479 results on '"Druker BJ"'
Search Results
2. Insights from pre-clinical studies for new combination treatment regimens with the Bcr-Abl kinase inhibitor imatinib mesylate (Gleevec/Glivec) in chronic myelogenous leukemia: a translational perspective
- Author
-
La Rosée, P, O'Dwyer, ME, and Druker, BJ
- Published
- 2002
- Full Text
- View/download PDF
3. High-throughput mutational screen of the tyrosine kinome in chronic myelomonocytic leukemia
- Author
-
Tyner, JW, Loriaux, MM, Erickson, H, Eide, CA, Deininger, J, MacPartlin, M, Willis, SG, Lange, T, Druker, BJ, Kovacsovics, T, Maziarz, R, Gattermann, N, and Deininger, MW
- Published
- 2009
4. Long-Term Outcomes of Imatinib Treatment for Chronic Myeloid Leukemia
- Author
-
Hochhaus A, Larson RA, Guilhot F, Radich JP, Branford S, Hughes TP, Baccarani M, Deininger MW, Cervantes F, Fujihara S, Ortmann CE, Menssen HD, Kantarjian H, O'Brien SG, Druker BJ, IRIS Investigators, and IRIS Investigators.
- Published
- 2017
5. Brian J. Druker, MD
- Author
-
Druker Bj
- Subjects
Gerontology ,Immunology ,Library science ,Hematology ,Psychology - Published
- 2001
- Full Text
- View/download PDF
6. Dasatinib induces notable hematologic and cytogenetic responses in chronic-phase chronic myeloid leukemia after failure of imatinib therapy
- Author
-
Hochhaus A, Kantarjian HM, Lipton JH, Apperley JF, Druker BJ, Facon T, Goldberg SL, Cervantes F, Niederwieser D, Silver RT, Stone RM, Hughes TP, Muller MC, Ezzeddine R, Countouriotis AM, Shah N.P., BACCARANI, MICHELE, Hochhaus A, Kantarjian HM, Baccarani M, Lipton JH, Apperley JF, Druker BJ, Facon T, Goldberg SL, Cervantes F, Niederwieser D, Silver RT, Stone RM, Hughes TP, Muller MC, Ezzeddine R, Countouriotis AM, and Shah NP.
- Published
- 2007
7. Monitoring CML patients responding to treatment with tyrosine kinase inhibitors: review and recommendations for harmonizing current methodology for detectingBCR-ABL transcripts and kinase domain mutations and for expressing results
- Author
-
Hughes, T, Deininger, M, Hochhaus, A, Branford, S, Radich, J, Kaeda, J, Baccarani, M, Cortes, J, Cross, Nc, Druker, Bj, Gabert, J, Grimwade, D, Hehlmann, R, Kamel Reid, S, Lipton, Jh, Longtine, J, Martinelli, G, Saglio, Giuseppe, Soverini, S, Stock, W, and Goldman, J. M.
- Published
- 2006
8. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia
- Author
-
Druker, Bj, Guilhot, F, O'Brien, Sg, Gathmann, I, Kantarjian, H, Gattermann, N, Deininger, Mw, Silver, Rt, Goldman, Jm, Stone, Rm, Cervantes, F, Hochhaus, A, Powell, Bl, Gabrilove, Jl, Rousselot, P, Reiffers, J, Cornelissen, Jj, Hughes, T, Agis, H, Fischer, T, Verhoef, G, Shepherd, J, Saglio, Giuseppe, Gratwohl, A, Nielsen, Jl, Radich, Jp, Simonsson, B, Taylor, K, Baccarani, M, So, C, Letvak, L, Larson, Ra, and Iris, Investigators
- Published
- 2006
9. Six-year follow-up of patients receiving imatinib for the first-line treatment of chronic myeloid leukemia
- Author
-
Hochhaus, A., O'Brien, SG, Guilhot, François, Druker, BJ, Branford, S., Foroni, L., Goldman, JM, Müller, MC, Radich, JP, Rudoltz, M., Mone, M., Gathmann, I., Hughes, TP, Larson, RA, Martiat, Philippe, Hochhaus, A., O'Brien, SG, Guilhot, François, Druker, BJ, Branford, S., Foroni, L., Goldman, JM, Müller, MC, Radich, JP, Rudoltz, M., Mone, M., Gathmann, I., Hughes, TP, Larson, RA, and Martiat, Philippe
- Abstract
info:eu-repo/semantics/published
- Published
- 2009
10. Imatinib pharmacokinetics and its correlation with response and safety in chronic-phase chronic myeloid leukemia: a subanalysis of the IRIS study
- Author
-
Larson, RA, Druker, BJ, Guilhot, François, O'Brien, SG, Riviere, GJ, Krahnke, T., Gathmann, I., Wang, Yue, Martiat, Philippe, Larson, RA, Druker, BJ, Guilhot, François, O'Brien, SG, Riviere, GJ, Krahnke, T., Gathmann, I., Wang, Yue, and Martiat, Philippe
- Abstract
info:eu-repo/semantics/published
- Published
- 2008
11. FTY720, a new alternative for treating blast crisis chronic myelogenous leukemia and Philadelphia chromosome-positive acute lymphocytic leukemia
- Author
-
Neviani, P, Santhanam, R, Oaks, J, Eiring, A, Notari, M, Blaser, B, Liu, S, Trotta, R, Muthusamy, N, GAMBACORTI PASSERINI, C, Druker, B, Cortes, J, Marcucci, G, Chen, C, Verrills, N, Roy, D, Caligiuri, M, Bloomfield, C, Byrd, J, Perrotti, D, Oaks, JJ, Eiring, AM, Blaser, BW, Druker, BJ, Chen, CS, Verrills, NM, Roy, DC, Caligiuri, MA, Bloomfield, CD, Byrd, JC, Perrotti, D., GAMBACORTI PASSERINI, CARLO, Neviani, P, Santhanam, R, Oaks, J, Eiring, A, Notari, M, Blaser, B, Liu, S, Trotta, R, Muthusamy, N, GAMBACORTI PASSERINI, C, Druker, B, Cortes, J, Marcucci, G, Chen, C, Verrills, N, Roy, D, Caligiuri, M, Bloomfield, C, Byrd, J, Perrotti, D, Oaks, JJ, Eiring, AM, Blaser, BW, Druker, BJ, Chen, CS, Verrills, NM, Roy, DC, Caligiuri, MA, Bloomfield, CD, Byrd, JC, Perrotti, D., and GAMBACORTI PASSERINI, CARLO
- Abstract
