistamab, and 1 of whom also did not respond to ciltacabtagene autoleucel. Whole exome sequencing of tumor DNA from 1 patient revealed biallelic loss of TNFRSF17 following treatment with belantamab mafodotin. Low-to-undetectable peripheral blood soluble BCMA levels correlated with the absence of BCMA expression by bone marrow plasma cells. Thus, although rare, loss of BCMA expression following TNFRSF17 gene deletions can occur following any BCMA-directed therapy and prevents response to subsequent anti-BCMA-directed treatments, underscoring the importance of verifying the presence of a target antigen., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
21. Cancer-associated Histone H3 N-terminal arginine mutations disrupt PRC2 activity and impair differentiation.
- Author
-
Nacev BA, Dabas Y, Paul MR, Pacheco C, Mitchener M, Perez Y, Fang Y, Soshnev AA, Barrows D, Carroll T, Socci ND, St Jean SC, Tiwari S, Gruss MJ, Monette S, Tap WD, Garcia BA, Muir T, and Allis CD
- Subjects
- Animals, Humans, Mice, Chromatin metabolism, Epigenesis, Genetic, Mesenchymal Stem Cells metabolism, Cell Line, Tumor, Histones metabolism, Histones genetics, Cell Differentiation genetics, Arginine metabolism, Polycomb Repressive Complex 2 metabolism, Polycomb Repressive Complex 2 genetics, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology, Mutation
- Abstract
Dysregulated epigenetic states are a hallmark of cancer and often arise from genetic alterations in epigenetic regulators. This includes missense mutations in histones, which, together with associated DNA, form nucleosome core particles. However, the oncogenic mechanisms of most histone mutations are unknown. Here, we demonstrate that cancer-associated histone mutations at arginines in the histone H3 N-terminal tail disrupt repressive chromatin domains, alter gene regulation, and dysregulate differentiation. We find that histone H3R2C and R26C mutants reduce transcriptionally repressive H3K27me3. While H3K27me3 depletion in cells expressing these mutants is exclusively observed on the minor fraction of histone tails harboring the mutations, the same mutants recurrently disrupt broad H3K27me3 domains in the chromatin context, including near developmentally regulated promoters. H3K27me3 loss leads to de-repression of differentiation pathways, with concordant effects between H3R2 and H3R26 mutants despite different proximity to the PRC2 substrate, H3K27. Functionally, H3R26C-expressing mesenchymal progenitor cells and murine embryonic stem cell-derived teratomas demonstrate impaired differentiation. Collectively, these data show that cancer-associated H3 N-terminal arginine mutations reduce PRC2 activity and disrupt chromatin-dependent developmental functions, a cancer-relevant phenotype., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
22. ACK1 and BRK non-receptor tyrosine kinase deficiencies are associated with familial systemic lupus and involved in efferocytosis.
- Author
-
Guillet S, Lazarov T, Jordan N, Boisson B, Tello M, Craddock B, Zhou T, Nishi C, Bareja R, Yang H, Rieux-Laucat F, Lorenzo RIF, Dyall SD, Isenberg D, D'Cruz D, Lachmann N, Elemento O, Viale A, Socci ND, Abel L, Nagata S, Huse M, Miller WT, Casanova JL, and Geissmann F
- Abstract
Systemic Lupus Erythematosus (SLE) is an autoimmune disease, the pathophysiology and genetic basis of which are incompletely understood. Using a forward genetic screen in multiplex families with systemic lupus erythematosus (SLE) we identified an association between SLE and compound heterozygous deleterious variants in the non-receptor tyrosine kinases (NRTKs) ACK1 and BRK. Experimental blockade of ACK1 or BRK increased circulating autoantibodies in vivo in mice and exacerbated glomerular IgG deposits in an SLE mouse model. Mechanistically, non-receptor tyrosine kinases (NRTKs) regulate activation, migration, and proliferation of immune cells. We found that the patients' ACK1 and BRK variants impair efferocytosis, the MERTK-mediated anti-inflammatory response to apoptotic cells, in human induced Pluripotent Stem Cell (hiPSC)-derived macrophages, which may contribute to SLE pathogenesis. Overall, our data suggest that ACK1 and BRK deficiencies are associated with human SLE and impair efferocytosis in macrophages.
- Published
- 2024
- Full Text
- View/download PDF
23. Tumour-selective activity of RAS-GTP inhibition in pancreatic cancer.
- Author
-
Wasko UN, Jiang J, Dalton TC, Curiel-Garcia A, Edwards AC, Wang Y, Lee B, Orlen M, Tian S, Stalnecker CA, Drizyte-Miller K, Menard M, Dilly J, Sastra SA, Palermo CF, Hasselluhn MC, Decker-Farrell AR, Chang S, Jiang L, Wei X, Yang YC, Helland C, Courtney H, Gindin Y, Muonio K, Zhao R, Kemp SB, Clendenin C, Sor R, Vostrejs WP, Hibshman PS, Amparo AM, Hennessey C, Rees MG, Ronan MM, Roth JA, Brodbeck J, Tomassoni L, Bakir B, Socci ND, Herring LE, Barker NK, Wang J, Cleary JM, Wolpin BM, Chabot JA, Kluger MD, Manji GA, Tsai KY, Sekulic M, Lagana SM, Califano A, Quintana E, Wang Z, Smith JAM, Holderfield M, Wildes D, Lowe SW, Badgley MA, Aguirre AJ, Vonderheide RH, Stanger BZ, Baslan T, Der CJ, Singh M, and Olive KP
- Subjects
- Animals, Female, Humans, Mice, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Disease Models, Animal, DNA Copy Number Variations, Drug Resistance, Neoplasm drug effects, Genes, myc, Mice, Inbred BALB C, Mice, Inbred C57BL, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics, Treatment Outcome, Xenograft Model Antitumor Assays, Mutation, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Guanosine Triphosphate metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms pathology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Proto-Oncogene Proteins p21(ras) antagonists & inhibitors
- Abstract
Broad-spectrum RAS inhibition has the potential to benefit roughly a quarter of human patients with cancer whose tumours are driven by RAS mutations
1,2 . RMC-7977 is a highly selective inhibitor of the active GTP-bound forms of KRAS, HRAS and NRAS, with affinity for both mutant and wild-type variants3 . More than 90% of cases of human pancreatic ductal adenocarcinoma (PDAC) are driven by activating mutations in KRAS4 . Here we assessed the therapeutic potential of RMC-7977 in a comprehensive range of PDAC models. We observed broad and pronounced anti-tumour activity across models following direct RAS inhibition at exposures that were well-tolerated in vivo. Pharmacological analyses revealed divergent responses to RMC-7977 in tumour versus normal tissues. Treated tumours exhibited waves of apoptosis along with sustained proliferative arrest, whereas normal tissues underwent only transient decreases in proliferation, with no evidence of apoptosis. In the autochthonous KPC mouse model, RMC-7977 treatment resulted in a profound extension of survival followed by on-treatment relapse. Analysis of relapsed tumours identified Myc copy number gain as a prevalent candidate resistance mechanism, which could be overcome by combinatorial TEAD inhibition in vitro. Together, these data establish a strong preclinical rationale for the use of broad-spectrum RAS-GTP inhibition in the setting of PDAC and identify a promising candidate combination therapeutic regimen to overcome monotherapy resistance., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
