304 results on '"Ley, TJ"'
Search Results
2. Obesity is a risk factor for acute promyelocytic leukemia: evidence from population and cross-sectional studies studies and correlation with flt3 mutations and polyunsaturated fatty acid metabolism
- Author
-
Mazzarella L, Botteri E, Matthews A, Gatti E, Di Salvatore D, Bagnardi V, Breccia M, Montesinos P, Bernal T, Gil C, Ley TJ, Sanz M, Bhaskaran K, Lo Coco F, and Pelicci PG
- Subjects
Acute Myeloid Leukemia, Acute Promyelocytic Leukemia, Clinical and Molecular Epidemiology, Cytogenetics and Molecular Genetics ,hemic and lymphatic diseases - Abstract
Obesity correlates with hematological malignances including leukemias, but risk of specific leukemia subtypes like Acute Promyelocytic Leukemia and underlying molecular mechanisms are poorly understood. We explored multiple datasets for correlation between leukemia, Body Mass Index and molecular features. In a population-based study (n=5.2 million), we correlated Body Mass Index with promyelocytic, other acute myeloid, lymphoid or other leukemias. In cross-sectional studies, we tested body mass index deviation in promyelocytic leukemia trial cohorts from what expected based on national surveys. We interrogated The Cancer Genome Atlas for transcriptional signatures and mutations enriched in promyelocytic leukemia and/or obesity and confirmed correlation between body mass and FLT3 mutations in promyelocytic leukemia cohorts by logistic regression. In the population-based study, Hazard Ratio per 5 kg/m2 increase was: promyelocytic leukemia 1.44 (95% CI 1.0-2.08); non-promyelocytic acute myeloid leukemias 1.17 (1.10-1.26); lymphoid leukemias 1.04 (1.0-1.09); other 1.10 (1.04-1.15). In cross-sectional studies, body mass deviated significantly from expected (Italy p
- Published
- 2020
3. An Inv(16)(p13.3q24.3)-encoded CBFA2T3-GLIS2 fusion protein defines an aggressive subtype of pediatric acute megakaryoblastic leukemia
- Author
-
Gruber, T, Larson Gedman, A, Zhang, J, Koss, C, Marada, S, Ta, H, Chen, S, Su, X, Ogden, S, Dang, J, Wu, G, Gupta, V, Andersson, A, Pounds, S, Sh, L, Easton, J, Barbato, M, Mulder, H, Manne, J, Wang, J, Rusch, M, Ranade, S, Ganti, R, Parker, M, Ma, J, Radtke, I, Ding, L, Cazzaniga, G, Biondi, A, Kornblau, S, Ravandi, F, Kantarjian, H, Nimer, S, Döhner, K, Döhner, H, Ley, T, Ballerini, P, Shurtleff, S, Tomizawa, D, Adachi, S, Hayashi, Y, Tawa, A, Shih, L, Liang, D, Rubnitz, J, Pui, C, Mardis, E, Wilson, R, Downing, J, Gruber, TA, Koss, CS, Ta, HQ, Chen, SC, Ogden, SK, Andersson, AK, Barbato, MI, Mulder, HL, Kornblau, SM, Nimer, SD, Ley, TJ, Shih, LY, Liang, DC, Rubnitz, JE, Pui, CH, Mardis, ER, Wilson, RK, Downing, JR, BIONDI, ANDREA, Gruber, T, Larson Gedman, A, Zhang, J, Koss, C, Marada, S, Ta, H, Chen, S, Su, X, Ogden, S, Dang, J, Wu, G, Gupta, V, Andersson, A, Pounds, S, Sh, L, Easton, J, Barbato, M, Mulder, H, Manne, J, Wang, J, Rusch, M, Ranade, S, Ganti, R, Parker, M, Ma, J, Radtke, I, Ding, L, Cazzaniga, G, Biondi, A, Kornblau, S, Ravandi, F, Kantarjian, H, Nimer, S, Döhner, K, Döhner, H, Ley, T, Ballerini, P, Shurtleff, S, Tomizawa, D, Adachi, S, Hayashi, Y, Tawa, A, Shih, L, Liang, D, Rubnitz, J, Pui, C, Mardis, E, Wilson, R, Downing, J, Gruber, TA, Koss, CS, Ta, HQ, Chen, SC, Ogden, SK, Andersson, AK, Barbato, MI, Mulder, HL, Kornblau, SM, Nimer, SD, Ley, TJ, Shih, LY, Liang, DC, Rubnitz, JE, Pui, CH, Mardis, ER, Wilson, RK, Downing, JR, and BIONDI, ANDREA
- Abstract
To define the mutation spectrum in non-Down syndrome acute megakaryoblastic leukemia (non-DS-AMKL), we performed transcriptome sequencing on diagnostic blasts from 14 pediatric patients and validated our findings in a recurrency/validation cohort consisting of 34 pediatric and 28 adult AMKL samples. Our analysis identified a cryptic chromosome 16 inversion (inv(16)(p13.3q24.3)) in 27% of pediatric cases, which encodes a CBFA2T3-GLIS2 fusion protein. Expression of CBFA2T3-GLIS2 in Drosophila and murine hematopoietic cells induced bone morphogenic protein (BMP) signaling and resulted in a marked increase in the self-renewal capacity of hematopoietic progenitors. These data suggest that expression of CBFA2T3-GLIS2 directly contributes to leukemogenesis
- Published
- 2012
4. International network of cancer genome projects
- Author
-
Hudson, TJ, Anderson, W, Aretz, A, Barker, AD, Bell, C, Bernabe, RR, Bhan, MK, Calvo, F, Eerola, I, Gerhard, DS, Guttmacher, A, Guyer, M, Hemsley, FM, Jennings, JL, Kerr, D, Klatt, P, Kolar, P, Kusuda, J, Lane, DP, Laplace, F, Lu, Y, Nettekoven, G, Ozenberger, B, Peterson, J, Rao, TS, Remacle, J, Schafer, AJ, Shibata, T, Stratton, MR, Vockley, JG, Watanabe, K, Yang, H, Yuen, MMF, Knoppers, M, Bobrow, M, Cambon-Thomsen, A, Dressler, LG, Dyke, SOM, Joly, Y, Kato, K, Kennedy, KL, Nicolas, P, Parker, MJ, Rial-Sebbag, E, Romeo-Casabona, CM, Shaw, KM, Wallace, S, Wiesner, GL, Zeps, N, Lichter, P, Biankin, AV, Chabannon, C, Chin, L, Clement, B, de Alava, E, Degos, F, Ferguson, ML, Geary, P, Hayes, DN, Johns, AL, Nakagawa, H, Penny, R, Piris, MA, Sarin, R, Scarpa, A, van de Vijver, M, Futreal, PA, Aburatani, H, Bayes, M, Bowtell, DDL, Campbell, PJ, Estivill, X, Grimmond, SM, Gut, I, Hirst, M, Lopez-Otin, C, Majumder, P, Marra, M, Ning, Z, Puente, XS, Ruan, Y, Stunnenberg, HG, Swerdlow, H, Velculescu, VE, Wilson, RK, Xue, HH, Yang, L, Spellman, PT, Bader, GD, Boutros, PC, Flicek, P, Getz, G, Guigo, R, Guo, G, Haussler, D, Heath, S, Hubbard, TJ, Jiang, T, Jones, SM, Li, Q, Lopez-Bigas, N, Luo, R, Pearson, JV, Quesada, V, Raphael, BJ, Sander, C, Speed, TP, Stuart, JM, Teague, JW, Totoki, Y, Tsunoda, T, Valencia, A, Wheeler, DA, Wu, H, Zhao, S, Zhou, G, Stein, LD, Lathrop, M, Ouellette, BFF, Thomas, G, Yoshida, T, Axton, M, Gunter, C, McPherson, JD, Miller, LJ, Kasprzyk, A, Zhang, J, Haider, SA, Wang, J, Yung, CK, Cross, A, Liang, Y, Gnaneshan, S, Guberman, J, Hsu, J, Chalmers, DRC, Hasel, KW, Kaan, TSH, Knoppers, BM, Lowrance, WW, Masui, T, Rodriguez, LL, Vergely, C, Cloonan, N, Defazio, A, Eshleman, JR, Etemadmoghadam, D, Gardiner, BA, Kench, JG, Sutherland, RL, Tempero, MA, Waddell, NJ, Wilson, PJ, Gallinger, S, Tsao, M-S, Shaw, PA, Petersen, GM, Mukhopadhyay, D, DePinho, RA, Thayer, S, Muthuswamy, L, Shazand, K, Beck, T, Sam, M, Timms, L, Ballin, V, Ji, J, Zhang, X, Chen, F, Hu, X, Yang, Q, Tian, G, Zhang, L, Xing, X, Li, X, Zhu, Z, Yu, Y, Yu, J, Tost, J, Brennan, P, Holcatova, I, Zaridze, D, Brazma, A, Egevad, L, Prokhortchouk, E, Banks, RE, Uhlen, M, Viksna, J, Ponten, F, Skryabin, K, Birney, E, Borg, A, Borresen-Dale, A-L, Caldas, C, Foekens, JA, Martin, S, Reis-Filho, JS, Richardson, AL, Sotiriou, C, van't Veer, L, Birnbaum, D, Blanche, H, Boucher, P, Boyault, S, Masson-Jacquemier, JD, Pauporte, I, Pivot, X, Vincent-Salomon, A, Tabone, E, Theillet, C, Treilleux, I, Bioulac-Sage, P, Decaens, T, Franco, D, Gut, M, Samuel, D, Zucman-Rossi, J, Eils, R, Brors, B, Korbel, JO, Korshunov, A, Landgraf, P, Lehrach, H, Pfister, S, Radlwimmer, B, Reifenberger, G, Taylor, MD, von Kalle, C, Majumder, PP, Pederzoli, P, Lawlor, RT, Delledonne, M, Bardelli, A, Gress, T, Klimstra, D, Zamboni, G, Nakamura, Y, Miyano, S, Fujimoto, A, Campo, E, de Sanjose, S, Montserrat, E, Gonzalez-Diaz, M, Jares, P, Himmelbaue, H, Bea, S, Aparicio, S, Easton, DF, Collins, FS, Compton, CC, Lander, ES, Burke, W, Green, AR, Hamilton, SR, Kallioniemi, OP, Ley, TJ, Liu, ET, Wainwright, BJ, Hudson, TJ, Anderson, W, Aretz, A, Barker, AD, Bell, C, Bernabe, RR, Bhan, MK, Calvo, F, Eerola, I, Gerhard, DS, Guttmacher, A, Guyer, M, Hemsley, FM, Jennings, JL, Kerr, D, Klatt, P, Kolar, P, Kusuda, J, Lane, DP, Laplace, F, Lu, Y, Nettekoven, G, Ozenberger, B, Peterson, J, Rao, TS, Remacle, J, Schafer, AJ, Shibata, T, Stratton, MR, Vockley, JG, Watanabe, K, Yang, H, Yuen, MMF, Knoppers, M, Bobrow, M, Cambon-Thomsen, A, Dressler, LG, Dyke, SOM, Joly, Y, Kato, K, Kennedy, KL, Nicolas, P, Parker, MJ, Rial-Sebbag, E, Romeo-Casabona, CM, Shaw, KM, Wallace, S, Wiesner, GL, Zeps, N, Lichter, P, Biankin, AV, Chabannon, C, Chin, L, Clement, B, de Alava, E, Degos, F, Ferguson, ML, Geary, P, Hayes, DN, Johns, AL, Nakagawa, H, Penny, R, Piris, MA, Sarin, R, Scarpa, A, van de Vijver, M, Futreal, PA, Aburatani, H, Bayes, M, Bowtell, DDL, Campbell, PJ, Estivill, X, Grimmond, SM, Gut, I, Hirst, M, Lopez-Otin, C, Majumder, P, Marra, M, Ning, Z, Puente, XS, Ruan, Y, Stunnenberg, HG, Swerdlow, H, Velculescu, VE, Wilson, RK, Xue, HH, Yang, L, Spellman, PT, Bader, GD, Boutros, PC, Flicek, P, Getz, G, Guigo, R, Guo, G, Haussler, D, Heath, S, Hubbard, TJ, Jiang, T, Jones, SM, Li, Q, Lopez-Bigas, N, Luo, R, Pearson, JV, Quesada, V, Raphael, BJ, Sander, C, Speed, TP, Stuart, JM, Teague, JW, Totoki, Y, Tsunoda, T, Valencia, A, Wheeler, DA, Wu, H, Zhao, S, Zhou, G, Stein, LD, Lathrop, M, Ouellette, BFF, Thomas, G, Yoshida, T, Axton, M, Gunter, C, McPherson, JD, Miller, LJ, Kasprzyk, A, Zhang, J, Haider, SA, Wang, J, Yung, CK, Cross, A, Liang, Y, Gnaneshan, S, Guberman, J, Hsu, J, Chalmers, DRC, Hasel, KW, Kaan, TSH, Knoppers, BM, Lowrance, WW, Masui, T, Rodriguez, LL, Vergely, C, Cloonan, N, Defazio, A, Eshleman, JR, Etemadmoghadam, D, Gardiner, BA, Kench, JG, Sutherland, RL, Tempero, MA, Waddell, NJ, Wilson, PJ, Gallinger, S, Tsao, M-S, Shaw, PA, Petersen, GM, Mukhopadhyay, D, DePinho, RA, Thayer, S, Muthuswamy, L, Shazand, K, Beck, T, Sam, M, Timms, L, Ballin, V, Ji, J, Zhang, X, Chen, F, Hu, X, Yang, Q, Tian, G, Zhang, L, Xing, X, Li, X, Zhu, Z, Yu, Y, Yu, J, Tost, J, Brennan, P, Holcatova, I, Zaridze, D, Brazma, A, Egevad, L, Prokhortchouk, E, Banks, RE, Uhlen, M, Viksna, J, Ponten, F, Skryabin, K, Birney, E, Borg, A, Borresen-Dale, A-L, Caldas, C, Foekens, JA, Martin, S, Reis-Filho, JS, Richardson, AL, Sotiriou, C, van't