Blast crisis chronic myelogenous leukemia (CML-BC) and Philadelphia chromosome-positive (Ph1-positive) acute lymphocytic leukemia (ALL) are 2 fatal BCR/ABL-driven leukemias against which Abl kinase inhibitors fail to induce a long-term response. We recently reported that functional loss of protein phosphatase 2A (PP2A) activity is important for CML blastic transformation. We assessed the therapeutic potential of the PP2A activator FTY720 (2-amino-2-[2-(4-octylphenyl)ethyl]-1,3-propanediol hydrochloride), an immunomodulator in Phase III trials for patients with multiple sclerosis or undergoing organ transplantation, in CML-BC and Ph1 ALL patient cells and in in vitro and in vivo models of these BCR/ABL(+) leukemias. Our data indicate that FTY720 induces apoptosis and impairs clonogenicity of imatinib/dasatinib-sensitive and -resistant p210/p190(BCR/ABL) myeloid and lymphoid cell lines and CML-BCCD34+ and Ph1 ALL(CD34+/CD19+) progenitors but not of normal CD34(+) and CD34(+)/CD19(+) bone marrow cells. Furthermore, pharmacologic doses of FTY720 remarkably suppress in vivo p210/p190(BCR/ABL)-driven [including p210/p190(BCR/ABL) (T315I)] leukemogenesis without exerting any toxicity. Altogether, these results highlight the therapeutic relevance of rescuing PP2A tumor suppressor activity in Ph1 leukemias and strongly support the introduction of the PP2A activator FTY720 in the treatment of CML-BC and Ph1 ALL patients.
- Published
- 2007
12. Imatinib compared with interferon and low-dose cytarabine for newlydiagnosed chronic-phase chronic myeloid leukemia.
- Author
-
O'Brien, SG, Guilhot, F, Larson, RA, Gathmann, I, Baccarani, M, Cervantes, F, Cornelissen, JJ, Fischer, T, Hochhaus, A, Hughes, T, Lechner, K, Nielsen, JL, Rousselot, P, Reiffers, J, Saglio, G, Shepherd, J, Simonsson, B, Gratwohl, A, Goldman, JM, Kantarjian, H, Taylor, K, Verhoef, G, Bolton, AE, Capdeville, R, Druker, BJ, O'Brien, SG, Guilhot, F, Larson, RA, Gathmann, I, Baccarani, M, Cervantes, F, Cornelissen, JJ, Fischer, T, Hochhaus, A, Hughes, T, Lechner, K, Nielsen, JL, Rousselot, P, Reiffers, J, Saglio, G, Shepherd, J, Simonsson, B, Gratwohl, A, Goldman, JM, Kantarjian, H, Taylor, K, Verhoef, G, Bolton, AE, Capdeville, R, and Druker, BJ
- Published
- 2003
13. Sceptical scientists
- Author
-
Druker Bj
- Subjects
business.industry ,Medicine ,Myeloid leukemia ,General Medicine ,business ,Bioinformatics - Published
- 2001
- Full Text
- View/download PDF
14. Crkl is constitutively tyrosine phosphorylated in platelets from chronic myelogenous leukemia patients and inducibly phosphorylated in normal platelets stimulated by thrombopoietin
- Author
-
Oda, A, primary, Miyakawa, Y, additional, Druker, BJ, additional, Ishida, A, additional, Ozaki, K, additional, Ohashi, H, additional, Wakui, M, additional, Handa, M, additional, Watanabe, K, additional, Okamoto, S, additional, and Ikeda, Y, additional
- Published
- 1996
- Full Text
- View/download PDF
15. p120c-cbl is present in human blood platelets and is differentially involved in signaling by thrombopoietin and thrombin
- Author
-
Oda, A, primary, Ozaki, K, additional, Druker, BJ, additional, Miyakawa, Y, additional, Miyazaki, H, additional, Handa, M, additional, Morita, H, additional, Ohashi, H, additional, and Ikeda, Y, additional
- Published
- 1996
- Full Text
- View/download PDF
16. Thrombopoietin primes human platelet aggregation induced by shear stress and by multiple agonists
- Author
-
Oda, A, primary, Miyakawa, Y, additional, Druker, BJ, additional, Ozaki, K, additional, Yabusaki, K, additional, Shirasawa, Y, additional, Handa, M, additional, Kato, T, additional, Miyazaki, H, additional, Shimosaka, A, additional, and Ikeda, Y, additional
- Published
- 1996
- Full Text
- View/download PDF
17. Thrombopoietin induces tyrosine phosphorylation of Stat3 and Stat5 in human blood platelets
- Author
-
Miyakawa, Y, primary, Oda, A, additional, Druker, BJ, additional, Miyazaki, H, additional, Handa, M, additional, Ohashi, H, additional, and Ikeda, Y, additional
- Published
- 1996
- Full Text
- View/download PDF
18. Recombinant thrombopoietin induces rapid protein tyrosine phosphorylation of Janus kinase 2 and Shc in human blood platelets
- Author
-
Miyakawa, Y, primary, Oda, A, additional, Druker, BJ, additional, Kato, T, additional, Miyazaki, H, additional, Handa, M, additional, and Ikeda, Y, additional
- Published
- 1995
- Full Text
- View/download PDF
19. Point mutations in the conserved box 1 region inactivate the human granulocyte colony-stimulating factor receptor for growth signal transduction and tyrosine phosphorylation of p75c-rel
- Author
-
Avalos, BR, primary, Hunter, MG, additional, Parker, JM, additional, Ceselski, SK, additional, Druker, BJ, additional, Corey, SJ, additional, and Mehta, VB, additional
- Published
- 1995
- Full Text
- View/download PDF
20. Abnormal response to granulocyte colony-stimulating factor (G-CSF) in canine cyclic hematopoiesis is not caused by altered G-CSF receptor expression
- Author
-
Avalos, BR, primary, Broudy, VC, additional, Ceselski, SK, additional, Druker, BJ, additional, Griffin, JD, additional, and Hammond, WP, additional
- Published
- 1994
- Full Text
- View/download PDF
21. Granulocyte-macrophage colony-stimulating factor, interleukin-3, and steel factor induce rapid tyrosine phosphorylation of p42 and p44 MAP kinase
- Author
-
Okuda, K, primary, Sanghera, JS, additional, Pelech, SL, additional, Kanakura, Y, additional, Hallek, M, additional, Griffin, JD, additional, and Druker, BJ, additional
- Published
- 1992
- Full Text
- View/download PDF
22. Phase III, randomized, open-label study of daily imatinib mesylate 400 mg versus 800 mg in patients with newly diagnosed, previously untreated chronic myeloid leukemia in chronic phase using molecular end points: tyrosine kinase inhibitor optimization...
