25 results on '"Jessica Schulman"'
Search Results
2. Small-molecule targeting of MUSASHI RNA-binding activity in acute myeloid leukemia
- Author
-
Gerard Minuesa, Steven K. Albanese, Wei Xie, Yaniv Kazansky, Daniel Worroll, Arthur Chow, Alexandra Schurer, Sun-Mi Park, Christina Z. Rotsides, James Taggart, Andrea Rizzi, Levi N. Naden, Timothy Chou, Saroj Gourkanti, Daniel Cappel, Maria C. Passarelli, Lauren Fairchild, Carolina Adura, J. Fraser Glickman, Jessica Schulman, Christopher Famulare, Minal Patel, Joseph K. Eibl, Gregory M. Ross, Shibani Bhattacharya, Derek S. Tan, Christina S. Leslie, Thijs Beuming, Dinshaw J. Patel, Yehuda Goldgur, John D. Chodera, and Michael G. Kharas
- Subjects
Science - Abstract
The RNA binding protein MUSASHI-2 (MSI2) is a potential therapeutic target for acute myeloid leukemia. Here the authors identify a small molecule inhibitor of MSI2 and characterize its effects in a murine leukemia model.
- Published
- 2019
- Full Text
- View/download PDF
3. Loss of plasmacytoid dendritic cell differentiation is highly predictive for post-induction measurable residual disease and inferior outcomes in acute myeloid leukemia
- Author
-
Wenbin Xiao, Aaron D. Goldberg, Christopher A. Famulare, Sean M. Devlin, Nghia T. Nguyen, Sinnifer Sim, Charlene C. Kabel, Minal A. Patel, Erin M. McGovern, Akshar Patel, Jessica Schulman, Andrew J. Dunbar, Zachary D. Epstein-Peterson, Kamal N. Menghrajani, Bartlomiej M. Getta, Sheng F. Cai, Mark B. Geyer, Jacob L. Glass, Justin Taylor, Aaron D. Viny, Ross L. Levine, Yanming Zhang, Sergio A. Giralt, Virginia Klimek, Martin S. Tallman, and Mikhail Roshal
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Measurable residual disease is associated with inferior outcomes in patients with acute myeloid leukemia (AML). Measurable residual disease monitoring enhances risk stratification and may guide therapeutic intervention. The European LeukemiaNet working party recently came to a consensus recommendation incorporating leukemia associated immunophenotype-based different from normal approach by multi-color flow cytometry for measurable residual disease evaluation. However, the analytical approach is highly expertise-dependent and difficult to standardize. Here we demonstrate that loss of plasmacytoid dendritic cell differentiation after 7+3 induction in AML is highly specific for measurable residual disease positivity (specificity 97.4%) in a uniformly treated patient cohort. Moreover, loss of plasmacytoid dendritic cell differentiation as determined by a blast-to-plasmacytoid dendritic cell ratio >10 was strongly associated with inferior overall and relapse-free survival (RFS) [Hazard ratio 2.79, 95% confidence interval (95%CI): 0.98-7.97; P=0.077) and 3.83 (95%CI: 1.51-9.74; P=0.007), respectively), which is similar in magnitude to measurable residual disease positivity. Importantly, measurable residual disease positive patients who reconstituted plasmacytoid dendritic cell differentiation (blast/ plasmacytoid dendritic cell ratio
- Published
- 2019
- Full Text
- View/download PDF
4. Molecular predictors of immunophenotypic measurable residual disease clearance in acute myeloid leukemia
- Author
-
Maximilian Stahl, Andriy Derkach, Noushin Farnoud, Jan Philipp Bewersdorf, Troy Robinson, Christopher Famulare, Christina Cho, Sean Devlin, Kamal Menghrajani, Minal A. Patel, Sheng F. Cai, Linde A. Miles, Robert L. Bowman, Mark B. Geyer, Andrew Dunbar, Zachary D. Epstein‐Peterson, Erin McGovern, Jessica Schulman, Jacob L. Glass, Justin Taylor, Aaron D. Viny, Eytan M. Stein, Bartlomiej Getta, Maria E. Arcila, Qi Gao, Juliet Barker, Brian C. Shaffer, Esperanza B. Papadopoulos, Boglarka Gyurkocza, Miguel‐Angel Perales, Omar Abdel‐Wahab, Ross L. Levine, Sergio A. Giralt, Yanming Zhang, Wenbin Xiao, Nidhi Pai, Elli Papaemmanuil, Martin S. Tallman, Mikhail Roshal, and Aaron D. Goldberg
- Subjects
Hematology - Abstract
Measurable residual disease (MRD) is a powerful prognostic factor in acute myeloid leukemia (AML). However, pre-treatment molecular predictors of immunophenotypic MRD clearance remain unclear. We analyzed a dataset of 211 patients with pre-treatment next-generation sequencing who received induction chemotherapy and had MRD assessed by serial immunophenotypic monitoring after induction, subsequent therapy, and allogeneic stem cell transplant (allo-SCT). Induction chemotherapy led to MRD- remission, MRD+ remission, and persistent disease in 35%, 27%, and 38% of patients, respectively. With subsequent therapy, 34% of patients with MRD+ and 26% of patients with persistent disease converted to MRD-. Mutations in CEBPA, NRAS, KRAS, and NPM1 predicted high rates of MRD- remission, while mutations in TP53, SF3B1, ASXL1, and RUNX1 and karyotypic abnormalities including inv (3), monosomy 5 or 7 predicted low rates of MRD- remission. Patients with fewer individual clones were more likely to achieve MRD- remission. Among 132 patients who underwent allo-SCT, outcomes were favorable whether patients achieved early MRD- after induction or later MRD- after subsequent therapy prior to allo-SCT. As MRD conversion with chemotherapy prior to allo-SCT is rarely achieved in patients with specific baseline mutational patterns and high clone numbers, upfront inclusion of these patients into clinical trials should be considered.
- Published
- 2022
- Full Text
- View/download PDF
5. Clinical, genomic, and neurophysiological correlates of lifetime suicide attempts among individuals with alcohol dependence
- Author
-
Peter B. Barr, Zoe Neale, Jessica Schulman, Niamh Mullins, Jian Zhang, David B. Chorlian, Chella Kamarajan, Sivan Kinreich, Ashwini K. Pandey, Gayathri Pandey, Stacey Saenz de Viteri, Laura Acion, Lance Bauer, Kathleen K. Bucholz, Grace Chan, Michael Chao, Danielle M. Dick, Howard J. Edenberg, Tatiana Foroud, Alison Goate, Victor Hesselbrock, Emma C. Johnson, John Kramer, Dongbing Lai, Martin H. Plawecki, Jessica E. Salvatore, Leah Wetherill, Arpana Agrawal, Bernice Porjesz, and Jacquelyn L. Meyers
- Subjects
Article - Abstract
Research has identified clinical, genomic, and neurophysiological markers associated with suicide attempts (SA) among individuals with psychiatric illness. However, there is limited research among those with an alcohol use disorder, despite their disproportionately higher rates of SA. We examined lifetime SA in 4,068 individuals with DSM-IV alcohol dependence from the Collaborative Study on the Genetics of Alcoholism (23% lifetime suicide attempt; 53% female; 17% Admixed African American ancestries; mean age: 38). We 1) explored clinical risk factors associated with SA, 2) conducted a genome-wide association study of SA, 3) examined whether individuals with a SA had elevated polygenic scores for comorbid psychiatric conditions (e.g., alcohol use disorders, lifetime suicide attempt, and depression), and 4) explored differences in electroencephalogram neural functional connectivity between those with and without a SA. One gene-based finding emerged,RFX3(Regulatory Factor X, located on 9p24.2) which had supporting evidence in prior research of SA among individuals with major depression. Only the polygenic score for suicide attempts was associated with reporting a suicide attempt (OR = 1.20, 95% CI = 1.06, 1.37). Lastly, we observed decreased right hemispheric frontal-parietal theta and decreased interhemispheric temporal-parietal alpha electroencephalogram resting-state coherences among those participants who reported a SA relative to those who did not, but differences were small. Overall, individuals with alcohol dependence who report SA appear to experience a variety of severe comorbidities and elevated polygenic risk for SA. Our results demonstrate the need to further investigate suicide attempts in the presence of substance use disorders.