Veer, L, Birnbaum, D, Blanche, H, Boucher, P, Boyault, S, Masson-Jacquemier, JD, Pauporte, I, Pivot, X, Vincent-Salomon, A, Tabone, E, Theillet, C, Treilleux, I, Bioulac-Sage, P, Decaens, T, Franco, D, Gut, M, Samuel, D, Zucman-Rossi, J, Eils, R, Brors, B, Korbel, JO, Korshunov, A, Landgraf, P, Lehrach, H, Pfister, S, Radlwimmer, B, Reifenberger, G, Taylor, MD, von Kalle, C, Majumder, PP, Pederzoli, P, Lawlor, RT, Delledonne, M, Bardelli, A, Gress, T, Klimstra, D, Zamboni, G, Nakamura, Y, Miyano, S, Fujimoto, A, Campo, E, de Sanjose, S, Montserrat, E, Gonzalez-Diaz, M, Jares, P, Himmelbaue, H, Bea, S, Aparicio, S, Easton, DF, Collins, FS, Compton, CC, Lander, ES, Burke, W, Green, AR, Hamilton, SR, Kallioniemi, OP, Ley, TJ, Liu, ET, and Wainwright, BJ
- Abstract
The International Cancer Genome Consortium (ICGC) was launched to coordinate large-scale cancer genome studies in tumours from 50 different cancer types and/or subtypes that are of clinical and societal importance across the globe. Systematic studies of more than 25,000 cancer genomes at the genomic, epigenomic and transcriptomic levels will reveal the repertoire of oncogenic mutations, uncover traces of the mutagenic influences, define clinically relevant subtypes for prognosis and therapeutic management, and enable the development of new cancer therapies.
- Published
- 2010
5. The role of granzyme B in murine models of acute graft-versus-host disease and graft rejection
- Author
-
Graubert, TA, primary, Russell, JH, additional, and Ley, TJ, additional
- Published
- 1996
- Full Text
- View/download PDF
6. Consensus AP-1 and CRE motifs upstream from the human cytotoxic serine protease B (CSP-B/CGL-1) gene synergize to activate transcription
- Author
-
Hanson, RD, primary, Grisolano, JL, additional, and Ley, TJ, additional
- Published
- 1993
- Full Text
- View/download PDF
7. Molecular cloning, chromosomal location, and tissue-specific expression of the murine cathepsin G gene
- Author
-
Heusel, JW, primary, Scarpati, EM, additional, Jenkins, NA, additional, Gilbert, DJ, additional, Copeland, NG, additional, Shapiro, SD, additional, and Ley, TJ, additional
- Published
- 1993
- Full Text
- View/download PDF
8. A-T-rich scaffold attachment regions flank the hematopoietic serine protease genes clustered on chromosome 14q11.2
- Author
-
Hanson, RD, primary and Ley, TJ, additional
- Published
- 1992
- Full Text
- View/download PDF
9. Functional properties of the beta-globin locus control region in K562 erythroleukemia cells
- Author
-
Moon, AM, primary and Ley, TJ, additional
- Published
- 1991
- Full Text
- View/download PDF
10. The pharmacology of hemoglobin switching: of mice and men
- Author
-
Ley, TJ, primary
- Published
- 1991
- Full Text
- View/download PDF
11. Function of normal and mutated gamma-globin gene promoters in electroporated K562 erythroleukemia cells
- Author
-
Ulrich, MJ, primary and Ley, TJ, additional
- Published
- 1990
- Full Text
- View/download PDF
12. Advances in thalassemia research
- Author
-
Nienhuis, AW, Anagnou, NP, and Ley, TJ
- Published
- 1984
- Full Text
- View/download PDF
13. High-resolution analysis of the human gamma-globin gene promoter in K562 erythroleukemia cell chromatin
- Author
-
Gimble, JM, Max, EE, and Ley, TJ
- Abstract
We performed high-resolution mapping studies of the DNAse I- hypersensitive sites located just 5' to the human G gamma- and A gamma- globin genes of K562 erythroleukemia cells, in which these genes are constitutively expressed at low levels. This analysis revealed that the hypersensitive site extends from approximately -210 +/- 5 to -25 +/- 5 base pairs (bp) upstream from the transcription initiation site. Within this region, a GC-rich region located between the proximal CCAAT box and the TATA box is particularly accessible to nuclease digestion; however, the 5' end of the hypersensitive site is less accessible to nucleases. The pattern of DNAse I cleavage does not change on either strand with hemin induction of K562 cells, which increases the rate of gamma-globin gene transcription about threefold. The region within the hypersensitive site includes all the consensus promoter elements of the gamma-globin genes as well as an octamer sequence located between -182 and -175, and a region associated with a variety of mutations that may cause hereditary persistence of fetal hemoglobin (HPFH).
- Published
- 1988
- Full Text
- View/download PDF
14. Beta O-39 thalassemia gene: a premature termination codon causes beta- mRNA deficiency without affecting cytoplasmic beta-mRNA stability
- Author
-
Humphries, RK, Ley, TJ, Anagnou, NP, Baur, AW, and Nienhuis, AW
- Abstract
The function of a beta O-thalassemia globin gene with a premature termination codon at position 39 was studied in tissue culture cells using plasmid expression vectors. The thalassemic globin gene was isolated by molecular cloning from the bone marrow DNA of an Italian patient with severe thalassemia. Sequences upstream of the normal beta- globin gene and the beta O-39 globin gene were removed to 127 nucleotides (nt) 5' to the start site of transcription, thereby eliminating uncharacterized DNA sequences but preserving promoter function. To create a hybrid gene differing from the normal by only the single nt change in codon 39, the truncated 5' end of the beta O-39 gene was recombined with the 3' end of the normal gene. The beta O-39 substitution resulted in a 6–14-fold reduction in cytoplasmic RNA accumulation in transfected monkey kidney or HeLa cells. This decrease in mRNA did not appear to be due to instability, as the beta-mRNA present 24–36 hours after transfection was stable for up to 24 hours in the presence of actinomycin D. In the presence of actinomycin D, the globin mRNA precursor disappeared, suggesting that globin gene transcription had been effectively blocked. We also examined a second globin mRNA with a premature termination codon; this RNA that arises from incorrect splicing of the transcript of a beta + -24 thalassemia gene was as stable as the correctly spliced species. Thus, the presence of a premature termination codon does not necessarily alter cytoplasmic mRNA stability, nor does cytoplasmic instability account for the quantitative deficiency of beta-globin mRNA. Our observations suggest that a premature termination codon alters intranuclear stability and/or nuclear to cytoplasmic transport of the beta-globin mRNA.
- Published
- 1984
- Full Text
- View/download PDF
15. Hydroxyurea induction of hemoglobin F production in sickle cell disease: relationship between cytotoxicity and F cell production
- Author
-
Dover, GJ, Humphries, RK, Moore, JG, Ley, TJ, Young, NS, Charache, S, and Nienhuis, AW
- Abstract
Initial alterations in fetal hemoglobin (HbF) production among eight sickle cell anemia subjects treated with hydroxyurea (Hu) are summarized. Four of these subjects had been previously treated with 5- azacytidine (5-aza). All subjects treated with Hu (50 mg/kg/d for three to five days) had suppression of their total reticulocyte counts by seven days, whereas the four subjects previously treated with 5-aza (2 mg/kg/d for three to five days) had increased reticulocyte counts at day 7. The effect of Hu on increasing the number of HbF-containing reticulocytes (F reticulocytes) is extremely variable, ranging from ten- to less than onefold differences in maximal posttherapy v pretherapy levels. Recovery from marrow suppression did not result in greater than twofold increases in F reticulocyte counts. Mean day 7 F reticulocyte levels in the four subjects treated with both Hu and 5-aza were 4.1 X 10/microL and 15.4 X 10(4)/microL, respectively. Among Hu-treated subjects, increased F reticulocyte production was correlated with low serum creatinine levels and rapid removal of Hu from the plasma. Furthermore, suppression of CFU-E colony formation on day 2 of therapy with Hu was inversely correlated with maximal F reticulocyte response. We conclude that where Hu treatment results in marrow toxicity (decreased reticulocyte counts, decreased CFU-E colony formation) HbF production is less likely to increase. Those sickle cell anemia subjects with minimal renal dysfunction (serum creatinine level, greater than 1.0 mg/dL) exhibit the most cytotoxicity and least F reticulocyte response to Hu.