24. Childhood cancer mutagenesis caused by transposase-derived PGBD5.
- Author
-
Yamada M, Keller RR, Gutierrez RL, Cameron D, Suzuki H, Sanghrajka R, Vaynshteyn J, Gerwin J, Maura F, Hooper W, Shah M, Robine N, Demarest P, Bayin NS, Zapater LJ, Reed C, Hébert S, Masilionis I, Chaligne R, Socci ND, Taylor MD, Kleinman CL, Joyner AL, Raju GP, and Kentsis A
- Subjects
- Humans, Child, Animals, Mice, Transposases genetics, Transposases metabolism, Hedgehog Proteins metabolism, Transcription Factors genetics, Mutagenesis, Medulloblastoma genetics, Cerebellar Neoplasms genetics
- Abstract
Genomic rearrangements are a hallmark of most childhood tumors, including medulloblastoma, one of the most common brain tumors in children, but their causes remain largely unknown. Here, we show that PiggyBac transposable element derived 5 (Pgbd5) promotes tumor development in multiple developmentally accurate mouse models of Sonic Hedgehog (SHH) medulloblastoma. Most Pgbd5-deficient mice do not develop tumors, while maintaining normal cerebellar development. Ectopic activation of SHH signaling is sufficient to enforce cerebellar granule cell progenitor-like cell states, which exhibit Pgbd5-dependent expression of distinct DNA repair and neurodevelopmental factors. Mouse medulloblastomas expressing Pgbd5 have increased numbers of somatic structural DNA rearrangements, some of which carry PGBD5-specific sequences at their breakpoints. Similar sequence breakpoints recurrently affect somatic DNA rearrangements of known tumor suppressors and oncogenes in medulloblastomas in 329 children. This identifies PGBD5 as a medulloblastoma mutator and provides a genetic mechanism for the generation of oncogenic DNA rearrangements in childhood cancer.
- Published
- 2024
- Full Text
- View/download PDF
25. Immune-Related Colitis Is Associated with Fecal Microbial Dysbiosis and Can Be Mitigated by Fecal Microbiota Transplantation.
- Author
-
Elkrief A, Waters NR, Smith N, Dai A, Slingerland J, Aleynick N, Febles B, Gogia P, Socci ND, Lumish M, Giardina PA, Chaft JE, Eng J, Motzer RJ, Mendelsohn RB, Markey KA, Zhuang M, Li Y, Yang Z, Hollmann TJ, Rudin CM, van den Brink MRM, Shia J, DeWolf S, Schoenfeld AJ, Hellmann MD, Babady NE, Faleck DM, and Peled JU
- Subjects
- Humans, Fecal Microbiota Transplantation adverse effects, Prospective Studies, Dysbiosis therapy, Dysbiosis etiology, Treatment Outcome, Gastrointestinal Microbiome, Colitis therapy, Colitis complications, Biological Products
- Abstract
Colitis induced by treatment with immune-checkpoint inhibitors (ICI), termed irColitis, is a substantial cause of morbidity complicating cancer treatment. We hypothesized that abnormal fecal microbiome features would be present at the time of irColitis onset and that restoring the microbiome with fecal transplant from a healthy donor would mitigate disease severity. Herein, we present fecal microbiota profiles from 18 patients with irColitis from a single center, 5 of whom were treated with healthy-donor fecal microbial transplantation (FMT). Although fecal samples collected at onset of irColitis had comparable α-diversity to that of comparator groups with gastrointestinal symptoms, irColitis was characterized by fecal microbial dysbiosis. Abundances of Proteobacteria were associated with irColitis in multivariable analyses. Five patients with irColitis refractory to steroids and biologic anti-inflammatory agents received healthy-donor FMT, with initial clinical improvement in irColitis symptoms observed in four of five patients. Two subsequently exhibited recurrence of irColitis symptoms following courses of antibiotics. Both received a second "salvage" FMT that was, again, followed by clinical improvement of irColitis. In summary, we observed distinct microbial community changes that were present at the time of irColitis onset. FMT was followed by clinical improvements in several cases of steroid- and biologic-agent-refractory irColitis. Strategies to restore or prevent microbiome dysbiosis in the context of immunotherapy toxicities should be further explored in prospective clinical trials., (©2023 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
26. PDGFRβ Signaling Cooperates with β-Catenin to Modulate c-Abl and Biologic Behavior of Desmoid-Type Fibromatosis.
- Author
-
Hu J, Hameed MR, Agaram NP, Whiting KA, Qin LX, Villano AM, O'Connor RB, Rozenberg JM, Cohen S, Prendergast K, Kryeziu S, White RL Jr, Posner MC, Socci ND, Gounder MM, Singer S, and Crago AM
- Subjects
- Humans, beta Catenin genetics, beta Catenin metabolism, Sorafenib pharmacology, Signal Transduction, Fibromatosis, Aggressive drug therapy, Fibromatosis, Aggressive genetics, Biological Products
- Abstract
Purpose: This study sought to identify β-catenin targets that regulate desmoid oncogenesis and determine whether external signaling pathways, particularly those inhibited by sorafenib (e.g., PDGFRβ), affect these targets to alter natural history or treatment response in patients., Experimental Design: In vitro experiments utilized primary desmoid cell lines to examine regulation of β-catenin targets. Relevance of results was assessed in vivo using Alliance trial A091105 correlative biopsies., Results: CTNNB1 knockdown inhibited hypoxia-regulated gene expression in vitro and reduced levels of HIF1α protein. ChIP-seq identified ABL1 as a β-catenin transcriptional target that modulated HIF1α and desmoid cell proliferation. Abrogation of either CTNNB1 or HIF1A inhibited desmoid cell-induced VEGFR2 phosphorylation and tube formation in endothelial cell co-cultures. Sorafenib inhibited this activity directly but also reduced HIF1α protein expression and c-Abl activity while inhibiting PDGFRβ signaling in desmoid cells. Conversely, c-Abl activity and desmoid cell proliferation were positively regulated by PDGF-BB. Reduction in PDGFRβ and c-Abl phosphorylation was commonly observed in biopsy samples from patients after treatment with sorafenib; markers of PDGFRβ/c-Abl pathway activation in baseline samples were associated with tumor progression in patients on the placebo arm and response to sorafenib in patients receiving treatment., Conclusions: The β-catenin transcriptional target ABL1 is necessary for proliferation and maintenance of HIF1α in desmoid cells. Regulation of c-Abl activity by PDGF signaling and targeted therapies modulates desmoid cell proliferation, thereby suggesting a reason for variable biologic behavior between tumors, a mechanism for sorafenib activity in desmoids, and markers predictive of outcome in patients., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
27. Tissue-specific features of the T cell repertoire after allogeneic hematopoietic cell transplantation in human and mouse.
- Author
-
DeWolf S, Elhanati Y, Nichols K, Waters NR, Nguyen CL, Slingerland JB, Rodriguez N, Lyudovyk O, Giardina PA, Kousa AI, Andrlová H, Ceglia N, Fei T, Kappagantula R, Li Y, Aleynick N, Baez P, Murali R, Hayashi A, Lee N, Gipson B, Rangesa M, Katsamakis Z, Dai A, Blouin AG, Arcila M, Masilionis I, Chaligne R, Ponce DM, Landau HJ, Politikos I, Tamari R, Hanash AM, Jenq RR, Giralt SA, Markey KA, Zhang Y, Perales MA, Socci ND, Greenbaum BD, Iacobuzio-Donahue CA, Hollmann TJ, van den Brink MRM, and Peled JU
- Subjects
- Humans, Mice, Animals, T-Lymphocytes pathology, Receptors, Antigen, T-Cell, Hematopoietic Stem Cell Transplantation, Graft vs Host Disease pathology
- Abstract
T cells are the central drivers of many inflammatory diseases, but the repertoire of tissue-resident T cells at sites of pathology in human organs remains poorly understood. We examined the site-specificity of T cell receptor (TCR) repertoires across tissues (5 to 18 tissues per patient) in prospectively collected autopsies of patients with and without graft-versus-host disease (GVHD), a potentially lethal tissue-targeting complication of allogeneic hematopoietic cell transplantation, and in mouse models of GVHD. Anatomic similarity between tissues was a key determinant of TCR repertoire composition within patients, independent of disease or transplant status. The T cells recovered from peripheral blood and spleens in patients and mice captured a limited portion of the TCR repertoire detected in tissues. Whereas few T cell clones were shared across patients, motif-based clustering revealed shared repertoire signatures across patients in a tissue-specific fashion. T cells at disease sites had a tissue-resident phenotype and were of donor origin based on single-cell chimerism analysis. These data demonstrate the complex composition of T cell populations that persist in human tissues at the end stage of an inflammatory disorder after lymphocyte-directed therapy. These findings also underscore the importance of studying T cell in tissues rather than blood for tissue-based pathologies and suggest the tissue-specific nature of both the endogenous and posttransplant T cell landscape.