- Published
- 2023
6. Polygenic contributions to suicidal thoughts and behaviors in a sample ascertained for alcohol use disorders
- Author
-
Sarah Colbert, Niamh Mullins, Grace Chan, Jacquelyn L. Meyers, Jessica Schulman, Samuel Kuperman, Dongbing Lai, John Nurnberger, Martin H. Plawecki, Chella Kamarajan, Andrey P. Anokhin, Kathleen K. Bucholz, Victor Hesselbrock, Howard J. Edenberg, John Kramer, Danielle M. Dick, Bernice Porjesz, Arpana Agrawal, and Emma C. Johnson
- Subjects
General Medicine - Abstract
Suicidal thoughts and behaviors have partially distinct genetic etiologies. We used PRS-CS to create polygenic risk scores (PRS) from GWAS of non-suicidal self-injury, broad sense self-harm ideation, non-fatal suicide attempt, death by suicide, and depression. Using mixed-effect models, we estimated whether these PRS were associated with a range of suicidal thoughts and behaviors in the Collaborative Study on the Genetics of Alcoholism (N = 7,526). All PRS were significantly associated with suicidal ideation and suicide attempt (betas=0.08-0.44, FDR
- Published
- 2023
- Full Text
- View/download PDF
7. VenomFlow: An Automated Bioinformatic Pipeline for Identification of Disulfide-Rich Peptides from Venom Arsenals
- Author
-
Eleonora, Achrak, Jennifer, Ferd, Jessica, Schulman, Trami, Dang, Konstantinos, Krampis, and Mande, Holford
- Subjects
Proteomics ,Venoms ,Snails ,Animals ,Computational Biology ,Cysteine ,Disulfides ,Peptides ,Toxins, Biological - Abstract
Animal venoms are among the most complex natural secretions known, comprising a mixture of bioactive compounds often referred to as toxins. Venom arsenals are predominately made up of cysteine-rich peptide toxins that manipulate molecular targets, such as ion channels and receptors, making these venom peptides attractive candidates for the development of therapeutics to benefit human health. With the rise of omic strategies that utilize transcriptomic, proteomic, and bioinformatic methods, we are able to identify more venom proteins and peptides than ever before. However, identification and characterization of bioactive venom peptides remains a significant challenge due to the unique chemical structure and enormous number of peptides found in each venom arsenal (upward of 200 per organism). Here, we introduce a rapid and user-friendly in silico bioinformatic pipeline for the de novo identification and characterization of raw RNAseq reads from venom glands to elucidate cysteine-rich peptides from the arsenal of venomous organisms.Implementation: This project develops a user-friendly automated bioinformatics pipeline via a Galaxy workflow to identify novel venom peptides from raw RNAseq reads of terebrid snails. While designed for venomous terebrid snails, with minor adjustments, this pipeline can be made universal to identify secreted disulfide-rich peptide toxins from any venomous organism.
- Published
- 2022
8. VenomFlow: An Automated Bioinformatic Pipeline for Identification of Disulfide-Rich Peptides from Venom Arsenals
- Author
-
Eleonora Achrak, Jennifer Ferd, Jessica Schulman, Trami Dang, Konstantinos Krampis, and Mande Holford
- Published
- 2022
- Full Text
- View/download PDF
9. Molecular Predictors and Effectiveness of Measurable Residual Disease (MRD) Eradication with Chemotherapy and Allogeneic Stem Cell Transplantation for Acute Myeloid Leukemia
- Author
-
Maria E. Arcila, Omar Abdel-Wahab, Miguel-Angel Perales, Kamal Menghrajani, Maximilian Stahl, Mikhail Roshal, Christina Cho, Elli Papaemmanuil, Juliet N. Barker, Jacob L. Glass, Sergio Giralt, Sean M. Devlin, Justin Taylor, Noushin Farnoud, Andrew Dunbar, Martin S. Tallman, Brian C. Shaffer, Andriy Derkach, Bartlomiej Getta, Esperanza B. Papadopoulos, Christopher Famulare, Eytan M. Stein, Boglarka Gyurkocza, Ross L. Levine, Aaron D. Viny, Sheng F. Cai, Jessica Schulman, Erin McGovern, Yanming Zhang, Minal Patel, Zachary D. Epstein-Peterson, Mark B. Geyer, and Aaron D Goldberg
- Subjects
Oncology ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Disease ,Biochemistry ,Transplantation ,Internal medicine ,Medicine ,Stem cell ,business - Abstract
Background: Measurable residual disease (MRD) is a powerful prognostic factor in AML, including in prediction of outcomes post allogeneic stem cell transplant (alloSCT). However, genomic predictors of successful MRD eradication with chemotherapy prior to alloSCT are unclear. Objectives: Here we provide an integrated analysis of 233 patients (pts) who underwent induction chemotherapy with baseline next-generation sequencing (NGS) followed by serial immunophenotypic monitoring for MRD while patients received additional therapy and alloSCT. Methods: All pts who received anthracycline + cytarabine, +/- investigational agents at Memorial Sloan Kettering Cancer Center starting in April 2014 were retrospectively studied (A). 142 out of 233 pts subsequently underwent alloSCT after induction or additional therapy (A). Immunophenotypic MRD was identified in bone marrow aspirates (BMA) by multiparameter flow cytometry. Any level of residual disease was considered MRD+. Molecular analysis was obtained from pre-induction BMA by NGS using 28 or 49 or 400 gene panels. Results: Patient and treatment characteristics for all pts are detailed in panel (B). Induction chemotherapy resulted in an MRD-CR/CRi and MRD+CR/CRi in 29% and 23% of all pts, respectively (C). Additional therapy included consolidation (n=51), intensive re-induction/salvage (n=47) and non-intensive therapy (n=9). Of 83 AML pts with persistent AML and 58 pts with MRD+CR/CRi after induction (R1), 38/141 (27%) were able to be converted to MRD-CR/CRi. While 33/38 of pts went on to alloSCT after conversion to MRD-CR/CRi, 22 and 36 pts went to alloSCT with persistent AML and MRD+CR/CRi AML, respectively. We focused on pre-induction molecular predictors for achieving an MRD-CR/CRi response prior to transplant for the 142 pts who underwent alloSCT (D). Pts with a NPM1 (79%, Odds ratio [OR] 3.7, p=0.01), IDH1 (92%, OR 3.9, p=0.01) and KRAS (100%, OR 5.0, p=0.03) mutations achieved high rates of MRD-CR/CRi prior to alloSCT. In contrast, RUNX1 (28%, OR 0.2, p=0.01), TP53 (12%, OR 0.1, p=0.02) and SF3B1 (14%, OR 0.1, p=0.04) mutations predicted decreased odds of achieving MRD-CR/CRi prior to alloSCT despite induction and post-induction therapy. AlloSCT resulted in high rates of conversion from MRD+ and persistent disease to MRD negativity. Most pts who entered transplant with CR/CRi MRD+ (28/36, 76%) or persistent AML (14/22, 64%) cleared MRD by the first post-transplant BMA at a median of 32 days (E). Post-alloSCT follow-up indicated value in converting MRD+ to MRD- prior to alloSCT. There was no significant difference in post-transplant cumulative incidence of relapse (F) and OS (G) between early MRD-CR/CRi immediately following induction versus later conversion to MRD-CR/CRi with additional therapy prior to alloSCT. Despite initial post-transplant MRD clearance, pts who entered alloSCT with persistent AML or MRD+ had higher incidence of relapse (p=0.00037, F) and poorer post-transplant OS (p=0.013, G) compared to pts who entered alloSCT with MRD-. Pts with persistent disease prior to alloSCT had shorter duration of MRD- induced by alloSCT compared to pts with MRD-CR/CRi after induction or converted MRD-CR/CRi prior to alloSCT (p=0.0042, H). Importantly, duration of MRD negativity after alloSCT for patients who achieved MRD- prior to alloSCT was not affected by whether patients received induction +/- consolidation (I: treatment type 1-3 from B) vs. induction and salvage treatment for refractory AML (I: treatment type 4-6 from B). Conclusion: We show that transplanted AML pts with specific molecular mutations (RUNX1, SF3B1, and TP53) are unlikely to achieve MRD-CR/CRi after induction, consolidation or salvage therapy, while other mutations (NPM1, IDH1, KRAS) predict high rates of MRD- prior to alloSCT. Additional post-induction therapy may be advantageous for some MRD+ pts to achieve MRD- prior to alloSCT. Post-transplant OS is improved in pts who are MRD- at time of transplant, regardless of whether they required additional therapy beyond induction to achieve this state. AlloSCT is highly effective at eradicating MRD, but post-transplant MRD- is more durable in pts who are MRD- pre-alloSCT. Our results suggest that development of MRD-eradicating therapies has the potential to improve post-transplant outcomes and argues for innovative trials for pts with adverse molecular features currently unlikely to achieve MRD- pre alloSCT. Figure Disclosures Cai: Imago Biosciences, Inc.: Consultancy, Current equity holder in private company; DAVA Oncology: Honoraria. Geyer:Amgen: Research Funding. Glass:Gerson Lehman Group: Consultancy. Stein:Syros: Membership on an entity's Board of Directors or advisory committees; PTC Therapeutics: Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Biotheryx: Consultancy; Bayer: Research Funding; Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees; Syndax: Consultancy, Research Funding; Seattle Genetics: Consultancy; Abbvie: Consultancy; Amgen: Consultancy; Celgene Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Agios Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astellas Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Daiichi-Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding. Levine:Gilead: Honoraria; Isoplexis: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Qiagen: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Honoraria, Research Funding; Janssen: Consultancy; Lilly: Consultancy, Honoraria; Imago: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; C4 Therapeutics: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Astellas: Consultancy; Novartis: Consultancy; Prelude Therapeutics: Research Funding; Loxo: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria; Morphosys: Consultancy; Roche: Consultancy, Honoraria, Research Funding. Gyurkocza:Actinium: Research Funding. Perales:Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Nektar Therapeutics: Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; MolMed: Membership on an entity's Board of Directors or advisory committees; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees; Medigene: Membership on an entity's Board of Directors or advisory committees, Other; Servier: Membership on an entity's Board of Directors or advisory committees, Other; Omeros: Honoraria, Membership on an entity's Board of Directors or advisory committees; Merck: Consultancy, Honoraria; NexImmune: Membership on an entity's Board of Directors or advisory committees; Cidara Therapeutics: Other; Miltenyi Biotec: Research Funding; Kite/Gilead: Honoraria, Research Funding; Incyte Corporation: Honoraria, Research Funding; Bristol Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria; Bellicum: Honoraria, Membership on an entity's Board of Directors or advisory committees. Abdel-Wahab:H3 Biomedicine Inc.: Consultancy, Research Funding; Janssen: Consultancy; Envisagenics Inc.: Current equity holder in private company; Merck: Consultancy. Papaemmanuil:Kyowa Hakko Kirin: Consultancy, Honoraria; Isabl: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; MSKCC: Patents & Royalties; Novartis: Consultancy, Honoraria; Illumina: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Research Funding; Prime Oncology: Consultancy, Honoraria. Giralt:KITE: Consultancy; NOVARTIS: Consultancy, Honoraria, Research Funding; OMEROS: Consultancy, Honoraria; AMGEN: Consultancy, Research Funding; TAKEDA: Research Funding; ACTINUUM: Consultancy, Research Funding; MILTENYI: Consultancy, Research Funding; CELGENE: Consultancy, Honoraria, Research Funding; JAZZ: Consultancy, Honoraria. Tallman:Glycomimetics: Research Funding; Rafael: Research Funding; Amgen: Research Funding; Bioline rx: Membership on an entity's Board of Directors or advisory committees; Daiichi-Sankyo: Membership on an entity's Board of Directors or advisory committees; KAHR: Membership on an entity's Board of Directors or advisory committees; UpToDate: Patents & Royalties; Rigel: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Roche: Membership on an entity's Board of Directors or advisory committees; Jazz Pharma: Membership on an entity's Board of Directors or advisory committees; Oncolyze: Membership on an entity's Board of Directors or advisory committees; Delta Fly Pharma: Membership on an entity's Board of Directors or advisory committees; BioSight: Membership on an entity's Board of Directors or advisory committees, Research Funding; ADC Therapeutics: Research Funding; Orsenix: Research Funding; Cellerant: Research Funding; Abbvie: Research Funding. Goldberg:AROG: Research Funding; Aprea: Research Funding; ADC Therapeutics: Research Funding; Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees; Daiichi Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Consultancy; Aptose: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Dava Oncology: Honoraria; Pfizer: Research Funding; Celularity: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
10. Small-molecule targeting of MUSASHI RNA-binding activity in acute myeloid leukemia
- Author
-
Dinshaw J. Patel, Christina Z. Rotsides, Shibani Bhattacharya, Derek S. Tan, Sun Mi Park, Steven K. Albanese, Fraser Glickman, Alexandra Schurer, Minal Patel, Yaniv Kazansky, Daniel Worroll, Gerard Minuesa, Michael G. Kharas, Yehuda Goldgur, John D. Chodera, Arthur Chow, Gregory M. Ross, Lauren Fairchild, Thijs Beuming, Carolina Adura, James Taggart, Saroj Gourkanti, Wei Xie, Jessica Schulman, Maria C. Passarelli, Levi N. Naden, Andrea Rizzi, Timothy Chou, Joseph K. Eibl, Christopher Famulare, Daniel Cappel, and Christina S. Leslie
- Subjects
0301 basic medicine ,Male ,Myeloid ,Molecular biology ,General Physics and Astronomy ,RNA-binding protein ,Apoptosis ,02 engineering and technology ,Small hairpin RNA ,Mice ,hemic and lymphatic diseases ,Gene expression ,Tumor Cells, Cultured ,RNA, Small Interfering ,lcsh:Science ,Regulation of gene expression ,Multidisciplinary ,Chemistry ,Gene Expression Regulation, Leukemic ,Pteridines ,Small molecules ,Myeloid leukemia ,RNA-Binding Proteins ,021001 nanoscience & nanotechnology ,3. Good health ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,0210 nano-technology ,RNA Recognition Motif ,Science ,Primary Cell Culture ,Drug development ,General Biochemistry, Genetics and Molecular Biology ,Article ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Flavins ,medicine ,Animals ,Humans ,Leukemia, Experimental ,Gene Expression Profiling ,RNA ,General Chemistry ,medicine.disease ,030104 developmental biology ,Cancer research ,lcsh:Q ,Transcriptome - Abstract
The MUSASHI (MSI) family of RNA binding proteins (MSI1 and MSI2) contribute to a wide spectrum of cancers including acute myeloid leukemia. We find that the small molecule Ro 08–2750 (Ro) binds directly and selectively to MSI2 and competes for its RNA binding in biochemical assays. Ro treatment in mouse and human myeloid leukemia cells results in an increase in differentiation and apoptosis, inhibition of known MSI-targets, and a shared global gene expression signature similar to shRNA depletion of MSI2. Ro demonstrates in vivo inhibition of c-MYC and reduces disease burden in a murine AML leukemia model. Thus, we identify a small molecule that targets MSI’s oncogenic activity. Our study provides a framework for targeting RNA binding proteins in cancer., The RNA binding protein MUSASHI-2 (MSI2) is a potential therapeutic target for acute myeloid leukemia. Here the authors identify a small molecule inhibitor of MSI2 and characterize its effects in a murine leukemia model.
- Published
- 2019
- Full Text
- View/download PDF
11. Multi-omics integration analysis identifies novel genes for alcoholism with potential overlap with neurodegenerative diseases
- Author
-
Manav Kapoor, Michael J. Chao, Emma C. Johnson, Gloriia Novikova, Dongbing Lai, Jacquelyn L. Meyers, Jessica Schulman, John I. Nurnberger, Bernice Porjesz, Yunlong Liu, The Collaborative Study on the Genetics of Alcoholism (COGA), Tatiana Foroud, Howard J. Edenberg, Edoardo Marcora, Arpana Agrawal, and Alison Goate
- Subjects
Genetic Markers ,Science ,Quantitative Trait Loci ,General Physics and Astronomy ,Genome-wide association study ,Computational biology ,Alcohol use disorder ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Linkage Disequilibrium ,Article ,Epigenesis, Genetic ,Novel gene ,Fetus ,medicine ,SNP ,Humans ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Promoter Regions, Genetic ,Gene ,Behavioural genetics ,Multidisciplinary ,Brain ,Neurodegenerative Diseases ,General Chemistry ,Genomics ,Mendelian Randomization Analysis ,medicine.disease ,Physical Chromosome Mapping ,Gene expression profiling ,Alcoholism ,Genetic Loci ,Multi omics ,Data integration ,Gene expression ,Genome-Wide Association Study - Abstract
Identification of causal variants and genes underlying genome-wide association study (GWAS) loci is essential to understand the biology of alcohol use disorder (AUD) and drinks per week (DPW). Multi-omics integration approaches have shown potential for fine mapping complex loci to obtain biological insights to disease mechanisms. In this study, we use multi-omics approaches, to fine-map AUD and DPW associations at single SNP resolution to demonstrate that rs56030824 on chromosome 11 significantly reduces SPI1 mRNA expression in myeloid cells and lowers risk for AUD and DPW. Our analysis also identifies MAPT as a candidate causal gene specifically associated with DPW. Genes prioritized in this study show overlap with causal genes associated with neurodegenerative disorders. Multi-omics integration analyses highlight, genetic similarities and differences between alcohol intake and disordered drinking, suggesting molecular heterogeneity that might inform future targeted functional and cross-species studies., Alcohol use disorder and drinks per week both have been studied genetically and have different correlations with psychiatric diseases. Here the authors integrate multi-omics data to identify unique and shared variants, genes and pathways for alcohol use disorder and drinks per week.
- Published
- 2020
12. Multi-omics integration analysis identifies novel genes for alcoholism with potential link to neurodegenerative diseases
- Author
-
Gloriia Novikova, Edoardo Marcora, Jacquelyn L. Meyers, Manav Kapoor, Bernice Porjesz, John I. Nurnberger, Emma C. Johnson, Howard J. Edenberg, Alison Goate, Yunlong Liu, Dongbing Lai, Michael Chao, Jessica Schulman, and Arpana Agrawal
- Subjects
Mendelian randomization ,medicine ,Single-nucleotide polymorphism ,Genome-wide association study ,Locus (genetics) ,Alcohol use disorder ,Computational biology ,Epigenetics ,Quantitative trait locus ,Biology ,medicine.disease ,Gene - Abstract
SignificanceIdentification of causal variants and genes underlying genome-wide association study (GWAS) loci is essential to understanding the biology of alcohol use disorder (AUD).MethodsIntegration of “multi-omics” data is often necessary to nominate candidate causal variants and genes and prioritize them for follow up studies. Here, we used Mendelian randomization to integrate AUD and drinks per week (DPW) GWAS summary statistics with the gene expression and methylation quantitative trait loci (eQTLs and mQTLs) in the largest brain and myeloid datasets. We also used AUD-related single cell epigenetic data to nominate candidate causal variants and genes associated with DPW and AUD.ResultsOur multi-omics integration analyses prioritized unique as well as shared genes and pathways among AUD and DPW. The GWAS variants associated with both AUD and DPW showed significant enrichment in the promoter regions of fetal and adult brains. The integration of GWAS SNPs with mQTLs from fetal brain prioritized variants on chromosome 11 in both AUD and DPW GWASs. The co-localized variants were found to be overlapping with promoter marks for SPI1, specifically in human microglia, the myeloid cells of the brain. The co-localized SNPs were also strongly associated with SPI1 mRNA expression in myeloid cells from peripheral blood. The prioritized variant at this locus is predicted to alter the binding site for a transcription factor, RXRA, a key player in the regulation of myeloid cell function. Our analysis also identified MAPT as a candidate causal gene specifically associated with DPW. mRNA expression of MAPT was also correlated with daily amounts of alcohol intake in post-mortem brains (frontal cortex) from alcoholics and controls (N = 92). Results may be queried and visualized in an online public resource of these integrative analysis (https://lcad.shinyapps.io/alc_multiomics/). These results highlight overlap between causal genes for neurodegenerative diseases, alcohol use disorder and alcohol consumption.In conclusionintegrating GWAS summary statistics with multi-omics datasets from multiple sources identified biological similarities and differences between typical alcohol intake and disordered drinking highlighting molecular heterogeneity that might inform future targeted functional and cross-species studies. Interestingly, overlap was also observed with causal genes for neurodegenerative diseases.
