204 results on '"Timothy A. Graubert"'
Search Results
2. Mutant U2AF1-expressing cells are sensitive to pharmacological modulation of the spliceosome
- Author
-
Cara Lunn Shirai, Brian S. White, Manorama Tripathi, Roberto Tapia, James N. Ley, Matthew Ndonwi, Sanghyun Kim, Jin Shao, Alexa Carver, Borja Saez, Robert S. Fulton, Catrina Fronick, Michelle O’Laughlin, Chandraiah Lagisetti, Thomas R. Webb, Timothy A. Graubert, and Matthew J. Walter
- Subjects
Science - Abstract
Spliceosome mutations occur in approximately 50% of patients with myelodysplastic syndromes. Here, the authors show that tumour cells harbouring theS34F mutation in the U2AFspliceosome gene is sensitive to compounds that further perturb the spliceosome.
- Published
- 2017
- Full Text
- View/download PDF
3. Biology of secondary leukemia
- Author
-
Andrew M. Brunner and Timothy A. Graubert
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2018
- Full Text
- View/download PDF
4. Phase I study of the aurora A kinase inhibitor alisertib with induction chemotherapy in patients with acute myeloid leukemia
- Author
-
Amir T. Fathi, Seth A. Wander, Traci M. Blonquist, Andrew M. Brunner, Philip C. Amrein, Jeffrey Supko, Nicole M. Hermance, Amity L. Manning, Hossein Sadrzadeh, Karen K. Ballen, Eyal C. Attar, Timothy A. Graubert, Gabriela Hobbs, Christelle Joseph, Ashley M. Perry, Meghan Burke, Regina Silver, Julia Foster, Meghan Bergeron, Aura Y. Ramos, Tina T. Som, Kaitlyn M. Fishman, Kristin L. McGregor, Christine Connolly, Donna S. Neuberg, and Yi-Bin Chen
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Aberrant expression of aurora kinase A is implicated in the genesis of various neoplasms, including acute myeloid leukemia. Alisertib, an aurora A kinase inhibitor, has demonstrated efficacy as monotherapy in trials of myeloid malignancy, and this efficacy appears enhanced in combination with conventional chemotherapies. In this phase I, dose-escalation study, newly diagnosed patients received conventional induction with cytarabine and idarubicin, after which alisertib was administered for 7 days. Dose escalation occurred via cohorts. Patients could then receive up to four cycles of consolidation, incorporating alisertib, and thereafter alisertib maintenance for up to 12 months. Twenty-two patients were enrolled. One dose limiting toxicity occurred at dose level 2 (prolonged thrombocytopenia), and the recommended phase 2 dose was established at 30mg twice daily. Common therapy-related toxicities included cytopenias and mucositis. Only three (14%) patients had persistent disease at mid-cycle, requiring “5+2” reinduction. The composite remission rate (complete remission and complete remission with incomplete neutrophil recovery) was 86% (nineteen of twenty-two patients; 90% CI 68–96%). Among those over age 65 and those with high-risk disease (secondary acute leukemia or cytogenetically high-risk disease), the composite remission rate was 88% and 100%, respectively. The median follow up was 13.5 months. Of those treated at the recommended phase 2 dose, the 12-month overall survival and progression-free survival were 62% (90% CI 33–81%) and 42% (90% CI 17–65%), respectively. Alisertib is well tolerated when combined with induction chemotherapy in acute myeloid leukemia, with a promising suggestion of efficacy. (clinicaltrials.gov Identifier:01779843).
- Published
- 2017
- Full Text
- View/download PDF
5. Subclonal KIT D816V Mutations Are Prevalent in Chronic Myelomonocytic Leukemia and Correlate with Distinct Phenotypic Features
- Author
-
Anthony Hunter, Hannah Newman, Eric Solary, Klaus Geissler, Laura Palomo, Lurdes Zamora, Francesc Solé, Valeria Santini, Timothy A. Graubert, Swapna Thota, Elizabeth A. Griffiths, Lisa Pleyer, Felicitas R. Thol, Rafael Bejar, Luis E. Aguirre, David A. Sallman, Andrew Kuykendall, Rami S. Komrokji, Tracy I. George, and Eric Padron
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
6. Data from Spliceosome Mutations Induce R Loop-Associated Sensitivity to ATR Inhibition in Myelodysplastic Syndromes
- Author
-
Timothy A. Graubert, Lee Zou, Matthew J. Walter, Jack D. Sullivan, Pavankumar N.G. Reddy, Weiling Li, Wan Yee Leong, and Hai Dang Nguyen
- Abstract
Heterozygous somatic mutations in spliceosome genes (U2AF1, SF3B1, ZRSR2, or SRSF2) occur in >50% of patients with myelodysplastic syndrome (MDS). These mutations occur early in disease development, suggesting that they contribute to MDS pathogenesis and may represent a unique genetic vulnerability for targeted therapy. Here, we show that RNA splicing perturbation by expression of the U2AF1(S34F) mutant causes accumulation of R loops, a transcription intermediate containing RNA:DNA hybrids and displaced single-stranded DNA, and elicits an ATR response. ATR inhibitors (ATRi) induced DNA damage and cell death in U2AF1(S34F)-expressing cells, and these effects of ATRi were enhanced by splicing modulating compounds. Moreover, ATRi-induced DNA damage was suppressed by overexpression of RNaseH1, an enzyme that specifically removes the RNA in RNA:DNA hybrids, suggesting that the ATRi sensitivity of U2AF1(S34F)-expressing cells arises from R loops. Taken together, our results demonstrate that ATR may represent a novel therapeutic target in patients with MDS carrying the U2AF1(S34F) mutation and potentially other malignancies harboring spliceosome mutations.Significance: This study provides preclinical evidence that patients with MDS or other myeloid malignancies driven by spliceosome mutations may benefit from ATR inhibition to exploit the R loop–associated vulnerability induced by perturbations in splicing. Cancer Res; 78(18); 5363–74. ©2018 AACR.
- Published
- 2023
7. Figure S1 from Spliceosome Mutations Induce R Loop-Associated Sensitivity to ATR Inhibition in Myelodysplastic Syndromes
- Author
-
Timothy A. Graubert, Lee Zou, Matthew J. Walter, Jack D. Sullivan, Pavankumar N.G. Reddy, Weiling Li, Wan Yee Leong, and Hai Dang Nguyen
- Abstract
U2AF1(S34F)-expressing cells induce R loop-associated ATR-mediated RPA32 phosphorylation.
- Published
- 2023
8. Table S1 from Spliceosome Mutations Induce R Loop-Associated Sensitivity to ATR Inhibition in Myelodysplastic Syndromes
- Author
-
Timothy A. Graubert, Lee Zou, Matthew J. Walter, Jack D. Sullivan, Pavankumar N.G. Reddy, Weiling Li, Wan Yee Leong, and Hai Dang Nguyen
- Abstract
List of primers used for RNA splicing assay
- Published
- 2023
9. Therapeutic Targeting of Spliceosome Mutant Myeloid Neoplasms Via PARP1 Inhibition
- Author
-
Sayantani Sinha, Zhiyan Silvia Liu, Maxwell Bannister, Erica Arriaga-Gomez, Axia Song, Dawei Zong, Martina Sarchi, Elizabeth Bonner, Victor Corral, Cassandra Leibson, Wannasiri Chiraphapphaiboon, Derek Stirewalt, Joachim Deeg, Sumit Rai, Matthew J Walter, Timothy A. Graubert, Sergei Doulatov, Dang Hai Nguyen, and Stanley Chun-Wei Lee
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
10. A Phase 2 Randomized Study Comparing Venetoclax and Azacitidine to Conventional Induction Chemotherapy for Newly Diagnosed Fit Adults with Acute Myeloid Leukemia
- Author
-
Amir T. Fathi, Geoffrey G Fell, Areej El-Jawahri, Alexander E. Perl, Brian A. Jonas, Ajoy L. Dias, Alice S. Mims, Uma Borate, Brittany Knick Ragon, Michael R. Grunwald, Mary Linton B. Peters, Timothy A. Graubert, Philip C. Amrein, Hanno R. Hock, Andrew M. Brunner, Gabriela S. Hobbs, Rupa Narayan, Donna S. Neuberg, and Ibrahim Aldoss
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
11. Supporting Patients with Cancer after Dobbs v. Jackson Women's Health Organization
- Author
-
Andrew G Shuman, Matti S Aapro, Benjamin Anderson, Katherine Arbour, Pedro C Barata, Aditya Bardia, Eduardo Bruera, Bruce A Chabner, Herbert Chen, Edwin Choy, Pierfranco Conte, Giuseppe Curigliano, Don Dizon, Eileen O’Reilly, Antonio Tito Fojo, Hans Gelderblom, Timothy A Graubert, Jayne S Gurtler, Evan Hall, Fred R Hirsch, Ahmed Idbaih, David H Ilson, Michael Kelley, Carlo La Vecchia, Heinz Ludwig, Beverly Moy, Hyman Muss, Frans Opdam, Rebecca D Pentz, Marshall R Posner, Jeffrey S Ross, Adrian Sacher, Suresh Senan, Enrique Soto-Perez-de-Celis, Kenneth K Tanabe, Jan B Vermorken, Eric Wehrenberg-Klee, Susan E Bates, Radiation Oncology, CCA - Cancer Treatment and quality of life, and CCA - Cancer biology and immunology
- Subjects
Cancer Research ,Oncology ,Human medicine - Abstract
In the context of cancer, whether or not to choose pregnancy termination represents a difficult and multifaceted decision. In this editorial, members of The Oncologist editorial team attempt to contextualize the potential implications of the recent Supreme Court decision in Dobbs v. Jackson Women’s Health Organizationfor patients with cancer.
- Published
- 2022
12. Abstract 6183: PARP inhibitors preferentially sensitize splicing factor mutant myeloid neoplasms
- Author
-
Dang Hai Nguyen, Sayantani Sinha, Zhiyan Silvia Liu, Maxwell Henry Bannister, Erica Arriaga-Gomez, Axia Song, Dawei Zong, Martina Sarchi, Victor Corral, Wannasiri Chiraphapphaiboon, Jennifer Yoo, Matthew McMahon, Cassandra Leibson, Derek L. Stirewalt, H Joachim Deeg, Sumit Rai, Matthew Walter, Timothy A. Graubert, Sergei Doulatov, and Stanley C. Lee
- Subjects
Cancer Research ,Oncology - Abstract
Somatic heterozygous mutations in genes encoding for RNA splicing factors (SF) SRSF2, U2AF1, and SF3B1 are frequently mutated in patients with hematologic malignancies, representing a unique genetic vulnerability for targeted therapy. In the current study, we performed a focused drug screen with inhibitors targeting different DNA damage response and DNA metabolic pathways to identify novel therapeutic vulnerabilities generated by SF mutations. We generated a murine leukemia model by overexpressing the MLL-AF9 fusion oncogene on an Srsf2P95H/+ background, a mutational combination that is found in ~10% of MLL-rearranged leukemias. We surprisingly found that MLL-AF9 Srsf2P95H/+ mutant leukemias are sensitive to inhibitors targeting ADP-ribosyltransferases (PARP). PARP inhibitor sensitivity was also observed in isogenic murine MLL-AF9 U2af1s34/+ cells compared to MLL-AF9 U2af1+/+ cells. Second, murine Srsf2P95H leukemias showed improved prolonged survival when treated with olaparib (PARPi) compared to vehicle treatment in vivo. Third, human primary AML patient samples that harbor SF mutations are sensitive to PARPi compared to non-SF mutant samples. These data highlight that both SRSF2P95H and U2AF1S34F mutations create a common vulnerability that is dependent on PARP activity for survival. To evaluate PARP activity, we used isogenic K562 leukemia cells expressing SRSF2P95H and U2AF1S34F mutations from their endogenous loci and monitored ADP-ribosylation (ADPr) levels, a marker of PARP activity. Both SRSF2P95H and U2AF1S34F cells exhibited elevated levels of ADPr compared to wildtype cells in a PARP1- dependent manner. PARPi preferentially induced DNA damage and cell death in SF mutant cells. Surprisingly, we found that SRSF2P95H and U2AF1S34F cells are not defective in homologous recombination repair. Instead, the increased PARP1-mediated ADPr in SF-mutant cells is caused by accumulated R loops, a group of transcription intermediates containing RNA:DNA hybrids and displaced single-stranded DNA. To determine whether PARPi sensitivity is due to R-loop accumulation, we overexpressed RNase H1, an enzyme that specifically cleaves the RNA moiety within RNA:DNA hybrids in U2AF1S34F cells. Overexpression of RNase H1 significantly reduced ADPr levels and suppressed the PARPi-induced U2AF1S34F cell growth inhibition. Collectively, these results suggest that spliceosome mutants induce R-loop accumulation and elicit an R-loop-associated PARP1 response to promote cell survival. In summary, our data establish a previously unknown link between R-loop-induced PARP1 response and RNA splicing perturbation and provide a mechanistic rationale to evaluate the clinical efficacy of PARP inhibitors in spliceosome-mutant malignancies. Furthermore, our study highlights a new therapeutic potential of targeting the R-loop tolerance pathways caused by different spliceosome gene mutations. Citation Format: Dang Hai Nguyen, Sayantani Sinha, Zhiyan Silvia Liu, Maxwell Henry Bannister, Erica Arriaga-Gomez, Axia Song, Dawei Zong, Martina Sarchi, Victor Corral, Wannasiri Chiraphapphaiboon, Jennifer Yoo, Matthew McMahon, Cassandra Leibson, Derek L. Stirewalt, H Joachim Deeg, Sumit Rai, Matthew Walter, Timothy A. Graubert, Sergei Doulatov, Stanley C. Lee. PARP inhibitors preferentially sensitize splicing factor mutant myeloid neoplasms. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6183.