- Published
- 1986
- Full Text
- View/download PDF
16. 5-Azacytidine increases gamma-globin synthesis and reduces the proportion of dense cells in patients with sickle cell anemia
- Author
-
Ley, TJ, DeSimone, J, Noguchi, CT, Turner, PH, Schechter, AN, Heller, P, and Nienhuis, AW
- Abstract
We previously demonstrated that 5-azacytidine can selectively increase gamma-globin synthesis in a patient with beta +-thalassemia, prompting us to treat two patients with sickle cell anemia and two additional patients with beta + thalassemia. 5-Azacytidine (2 mg/kg/day) was continuously infused for 7 days with no apparent clinical toxicity. The gamma/beta-globin biosynthetic ratio increased fourfold to sixfold in the bone marrow cells of each patient after treatment and remained elevated for 7–14 additional days. Hypomethylation of DNA near the gamma-globin genes in bone marrow cells was demonstrated 2 days after beginning the 5-azacytidine infusion. The peripheral blood fetal hemoglobin (HbF) level increased from 6.0% to 13.7% in one patient with sickle cell anemia and from 1.6% to 8.9% in the second. Stractan gradient analyses of peripheral blood from patients with sickle cell anemia revealed a marked decrease in the percentage of dense cells (cells that contain increased amounts of HbS polymer when deoxygenated) following treatment. These observations provide an impetus to investigate the effects of repeated courses of 5-azacytidine in a small group of severely ill patients to determine whether this drug may have a role in the treatment of patients with sickle cell anemia and beta- thalassemia.
- Published
- 1983
- Full Text
- View/download PDF
17. Debt repayment for trainees.
- Author
-
Chessler SD, Egan LW, Gallin EK, Sung NS, Gronski TJ Jr., Ley TJ, Rosenberg LE, and Nathan DG
- Published
- 2002
18. Molecular responses in decitabine- and decitabine/ venetoclax-treated patients with acute myeloid leukemia and myelodysplastic syndromes.
- Author
-
Gruszczynska A, Maiti A, Miller CA, Ramakrishnan SM, Link DC, Uy GL, Petti AA, Hayes K, DiNardo CD, Ravandi F, Ley TJ, Spencer DH, Gao F, Konopleva MY, and Welch JS
- Subjects
- Humans, Male, Aged, Female, Treatment Outcome, Middle Aged, Adult, Decitabine therapeutic use, Decitabine pharmacology, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute mortality, Sulfonamides therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use
- Published
- 2024
- Full Text
- View/download PDF
19. PML::RARA and GATA2 proteins interact via DNA templates to induce aberrant self-renewal in mouse and human hematopoietic cells.
- Author
-
Katerndahl CDS, Rogers ORS, Day RB, Xu Z, Helton NM, Ramakrishnan SM, Miller CA, and Ley TJ
- Subjects
- Animals, Humans, Mice, Binding Sites, Cell Self Renewal, Chromatin metabolism, DNA metabolism, Leukemia, Promyelocytic, Acute metabolism, Leukemia, Promyelocytic, Acute genetics, Leukemia, Promyelocytic, Acute pathology, Promyelocytic Leukemia Protein metabolism, Promyelocytic Leukemia Protein genetics, Protein Binding, Retinoic Acid Receptor alpha metabolism, Retinoic Acid Receptor alpha genetics, GATA2 Transcription Factor metabolism, GATA2 Transcription Factor genetics, Hematopoietic Stem Cells metabolism, Oncogene Proteins, Fusion metabolism, Oncogene Proteins, Fusion genetics
- Abstract
The underlying mechanism(s) by which the PML::RARA fusion protein initiates acute promyelocytic leukemia is not yet clear. We defined the genomic binding sites of PML::RARA in primary mouse and human hematopoietic progenitor cells with V5-tagged PML::RARA, using anti-V5-PML::RARA chromatin immunoprecipitation sequencing and CUT&RUN approaches. Most genomic PML::RARA binding sites were found in regions that were already chromatin-accessible (defined by ATAC-seq) in unmanipulated, wild-type promyelocytes, suggesting that these regions are "open" prior to PML::RARA expression. We found that GATA binding motifs, and the direct binding of the chromatin "pioneering factor" GATA2, were significantly enriched near PML::RARA binding sites. Proximity labeling studies revealed that PML::RARA interacts with ~250 proteins in primary mouse hematopoietic cells; GATA2 and 33 others require PML::RARA binding to DNA for the interaction to occur, suggesting that binding to their cognate DNA target motifs may stabilize their interactions. In the absence of PML::RARA , Gata2 overexpression induces many of the same epigenetic and transcriptional changes as PML::RARA . These findings suggested that PML::RARA may indirectly initiate its transcriptional program by activating Gata2 expression: Indeed, we demonstrated that inactivation of Gata2 prior to PML::RARA expression prevented its ability to induce self-renewal. These data suggested that GATA2 binding creates accessible chromatin regions enriched for both GATA and Retinoic Acid Receptor Element motifs, where GATA2 and PML::RARA can potentially bind and interact with each other. In turn, PML::RARA binding to DNA promotes a feed-forward transcriptional program by positively regulating Gata2 expression. Gata2 may therefore be required for PML::RARA to establish its transcriptional program., Competing Interests: Competing interests statement:The authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
20. Rapid and accurate remethylation of DNA in Dnmt3a- deficient hematopoietic cells with restoration of DNMT3A activity.
- Author
-
Li Y, Abel HJ, Cai M, LaValle TA, Yin T, Helton NM, Smith AM, Miller CA, and Ley TJ
- Subjects
- Humans, Mice, Animals, DNA Methyltransferase 3A, DNA, Mutation, DNA Methylation, DNA (Cytosine-5-)-Methyltransferases genetics, Leukemia, Myeloid, Acute genetics
- Abstract
Here, we characterize the DNA methylation phenotypes of bone marrow cells from mice with hematopoietic deficiency of Dnmt3a or Dnmt3b (or both enzymes) or expressing the dominant-negative Dnmt3a
R878H mutation [R882H in humans; the most common DNMT3A mutation found in acute myeloid leukemia (AML)]. Using these cells as substrates, we defined DNA remethylation after overexpressing wild-type (WT) DNMT3A1, DNMT3B1, DNMT3B3 (an inactive splice isoform of DNMT3B), or DNMT3L (a catalytically inactive "chaperone" for DNMT3A and DNMT3B in early embryogenesis). Overexpression of DNMT3A for 2 weeks reverses the hypomethylation phenotype of Dnmt3a-deficient cells or cells expressing the R878H mutation. Overexpression of DNMT3L (which is minimally expressed in AML cells) also corrects the hypomethylation phenotype of Dnmt3aR878H/+ marrow, probably by augmenting the activity of WT DNMT3A encoded by the residual WT allele. DNMT3L reactivation may represent a previously unidentified approach for restoring DNMT3A activity in hematopoietic cells with reduced DNMT3A function.- Published
- 2024
- Full Text
- View/download PDF
21. Stabilizing the Proteomes of Acute Myeloid Leukemia Cells: Implications for Cancer Proteomics.
- Author
-
Sprung RW, Zhang Q, Kramer MH, Christopher MC, Erdmann-Gilmore P, Mi Y, Malone JP, Ley TJ, and Townsend RR
- Subjects
- Humans, Proteomics, Endopeptidases metabolism, Serine Proteases, Peptides chemistry, Proteome, Leukemia, Myeloid, Acute
- Abstract
Previous work has shown that inhibition of abundant myeloid azurophil granule-associated serine proteases (ELANE [neutrophil elastase], PRTN3 [protease 3], and CTSG [Cathepsin G]) is required to stabilize some proteins in myeloid cells. We therefore hypothesized that effective inhibition of these proteases may be necessary for quantitative proteomic analysis of samples containing myeloid cells. To test this hypothesis, we thawed viably preserved acute myeloid leukemia cells from cryovials in the presence or the absence of diisopropyl fluorophosphate (DFP), a cell-permeable and irreversible serine protease inhibitor. Global proteomic analysis was performed, using label-free and isobaric peptide-labeling quantitation. The presence of DFP resulted in an increase of tryptic peptides (14-57%) and proteins (9-31%). In the absence of DFP, 11 to 31% of peptide intensity came from nontryptic peptides; 52 to 75% had cleavage specificity consistent with activities of ELANE-PRTN3. Treatment with DFP reduced the intensity of nontryptic peptides to 4-8% of the total. ELANE inhibition was 95%, based on diisopropyl phosphate modification of active site serine residue. Overall, the relative abundance of 20% of proteins was significantly altered by DFP treatment. These results suggest that active myeloid serine proteases, released during sample processing, can skew quantitative proteomic measurements. Finally, significant ELANE activity was also detected in Clinical Proteomics Tumor Analysis Consortium datasets of solid tumors (many of which have known myeloid infiltration). In the pancreatic cancer dataset, the median percentage of nontryptic intensity detected across patient samples was 34%, with many patient samples having more than half of their detected peptide intensity from nontryptic cleavage events consistent with ELANE-PRTN3 cleavage specificity. Our study suggests that in vitro cleavage of proteins by myeloid serine proteases may be relevant for proteomic studies of any tumor that contains infiltrating myeloid cells., Competing Interests: Conflict of interest The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
22. Proteogenomic analysis reveals cytoplasmic sequestration of RUNX1 by the acute myeloid leukemia-initiating CBFB::MYH11 oncofusion protein.
- Author
-
Day RB, Hickman JA, Xu Z, Katerndahl CD, Ferraro F, Ramakrishnan SM, Erdmann-Gilmore P, Sprung RW, Mi Y, Townsend RR, Miller CA, and Ley TJ
- Subjects
- Humans, Mice, Animals, Core Binding Factor Alpha 2 Subunit genetics, Translocation, Genetic, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism, Core Binding Factor beta Subunit, Myosin Heavy Chains genetics, Proteogenomics, Leukemia, Myeloid, Acute pathology
- Abstract
Several canonical translocations produce oncofusion genes that can initiate acute myeloid leukemia (AML). Although each translocation is associated with unique features, the mechanisms responsible remain unclear. While proteins interacting with each oncofusion are known to be relevant for how they act, these interactions have not yet been systematically defined. To address this issue in an unbiased fashion, we fused a promiscuous biotin ligase (TurboID) in-frame with 3 favorable-risk AML oncofusion cDNAs (PML::RARA, RUNX1::RUNX1T1, and CBFB::MYH11) and identified their interacting proteins in primary murine hematopoietic cells. The PML::RARA- and RUNX1::RUNX1T1-TurboID fusion proteins labeled common and unique nuclear repressor complexes, implying their nuclear localization. However, CBFB::MYH11-TurboID-interacting proteins were largely cytoplasmic, probably because of an interaction of the MYH11 domain with several cytoplasmic myosin-related proteins. Using a variety of methods, we showed that the CBFB domain of CBFB::MYH11 sequesters RUNX1 in cytoplasmic aggregates; these findings were confirmed in primary human AML cells. Paradoxically, CBFB::MYH11 expression was associated with increased RUNX1/2 expression, suggesting the presence of a sensor for reduced functional RUNX1 protein, and a feedback loop that may attempt to compensate by increasing RUNX1/2 transcription. These findings may have broad implications for AML pathogenesis.