- Published
- 2020
- Full Text
- View/download PDF
13. Cancer therapy shapes the fitness landscape of clonal hematopoiesis
- Author
-
David M. Hyman, Stuart Gardos, Gunes Gundem, Luis A. Diaz, James A. Fagin, Kelly L. Bolton, Catherine C. Coombs, Michael F. Berger, Nilanjan Chatterjee, Antonin Berthon, Nicole M. Caltabellotta, Konrad H. Stopsack, John Philip, Mariko Yabe, Barbara Spitzer, Virginia M. Klimek, Eder Paraiso, Max Levine, Ryan Ptashkin, Ahmet Zehir, Akshar Patel, Markus Ball, Zsofia K. Stadler, Juan E. Arango Ossa, Ross L. Levine, Lindsay M. Morton, Larry Norton, Lior Z. Braunstein, Minal Patel, Sean M. Devlin, Daniel Kelly, Noushin Farnoud, Mark E. Robson, Elsa Bernard, Laura Boucai, Maria E. Arcila, Kenneth Offit, Jessica Schulman, Montserrat Garcia-Closas, David B. Solit, Teng Gao, José Baselga, Ryma Benayed, Howard I. Scher, Simon Mantha, Martin S. Tallman, Elli Papaemmanuil, Andrew L. Young, Sonya Li, Dominik Glodzik, Nancy K. Gillis, Choonsik Lee, Juan S. Medina Martinez, Eric Padron, Benjamin L. Ebert, Marc Ladanyi, Michael Walsh, Aijazuddin Syed, Dean F. Bajorin, Paul D.P. Pharoah, Todd E. Druley, Koichi Takahashi, Christopher J. Gibson, Diana Mandelker, Philip, John [0000-0002-0535-7178], Bernard, Elsa [0000-0002-2057-7187], Levine, Max [0000-0001-5156-9086], Robson, Mark E [0000-0002-3109-1692], Pharoah, Paul DP [0000-0001-8494-732X], Stopsack, Konrad H [0000-0002-0722-1311], Fagin, James [0000-0002-2365-2517], Boucai, Laura [0000-0002-8852-1120], Ebert, Benjamin L [0000-0003-0197-5451], Takahashi, Koichi [0000-0002-8027-9659], Solit, David B [0000-0002-6614-802X], Garcia-Closas, Montserrat [0000-0003-1033-2650], Diaz, Luis A [0000-0002-7079-8914], Levine, Ross L [0000-0002-7884-1905], Zehir, Ahmet [0000-0001-5406-4104], Papaemmanuil, Elli [0000-0003-1709-8983], and Apollo - University of Cambridge Repository
- Subjects
Adult ,Male ,Myeloid ,Adolescent ,DNA damage ,Context (language use) ,Antineoplastic Agents ,Biology ,medicine.disease_cause ,Somatic evolution in cancer ,Models, Biological ,Clonal Evolution ,Cohort Studies ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Neoplasms ,Genetics ,medicine ,Humans ,Selection, Genetic ,Child ,CHEK2 ,Gene ,030304 developmental biology ,Aged ,Aged, 80 and over ,0303 health sciences ,Mutation ,Infant, Newborn ,Cancer ,Infant ,Neoplasms, Second Primary ,Middle Aged ,medicine.disease ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,Leukemia, Myeloid ,Child, Preschool ,Cancer research ,Female ,Genetic Fitness ,Clonal Hematopoiesis ,030217 neurology & neurosurgery - Abstract
Acquired mutations are pervasive across normal tissues. However, understanding of the processes that drive transformation of certain clones to cancer is limited. Here we study this phenomenon in the context of clonal hematopoiesis (CH) and the development of therapy-related myeloid neoplasms (tMNs). We find that mutations are selected differentially based on exposures. Mutations in ASXL1 are enriched in current or former smokers, whereas cancer therapy with radiation, platinum and topoisomerase II inhibitors preferentially selects for mutations in DNA damage response genes (TP53, PPM1D, CHEK2). Sequential sampling provides definitive evidence that DNA damage response clones outcompete other clones when exposed to certain therapies. Among cases in which CH was previously detected, the CH mutation was present at tMN diagnosis. We identify the molecular characteristics of CH that increase risk of tMN. The increasing implementation of clinical sequencing at diagnosis provides an opportunity to identify patients at risk of tMN for prevention strategies. Environmental exposures shape patterns of selection for mutations in clonal hematopoiesis. Cancer therapies promote the growth of clones with mutations that are strongly enriched in treatment-related myeloid neoplasms.
- Published
- 2020
14. The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells
- Author
-
Sara Zaccara, Diu T.T. Nguyen, Minal Patel, Jessica Schulman, Brian F. Pickering, Timothy Chou, Samie R. Jaffrey, Virginia M. Klimek, Gerard Minuesa, Michael G. Kharas, Ly P. Vu, Christopher Famulare, Yogesh Saletore, Christopher E. Mason, Yuanming Cheng, Ari Melnick, Francine E. Garrett-Bakelman, Martin Carroll, Matthew MacKay, and Arthur Chow
- Subjects
0301 basic medicine ,Myeloid ,Cell growth ,Chemistry ,Cellular differentiation ,MRNA modification ,Myeloid leukemia ,General Medicine ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,3. Good health ,03 medical and health sciences ,Leukemia ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,hemic and lymphatic diseases ,medicine ,Cancer research ,Progenitor cell - Abstract
N6-methyladenosine (m6A) is an abundant nucleotide modification in mRNA that is required for the differentiation of mouse embryonic stem cells. However, it remains unknown whether the m6A modification controls the differentiation of normal and/or malignant myeloid hematopoietic cells. Here we show that shRNA-mediated depletion of the m6A-forming enzyme METTL3 in human hematopoietic stem/progenitor cells (HSPCs) promotes cell differentiation, coupled with reduced cell proliferation. Conversely, overexpression of wild-type METTL3, but not of a catalytically inactive form of METTL3, inhibits cell differentiation and increases cell growth. METTL3 mRNA and protein are expressed more abundantly in acute myeloid leukemia (AML) cells than in healthy HSPCs or other types of tumor cells. Furthermore, METTL3 depletion in human myeloid leukemia cell lines induces cell differentiation and apoptosis and delays leukemia progression in recipient mice in vivo. Single-nucleotide-resolution mapping of m6A coupled with ribosome profiling reveals that m6A promotes the translation of c-MYC, BCL2 and PTEN mRNAs in the human acute myeloid leukemia MOLM-13 cell line. Moreover, loss of METTL3 leads to increased levels of phosphorylated AKT, which contributes to the differentiation-promoting effects of METTL3 depletion. Overall, these results provide a rationale for the therapeutic targeting of METTL3 in myeloid leukemia.
- Published
- 2017
- Full Text
- View/download PDF
15. Oncologic therapy shapes the fitness landscape of clonal hematopoiesis
- Author
-
Elli Papaemmanuil, Nicole M. Caltabellotta, Choonsik Lee, Montserrat Garcia-Closas, David B. Solit, José Baselga, Mariko Yabe, Sean M. Devlin, Larry Norton, Max Levine, Simon Mantha, Martin S. Tallman, Juan E. Arango, Christopher J. Gibson, Andrew L. Young, Elsa Bernard, Jessica Schulman, Minal Patel, Zsofia K. Stadler, Michael F. Berger, Benjamin L. Ebert, Dominik Glodzik, James A. Fagin, Sonya Li, Maria E. Arcila, Eric Padron, Noushin Farnoud, John Philip, Antonin Berthon, M.E. Robson, Nancy K. Gillis, Lior Z. Braunstein, Lindsay M. Morton, Teng Gao, Akshar Patel, Ryan Ptashkin, Ahmet Zehir, Nilanjan Chatterjee, Aijazuddin Syed, Ross L. Levine, Kenneth Offit, Konrad H. Stopsack, Koichi Takahasi, Paul D.P. Pharoah, Barbara Spitzer, Stuart Gardos, Eder Paraiso, Virginia M. Klimek, Ryma Benayed, Markus Ball, Howard I. Scher, Kelly L. Bolton, Dean F. Bajorin, David M. Hyman, Daniel Kelly, Gunes Gundem, Luis A. Diaz, Laura Boucai, Todd E. Druley, Diana Mandelker, Catherine C. Coombs, Juan Medina, Marc Ladanyi, and Michael Walsh
- Subjects
Myeloid ,business.industry ,medicine.medical_treatment ,Clone (cell biology) ,Cancer ,medicine.disease ,Myeloid Neoplasm ,Radiation therapy ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,Cancer research ,Cytotoxic T cell ,business ,CHEK2 - Abstract
Clonal hematopoiesis (CH) is frequent in cancer patients and associated with increased risk of therapy related myeloid neoplasms (tMN). To define the relationship between CH, oncologic therapy, and tMN progression, we studied 24,439 cancer patients. We show that previously treated patients have increased rates of CH, with enrichment of mutations in DNA Damage Response (DDR) genes (TP53, PPM1D, CHEK2). Exposure to radiation, platinum and topoisomerase II inhibitors have the strongest association with CH with evidence of dose-dependence and gene-treatment interactions. We validate these associations in serial sampling from 525 patients and show that exposure to cytotoxic and radiation therapy imparts a selective advantage specifically in hematopoietic cells with DDR mutations. In patients who progressed to tMN, the clone at CH demarcated the dominant clone at tMN diagnosis. CH mutational features predict risk of therapy-related myeloid neoplasm in solid tumor patients with clinical implications for early detection and treatment decisions.