- Published
- 2023
- Full Text
- View/download PDF
28. Overcoming Barriers to Tumor Genomic Profiling through Direct-to-Patient Outreach.
- Author
-
Doe-Tetteh SA, Camp SY, Reales D, Crowdis J, Noronha AM, Wolff B, Alano T, Galle J, Selcuklu SD, Viale A, Socci ND, Liu YL, Tew WP, Aghajanian C, Ladanyi M, He MX, AlDubayan SH, Mazor RD, Shpilberg O, Hershkovitz-Rokah O, Riancho JA, Hernandez JL, Gonzalez-Vela MC, Buthorn JJ, Wilson M, Webber AE, Yabe M, Petrova-Drus K, Rosenblum M, Durham BH, Abdel-Wahab O, Berger MF, Donoghue MTA, Kung AL, Glade Bender J, Shukla NN, Funt SA, Dogan A, Soslow RA, Al-Ahmadie H, Feldman DR, Van Allen EM, Diamond EL, and Solit DB
- Subjects
- Humans, Female, Mutation, Genomics, Exome, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics, Neoplasms, Germ Cell and Embryonal
- Abstract
Purpose: To overcome barriers to genomic testing for patients with rare cancers, we initiated a program to offer free clinical tumor genomic testing worldwide to patients with select rare cancer subtypes., Experimental Design: Patients were recruited through social media outreach and engagement with disease-specific advocacy groups, with a focus on patients with histiocytosis, germ cell tumors (GCT), and pediatric cancers. Tumors were analyzed using the MSK-IMPACT next-generation sequencing assay with the return of results to patients and their local physicians. Whole-exome recapture was performed for female patients with GCTs to define the genomic landscape of this rare cancer subtype., Results: A total of 333 patients were enrolled, and tumor tissue was received for 288 (86.4%), with 250 (86.8%) having tumor DNA of sufficient quality for MSK-IMPACT testing. Eighteen patients with histiocytosis have received genomically guided therapy to date, of whom 17 (94%) have had clinical benefit with a mean treatment duration of 21.7 months (range, 6-40+). Whole-exome sequencing of ovarian GCTs identified a subset with haploid genotypes, a phenotype rarely observed in other cancer types. Actionable genomic alterations were rare in ovarian GCT (28%); however, 2 patients with ovarian GCTs with squamous transformation had high tumor mutational burden, one of whom had a complete response to pembrolizumab., Conclusions: Direct-to-patient outreach can facilitate the assembly of cohorts of rare cancers of sufficient size to define their genomic landscape. By profiling tumors in a clinical laboratory, results could be reported to patients and their local physicians to guide treatment. See related commentary by Desai and Subbiah, p. 2339., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
29. Moving Spatially Resolved Multiplexed Protein Profiling toward Clinical Oncology.
- Author
-
Pourmaleki M, Socci ND, Hollmann TJ, and Mellinghoff IK
- Subjects
- Humans, Medical Oncology, Proteins, Neoplasms pathology
- Abstract
The shift in cancer therapy from broadly cytotoxic agents toward "personalized" treatments that target specific alterations in each patient's tumor requires diagnostic pathology approaches that are quantitative and biospecimen-friendly. Novel multiplexed antibody-based imaging technologies can measure single-cell expression of over 60 proteins in intact tumor sections and hold promise for clinical oncology., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
30. Innate immune signaling drives late cardiac toxicity following DNA-damaging cancer therapies.
- Author
-
Shamseddine A, Patel SH, Chavez V, Moore ZR, Adnan M, Di Bona M, Li J, Dang CT, Ramanathan LV, Oeffinger KC, Liu JE, Steingart RM, Piersigilli A, Socci ND, Chan AT, Yu AF, Bakhoum SF, and Schmitt AM
- Subjects
- Animals, Mice, Immunity, Innate, Inflammation, Nucleotidyltransferases genetics, Cardiotoxicity, Neoplasms drug therapy, DNA Damage, Antineoplastic Agents adverse effects
- Abstract
Late cardiac toxicity is a potentially lethal complication of cancer therapy, yet the pathogenic mechanism remains largely unknown, and few treatment options exist. Here we report DNA-damaging agents such as radiation and anthracycline chemotherapies inducing delayed cardiac inflammation following therapy due to activation of cGAS- and STING-dependent type I interferon signaling. Genetic ablation of cGAS-STING signaling in mice inhibits DNA damage-induced cardiac inflammation, rescues late cardiac functional decline, and prevents death from cardiac events. Treatment with a STING antagonist suppresses cardiac interferon signaling following DNA-damaging therapies and effectively mitigates cardiac toxicity. These results identify a therapeutically targetable, pathogenic mechanism for one of the most vexing treatment-related toxicities in cancer survivors., (© 2022 Shamseddine et al.)
- Published
- 2023
- Full Text
- View/download PDF
31. The endogenous repertoire harbors self-reactive CD4 + T cell clones that adopt a follicular helper T cell-like phenotype at steady state.
- Author
-
Lee V, Rodriguez DM, Ganci NK, Zeng S, Ai J, Chao JL, Walker MT, Miller CH, Klawon DEJ, Schoenbach MH, Kennedy DE, Maienschein-Cline M, Socci ND, Clark MR, and Savage PA
- Subjects
- Animals, Humans, Mice, Autoimmunity, Cell Differentiation, Clone Cells, Phenotype, T-Lymphocytes, Helper-Inducer, T Follicular Helper Cells, T-Lymphocytes, Regulatory, CD4-Positive T-Lymphocytes immunology
- Abstract
The T cell repertoire of healthy mice and humans harbors self-reactive CD4
+ conventional T (Tconv ) cells capable of inducing autoimmunity. Using T cell receptor profiling paired with in vivo clonal analysis of T cell differentiation, we identified Tconv cell clones that are recurrently enriched in non-lymphoid organs following ablation of Foxp3+ regulatory T (Treg ) cells. A subset of these clones was highly proliferative in the lymphoid organs at steady state and exhibited overt reactivity to self-ligands displayed by dendritic cells, yet were not purged by clonal deletion. These clones spontaneously adopted numerous hallmarks of follicular helper T (TFH ) cells, including expression of Bcl6 and PD-1, exhibited an elevated propensity to localize within B cell follicles at steady state, and produced interferon-γ in non-lymphoid organs following sustained Treg cell depletion. Our work identifies a naturally occurring population of self-reactive TFH -like cells and delineates a previously unappreciated fate for self-specific Tconv cells., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2023