- Published
- 2023
- Full Text
- View/download PDF
23. Genomic landscape of TP53-mutated myeloid malignancies.
- Author
-
Abel HJ, Oetjen KA, Miller CA, Ramakrishnan SM, Day RB, Helton NM, Fronick CC, Fulton RS, Heath SE, Tarnawsky SP, Nonavinkere Srivatsan S, Duncavage EJ, Schroeder MC, Payton JE, Spencer DH, Walter MJ, Westervelt P, DiPersio JF, Ley TJ, and Link DC
- Subjects
- Humans, Mutation, Chromosome Aberrations, Genomics, Tumor Suppressor Protein p53 genetics, Chromothripsis, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Myeloproliferative Disorders genetics, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes pathology
- Abstract
TP53-mutated myeloid malignancies are associated with complex cytogenetics and extensive structural variants, which complicates detailed genomic analysis by conventional clinical techniques. We performed whole-genome sequencing (WGS) of 42 acute myeloid leukemia (AML)/myelodysplastic syndromes (MDS) cases with paired normal tissue to better characterize the genomic landscape of TP53-mutated AML/MDS. WGS accurately determines TP53 allele status, a key prognostic factor, resulting in the reclassification of 12% of cases from monoallelic to multihit. Although aneuploidy and chromothripsis are shared with most TP53-mutated cancers, the specific chromosome abnormalities are distinct to each cancer type, suggesting a dependence on the tissue of origin. ETV6 expression is reduced in nearly all cases of TP53-mutated AML/MDS, either through gene deletion or presumed epigenetic silencing. Within the AML cohort, mutations of NF1 are highly enriched, with deletions of 1 copy of NF1 present in 45% of cases and biallelic mutations in 17%. Telomere content is increased in TP53-mutated AMLs compared with other AML subtypes, and abnormal telomeric sequences were detected in the interstitial regions of chromosomes. These data highlight the unique features of TP53-mutated myeloid malignancies, including the high frequency of chromothripsis and structural variation, the frequent involvement of unique genes (including NF1 and ETV6) as cooperating events, and evidence for altered telomere maintenance., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
24. Melanoma in a patient with DNMT3A overgrowth syndrome.
- Author
-
Chen DY, Sutton LA, Ramakrishnan SM, Duncavage EJ, Heath SE, Compton LA, Miller CA, and Ley TJ
- Subjects
- Humans, Cell Proliferation, DNA Modification Methylases, Genotype, Syndrome, Melanoma genetics, Neoplasms, Second Primary
- Abstract
Alterations in epigenetic regulators are increasingly recognized as early events in tumorigenesis; thus, patients with acquired or inherited variants in epigenetic regulators may be at increased risk for developing multiple types of cancer. DNMT3A overgrowth syndrome (DOS), caused by germline pathogenic variants in the DNA methyltransferase gene DNMT3A , has been associated with a predisposition toward development of hematopoietic and neuronal malignancies. DNMT3A deficiency has been described to promote keratinocyte proliferation in mice. Although altered DNA methylation patterns are well-recognized in melanoma, the role of DNA methyltransferases in melanoma pathogenesis is not clear. We report the case of an adult DOS patient with a germline DNMT3A loss-of-function mutation, who developed an early-onset melanoma with regional lymph node metastatic disease. Exome sequencing of the primary tumor identified an additional acquired, missense DNMT3A mutation in the dominant tumor clone, suggesting that the loss of DNMT3A function was relevant for the development of this tumor., (© 2023 Chen et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2023
- Full Text
- View/download PDF
25. Persistent Molecular Disease in Adult Patients With AML Evaluated With Whole-Exome and Targeted Error-Corrected DNA Sequencing.
- Author
-
Slade MJ, Ghasemi R, O'Laughlin M, Burton T, Fulton RS, Abel HJ, Duncavage EJ, Ley TJ, Jacoby MA, and Spencer DH
- Subjects
- Humans, Adult, Exome, Prognosis, Neoplasm Recurrence, Local genetics, Sequence Analysis, DNA, Leukemia, Myeloid, Acute genetics
- Abstract
Purpose: Persistent molecular disease (PMD) after induction chemotherapy predicts relapse in AML. In this study, we used whole-exome sequencing (WES) and targeted error-corrected sequencing to assess the frequency and mutational patterns of PMD in 30 patients with AML., Materials and Methods: The study cohort included 30 patients with adult AML younger than 65 years who were uniformly treated with standard induction chemotherapy. Tumor/normal WES was performed for all patients at presentation. PMD analysis was evaluated in bone marrow samples obtained during clinicopathologic remission using repeat WES and analysis of patient-specific mutations and error-corrected sequencing of 40 recurrently mutated AML genes (MyeloSeq)., Results: WES for patient-specific mutations detected PMD in 63% of patients (19/30) using a minimum variant allele fraction (VAF) of 2.5%. In comparison, MyeloSeq identified persistent mutations above 0.1% VAF in 77% of patients (23/30). PMD was usually present at relatively high levels (>2.5% VAFs), such that WES and MyeloSeq agreed for 73% of patients despite differences in detection limits. Mutations in DNMT3A , ASXL1 , and TET2 (ie, DTA mutations) were persistent in 16 of 17 patients, but WES also detected non-DTA mutations in 14 of these patients, which for some patients distinguished residual AML cells from clonal hematopoiesis. Surprisingly, MyeloSeq detected additional variants not identified at presentation in 73% of patients that were consistent with new clonal cell populations after chemotherapy., Conclusion: PMD and clonal hematopoiesis are both common in patients with AML in first remission. These findings demonstrate the importance of baseline testing for accurate interpretation of mutation-based tumor monitoring assays for patients with AML and highlight the need for clinical trials to determine whether these complex mutation patterns correlate with clinical outcomes in AML.
- Published
- 2023
- Full Text
- View/download PDF
26. Description of a novel subtype of acute myeloid leukemia defined by recurrent CBFB insertions.
- Author
-
Ryland GL, Umeda M, Holmfeldt L, Lehmann S, Herlin MK, Ma J, Khanlari M, Rubnitz JE, Ries RE, Kosasih HJ, Ekert PG, Goh HN, Tiong IS, Grimmond SM, Haferlach C, Day RB, Ley TJ, Meshinchi S, Ma X, Blombery P, and Klco JM
- Subjects
- Humans, Oncogene Proteins, Fusion, Core Binding Factor beta Subunit, Leukemia, Myeloid, Acute genetics
- Published
- 2023
- Full Text
- View/download PDF
27. IL-1β expression in bone marrow dendritic cells is induced by TLR2 agonists and regulates HSC function.
- Author
-
Li S, Yao JC, Oetjen KA, Krambs JR, Xia J, Zhang J, Schmidt AP, Helton NM, Fulton RS, Heath SE, Turnbull IR, Mbalaviele G, Ley TJ, Walter MJ, and Link DC
- Subjects
- Animals, Bone Marrow metabolism, Cytokines metabolism, Humans, Mice, Myeloid Differentiation Factor 88 metabolism, RNA metabolism, Toll-Like Receptor 1 metabolism, Toll-Like Receptor 2 agonists, Toll-Like Receptor 2 metabolism, Bone Marrow Cells cytology, Dendritic Cells cytology, Hematopoietic Stem Cells cytology, Interleukin-1beta metabolism, Myelodysplastic Syndromes metabolism
- Abstract
Hematopoietic stem/progenitor cells (HSPCs) reside in localized microenvironments, or niches, in the bone marrow that provide key signals regulating their activity. A fundamental property of hematopoiesis is the ability to respond to environmental cues such as inflammation. How these cues are transmitted to HSPCs within hematopoietic niches is not well established. Here, we show that perivascular bone marrow dendritic cells (DCs) express a high basal level of Toll-like receptor-1 (TLR1) and TLR2. Systemic treatment with a TLR1/2 agonist induces HSPC expansion and mobilization. It also induces marked alterations in the bone marrow microenvironment, including a decrease in osteoblast activity and sinusoidal endothelial cell numbers. TLR1/2 agonist treatment of mice in which Myd88 is deleted specifically in DCs using Zbtb46-Cre show that the TLR1/2-induced expansion of multipotent HPSCs, but not HSPC mobilization or alterations in the bone marrow microenvironment, is dependent on TLR1/2 signaling in DCs. Interleukin-1β (IL-1β) is constitutively expressed in both murine and human DCs and is further induced after TLR1/2 stimulation. Systemic TLR1/2 agonist treatment of Il1r1-/- mice show that TLR1/2-induced HSPC expansion is dependent on IL-1β signaling. Single-cell RNA-sequencing of low-risk myelodysplastic syndrome bone marrow revealed that IL1B and TLR1 expression is increased in DCs. Collectively, these data suggest a model in which TLR1/2 stimulation of DCs induces secretion of IL-1β and other inflammatory cytokines into the perivascular niche, which in turn, regulates multipotent HSPCs. Increased DC TLR1/2 signaling may contribute to altered HSPC function in myelodysplastic syndrome by increasing local IL-1β expression., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
28. Proteomic and phosphoproteomic landscapes of acute myeloid leukemia.
- Author
-
Kramer MH, Zhang Q, Sprung R, Day RB, Erdmann-Gilmore P, Li Y, Xu Z, Helton NM, George DR, Mi Y, Westervelt P, Payton JE, Ramakrishnan SM, Miller CA, Link DC, DiPersio JF, Walter MJ, Townsend RR, and Ley TJ
- Subjects
- Histone Demethylases metabolism, Humans, Jumonji Domain-Containing Histone Demethylases, Karyopherins genetics, Ketoglutaric Acids, Membrane Proteins genetics, Mutation, Nucleophosmin, Proteome metabolism, Proteomics, RNA, Messenger, Serine genetics, fms-Like Tyrosine Kinase 3 genetics, src-Family Kinases metabolism, Leukemia, Myeloid, Acute pathology, Nuclear Proteins genetics
- Abstract
We have developed a deep-scale proteome and phosphoproteome database from 44 representative acute myeloid leukemia (AML) patients from the LAML TCGA dataset and 6 healthy bone marrow-derived controls. After confirming data quality, we orthogonally validated several previously undescribed features of AML revealed by the proteomic data. We identified examples of posttranscriptionally regulated proteins both globally (ie, in all AML samples) and also in patients with recurrent AML driver mutations. For example, samples with IDH1/2 mutations displayed elevated levels of the 2-oxoglutarate-dependent histone demethylases KDM4A/B/C, despite no changes in messenger RNA levels for these genes; we confirmed this finding in vitro. In samples with NPMc mutations, we identified several nuclear importins with posttranscriptionally increased protein abundance and showed that they interact with NPMc but not wild-type NPM1. We identified 2 cell surface proteins (CD180 and MRC1/CD206) expressed on AML blasts of many patients (but not healthy CD34+ stem/progenitor cells) that could represent novel targets for immunologic therapies and confirmed these targets via flow cytometry. Finally, we detected nearly 30 000 phosphosites in these samples; globally, AML samples were associated with the abnormal phosphorylation of specific residues in PTPN11, STAT3, AKT1, and PRKCD. FLT3-TKD samples were associated with increased phosphorylation of activating tyrosines on the cytoplasmic Src-family tyrosine kinases FGR and HCK and related signaling proteins. PML-RARA-initiated AML samples displayed a unique phosphorylation signature, and TP53-mutant samples showed abundant phosphorylation of serine-183 on TP53 itself. This publicly available database will serve as a foundation for further investigations of protein dysregulation in AML pathogenesis., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
29. Author Correction: A general approach for detecting expressed mutations in AML cells using single cell RNA-sequencing.