- Published
- 2023
13. Mutant Srsf2 Diminishes Jak2V617F-Induced Erythrocytosis in Mice and Is Associated with Lower Hemoglobin in Patients with Chronic Phase JAK2-Mutant MPN
- Author
-
Anna E Marneth, Jonas S. Jutzi, Chulwoo J. Kim, Charles Laurore, Anastasia Tishena, Frederike Kramer, Azucena V. Rocha, Mohammed Wazir, Lachelle D. Weeks, Joan How, Maximilian F. Stahl, Marlise R. Luskin, R. Coleman Lindsley, Olga Pozdnyakova, Timothy A. Graubert, and Ann Mullally
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
14. Long: molecular tracking of CML with bilineal inv(16) myeloid and del(9) lymphoid blast crisis and durable response to CD19-directed CAR-T therapy
- Author
-
Jenna A. Moran, Philip C. Amrein, Chrisa Hunnewell, Amir T. Fathi, Meghan Bergeron, Timothy A. Graubert, Valentina Nardi, Julia Foster, Andrew M. Brunner, Keagan S Casey, Maristela L. Onozato, Vinayak Venkataraman, Steven L. McAfee, Matthew J. Frigault, Gabriela S. Hobbs, Hanno Hock, and Paola Dal Cin
- Subjects
Cancer Research ,Blast Crisis ,Myeloid ,biology ,business.industry ,medicine.medical_treatment ,Treatment outcome ,Hematology ,Immunotherapy ,medicine.disease ,CD19 ,Myelogenous ,Leukemia ,medicine.anatomical_structure ,Oncology ,biology.protein ,Cancer research ,Medicine ,Car t cells ,business - Published
- 2020
15. Clonal hematopoiesis and measurable residual disease assessment in acute myeloid leukemia
- Author
-
Robert P. Hasserjian, Timothy A. Graubert, David P. Steensma, and Benjamin L. Ebert
- Subjects
Neoplasm, Residual ,Transplantation Conditioning ,Myeloid ,medicine.medical_treatment ,Immunology ,Hematopoietic stem cell transplantation ,Medical Oncology ,Biochemistry ,Leukemogenic ,Myeloid Neoplasm ,hemic and lymphatic diseases ,medicine ,Humans ,Transplantation, Homologous ,Diagnostic Techniques and Procedures ,business.industry ,Remission Induction ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,Cell Biology ,Hematology ,Prognosis ,medicine.disease ,Transplantation ,Leukemia, Myeloid, Acute ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,Perspective ,Cancer research ,Clonal Hematopoiesis ,business - Abstract
Current objectives regarding treatment of acute myeloid leukemia (AML) include achieving complete remission (CR) by clinicopathological criteria followed by interrogation for the presence of minimal/measurable residual disease (MRD) by molecular genetic and/or flow cytometric techniques. Although advances in molecular genetic technologies have enabled highly sensitive detection of AML-associated mutations and translocations, determination of MRD is complicated by the fact that many treated patients have persistent clonal hematopoiesis (CH) that may not reflect residual AML. CH detected in AML patients in CR includes true residual or early recurrent AML, myelodysplastic syndrome or CH that is ancestral to the AML, and independent or newly emerging clones of uncertain leukemogenic potential. Although the presence of AML-related mutations has been shown to be a harbinger of relapse in multiple studies, the significance of other types of CH is less well understood. In patients who undergo allogeneic hematopoietic cell transplantation (HCT), post-HCT clones can be donor-derived and in some cases engender a new myeloid neoplasm that is clonally unrelated to the recipient’s original AML. In this article, we discuss the spectrum of CH that can be detected in treated AML patients, propose terminology to standardize nomenclature in this setting, and review clinical data and areas of uncertainty among the various types of posttreatment hematopoietic clones.
- Published
- 2020
16. Combined Targeted Therapy for
- Author
-
Seth A, Wander, Robert P, Hasserjian, Kwadwo, Oduro, Krzysztof, Glomski, Valentina, Nardi, Gregory M, Cote, Timothy A, Graubert, Andrew M, Brunner, Yi-Bin A, Chen, and Amir T, Fathi
- Abstract
A 44-year-old woman with a prior history of myxoid liposarcoma who was previously treated with radiation, chemotherapy, and resection, was admitted with syncope and pancytopenia. She was diagnosed with a high-grade therapy-related myelodysplastic syndrome and treated with decitabine. Her disease soon progressed to overt acute myeloid leukemia (AML). She was treated with cytotoxic chemotherapy including cytarabine and topotecan, but she was not a candidate for further anthracycline exposure given her prior treatment for sarcoma and concern for cardiotoxicity. Her AML was refractory to two sequential induction regimens. Given that targeted genomic sequencing had revealed a
- Published
- 2022
17. A phase 1 study of the antibody‐drug conjugate brentuximab vedotin with re‐induction chemotherapy in patients with CD30‐expressing relapsed/refractory acute myeloid leukemia
- Author
-
Rupa Narayan, Robert P. Hasserjian, Margaret C. Wey, Tanya T. Behnan, Meghan Bergeron, Jessica Rae, Mason L. Mann, Ashkan Emadi, Christina Bertoli, Eyal C. Attar, Traci M. Blonquist, Christopher Lescinskas, Jenna A. Moran, Yi-Bin Chen, Steven L. McAfee, Vu H. Duong, Patricia Lesho, Timothy A. Graubert, Jennifer Lombardi Story, Andrew M. Brunner, Meghan Burke, Megan K. Vartanian, Gabriela S. Hobbs, Tina T. Som, Donna Neuberg, Hanno Hock, Molly Macrae, Julia Foster, Kristin McGregor, Philip C. Amrein, Amir T. Fathi, Karen K. Ballen, Frederic I. Preffer, Aura Y. Ramos, and Christine Connolly
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,Antibody-drug conjugate ,medicine.medical_specialty ,Immunoconjugates ,Maximum Tolerated Dose ,Population ,Ki-1 Antigen ,Disease-Free Survival ,Drug Administration Schedule ,Young Adult ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Recurrence ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,030212 general & internal medicine ,Brentuximab vedotin ,education ,Aged ,Etoposide ,Brentuximab Vedotin ,Mitoxantrone ,education.field_of_study ,business.industry ,Cytarabine ,Induction chemotherapy ,Induction Chemotherapy ,Middle Aged ,medicine.disease ,Chemotherapy regimen ,Leukemia, Myeloid, Acute ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Female ,business ,Febrile neutropenia ,medicine.drug - Abstract
Background Outcomes for patients with relapsed/refractory acute myeloid leukemia (R/R AML) remain poor. Novel therapies specifically targeting AML are of high interest. Brentuximab vedotin (BV) is an antibody-drug conjugate that is specific for human CD30. In this phase 1 dose escalation study, the authors evaluated the safety of BV combined with mitoxantrone, etoposide, and cytarabine (MEC) re-induction chemotherapy for patients with CD30-expressing R/R AML. Methods Using a standard dose escalation design, the authors evaluated 3 dose levels of BV (0.9 mg/kg, 1.2 mg/kg, and 1.8 mg/kg) administered once on day 1 followed by MEC on days 3 through 7. Results There were no dose-limiting toxicities noted and the maximum tolerated dose was not reached. The recommended phase 2 dose of BV was determined to be 1.8 mg/kg when combined with MEC. The side effect profile was similar to that expected from MEC chemotherapy alone, with the most common grade ≥3 toxicities being febrile neutropenia, thrombocytopenia, and anemia (toxicities were graded using the National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.0]). Among the 22 patients enrolled on the trial, the composite response rate was 36%, with a composite response rate of 42% noted among those who received the highest dose of BV. The median overall survival was 9.5 months, with a median disease-free survival of 6.8 months observed among responders. Approximately 55% of patients were able to proceed with either allogeneic hematopoietic stem cell transplantation or donor lymphocyte infusion. Conclusions The combination of BV with MEC was found to be safe in patients with CD30-expressing R/R AML and warrants further study comparing this combination with the use of MEC alone in this population (ClinicalTrials.gov identifier NCT01830777). Lay summary The outcomes for patients with relapsed/refractory acute myeloid leukemia (R/R AML) are exceptionally poor. New and emerging treatment combinations are actively being studied in an effort to improve outcomes. The authors examined the combination of brentuximab vedotin, an antibody product that recognizes a marker called CD30, with mitoxantrone, etoposide, and cytarabine (MEC), a common chemotherapy regimen, in patients with R/R AML that expressed the CD30 marker. The authors found that the combination was safe and well tolerated. Future studies comparing this new combination with the use of MEC alone can help to inform its effectiveness for this patient population.
- Published
- 2019
18. U2af1 is a haplo-essential gene required for hematopoietic cancer cell survival in mice
- Author
-
Ajay Khanna, Timothy A. Graubert, Brian A. Wadugu, Matthew Ndonwi, Michael O. Alberti, Tanzir Ahmed, Matthew J. Walter, Jie Liu, Jin Shao, Dennis L. Fei, Amanda Heard, Christopher A. Miller, Cara Lunn Shirai, Joseph Bradley, Sarah Grieb, and Sridhar Nonavinkere Srivatsan
- Subjects
Mice, Knockout ,Heterozygote ,Leukemia ,Somatic cell ,Mutant ,Neoplasms, Experimental ,General Medicine ,Biology ,Splicing Factor U2AF ,medicine.disease ,Molecular biology ,Neoplasm Proteins ,Mice ,Haematopoiesis ,Essential gene ,Hematologic Neoplasms ,Cancer cell ,medicine ,Animals ,Allele ,Gene ,Alleles ,Research Article - Abstract
Somatic mutations in the spliceosome gene U2AF1 are common in patients with myelodysplastic syndromes. U2AF1 mutations that code for the most common amino acid substitutions are always heterozygous, and the retained wild-type allele is expressed, suggesting that mutant hematopoietic cells may require the residual wild-type allele to be viable. We show that hematopoiesis and RNA splicing in U2af1 heterozygous knock-out mice was similar to control mice, but that deletion of the wild-type allele in U2AF1(S34F) heterozygous mutant expressing hematopoietic cells (i.e., hemizygous mutant) was lethal. These results confirm that U2AF1 mutant hematopoietic cells are dependent on the expression of wild-type U2AF1 for survival in vivo and that U2AF1 is a haplo-essential cancer gene. Mutant U2AF1 (S34F) expressing cells were also more sensitive to reduced expression of wild-type U2AF1 than non-mutant cells. Furthermore, mice transplanted with leukemia cells expressing mutant U2AF1 had significantly reduced tumor burden and improved survival after the wild-type U2af1 allele was deleted compared to when it was not deleted. These results suggest that selectively targeting the wild-type U2AF1 allele in heterozygous mutant cells could induce cancer cell death and be a therapeutic strategy for patients harboring U2AF1 mutations.
- Published
- 2021
19. A synthetic small molecule stalls pre-mRNA splicing by promoting an early-stage U2AF2-RNA complex
- Author
-
Mary J. Pulvino, Zackary Falls, Matthew J. Walter, Rakesh Chatrikhi, Andrew J. MacRae, Jermaine L. Jenkins, Callen F. Feeney, Clara L. Kielkopf, Melissa S. Jurica, William W. Brennessel, Sumit Rai, Georgios Alachouzos, Alison J. Frontier, Timothy A. Graubert, and Ram Samudrala
- Subjects
Clinical Biochemistry ,Mutant ,U2AF(65) ,01 natural sciences ,Biochemistry ,Drug Discovery ,RNA Precursors ,RNA, Neoplasm ,Cancer ,U2AF2 ,Molecular Structure ,Chemistry ,ribonucleoprotein targeting ,S34F mutant ,spliceosome inhibition ,Small molecule ,Cell biology ,Molecular Docking Simulation ,5.1 Pharmaceuticals ,RNA splicing ,Molecular Medicine ,Female ,Development of treatments and therapeutic interventions ,Macromolecule ,Protein subunit ,RNA Splicing ,Mutagenesis (molecular biology technique) ,Biology ,splicing factor mutation ,Article ,U2AF(35) ,Small Molecule Libraries ,U2AF1 ,Genetics ,Humans ,therapeutic strategy ,Molecular Biology ,Pharmacology ,010405 organic chemistry ,Mutagenesis ,RNA ,Splicing Factor U2AF ,0104 chemical sciences ,myelodysplastic syndrome ,HEK293 Cells ,Docking (molecular) ,Neoplasm ,K562 Cells ,Function (biology) - Abstract
Dysregulated pre-mRNA splicing is an emerging Achilles heel of cancers and myelodysplasias. To expand the currently limited portfolio of small molecule drug leads, we screened for chemical modulators of the U2AF complex, which nucleates spliceosome assembly and is mutated in myelodysplasias. A hit compound specifically enhances RNA binding by a U2AF2 subunit. Remarkably, the compound inhibits splicing of representative substrates in cells and stalls spliceosome assembly at the stage of U2AF function. Computational docking, together with structure-guided mutagenesis, indicates that the compound bridges an active conformation of the U2AF2 RNA recognition motifs via hydrophobic and electrostatic moieties. Altogether, our results highlight the potential of trapping early spliceosome assembly as an effective pharmacological means to manipulate pre-mRNA splicing. By extension, we suggest that stabilizing inactive checkpoints may offer a breakthrough approach for small molecule inhibition of multi-stage macromolecular assemblies.
- Published
- 2021
20. Immune Escape of Relapsed AML Cells after Allogeneic Transplantation
- Author
-
Jacqueline E. Payton, Michael P. Rettig, Christopher A. Miller, Nicole M. Helton, Matthew J. Walter, Jeffery M. Klco, Catrina Fronick, Peter Westervelt, John S. Welch, Daniel C. Link, Matthew J. Christopher, Allegra A. Petti, Sharon Heath, Timothy J. Ley, Michelle O'Laughlin, Richard K. Wilson, Lukas D. Wartman, John F. DiPersio, Timothy A. Graubert, Jack Baty, Eric J. Duncavage, Ezhilarasi Chendamarai, and Robert S. Fulton
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Chemotherapy ,Allogeneic transplantation ,Myeloid ,business.industry ,medicine.medical_treatment ,Myeloid leukemia ,General Medicine ,Hematopoietic stem cell transplantation ,medicine.disease ,Article ,Transplantation ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,business ,Exome sequencing - Abstract
BACKGROUND: As consolidation therapy for acute myeloid leukemia (AML), allogeneic hematopoietic stem-cell transplantation provides a benefit in part by means of an immune-mediated graft-versus-leukemia effect. We hypothesized that the immune-mediated selective pressure imposed by allogeneic transplantation may cause distinct patterns of tumor evolution in relapsed disease. METHODS: We performed enhanced exome sequencing on paired samples obtained at initial presentation with AML and at relapse from 15 patients who had a relapse after hematopoietic stem-cell transplantation (with transplants from an HLA-matched sibling, HLA-matched unrelated donor, or HLA-mismatched unrelated donor) and from 20 patients who had a relapse after chemotherapy. We performed RNA sequencing and flow cytometry on a subgroup of these samples and on additional samples for validation. RESULTS: On exome sequencing, the spectrum of gained and lost mutations observed with relapse after transplantation was similar to the spectrum observed with relapse after chemotherapy. Specifically, relapse after transplantation was not associated with the acquisition of previously unknown AML-specific mutations or structural variations in immune-related genes. In contrast, RNA sequencing of samples obtained at relapse after transplantation revealed dysregulation of pathways involved in adaptive and innate immunity, including down-regulation of major histocompatibility complex (MHC) class II genes (HLA-DPA1, HLA-DPB1, HLA-DQB1, and HLA-DRB1) to levels that were 3 to 12 times lower than the levels seen in paired samples obtained at presentation. Flow cytometry and immunohistochemical analysis confirmed decreased expression of MHC class II at relapse in 17 of 34 patients who had a relapse after transplantation. Evidence suggested that interferon-γ treatment could rapidly reverse this phenotype in AML blasts in vitro. CONCLUSIONS: AML relapse after transplantation was not associated with the acquisition of relapse-specific mutations in immune-related genes. However, it was associated with dysregulation of pathways that may influence immune function, including down-regulation of MHC class II genes, which are involved in antigen presentation. These epigenetic changes may be reversible with appropriate therapy. (Funded by the National Cancer Institute and others.)