- Full Text
- View/download PDF
32. Molecular Characterization of Acquired Resistance to KRASG12C-EGFR Inhibition in Colorectal Cancer.
- Author
-
Yaeger R, Mezzadra R, Sinopoli J, Bian Y, Marasco M, Kaplun E, Gao Y, Zhao H, Paula ADC, Zhu Y, Perez AC, Chadalavada K, Tse E, Chowdhry S, Bowker S, Chang Q, Qeriqi B, Weigelt B, Nanjangud GJ, Berger MF, Der-Torossian H, Anderes K, Socci ND, Shia J, Riely GJ, Murciano-Goroff YR, Li BT, Christensen JG, Reis-Filho JS, Solit DB, de Stanchina E, Lowe SW, Rosen N, and Misale S
- Subjects
- Animals, Humans, Signal Transduction, Disease Models, Animal, ErbB Receptors, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Mutation, Drug Resistance, Neoplasm genetics, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism
- Abstract
With the combination of KRASG12C and EGFR inhibitors, KRAS is becoming a druggable target in colorectal cancer. However, secondary resistance limits its efficacy. Using cell lines, patient-derived xenografts, and patient samples, we detected a heterogeneous pattern of putative resistance alterations expected primarily to prevent inhibition of ERK signaling by drugs at progression. Serial analysis of patient blood samples on treatment demonstrates that most of these alterations are detected at a low frequency except for KRASG12C amplification, a recurrent resistance mechanism that rises in step with clinical progression. Upon drug withdrawal, resistant cells with KRASG12C amplification undergo oncogene-induced senescence, and progressing patients experience a rapid fall in levels of this alteration in circulating DNA. In this new state, drug resumption is ineffective as mTOR signaling is elevated. However, our work exposes a potential therapeutic vulnerability, whereby therapies that target the senescence response may overcome acquired resistance., Significance: Clinical resistance to KRASG12C-EGFR inhibition primarily prevents suppression of ERK signaling. Most resistance mechanisms are subclonal, whereas KRASG12C amplification rises over time to drive a higher portion of resistance. This recurrent resistance mechanism leads to oncogene-induced senescence upon drug withdrawal and creates a potential vulnerability to senolytic approaches. This article is highlighted in the In This Issue feature, p. 1., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
33. Author Correction: FOXA1 repression drives lineage plasticity and immune heterogeneity in bladder cancers with squamous differentiation.
- Author
-
Warrick JI, Hu W, Yamashita H, Walter V, Shuman L, Craig JM, Gellert LL, Castro MAA, Robertson AG, Kuo F, Ostrovnaya I, Sarungbam J, Chen YB, Gopalan A, Sirintrapun SJ, Fine SW, Tickoo SK, Kim K, Thomas J, Karan N, Gao SP, Clinton TN, Lenis AT, Chan TA, Chen Z, Rao M, Hollman TJ, Li Y, Socci ND, Chavan S, Viale A, Mohibullah N, Bochner BH, Pietzak EJ, Teo MY, Iyer G, Rosenberg JE, Bajorin DF, Kaag M, Merrill SB, Joshi M, Adam R, Taylor JA 3rd, Clark PE, Raman JD, Reuter VE, Chen Y, Funt SA, Solit DB, DeGraff DJ, and Al-Ahmadie HA
- Published
- 2022
- Full Text
- View/download PDF
34. Increased tumor glycolysis is associated with decreased immune infiltration across human solid tumors.
- Author
-
Cohen IJ, Pareja F, Socci ND, Shen R, Doane AS, Schwartz J, Khanin R, Morris EA, Sutton EJ, and Blasberg RG
- Subjects
- Humans, Female, Immunotherapy, Glycolysis, Tumor Microenvironment, Carcinoma, Non-Small-Cell Lung, Lung Neoplasms, Breast Neoplasms
- Abstract
Response to immunotherapy across multiple cancer types is approximately 25%, with some tumor types showing increased response rates compared to others (i.e. response rates in melanoma and non-small cell lung cancer (NSCLC) are typically 30-60%). Patients whose tumors are resistant to immunotherapy often lack high levels of pre-existing inflammation in the tumor microenvironment. Increased tumor glycolysis, acting through glucose deprivation and lactic acid accumulation, has been shown to have pleiotropic immune suppressive effects using in-vitro and in-vivo models of disease. To determine whether the immune suppressive effect of tumor glycolysis is observed across human solid tumors, we analyzed glycolytic and immune gene expression patterns in multiple solid malignancies. We found that increased expression of a glycolytic signature was associated with decreased immune infiltration and a more aggressive disease across multiple tumor types. Radiologic and pathologic analysis of untreated estrogen receptor (ER)-negative breast cancers corroborated these observations, and demonstrated that protein expression of glycolytic enzymes correlates positively with glucose uptake and negatively with infiltration of CD3
+ and CD8+ lymphocytes. This study reveals an inverse relationship between tumor glycolysis and immune infiltration in a large cohort of multiple solid tumor types., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Cohen, Pareja, Socci, Shen, Doane, Schwartz, Khanin, Morris, Sutton and Blasberg.)- Published
- 2022
- Full Text
- View/download PDF
35. FOXA1 repression drives lineage plasticity and immune heterogeneity in bladder cancers with squamous differentiation.
- Author
-
Warrick JI, Hu W, Yamashita H, Walter V, Shuman L, Craig JM, Gellert LL, Castro MAA, Robertson AG, Kuo F, Ostrovnaya I, Sarungbam J, Chen YB, Gopalan A, Sirintrapun SJ, Fine SW, Tickoo SK, Kim K, Thomas J, Karan N, Gao SP, Clinton TN, Lenis AT, Chan TA, Chen Z, Rao M, Hollman TJ, Li Y, Socci ND, Chavan S, Viale A, Mohibullah N, Bochner BH, Pietzak EJ, Teo MY, Iyer G, Rosenberg JE, Bajorin DF, Kaag M, Merrill SB, Joshi M, Adam R, Taylor JA 3rd, Clark PE, Raman JD, Reuter VE, Chen Y, Funt SA, Solit DB, DeGraff DJ, and Al-Ahmadie HA
- Subjects
- Humans, Biomarkers, Tumor genetics, Hepatocyte Nuclear Factor 3-alpha genetics, Phylogeny, Cell Lineage, Carcinoma, Squamous Cell pathology, Carcinoma, Transitional Cell metabolism, Urinary Bladder Neoplasms pathology
- Abstract
Cancers arising from the bladder urothelium often exhibit lineage plasticity with regions of urothelial carcinoma adjacent to or admixed with regions of divergent histomorphology, most commonly squamous differentiation. To define the biologic basis for and clinical significance of this morphologic heterogeneity, here we perform integrated genomic analyses of mixed histology bladder cancers with separable regions of urothelial and squamous differentiation. We find that squamous differentiation is a marker of intratumoral genomic and immunologic heterogeneity in patients with bladder cancer and a biomarker of intrinsic immunotherapy resistance. Phylogenetic analysis confirms that in all cases the urothelial and squamous regions are derived from a common shared precursor. Despite the presence of marked genomic heterogeneity between co-existent urothelial and squamous differentiated regions, no recurrent genomic alteration exclusive to the urothelial or squamous morphologies is identified. Rather, lineage plasticity in bladder cancers with squamous differentiation is associated with loss of expression of FOXA1, GATA3, and PPARG, transcription factors critical for maintenance of urothelial cell identity. Of clinical significance, lineage plasticity and PD-L1 expression is coordinately dysregulated via FOXA1, with patients exhibiting morphologic heterogeneity pre-treatment significantly less likely to respond to immune checkpoint inhibitors., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