- Author
-
Petti AA, Williams SR, Miller CA, Fiddes IT, Srivatsan SN, Chen DY, Fronick CC, Fulton RS, Church DM, and Ley TJ
- Published
- 2022
- Full Text
- View/download PDF
30. Convergent Clonal Evolution of Signaling Gene Mutations Is a Hallmark of Myelodysplastic Syndrome Progression.
- Author
-
Menssen AJ, Khanna A, Miller CA, Nonavinkere Srivatsan S, Chang GS, Shao J, Robinson J, O'Laughlin M, Fronick CC, Fulton RS, Brendel K, Heath SE, Saba R, Welch JS, Spencer DH, Payton JE, Westervelt P, DiPersio JF, Link DC, Schuelke MJ, Jacoby MA, Duncavage EJ, Ley TJ, and Walter MJ
- Subjects
- Clonal Evolution genetics, Disease Progression, Humans, Mutation genetics, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes genetics, Neoplasms, Second Primary
- Abstract
Progression from myelodysplastic syndromes (MDS) to secondary acute myeloid leukemia (AML) is associated with the acquisition and expansion of subclones. Our understanding of subclone evolution during progression, including the frequency and preferred order of gene mutation acquisition, remains incomplete. Sequencing of 43 paired MDS and secondary AML samples identified at least one signaling gene mutation in 44% of MDS and 60% of secondary AML samples, often below the level of standard sequencing detection. In addition, 19% of MDS and 47% of secondary AML patients harbored more than one signaling gene mutation, almost always in separate, coexisting subclones. Signaling gene mutations demonstrated diverse patterns of clonal evolution during disease progression, including acquisition, expansion, persistence, and loss of mutations, with multiple patterns often coexisting in the same patient. Multivariate analysis revealed that MDS patients who had a signaling gene mutation had a higher risk of AML progression, potentially providing a biomarker for progression., Significance: Subclone expansion is a hallmark of progression from MDS to secondary AML. Subclonal signaling gene mutations are common at MDS (often at low levels), show complex and convergent patterns of clonal evolution, and are associated with future progression to secondary AML. See related article by Guess et al., p. 316 (33). See related commentary by Romine and van Galen, p. 270. This article is highlighted in the In This Issue feature, p. 265., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
31. Recurrent transcriptional responses in AML and MDS patients treated with decitabine.
- Author
-
Upadhyay P, Beales J, Shah NM, Gruszczynska A, Miller CA, Petti AA, Ramakrishnan SM, Link DC, Ley TJ, and Welch JS
- Subjects
- Humans, Interferons, Recurrence, Decitabine therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes genetics
- Abstract
The molecular events responsible for decitabine responses in myelodysplastic syndrome and acute myeloid leukemia patients are poorly understood. Decitabine has a short serum half-life and limited stability in tissue culture. Therefore, theoretical pharmacologic differences may exist between patient molecular changes in vitro and the consequences of in vivo treatment. To systematically identify the global genomic and transcriptomic alterations induced by decitabine in vivo, we evaluated primary bone marrow samples that were collected during patient treatment and applied whole-genome bisulfite sequencing, RNA-sequencing, and single-cell RNA sequencing. Decitabine induced global, reversible hypomethylation after 10 days of therapy in all patients, which was associated with induction of interferon-induced pathways, the expression of endogenous retroviral elements, and inhibition of erythroid-related transcripts, recapitulating many effects seen previously in in vitro studies. However, at relapse after decitabine treatment, interferon-induced transcripts remained elevated relative to day 0, but erythroid-related transcripts now were more highly expressed than at day 0. Clinical responses were not correlated with epigenetic or transcriptional signatures, although sample size and interpatient variance restricted the statistical power required for capturing smaller effects. Collectively, these data define global hypomethylation by decitabine and find that erythroid-related pathways may be relevant because they are inhibited by therapy and reverse at relapse., Competing Interests: Conflict of interest disclosure JSW receives research funding from Janssen Pharmaceuticals and Notable Labs., (Copyright © 2022 ISEH -- Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
32. Decitabine salvage for TP53 -mutated, relapsed/refractory acute myeloid leukemia after cytotoxic induction therapy.
- Author
-
Ferraro F, Gruszczynska A, Ruzinova MB, Miller CA, Percival ME, Uy GL, Pusic I, Jacoby MA, Christopher MJ, Kim MY, Westervelt P, Cashen AF, Schroeder MA, DiPersio JF, Abboud CN, Wartman LD, Gao F, Link DC, Ley TJ, and Welch JS
- Subjects
- Antineoplastic Combined Chemotherapy Protocols, Decitabine therapeutic use, Humans, Induction Chemotherapy, Remission Induction, Salvage Therapy, Treatment Outcome, Tumor Suppressor Protein p53 genetics, Antineoplastic Agents therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Published
- 2022
- Full Text
- View/download PDF
33. Physician-scientists in the United States at 2020: Trends and concerns.
- Author
-
Garrison HH and Ley TJ
- Subjects
- Career Choice, Humans, United States, Workforce, Biomedical Research education, Physicians
- Abstract
Physician-scientists comprise a unique and valuable part of the biomedical workforce, but for decades there has been concern about the number of physicians actively engaged in research. Reports have outlined the challenges facing physician-scientists, and programs have been initiated to encourage and facilitate research careers for medically trained scientists. Many of these initiatives have demonstrated successful outcomes, but there has not been a recent summary of the impact of the past decade of effort. This report compiles available data from surveys of medical education and physician research participation to assess changes in the physician-scientist workforce from 2011-2020. Several trends are positive: rising enrollments in MD-PhD programs, greater levels of interest in research careers among matriculating medical students, more research experience during medical school and rising numbers of physicians in academic medicine, and an increase in first R01 grants to physician-scientists. However, there are now decreased levels of interest in research careers among graduating medical students, a steady decline in MDs applying for NIH loan repayment program support, an increased age at first R01 grant success for physicians, and fewer physicians reporting research as their primary work activity: all of these indicators create concern for the stability of the career path. Despite a recommendation by the Physician-Scientist Workforce in 2014 to create "real-time" reporting on NIH grants and grantees to help the public assess trends, this initiative has not been completed. Better information is still needed to fully understand the status of the physician-scientist workforce, and to assess efforts to stabilize this vulnerable career path., (© 2022 The Authors. The FASEB Journal published by Wiley Periodicals LLC on behalf of Federation of American Societies for Experimental Biology.)
- Published
- 2022
- Full Text
- View/download PDF
34. Focal disruption of DNA methylation dynamics at enhancers in IDH-mutant AML cells.
- Author
-
Wilson ER, Helton NM, Heath SE, Fulton RS, Payton JE, Welch JS, Walter MJ, Westervelt P, DiPersio JF, Link DC, Miller CA, Ley TJ, and Spencer DH
- Subjects
- Humans, Isocitrate Dehydrogenase genetics, Mutation, Regulatory Sequences, Nucleic Acid, DNA Methylation, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology
- Abstract
Recurrent mutations in IDH1 or IDH2 in acute myeloid leukemia (AML) are associated with increased DNA methylation, but the genome-wide patterns of this hypermethylation phenotype have not been comprehensively studied in AML samples. We analyzed whole-genome bisulfite sequencing data from 15 primary AML samples with IDH1 or IDH2 mutations, which identified ~4000 focal regions that were uniquely hypermethylated in IDH
mut samples vs. normal CD34+ cells and other AMLs. These regions had modest hypermethylation in AMLs with biallelic TET2 mutations, and levels of 5-hydroxymethylation that were diminished in IDH and TET-mutant samples, indicating that this hypermethylation results from inhibition of TET-mediated demethylation. Focal hypermethylation in IDHmut AMLs occurred at regions with low methylation in CD34+ cells, implying that DNA methylation and demethylation are active at these loci. AML samples containing IDH and DNMT3AR882 mutations were significantly less hypermethylated, suggesting that IDHmut -associated hypermethylation is mediated by DNMT3A. IDHmut -specific hypermethylation was highly enriched for enhancers that form direct interactions with genes involved in normal hematopoiesis and AML, including MYC and ETV6. These results suggest that focal hypermethylation in IDH-mutant AML occurs by altering the balance between DNA methylation and demethylation, and that disruption of these pathways at enhancers may contribute to AML pathogenesis., (© 2021. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
35. Somatic Dnmt3a inactivation leads to slow, canonical DNA methylation loss in murine hematopoietic cells.
- Author
-
Smith AM, Verdoni AM, Abel HJ, Chen DY, Ketkar S, Leight ER, Miller CA, and Ley TJ
- Abstract
Mutations in the gene encoding DNA methyltransferase 3A ( DNMT3A ) are the most common cause of clonal hematopoiesis and are among the most common initiating events of acute myeloid leukemia (AML). Studies in germline and somatic Dnmt3a knockout mice have identified focal, canonical hypomethylation phenotypes in hematopoietic cells; however, the kinetics of methylation loss following acquired DNMT3A inactivation in hematopoietic cells is essentially unknown. Therefore, we evaluated a somatic, inducible model of hematopoietic Dnmt3a loss, and show that inactivation of Dnmt3a in murine hematopoietic cells results in a relatively slow loss of methylation at canonical sites throughout the genome; in contrast, remethylation of Dnmt3a deficient genomes in hematopoietic cells occurs much more quickly. This data suggests that slow methylation loss may contribute, at least in part, to the long latent period that characterizes clonal expansion and leukemia development in individuals with acquired DNMT3A mutations in hematopoietic stem cells., Competing Interests: A.M.S. is an employee of Incyte Corporation, A.M.V. is an employee of Vitalant Corporation, and E.R.L. is the owner of Leight Medical Communications, LLC. All studies in the manuscript were performed before the authors joined these companies. The other authors declare no competing interests., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
36. Failure to Detect Mutations in U2AF1 due to Changes in the GRCh38 Reference Sequence.
- Author
-
Miller CA, Walker JR, Jensen TL, Hooper WF, Fulton RS, Painter JS, Sekeres MA, Ley TJ, Spencer DH, Goll JB, and Walter MJ
- Subjects
- Humans, Mutation, Splicing Factor U2AF genetics, Leukemia, Myeloid, Acute genetics
- Abstract
The U2AF1 gene is a core part of mRNA splicing machinery and frequently contains somatic mutations that contribute to oncogenesis in myelodysplastic syndrome, acute myeloid leukemia, and other cancers. A change introduced in the GRCh38 version of the human reference build prevents detection of mutations in this gene, and others, by variant calling pipelines. This study describes the problem in detail and shows that a modified GRCh38 reference build with unchanged coordinates can be used to ameliorate the issue., (Copyright © 2022 Association for Molecular Pathology and American Society for Investigative Pathology. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
37. DNMT3A overgrowth syndrome is associated with the development of hematopoietic malignancies in children and young adults.