- Published
- 2018
21. Discovery and Pharmacological Characterization of JNJ-64619178, a Novel Small-Molecule Inhibitor of PRMT5 with Potent Antitumor Activity
- Author
-
Tinne Verhulst, Lieven Meerpoel, Emmanuel Gustin, Weimei Sun, Matthew V. Lorenzi, Junchen Gu, Longen Zhou, Wim Bert Griet Schepens, Hillary Millar, Tongfei Wu, Viellevoye Marcel, Vineet Pande, Petra Vinken, Desiree De Lange, An Boeckx, Christopher Moy, Friederike Pastore, Geert Mannens, Sylvie Laquerre, Ulrike Philippar, Vipul Bhargava, Gaston Stanislas Marcella Diels, Thomas Nys, Kathryn Packman, Jan Willem Thuring, Erika van Heerde, Bie Verbist, Sumit Rai, Lijs Beke, Pegah Safabakhsh, Timothy A. Graubert, Yue Fan, Angelique N Gilbert, Dirk Brehmer, Vikki Keersmaekers, Barbara Morschhäuser, Danilo Fiore, David Walker, Amy J. Johnson, Brehmer, D., Beke, L., Wu, T., Millar, H. J., Moy, C., Sun, W., Mannens, G., Pande, V., Boeckx, A., van Heerde, E., Nys, T., Gustin, E. M., Verbist, B., Zhou, L., Fan, Y., Bhargava, V., Safabakhsh, P., Vinken, P., Verhulst, T., Gilbert, A., Rai, S., Graubert, T. A., Pastore, F., Fiore, D., Gu, J., Johnson, A., Philippar, U., Morschhauser, B., Walker, D., de Lange, D., Keersmaekers, V., Viellevoye, M., Diels, G., Schepens, W., Thuring, J. W., Meerpoel, L., Packman, K., Lorenzi, M. V., and Laquerre, S.
- Subjects
Cancer Research ,Protein-Arginine N-Methyltransferases ,Lung Neoplasms ,PROTEIN ,Splicing factor ,Mice ,In vivo ,REVEALS ,medicine ,Animals ,Humans ,Pyrroles ,Enzyme Inhibitors ,Lung cancer ,VULNERABILITY ,Science & Technology ,business.industry ,Protein arginine methyltransferase 5 ,PRE-MESSENGER-RNA ,METHYLATION ,Myeloid leukemia ,SELECTIVE INHIBITOR ,medicine.disease ,Lymphoma ,Disease Models, Animal ,Pyrimidines ,Oncology ,Cancer research ,ARGININE METHYLTRANSFERASE PRMT5 ,Signal transduction ,business ,Life Sciences & Biomedicine ,Ex vivo - Abstract
The protein arginine methyltransferase 5 (PRMT5) methylates a variety of proteins involved in splicing, multiple signal transduction pathways, epigenetic control of gene expression, and mechanisms leading to protein expression required for cellular proliferation. Dysregulation of PRMT5 is associated with clinical features of several cancers, including lymphomas, lung cancer, and breast cancer. Here, we describe the characterization of JNJ-64619178, a novel, selective, and potent PRMT5 inhibitor, currently in clinical trials for patients with advanced solid tumors, non-Hodgkin's lymphoma, and lower-risk myelodysplastic syndrome. JNJ-64619178 demonstrated a prolonged inhibition of PRMT5 and potent antiproliferative activity in subsets of cancer cell lines derived from various histologies, including lung, breast, pancreatic, and hematological malignancies. In primary acute myelogenous leukemia samples, the presence of splicing factor mutations correlated with a higher ex vivo sensitivity to JNJ-64619178. Furthermore, the potent and unique mechanism of inhibition of JNJ-64619178, combined with highly optimized pharmacological properties, led to efficient tumor growth inhibition and regression in several xenograft models in vivo, with once-daily or intermittent oral-dosing schedules. An increase in splicing burden was observed upon JNJ-64619178 treatment. Overall, these observations support the continued clinical evaluation of JNJ-64619178 in patients with aberrant PRMT5 activity-driven tumors. ispartof: MOLECULAR CANCER THERAPEUTICS vol:20 issue:12 pages:2317-2328 ispartof: location:United States status: published
- Published
- 2021
22. U2AF1is a haplo-essential gene required for cancer cell survival
- Author
-
Michael O. Alberti, Ajay Khanna, Cara Lunn Shirai, Christopher A. Miller, Sarah Grieb, Dennis L. Fei, Jin Shao, Matthew J. Walter, Tanzir Ahmed, Matthew Ndonwi, Timothy A. Graubert, Brian A. Wadugu, Sridhar Nonavinkere Srivatsan, Amanda Heard, and Joseph Bradley
- Subjects
Haematopoiesis ,Leukemia ,Essential gene ,Somatic cell ,Cancer cell ,Mutant ,medicine ,Allele ,Biology ,medicine.disease ,Gene ,Molecular biology - Abstract
Somatic mutations in the spliceosome geneU2AF1are common in patients with myelodysplastic syndromes.U2AF1mutations that code for the most common amino acid substitutions are always heterozygous, and the retained wild-type allele is expressed, suggesting that mutant hematopoietic cells may require the residual wild-type allele to be viable and cause disease. We show that hematopoiesis and RNA splicing inU2af1heterozygous knock-out mice was similar to control mice, but that deletion of the wild-type allele in U2AF1(S34F) heterozygous mutant expressing hematopoietic cells (i.e., hemizygous mutant) was lethal. These results confirm that U2AF1 mutant hematopoietic cells are dependent on the expression of wild-type U2AF1 for survivalin vivoand thatU2AF1is a haplo-essential cancer gene. Mutant U2AF1 (S34F) expressing cells were also more sensitive to reduced, but not absent, expression of wild-type U2AF1 than non-mutant cells. Furthermore, mice transplanted with leukemia cells expressing mutant U2AF1 had significantly reduced tumor burden and improved survival after the wild-typeU2af1allele was deleted compared to when it was not deleted. These results suggest that selectively targeting the wild-typeU2AF1allele in heterozygous mutant cells could induce cancer cell death and be a therapeutic strategy for patients harboringU2AF1mutations.
- Published
- 2020
23. SF3B1-mutant MDS as a distinct disease subtype: a proposal from the International Working Group for the Prognosis of MDS
- Author
-
Jacqueline Boultwood, Manja Meggendorfer, Seishi Ogawa, Donna Neuberg, Luca Malcovati, David T. Bowen, Felicitas Thol, Michael Heuser, Elli Papaemmanuil, David P. Steensma, Michaela Fontenay, Kristen E. Stevenson, Mario Cazzola, David A. Sallman, Heinz Tüchler, Mikkael A. Sekeres, Michael R. Savona, Rami S. Komrokji, Detlef Haase, Sudhir Tauro, Andrea Pellagatti, Torsten Haferlach, Peter J. Campbell, Pierre Fenaux, Rafael Bejar, Benjamin L. Ebert, Paresh Vyas, Matthew J. Walter, Joop H. Jansen, Aly Karsan, Timothy A. Graubert, Peter L. Greenberg, Jaroslaw P. Maciejewski, Arjan A. van de Loosdrecht, Eva Hellström-Lindberg, Guillermo Garcia-Manero, Hematology, and CCA - Cancer biology and immunology
- Subjects
Ineffective erythropoiesis ,Oncology ,medicine.medical_specialty ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Immunology ,Plenary Paper ,medicine.disease_cause ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,All institutes and research themes of the Radboud University Medical Center ,Internal medicine ,Molecular genetics ,hemic and lymphatic diseases ,medicine ,Humans ,030304 developmental biology ,0303 health sciences ,Cytopenia ,business.industry ,Leukemia, Myelomonocytic, Chronic ,Cell Biology ,Hematology ,medicine.disease ,Phosphoproteins ,Prognosis ,3. Good health ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Dysplasia ,030220 oncology & carcinogenesis ,Concomitant ,Mutation (genetic algorithm) ,Erythropoiesis ,Bone marrow ,RNA Splicing Factors ,business - Abstract
The 2016 revision of the World Health Organization classification of tumors of hematopoietic and lymphoid tissues is characterized by a closer integration of morphology and molecular genetics. Notwithstanding, the myelodysplastic syndrome (MDS) with isolated del(5q) remains so far the only MDS subtype defined by a genetic abnormality. Approximately half of MDS patients carry somatic mutations in spliceosome genes, with SF3B1 being the most commonly mutated one. SF3B1 mutation identifies a condition characterized by ring sideroblasts (RS), ineffective erythropoiesis, and indolent clinical course. A large body of evidence supports recognition of SF3B1-mutant MDS as a distinct nosologic entity. To further validate this notion, we interrogated the data set of the International Working Group for the Prognosis of MDS (IWG-PM). Based on the findings of our analyses, we propose the following diagnostic criteria for SF3B1-mutant MDS: (1) cytopenia as defined by standard hematologic values, (2) somatic SF3B1 mutation, (3) morphologic dysplasia (with or without RS), and (4) bone marrow blasts
- Published
- 2020
24. Isocitrate dehydrogenase 1 and 2 mutations, 2‐hydroxyglutarate levels, and response to standard chemotherapy for patients with newly diagnosed acute myeloid leukemia
- Author
-
Yi-Bin Chen, Gabriela S. Hobbs, Meghan Burke, Philip C. Amrein, Seth A. Wander, Ilene Galinsky, Donna Neuberg, Christelle Joseph, Darrell R. Borger, Kimberly Straley, Aura Y. Ramos, Richard Stone, Hua Yang, Eyal C. Attar, Karen K. Ballen, Hossein Sadrzadeh, Sam Agresta, Ashley M. Perry, Andrew M. Brunner, Katharine E. Yen, Timothy A. Graubert, Anthony J. Iafrate, Amir T. Fathi, Christine Connolly, and Sophia Adamia
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,IDH1 ,medicine.medical_treatment ,Gastroenterology ,IDH2 ,Glutarates ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,medicine ,Humans ,Prospective Studies ,030212 general & internal medicine ,Aged ,Aged, 80 and over ,Chemotherapy ,business.industry ,Proportional hazards model ,Myeloid leukemia ,Middle Aged ,Prognosis ,Isocitrate Dehydrogenase ,Survival Rate ,Leukemia, Myeloid, Acute ,Exact test ,medicine.anatomical_structure ,Isocitrate dehydrogenase ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Female ,Bone marrow ,business ,Follow-Up Studies - Abstract
Background Acute myeloid leukemia (AML) cells harboring mutations in isocitrate dehydrogenase 1 (IDH1) and isocitrate dehydrogenase 2 (IDH2) produce the oncometabolite 2-hydroxyglutarate (2HG). This study prospectively evaluated the 2HG levels, IDH1/2 mutational status, and outcomes of patients receiving standard chemotherapy for newly diagnosed AML. Methods Serial samples of serum, urine, and bone marrow aspirates were collected from patients newly diagnosed with AML, and 2HG levels were measured with mass spectrometry. Patients with baseline serum 2HG levels greater than 1000 ng/mL or marrow pellet 2HG levels greater than 1000 ng/2 × 106 cells, which suggested the presence of an IDH1/2 mutation, underwent serial testing. IDH1/2 mutations and estimated variant allele frequencies were identified. AML characteristics were compared with the Wilcoxon test and Fisher's exact test. Disease-free survival and overall survival (OS) were evaluated with log-rank tests and Cox regression. Results Two hundred and two patients were treated for AML; 51 harbored IDH1/2 mutations. IDH1/2-mutated patients had significantly higher 2HG levels in serum, urine, bone marrow aspirates, and aspirate cell pellets than wild-type patients. A serum 2HG level greater than 534.5 ng/mL was 98.8% specific for the presence of an IDH1/2 mutation. Patients with IDH1/2-mutated AML treated with 7+3-based induction had a 2-year event-free survival (EFS) rate of 44% and a 2-year OS rate of 57%. There was no difference in complete remission rates, EFS, or OS between IDH1/2-mutated and wild-type patients. Decreased serum 2HG levels on day 14 as a proportion of the baseline were significantly associated with improvements in EFS (P = .047) and OS (P = .019) in a multivariate analysis. Conclusions Among patients with IDH1/2-mutated AML, 2HG levels are highly specific for the mutational status at diagnosis, and they have prognostic relevance in patients receiving standard chemotherapy.
- Published
- 2018
25. Impact of lenalidomide use among non-transfusion dependent patients with myelodysplastic syndromes
- Author
-
Gregory A. Abel, Richard Stone, Amir T. Fathi, Anand R. Habib, Shicheng Weng, Angel M. Cronin, Timothy A. Graubert, Andrew M. Brunner, and David P. Steensma
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Blood transfusion ,medicine.medical_treatment ,Disease ,Off-label use ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Blood Transfusion ,Lenalidomide ,Survival analysis ,Aged ,Aged, 80 and over ,business.industry ,Myelodysplastic syndromes ,Off-Label Use ,Hematology ,medicine.disease ,Survival Analysis ,Hematologic Response ,030104 developmental biology ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Transfusion dependence ,Female ,business ,medicine.drug - Abstract
Chemotherapies approved for defined subgroups promise personalized oncologic care, but their off-label impact is unclear. Lenalidomide is approved for lower-risk, transfusion-dependent (TD) myelodysplastic syndromes (MDS) with del(5q), but frequently used in MDS outside this indication. We characterized lenalidomide use and outcomes among non-TD patients with MDS. Patients 65 or older diagnosed with MDS between 2007 and 2013 were identified using SEER; linked Medicare claims were evaluated for transfusions, lenalidomide use, and incident toxicities. TD was ≥2 transfusion episodes within an 8-week period; responses were transfusion independence (TI) and ≥50% transfusion reduction (minor response). We compared overall survival for non-TD patients receiving lenalidomide versus those not receiving lenalidomide, matched on disease and patient characteristics. We identified 676 patients who had received lenalidomide, including 275 (40.7%) TD and 401 (59.3%) non-TD; 18.5% (125/676) had zero claims for RBC transfusion prior to receiving lenalidomide. Incident toxicities among patients prescribed lenalidomide were similar in TD and non-TD groups, except incident thromboembolic events were higher among non-TD patients (10.8% vs. 6.0%, P = .04). Comparing 191 non-TD patients receiving lenalidomide within 6 months of MDS diagnosis to risk-matched MDS controls, lenalidomide was not associated with improved OS (P = .78). Among TD patients (n = 275), 31% achieved TI, and 30% achieved minor hematologic response, with a median time to TI of 4.1 weeks. In conclusion, we confirmed the benefit of lenalidomide among TD patients with MDS; however, many non-TD patients also received lenalidomide. These patients experienced accompanying toxicity without evidence of benefit in terms of transfusion needs or overall survival.