36. Ordered and deterministic cancer genome evolution after p53 loss.
- Author
-
Baslan T, Morris JP 4th, Zhao Z, Reyes J, Ho YJ, Tsanov KM, Bermeo J, Tian S, Zhang S, Askan G, Yavas A, Lecomte N, Erakky A, Varghese AM, Zhang A, Kendall J, Ghiban E, Chorbadjiev L, Wu J, Dimitrova N, Chadalavada K, Nanjangud GJ, Bandlamudi C, Gong Y, Donoghue MTA, Socci ND, Krasnitz A, Notta F, Leach SD, Iacobuzio-Donahue CA, and Lowe SW
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma pathology, Animals, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Evolution, Molecular, Gene Deletion, Mice, Models, Genetic, Carcinogenesis genetics, Carcinogenesis pathology, Disease Progression, Genes, p53 genetics, Genome genetics, Loss of Heterozygosity, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Tumor Suppressor Protein p53 genetics
- Abstract
Although p53 inactivation promotes genomic instability
1 and presents a route to malignancy for more than half of all human cancers2,3 , the patterns through which heterogenous TP53 (encoding human p53) mutant genomes emerge and influence tumorigenesis remain poorly understood. Here, in a mouse model of pancreatic ductal adenocarcinoma that reports sporadic p53 loss of heterozygosity before cancer onset, we find that malignant properties enabled by p53 inactivation are acquired through a predictable pattern of genome evolution. Single-cell sequencing and in situ genotyping of cells from the point of p53 inactivation through progression to frank cancer reveal that this deterministic behaviour involves four sequential phases-Trp53 (encoding mouse p53) loss of heterozygosity, accumulation of deletions, genome doubling, and the emergence of gains and amplifications-each associated with specific histological stages across the premalignant and malignant spectrum. Despite rampant heterogeneity, the deletion events that follow p53 inactivation target functionally relevant pathways that can shape genomic evolution and remain fixed as homogenous events in diverse malignant populations. Thus, loss of p53-the 'guardian of the genome'-is not merely a gateway to genetic chaos but, rather, can enable deterministic patterns of genome evolution that may point to new strategies for the treatment of TP53-mutant tumours., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
37. Clinical sequencing of soft tissue and bone sarcomas delineates diverse genomic landscapes and potential therapeutic targets.
- Author
-
Nacev BA, Sanchez-Vega F, Smith SA, Antonescu CR, Rosenbaum E, Shi H, Tang C, Socci ND, Rana S, Gularte-Mérida R, Zehir A, Gounder MM, Bowler TG, Luthra A, Jadeja B, Okada A, Strong JA, Stoller J, Chan JE, Chi P, D'Angelo SP, Dickson MA, Kelly CM, Keohan ML, Movva S, Thornton K, Meyers PA, Wexler LH, Slotkin EK, Glade Bender JL, Shukla NN, Hensley ML, Healey JH, La Quaglia MP, Alektiar KM, Crago AM, Yoon SS, Untch BR, Chiang S, Agaram NP, Hameed MR, Berger MF, Solit DB, Schultz N, Ladanyi M, Singer S, and Tap WD
- Subjects
- Genomics, Humans, Bone Neoplasms, Osteosarcoma, Sarcoma drug therapy, Sarcoma therapy, Soft Tissue Neoplasms genetics
- Abstract
The genetic, biologic, and clinical heterogeneity of sarcomas poses a challenge for the identification of therapeutic targets, clinical research, and advancing patient care. Because there are > 100 sarcoma subtypes, in-depth genetic studies have focused on one or a few subtypes. Herein, we report a comparative genetic analysis of 2,138 sarcomas representing 45 pathological entities. This cohort is prospectively analyzed using targeted sequencing to characterize subtype-specific somatic alterations in targetable pathways, rates of whole genome doubling, mutational signatures, and subtype-agnostic genomic clusters. The most common alterations are in cell cycle control and TP53, receptor tyrosine kinases/PI3K/RAS, and epigenetic regulators. Subtype-specific associations include TERT amplification in intimal sarcoma and SWI/SNF alterations in uterine adenosarcoma. Tumor mutational burden, while low compared to other cancers, varies between and within subtypes. This resource will improve sarcoma models, motivate studies of subtype-specific alterations, and inform investigations of genetic factors and their correlations with treatment response., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
38. Genomic and transcriptomic analysis of a library of small cell lung cancer patient-derived xenografts.
- Author
-
Caeser R, Egger JV, Chavan S, Socci ND, Jones CB, Kombak FE, Asher M, Roehrl MH, Shah NS, Allaj V, Manoj P, Tischfield SE, Kulick A, Meneses M, Iacobuzio-Donahue CA, Lai WV, Bhanot U, Baine MK, Rekhtman N, Hollmann TJ, de Stanchina E, Poirier JT, Rudin CM, and Sen T
- Subjects
- Heterografts, Humans, Proteomics, Transcriptome genetics, Lung Neoplasms genetics, Lung Neoplasms pathology, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma pathology
- Abstract
Access to clinically relevant small cell lung cancer (SCLC) tissue is limited because surgical resection is rare in metastatic SCLC. Patient-derived xenografts (PDX) and circulating tumor cell-derived xenografts (CDX) have emerged as valuable tools to characterize SCLC. Here, we present a resource of 46 extensively annotated PDX/CDX models derived from 33 patients with SCLC. We perform multi-omic analyses, using targeted tumor next-generation sequencing, RNA-sequencing, and immunohistochemistry to deconvolute the mutational landscapes, global expression profiles, and molecular subtypes of these SCLC models. SCLC subtypes characterized by transcriptional regulators, ASCL1, NEUROD1 and POU2F3 are confirmed in this cohort. A subset of SCLC clinical specimens, including matched PDX/CDX and clinical specimen pairs, confirm that the primary features and genomic and proteomic landscapes of the tumors of origin are preserved in the derivative PDX models. This resource provides a powerful system to study SCLC biology., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
39. Tumor MHC Class I Expression Associates with Intralesional IL2 Response in Melanoma.
- Author
-
Pourmaleki M, Jones CJ, Ariyan CE, Zeng Z, Pirun M, Navarrete DA, Li Y, Zhang M, Nandakumar S, Campos C, Nadeem S, Klimstra DS, Temple-Oberle CF, Brenn T, Lipson EJ, Schenk KM, Stein JE, Taube JM, White MG, Traweek R, Wargo JA, Kirkwood JM, Gasmi B, Goff SL, Corwin AD, McDonough E, Ginty F, Callahan MK, Schietinger A, Socci ND, Mellinghoff IK, and Hollmann TJ
- Subjects
- Hepatitis A Virus Cellular Receptor 2, Humans, Immunotherapy methods, Programmed Cell Death 1 Receptor metabolism, Interleukin-2 therapeutic use, Melanoma drug therapy, Melanoma genetics, Melanoma pathology
- Abstract
Cancer immunotherapy can result in lasting tumor regression, but predictive biomarkers of treatment response remain ill-defined. Here, we performed single-cell proteomics, transcriptomics, and genomics on matched untreated and IL2 injected metastases from patients with melanoma. Lesions that completely regressed following intralesional IL2 harbored increased fractions and densities of nonproliferating CD8+ T cells lacking expression of PD-1, LAG-3, and TIM-3 (PD-1-LAG-3-TIM-3-). Untreated lesions from patients who subsequently responded with complete eradication of all tumor cells in all injected lesions (individuals referred to herein as "extreme responders") were characterized by proliferating CD8+ T cells with an exhausted phenotype (PD-1+LAG-3+TIM-3+), stromal B-cell aggregates, and expression of IFNγ and IL2 response genes. Loss of membranous MHC class I expression in tumor cells of untreated lesions was associated with resistance to IL2 therapy. We validated this finding in an independent cohort of metastatic melanoma patients treated with intralesional or systemic IL2. Our study suggests that intact tumor-cell antigen presentation is required for melanoma response to IL2 and describes a multidimensional and spatial approach to develop immuno-oncology biomarker hypotheses using routinely collected clinical biospecimens., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