- Author
-
Ferris MA, Smith AM, Heath SE, Duncavage EJ, Oberley M, Freyer D, Wynn R, Douzgou S, Maris JM, Reilly AF, Wu MD, Choo F, Fiets RB, Koene S, Spencer DH, Miller CA, Shinawi M, and Ley TJ
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Congenital Abnormalities genetics, Female, Germ-Line Mutation, Humans, Male, Syndrome, Young Adult, DNA Methyltransferase 3A genetics, Hematologic Neoplasms genetics, Mutation
- Published
- 2022
- Full Text
- View/download PDF
38. Tumor suppressor function of WT1 in acute promyelocytic leukemia.
- Author
-
Christopher MJ, Katerndahl CDS, LeBlanc HR, Elmendorf TT, Basu V, Gang M, Menssen AJ, Spencer DH, Duncavage EJ, Ketkar S, Wartman LD, Ramakrishnan SM, Miller CA, and Ley TJ
- Subjects
- Genes, Tumor Suppressor, Humans, Leukemia, Promyelocytic, Acute genetics, WT1 Proteins genetics
- Published
- 2022
- Full Text
- View/download PDF
39. Genetic and Transcriptional Contributions to Relapse in Normal Karyotype Acute Myeloid Leukemia.
- Author
-
Petti AA, Khan SM, Xu Z, Helton N, Fronick CC, Fulton R, Ramakrishnan SM, Nonavinkere Srivatsan S, Heath SE, Westervelt P, Payton JE, Walter MJ, Link DC, DiPersio J, Miller C, and Ley TJ
- Subjects
- Clonal Evolution, Humans, Karyotype, Mutation, Recurrence, Tumor Microenvironment, Leukemia, Myeloid, Acute diagnosis
- Abstract
To better understand clonal and transcriptional adaptations after relapse in patients with acute myeloid leukemia (AML), we collected presentation and relapse samples from six normal karyotype AML cases. We performed enhanced whole-genome sequencing to characterize clonal evolution, and deep-coverage single-cell RNA sequencing on the same samples, which yielded 142,642 high-quality cells for analysis. Identifying expressed mutations in individual cells enabled us to discriminate between normal and AML cells, to identify coordinated changes in the genome and transcriptome, and to identify subclone-specific cell states. We quantified the coevolution of genetic and transcriptional heterogeneity during AML progression, and found that transcriptional changes were significantly correlated with genetic changes. However, transcriptional adaptation sometimes occurred independently, suggesting that clonal evolution does not represent all relevant biological changes. In three cases, we identified cells at diagnosis that likely seeded the relapse. Finally, these data revealed a conserved relapse-enriched leukemic cell state bearing markers of stemness, quiescence, and adhesion. SIGNIFICANCE: These data enabled us to identify a relapse-enriched leukemic cell state with distinct transcriptional properties. Detailed case-by-case analyses elucidated the complex ways in which the AML genome, transcriptome, and immune microenvironment interact to evade chemotherapy. These analyses provide a blueprint for evaluating these factors in larger cohorts. This article is highlighted in the In This Issue feature, p. 1 ., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
40. Immunosuppression and outcomes in adult patients with de novo acute myeloid leukemia with normal karyotypes.
- Author
-
Ferraro F, Miller CA, Christensen KA, Helton NM, O'Laughlin M, Fronick CC, Fulton RS, Kohlschmidt J, Eisfeld AK, Bloomfield CD, Ramakrishnan SM, Day RB, Wartman LD, Uy GL, Welch JS, Christopher MJ, Heath SE, Baty JD, Schuelke MJ, Payton JE, Spencer DH, Rettig MP, Link DC, Walter MJ, Westervelt P, DiPersio JF, and Ley TJ
- Subjects
- Adult, CD4-Positive T-Lymphocytes immunology, Female, Humans, Immune Tolerance immunology, Karyotype, Leukemia, Myeloid, Acute therapy, Male, Middle Aged, Prognosis, Recurrence, Remission Induction, Risk Factors, Sequence Analysis, RNA methods, Th1 Cells immunology, Transcriptome genetics, Treatment Outcome, Immune Tolerance genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute immunology
- Abstract
Acute myeloid leukemia (AML) patients rarely have long first remissions (LFRs; >5 y) after standard-of-care chemotherapy, unless classified as favorable risk at presentation. Identification of the mechanisms responsible for long vs. more typical, standard remissions may help to define prognostic determinants for chemotherapy responses. Using exome sequencing, RNA-sequencing, and functional immunologic studies, we characterized 28 normal karyotype (NK)-AML patients with >5 y first remissions after chemotherapy (LFRs) and compared them to a well-matched group of 31 NK-AML patients who relapsed within 2 y (standard first remissions [SFRs]). Our combined analyses indicated that genetic-risk profiling at presentation (as defined by European LeukemiaNet [ELN] 2017 criteria) was not sufficient to explain the outcomes of many SFR cases. Single-cell RNA-sequencing studies of 15 AML samples showed that SFR AML cells differentially expressed many genes associated with immune suppression. The bone marrow of SFR cases had significantly fewer CD4
+ Th1 cells; these T cells expressed an exhaustion signature and were resistant to activation by T cell receptor stimulation in the presence of autologous AML cells. T cell activation could be restored by removing the AML cells or blocking the inhibitory major histocompatibility complex class II receptor, LAG3. Most LFR cases did not display these features, suggesting that their AML cells were not as immunosuppressive. These findings were confirmed and extended in an independent set of 50 AML cases representing all ELN 2017 risk groups. AML cell-mediated suppression of CD4+ T cell activation at presentation is strongly associated with unfavorable outcomes in AML patients treated with standard chemotherapy., Competing Interests: The authors declare no competing interest.- Published
- 2021
- Full Text
- View/download PDF
41. Tumor suppressor function of Gata2 in acute promyelocytic leukemia.
- Author
-
Katerndahl CDS, Rogers ORS, Day RB, Cai MA, Rooney TP, Helton NM, Hoock M, Ramakrishnan SM, Nonavinkere Srivatsan S, Wartman LD, Miller CA, and Ley TJ
- Subjects
- Animals, CRISPR-Cas Systems, Cell Line, Tumor, Disease Progression, GATA2 Transcription Factor metabolism, Gene Expression Regulation, Leukemic, Genes, Tumor Suppressor, Humans, Leukemia, Promyelocytic, Acute metabolism, Leukemia, Promyelocytic, Acute pathology, Mice, Mutation, GATA2 Transcription Factor genetics, Leukemia, Promyelocytic, Acute genetics
- Abstract
Most patients with acute promyelocytic leukemia (APL) can be cured with combined all-trans retinoic acid (ATRA) and arsenic trioxide therapy, which induces the destruction of PML-RARA, the initiating fusion protein for this disease. However, the underlying mechanisms by which PML-RARA initiates and maintains APL cells are still not clear. Therefore, we identified genes that are dysregulated by PML-RARA in mouse and human APL cells and prioritized GATA2 for functional studies because it is highly expressed in preleukemic cells expressing PML-RARA, its high expression persists in transformed APL cells, and spontaneous somatic mutations of GATA2 occur during APL progression in mice and humans. These and other findings suggested that GATA2 may be upregulated to thwart the proliferative signal generated by PML-RARA and that its inactivation by mutation (and/or epigenetic silencing) may accelerate disease progression in APL and other forms of acute myeloid leukemia (AML). Indeed, biallelic knockout of Gata2 with CRISPR/Cas9-mediated gene editing increased the serial replating efficiency of PML-RARA-expressing myeloid progenitors (as well as progenitors expressing RUNX1-RUNX1T1, or deficient for Cebpa), increased mouse APL penetrance, and decreased latency. Restoration of Gata2 expression suppressed PML-RARA-driven aberrant self-renewal and leukemogenesis. Conversely, addback of a mutant GATA2R362G protein associated with APL and AML minimally suppressed PML-RARA-induced aberrant self-renewal, suggesting that it is a loss-of-function mutation. These studies reveal a potential role for Gata2 as a tumor suppressor in AML and suggest that restoration of its function (when inactivated) may provide benefit for AML patients., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
42. Bam-readcount - rapid generation of basepair-resolution sequence metrics.
- Author
-
Khanna A, Larson DE, Srivatsan SN, Mosior M, Abbott TE, Kiwala S, Ley TJ, Duncavage EJ, Walter MJ, Walker JR, Griffith OL, Griffith M, and Miller CA
- Abstract
Bam-readcount is a utility for generating low-level information about sequencing data at specific nucleotide positions. Originally designed to help filter genomic mutation calls, the metrics it outputs are useful as input for variant detection tools and for resolving ambiguity between variant callers
1,2 . In addition, it has found broad applicability in diverse fields including tumor evolution, single-cell genomics, climate change ecology, and tracking community spread of SARS-CoV-2.3-6 .- Published
- 2021
43. Functional and epigenetic phenotypes of humans and mice with DNMT3A Overgrowth Syndrome.
- Author
-
Smith AM, LaValle TA, Shinawi M, Ramakrishnan SM, Abel HJ, Hill CA, Kirkland NM, Rettig MP, Helton NM, Heath SE, Ferraro F, Chen DY, Adak S, Semenkovich CF, Christian DL, Martin JR, Gabel HW, Miller CA, and Ley TJ
- Subjects
- Abnormalities, Multiple blood, Adolescent, Adult, Animals, Behavior, Animal, Body Weight genetics, Bone Marrow Cells metabolism, Child, Child, Preschool, CpG Islands genetics, DNA Methylation genetics, DNA Methyltransferase 3A, Female, Gene Expression Profiling, Germ-Line Mutation genetics, Hematopoiesis genetics, Hematopoietic Stem Cells metabolism, Humans, Infant, Leukemia genetics, Leukemia pathology, Male, Mice, Inbred C57BL, Obesity genetics, Phenotype, Syndrome, Transcription, Genetic, Mice, Abnormalities, Multiple genetics, DNA (Cytosine-5-)-Methyltransferases genetics, Epigenesis, Genetic
- Abstract
Germline pathogenic variants in DNMT3A were recently described in patients with overgrowth, obesity, behavioral, and learning difficulties (DNMT3A Overgrowth Syndrome/DOS). Somatic mutations in the DNMT3A gene are also the most common cause of clonal hematopoiesis, and can initiate acute myeloid leukemia (AML). Using whole genome bisulfite sequencing, we studied DNA methylation in peripheral blood cells of 11 DOS patients and found a focal, canonical hypomethylation phenotype, which is most severe with the dominant negative DNMT3A
R882H mutation. A germline mouse model expressing the homologous Dnmt3aR878H mutation phenocopies most aspects of the human DOS syndrome, including the methylation phenotype and an increased incidence of spontaneous hematopoietic malignancies, suggesting that all aspects of this syndrome are caused by this mutation., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