- Author
-
Cortes JE, Baccarani M, Guilhot F, Druker BJ, Branford S, Kim DW, Pane F, Pasquini R, Goldberg SL, Kalaycio M, Moiraghi B, Rowe JM, Tothova E, De Souza C, Rudoltz M, Yu R, Krahnke T, Kantarjian HM, Radich JP, and Hughes TP
- Published
- 2010
- Full Text
- View/download PDF
23. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia.
- Author
-
Druker BJ, Guilhot F, O'Brien SG, Gathmann I, Kantarjian H, Gattermann N, Deininger MWN, Silver RT, Goldman JM, Stone RM, Cervantes F, Hochhaus A, Powell BL, Gabrilove JL, Rousselot P, Reiffers J, Cornelissen JJ, Hughes T, Agis H, and Fischer T
- Published
- 2006
24. The cancer chemotherapy drug etoposide (VP-16) induces proinflammatory cytokine production and sickness behavior-like symptoms in a mouse model of cancer chemotherapy-related symptoms.
- Author
-
Wood LJ, Nail LM, Perrin NA, Elsea CR, Fischer A, and Druker BJ
- Abstract
Cancer chemotherapy-related symptoms such as fatigue, malaise, loss of interest in social activities, difficulty concentrating, and changes in sleep patterns can lead to treatment delays, dose reductions, or termination and have a profound effect on the physical, psychosocial, and economic aspects of quality of life. Clinicians have long suspected that these symptoms are similar to those associated with 'sickness behavior,' which is triggered by the production of the inflammatory cytokines IL-1beta, TNF-alpha, and IL-6 by macrophages and other cells of the innate immune system in response to immune challenge. The p38 mitogen-activated protein kinase (p38 MAPK) plays a central role in the production of these cytokines and consequently the induction of sickness behavior. Several cancer chemotherapy drugs have been shown to activate p38 MAPK, but whether these drugs can also induce the production of inflammatory cytokines to cause sickness behavior is unknown. The aim of this study was to determine whether the cancer chemotherapy drug etoposide (VP-16), which is known to activate p38 MAPK, could induce inflammatory cytokine production by murine macrophages and sickness-like behaviors when injected into mice. VP-16 activated p38 MAPK and induced IL-6 production in murine macrophages in a p38 MAPK- dependent manner. VP-16 administration rapidly increased serum levels of IL-6 in healthy mice and induced sickness-like behaviors as evidenced by a decrease in food intake, body weight, hemoglobin level, and voluntary wheel-running activity. These findings support the idea that the induction of IL-1beta, TNF-alpha, and IL-6 by cancer chemotherapy drugs underlies the fatigue and associated symptoms experienced by people undergoing cancer chemotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
25. Circumventing resistance to kinase-inhibitor therapy.
- Author
-
Druker BJ and Druker, Brian J
- Published
- 2006
26. Signal transduction of interleukin-6 involves tyrosine phosphorylation of multiple cytosolic proteins and activation of Src-family kinases Fyn, Hck, and Lyn in multiple myeloma cell lines
- Author
-
Michael Hallek, Neumann, C., Schaffer, M., Danhauserriedl, S., Vonbubnoff, N., Devos, G., Druker, Bj, Yasukawa, K., Griffin, Jd, and Emmerich, B.