40. TCR signal strength defines distinct mechanisms of T cell dysfunction and cancer evasion.
- Author
-
Shakiba M, Zumbo P, Espinosa-Carrasco G, Menocal L, Dündar F, Carson SE, Bruno EM, Sanchez-Rivera FJ, Lowe SW, Camara S, Koche RP, Reuter VP, Socci ND, Whitlock B, Tamzalit F, Huse M, Hellmann MD, Wells DK, Defranoux NA, Betel D, Philip M, and Schietinger A
- Subjects
- Animals, Antigens, Neoplasm immunology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Cell Line, Tumor, Cytokines metabolism, Disease Models, Animal, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Humans, Immunotherapy, Adoptive methods, Lymphocyte Activation immunology, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Lymphocytes, Tumor-Infiltrating pathology, Mice, Neoplasms pathology, Neoplasms therapy, T-Cell Antigen Receptor Specificity, Neoplasms etiology, Neoplasms metabolism, Receptors, Antigen, T-Cell metabolism, Signal Transduction, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Tumor Escape
- Abstract
T cell receptor (TCR) signal strength is a key determinant of T cell responses. We developed a cancer mouse model in which tumor-specific CD8 T cells (TST cells) encounter tumor antigens with varying TCR signal strength. High-signal-strength interactions caused TST cells to up-regulate inhibitory receptors (IRs), lose effector function, and establish a dysfunction-associated molecular program. TST cells undergoing low-signal-strength interactions also up-regulated IRs, including PD1, but retained a cell-intrinsic functional state. Surprisingly, neither high- nor low-signal-strength interactions led to tumor control in vivo, revealing two distinct mechanisms by which PD1hi TST cells permit tumor escape; high signal strength drives dysfunction, while low signal strength results in functional inertness, where the signal strength is too low to mediate effective cancer cell killing by functional TST cells. CRISPR-Cas9-mediated fine-tuning of signal strength to an intermediate range improved anti-tumor activity in vivo. Our study defines the role of TCR signal strength in TST cell function, with important implications for T cell-based cancer immunotherapies., Competing Interests: Disclosures: N.A. Defranoux reports personal fees from Alector outside the submitted work. M.D. Hellman receives institutional research funding from Bristol-Myers Squibb; has been a compensated consultant for Merck, Bristol-Myers Squibb, AstraZeneca, Genentech/Roche, Nektar, Syndax, Mirati, Shattuck Labs, Immunai, Blueprint Medicines, Achilles, and Arcus; received travel support/honoraria from AstraZeneca, Eli Lilly, and Bristol-Myers Squibb; has options from Factorial, Shattuck Labs, Immunai, and Arcus; reported personal fees from Adicet, DaVolterra, Genzyme/Sanofi, Janssen, Immunai, Instil Bio, Mana Therapeutics, Natera, Pact Pharms, Chattuck Labs, and Regenron out side the submitted work; and has a patent filed by his institution related to the use of tumor mutation burden to predict response to immunotherapy (PCT/US2015/062208), which has received licensing fees from PGDx. D.K. Wells is a founder, equity holder, and receives advisory fees from Immunai. No other disclosures were reported., (© 2021 Shakiba et al.)
- Published
- 2022
- Full Text
- View/download PDF
41. Recurrent Mutations in Cyclin D3 Confer Clinical Resistance to FLT3 Inhibitors in Acute Myeloid Leukemia.
- Author
-
Smith CC, Viny AD, Massi E, Kandoth C, Socci ND, Rapaport F, Najm M, Medina-Martinez JS, Papaemmanuil E, Tarver TC, Hsu HH, Le MH, West B, Bollag G, Taylor BS, Levine RL, and Shah NP
- Subjects
- Cell Line, Tumor, Humans, Aminopyridines therapeutic use, Cyclin D3 genetics, Drug Resistance, Neoplasm genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Mutation, Protein Kinase Inhibitors therapeutic use, Pyrroles therapeutic use, fms-Like Tyrosine Kinase 3 antagonists & inhibitors
- Abstract
Purpose: Biomarkers of response and resistance to FLT3 tyrosine kinase inhibitors (TKI) are still emerging, and optimal clinical combinations remain unclear. The purpose of this study is to identify co-occurring mutations that influence clinical response to the novel FLT3 inhibitor pexidartinib (PLX3397)., Experimental Design: We performed targeted sequencing of pretreatment blasts from 29 patients with FLT3 internal tandem duplication (ITD) mutations treated on the phase I/II trial of pexidartinib in relapsed/refractory FLT3 -ITD+ acute myeloid leukemia (AML). We sequenced 37 samples from 29 patients with available material, including 8 responders and 21 non-responders treated at or above the recommended phase II dose of 3,000 mg., Results: Consistent with other studies, we identified mutations in NRAS, TP53, IDH2 , and a variety of epigenetic and transcriptional regulators only in non-responders. Among the most frequently mutated genes in non-responders was Cyclin D3 ( CCND3) . A total of 3 individual mutations in CCND3 (Q276*, S264R, and T283A) were identified in 2 of 21 non-responders (one patient had both Q276* and S264R). No CCND3 mutations were found in pexidartinib responders. Expression of the Q276* and T283A mutations in FLT3 -ITD MV4;11 cells conferred resistance to apoptosis, decreased cell-cycle arrest, and increased proliferation in the presence of pexidartinib and other FLT3 inhibitors. Inhibition of CDK4/6 activity in CCND3 mutant MV4;11 cells restored pexidartinib-induced cell-cycle arrest but not apoptosis., Conclusions: Mutations in CCND3 , a gene not commonly mutated in AML, are a novel cause of clinical primary resistance to FLT3 inhibitors in AML and may have sensitivity to CDK4/6 inhibition., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
42. Tumor fraction-guided cell-free DNA profiling in metastatic solid tumor patients.
- Author
-
Tsui DWY, Cheng ML, Shady M, Yang JL, Stephens D, Won H, Srinivasan P, Huberman K, Meng F, Jing X, Patel J, Hasan M, Johnson I, Gedvilaite E, Houck-Loomis B, Socci ND, Selcuklu SD, Seshan VE, Zhang H, Chakravarty D, Zehir A, Benayed R, Arcila M, Ladanyi M, Funt SA, Feldman DR, Li BT, Razavi P, Rosenberg J, Bajorin D, Iyer G, Abida W, Scher HI, Rathkopf D, Viale A, Berger MF, and Solit DB
- Subjects
- DNA Copy Number Variations, Genomics methods, Humans, Mutation, ROC Curve, Exome Sequencing, Whole Genome Sequencing, Biomarkers, Tumor, Circulating Tumor DNA, Liquid Biopsy methods, Neoplasms diagnosis, Neoplasms genetics
- Abstract
Background: Cell-free DNA (cfDNA) profiling is increasingly used to guide cancer care, yet mutations are not always identified. The ability to detect somatic mutations in plasma depends on both assay sensitivity and the fraction of circulating DNA in plasma that is tumor-derived (i.e., cfDNA tumor fraction). We hypothesized that cfDNA tumor fraction could inform the interpretation of negative cfDNA results and guide the choice of subsequent assays of greater genomic breadth or depth., Methods: Plasma samples collected from 118 metastatic cancer patients were analyzed with cf-IMPACT, a modified version of the FDA-authorized MSK-IMPACT tumor test that can detect genomic alterations in 410 cancer-associated genes. Shallow whole genome sequencing (sWGS) was also performed in the same samples to estimate cfDNA tumor fraction based on genome-wide copy number alterations using z-score statistics. Plasma samples with no somatic alterations detected by cf-IMPACT were triaged based on sWGS-estimated tumor fraction for analysis with either a less comprehensive but more sensitive assay (MSK-ACCESS) or broader whole exome sequencing (WES)., Results: cfDNA profiling using cf-IMPACT identified somatic mutations in 55/76 (72%) patients for whom MSK-IMPACT tumor profiling data were available. A significantly higher concordance of mutational profiles and tumor mutational burden (TMB) was observed between plasma and tumor profiling for plasma samples with a high tumor fraction (z-score≥5). In the 42 patients from whom tumor data was not available, cf-IMPACT identified mutations in 16/42 (38%). In total, cf-IMPACT analysis of plasma revealed mutations in 71/118 (60%) patients, with clinically actionable alterations identified in 30 (25%), including therapeutic targets of FDA-approved drugs. Of the 47 samples without alterations detected and low tumor fraction (z-score<5), 29 had sufficient material to be re-analyzed using a less comprehensive but more sensitive assay, MSK-ACCESS, which revealed somatic mutations in 14/29 (48%). Conversely, 5 patients without alterations detected by cf-IMPACT and with high tumor fraction (z-score≥5) were analyzed by WES, which identified mutational signatures and alterations in potential oncogenic drivers not covered by the cf-IMPACT panel. Overall, we identified mutations in 90/118 (76%) patients in the entire cohort using the three complementary plasma profiling approaches., Conclusions: cfDNA tumor fraction can inform the interpretation of negative cfDNA results and guide the selection of subsequent sequencing platforms that are most likely to identify clinically-relevant genomic alterations.