- Published
- 2019
- Full Text
- View/download PDF
16. Prognostic impact of RAS-pathway mutations in patients with myelofibrosis
- Author
-
Nelson Hamerschlak, Paulo Vidal Campregher, Jessica Schulman, Srdan Verstovsek, Hagop M. Kantarjian, Lucia Masarova, Maria E. Arcila, Ross L. Levine, Tarcila Santos Datoguia, Bartlomiej Getta, Raajit K. Rampal, Christopher Famulare, Renato David Puga, Raquel de Melo Alves Paiva, and Fabio P.S. Santos
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Ruxolitinib ,Myeloid ,DNA Mutational Analysis ,Protein Tyrosine Phosphatase, Non-Receptor Type 11 ,medicine.disease_cause ,0302 clinical medicine ,Leukocytosis ,Polycythemia Vera ,Aged, 80 and over ,Myeloid leukemia ,High-Throughput Nucleotide Sequencing ,Hematology ,Middle Aged ,Prognosis ,medicine.anatomical_structure ,Treatment Outcome ,030220 oncology & carcinogenesis ,Female ,KRAS ,medicine.symptom ,medicine.drug ,Thrombocythemia, Essential ,Adult ,Risk ,medicine.medical_specialty ,Adolescent ,Article ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Young Adult ,Germline mutation ,Internal medicine ,Nitriles ,medicine ,Humans ,Myelofibrosis ,Aged ,Proportional Hazards Models ,Retrospective Studies ,Proportional hazards model ,business.industry ,Genetic Variation ,medicine.disease ,030104 developmental biology ,Genes, ras ,Pyrimidines ,fms-Like Tyrosine Kinase 3 ,Primary Myelofibrosis ,Mutation ,Pyrazoles ,business - Abstract
RAS-pathway mutations are recurrent events in myeloid malignancies. However, there is limited data on the significance of RAS-pathway mutations in patients with myelofibrosis (MF). We analyzed next-generation sequencing data of 16 genes, including RAS-pathway genes, from 723 patients with primary and secondary MF across three international centers and evaluated their significance. N/KRAS variants were present in 6% of patients and were typically sub-clonal (median VAF = 20%) relative to other genes variants. RAS variants were associated with advanced MF features including leukocytosis (p = 0.02), high somatic mutation burden (p
- Published
- 2019
17. Loss of plasmacytoid dendritic cell differentiation is highly predictive for post-induction measurable residual disease and inferior outcomes in acute myeloid leukemia
- Author
-
Jessica Schulman, Akshar Patel, Ross L. Levine, Sergio Giralt, Sheng F. Cai, Erin McGovern, Justin Taylor, Virginia M. Klimek, Martin S. Tallman, Sinnifer Sim, Sean M. Devlin, Andrew Dunbar, Charlene C. Kabel, Mark B. Geyer, Nghia T. Nguyen, Aaron D. Viny, Yanming Zhang, Christopher Famulare, Wenbin Xiao, Kamal Menghrajani, Zachary D. Epstein-Peterson, Minal Patel, Aaron D Goldberg, Mikhail Roshal, Jacob L. Glass, and Bartlomiej Getta
- Subjects
Oncology ,Adult ,Male ,Acute Myeloid Leukemia ,medicine.medical_specialty ,Myeloid ,Neoplasm, Residual ,Plasmacytoid dendritic cell ,Article ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,Immunophenotyping ,Internal medicine ,Medicine ,Humans ,Plasmacytoid dendritic cell differentiation ,Aged ,Retrospective Studies ,medicine.diagnostic_test ,business.industry ,Myeloid leukemia ,Hematology ,Dendritic cell ,Dendritic Cells ,Middle Aged ,medicine.disease ,Prognosis ,Combined Modality Therapy ,3. Good health ,Survival Rate ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Case-Control Studies ,Female ,Neoplasm Recurrence, Local ,business ,030215 immunology ,Follow-Up Studies - Abstract
Measurable residual disease is associated with inferior outcomes in patients with acute myeloid leukemia (AML). Measurable residual disease monitoring enhances risk stratification and may guide therapeutic intervention. The European LeukemiaNet working party recently came to a consensus recommendation incorporating leukemia associated immunophenotype-based different from normal approach by multi-color flow cytometry for measurable residual disease evaluation. However, the analytical approach is highly expertise-dependent and difficult to standardize. Here we demonstrate that loss of plasmacytoid dendritic cell differentiation after 7+3 induction in AML is highly specific for measurable residual disease positivity (specificity 97.4%) in a uniformly treated patient cohort. Moreover, loss of plasmacytoid dendritic cell differentiation as determined by a blast-to-plasmacytoid dendritic cell ratio >10 was strongly associated with inferior overall and relapse-free survival (RFS) [Hazard ratio 2.79, 95% confidence interval (95%CI): 0.98-7.97; P=0.077) and 3.83 (95%CI: 1.51-9.74; P=0.007), respectively), which is similar in magnitude to measurable residual disease positivity. Importantly, measurable residual disease positive patients who reconstituted plasmacytoid dendritic cell differentiation (blast/ plasmacytoid dendritic cell ratio
- Published
- 2018
18. Small-molecule targeting of MUSASHI RNA-binding activity in acute myeloid leukemia
- Author
-
Timothy Chou, Joseph K. Eibl, Andrea Rizzi, Steven K. Albanese, J.F. Glickman, James Taggart, John D. Chodera, Saroj Gourkanti, Christina Z. Rotsides, Minal Patel, Gerard Minuesa Dinares, Lauren Fairchild, Thijs Beuming, Alexandra Schurer, Gregory M. Ross, Derek S. Tan, Michael G. Kharas, Arthur Chow, Daniel Cappel, Christina S. Leslie, Christopher Famulare, Sun Mi Park, Carolina Adura, Yehuda Goldgur, Levi N. Naden, Jessica Schulman, and Maria C. Passarelli
- Subjects
Immunology ,RNA-binding protein ,medicine.disease_cause ,Biochemistry ,Small hairpin RNA ,03 medical and health sciences ,0302 clinical medicine ,Acute lymphocytic leukemia ,hemic and lymphatic diseases ,Gene expression ,medicine ,Electrophoretic mobility shift assay ,030304 developmental biology ,0303 health sciences ,Chemistry ,Myeloid leukemia ,RNA ,Cell Biology ,Hematology ,medicine.disease ,Small molecule ,Leukemia ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer research ,Carcinogenesis ,030215 immunology - Abstract
RNA-binding proteins (RBPs) play critical roles in cell homeostasis by controlling gene expression post-transcriptionally, contributing to mRNA processing events (splicing, polyadenylation, localization, stability, export and translation). The involvement of RBPs to tumorigenesis, through genetic perturbation or epigenetic dysregulation, has been found in a variety of human cancers. The RBP MUSASHI-2 (MSI2) contributes to the pathogenesis of a spectrum of solid tumors and hematologic malignancies and predicts a worse clinical outcome in patients with myeloid and acute lymphoblastic leukemia (MDS, AML and ALL). Thus, MSI2 has been proposed as a putative biomarker for diagnosis as well as a potential therapeutic target for AML. However, there are currently no specific inhibitors for MSI. Previous work from our lab reported a Fluorescent Polarization (FP) screen with 6,208 compounds identifying small-molecules with MSI RNA-binding inhibition activity. Here, we characterize Ro 08-2750 (Ro), best FP screen hit, as a MSI RNA-competitive inhibitor. Electrophoresis Mobility Shift Assays (EMSA) demonstrated Ro inhibition of MSI2-RNA complexes formation. MicroScale Thermophoresis (MST) interaction studies showed that the compound interacts with MSI2 full-length and RNA-Recognition Motif 1 (RRM1) with μM affinity and with nearly 20-fold lower KD to an RBP control (SYNCRIP). We obtained the crystal structure of MSI2 RRM1 at 1.7Å and docking and mutagenesis validation confirmed K22, F66, F97 and R100 as crucial binding residues in the RNA-binding pocket. To further prove structure activity relationship, we used two chemical analogs: Ro-OH, an alcohol derivative of the Ro's aldehyde, showed 10-fold reduced activity and Ro-NGF, containing the Ro isoalloxazine scaffold, showed no binding or activity in vitro. Of note, in proliferation assays Ro EC50 was 2.6±0.1 μM in MLL-AF9 bone marrow cells and an average of 8.4±1.1 μM in MOLM13 and K562 human AML cells, whereas RoOH and RoNGF showed 10-fold or >50 μM EC50, respectively. Ro significantly reduced binding of MSI2 to its mRNA targets (such as cMYC, CDKN1A or SMAD3) in an RNA-IP and a direct effect in their protein translation in human leukemia cells. RNA-sequencing of 4h Ro treated MOLM13 and K562 AML cells resulted in gene expression changes that enriched for the gene expression profiling after shRNA mediated depletion of MSI2 in CML-BC and AML cell lines. Ro demonstrated a significant therapeutic index abolishing MLL-AF9+ BM colony formation at concentrations that did not affect the plating efficiency of normal Lin-Sca+cKit+ (LSK) cells. Similarly, Ro demonstrated differential sensitivity in three AML patient samples colony formation compared to normal human CD34+ cord blood cells. Finally, we sought to determine Ro in vivo activity by using an aggressive murine MLL-AF9 murine leukemia model. Acute treatment (4h and 12hr) with 13.75 mg/kg Ro in DMSO reduced c-KIT protein abundance and intracellular c-MYC. Administration of the same Ro dose every 3 days was well tolerated and showed a significant reduction in spleen weights, white blood cell counts and c-MYC levels compared to the controls. These data provide the feasibility that targeting MSI in vivo could have therapeutic efficacy in AML. This study identifies and characterizes Ro 08-2750 as a compound selectively inhibiting the oncogenic RNA-binding activity of MSI in myeloid leukemia. Ro targeting an RRM motif to block RNA activity represents a valuable proof of concept for the general inhibition of these class of RNA regulators. Overall, we provide a framework to identify and test novel RBP inhibitors thus validating this class of proteins as chemically "druggable" novel therapeutic targets in cancer. Disclosures Chodera: Schrödinger: Consultancy, Membership on an entity's Board of Directors or advisory committees.