27. Chronic myeloid leukemia. Sceptical scientists.
- Author
-
Druker BJ and Druker, B J
- Published
- 2001
28. Phase III, randomized, open-label study of daily imatinib mesylate 400 mg versus 800 mg in patients with newly diagnosed, previously untreated chronic myeloid leukemia in chronic phase using molecular end points: tyrosine kinase inhibitor optimization and selectivity study
- Author
-
Richard N. Yu, Tillmann Krahnke, Timothy P. Hughes, Ricardo Pasquini, François Guilhot, Michele Baccarani, Stuart L. Goldberg, Jacob M. Rowe, Matt Kalaycio, Hagop M. Kantarjian, Fabrizio Pane, Beatriz Moiraghi, Jorge E. Cortes, Carmino Antonio De Souza, Dong-Wook Kim, Marc Rudoltz, Elena Tothova, Brian J. Druker, Susan Branford, Jerald P. Radich, Cortes, Je, Baccarani, M, Guilhot, F, Druker, Bj, Branford, S, Kim, Dw, Pane, Fabrizio, Pasquini, R, Goldberg, Sl, Kalaycio, M, Moiraghi, B, Rowe, Jm, Tothova, E, De Souza, C, Rudoltz, M, Yu, R, Krahnke, T, Kantarjian, Hm, Radich, Jp, Hughes, T. P., Hughes, Tp, Cortes JE, Baccarani M, Guilhot F, Druker BJ, Branford S, Kim DW, Pane F, Pasquini R, Goldberg SL, Kalaycio M, Moiraghi B, Rowe JM, Tothova E, De Souza C, Rudoltz M, Yu R, Krahnke T, and Kantarjian HM
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Adolescent ,medicine.drug_class ,Antineoplastic Agents ,Gastroenterology ,Tyrosine-kinase inhibitor ,Piperazines ,Young Adult ,Internal medicine ,medicine ,Humans ,Adverse effect ,Protein Kinase Inhibitors ,Aged ,Dose-Response Relationship, Drug ,business.industry ,Myeloid leukemia ,Imatinib ,ORIGINAL REPORTS ,Middle Aged ,Protein-Tyrosine Kinases ,medicine.disease ,Surgery ,Leukemia ,medicine.anatomical_structure ,Imatinib mesylate ,Pyrimidines ,Treatment Outcome ,Oncology ,Benzamides ,Leukemia, Myeloid, Chronic-Phase ,Imatinib Mesylate ,Female ,business ,Chronic myelogenous leukemia ,medicine.drug - Abstract
Purpose To evaluate the safety and efficacy of initial treatment with imatinib mesylate 800 mg/d (400 mg twice daily) versus 400 mg/d in patients with newly diagnosed chronic myeloid leukemia in chronic phase. Patients and Methods A total of 476 patients were randomly assigned 2:1 to imatinib 800 mg (n = 319) or 400 mg (n = 157) daily. The primary end point was the major molecular response (MMR) rate at 12 months. Results At 12 months, differences in MMR and complete cytogenetic response (CCyR) rates were not statistically significant (MMR, 46% v 40%; P = .2035; CCyR, 70% v 66%; P = .3470). However, MMR occurred faster among patients randomly assigned to imatinib 800 mg/d, who had higher rates of MMR at 3 and 6 months compared with those in the imatinib 400-mg/d arm (P = .0035 by log-rank test). CCyR also occurred faster in the 800-mg/d arm (CCyR at 6 months, 57% v 45%; P = .0146). The most common adverse events were edema, gastrointestinal problems, and rash, and all were more common in patients in the 800-mg/d arm. Grades 3 to 4 hematologic toxicity also occurred more frequently in patients receiving imatinib 800 mg/d. Conclusion MMR rates at 1 year were similar with imatinib 800 mg/d and 400 mg/d, but MMR and CCyR occurred earlier in patients treated with 800 mg/d. Continued follow-up is needed to determine the clinical significance of earlier responses on high-dose imatinib.
- Published
- 2010
29. Ponatinib in refractory Philadelphia chromosome-positive leukemias.
- Author
-
Cortes JE, Kantarjian H, Shah NP, Bixby D, Mauro MJ, Flinn I, O'Hare T, Hu S, Narasimhan NI, Rivera VM, Clackson T, Turner CD, Haluska FG, Druker BJ, Deininger MW, Talpaz M, Cortes, Jorge E, Kantarjian, Hagop, Shah, Neil P, and Bixby, Dale
- Abstract
Background: Resistance to tyrosine kinase inhibitors in patients with chronic myeloid leukemia (CML) and Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph-positive ALL) is frequently caused by mutations in the BCR-ABL kinase domain. Ponatinib (AP24534) is a potent oral tyrosine kinase inhibitor that blocks native and mutated BCR-ABL, including the gatekeeper mutant T315I, which is uniformly resistant to tyrosine kinase inhibitors.Methods: In this phase 1 dose-escalation study, we enrolled 81 patients with resistant hematologic cancers, including 60 with CML and 5 with Ph-positive ALL. Ponatinib was administered once daily at doses ranging from 2 to 60 mg. Median follow-up was 56 weeks (range, 2 to 140).Results: Dose-limiting toxic effects included elevated lipase or amylase levels and pancreatitis. Common adverse events were rash, myelosuppression, and constitutional symptoms. Among Ph-positive patients, 91% had received two or more approved tyrosine kinase inhibitors, and 51% had received all three approved tyrosine kinase inhibitors. Of 43 patients with chronic-phase CML, 98% had a complete hematologic response, 72% had a major cytogenetic response, and 44% had a major molecular response. Of 12 patients who had chronic-phase CML with the T315I mutation, 100% had a complete hematologic response and 92% had a major cytogenetic response. Of 13 patients with chronic-phase CML without detectable mutations, 100% had a complete hematologic response and 62% had a major cytogenetic response. Responses among patients with chronic-phase CML were durable. Of 22 patients with accelerated-phase or blast-phase CML or Ph-positive ALL, 36% had a major hematologic response and 32% had a major cytogenetic response.Conclusions: Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. (Funded by Ariad Pharmaceuticals and others; ClinicalTrials.gov number, NCT00660920.). [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
30. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors.