- Published
- 2021
- Full Text
- View/download PDF
43. Impact of tumor heterogeneity and microenvironment in identifying neoantigens in a patient with ovarian cancer.
- Author
-
Dao T, Klatt MG, Korontsvit T, Mun SS, Guzman S, Mattar M, Zivanovic O, Kyi CK, Socci ND, O'Cearbhaill RE, and Scheinberg DA
- Subjects
- Antigens, Neoplasm genetics, Cells, Cultured, Epitopes, T-Lymphocyte genetics, Female, HLA-A2 Antigen genetics, Humans, Middle Aged, Neoplasm Staging, Tumor Microenvironment, Tumor Suppressor Protein p53 genetics, Exome Sequencing, Antigens, Neoplasm metabolism, Epitopes, T-Lymphocyte metabolism, HLA-A2 Antigen metabolism, Immunotherapy, Adoptive methods, Mutation genetics, Ovarian Neoplasms immunology, T-Lymphocytes immunology, Tumor Suppressor Protein p53 metabolism
- Abstract
Identification of neoepitopes as tumor-specific targets remains challenging, especially for cancers with low mutational burden, such as ovarian cancer. To identify mutated human leukocyte antigen (HLA) ligands as potential targets for immunotherapy in ovarian cancer, we combined mass spectrometry analysis of the major histocompatibility complex (MHC) class I peptidomes of ovarian cancer cells with parallel sequencing of whole exome and RNA in a patient with high-grade serous ovarian cancer. Four of six predicted mutated epitopes capable of binding to HLA-A*02:01 induced peptide-specific T cell responses in blood from healthy donors. In contrast, all six peptides failed to induce autologous peptide-specific response by T cells in peripheral blood or tumor-infiltrating lymphocytes from ascites of the patient. Surprisingly, T cell responses against a low-affinity p53-mutant Y220C epitope were consistently detected in the patient with either unprimed or in vitro peptide-stimulated T cells even though the patient's primary tumor did not bear this mutation. Our results demonstrated that tumor heterogeneity and distinct immune microenvironments within a patient should be taken into consideration for identification of immunogenic neoantigens. T cell responses to a driver gene-derived p53 Y220C mutation in ovarian cancer warrant further study.
- Published
- 2021
- Full Text
- View/download PDF
44. The Genetic Evolution of Treatment-Resistant Cutaneous, Acral, and Uveal Melanomas.
- Author
-
Makohon-Moore AP, Lipson EJ, Hooper JE, Zucker A, Hong J, Bielski CM, Hayashi A, Tokheim C, Baez P, Kappagantula R, Kohutek Z, Makarov V, Riaz N, Postow MA, Chapman PB, Karchin R, Socci ND, Solit DB, Chan TA, Taylor BS, Topalian SL, and Iacobuzio-Donahue CA
- Subjects
- Biomarkers, Tumor, Humans, Melanoma drug therapy, Melanoma genetics, Melanoma immunology, Prognosis, Skin Neoplasms drug therapy, Skin Neoplasms genetics, Skin Neoplasms immunology, Uveal Neoplasms drug therapy, Uveal Neoplasms genetics, Uveal Neoplasms immunology, Drug Resistance, Neoplasm genetics, Evolution, Molecular, Immunotherapy methods, Melanoma pathology, Mutation, Skin Neoplasms pathology, Uveal Neoplasms pathology
- Abstract
Purpose: Melanoma is a biologically heterogeneous disease composed of distinct clinicopathologic subtypes that frequently resist treatment. To explore the evolution of treatment resistance and metastasis, we used a combination of temporal and multilesional tumor sampling in conjunction with whole-exome sequencing of 110 tumors collected from 7 patients with cutaneous ( n = 3), uveal ( n = 2), and acral ( n = 2) melanoma subtypes., Experimental Design: Primary tumors, metastases collected longitudinally, and autopsy tissues were interrogated. All but 1 patient died because of melanoma progression., Results: For each patient, we generated phylogenies and quantified the extent of genetic diversity among tumors, specifically among putative somatic alterations affecting therapeutic resistance., Conclusions: In 4 patients who received immunotherapy, we found 1-3 putative acquired and intrinsic resistance mechanisms coexisting in the same patient, including mechanisms that were shared by all tumors within each patient, suggesting that future therapies directed at overcoming intrinsic resistance mechanisms may be broadly effective., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
45. Biphasic Activation of WNT Signaling Facilitates the Derivation of Midbrain Dopamine Neurons from hESCs for Translational Use.
- Author
-
Kim TW, Piao J, Koo SY, Kriks S, Chung SY, Betel D, Socci ND, Choi SJ, Zabierowski S, Dubose BN, Hill EJ, Mosharov EV, Irion S, Tomishima MJ, Tabar V, and Studer L
- Subjects
- Animals, Cell Differentiation, Mesencephalon, Rats, Wnt Signaling Pathway, Dopaminergic Neurons, Human Embryonic Stem Cells
- Abstract
Human pluripotent stem cells show considerable promise for applications in regenerative medicine, including the development of cell replacement paradigms for the treatment of Parkinson's disease. Protocols have been developed to generate authentic midbrain dopamine (mDA) neurons capable of reversing dopamine-related deficits in animal models of Parkinson's disease. However, the generation of mDA neurons at clinical scale suitable for human application remains an important challenge. Here, we present an mDA neuron derivation protocol based on a two-step WNT signaling activation strategy that improves expression of midbrain markers, such as Engrailed-1 (EN1), while minimizing expression of contaminating posterior (hindbrain) and anterior (diencephalic) lineage markers. The resulting neurons exhibit molecular, biochemical, and electrophysiological properties of mDA neurons. Cryopreserved mDA neuron precursors can be successfully transplanted into 6-hydroxydopamine (6OHDA) lesioned rats to induce recovery of amphetamine-induced rotation behavior. The protocol presented here is the basis for clinical-grade mDA neuron production and preclinical safety and efficacy studies., Competing Interests: Declaration of Interests L.S. is a scientific co-founder and consultant, and V.T. is a founding investigator and consultant, of Bluerock Therapeutics, Inc. M.T. and S.I. are employed by BlueRock Therapeutics, Inc. L.S., S.K., S.I., and M.T. are inventors of a patent WO2016196661A1 filed by Memorial Sloan Kettering Cancer Center on the methods described in this study. All other authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