- Published
- 2018
26. High NPM1-mutant allele burden at diagnosis predicts unfavorable outcomes in de novo AML
- Author
-
Olga K. Weinberg, Richard Stone, Amir T. Fathi, Yi-Bin Chen, Robert P. Hasserjian, Edwin P. Alyea, Sanjay S. Patel, Robert J. Soiffer, Daniel J. DeAngelo, Frank C. Kuo, David P. Steensma, Martha Wadleigh, Valentina Nardi, Andrew M. Brunner, Christopher J. Gibson, and Timothy A. Graubert
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,NPM1 ,Myeloid ,Adolescent ,Immunology ,Biochemistry ,Disease-Free Survival ,DNA Methyltransferase 3A ,Mutation Accumulation ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,Internal medicine ,medicine ,Humans ,DNA (Cytosine-5-)-Methyltransferases ,Allele ,Allele frequency ,Survival analysis ,Aged ,Retrospective Studies ,Aged, 80 and over ,Myeloid Neoplasia ,business.industry ,Nuclear Proteins ,Cell Biology ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Acute Myeloid Leukemia with Mutated NPM1 ,Survival Analysis ,Transplantation ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Female ,business ,Nucleophosmin ,Stem Cell Transplantation ,030215 immunology - Abstract
Acute myeloid leukemia (AML) with mutated NPM1 is a newly recognized separate entity in the revised 2016 World Health Organization classification and is associated with a favorable prognosis. Although previous studies have evaluated NPM1 in a binary fashion, little is known about the significance of its mutant allele burden at diagnosis, nor has the effect of comutations (other than FLT3) been extensively evaluated. We retrospectively used targeted sequencing data from 109 patients with de novo AML with mutated NPM1 to evaluate the potential significance of NPM1 variant allele frequency (VAF), comutations, and clinical parameters with regard to patient outcomes. We observed that high NPM1 VAF (uppermost quartile) correlated with shortened overall survival (median, 12.1 months vs not reached; P < .0001) as well as event-free survival (median, 7.5 vs 65.44 months; P < .0001) compared with the other NPM1-mutated cases. In both univariate and multivariable analyses, high NPM1 VAF had a particularly adverse prognostic effect in the subset of patients treated with stem-cell transplantation in first remission (P = .0004) and in patients with mutated DNMT3A (P < .0001). Our findings indicate that the prognostic effect of NPM1 mutation in de novo AML may be influenced by the relative abundance of the mutated allele.
- Published
- 2018
27. ATR/CHK1/WEE1 Dependency in SRSF2-Mutated MDS/AML
- Author
-
Timothy A. Graubert, Samuli Eldfors, Kimmo Porkka, Vineet Sharma, Angelique N Gilbert, and Sumit Rai
- Subjects
0303 health sciences ,Dependency (UML) ,biology ,Chemistry ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,3. Good health ,03 medical and health sciences ,Wee1 ,0302 clinical medicine ,biology.protein ,Cancer research ,030304 developmental biology ,030215 immunology - Abstract
Background: Mutations in splicing factor gene SRSF2 are recurrent drivers in 5-15 % of patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). A key property of SRSF2 mutations is that they occur early in the pathogenesis of MDS and are therefore present in all tumor cells in a patient. This property makes targetable vulnerabilities caused by SRSF2 mutations exceptionally important as they provide a way to inhibit the whole tumor. We previously demonstrated that splicing factor mutations induce R-loop-dependent activation of ATR, rendering cells sensitive to ATR inhibition. R-loops are transcription intermediates consisting of an RNA:DNA hybrid and a displaced single-stranded DNA. Accumulation of aberrant R-loops induces the ATR kinase, which activates the G2-M cell cycle checkpoint via CHK1- and WEE1-mediated signaling. In normal cells, activation of the G2-M checkpoint halts the cell cycle until R-loops have been resolved. When the ATR pathway is inhibited, checkpoint activation does not occur, causing cells with unresolved R-loops to proceed to mitosis, resulting in DNA damage and cell death. We, therefore, sought to assess primary human MDS/AML samples for sensitivity to perturbation of the ATR/CHK1/WEE1 pathway and identify mechanisms of resistance. Methods: Sensitivity of 147 AML patient samples to 515 oncology drugs was tested ex vivo. Bone marrow mononuclear cells were incubated with 5 concentrations of each drug for 72 h followed by measurement of cell viability by CTG assay. Somatic mutations were identified by exome sequencing of matched leukemic bone marrow and skin biopsy samples. Isogenic K562 cell line clones carrying SRSF2 P95H/L/R mutations were generated using CRISPR/Cas9 editing. The presence of SRSF2 mutation was confirmed by CRISPR-sequencing and expression by whole transcriptome RNA-sequencing. Drug sensitivity of the K562 clones with and without SRSF2 mutation was determined by incubating cells with 16 concentrations of prexasertib, SRA-737, adavosertib, or BAY-1895344, followed by determination of cell viability by the MTS assay. Results: Analysis of ex vivo drug sensitivities in AML patient samples identified vulnerability to CHK1 and WEE1 inhibition in SRSF2-mutated AML: SRSF2 mutation is associated with sensitivity to the CHK1 inhibitors prexasertib (p = 0.006) (Fig 1 A and B) and PF-00477736 (p = 0.002) and the WEE1 inhibitor adavosertib (p = 0.003). To establish whether the isogenic SRSF2-mutant K562 cell line models recapitulate known downstream aberrations associated with SRSF2 mutations in patients, we analyzed gene expression and splicing. SRSF2 contains an RNA binding domain with affinity to CCNG or GGNG exonic splicing enhancer sequences. Similar to what has been observed in patients, the K562 clones with SRSF2 mutation show reduced use of GGNG sequence motifs at skipped exons. These results demonstrate that isogenic K562 clones recapitulate known alterations caused by mutant SRSF2. To determine whether SRSF2 mutations induce sensitivity to inhibition of ATR, CHK1, and WEE1, we tested 10 isogenic SRSF2 mutant and 4 wild-type K562 clones. Cells with SRSF2 mutation show increased sensitivity to ATR/CHK1/WEE1 inhibition (Fig 1C). We found no significant difference in drug sensitivity between clones carrying SRSF2 P95H/L/R substitutions. Clones with higher SRSF2 mutant allele dosage are more sensitive (Fig 1D). We identified a subset of SRSF2 mutated AML samples that were resistant to CHK1 and WEE1 inhibition. All resistant AML have co-occurring RUNX1 mutations (Fig 1B). In AML, RUNX1 mutations are associated with therapy resistance, suggesting that these mutations contribute to drug resistance. To test whether RUNX1 mutations induce resistance to ATR/CHK1/WEE1 inhibition in SRSF2-mutant leukemia, we introduced RUNX1 loss-of-function mutations in isogenic K562 carrying SRSF2 mutations. Candidate resistance factors identified by ATAC and RNA-sequencing will be validated in functional assays. Conclusions: Our results indicate that SRSF2-mutated leukemia harbor a vulnerability to the inhibition of ATR, CHK1, and WEE1 kinases. Cell line models indicate that sensitivity is similar across mutant alleles and dependent on allelic copy number. Several ATR/CHK1 and WEE1 inhibitors are in development, and our results suggest that these compounds could be effective treatments for SRSF2-mutated MDS and AML. Figure 1 Figure 1. Disclosures Graubert: astrazeneca: Research Funding; Janssen: Research Funding; Calico: Research Funding.
- Published
- 2021
28. Spliceosome Mutant Myeloid Malignancies Are Preferentially Sensitive to PARP Inhibition
- Author
-
Maxwell Bannister, Victor M. Corral, Cassandra Leibson, Zhiyan Silvia Liu, Axia Song, Timothy A. Graubert, Sayantani Sinha, Erica Arriaga-Gomez, Sumit Rai, Dang Hai Nguyen, Dawei Zong, and Stanley Chun-Wei Lee
- Subjects
Spliceosome ,Myeloid ,medicine.anatomical_structure ,Chemistry ,Poly ADP ribose polymerase ,Immunology ,Mutant ,Cancer research ,medicine ,Cell Biology ,Hematology ,Biochemistry - Abstract
Somatic heterozygous mutations in spliceosome genes SRSF2, U2AF1, and SF3B1 commonly occur in patients with myeloid malignancies such as myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Moreover, SRSF2 and U2AF1 mutations associate with poor survival and high risk of progression to AML, representing a unique genetic vulnerability for targeted therapy. We and others previously found that R-loops, a group of transcription intermediates containing RNA:DNA hybrids and displaced single-stranded DNA, are a source of genomic instability induced by different spliceosome mutants. We further showed that inhibition of ATR kinase activity preferentially kills spliceosome mutant cells in an R-loop-dependent manner. Inspired by ATR inhibition results, we performed a focused drug screen with inhibitors targeting additional DNA damage response pathways to identify novel therapeutic vulnerabilities generated by spliceosome mutations. We generated a murine leukemia model by overexpressing the MLL-AF9 fusion oncogene on an Srsf2 P95H/+background, a mutational combination that is found in ~10% of MLL-rearranged leukemias. Surprisingly, we found that Srsf2 P95H/+cells are more sensitive to five inhibitors targeting ADP-ribosyltransferases or PARP (olaparib, talazoparib, rucaparib, niraparib, veliparib) (Figs 1A-B). Olaparib (PARPi)-treated Srsf2 P95H/+cells exhibited increased apoptosis compared to Srsf2 +/+ cells as determined by AnnexinV (Fig 1C). PARPi sensitivity was also observed in isogenic murine MLL-AF9 U2af1 S34F/+cells compared to MLL-AF9 U2af1 +/+ cells (Fig 1D). These data highlight that both SRSF2 P95H and U2AF1 S34F mutations create a common vulnerability that is dependent on PARP activity for survival. To evaluate PARP activity in cells, we used isogenic K562 leukemia cells expressing SRSF2 P95H and U2AF1 S34F mutations from their endogenous loci and monitored PAR (poly(ADP-ribose)) chain levels, a marker of PARP activity. Both SRSF2 P95H and U2AF1 S34F cells exhibited elevated PAR levels compared to wildtype cells (Figs 1E-F). PARPi treatment significantly suppressed PAR signals in SRSF2 P95H and U2AF1 S34F cells. PARP inhibitors target both PARP1 and PARP2 enzymes, of which PARP1 plays a key role in DNA damage response. We used CRISPR-Cas9 to knockout PARP1 gene to determine the major PARP responsible for elevated PAR level in these leukemia cells. PARP1 deletion abrogated elevated PAR levels in U2AF1 S34F (Fig 1G) and SRSF2 P95H cells (data not shown). Altogether, we demonstrated that SRSF2 P95H and U2AF1 S34F cells trigger a PARP1 response critical for cell survival. To test whether increased PAR level arises from U2AF1 S34F-induced R-loops, we generated U2AF1 S34F cells that inducibly express RNaseH1, an enzyme that specifically cleaves the RNA moiety within RNA:DNA hybrids. Induction of RNaseH1 in U2AF1 S34F cells significantly reduced PAR levels, showing that U2AF1 S34F-induced PAR chains is R-loop-dependent (Fig 1H). Moreover, RNaseH1 overexpression suppressed the growth inhibition of PARPi-treated U2AF1 S34F cells (Fig 1I). Collectively, these results suggest that U2AF1 S34F mutants induce R-loop accumulation and elicit an R-loop-associated PARP1 signaling to promote cell survival. We next tested whether combining ATR inhibitor (ATRi) can further exacerbate PARPi sensitivity in spliceosome mutant cells. To examine ATR activity, we monitored phosphorylated RPA (Replication Protein A, or pRPA), a known ATR substrate. pRPA level was enhanced in PARPi-treated SRSF2 P95H cells compared to PARPi-treated SRSF2 WT cells but was suppressed when treated with ATRi (Fig 1J), suggesting that splicing factor mutant cells are more reliant on ATR function in the context of PARPi. Importantly, the combination of PARPi with ATRi, but not with ATMi, significantly promoted cell growth inhibition in SRSF2 P95H cells compared to SRSF2 WT cells or to SRSF2 P95H cells treated with individual compounds alone (Fig 1K). Collectively, these data provide a pre-clinical rationale that splicing factor mutant leukemias are preferentially sensitive to PARP1 modulation compared to their wildtype counterpart. Moreover, combining PARPi and ATRi may further sensitize spliceosome mutant cells and could represent a new therapeutic strategy in myeloid leukemia patients harboring these mutations (Fig 1L). Figure 1 Figure 1. Disclosures Graubert: Calico: Research Funding; Janssen: Research Funding; astrazeneca: Research Funding.