44. Co-evolution of tumor and immune cells during progression of multiple myeloma.
- Author
-
Liu R, Gao Q, Foltz SM, Fowles JS, Yao L, Wang JT, Cao S, Sun H, Wendl MC, Sethuraman S, Weerasinghe A, Rettig MP, Storrs EP, Yoon CJ, Wyczalkowski MA, McMichael JF, Kohnen DR, King J, Goldsmith SR, O'Neal J, Fulton RS, Fronick CC, Ley TJ, Jayasinghe RG, Fiala MA, Oh ST, DiPersio JF, Vij R, and Ding L
- Subjects
- Aged, B-Lymphocytes cytology, B-Lymphocytes immunology, Cell Lineage, Clonal Evolution genetics, Cohort Studies, Disease Progression, Female, Gene Expression Regulation, Neoplastic immunology, Haplotypes, Humans, Interleukin-1beta blood, Interleukin-6 blood, Male, Mass Spectrometry, Middle Aged, Multigene Family, Multiple Myeloma blood, Multiple Myeloma pathology, Mutation, Neoplasm Recurrence, Local blood, Neoplasm Recurrence, Local immunology, Proto-Oncogene Proteins c-fos blood, Proto-Oncogene Proteins c-jun blood, RNA-Seq, Signal Transduction genetics, Signal Transduction immunology, Single-Cell Analysis, B-Lymphocytes metabolism, Gene Expression Regulation, Neoplastic genetics, Multiple Myeloma genetics, Multiple Myeloma immunology, Neoplasm Recurrence, Local genetics, Tumor Microenvironment immunology
- Abstract
Multiple myeloma (MM) is characterized by the uncontrolled proliferation of plasma cells. Despite recent treatment advances, it is still incurable as disease progression is not fully understood. To investigate MM and its immune environment, we apply single cell RNA and linked-read whole genome sequencing to profile 29 longitudinal samples at different disease stages from 14 patients. Here, we collect 17,267 plasma cells and 57,719 immune cells, discovering patient-specific plasma cell profiles and immune cell expression changes. Patients with the same genetic alterations tend to have both plasma cells and immune cells clustered together. By integrating bulk genomics and single cell mapping, we track plasma cell subpopulations across disease stages and find three patterns: stability (from precancer to diagnosis), and gain or loss (from diagnosis to relapse). In multiple patients, we detect "B cell-featured" plasma cell subpopulations that cluster closely with B cells, implicating their cell of origin. We validate AP-1 complex differential expression (JUN and FOS) in plasma cell subpopulations using CyTOF-based protein assays, and integrated analysis of single-cell RNA and CyTOF data reveals AP-1 downstream targets (IL6 and IL1B) potentially leading to inflammation regulation. Our work represents a longitudinal investigation for tumor and microenvironment during MM progression and paves the way for expanding treatment options.
- Published
- 2021
- Full Text
- View/download PDF
45. Dnmt3a deficiency in the skin causes focal, canonical DNA hypomethylation and a cellular proliferation phenotype.
- Author
-
Chen DY, Ferguson IM, Braun KA, Sutton LA, Helton NM, Ramakrishnan SM, Smith AM, Miller CA, and Ley TJ
- Subjects
- Abnormalities, Multiple genetics, Adolescent, Animals, Child, DNA metabolism, DNA (Cytosine-5-)-Methyltransferases genetics, DNA Methyltransferase 3A metabolism, DNA Modification Methylases metabolism, Germ-Line Mutation, Heterozygote, Humans, Intellectual Disability genetics, Keratinocytes physiology, Male, Methyltransferases genetics, Mice, Mutation, Phenotype, Skin metabolism, Syndrome, DNA Methylation physiology, DNA Methyltransferase 3A genetics, Keratinocytes metabolism
- Abstract
DNA hypomethylation is a feature of epidermal cells from aged and sun-exposed skin, but the mechanisms responsible for this methylation loss are not known. Dnmt3a is the dominant de novo DNA methyltransferase in the skin; while epidermal Dnmt3a deficiency creates a premalignant state in which keratinocytes are more easily transformed by topical mutagens, the conditions responsible for this increased susceptibility to transformation are not well understood. Using whole genome bisulfite sequencing, we identified a focal, canonical DNA hypomethylation phenotype in the epidermal cells of Dnmt3a-deficient mice. Single-cell transcriptomic analysis revealed an increased proportion of cells with a proliferative gene expression signature, while other populations in the skin were relatively unchanged. Although total DNMT3A deficiency has not been described in human disease states, rare patients with an overgrowth syndrome associated with behavioral abnormalities and an increased risk of cancer often have heterozygous, germline mutations in DNMT3A that reduce its function (Tatton-Brown Rahman syndrome [TBRS]). We evaluated the DNA methylation phenotype of the skin from a TBRS patient with a germline DNMT3A
R882H mutation, which encodes a dominant-negative protein that reduces its methyltransferase function by ∼80%. We detected a focal, canonical hypomethylation phenotype that revealed considerable overlap with hypomethylated regions found in Dnmt3a-deficient mouse skin. Together, these data suggest that DNMT3A loss creates a premalignant epigenetic state associated with a hyperproliferative phenotype in the skin and further suggest that DNMT3A acts as a tumor suppressor in the skin., Competing Interests: The authors declare no competing interest., (Copyright © 2021 the Author(s). Published by PNAS.)- Published
- 2021
- Full Text
- View/download PDF
46. Genome Sequencing as an Alternative to Cytogenetic Analysis in Myeloid Cancers.
- Author
-
Duncavage EJ, Schroeder MC, O'Laughlin M, Wilson R, MacMillan S, Bohannon A, Kruchowski S, Garza J, Du F, Hughes AEO, Robinson J, Hughes E, Heath SE, Baty JD, Neidich J, Christopher MJ, Jacoby MA, Uy GL, Fulton RS, Miller CA, Payton JE, Link DC, Walter MJ, Westervelt P, DiPersio JF, Ley TJ, and Spencer DH
- Subjects
- Feasibility Studies, Female, Humans, Male, Middle Aged, Proportional Hazards Models, Survival Analysis, Cytogenetic Analysis, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes genetics, Whole Genome Sequencing methods
- Abstract
Background: Genomic analysis is essential for risk stratification in patients with acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS). Whole-genome sequencing is a potential replacement for conventional cytogenetic and sequencing approaches, but its accuracy, feasibility, and clinical utility have not been demonstrated., Methods: We used a streamlined whole-genome sequencing approach to obtain genomic profiles for 263 patients with myeloid cancers, including 235 patients who had undergone successful cytogenetic analysis. We adapted sample preparation, sequencing, and analysis to detect mutations for risk stratification using existing European Leukemia Network (ELN) guidelines and to minimize turnaround time. We analyzed the performance of whole-genome sequencing by comparing our results with findings from cytogenetic analysis and targeted sequencing., Results: Whole-genome sequencing detected all 40 recurrent translocations and 91 copy-number alterations that had been identified by cytogenetic analysis. In addition, we identified new clinically reportable genomic events in 40 of 235 patients (17.0%). Prospective sequencing of samples obtained from 117 consecutive patients was performed in a median of 5 days and provided new genetic information in 29 patients (24.8%), which changed the risk category for 19 patients (16.2%). Standard AML risk groups, as defined by sequencing results instead of cytogenetic analysis, correlated with clinical outcomes. Whole-genome sequencing was also used to stratify patients who had inconclusive results by cytogenetic analysis into risk groups in which clinical outcomes were measurably different., Conclusions: In our study, we found that whole-genome sequencing provided rapid and accurate genomic profiling in patients with AML or MDS. Such sequencing also provided a greater diagnostic yield than conventional cytogenetic analysis and more efficient risk stratification on the basis of standard risk categories. (Funded by the Siteman Cancer Research Fund and others.)., (Copyright © 2021 Massachusetts Medical Society.)
- Published
- 2021
- Full Text
- View/download PDF
47. Enhanced Efficacy and Increased Long-Term Toxicity of CNS-Directed, AAV-Based Combination Therapy for Krabbe Disease.
- Author
-
Li Y, Miller CA, Shea LK, Jiang X, Guzman MA, Chandler RJ, Ramakrishnan SM, Smith SN, Venditti CP, Vogler CA, Ory DS, Ley TJ, and Sands MS
- Subjects
- Animals, Bone Marrow Transplantation methods, Carcinoma, Hepatocellular etiology, Combined Modality Therapy, Disease Models, Animal, Genetic Therapy methods, Genetic Vectors administration & dosage, Liver Neoplasms etiology, Mice, Dependovirus genetics, Genetic Therapy adverse effects, Genetic Vectors genetics, Leukodystrophy, Globoid Cell complications, Leukodystrophy, Globoid Cell therapy
- Abstract
Infantile globoid cell leukodystrophy (GLD, Krabbe disease) is a demyelinating disease caused by the deficiency of the lysosomal enzyme galactosylceramidase (GALC) and the progressive accumulation of the toxic metabolite psychosine. We showed previously that central nervous system (CNS)-directed, adeno-associated virus (AAV)2/5-mediated gene therapy synergized with bone marrow transplantation and substrate reduction therapy (SRT) to greatly increase therapeutic efficacy in the murine model of Krabbe disease (Twitcher). However, motor deficits remained largely refractory to treatment. In the current study, we replaced AAV2/5 with an AAV2/9 vector. This single change significantly improved several endpoints primarily associated with motor function. However, nearly all (14/16) of the combination-treated Twitcher mice and all (19/19) of the combination-treated wild-type mice developed hepatocellular carcinoma (HCC). 10 out of 10 tumors analyzed had AAV integrations within the Rian locus. Several animals had additional integrations within or near genes that regulate cell growth or death, are known or potential tumor suppressors, or are associated with poor prognosis in human HCC. Finally, the substrate reduction drug L-cycloserine significantly decreased the level of the pro-apoptotic ceramide 18:0. These data demonstrate the value of AAV-based combination therapy for Krabbe disease. However, they also suggest that other therapies or co-morbidities must be taken into account before AAV-mediated gene therapy is considered for human therapeutic trials., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The American Society of Gene and Cell Therapy. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