- Author
-
Demetri GD, von Mehren M, Blanke CD, Van den Abbeele AD, Eisenberg B, Roberts PJ, Heinrich MC, Tuveson DA, Singer S, Janicek M, Fletcher JA, Silverman SG, Silberman SL, Capdeville R, Kiese B, Peng B, Dimitrijevic S, Druker BJ, Corless C, and Fletcher CDM
- Published
- 2002
31. Long-term outcomes of imatinib treatment for chronic myeloid leukemia
- Author
-
Andreas, Hochhaus, Richard A, Larson, François, Guilhot, Jerald P, Radich, Susan, Branford, Timothy P, Hughes, Michele, Baccarani, Michael W, Deininger, Francisco, Cervantes, Satoko, Fujihara, Christine-Elke, Ortmann, Hans D, Menssen, Hagop, Kantarjian, Stephen G, O'Brien, Brian J, Druker, G, Verhoef, Hochhaus, Andreas, Larson, Richard A, Guilhot, François, Radich, Jerald P, Branford, Susan, Hughes, Timothy P, Baccarani, Michele, Deininger, Michael W, Cervantes, Francisco, Fujihara, Satoko, Ortmann, Christine Elke, Menssen, Hans D, Kantarjian, Hagop, O'Brien, Stephen G, Druker, Brian J, CCA - Cancer Treatment and quality of life, Hematology, Universitat de Barcelona, and Hochhaus A, Larson RA, Guilhot F, Radich JP, Branford S, Hughes TP, Baccarani M, Deininger MW, Cervantes F, Fujihara S, Ortmann CE, Menssen HD, Kantarjian H, O'Brien SG, Druker BJ, (plus IRIS Investigators: 180 collaborators), Martinelli G, et al.
- Subjects
0301 basic medicine ,Male ,Intention to Treat Analysi ,Antineoplastic Agent ,0302 clinical medicine ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Chronic ,CML ,Leukemia ,Cross-Over Studies ,Medicine (all) ,Cytarabine ,drug treatment ,Cytogenetic Analysi ,Enzyme inhibitors ,General Medicine ,Hematology ,Cross-Over Studie ,Middle Aged ,Intention to Treat Analysis ,Myeloid leukemia ,030220 oncology & carcinogenesis ,Cytogenetic Analysis ,Imatinib Mesylate ,Female ,Survival Analysi ,Human ,medicine.drug ,Adult ,medicine.medical_specialty ,Adolescent ,Leucèmia mieloide ,Protein Kinase Inhibitor ,Alpha interferon ,Antineoplastic Agents ,Follow-Up Studie ,03 medical and health sciences ,Young Adult ,Internal medicine ,Multicenter trial ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Humans ,Aged ,Follow-Up Studies ,Interferon-alpha ,Protein Kinase Inhibitors ,Survival Analysis ,Hematologia ,neoplasms ,Interferon alfa ,Antineoplastic Combined Chemotherapy Protocol ,Intention-to-treat analysis ,business.industry ,Imatinib ,Crossover study ,Surgery ,030104 developmental biology ,Imatinib mesylate ,Inhibidors enzimàtics ,BCR-ABL Positive ,business ,Myelogenous - Abstract
BACKGROUND Imatinib, a selective BCR-ABL1 kinase inhibitor, improved the prognosis for patients with chronic myeloid leukemia (CML). We conducted efficacy and safety analyses on the basis of more than 10 years of follow-up in patients with CML who were treated with imatinib as initial therapy. METHODS In this open-label, multicenter trial with crossover design, we randomly assigned patients with newly diagnosed CML in the chronic phase to receive either imatinib or interferon alfa plus cytarabine. Long-term analyses included overall survival, response to treatment, and serious adverse events. RESULTS The median follow-up was 10.9 years. Given the high rate of crossover among patients who had been randomly assigned to receive interferon alfa plus cytarabine (65.6%) and the short duration of therapy before crossover in these patients (median, 0.8 years), the current analyses focused on patients who had been randomly assigned to receive imatinib. Among the patients in the imatinib group, the estimated overall survival rate at 10 years was 83.3%. Approximately half the patients (48.3%) who had been randomly assigned to imatinib completed study treatment with imatinib, and 82.8% had a complete cytogenetic response. Serious adverse events that were considered by the investigators to be related to imatinib were uncommon and most frequently occurred during the first year of treatment. CONCLUSIONS Almost 11 years of follow-up showed that the efficacy of imatinib persisted over time and that long-term administration of imatinib was not associated with unacceptable cumulative or late toxic effects. (Funded by Novartis Pharmaceuticals; IRIS ClinicalTrials.gov numbers, NCT00006343 and NCT00333840.)
- Published
- 2017
- Full Text
- View/download PDF
32. Five-Year Follow-up of Patients Receiving Imatinib for Chronic Myeloid Leukemia
- Author
-
François Guilhot, Laurie Letvak, Richard A. Larson, Bayard L. Powell, Hermine Agis, Norbert Gattermann, Francisco Cervantes, Andreas Hochhaus, Brian J. Druker, Richard T. Silver, Hagop M. Kantarjian, Charlene So, Bengt Simonsson, Thea Kolsen Fischer, Jerald P. Radich, John D. Shepherd, Johan Lanng Nielsen, Alois Gratwohl, Jan J. Cornelissen, Insa Gathmann, Stephen G. O'Brien, Janice Gabrilove, Josy Reiffers, Michael W. Deininger, Kerry Taylor, Richard Stone, Michele Baccarani, Philippe Rousselot, Gregor Verhoef, John M. Goldman, Giuseppe Saglio, Timothy P. Hughes, Medical Oncology, Hematology, Druker BJ, Guilhot F, O'Brien SG, Gathmann I, Kantarjian H, Gattermann N, Deininger MW, Silver RT, Goldman JM, Stone RM, Cervantes F, Hochhaus A, Powell BL, Gabrilove JL, Rousselot P, Reiffers J, Cornelissen JJ, Hughes T, Agis H, Fischer T, Verhoef G, Shepherd J, Saglio G, Gratwohl A, Nielsen JL, Radich JP, Simonsson B, Taylor K, Baccarani M, So C, Letvak L, and Larson RA
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Fusion Proteins, bcr-abl ,Antineoplastic Agents ,Kaplan-Meier Estimate ,Chronic phase chronic myelogenous leukemia ,Disease-Free Survival ,Piperazines ,chemistry.chemical_compound ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Omacetaxine mepesuccinate ,medicine ,Humans ,neoplasms ,business.industry ,Ponatinib ,Cytarabine ,Interferon-alpha ,Myeloid leukemia ,Imatinib ,General Medicine ,Protein-Tyrosine Kinases ,Survival Analysis ,Survival Rate ,Dasatinib ,Pyrimidines ,Treatment Outcome ,Imatinib mesylate ,chemistry ,Nilotinib ,Benzamides ,Immunology ,Imatinib Mesylate ,Female ,business ,Follow-Up Studies ,medicine.drug - Abstract