- Published
- 2021
29. Inhibition of ATR with AZD6738 (Ceralasertib) for the Treatment of Progressive or Relapsed Myelodysplastic Syndromes and Chronic Myelomonocytic Leukemia: Safety and Preliminary Activity from a Phase Ib/II Study
- Author
-
Timothy A. Graubert, Lourdes M. Mendez, Meagan A. Jacoby, Andrew M. Brunner, Richard Stone, Donna Neuberg, Yiwen Liu, Amir T. Fathi, Philip C. Amrein, Jacqueline S. Garcia, Simon Smith, Emma Dean, and Matthew J. Walter
- Subjects
business.industry ,Myelodysplastic syndromes ,Immunology ,Cancer research ,Medicine ,Chronic myelomonocytic leukemia ,Cell Biology ,Hematology ,business ,medicine.disease ,Biochemistry - Abstract
Introduction: Myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML) are advanced myeloid neoplasms with poor survival and no curative therapy except allogeneic transplant; they are enriched for mutations in the spliceosome complex, including the genes SF3B1, SRSF2, ZRSR2, and U2AF1. Ceralasertib (AZD6738) is a selective and potent orally bioavailable inhibitor of Ataxia Telangiectasis and Rad3 Related (ATR) kinase. Expression of mutant spliceosome genes induces alternative splicing and R loops (DNA:RNA hybrids) in human hematopoietic cells. ATR is a critical mediator for R loop resolution; if ATR is inhibited, cells that have excess R loops undergo apoptosis, whereas normal cells are spared. We hypothesized that inhibition of ATR may be useful in patients with MDS or CMML, particularly those who harbor splicing factor gene mutations. Methods: We designed a 2 part study to evaluate ceralasertib monotherapy in adult patients with MDS or CMML progressing on, not responsive to, or ineligible to receive front-line therapy. Enrollment cohorts included SF mut (SF3B1, SRSF2, U2AF1, or ZRSR2) and SF wt. Part 1 was dose finding, enrolling up to 6 patients at de-escalating dose levels starting at the tolerable dose in solid tumors: 160mg BID days 1-14 of 28 d cycles, 120mg BID d1-14, or 80mg BID d1-14. Higher-risk MDS (IPSS-R > 3.5) and CMML patients were enrolled first, and once a dose was deemed safe in this group, patients with lower-risk MDS (IPSS-R 3.5, TD post ESA or severe neutropenia/thrombocytopenia) were evaluated for safety starting at that dose. Part 2 of the study is ongoing and evaluating a total of up to 20 SF mut patients and 20 SF wt patients for efficacy. A Simon 2-stage design mandated that at least 1 of the first 10 patients in each group needed to respond (IWG) to enroll the final 10 patients. We report here the toxicity and preliminary response data. Results: At the data cut-off (7/15/21) a total of 31 patients have been enrolled on study with 29 evaluable: 21 with higher-risk MDS or CMML, and 8 with lower-risk MDS. 2 higher-risk MDS patients were not evaluable for toxicity or response and replaced; one did not start the study drug, and one patient mistakenly took a home chemotherapy concurrently. The median age was 73 (range 43-88) and the cohort was predominantly male (26/29). Baseline blood counts evaluable for hematologic lineage response: 16/27 had ANC < 1000 (median ANC 700, range 100-7760), 27/29 had Hgb < 11 (median 8.5, range 5.9-12.4), and 17/29 had platelets < 100 (median 66, range 11-415). The most frequently seen ≥ grade 3 and higher all cause adverse events (AEs) for higher-risk patients (n=20) included anemia (n=6), febrile neutropenia (n=4), neutropenia (n=5), thrombocytopenia (n=6). In lower-risk MDS (n=8), ≥ grade 3 AEs included thrombocytopenia (n=2), and neutropenia (n=2). The most frequent AEs possibly related to therapy were cytopenias (Table 1). Blood counts typically declined during treatment with recovery or improvement at the end of the 28d cycle. 4 patients had dose modifications to treat with ceralasertib on days 1-10 of 28, with improved blood parameters. 5 deaths occurred within 30d of study treatment; 1 patient had an intracranial bleed with thrombocytopenia and active CMML, possibly related to study treatment, while 4 deaths were related to underlying disease (3 with AML, 1 with sepsis and MDS). No DLTs were observed during the DLT period and the starting dose of 160mg BID days 1-14 of 28 was deemed the RP2D. The Simon 2-stage design assessed responses after the first 10 SF mut patients were treated (n=8 higher-risk, n=2 lower-risk MDS). Three patients had responses as per IWG criteria (Fig 1), including one patient each showing improvement in neutrophil count, hemoglobin improvement, and one patient with blast decrease and blood counts that met CR. In addition, one patient had evidence for clinical activity with platelet count improvement. Out of 7 SF wt patients evaluable for response, no responses have been observed. Study enrollment and follow-up continues in both cohorts. Discussion: Ceralasertib is an oral ATR inhibitor which can be safely administered to patients with progressive/refractory MDS and CMML at doses of 160mg BID on days 1-14 of a 28 day cycle; toxicity was manageable with decreased dosing. The study is ongoing, with preliminary evidence of activity in splicing factor mutated disease that may permit further development of mutation-directed therapy. Figure 1 Figure 1. Disclosures Brunner: Keros Therapeutics: Consultancy; AstraZeneca: Research Funding; GSK: Research Funding; Aprea: Research Funding; Agios: Consultancy; Acceleron: Consultancy; Janssen: Research Funding; Takeda: Consultancy, Research Funding; BMS/Celgene: Consultancy, Research Funding; Novartis: Consultancy, Research Funding. Garcia: Prelude: Research Funding; Pfizer: Research Funding; Genentech: Research Funding; Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astellas: Consultancy, Membership on an entity's Board of Directors or advisory committees; AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; AstraZeneca: Research Funding. Neuberg: Madrigal Pharmaceuticals: Other: Stock ownership; Pharmacyclics: Research Funding. Dean: AstraZeneca: Current Employment. Smith: AstraZeneca: Current Employment. Stone: Arog: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Astellas: Membership on an entity's Board of Directors or advisory committees; Onconova: Consultancy; Gemoab: Membership on an entity's Board of Directors or advisory committees; Innate: Consultancy; Janssen: Consultancy; Foghorn Therapeutics: Consultancy; AbbVie: Consultancy; Actinium: Membership on an entity's Board of Directors or advisory committees; Aprea: Consultancy; Amgen: Membership on an entity's Board of Directors or advisory committees; Jazz: Consultancy; Syndax: Membership on an entity's Board of Directors or advisory committees; Syntrix/ACI: Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Consultancy; Boston Pharmaceuticals: Consultancy; GlaxoSmithKline: Consultancy; Elevate Bio: Membership on an entity's Board of Directors or advisory committees; BerGen Bio: Membership on an entity's Board of Directors or advisory committees; Syros: Membership on an entity's Board of Directors or advisory committees; Takeda: Consultancy; Agios: Consultancy, Research Funding; Celgene: Consultancy; Macrogenics: Consultancy. Fathi: Foghorn: Consultancy, Honoraria; Kite: Consultancy, Honoraria; Morphosys: Consultancy, Honoraria; Ipsen: Consultancy, Honoraria; Kura: Consultancy, Honoraria; Trillium: Consultancy, Honoraria; Genentech: Consultancy, Honoraria; Daiichi Sankyo: Consultancy, Honoraria; Astellas: Consultancy, Honoraria; Seattle Genetics: Consultancy, Honoraria; Blueprint: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Celgene/BMS: Consultancy, Honoraria, Research Funding; Servier: Research Funding; Agios: Consultancy, Honoraria, Research Funding; AbbVie: Consultancy, Honoraria, Research Funding. Graubert: Janssen: Research Funding; astrazeneca: Research Funding; Calico: Research Funding. OffLabel Disclosure: AZD6738 is being investigated in a number of malignancies including MDS and CMML
- Published
- 2021
30. Combined Targeted Therapy for BRAF-Mutant, Treatment-Related Acute Myeloid Leukemia
- Author
-
Robert P. Hasserjian, Amir T. Fathi, Yi-Bin Chen, Valentina Nardi, Gregory M. Cote, Andrew M. Brunner, Timothy A. Graubert, Kwadwo A. Oduro, Seth A. Wander, and Krzysztof Glomski
- Subjects
0301 basic medicine ,Trametinib ,Cancer Research ,Cardiotoxicity ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Myeloid leukemia ,Decitabine ,Dabrafenib ,Targeted therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,medicine ,Cancer research ,Cytarabine ,business ,neoplasms ,medicine.drug - Abstract
A 44-year-old woman with a prior history of myxoid liposarcoma who was previously treated with radiation, chemotherapy, and resection, was admitted with syncope and pancytopenia. She was diagnosed with a high-grade therapy-related myelodysplastic syndrome and treated with decitabine. Her disease soon progressed to overt acute myeloid leukemia (AML). She was treated with cytotoxic chemotherapy including cytarabine and topotecan, but she was not a candidate for further anthracycline exposure given her prior treatment for sarcoma and concern for cardiotoxicity. Her AML was refractory to two sequential induction regimens. Given that targeted genomic sequencing had revealed a BRAF V600E mutation with a high allelic fraction, she was then placed on combined targeted BRAF/MEK therapy with dabrafenib and trametinib for her refractory disease. This resulted in a dramatic response with clearance of circulating myeloblasts, restoration of normal hematopoiesis, a significant decrease in marrow leukemic burden, and a concordant decrease in the BRAF V600E allelic burden. The response was transient, however, with a rapid increase in circulating blasts a few weeks later. At the time of subsequent progression, four separate KRAS mutations were identified. She died approximately 4 months after her diagnosis from rapidly progressive AML.
- Published
- 2017
31. Phase I Study of Ixazomib Added to Chemotherapy in the Treatment of Acute Lymphoblastic Leukemia in Older Adults
- Author
-
Philip C. Amrein, Karen K. Ballen, Kristen E. Stevenson, Traci M. Blonquist, Andrew M. Brunner, Gabriela S. Hobbs, Hanno R. Hock, Steven L. McAfee, Jenna A. Moran, Meghan Bergeron, Julia E. Foster, Christina Bertoli, Kristin McGregor, Molly Macrae, Meghan Burke, Tanya T. Behnan, Tina T. Som, Aura Y. Ramos, Megan K. Vartanian, Jennifer Lombardi Story, Christine Connolly, Timothy A. Graubert, Donna S. Neuberg, and Amir T. Fathi
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Introduction: While progress has been made in the treatment of childhood leukemia, the outlook for patients >60 years of age with acute lymphoblastic leukemia (ALL) is poor with complete remission rates (CR) of approximately 60% and 3-year survivals (OS) of less than 15%. Intensified treatment in a later CALGB trial showed little improvement with a CR=61% and 5-year OS=6% (Stock, Cancer 2013). Ixazomib is an oral proteasome inhibitor, which has shown single agent activity and promising combination activity in pediatric ALL patients (Messinger, Blood 2012). We sought to assess the safety and tolerability, as well as early efficacy of adding ixazomib to a current MGH-DFCI/HCC multi-agent regimen for older adults with ALL. Methods: Patients aged 51 to 75 years of age with newly diagnosed B-ALL and T-ALL were screened for eligibility. Patients with mature ALL (including Burkitt's) were excluded. Patients with Philadelphia chromosome positive ALL (BCR-ABL1+) were eligible, and dasatinib was added to the chemotherapy on Day 10 for these patients. The chemotherapy treatment schedule from induction through maintenance is outlined in Table 1. A standard 3 + 3 patient cohort dose escalation design was used to determine the maximum tolerated dose (MTD) of ixazomib during induction for these patients, the primary objective of the trial. After consolidation I, patients in complete remission (CR) with a suitable donor were offered a hematopoietic stem cell transplantation (HSCT) as per institutional guidelines. Those not going to HSCT continued therapy as noted in the table. Results: There were 19 patients with B-ALL enrolled, none with T-ALL. Among these patients, 7 harbored BCR-ABL1 rearrangements. The median age was 65 years, 74% were male, and 90% had a performance status 0 or 1. The MTD was 2.3 mg of ixazomib, as 2 patients at 3.0 mg developed DLT's: a grade 3 peripheral neuropathy and a grade 5 acute kidney injury (Table 2). Grade 3 and 4 toxicities encountered at any time consisted mainly of grade 4 neutropenia in 13 patients and grade 4 thrombocytopenia in 12 patients. One patient experienced grade 3 neutropenia and 5 patients experienced grade 3 thrombocytopenia. Two patients with grade 2 neuropathy did not meet the definition of DLT. Among the 19 patients, 15 (79%, [95% confidence interval (CI), 54-94%]) achieved CR (14) or CRi (1), and 5 patients went on to HSCT. The median follow-up time was 2 years (range, 1-5) for 8 patients remaining alive. The 1-year overall survival estimate was 53% [95% CI, 29-72%], while the 2-year overall survival estimate was 47% [95% CI, 24-67%]. Conclusions: A dose of 2.3 mg of ixazomib in combination with induction chemotherapy among older patients with ALL was well-tolerated and associated with a promising rate of complete remission. Disclosures Amrein: Takeda: Research Funding; AstraZeneca: Consultancy, Research Funding; Amgen: Research Funding. Brunner:Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Research Funding; AstraZeneca: Research Funding; Forty-Seven Inc: Membership on an entity's Board of Directors or advisory committees; Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Novartis: Research Funding. Hobbs:Novartis: Honoraria; Celgene/BMS: Honoraria; Jazz: Honoraria; Constellation: Honoraria, Research Funding; Incyte: Research Funding; Merck: Research Funding; Bayer: Research Funding. Neuberg:Celgene: Research Funding; Pharmacyclics: Research Funding; Madrigak Pharmaceuticals: Current equity holder in publicly-traded company. Fathi:Takeda: Consultancy, Research Funding; Agios: Consultancy, Research Funding; PTC Therapeutics: Consultancy; Amphivena: Consultancy; Astellas: Consultancy; Daiichi Sankyo: Consultancy; Novartis: Consultancy; Newlink Genetics: Consultancy; Pfizer: Consultancy; Blueprint: Consultancy; Trillium: Consultancy; Kura Oncology: Consultancy; Forty Seven: Consultancy; Jazz: Consultancy; Boston Biomedical: Consultancy; BMS/Celgene: Consultancy, Research Funding; Kite: Consultancy; Trovagene: Consultancy; Amgen: Consultancy; Seattle Genetics: Consultancy, Research Funding; Abbvie: Consultancy. OffLabel Disclosure: MLN 9708, ixazomib is FDA approved for multiple myeloma. In this trial it is used to treat acute lymphoblastic leukemia.
- Published
- 2020
32. Case 37-2016
- Author
-
Naveen M. Kulkarni, Timothy A. Graubert, Robert P. Hasserjian, Amir T. Fathi, and Frank C. Kuo
- Subjects
medicine.medical_specialty ,Leukocytosis ,Neutrophils ,Sweating ,Diagnosis, Differential ,03 medical and health sciences ,0302 clinical medicine ,Monocytosis ,Bone Marrow ,Receptors, Colony-Stimulating Factor ,medicine ,Humans ,Fatigue ,Leukemia, Neutrophilic, Chronic ,Aged, 80 and over ,Blood Cells ,business.industry ,General Medicine ,medicine.disease ,Dermatology ,Neutrophilia ,Surgery ,030220 oncology & carcinogenesis ,Mutation ,Splenomegaly ,Female ,medicine.symptom ,business ,030215 immunology - Abstract
An 86-year-old woman was seen at this hospital because of fatigue, night sweats, leukocytosis, and splenomegaly. Review of the peripheral-blood smear revealed neutrophilia without dysplastic features, immature forms, or monocytosis. A diagnostic procedure was performed.
- Published
- 2016
33. Genomics in childhood acute myeloid leukemia comes of age
- Author
-
Andrew M. Brunner and Timothy A. Graubert
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Myeloid ,Group study ,business.industry ,Pediatric acute myeloid leukemia ,Childhood Acute Myeloid Leukemia ,Genomics ,General Medicine ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,hemic and lymphatic diseases ,Internal medicine ,medicine ,business ,neoplasms - Abstract
A Children's Oncology Group study of nearly 1,000 pediatric acute myeloid leukemia (AML) cases reveals marked differences between the genomic landscapes of pediatric and adult AML and offers directions for future work.
- Published
- 2018
34. Alisertib plus induction chemotherapy in previously untreated patients with high-risk, acute myeloid leukaemia: a single-arm, phase 2 trial
- Author
-
Gabriela S. Hobbs, Kristin McGregor, Donna Neuberg, R. Coleman Lindsley, Nicole Nelson, Daniel J. DeAngelo, Margaret C. Wey, Richard Stone, Kaitlyn M. Fishman, Philip C. Amrein, Jessica Rae, Jacqueline S. Garcia, Emma Logan, Traci M. Blonquist, Amity L. Manning, Myrna Nahas, Aura Y. Ramos, David P. Steensma, Lourdes M. Mendez, Meghan Bergeron, Tina T. Som, Meghan Burke, Jenna A. Moran, Yi Bin Chen, Julia Foster, Steven L. McAfee, Andrew M. Brunner, Molly Macrae, Hanno Hock, Eric S. Winer, Marlise R. Luskin, David Avigan, Karen K. Ballen, Michele Baltay, Jacalyn Rosenblatt, Nicole M. Hermance, Malgorzata McMasters, Timothy A. Graubert, Frank C. Kuo, Jennifer Lombardi Story, Amir T. Fathi, Geoffrey Fell, Christina Bertoli, Tanya T. Behnan, and Christine Connolly
- Subjects
Male ,medicine.medical_specialty ,Myeloid ,Neutropenia ,Gastroenterology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Risk Factors ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Idarubicin ,Humans ,Myeloproliferative neoplasm ,Aged ,business.industry ,Cytarabine ,Induction chemotherapy ,Hematology ,Azepines ,Induction Chemotherapy ,Middle Aged ,medicine.disease ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Pyrimidines ,chemistry ,030220 oncology & carcinogenesis ,Alisertib ,Female ,business ,Febrile neutropenia ,030215 immunology ,medicine.drug ,Follow-Up Studies - Abstract
Summary Background Increased aurora A kinase (AAK) expression occurs in acute myeloid leukaemia; AAK inhibition is a promising therapeutic target in this disease. We therefore aimed to assess the activity of alisertib combined with 7 + 3 induction chemotherapy in previously untreated patients with high-risk acute myeloid leukaemia. Methods We did a single-arm, phase 2 trial of patients recruited from the Dana–Farber/Harvard Cancer Center in the USA. Eligible patients had previously untreated acute myeloid leukaemia, an Eastern Cooperative Oncology Group performance status of 0–2, and were at high risk of disease as defined by the presence of an adverse-risk karyotype, the presence of secondary acute myeloid leukaemia arising from previous myelodysplastic syndrome or myeloproliferative neoplasm, the presence of therapy-related acute myeloid leukaemia, or being 65 years or older. Enrolled patients received 7 + 3 induction chemotherapy of continuous infusion of cytarabine (100 mg/m2 per day on days 1–7) and intravenous bolus of idarubicin (12 mg/m2 per day on days 1–3). Oral alisertib (30 mg) was given twice per day on days 8–15. Patients could receive up to four consolidation cycles with cytarabine and alisertib, and alisertib maintenance for 12 months. The primary endpoint was a composite including the proportion of patients achieving complete remission and those with a complete remission with incomplete neutrophil or platelet count recovery. Analyses were per-protocol. This study is registered with Clinicaltrials.gov , number NCT02560025 , and has completed enrolment. Findings Between Dec 31, 2015, and Aug 1, 2017, we enrolled a total of 39 eligible patients. 19 (49%) of 39 patients had secondary acute myeloid leukaemia and three (8%) had therapy-related acute myeloid leukaemia. At mid-induction, 33 (85%) of 39 patients showed marrow aplasia, six (15%) received re-induction. The median follow-up was 13·7 months (IQR 12·7–14·4). Composite remission was 64% (two-stage 95% CI 48–79), with 20 (51%) of 39 patients achieving complete remission and five (13%) achieving complete remission with incomplete neutrophil or platelet count recovery. The most common grade 3 or 4 adverse events included febrile neutropenia (16 [41%] of 39), neutropenia (12 [31%]), thrombocytopenia (13 [33%]), anaemia (11 [28%]), anorexia (nine [23%]), and oral mucositis (four [10%]). No treatment-related deaths were observed. Interpretation These results suggest that alisertib combined with induction chemotherapy is active and safe in previously untreated patients with high-risk acute myeloid leukaemia. This study met criteria to move forward to a future randomised trial. Funding Millennium Pharmaceuticals.