- Published
- 2018
- Full Text
- View/download PDF
19. The N
- Author
-
Ly P, Vu, Brian F, Pickering, Yuanming, Cheng, Sara, Zaccara, Diu, Nguyen, Gerard, Minuesa, Timothy, Chou, Arthur, Chow, Yogesh, Saletore, Matthew, MacKay, Jessica, Schulman, Christopher, Famulare, Minal, Patel, Virginia M, Klimek, Francine E, Garrett-Bakelman, Ari, Melnick, Martin, Carroll, Christopher E, Mason, Samie R, Jaffrey, and Michael G, Kharas
- Subjects
Leukemia, Myeloid, Acute ,Adenosine ,Tumor Cells, Cultured ,Humans ,Bone Marrow Cells ,Cell Differentiation ,Clustered Regularly Interspaced Short Palindromic Repeats ,Methyltransferases ,Cells, Cultured - Abstract
N
- Published
- 2017
20. Abstract LB-304: Oncologic therapy for solid tumors alters the risk of clonal hematopoiesis
- Author
-
Maria E. Arcila, Marc Ladanyi, Teng Gao, Kenneth Offit, Stuart Gardos, Michael Walsh, Michael F. Berger, Antonin Berthon, Lindsay M. Morton, Matahi Moarii, Laura Boucai, Nilanjan Chatterjee, Ryan Ptashkin, Mariko Yabe, Diana Mandelker, Ahmet Zehir, Minal Patel, Elli Papaemmanuil, Ross L. Levine, Jessica Schulman, Martin S. Tallman, David M. Hyman, Sean M. Devlin, Gunes Gundem, Daniel Kelly, Catherine C. Coombs, Aijazuddin Syed, Lior Z. Braunstein, Virginia M. Klimek, Luis A. Diaz, Dean F. Bajorin, Axel Martin, Kelly L. Bolton, John Philip, Dominik Glodzik, Choonsik Lee, Mark E. Robson, Elsa Bernard, Zsofia K. Stadler, Howard I. Scher, Montserrat Garcia-Closas, David B. Solit, and Akshar Patel
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Myeloid ,business.industry ,Clonal hematopoiesis ,Chatterjee ,Sequencing data ,Cancer ,medicine.disease ,Leukemia ,medicine.anatomical_structure ,Internal medicine ,Cohort ,medicine ,Mutation detection ,business - Abstract
Solid tumor patients often suffer from cytopenias and are at risk for therapy-related myeloid neoplasms (tMN). Somatic mutations in leukemia-associated genes can occur in normal healthy individuals, referred to as clonal hematopoiesis (CH). CH is associated with cytopenias, risk of leukemia and cardiovascular disease. We and others have shown that CH is frequent in cancer patients. Characterization of the relationship between exposure to specific oncologic regimens and CH and how these relate to cytopenias and tMN risk would inform treatment decisions and tMN prevention strategies. To determine the relationship between CH and oncologic therapy we interrogated CH in a cohort of 9045 solid tumor patients. Subjects were sequenced using a targeted panel of cancer-associated mutations used to screen tumor samples against a blood control sample. Mutation detection was performed on blood-derived sequencing data using the matched tumor as a comparator and accounted for background sequencing error rates. CH was identified in 23% of patients. In multivariate regression analyses adjusted by age, CH was more often found in current smokers (OR=1.20, 95%CI=1.07-1.35, p CH is frequent in solid tumor patients and can be reliably detected when a matched tumor normal targeted gene sequencing approach is performed. Beyond age, CH is strongly associated with race, smoking and importantly prior exposure to oncologic therapy with evidence of specific treatment effects. Screening of CH in cancer cohorts is critical to the development of future clinical guidelines and risk-adapted prevention strategies for tMN. Note: This abstract was not presented at the meeting. Citation Format: Kelly Bolton, Ryan Ptashkin, Lior Braunstein, Teng Gao, Sean M. Devlin, Daniel Kelly, Catherine Coombs, Minal Patel, Matahi Moarii, Elsa Bernard, Antonin Berthon, Laura Boucai, Dominik Glodzik, Axel Martin, Zsofia Stadler, Michael Walsh, Diana Mandelker, Akshar Patel, Jessica Schulman, Gunes Gundem, Aijazuddin Syed, Maria Arcila, David B. Solit, Mark E. Robson, Marc Ladanyi, Choonsik Lee, John Philip, Dean Bajorin, Montserrat Garcia-Closas, Stuart Gardos, David Hyman, Martin Tallman, Mariko Yabe, Kenneth Offit, Howard Scher, Virginia Klimek, Luis Diaz, Nilanjan Chatterjee, Michael F. Berger, Lindsay Morton, Ross Levine, Ahmet Zehir, Elli Papaemmanuil. Oncologic therapy for solid tumors alters the risk of clonal hematopoiesis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr LB-304.
- Published
- 2019
- Full Text
- View/download PDF
21. A Novel Prognostic Model Including Cytogenetic and Molecular Data in Patients with Primary and Post-Essential Thrombocythemia (ET)/Polycythemia Vera (PV) Myelofibrosis (MF)
- Author
-
Fabio Ps Santos, Nelson Hamerschlak, Paulo Vidal Campregher, Giulliana Ar Goncalves, Srdan Verstovsek, Tarcila Santos Datoguia, Christopher Famulare, Jessica Schulman, Raajit K. Rampal, Ross L. Levine, Hagop M. Kantarjian, Lucia Masarova, and Bartlomiej Getta
- Subjects
Oncology ,Neuroblastoma RAS viral oncogene homolog ,medicine.medical_specialty ,business.industry ,Essential thrombocythemia ,Proportional hazards model ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,medicine.disease_cause ,Biochemistry ,Internal medicine ,medicine ,Sample collection ,KRAS ,business ,Myelofibrosis ,Myeloproliferative neoplasm ,Exome sequencing - Abstract
Introduction: MF is a Philadelphia-negative myeloproliferative neoplasm (Ph-negative MPN) with an heterogeneous outcome. In 2009, Cervantes et al. published the International Prognostic Score System (IPSS) to better determine outcomes in this disease. In the last decade, several recurrently mutated genes have been described in MF, some of them associated with prognostic impact in survival. We propose a novel prognostic score that incorporates molecular and cytogenetic data in patients with MF. Methods: We analyzed clinical, cytogenetic and molecular data from 623 patients with a diagnosis of primary MF (N=445), post-PV MF (N=109) and post-ET MF (N=69). Data was extracted from medical records at time of sample collection for analysis. Mutation data was obtained by next-generation sequencing analysis, performed with either paired tumor-normal whole exome sequencing (N=46) or selected gene panel for genes associated with myeloid malignancies (N=577). The following 16 genes were analyzed in all 623 patients and were considered as the common denominator for analysis: ASXL1, CALR, DNMT3A, EZH2, FLT3, IDH1, IDH2, JAK2, KIT, KRAS, MPL, NRAS, RUNX1, TET2, TP53, WT1. RAS mutations were considered as oncogenic mutations in NRAS and/or KRAS. Molecular high risk (MHR) mutations were considered as mutations in any one of the 4 genes: ASXL1, EZH2, IDH1, IDH2 (SRSF2 mutations were not included since they were not evaluated in all cases). Cytogenetic data was stratified into 4 risk categories (based on Tam et al, Blood 2009): (1) Diploid; (2) Del(13q)/Del(20q)/Trisomy 9; (3) Abnormalities of chromosomes 5, 7, 17 and complex karyotype; (4) Other abnormalities. To develop the model, the data was split into a training dataset (N=434) and a test dataset (N=189). Variables initially included in the initial training model were those with a p-value Results: In the training cohort, after a median follow-up of 30.8 months, there were 176 deaths (40.5%). The initial variables included in the multivariate Cox model were: age (>65 years), hemoglobin (25x109/L), peripheral blood blasts (>1%), presence of constitutional symptoms, sex (male vs female), platelet count ( Conclusion: Incorporating molecular data, including MHR mutations, karyotype and RAS mutations leads to the development of an improved prognostic model. Further validation of this model in other cohorts is necessary. The impact of novel therapies should be evaluated among the different risk categories. Disclosures Rampal: Stemline: Research Funding; Celgene: Honoraria; Constellation: Research Funding; Incyte: Honoraria, Research Funding; Jazz: Consultancy, Honoraria. Verstovsek:Celgene: Membership on an entity's Board of Directors or advisory committees; Italfarmaco: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Incyte: Consultancy.