The cause of chronic myeloid leukemia (CML) is a constitutively active BCR-ABL tyrosine kinase. Imatinib inhibits this kinase, and in a short-term study was superior to interferon alfa plus cytarabine for newly diagnosed CML in the chronic phase. For 5 years, we followed patients with CML who received imatinib as initial therapy.We randomly assigned 553 patients to receive imatinib and 553 to receive interferon alfa plus cytarabine and then evaluated them for overall and event-free survival; progression to accelerated-phase CML or blast crisis; hematologic, cytogenetic, and molecular responses; and adverse events.The median follow-up was 60 months. Kaplan-Meier estimates of cumulative best rates of complete cytogenetic response among patients receiving imatinib were 69% by 12 months and 87% by 60 months. An estimated 7% of patients progressed to accelerated-phase CML or blast crisis, and the estimated overall survival of patients who received imatinib as initial therapy was 89% at 60 months. Patients who had a complete cytogenetic response or in whom levels of BCR-ABL transcripts had fallen by at least 3 log had a significantly lower risk of disease progression than did patients without a complete cytogenetic response (P0.001). Grade 3 or 4 adverse events diminished over time, and there was no clinically significant change in the profile of adverse events.After 5 years of follow-up, continuous treatment of chronic-phase CML with imatinib as initial therapy was found to induce durable responses in a high proportion of patients. (ClinicalTrials.gov number, NCT00006343 [ClinicalTrials.gov].)
- Published
- 2006
- Full Text
- View/download PDF
33. e8a2 BCR–ABL: more frequent than other atypical BCR–ABL variants?
- Author
-
Mw Deininger, T Kegel, Susan Branford, Shadmehr Demehri, Giovanni Martinelli, Lc Lim, Peter Paschka, Thoralf Lange, T Koizumi, Beate Schultheis, Bj Druker, Andreas Hochhaus, Takeshi Sugimoto, DEMEHRI S, PASCHKA P, SCHULTHEIS B, LANGE T, KOIZUMI T, SUGIMOTO T, BRANFORD S, LIM LC, KEGEL T, MARTINELLI G., HOCHHAUS A, DRUKER BJ, and DEININGER MW
- Subjects
Cancer Research ,CHIMERIC GENE ,Imatinib ,Hematology ,Chimeric gene ,Biology ,medicine.disease ,IMATINIB ,Fusion protein ,E8/A2 ,Leukemia ,Myelogenous ,Oncology ,medicine ,Cancer research ,LEUKEMIA ,BCR/ABL ,medicine.drug - Published
- 2005
- Full Text
- View/download PDF
34. The prognosis for patients with chronic myeloid leukemia who have clonal cytogenetic abnormalities in philadelphia chromosome-negative cells
- Author
-
Michael W. Deininger, Dietger Niederwieser, Byung Park, Douglas R. Smith, Michele Baccarani, Brian J. Druker, François Guilhot, Insa Gathmann, Thomas Fischer, Andreas Hochhaus, Richard Stone, John M. Goldman, Hagop M. Kantarjian, Jorge E. Cortes, Charlene So, Carlo Gambacorti-Passerini, Ron Paquette, Deininger, M, Cortes, J, Paquette, R, Park, B, Hochhaus, A, Baccarani, M, Stone, R, Fischer, T, Kantarjian, H, Niederwieser, D, GAMBACORTI PASSERINI, C, So, C, Gathmann, I, Goldman, J, Smith, D, Druker, B, Guilhot, F, Deininger MW, Cortes J, Paquette R, Park B, Hochhaus A, Baccarani M, Stone R, Fischer T, Kantarjian H, Niederwieser D, Gambacorti-Passerini C, So C, Gathmann I, Goldman JM, Smith D, Druker BJ, and Guilhot F.
- Subjects
Oncology ,Male ,Cancer Research ,Myeloid ,Kaplan-Meier Estimate ,Piperazines ,hemic and lymphatic diseases ,Treatment Failure ,Aged, 80 and over ,Myeloid leukemia ,Middle Aged ,Prognosis ,Leukemia ,medicine.anatomical_structure ,Treatment Outcome ,Benzamides ,Cytogenetic Analysis ,Imatinib Mesylate ,Female ,medicine.drug ,Adult ,medicine.medical_specialty ,Neutropenia ,Antineoplastic Agents ,Philadelphia chromosome ,Disease-Free Survival ,Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative ,chronic myeloid leukemia ,Internal medicine ,parasitic diseases ,medicine ,Humans ,Aged ,Chromosome Aberrations ,Chi-Square Distribution ,business.industry ,Myelodysplastic syndromes ,Cancer ,Interferon-alpha ,Imatinib ,medicine.disease ,Thrombocytopenia ,Imatinib mesylate ,Logistic Models ,Pyrimidines ,Myelodysplastic Syndromes ,Chronic Disease ,Cancer research ,business ,Follow-Up Studies - Abstract
BACKGROUND. Clonal cytogenetic abnormalities (CCA) were detected in Philadelphia chromosome (Ph)-negative cells in some patients with chronic myeloid leukemia (CML) who attained a cytogenetic response to imatinib mesylate. In some patients, CCA/Ph-negative status was associated with myelodysplasia or acute myeloid leukemia. The objective of the current study was to determine the prognostic impact of CCA/Ph-negative cells. METHODS. The authors compared the pretherapeutic risk factors (Kruskall-Wallis test), exposure to cytotoxic drugs (chi-square test), and overall and progression-free survival (Kaplan-Meyer and logistic regression analysis, respectively) of 515 patients with mostly chronic-phase CML who were treated with imatinib mesylate after failure of interferon-α according to whether they attained a major cytogenetic response (MCR) (n = 324 patients), an MCR with CCA/Ph-negative status (n = 30 patients), or no MCR (n = 161 patients). RESULTS. CCA/Ph-negative status most frequently involved chromosomes Y, 8, and 7. No significant differences in pretherapeutic risk factors were detected between patients who attained an MCR with and without CCA/Ph-negative cells, except that exposure to alkylating agents was more frequent in patients with CCA/Ph-negative cells, and overall and progression-free survival were identical. With a median follow-up of 51 months, only 2 patients developed myelodysplastic syndromes (MDS). CONCLUSIONS. The overall prognosis for patients who had CML with CCA/Ph-negative status was good and was driven by the CML response to imatinib mesylate. Isolated CCA/Ph-negative cells in the absence of morphologic evidence of MDS do not justify a change in therapy. Cancer 2007. © 2007 American Cancer Society.