- Published
- 2019
35. Single-Cell RNA-Seq Reveals AML Hierarchies Relevant to Disease Progression and Immunity
- Author
-
Jason Stephansky, Timothy J. Pastika, Peter van Galen, Alex K. Shalek, Jennifer Lombardi Story, Richard Stone, Travis K. Hughes, Marc H. Wadsworth, Andrew A. Lane, Julia A. Verga, Gabriel K. Griffin, Olga Pozdnyakova, Timothy A. Graubert, Sofia Battaglia, Volker Hovestadt, Bradley E. Bernstein, Ilene Galinsky, Jon C. Aster, Geraldine S. Pinkus, Massachusetts Institute of Technology. Department of Chemistry, Massachusetts Institute of Technology. Institute for Medical Engineering & Science, and Koch Institute for Integrative Cancer Research at MIT
- Subjects
Adult ,Male ,Cell type ,Myeloid ,Genotype ,Cell ,Bone Marrow Cells ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Machine Learning ,03 medical and health sciences ,0302 clinical medicine ,Bone Marrow ,Cell Line, Tumor ,hemic and lymphatic diseases ,Exome Sequencing ,medicine ,Tumor Microenvironment ,Humans ,Genotyping ,Gene ,030304 developmental biology ,0303 health sciences ,Base Sequence ,RNA ,Myeloid leukemia ,Middle Aged ,Prognosis ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Mutation ,Cancer research ,Disease Progression ,Female ,Bone marrow ,Single-Cell Analysis ,Transcriptome ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease that resides within a complex microenvironment, complicating efforts to understand how different cell types contribute to disease progression. We combined single-cell RNA sequencing and genotyping to profile 38,410 cells from 40 bone marrow aspirates, including 16 AML patients and five healthy donors. We then applied a machine learning classifier to distinguish a spectrum of malignant cell types whose abundances varied between patients and between subclones in the same tumor. Cell type compositions correlated with prototypic genetic lesions, including an association of FLT3-ITD with abundant progenitor-like cells. Primitive AML cells exhibited dysregulated transcriptional programs with co-expression of stemness and myeloid priming genes and had prognostic significance. Differentiated monocyte-like AML cells expressed diverse immunomodulatory genes and suppressed T cell activity in vitro. In conclusion, we provide single-cell technologies and an atlas of AML cell states, regulators, and markers with implications for precision medicine and immune therapies. Video Abstract: A combination of transcriptomics and mutational analyses in single cells from acute myeloid leukemia patients reveals the existence of distinct functional subsets and their associated drivers.
- Published
- 2018
36. TP53 and Decitabine in Acute Myeloid Leukemia and Myelodysplastic Syndromes
- Author
-
Keith Stockerl-Goldstein, Stephen T. Oh, Catrina Fronick, Bevan Tandon, Ravi Vij, John S. Welch, Daniel C. Link, Camille N. Abboud, Armin Ghobadi, Mark A. Schroeder, Jack Baty, Amanda F. Cashen, Christopher A. Miller, Madina Sukhanova, Peter Westervelt, Michelle O'Laughlin, Wendy Stock, Timothy J. Ley, Randall W. Knoebel, Eric J. Duncavage, John F. DiPersio, Michael H. Tomasson, Robert S. Fulton, Meagan A. Jacoby, Kierstin Luber, Yi-Shan Lee, Lukas D. Wartman, Geoffrey L. Uy, Todd A. Fehniger, Timothy A. Graubert, Andrew Hantel, Matthew J. Walter, Rizwan Romee, Allegra A. Petti, Megan Janke, Iskra Pusic, Sharon Heath, Richard K. Wilson, and Niloufer Khan
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Myeloid ,business.industry ,Myelodysplastic syndromes ,Decitabine ,Myeloid leukemia ,General Medicine ,medicine.disease ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Internal medicine ,Immunology ,medicine ,Bone marrow ,business ,Prospective cohort study ,Survival rate ,medicine.drug - Abstract
BackgroundThe molecular determinants of clinical responses to decitabine therapy in patients with acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS) are unclear. MethodsWe enrolled 84 adult patients with AML or MDS in a single-institution trial of decitabine to identify somatic mutations and their relationships to clinical responses. Decitabine was administered at a dose of 20 mg per square meter of body-surface area per day for 10 consecutive days in monthly cycles. We performed enhanced exome or gene-panel sequencing in 67 of these patients and serial sequencing at multiple time points to evaluate patterns of mutation clearance in 54 patients. An extension cohort included 32 additional patients who received decitabine in different protocols. ResultsOf the 116 patients, 53 (46%) had bone marrow blast clearance (
- Published
- 2016
37. Dynamic changes in the clonal structure of MDS and AML in response to epigenetic therapy
- Author
-
Eric J. Duncavage, Kevin Elliott, Robert S. Fulton, Meagan A. Jacoby, Camille N. Abboud, Jin Shao, Christopher A. Miller, Timothy J. Ley, Geoffrey L. Uy, Catrina Fronick, Peter Westervelt, Amanda F. Cashen, Sharon Heath, L. Ganel, Matthew J. Walter, Gue Su Chang, John S. Welch, Daniel C. Link, T. Reineck, Michelle O'Laughlin, Richard K. Wilson, Timothy A. Graubert, and John F. DiPersio
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,Myeloid ,Epigenesis, Genetic ,0302 clinical medicine ,Bone Marrow ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Exome ,Aged, 80 and over ,Hematology ,Remission Induction ,High-Throughput Nucleotide Sequencing ,Myeloid leukemia ,Middle Aged ,Tumor Burden ,3. Good health ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Epigenetic therapy ,medicine.drug ,medicine.medical_specialty ,Decitabine ,Polymorphism, Single Nucleotide ,Article ,Clonal Evolution ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,Aged ,business.industry ,Genes, p53 ,medicine.disease ,Histone Deacetylase Inhibitors ,030104 developmental biology ,Myelodysplastic Syndromes ,Mutation ,Immunology ,Bone marrow ,business - Abstract
Traditional response criteria in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are based on bone marrow morphology and may not accurately reflect clonal tumor burden in patients treated with non-cytotoxic chemotherapy. We used next-generation sequencing of serial bone marrow samples to monitor MDS and AML tumor burden during treatment with epigenetic therapy (decitabine and panobinostat). Serial bone marrow samples (and skin as a source of normal DNA) from 25 MDS and AML patients were sequenced (exome or 285 gene panel). We observed that responders, including those in complete remission (CR), can have persistent measurable tumor burden (that is, mutations) for at least 1 year without disease progression. Using an ultrasensitive sequencing approach, we detected extremely rare mutations (equivalent to 1 heterozygous mutant cell in 2000 non-mutant cells) months to years before their expansion at disease relapse. While patients can live with persistent clonal hematopoiesis in a CR or stable disease, ultimately we find evidence that expansion of a rare subclone occurs at relapse or progression. Here we demonstrate that sequencing of serial samples provides an alternative measure of tumor burden in MDS or AML patients and augments traditional response criteria that rely on bone marrow blast percentage.
- Published
- 2016
38. Comprehensive genomic analysis reveals FLT3 activation and a therapeutic strategy for a patient with relapsed adult B-lymphoblastic leukemia
- Author
-
Jeffery M. Klco, Benjamin J. Ainscough, Katie M. Campbell, Vincent Magrini, Malachi Griffith, Jason Walker, Nicholas C. Spies, John F. DiPersio, Peter Westervelt, David E. Larson, Sharon Heath, Jin Zhang, Catrina Fronick, Jasreet Hundal, Elaine R. Mardis, Shelly O'Laughlin, Timothy J. Ley, Sean McGrath, Christopher G. Maher, Christopher A. Miller, Kilannin Krysiak, Robert Lesurf, Timothy A. Graubert, Matthew J. Christopher, Robert S. Fulton, Daniel C. Link, Jacqueline E. Payton, James M. Eldred, Alex H. Wagner, Zachary L. Skidmore, Tamara Lamprecht, Avinash Ramu, Rick K. Wilson, Scott M. Smith, Matthew J. Walter, Obi L. Griffith, and Shashikant Kulkarni
- Subjects
Adult ,Male ,Transcriptional Activation ,0301 basic medicine ,Cancer Research ,Biopsy ,Graft vs Host Disease ,Salvage therapy ,Biology ,Bioinformatics ,Dexamethasone ,Article ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Bone Marrow ,Recurrence ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Antineoplastic Combined Chemotherapy Protocols ,Genetics ,medicine ,Humans ,Transplantation, Homologous ,Cyclophosphamide ,Molecular Biology ,Bone Marrow Transplantation ,Sunitinib ,Gene Expression Profiling ,Genetic Variation ,Genomics ,Cell Biology ,Hematology ,Flow Cytometry ,medicine.disease ,3. Good health ,Transplantation ,Gene expression profiling ,Leukemia ,ETV6 ,030104 developmental biology ,fms-Like Tyrosine Kinase 3 ,Doxorubicin ,Vincristine ,030220 oncology & carcinogenesis ,Cytogenetic Analysis ,Fms-Like Tyrosine Kinase 3 ,Cancer research ,medicine.drug - Abstract
The genomic events responsible for the pathogenesis of relapsed adult B-lymphoblastic leukemia (B-ALL) are not yet clear. We performed integrative analysis of whole-genome, whole-exome, custom capture, whole-transcriptome (RNA-seq), and locus-specific genomic assays across nine time points from a patient with primary de novo B-ALL. Comprehensive genome and transcriptome characterization revealed a dramatic tumor evolution during progression, yielding a tumor with complex clonal architecture at second relapse. We observed and validated point mutations in EP300 and NF1, a highly expressed EP300-ZNF384 gene fusion, a microdeletion in IKZF1, a focal deletion affecting SETD2, and large deletions affecting RB1, PAX5, NF1, and ETV6. Although the genome analysis revealed events of potential biological relevance, no clinically actionable treatment options were evident at the time of the second relapse. However, transcriptome analysis identified aberrant overexpression of the targetable protein kinase encoded by the FLT3 gene. Although the patient had refractory disease after salvage therapy for the second relapse, treatment with the FLT3 inhibitor sunitinib rapidly induced a near complete molecular response, permitting the patient to proceed to a matched-unrelated donor stem cell transplantation. The patient remains in complete remission more than 4 years later. Analysis of this patient's relapse genome revealed an unexpected, actionable therapeutic target that led to a specific therapy associated with a rapid clinical response. For some patients with relapsed or refractory cancers, this approach may indicate a novel therapeutic intervention that could alter outcome.