46. Author Correction: PGBD5 promotes site-specific oncogenic mutations in human tumors.
- Author
-
Henssen AG, Koche R, Zhuang J, Jiang E, Reed C, Eisenberg A, Still E, MacArthur IC, Rodríguez-Fos E, Gonzalez S, Puiggròs M, Blackford AN, Mason CE, de Stanchina E, Gönen M, Emde AK, Shah M, Arora K, Reeves C, Socci ND, Perlman E, Antonescu CR, Roberts CWM, Steen H, Mullen E, Jackson SP, Torrents D, Weng Z, Armstrong SA, and Kentsis A
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
47. Rb and p53-Deficient Myxofibrosarcoma and Undifferentiated Pleomorphic Sarcoma Require Skp2 for Survival.
- Author
-
Li GZ, Okada T, Kim YM, Agaram NP, Sanchez-Vega F, Shen Y, Tsubokawa N, Rios J, Martin AS, Dickson MA, Qin LX, Socci ND, and Singer S
- Subjects
- Adult, Animals, Comparative Genomic Hybridization, Humans, Mice, Tumor Suppressor Protein p53 genetics, Fibrosarcoma genetics, Sarcoma genetics, Soft Tissue Neoplasms
- Abstract
Myxofibrosarcoma (MFS) and undifferentiated pleomorphic sarcoma (UPS) are highly genetically complex soft tissue sarcomas. Up to 50% of patients develop distant metastases, but current systemic therapies have limited efficacy. MFS and UPS have recently been shown to commonly harbor copy number alterations or mutations in the tumor suppressor genes RB1 and TP53 . As these alterations have been shown to engender dependence on the oncogenic protein Skp2 for survival of transformed cells in mouse models, we sought to examine its function and potential as a therapeutic target in MFS/UPS. Comparative genomic hybridization and next-generation sequencing confirmed that a significant fraction of MFS and UPS patient samples ( n = 94) harbor chromosomal deletions and/or loss-of-function mutations in RB1 and TP53 (88% carry alterations in at least one gene; 60% carry alterations in both). Tissue microarray analysis identified a correlation between absent Rb and p53 expression and positive expression of Skp2. Downregulation of Skp2 or treatment with the Skp2-specific inhibitor C1 revealed that Skp2 drives proliferation of patient-derived MFS/UPS cell lines deficient in both Rb and p53 by degrading p21 and p27. Inhibition of Skp2 using the neddylation-activating enzyme inhibitor pevonedistat decreased growth of Rb/p53-negative patient-derived cell lines and mouse xenografts. These results demonstrate that loss of both Rb and p53 renders MFS and UPS dependent on Skp2, which can be therapeutically exploited and could provide the basis for promising novel systemic therapies for MFS and UPS. SIGNIFICANCE: Loss of both Rb and p53 renders myxofibrosarcoma and undifferentiated pleomorphic sarcoma dependent on Skp2, which could provide the basis for promising novel systemic therapies. See related commentary by Lambert and Jones, p. 2437 ., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
48. The Evolutionary Origins of Recurrent Pancreatic Cancer.
- Author
-
Sakamoto H, Attiyeh MA, Gerold JM, Makohon-Moore AP, Hayashi A, Hong J, Kappagantula R, Zhang L, Melchor JP, Reiter JG, Heyde A, Bielski CM, Penson AV, Gönen M, Chakravarty D, O'Reilly EM, Wood LD, Hruban RH, Nowak MA, Socci ND, Taylor BS, and Iacobuzio-Donahue CA
- Subjects
- Carcinoma, Pancreatic Ductal secondary, Evolution, Molecular, Humans, Neoplasm Recurrence, Local pathology, Pancreatic Neoplasms pathology, Exome Sequencing, Carcinoma, Pancreatic Ductal genetics, Neoplasm Metastasis genetics, Neoplasm Recurrence, Local genetics, Pancreatic Neoplasms genetics
- Abstract
Surgery is the only curative option for stage I/II pancreatic cancer; nonetheless, most patients will experience a recurrence after surgery and die of their disease. To identify novel opportunities for management of recurrent pancreatic cancer, we performed whole-exome or targeted sequencing of 10 resected primary cancers and matched intrapancreatic recurrences or distant metastases. We identified that recurrent disease after adjuvant or first-line platinum therapy corresponds to an increased mutational burden. Recurrent disease is enriched for genetic alterations predicted to activate MAPK/ERK and PI3K-AKT signaling and develops from a monophyletic or polyphyletic origin. Treatment-induced genetic bottlenecks lead to a modified genetic landscape and subclonal heterogeneity for driver gene alterations in part due to intermetastatic seeding. In 1 patient what was believed to be recurrent disease was an independent (second) primary tumor. These findings suggest routine post-treatment sampling may have value in the management of recurrent pancreatic cancer. SIGNIFICANCE: The biological features or clinical vulnerabilities of recurrent pancreatic cancer after pancreaticoduodenectomy are unknown. Using whole-exome sequencing we find that recurrent disease has a distinct genomic landscape, intermetastatic genetic heterogeneity, diverse clonal origins, and higher mutational burden than found for treatment-naïve disease. See related commentary by Bednar and Pasca di Magliano, p. 762 . This article is highlighted in the In This Issue feature, p. 747 ., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
49. Eomes identifies thymic precursors of self-specific memory-phenotype CD8 + T cells.
- Author
-
Miller CH, Klawon DEJ, Zeng S, Lee V, Socci ND, and Savage PA
- Subjects
- Animals, Autoantigens immunology, Cell Differentiation, Clonal Selection, Antigen-Mediated, Clone Cells, Immunologic Memory, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Programmed Cell Death 1 Receptor metabolism, T-Box Domain Proteins genetics, Up-Regulation, CD8-Positive T-Lymphocytes immunology, Prostatic Neoplasms immunology, Receptors, Antigen, T-Cell genetics, T-Box Domain Proteins metabolism, Thymus Gland physiology
- Abstract
Unprimed mice harbor a substantial population of 'memory-phenotype' CD8
+ T cells (CD8-MP cells) that exhibit hallmarks of activation and innate-like functional properties. Due to the lack of faithful markers to distinguish CD8-MP cells from bona fide CD8+ memory T cells, the developmental origins and antigen specificities of CD8-MP cells remain incompletely defined. Using deep T cell antigen receptor (TCR) sequencing, we found that the TCRs expressed by CD8-MP cells are highly recurrent and distinct from the TCRs expressed by naive-phenotype CD8+ T cells. CD8-MP clones exhibited reactivity to widely expressed self-ligands. T cell precursors expressing CD8-MP TCRs showed upregulation of the transcription factor Eomes during maturation in the thymus, prior to induction of the full memory phenotype, which is suggestive of a unique program triggered by recognition of self-ligands. Moreover, CD8-MP cells infiltrate oncogene-driven prostate tumors and express high densities of PD-1, which suggests potential roles in antitumor immunity and the response to immunotherapy.- Published
- 2020
- Full Text
- View/download PDF
50. Author Correction: Tumour lineage shapes BRCA-mediated phenotypes.
- Author
-
Jonsson P, Bandlamudi C, Cheng ML, Srinivasan P, Chavan SS, Friedman ND, Rosen EY, Richards AL, Bouvier N, Selcuklu SD, Bielski CM, Abida W, Mandelker D, Birsoy O, Zhang L, Zehir A, Donoghue MTA, Baselga J, Offit K, Scher HI, O'Reilly EM, Stadler ZK, Schultz N, Socci ND, Viale A, Ladanyi M, Robson ME, Hyman DM, Berger MF, Solit DB, and Taylor BS
- Abstract
An Amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.