48. Impact of a 40-Gene Targeted Panel Test on Physician Decision Making for Patients With Acute Myeloid Leukemia.
- Author
-
Barnell EK, Newcomer KF, Skidmore ZL, Krysiak K, Anderson SR, Wartman LD, Oh ST, Welch JS, Stockerl-Goldstein KE, Vij R, Cashen AF, Pusic I, Westervelt P, Abboud CN, Ghobadi A, Uy GL, Schroeder MA, Dipersio JF, Politi MC, Spencer DH, Duncavage EJ, Ley TJ, Griffith M, Jacoby MA, and Griffith OL
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Young Adult, Clinical Decision-Making, Genetic Testing methods, Leukemia, Myeloid, Acute genetics
- Abstract
Purpose: Physicians treating hematologic malignancies increasingly order targeted sequencing panels to interrogate recurrently mutated genes. The precise impact of these panels on clinical decision making is not well understood., Methods: Here, we report our institutional experience with a targeted 40-gene panel (MyeloSeq) that is used to generate a report for both genetic variants and variant allele frequencies for the treating physician (the limit of mutation detection is approximately one AML cell in 50)., Results: In total, 346 sequencing reports were generated for 325 patients with suspected hematologic malignancies over an 8-month period (August 2018 to April 2019). To determine the influence of genomic data on clinical care for patients with acute myeloid leukemia (AML), we analyzed 122 consecutive reports from 109 patients diagnosed with AML and surveyed the treating physicians with a standardized questionnaire. The panel was ordered most commonly at diagnosis (61.5%), but was also used to assess response to therapy (22.9%) and to detect suspected relapse (15.6%). The panel was ordered at multiple timepoints during the disease course for 11% of patients. Physicians self-reported that 50 of 114 sequencing reports (44%) influenced clinical care decisions in 44 individual patients. Influences were often nuanced and extended beyond identifying actionable genetic variants with US Food and Drug Administration-approved drugs., Conclusion: This study provides insights into how physicians are currently using multigene panels capable of detecting relatively rare AML cells. The most influential way to integrate these tools into clinical practice will be to perform prospective clinical trials that assess patient outcomes in response to genomically driven interventions., Competing Interests: The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/author-center. Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments). Erica K. BarnellEmployment: Geneoscopy Stock and Other Ownership Interests: Geneoscopy Patents, Royalties, Other Intellectual Property: Inventor of intellectual property in start-up company (Geneoscopy) Travel, Accommodations, Expenses: GeneoscopyKenneth F. NewcomerEmployment: Geneoscopy (I) Stock and Other Ownership Interests: Geneoscopy (I) Patents, Royalties, Other Intellectual Property: Inventor of intellectual property owned by Geneoscopy (I)Zachary L. SkidmoreStock and Other Ownership Interests: Aim Immunotech, Catalyst PharmaceuticalsKilannin KrysiakConsulting or Advisory Role: Gerson Lehrman GroupLukas D. WartmanConsulting or Advisory Role: Novartis, IncyteStephen T. OhConsulting or Advisory Role: Incyte, Novartis, Blueprint Medicines, Celgene, Kartos, CTI BioPharma Corp, PharmaEssentia, Disc Medicine, Constellation Pharmaceuticals Research Funding: Incyte (Inst), Gilead Sciences (Inst), CTI BioPharma Corp (Inst), Kartos (Inst), Celgene (Inst), Sierra Oncology (Inst), Blueprint Medicines (Inst), Constellation Pharmaceuticals (Inst)John S. WelchConsulting or Advisory Role: Archer Biosciences, Agios Research Funding: Janssen Oncology (Inst), Notable Labs (Inst)Keith E. Stockerl-GoldsteinStock and Other Ownership Interests: Abbott Laboratories, AbbVie Consulting or Advisory Role: Celgene Research Funding: GlaxoSmithKline, Takeda, BiolineRx, Janssen Other Relationship: CellerantRavi VijConsulting or Advisory Role: Bristol Myers Squibb, Celgene, Janssen, Sanofi, Karyopharm Therapeutics, Takeda, Genentech, AbbVie, Oncopeptides Research Funding: Takeda, Celgene, Bristol Myers Squibb Travel, Accommodations, Expenses: Celgene, Bristol Myers Squibb, Sanofi, Janssen, DAVAOncology, Karyopharm Therapeutics, Amgen, Takeda, AbbVieAmanda F. CashenConsulting or Advisory Role: Agios Speakers' Bureau: Novartis, CelgeneIskra PusicConsulting or Advisory Role: Kadmon, Incyte, SyndaxCamille N. AbboudStock and Other Ownership Interests: AbbVie (I), Abbott Laboratories (I), Gilead Sciences (I), Bristol Myers Squibb (I), Johnson & Johnson (I) Honoraria: Cardinal Health, Dova Pharmaceuticals, NkartaTherapeutics, Archer Biosciences Research Funding: Actinium Pharmaceuticals, Selvita (Inst), AlloVir (Inst), Forty-Seven (Inst)Armin GhobadiHonoraria: Kite Pharma Consulting or Advisory Role: Kite Pharma, Celgene, Amgen, Wugen Speakers' Bureau: Kite Pharma, AmgenGeoffrey L. UyConsulting or Advisory Role: Astellas Pharma, Genentech, Jazz PharmaceuticalsMark A. SchroederConsulting or Advisory Role: Astellas Pharma, Dova Pharmaceuticals, Flatiron Health, GlaxoSmithKline, Incyte, Partners Therapeutics, Partners Therapeutics, Pfizer Speakers' Bureau: AbbVie, Merck, TakedaJohn F. DiPersioStock and Other Ownership Interests: Magenta Therapeutics, Wugen Honoraria: Incyte Consulting or Advisory Role: Cellworks, Rivervest, Magenta Therapeutics, Incyte Research Funding: Amphivena Therapeutics (Inst), Macrogenics (Inst), Incyte (Inst), Wugen (Inst), BiolineRx (Inst), Maxcyte (Inst), Bigelow Aerospace (Inst) Patents, Royalties, Other Intellectual Property: Patents: CD7 and CD2 knockout for CART to CD7 and CDL; Duvelisib for treatment of cytokine release syndrome; NT-17 to enhance CART survival; Novel WU mobilizing compounds (Inst); Selection of IMPDH mutant stem cells; IFNγ, upregulate MHCII for relapsed AML; Dextran-based molecules to detect CAR-T cells; Combining integrin inhibitor with chemokine binders, 016131; JAK and calcineurin inhibition, solid organ transplant; VLA4, gro-b; Triple combination–CXCR2, VLA-4, gro-b; Targeting IFNR/CSCR3 in GvHD; WU/SLU compounds VLA4 and CXCR2 (Inst) Travel, Accommodations, Expenses: Incyte, Macrogenics, Magenta TherapeuticsMary C. PolitiResearch Funding: MerckDavid H. SpencerConsulting or Advisory Role: Wugen Research Funding: IlluminaEric J. DuncavageStock and Other Ownership Interests: P&V Licensing Consulting or Advisory Role: Cofactor Genomics, Bristol Myers Squibb, Eli Lilly Open Payments Link: https://openpaymentsdata.cms.gov/physician/317379Timothy J. LeyResearch Funding: Rigel Patents, Royalties, Other Intellectual Property: Licenses for monoclonal antibodies and recombinant granzymesMeagan A. JacobyHonoraria: Takeda Research Funding: Geron (Inst), AbbVie (Inst), Jazz Pharmaceuticals (Inst), Aprea AB (Inst), Amphivena (Inst), Janssen (Inst) No other potential conflicts of interest were reported., (© 2021 by American Society of Clinical Oncology.)
- Published
- 2021
- Full Text
- View/download PDF
49. The clonal evolution of metastatic colorectal cancer.
- Author
-
Dang HX, Krasnick BA, White BS, Grossman JG, Strand MS, Zhang J, Cabanski CR, Miller CA, Fulton RS, Goedegebuure SP, Fronick CC, Griffith M, Larson DE, Goetz BD, Walker JR, Hawkins WG, Strasberg SM, Linehan DC, Lim KH, Lockhart AC, Mardis ER, Wilson RK, Ley TJ, Maher CA, and Fields RC
- Subjects
- Animals, Disease Models, Animal, Exome genetics, Genomics, Humans, Neoplasm Metastasis, Exome Sequencing, Clonal Evolution genetics, Colonic Neoplasms genetics
- Abstract
Tumor heterogeneity and evolution drive treatment resistance in metastatic colorectal cancer (mCRC). Patient-derived xenografts (PDXs) can model mCRC biology; however, their ability to accurately mimic human tumor heterogeneity is unclear. Current genomic studies in mCRC have limited scope and lack matched PDXs. Therefore, the landscape of tumor heterogeneity and its impact on the evolution of metastasis and PDXs remain undefined. We performed whole-genome, deep exome, and targeted validation sequencing of multiple primary regions, matched distant metastases, and PDXs from 11 patients with mCRC. We observed intricate clonal heterogeneity and evolution affecting metastasis dissemination and PDX clonal selection. Metastasis formation followed both monoclonal and polyclonal seeding models. In four cases, metastasis-seeding clones were not identified in any primary region, consistent with a metastasis-seeding-metastasis model. PDXs underrepresented the subclonal heterogeneity of parental tumors. These suggest that single sample tumor sequencing and current PDX models may be insufficient to guide precision medicine., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2020
- Full Text
- View/download PDF
50. Obesity is a risk factor for acute promyelocytic leukemia: evidence from population and cross-sectional studies and correlation with FLT3 mutations and polyunsaturated fatty acid metabolism.
- Author
-
Mazzarella L, Botteri E, Matthews A, Gatti E, Di Salvatore D, Bagnardi V, Breccia M, Montesinos P, Bernal T, Gil C, Ley TJ, Sanz M, Bhaskaran K, Coco FL, and Pelicci PG
- Subjects
- Cross-Sectional Studies, Humans, Italy, Mutation, Risk Factors, Spain, Leukemia, Promyelocytic, Acute, Obesity epidemiology, fms-Like Tyrosine Kinase 3 genetics
- Abstract
Obesity correlates with hematologic malignancies including leukemias, but risk of specific leukemia subtypes like acute promyelocytic leukemia and underlying molecular mechanisms are poorly understood. We explored multiple datasets for correlation between leukemia, body mass index (BMI) and molecular features. In a population-based study (n=5.2 million), we correlated BMI with promyelocytic leukemia, and other acute myeloid, lymphoid or other leukemias. In cross-sectional studies, we tested BMI deviation in promyelocytic leukemia trial cohorts from that expected based on national surveys. We explored The Cancer Genome Atlas for transcriptional signatures and mutations enriched in promyelocytic leukemia and/or obesity, and confirmed a correlation between body mass and FLT3 mutations in promyelocytic leukemia cohorts by logistic regression. In the population-based study, hazard ratio per 5 kg/m
2 increase was: promyelocytic leukemia 1.44 (95%CI: 1.0-2.08), non-promyelocytic acute myeloid leukemias 1.17 (95%CI: 1.10-1.26), lymphoid leukemias 1.04 (95%CI: 1.0-1.09), other 1.10 (95%CI: 1.04-1.15). In cross-sectional studies, body mass deviated significantly from that expected (Italy: P <0.001; Spain: P =0.011; USA: P <0.001). Promyelocytic leukemia showed upregulation of polyunsaturated fatty acid metabolism genes. Odds of FLT3 mutations were higher in obese acute myeloid leukemias (odds ratio=2.4, P =0.007), whether promyelocytic or not, a correlation confirmed in the pooled promyelocytic leukemia cohorts (OR=1.22, 1.05-1.43 per 5 kg/m2 ). These results strengthen the evidence for obesity as a bona fide risk factor for myeloid leukemias, and in particular APL. FLT3 mutations and polyunsaturated fatty acid metabolism may play a previously under-appreciated role in obesity-associated leukemogenesis., (Copyright© 2020 Ferrata Storti Foundation.)- 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.