- Published
- 2018
- Full Text
- View/download PDF
22. Prognostic Impact of Activating Mutations of the NRAS/KRAS Genes in Patients with Primary and Post-Essential Thrombocythemia (ET)/Polycythemia Vera (PV) Myelofibrosis (MF)
- Author
-
Raquel de Melo Alves Paiva, Bartlomiej Getta, Christopher Famulare, Srdan Verstovsek, Renato David Puga, Raajit K. Rampal, Paulo Vidal Campregher, Lucia Masarova, Hagop M. Kantarjian, Ross L. Levine, Fabio Ps Santos, Jessica Schulman, and Nelson Hamerschlak
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Essential thrombocythemia ,business.industry ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,medicine.disease_cause ,Biochemistry ,Polycythemia vera ,medicine ,Cancer research ,In patient ,KRAS ,Myelofibrosis ,business ,Gene - Abstract
Introduction: MF is a Philadelphia-negative myeloproliferative neoplasm (Ph-negative MPN) associated with driver mutations in the JAK-STAT pathway (e.g. JAK2, CALR, MPL) and other mutations in genes that lead to epigenetic changes and altered RNA splicing (e.g. TET2, SRSF2, ASXL1, EZH2). The RAS-signaling pathway is frequently altered in acute myeloid leukemia (AML) and other myeloid malignancies, but few studies have evaluated the prevalence of such mutations in patients with MF. We sought to describe the frequency and clinical features of RAS mutations in patients with MF. Methods: We analyzed next-generation sequencing data from 723 patients with a diagnosis of primary MF (N=520), post-PV MF (N=119) and post-ET MF (N=84). Sequencing was performed with either paired tumor-normal whole exome sequencing (WES; N=56) or selected gene panel for genes associated with myeloid malignancies (N=667). The following 16 genes were analyzed in all 723 patients and were considered as the common denominator for analysis: ASXL1, CALR, DNMT3A, EZH2, FLT3, IDH1, IDH2, JAK2, KIT, KRAS, MPL, NRAS, RUNX1, TET2, TP53, WT1. RAS mutations were considered as oncogenic mutations in NRAS and/or KRAS. Molecular high risk (MHR) mutations were considered as mutations in any one of the 4 genes: ASXL1, EZH2, IDH1, IDH2 (SRSF2 mutations were not included since they were not evaluated in all cases). Odds ratio (OR) and P-values were estimated using Fisher's exact test in pairwise comparisons among genetic features, and P-values were adjusted for multiple comparisons using the Benjamini-Hochberg procedure, with significant Q-values considered as those Results: Mutations in RAS genes were found in 44 patients (6.1%; 95% confidence interval [CI] 4.5-8.1%). There were 32 cases with NRAS mutations (4.4%; 95% CI 3-6.2%), 15 cases with KRAS mutations (2%; 95% CI 1.2-3.4%), and there were 3 cases with mutations in both genes (0.4%; 95% CI 0.1-1.3%). JAK-STAT driver genes found in the cohort included JAK2 (N=486), CALR (N=94), MPL (N=35) and Triple-negative status (N=108), and there was no difference in the distribution of JAK-STAT driver genes among patients with NRAS/KRAS mutations (p=0.96). Patients with NRAS/KRAS mutations had a higher number of non-driver mutations (median 3 vs 1; p=8.5e-20), higher white blood cell counts (median 15.1x109/L vs 10.5x109/L, p=0.02) and more frequently harbored ASXL1 mutations (OR 3.00, q=0.01), EZH2 mutations (OR 3.40, q=0.11) and MHR mutations (OR 3.13, q=0.004). There was a negative association of NRAS/KRAS mutations with del(20q) changes in karyotype (OR Conclusion: Patients with a diagnosis of MF who harbor NRAS/KRAS mutations comprise a high-risk subgroup with poor outcomes. NRAS/KRAS mutational status should be evaluated in future prognostic models in MF. The efficacy of JAK2 inhibitors needs to be further studied in this subgroup of patients, as well as the possibility of targeting the RAS pathway with MEK inhibitors. Disclosures Rampal: Incyte: Honoraria, Research Funding; Jazz: Consultancy, Honoraria; Celgene: Honoraria; Stemline: Research Funding; Constellation: Research Funding. Verstovsek:Italfarmaco: Membership on an entity's Board of Directors or advisory committees; Incyte: Consultancy; Celgene: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau.
- Published
- 2018
- Full Text
- View/download PDF
23. Molecular Predictors and Current Management of Minimal Residual Disease (MRD) Following Induction Chemotherapy for Acute Myeloid Leukemia (AML)
- Author
-
Justin Taylor, Martin S. Tallman, Andrew Dunbar, Ross L. Levine, Elli Papaemmanuil, Maria E. Arcila, Kamal Menghrajani, Miguel-Angel Perales, Esperanza B. Papadopoulos, Sean M. Devlin, Sergio Giralt, Erin McGovern, Christopher Famulare, Bartlomiej Getta, Mikhail Roshal, Jacob L. Glass, Noushin Farnoud, Aaron D. Viny, Jessica Schulman, Brian C. Shaffer, Boglarka Gyurkocza, Aaron D Goldberg, Minal Patel, Sheng Cai, Yanming Zhang, and Zachary D. Epstein-Peterson
- Subjects
Oncology ,medicine.medical_specialty ,Anthracycline ,business.industry ,Immunology ,Induction chemotherapy ,Cancer ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Minimal residual disease ,body regions ,Clinical trial ,Transplantation ,medicine.anatomical_structure ,hemic and lymphatic diseases ,Internal medicine ,Cytarabine ,medicine ,Bone marrow ,business ,medicine.drug - Abstract
Background: MRD is a powerful prognostic factor in AML. Emerging data indicate that allogeneic stem cell transplant (alloSCT) with MRD results in outcomes equivalently poor to alloSCT with morphologic AML (Araki et al., JCO 2016). Genomic predictors of MRD are unclear, and relative efficacy of therapies for MRD remains elusive. Objectives: Here we provide an integrated analysis of responses for 163 patients (pts) who underwent induction chemotherapy with baseline next-generation sequencing (NGS) followed by serial immunophenotypic monitoring for MRD. Methods:163 patients starting in April 2014 who underwent induction chemotherapy at Memorial Sloan Kettering Cancer Center were retrospectively studied. All received anthracycline + cytarabine, with or without investigational agents. Immunophenotypic MRD was identified in bone marrow aspirates (BMA) by multiparameter flow cytometry. Any level of residual disease was considered MRD+. Molecular analysis was obtained from pre-induction BMA by NGS using 28 or 49 gene panels. Cytogenetics/FISH were performed using standard techniques. Results: Patient characteristics are in Table 1. 7/163 (4.9%) died within 30 days of induction.153 pts had BM biopsy after induction prior to further therapy. 124/153 underwent flow after induction. 65/124 (52.4%) achieved CR/CRi after induction alone, 31/124 (25%) MRD+CR/CRi, and 34/124 (27.4%) MRD-CR/CRi. Pre-induction molecular analysis from 126 suggests that certain cytogenetic and molecular abnormalities correlate with achievement of MRD-CR. (Figure 1) Only 2/25 (8%) with RUNX1, 0/13 with SF3B1, and 0/11 with TP53 mutations achieved MRD-CR/CRi as best response after 1 cycle of induction. Only 3 additional RUNX1, 2 SF3B1, and 0/11 TP53 achieved MRD-CR/CRi as best response after a second cycle of therapy. In contrast, 7/8 with CBF AML (inv16 and no KIT mutation, n=4) or (t(8;21), n=3) achieved MRD-CR/CRi (n=5) or CR without flow (n=2) after 1 cycle of induction. 91/163 (55.8%) underwent alloSCT following induction or additional therapy. Post-alloSCT follow-up indicates potential value in converting MRD+ to MRD-. 84/91 were evaluable for MRD with flow cytometry prior to alloSCT. 41/84 (48.8%) were MRD-, 30/84 (35.7%) MRD+, and 13/84 (15.4%) persistent AML. 13/41 (31.7%) MRD-pre-alloSCT were MRD- post-induction. 28/41 (68.2%) MRD+ or persistent AML converted to MRD- prior to alloSCT following additional therapy. 23/29 MRD+CR/CRi pts after induction were intermediate/unfavorable and therefore transplant candidates. 19/23 MRD+CR/CRi intermediate/unfavorable underwent transplant (9 without post-induction therapy, 10 after consolidation), while 4 did not proceed to transplant due to relapse after induction (n=1), relapse after consolidation (n=2), and patient preference. There was no significant difference in post-transplant OS between early MRD-CR immediately following induction and later conversion to MRD-CR prior to alloSCT (Figure 1B). Post-transplant analysis reveals that most pts who enter transplant with persistent AML (n=13) or MRD+ (n=30) clear MRD (30/43, 69.7%) by the first post-transplant BM (median 32 days, Figure 1C). Despite initial post-transplant MRD clearance, pts who entered alloSCT with persistent AML or MRD+ had poorer post-transplant OS compared to pts who entered alloSCT with MRD- (p=0.02, Figure 1D). Conclusion: Our data show that AML pts with specific molecular mutations (RUNX1, SF3B1, and TP53) are unlikely to achieve MRD-CR/CRi after induction chemotherapy. We further show that additional therapy such as consolidation may be advantageous for some MRD+ pts to achieve MRD-CR prior to alloSCT, although others remain resistant to MRD clearance. Post-transplant OS is improved in pts who are MRD- at time of transplant, regardless of whether they required additional therapy beyond induction to achieve this state. Our results suggest that development of MRD-eradicating therapies after AML induction has the potential to improve post-transplant outcomes. Disclosures Goldberg: AROG: Research Funding; Pfizer: Research Funding; Celgene: Consultancy. Arcila:Invivoscribe, Inc.: Consultancy, Honoraria. Perales:Takeda: Other: Personal fees; Merck: Other: Personal fees; Abbvie: Other: Personal fees; Incyte: Membership on an entity's Board of Directors or advisory committees, Other: Personal fees and Clinical trial support; Novartis: Other: Personal fees. Tallman:ADC Therapeutics: Research Funding; Daiichi-Sankyo: Other: Advisory board; Orsenix: Other: Advisory board; Cellerant: Research Funding; BioSight: Other: Advisory board; AROG: Research Funding; AbbVie: Research Funding.