- Published
- 2016
39. Mutation Clearance after Transplantation for Myelodysplastic Syndrome
- Author
-
Haley J. Abel, John S. Welch, Daniel C. Link, Timothy A. Graubert, Christopher A. Miller, Gue Su Chang, Matthew J. Walter, Eric J. Duncavage, Matthew J. Christopher, Peter Westervelt, Timothy J. Ley, Catrina Fronick, Kevin Elliott, Robert S. Fulton, Meagan A. Jacoby, Jin Shao, Lukas D. Wartman, John F. DiPersio, Michelle O'Laughlin, Sharon Heath, Kathryn Trinkaus, Raya Saba, Kimberly J Brendel, Natasha Catherine Edwin, Joshua Robinson, Iskra Pusic, and Geoffrey L. Uy
- Subjects
Oncology ,Adult ,medicine.medical_specialty ,Myeloid ,Transplantation Conditioning ,medicine.medical_treatment ,DNA Mutational Analysis ,Hematopoietic stem cell transplantation ,Disease ,Article ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Survival analysis ,Skin ,medicine.diagnostic_test ,business.industry ,Hematopoietic Stem Cell Transplantation ,Bone Marrow Examination ,General Medicine ,Middle Aged ,medicine.disease ,Survival Analysis ,Transplantation ,Bone marrow examination ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Myelodysplastic Syndromes ,Mutation ,Disease Progression ,business ,030215 immunology - Abstract
BACKGROUND: Allogeneic hematopoietic stem-cell transplantation is the only curative treatment for patients with myelodysplastic syndrome (MDS). The molecular predictors of disease progression after transplantation are unclear. METHODS: We sequenced bone marrow and skin samples from 90 adults with MDS who underwent allogeneic hematopoietic stem-cell transplantation after a myeloablative or reduced-intensity conditioning regimen. We detected mutations before transplantation using enhanced exome sequencing, and we evaluated mutation clearance by using error-corrected sequencing to genotype mutations in bone marrow samples obtained 30 days after transplantation. In this exploratory study, we evaluated the association of a mutation detected after transplantation with disease progression and survival. RESULTS: Sequencing identified at least one validated somatic mutation before transplantation in 86 of 90 patients (96%); 32 of these patients (37%) had at least one mutation with a maximum variant allele frequency of at least 0.5% (equivalent to 1 heterozygous mutant cell in 100 cells) 30 days after transplantation. Patients with disease progression had mutations with a higher maximum variant allele frequency at 30 days than those who did not (median maximum variant allele frequency, 0.9% vs. 0%; P
- Published
- 2018
40. Pathobiology of Acute Myeloid Leukemia
- Author
-
Andrew M. Brunner and Timothy A. Graubert
- Subjects
0301 basic medicine ,Pathogenesis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,business.industry ,Cancer research ,Medicine ,Myeloid leukemia ,Epigenetics ,030204 cardiovascular system & hematology ,business - Published
- 2018
41. Contributors
- Author
-
Omar Abdel-Wahab, Janet L. Abrahm, Sharon Adams, Adeboye H. Adewoye, Carl Allen, Richard F. Ambinder, Claudio Anasetti, John Anastasi, Julia A. Anderson, Joseph H. Antin, Aśok C. Antony, David J. Araten, Philippe Armand, Gillian Armstrong, Scott A. Armstrong, Donald M. Arnold, Andrew S. Artz, Farrukh T. Awan, Trevor P. Baglin, Don M. Benson, Edward J. Benz, Nancy Berliner, Govind Bhagat, Nina Bhardwaj, Ravi Bhatia, Smita Bhatia, Mihir D. Bhatt, Vijaya Raj Bhatt, Menachem Bitan, Craig D. Blinderman, Catherine M. Bollard, Benjamin S. Braun, Malcolm K. Brenner, Gary M. Brittenham, Robert A. Brodsky, Myles Brown, Hal E. Broxmeyer, Kathleen Brummel-Ziedins, Andrew M. Brunner, Francis K. Buadi, Birgit Burkhardt, Melissa Burns, John C. Byrd, Paolo F. Caimi, Michael A. Caligiuri, Michelle Canavan, Alan B. Cantor, Manuel Carcao, Michael C. Carroll, Shannon A. Carty, Jorge J. Castillo, Anthony K.C. Chan, John Chapin, April Chiu, John P. Chute, David B. Clark, Thomas D. Coates, Christopher R. Cogle, Nathan T. Connell, Elizabeth Cooke, Sarah Cooley, Paolo Corradini, Mark A. Creager, Richard J. Creger, Caroline Cromwell, Mark A. Crowther, Melissa M. Cushing, Corey Cutler, Chi V. Dang, Nika N. Danial, Sandeep S. Dave, James A. DeCaprio, Mary C. Dinauer, Shira Dinner, Reyhan Diz-Küçükkaya, Roger Y. Dodd, Michele L. Donato, Kenneth Dorshkind, Gianpietro Dotti, Yigal Dror, Kieron Dunleavy, Christopher C. Dvorak, Benjamin L. Ebert, Michael J. Eck, John W. Eikelboom, Narendranath Epperla, William B. Ershler, William E. Evans, Stefan Faderl, James L.M. Ferrara, Alexandra Hult Filipovich, Martin Fischer, James C. Fredenburgh, Kenneth D. Friedman, Ephraim Fuchs, Stephen J. Fuller, David Gailani, Jacques Galipeau, Patrick G. Gallagher, Karthik A. Ganapathi, Lawrence B. Gardner, Adrian P. Gee, Stanton L. Gerson, Morie A. Gertz, Patricia J. Giardina, Christopher J. Gibson, Karin Golan, Todd R. Golub, Matthew J. Gonzales, Jason Gotlib, Stephen Gottschalk, Marianne A. Grant, Timothy A. Graubert, Xylina T. Gregg, John G. Gribben, Dawn M. Gross, Tanja A. Gruber, Joan Guitart, Sandeep Gurbuxani, Shiri Gur-Cohen, Alejandro Gutierrez, Mehdi Hamadani, Parameswaran N. Hari, John H. Hartwig, Suzanne R. Hayman, Catherine P.M. Hayward, Robert P. Hebbel, Helen E. Heslop, Christopher Hillis, Christopher D. Hillyer, Karin Ho, David M. Hockenbery, Ronald Hoffman, Kerstin E. Hogg, Shernan G. Holtan, Hans-Peter Horny, Yen-Michael S. Hsu, Zachary R. Hunter, James A. Huntington, Camelia Iancu-Rubin, Ali Iqbal, David E. Isenman, Sara J. Israels, Joseph E. Italiano, Elaine S. Jaffe, Iqbal H. Jaffer, Sundar Jagannath, Ulrich Jäger, Nitin Jain, Paula James, Sima Jeha, Michael B. Jordan, Cassandra D. Josephson, Moonjung Jung, Leo Kager, Taku Kambayashi, Jennifer A. Kanakry, Hagop M. Kantarjian, Jason Kaplan, Matthew S. Karafin, Aly Karsan, Randal J. Kaufman, Richard M. Kaufman, Frank G. Keller, Kara M. Kelly, Craig M. Kessler, Nigel S. Key, Alla Keyzner, Alexander G. Khandoga, Arati Khanna-Gupta, Eman Khatib-Massalha, Harvey G. Klein, Birgit Knoechel, Orit Kollet, Barbara A. Konkle, Dimitrios P. Kontoyiannis, John Koreth, Gary A. Koretzky, Dipak Kotecha, Marina Kremyanskaya, Anju Kumari, Timothy M. Kuzel, Ralf Küppers, Martha Q. Lacy, Elana Ladas, Wendy Landier, Kfir Lapid, Tsvee Lapidot, Peter J. Larson, Marcel Levi, Russell E. Lewis, Howard A. Liebman, David Lillicrap, Wendy Lim, Judith C. Lin, Robert Lindblad, Gregory Y.H. Lip, Jane A. Little, Jens G. Lohr, José A. López, Francis W. Luscinskas, Jaroslaw P. Maciejewski, Navneet S. Majhail, Olivier Manches, Robert J. Mandle, Kenneth G. Mann, Catherine S. Manno, Andrea N. Marcogliese, Guglielmo Mariani, Francesco M. Marincola, John Mascarenhas, Steffen Massberg, Rodger P. McEver, Emer McGrath, Matthew S. McKinney, Rohtesh S. Mehta, William C. Mentzer, Giampaolo Merlini, Reid Merryman, Marc Michel, Anna Rita Migliaccio, Jeffrey S. Miller, Martha P. Mims, Traci Heath Mondoro, Paul Moorehead, Luciana R. Muniz, Nikhil C. Munshi, Vesna Najfeld, Lalitha Nayak, Ishac Nazy, Anne T. Neff, Paul M. Ness, Luigi D. Notarangelo, Sarah H. O'Brien, Owen A. O'Connor, Martin O'Donnell, Amanda Olson, Stuart H. Orkin, Menaka Pai, Sung-Yun Pai, Michael Paidas, Sandhya R. Panch, Reena L. Pande, Thalia Papayannopoulou, Rahul Parikh, Effie W. Petersdorf, Shane E. Peterson, Stefania Pittaluga, Doris M. Ponce, Laura Popolo, Josef T. Prchal, Ching-Hon Pui, Pere Puigserver, Janusz Rak, Carlos A. Ramos, Jacob H. Rand, Margaret L. Rand, Dinesh S. Rao, Farhad Ravandi, David J. Rawlings, Pavan Reddy, Mark T. Reding, Andreas Reiter, Lawrence Rice, Matthew J. Riese, Arthur Kim Ritchey, David J. Roberts, Elizabeth Roman, Cliona M. Rooney, Steven T. Rosen, David S. Rosenthal, Marlies P. Rossmann, Antal Rot, Scott D. Rowley, Jeffrey E. Rubnitz, Natalia Rydz, Mohamed E. Salama, Steven Sauk, Yogen Saunthararajah, William Savage, David Scadden, Kristen G. Schaefer, Fred Schiffman, Robert Schneidewend, Stanley L. Schrier, Edward H. Schuchman, Bridget Fowler Scullion, Kathy J. Selvaggi, Keitaro Senoo, Montaser Shaheen, Beth H. Shaz, Samuel A. Shelburne, Elizabeth J. Shpall, Susan B. Shurin, Deborah Siegal, Leslie E. Silberstein, Lev Silberstein, Roy L. Silverstein, Steven R. Sloan, Franklin O. Smith, James W. Smith, Katy Smith, David P. Steensma, Martin H. Steinberg, Wendy Stock, Jill R. Storry, Susan L. Stramer, Ronald G. Strauss, David F. Stroncek, Justin Taylor, Swapna Thota, Steven P. Treon, Anil Tulpule, Roberto Ferro Valdes, Peter Valent, Suresh Vedantham, Gregory M. Vercellotti, Michael R. Verneris, Elliott P. Vichinsky, Ulrich H. von Andrian, Julie M. Vose, Andrew J. Wagner, Ena Wang, Jia-huai Wang, Theodore E. Warkentin, Melissa P. Wasserstein, Ann Webster, Daniel J. Weisdorf, Jeffrey I. Weitz, Connie M. Westhoff, Allison P. Wheeler, Page Widick, James S. Wiley, Basem M. William, David A. Williams, Wyndham H. Wilson, Joanne Wolfe, Lucia R. Wolgast, Deborah Wood, Jennifer Wu, Joachim Yahalom, Donald L. Yee, Anas Younes, Neal S. Young, and Michelle P. Zeller
- Published
- 2018
42. Spliceosome Mutations Induce R Loop-Associated Sensitivity to ATR Inhibition in Myelodysplastic Syndromes
- Author
-
Timothy A. Graubert, Weiling Li, Hai Dang Nguyen, Lee Zou, Wan Yee Leong, Jack D. Sullivan, Pavankumar N. G. Reddy, and Matthew J. Walter
- Subjects
0301 basic medicine ,Cancer Research ,Spliceosome ,R-loop ,DNA damage ,Cell Survival ,RNA Splicing ,Ribonuclease H ,Antigens, CD34 ,Apoptosis ,Ataxia Telangiectasia Mutated Proteins ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Transcription (biology) ,Animals ,Humans ,Gene ,Chemistry ,RNA ,Molecular biology ,030104 developmental biology ,Oncology ,Myelodysplastic Syndromes ,RNA splicing ,Mutation ,Spliceosomes ,RNA Splicing Factors ,K562 Cells ,DNA ,DNA Damage ,HeLa Cells - Abstract
Heterozygous somatic mutations in spliceosome genes (U2AF1, SF3B1, ZRSR2, or SRSF2) occur in >50% of patients with myelodysplastic syndrome (MDS). These mutations occur early in disease development, suggesting that they contribute to MDS pathogenesis and may represent a unique genetic vulnerability for targeted therapy. Here, we show that RNA splicing perturbation by expression of the U2AF1(S34F) mutant causes accumulation of R loops, a transcription intermediate containing RNA:DNA hybrids and displaced single-stranded DNA, and elicits an ATR response. ATR inhibitors (ATRi) induced DNA damage and cell death in U2AF1(S34F)-expressing cells, and these effects of ATRi were enhanced by splicing modulating compounds. Moreover, ATRi-induced DNA damage was suppressed by overexpression of RNaseH1, an enzyme that specifically removes the RNA in RNA:DNA hybrids, suggesting that the ATRi sensitivity of U2AF1(S34F)-expressing cells arises from R loops. Taken together, our results demonstrate that ATR may represent a novel therapeutic target in patients with MDS carrying the U2AF1(S34F) mutation and potentially other malignancies harboring spliceosome mutations. Significance: This study provides preclinical evidence that patients with MDS or other myeloid malignancies driven by spliceosome mutations may benefit from ATR inhibition to exploit the R loop–associated vulnerability induced by perturbations in splicing. Cancer Res; 78(18); 5363–74. ©2018 AACR.
- Published
- 2017
43. Genomic analysis of germ line and somatic variants in familial myelodysplasia/acute myeloid leukemia
- Author
-
Richard Wilson, Catrina Fronick, Mark J. Levis, Allan Jiang, Khateriaa Pyrtel, Iskra Pusic, Lucy A. Godley, Kevin Elliott, Julie A. Ross, Timothy J. Ley, Jin Shao, Robert S. Fulton, John F. DiPersio, Jane E. Churpek, Dong Shen, Sharon Heath, Matthew J. Walter, Krishna L. Kanchi, Peter Westervelt, Evan M. Braunstein, Geoffrey L. Uy, Daniel C. Koboldt, Christopher A. Miller, Michelle O'Laughlin, Timothy A. Graubert, John S. Welch, and Daniel C. Link
- Subjects
Adult ,Male ,Adolescent ,Molecular Sequence Data ,Immunology ,Biology ,medicine.disease_cause ,Biochemistry ,Germline ,chemistry.chemical_compound ,Germline mutation ,hemic and lymphatic diseases ,medicine ,Humans ,Exome ,Child ,neoplasms ,Germ-Line Mutation ,Exome sequencing ,Aged ,Genetics ,Mutation ,Myeloid Neoplasia ,Base Sequence ,Myelodysplastic syndromes ,Genetic Diseases, Inborn ,Myeloid leukemia ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Hematopoiesis ,Neoplasm Proteins ,DNA-Binding Proteins ,Repressor Proteins ,Leukemia, Myeloid, Acute ,RUNX1 ,chemistry ,Myelodysplastic Syndromes ,Core Binding Factor Alpha 2 Subunit ,Female ,Transcription Factors - Abstract
Familial clustering of myelodysplastic syndromes (MDSs) and acute myeloid leukemia (AML) can be caused by inherited factors. We screened 59 individuals from 17 families with 2 or more biological relatives with MDS/AML for variants in 12 genes with established roles in predisposition to MDS/AML, and identified a pathogenic germ line variant in 5 families (29%). Extending the screen with a panel of 264 genes that are recurrently mutated in de novo AML, we identified rare, nonsynonymous germ line variants in 4 genes, each segregating with MDS/AML in 2 families. Somatic mutations are required for progression to MDS/AML in these familial cases. Using a combination of targeted and exome sequencing of tumor and matched normal samples from 26 familial MDS/AML cases and asymptomatic carriers, we identified recurrent frameshift mutations in the cohesin-associated factor PDS5B, co-occurrence of somatic ASXL1 mutations with germ line GATA2 mutations, and recurrent mutations in other known MDS/AML drivers. Mutations in genes that are recurrently mutated in de novo AML were underrepresented in the familial MDS/AML cases, although the total number of somatic mutations per exome was the same. Lastly, clonal skewing of hematopoiesis was detected in 67% of young, asymptomatic RUNX1 carriers, providing a potential biomarker that could be used for surveillance in these high-risk families.