- Published
- 2007
35. Monitoring CML patients responding to treatment with tyrosine kinase inhibitors: review and recommendations for harmonizing current methodology for detecting BCR-ABL transcripts and kinase domain mutations and for expressing results
- Author
-
Jeffrey H. Lipton, David Grimwade, Jaspal Kaeda, Giuseppe Saglio, Janina A. Longtine, Brian J. Druker, Timothy P. Hughes, Andreas Hochhaus, Nicholas C.P. Cross, Giovanni Martinelli, Suzanne Kamel-Reid, Michael W. Deininger, Simona Soverini, Rüdiger Hehlmann, Michele Baccarani, John M. Goldman, Wendy Stock, Jerald P. Radich, Susan Branford, Jean Gabert, Jorge E. Cortes, Hughes T, Deininger M, Hochhaus A, Branford S, Radich J, Kaeda J, Baccarani M, Cortes J, Cross NC, Druker BJ, Gabert J, Grimwade D, Hehlmann R, Kamel-Reid S, Lipton JH, Longtine J, Martinelli G, Saglio G, Soverini S, Stock W, and Goldman JM.
- Subjects
medicine.medical_specialty ,Immunology ,Fusion Proteins, bcr-abl ,Biology ,Biochemistry ,Sensitivity and Specificity ,hemic and lymphatic diseases ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Humans ,IMATINIB MESYLATE (IM) ,Protein Kinase Inhibitors ,medicine.diagnostic_test ,Reverse Transcriptase Polymerase Chain Reaction ,Cytogenetics ,Myeloid leukemia ,Cell Biology ,Hematology ,Protein-Tyrosine Kinases ,Review in Translational Hematology ,medicine.disease ,Protein Structure, Tertiary ,BCR-ABL TRANSCRIPTS ,Leukemia ,Real-time polymerase chain reaction ,Imatinib mesylate ,medicine.anatomical_structure ,Mutation ,Cancer research ,Bone marrow ,Drug Monitoring ,CHRONIC MYELOID LEUKEMIA (CML) ,Tyrosine kinase ,Fluorescence in situ hybridization - Abstract
The introduction in 1998 of imatinib mesylate (IM) revolutionized management of patients with chronic myeloid leukemia (CML) and the second generation of tyrosine kinase inhibitors may prove superior to IM. Real-time quantitative polymerase chain reaction (RQ-PCR) provides an accurate measure of the total leukemiacell mass and the degree to which BCR-ABL transcripts are reduced by therapy correlates with progression-free survival. Because a rising level of BCR-ABL is an early indication of loss of response and thus the need to reassess therapeutic strategy, regular molecular monitoring of individual patients is clearly desirable. Here we summarize the results of a consensus meeting that took place at the National Institutes of Health (NIH) in Bethesda in October 2005. We make suggestions for (1) harmonizing the differing methodologies for measuring BCR-ABL transcripts in patients with CML undergoing treatment and using a conversion factor whereby individual laboratories can express BCR-ABL transcript levels on an internationally agreed scale; (2) using serial RQ-PCR results rather than bone marrow cytogenetics or fluorescence in situ hybridization (FISH) for the BCR-ABL gene to monitor individual patients responding to treatment; and (3) detecting and reporting Philadelphia (Ph) chromosome-positive subpopulations bearing BCR-ABL kinase domain mutations. We recognize that our recommendations are provisional and will require revision as new evidence emerges. (Blood. 2006;108:28-37)
- Published
- 2006
36. Beat-AML 2024 ELN-refined risk stratification for older adults with newly diagnosed AML given lower-intensity therapy.