- Published
- 2018
- Full Text
- View/download PDF
24. Loss of Plasmacytoid Dendritic Cell Differentiation Is Highly Predictive for Persistent Measurable Residual Disease and Poor Outcomes in Acute Myeloid Leukemia
- Author
-
Kamal Menghrajani, Andrew Dunbar, Mark B. Geyer, Jacob L. Glass, Justin Taylor, Aaron D Goldberg, Yanming Zhang, Martin S. Tallman, Zachary D. Epstein-Peterson, Bartlomiej Getta, Erin McGovern, Wenbin Xiao, Christopher Famulare, Jessica Schulman, Minal Patel, Akshar Patel, Aaron D. Viny, Sheng Cai, Mikhail Roshal, Sean Delvin, and Ross L. Levine
- Subjects
business.industry ,hemic and lymphatic diseases ,Immunology ,Cancer research ,Myeloid leukemia ,Medicine ,Cell Biology ,Hematology ,Disease ,Plasmacytoid dendritic cell differentiation ,business ,Biochemistry - Abstract
Background Measurable residual disease (MRD) is associated with inferior outcomes in patients with acute myeloid leukemia (AML). MRD monitoring enhances risk stratification and may guide therapeutic intervention. Post-induction MRD is frequently cleared with further therapy and the clearance may lead to better outcomes. In contrast, persistent MRD is associated with poor outcomes. At present it is not possible to predict which patients are likely to clear MRD with further therapy. Here we report a simple, objective, widely applicable and quantitative MFC approach using the ratio of blast/PDC to predict persistent MRD and poor outcomes in AML. Patients and Methods A cohort of 136 adult patients with a confirmed diagnosis of AML by WHO criteria who underwent standard induction therapy at a single center between 4/2014 and 9/2017 was initially included. 69 patients achieved complete morphologic remission (36 MRD-neg. and 33 MRD-pos.). MRD status was assessed by MFC using a different from normal (DfN) approach. PDC were quantified as the percent of total WBC by flow cytometry based on low side scatter, moderate CD45, CD303, bright CD123 and HLA-DR expression. Results The proportion of PDC was markedly decreased in patients with AML (≥20% blasts) (N=136) with a median of 0.016% (interquartile range IQR: 0.0019%-0.071%, Figure 1A), more than 10-fold lower than observed in normal controls (median 0.23%, IQR 0.17%-0.34%) (N=20). While there was no difference between MRD-neg. and normal control groups (median 0.31%, IQR: 0.17%-0.49%; vs. 0.28%, IQR: 0.17%-0.34%), MRD-pos. group had significantly reduced PDC proportion compared to the control (median 0.074%, IQR: 0.022%-0.33%, Wilcoxon rank sum, p=0.019). In an attempt to achieve better separation and to eliminate possible effects of hemodilution, the ratio of blast/PDC was calculated by using the proportions of blasts and PDCs out of total WBCs as quantitated by flow cytometry. A cut-off threshold of the blast/PDC ratio of 10 was chosen to separate each group (Figure 1B). Importantly, a ratio cut-off of 10 had a corresponding specificity of 97.4% for predicting MRD positivity status. MRD positivity was significantly associated with inferior overall survival (OS) and relapse-free survival (RFS) in our study cohort (OS HR 4.11 (95% CI: 1.30-13.03), p=0.016; RFS HR 4.20 (95% CI: 1.49-11.82), p=0.007, Figure 1C and D). The 2-year cumulative incidence of relapse in the MRD-neg. group compared to MRD-pos. group was 10% (95% CI: 2-24%) vs. 37% (95% CI: 18-56%, p=0.014). Importantly, blast/PDC ratio ≥10 was also strongly associated with inferior OS and RFS (OS HR 3.12 (95% CI: 1.13-8.60), p= 0.028; RFS HR 4.05 (95% CI: 1.63-10.11), p=0.003, Figure 1E and F), which is similar in magnitude to MRD positivity. Furthermore, MRD-pos. patients with blast/PDC ratio Conclusion We have established an objective and quantitative MFC method to risk stratify post induction AML patients by risk for relapse, MRD clearance and likelihood of survival. Loss of PDC correlates with residual leukemia, is highly specific for MRD positivity in post-induction patients, and strongly predicts poorer overall survival and higher likelihood of relapse. Loss of PDC also predicts persistent MRD in post-induction MRD-pos. patients despite further therapy, suggesting that MRD-pos. patients with normal PDC may benefit from further therapy prior to transplant, while MRD-pos. patients with loss of PDC may not. Figure 1. Figure 1. Disclosures Goldberg: AROG: Research Funding; Pfizer: Research Funding; Celgene: Consultancy. Geyer:Dava Oncology: Honoraria. Levine:Isoplexis: Equity Ownership; C4 Therapeutics: Equity Ownership; Gilead: Honoraria; Qiagen: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Prelude: Research Funding; Imago: Equity Ownership; Roche: Consultancy, Research Funding; Loxo: Consultancy, Equity Ownership; Celgene: Consultancy, Research Funding; Novartis: Consultancy; Epizyme: Patents & Royalties; Janssen: Consultancy, Honoraria. Tallman:BioSight: Other: Advisory board; AROG: Research Funding; AbbVie: Research Funding; Cellerant: Research Funding; ADC Therapeutics: Research Funding; Orsenix: Other: Advisory board; Daiichi-Sankyo: Other: Advisory board.
- Published
- 2018
- Full Text
- View/download PDF
25. Oncologic therapy for solid tumors alters the risk of clonal hematopoiesis
- Author
-
Elsa Bernard, Elli Papaemmanuil, Jessica Schulman, Mariko Yabe, Howard I. Scher, Lior Z. Braunstein, Michael F. Berger, Catherine C. Coombs, Sean M. Devlin, David M. Hyman, Stuart Gardos, Martin S. Tallman, Luis A. Diaz, Daniel Kelly, Ryan Ptashkin, Kelly L. Bolton, Dominik Glodzik, Matahi Moarii, Akshar Patel, Antonin Berthon, Dean F. Bajorin, Ross L. Levine, Nilanjan Chatterjee, Michael Francis Walsh, José Baselga, Ahmet Zehir, Minal Patel, Choonsik Lee, Montserrat Garcia-Closas, Virginia M. Klimek, Diana Mandelker, Lindsay M. Morton, and Gunes Gundem
- Subjects
business.industry ,medicine.medical_treatment ,Immunology ,Clonal hematopoiesis ,Cancer ,Cell Biology ,Hematology ,Immunotherapy ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Radiation therapy ,03 medical and health sciences ,Leukemia ,Haematopoiesis ,0302 clinical medicine ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,business ,Protein p53 ,030215 immunology - Abstract
Background: Solid tumor patients are at a heightened risk for developing therapy-related myeloid neoplasms (tMN). Recent studies show evidence of somatic mutations in leukemia-associated genes in normal healthy individuals, referred to as clonal hematopoiesis (CH). We and others have shown that clonal hematopoiesis (CH) is also frequent in cancer patients. A detailed characterization of the relationship between exposure to specific oncologic regimens, the molecular features of CH presentation and how these relate to tMN risk is warranted to inform treatment decisions, early detection and prevention strategies. Methods: To determine the relationship between CH and oncologic therapy, we performed a systematic interrogation of CH in a cohort of 17,478 solid tumor patients with clinical, outcome and molecular profiling by MSK-IMPACT. MSK-IMPACT is a targeted panel of cancer-associated mutations used to screen tumor samples against a blood control sample. Mutation detection was performed on blood derived sequencing data (median coverage at 600x) using the matched tumor as a comparator and accounted for background sequencing error rates. Results: Overall, 40% of the 17,478 patients were treatment naïve prior to IMPACT testing, 37% had received chemotherapy alone, 17% had received radiation therapy and 18% had received both. CH was identified in 4013 (23%) of patients, median VAF was 4% (range=1-80%). The vast majority (76%) had a single mutation whereas 9% had two and 5% had three or more. The number of mutations correlated with clone size (p-value= Conclusions: CH is frequent in solid tumor patients and can be reliably detected when a matched tumor normal targeted gene sequencing approach is performed. Beyond age, CH is strongly associated with race, smoking and importantly prior exposure to oncologic therapy with evidence of specific treatment effects. Taken together, we show that screening of CH in cancer cohorts is critical to the development of future clinical guidelines, the development of risk-adapted treatment decisions, surveillance programs and definition of patient subsets at highest risk for tMN. Disclosures Coombs: Incyte: Other: Travel fees; DAVA Oncology: Honoraria; Abbvie: Consultancy; H3 Biomedicine: Honoraria; AROG: Other: Travel fees. Tallman:Daiichi-Sankyo: Other: Advisory board; Cellerant: Research Funding; BioSight: Other: Advisory board; AROG: Research Funding; Orsenix: Other: Advisory board; AbbVie: Research Funding; ADC Therapeutics: Research Funding. Yabe:Y-mAbs Therapeutics: Consultancy. Levine:Celgene: Consultancy, Research Funding; Novartis: Consultancy; Gilead: Honoraria; Isoplexis: Equity Ownership; Epizyme: Patents & Royalties; Prelude: Research Funding; C4 Therapeutics: Equity Ownership; Janssen: Consultancy, Honoraria; Imago: Equity Ownership; Qiagen: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Roche: Consultancy, Research Funding; Loxo: Consultancy, Equity Ownership.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.