- Published
- 2015
44. Health care utilization and end-of-life care for older patients with acute myeloid leukemia
- Author
-
Richard Stone, Julia Foster, Amir T. Fathi, Karen K. Ballen, Gregory A. Abel, David P. Steensma, Martha Wadleigh, Jennifer S. Temel, Eyal C. Attar, Daniel J. DeAngelo, Thomas W. LeBlanc, Areej El-Jawahri, Timothy A. Graubert, Philip C. Amrein, and Andrew M. Brunner
- Subjects
Cancer Research ,Pediatrics ,medicine.medical_specialty ,Palliative care ,business.industry ,Induction chemotherapy ,Retrospective cohort study ,Oncology ,Ambulatory care ,Health care ,Cohort ,medicine ,Outpatient clinic ,business ,Intensive care medicine ,End-of-life care - Abstract
BACKGROUND Health care utilization in older adults (age ≥60 years) with acute myeloid leukemia (AML) has not been well studied. METHODS We conducted a retrospective analysis of 330 consecutive older patients who were diagnosed with AML between May 1, 2005 and December 23, 2011, at 2 hospitals in Boston to examine their health care utilization and end-of-life care. Using multivariable logistic and linear regression models adjusting for covariates, we also compared health care utilization between patients who received intensive induction chemotherapy (n = 197; cytarabine/ anthracycline combination) versus nonintensive chemotherapy (n = 133; single-agent therapy). RESULTS The median number of hospitalizations for the entire cohort was 4.2 (range, 1-18 hospitalizations). Patients who died spent a mean of 28.3% of their life after diagnosis in the hospital and 13.8% of their life attending outpatient clinic appointments. Although the majority of patients (87.9%) died during the 2-year follow-up period, a minority received palliative care (16.2%) or hospice (23.1%) services. Within 30 days of death, 84.5% of patients were hospitalized, and 61% died in the hospital. Among the patients who died, those who received intensive induction therapy (vs nonintensive therapy) spent 30% more of their life after diagnosis in the hospital (P
- Published
- 2015
45. Functional analysis of a chromosomal deletion associated with myelodysplastic syndromes using isogenic human induced pluripotent stem cells
- Author
-
Danwei Huangfu, Stephen D. Nimer, Andriana G. Kotini, Fabiana Perna, Chan Jung Chang, Abhinav B. Nagulapally, Ibrahim Boussaad, R. David Hawkins, Eirini P. Papapetrou, Charles E. Murry, Virginia M. Klimek, Gregory A. Fishbein, Emily K. Dolezal, Jeffrey J. Delrow, and Timothy A. Graubert
- Subjects
Somatic cell ,Induced Pluripotent Stem Cells ,Biomedical Engineering ,Bioengineering ,Biology ,Applied Microbiology and Biotechnology ,Article ,Mice ,medicine ,Animals ,Humans ,Induced pluripotent stem cell ,Chromosomal Deletion ,Genetics ,Myelodysplastic syndromes ,medicine.disease ,Phenotype ,3. Good health ,Haematopoiesis ,Karyotyping ,Myelodysplastic Syndromes ,Molecular Medicine ,Chromosome Deletion ,Haploinsufficiency ,Genetic Engineering ,Reprogramming ,Chromosomes, Human, Pair 7 ,Biotechnology - Abstract
Chromosomal deletions associated with human diseases, such as cancer, are common, but synteny issues complicate modeling of these deletions in mice. We use cellular reprogramming and genome engineering to functionally dissect the loss of chromosome 7q (del(7q)), a somatic cytogenetic abnormality present in myelodysplastic syndromes (MDS). We derive del(7q)- and isogenic karyotypically normal induced pluripotent stem cells (iPSCs) from hematopoietic cells of MDS patients and show that the del(7q) iPSCs recapitulate disease-associated phenotypes, including impaired hematopoietic differentiation. These disease phenotypes are rescued by spontaneous dosage correction and can be reproduced in karyotypically normal cells by engineering hemizygosity of defined chr7q segments in a 20-Mb region. We use a phenotype-rescue screen to identify candidate haploinsufficient genes that might mediate the del(7q)- hematopoietic defect. Our approach highlights the utility of human iPSCs both for functional mapping of disease-associated large-scale chromosomal deletions and for discovery of haploinsufficient genes.
- Published
- 2015
46. Caspase-9 is required for normal hematopoietic development and protection from alkylator-induced DNA damage in mice
- Author
-
Robert S. Fulton, Matthew J. Walter, Elaine R. Mardis, Richard K. Wilson, Daniel C. Link, Christopher A. Miller, Li Ding, Michael D. McLellan, Elise Peterson Lu, Timothy J. Ley, John F. DiPersio, Peter Westervelt, and Timothy A. Graubert
- Subjects
Alkylating Agents ,Hematopoiesis and Stem Cells ,DNA damage ,Immunology ,Apoptosis ,Bone Marrow Cells ,Mice, Transgenic ,Biochemistry ,Mice ,Bone Marrow ,hemic and lymphatic diseases ,White blood cell ,medicine ,Animals ,Crosses, Genetic ,business.industry ,Myelodysplastic syndromes ,Gene Expression Regulation, Developmental ,Cell Biology ,Hematology ,Flow Cytometry ,Hematopoietic Stem Cells ,medicine.disease ,Caspase 9 ,Hematopoiesis ,Mice, Inbred C57BL ,Transplantation ,Haematopoiesis ,Leukemia ,Phenotype ,medicine.anatomical_structure ,Ethylnitrosourea ,Myelodysplastic Syndromes ,Mutation ,Bone marrow ,Stem cell ,business ,DNA Damage - Abstract
Apoptosis and the DNA damage responses have been implicated in hematopoietic development and differentiation, as well as in the pathogenesis of myelodysplastic syndromes (MDS) and leukemia. However, the importance of late-stage mediators of apoptosis in hematopoiesis and leukemogenesis has not been elucidated. Here, we examine the role of caspase-9 (Casp9), the initiator caspase of the intrinsic apoptotic cascade, in murine fetal and adult hematopoiesis. Casp9 deficiency resulted in decreased erythroid and B-cell progenitor abundance and impaired function of hematopoietic stem cells after transplantation. Mouse bone marrow chimeras lacking Casp9 or its cofactor Apaf1 developed low white blood cell counts, decreased B-cell numbers, anemia, and reduced survival. Defects in apoptosis have also been previously implicated in susceptibility to therapy-related leukemia, a disease caused by exposure to DNA-damaging chemotherapy. We found that the burden of DNA damage was increased in Casp9-deficient cells after exposure to the alkylator, N-ethyl-nitrosourea (ENU). Furthermore, exome sequencing revealed that oligoclonal hematopoiesis emerged in Casp9-deficient bone marrow chimeras after alkylator exposure. Taken together, these findings suggest that defects in apoptosis could be a key step in the pathogenesis of alkylator-associated secondary malignancies.
- Published
- 2014
47. Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia
- Author
-
Giridharan Ramsingh, Jeffery M. Klco, Andrew L. Young, Richard K. Wilson, Terrence N. Wong, John S. Welch, Daniel C. Link, Matthew J. Walter, Li Ding, Tamara Lamprecht, Robert S. Fulton, Sharon Heath, Dong Shen, Todd E. Druley, Elaine R. Mardis, Jasreet Hundal, Timothy J. Ley, Waseem Touma, John F. DiPersio, Jack Baty, Timothy A. Graubert, Peter Westervelt, and Christopher A. Miller
- Subjects
Heterozygote ,medicine.medical_specialty ,Myeloid ,medicine.medical_treatment ,Clone (cell biology) ,Biology ,Models, Biological ,Article ,Evolution, Molecular ,Mice ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Cell Lineage ,Progenitor cell ,neoplasms ,Alleles ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,Chemotherapy ,Multidisciplinary ,Models, Genetic ,Cytogenetics ,Hematopoietic Stem Cells ,Genes, p53 ,medicine.disease ,Clone Cells ,3. Good health ,Leukemia, Myeloid, Acute ,Leukemia ,Haematopoiesis ,medicine.anatomical_structure ,Drug Resistance, Neoplasm ,Ethylnitrosourea ,030220 oncology & carcinogenesis ,Mutation ,Immunology ,Bone marrow ,DNA Damage - Abstract
Therapy-related acute myeloid leukaemia (t-AML) and therapy-related myelodysplastic syndrome (t-MDS) are well-recognized complications of cytotoxic chemotherapy and/or radiotherapy. There are several features that distinguish t-AML from de novo AML, including a higher incidence of TP53 mutations, abnormalities of chromosomes 5 or 7, complex cytogenetics and a reduced response to chemotherapy. However, it is not clear how prior exposure to cytotoxic therapy influences leukaemogenesis. In particular, the mechanism by which TP53 mutations are selectively enriched in t-AML/t-MDS is unknown. Here, by sequencing the genomes of 22 patients with t-AML, we show that the total number of somatic single-nucleotide variants and the percentage of chemotherapy-related transversions are similar in t-AML and de novo AML, indicating that previous chemotherapy does not induce genome-wide DNA damage. We identified four cases of t-AML/t-MDS in which the exact TP53 mutation found at diagnosis was also present at low frequencies (0.003-0.7%) in mobilized blood leukocytes or bone marrow 3-6 years before the development of t-AML/t-MDS, including two cases in which the relevant TP53 mutation was detected before any chemotherapy. Moreover, functional TP53 mutations were identified in small populations of peripheral blood cells of healthy chemotherapy-naive elderly individuals. Finally, in mouse bone marrow chimaeras containing both wild-type and Tp53(+/-) haematopoietic stem/progenitor cells (HSPCs), the Tp53(+/-) HSPCs preferentially expanded after exposure to chemotherapy. These data suggest that cytotoxic therapy does not directly induce TP53 mutations. Rather, they support a model in which rare HSPCs carrying age-related TP53 mutations are resistant to chemotherapy and expand preferentially after treatment. The early acquisition of TP53 mutations in the founding HSPC clone probably contributes to the frequent cytogenetic abnormalities and poor responses to chemotherapy that are typical of patients with t-AML/t-MDS.
- Published
- 2014
48. U2AF1 Mutations Alter Sequence Specificity of pre-mRNA Binding and Splicing
- Author
-
Malachi Griffith, Jasreet Hundal, Shamika Ketkar-Kulkarni, Theresa Okeyo-Owuor, Li Ding, Timothy J. Ley, Brian S. White, Kholiswa M. Laird, Dipika R. Mohan, Clara L. Kielkopf, Timothy A. Graubert, Matthew J. Walter, Rakesh Chatrikhi, and Sang-hyun Kim
- Subjects
Cancer Research ,Molecular Sequence Data ,Primary Cell Culture ,Exonic splicing enhancer ,Antigens, CD34 ,Biology ,Transfection ,Article ,03 medical and health sciences ,Splicing factor ,0302 clinical medicine ,RNA Precursors ,Humans ,snRNP ,Gene ,030304 developmental biology ,U2AF2 ,0303 health sciences ,Binding Sites ,Base Sequence ,Alternative splicing ,De novo Myelodysplastic Syndrome ,Nuclear Proteins ,Hematology ,Fetal Blood ,Splicing Factor U2AF ,Molecular biology ,3. Good health ,Alternative Splicing ,Oncology ,Ribonucleoproteins ,030220 oncology & carcinogenesis ,Myelodysplastic Syndromes ,RNA splicing ,Mutation ,Leukocytes, Mononuclear ,Spliceosomes ,Plasmids ,Protein Binding ,Signal Transduction - Abstract
We previously identified missense mutations in the U2AF1 splicing factor affecting codons S34 (S34F and S34Y) or Q157 (Q157R and Q157P) in 11% of the patients with de novo myelodysplastic syndrome (MDS). Although the role of U2AF1 as an accessory factor in the U2 snRNP is well established, it is not yet clear how these mutations affect splicing or contribute to MDS pathophysiology. We analyzed splice junctions in RNA-seq data generated from transfected CD34+ hematopoietic cells and found significant differences in the abundance of known and novel junctions in samples expressing mutant U2AF1 (S34F). For selected transcripts, splicing alterations detected by RNA-seq were confirmed by analysis of primary de novo MDS patient samples. These effects were not due to impaired U2AF1 (S34F) localization as it co-localized normally with U2AF2 within nuclear speckles. We further found evidence in the RNA-seq data for decreased affinity of U2AF1 (S34F) for uridine (relative to cytidine) at the e-3 position immediately upstream of the splice acceptor site and corroborated this finding using affinity-binding assays. These data suggest that the S34F mutation alters U2AF1 function in the context of specific RNA sequences, leading to aberrant alternative splicing of target genes, some of which may be relevant for MDS pathogenesis.
- Published
- 2014
49. AML Genomics for the Clinician
- Author
-
Timothy A. Graubert and Richard Stone
- Subjects
Chemotherapy ,Genome, Human ,business.industry ,medicine.medical_treatment ,Myeloid leukemia ,Cancer ,Genomics ,Sequence Analysis, DNA ,Hematology ,Disease ,Bioinformatics ,medicine.disease ,Transplantation ,Leukemia, Myeloid, Acute ,Gene panel ,Mutation ,Humans ,Medicine ,Genetic Predisposition to Disease ,Stem cell ,business - Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease, characterized by frequent resistance to available chemotherapeutic agents. The basic therapy for patients with AML has changed little over the past 30 years. Improvements in outcome in recent decades in younger adult cohorts have generally been ascribed to better supportive care (ie, transfusion and antimicrobial therapy); older adults with AML continue to fare poorly. The explosion of new knowledge regarding the AML genome has yet to be translated into therapeutic benefit, but analysis of specific molecular features in AML samples has enabled the field to more accurately parse out prognosis and assign appropriate therapies (eg, chemotherapy vs allogeneic stem cell transplantation) for groups of patients. Cytogenetic analysis, whether by metaphase or interphase analysis, has been the main tool used to divide patients into varying prognostic subsets, but it has been modified in recent years to include assessment of mutations in a small number of genes. In the past several years, new technologies have provided strategies to interrogate individual cancer genomes in a broad and in-depth fashion. The present article discusses the potential of these new technologies, particularly gene panel and whole-exome or whole-genome sequencing, to improve diagnosis, prognosis, and therapeutic outcome in AML.
- Published
- 2014
50. New Molecular Abnormalities and Clonal Architecture in AML: From Reciprocal Translocations to Whole-Genome Sequencing
- Author
-
Andrew M. Brunner, Timothy A. Graubert, and Amir T. Fathi
- Subjects
NPM1 ,Myeloid ,Biology ,Somatic evolution in cancer ,Translocation, Genetic ,Dioxygenases ,Epigenesis, Genetic ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,CEBPA ,medicine ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Molecular Targeted Therapy ,Epigenetics ,Epigenomics ,Genetics ,Genome, Human ,Polycomb Repressive Complex 2 ,Nuclear Proteins ,Myeloid leukemia ,General Medicine ,medicine.disease ,Isocitrate Dehydrogenase ,DNA-Binding Proteins ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Mutation ,CCAAT-Enhancer-Binding Proteins ,Nucleophosmin - Abstract
Acute myeloid leukemia (AML) is characterized by recurrent genetic alterations, including amplifications, deletions, rearrangements, and point mutations. Clinically, these lesions can be used to stratify patients into categories of risk, which directs further clinical management and prognostication. Patient risk categories were first described based on recurrent karyotypic abnormalities; most patients with AML, however, fall into intermediate cytogenetic risk, the majority harboring a normal karyotype. Subsequently, identification of recurrently mutated genes, including FLT3, NPM1, and CEBPA, allowed further stratification of patients with a normal karyotype. More extensive genomic and epigenomic analysis of AML samples has expanded the number of known molecular alterations present in this disease. The further understanding of this mutational landscape has shed light into the pathogenesis of AML. AML arises in a founding clone that often gives rise to subclones. Clonal evolution is a feature of the natural history of the disease but may also be influenced by the selective pressure of chemotherapy. The complex network of genetic and epigenetic alterations in this disease has yielded numerous new targets for intervention. In the future, further understanding of this mutational framework, along with the development of novel therapeutic targets, may lead to improved outcomes for patients with AML.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.