- Author
-
Hoff FW, Blum WG, Huang Y, Welkie RL, Swords RT, Traer E, Stein EM, Lin TL, Archer KJ, Patel PA, Collins RH, Baer MR, Duong VH, Arellano ML, Stock W, Odenike O, Redner RL, Kovacsovics T, Deininger MW, Zeidner JF, Olin RL, Smith CC, Foran JM, Schiller GJ, Curran EK, Koenig KL, Heerema NA, Chen T, Martycz M, Stefanos M, Marcus SG, Rosenberg L, Druker BJ, Levine RL, Burd A, Yocum AO, Borate UM, Mims AS, Byrd JC, and Madanat YF
- Subjects
- Humans, Aged, Female, Male, Middle Aged, Aged, 80 and over, Prognosis, Risk Assessment, Mutation, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute diagnosis
- Abstract
Abstract: Although the 2022 European LeukemiaNet (ELN) acute myeloid leukemia (AML) risk classification reliably predicts outcomes in younger patients treated with intensive chemotherapy, it is unclear whether it applies to adults ≥60 years treated with lower-intensity treatment (LIT). We aimed to test the prognostic impact of ELN risk in patients with newly diagnosed (ND) AML aged ≥60 years given LIT and to further refine risk stratification for these patients. A total of 595 patients were included: 11% had favorable-, 11% intermediate-, and 78% had adverse-risk AML. ELN risk was prognostic for overall survival (OS) (P < .001) but did not stratify favorable- from intermediate-risk (P = .71). Within adverse-risk AML, the impact of additional molecular abnormalities was further evaluated. Multivariable analysis was performed on a training set (n = 316) and identified IDH2 mutation as an independent favorable prognostic factor, and KRAS, MLL2, and TP53 mutations as unfavorable (P < .05). A "mutation score" was calculated for each combination of these mutations, assigning adverse-risk patients to 2 risk groups: -1 to 0 points ("Beat-AML intermediate") vs 1+ points ("Beat-AML adverse"). In the final refined risk classification, ELN favorable- and intermediate-risk were combined into a newly defined "Beat-AML favorable-risk" group, in addition to mutation scoring within the ELN adverse-risk group. This approach redefines risk for older patients with ND AML and proposes refined Beat-AML risk groups with improved discrimination for OS (2-year OS, 48% vs 33% vs 11%, respectively; P < .001), providing patients and providers additional information for treatment decision-making., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
37. Overcoming clinical BCR-ABL1 compound mutant resistance with combined ponatinib and asciminib therapy.
- Author
-
Eide CA, Brewer D, Xie T, Schultz AR, Savage SL, Muratcioglu S, Merz N, Press RD, O'Hare T, Jacob T, Vu TQ, Tognon CE, Macey TA, Kuriyan J, Kalodimos CG, and Druker BJ
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Male, Female, Niacinamide analogs & derivatives, Pyrazoles, Pyridazines therapeutic use, Pyridazines pharmacology, Fusion Proteins, bcr-abl genetics, Fusion Proteins, bcr-abl antagonists & inhibitors, Imidazoles pharmacology, Imidazoles therapeutic use, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Drug Resistance, Neoplasm genetics, Drug Resistance, Neoplasm drug effects, Mutation, Protein Kinase Inhibitors therapeutic use, Protein Kinase Inhibitors pharmacology
- Abstract
BCR-ABL1 compound mutations can lead to resistance to ABL1 inhibitors in chronic myeloid leukemia (CML), which could be targeted by combining the ATP-site inhibitor ponatinib and the allosteric inhibitor asciminib. Here, we report the clinical validation of this approach in a CML patient, providing a basis for combination therapy to overcome such resistance., Competing Interests: Declaration of interests B.J.D. serves on scientific advisory boards for Aileron Therapeutics, Therapy Architects (ALLCRON), Cepheid, Vivid Biosciences, Celgene, RUNX1 Research Program, Novartis, Gilead Sciences (inactive), and Monojul (inactive); serves on scientific advisory boards and receives stock from Aptose Biosciences, Blueprint Medicines, EnLiven Therapeutics, Iterion Therapeutics, Third Coast Therapeutics, and GRAIL (inactive on scientific advisory board); is scientific founder of MolecularMD (inactive, acquired by ICON); serves on the board of directors and receives stock from Amgen and Vincera Pharma; serves on the board of directors for Burroughs Wellcome Fund and CureOne; serves on the joint steering committee for Beat AML LLS; is founder of VB Therapeutics; has a sponsored research agreement with EnLiven Therapeutics; receives clinical trial funding from Novartis, Bristol-Myers Squibb, and Pfizer., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
38. Mass Spectrometry-Based Proteogenomics: New Therapeutic Opportunities for Precision Medicine.
- Author
-
Joshi SK, Piehowski P, Liu T, Gosline SJC, McDermott JE, Druker BJ, Traer E, Tyner JW, Agarwal A, Tognon CE, and Rodland KD
- Subjects
- Humans, Proteomics, Genomics, Mass Spectrometry, Precision Medicine, Proteogenomics
- Abstract
Proteogenomics refers to the integration of comprehensive genomic, transcriptomic, and proteomic measurements from the same samples with the goal of fully understanding the regulatory processes converting genotypes to phenotypes, often with an emphasis on gaining a deeper understanding of disease processes. Although specific genetic mutations have long been known to drive the development of multiple cancers, gene mutations alone do not always predict prognosis or response to targeted therapy. The benefit of proteogenomics research is that information obtained from proteins and their corresponding pathways provides insight into therapeutic targets that can complement genomic information by providing an additional dimension regarding the underlying mechanisms and pathophysiology of tumors. This review describes the novel insights into tumor biology and drug resistance derived from proteogenomic analysis while highlighting the clinical potential of proteogenomic observations and advances in technique and analysis tools.
- Published
- 2024
- Full Text
- View/download PDF
39. A study to assess the efficacy of enasidenib and risk-adapted addition of azacitidine in newly diagnosed IDH2-mutant AML.
- Author
-
Cai SF, Huang Y, Lance JR, Mao HC, Dunbar AJ, McNulty SN, Druley T, Li Y, Baer MR, Stock W, Kovacsovics T, Blum WG, Schiller GJ, Olin RL, Foran JM, Litzow M, Lin T, Patel P, Foster MC, Boyiadzis M, Collins RH, Chervin J, Shoben A, Vergilio JA, Heerema NA, Rosenberg L, Chen TL, Yocum AO, Druggan F, Marcus S, Stefanos M, Druker BJ, Mims AS, Borate U, Burd A, Byrd JC, Levine RL, and Stein EM
- Subjects
- Humans, Isocitrate Dehydrogenase genetics, Mutation, Pathologic Complete Response, Azacitidine adverse effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Aminopyridines, Triazines
- Abstract
Abstract: Enasidenib (ENA) is an inhibitor of isocitrate dehydrogenase 2 (IDH2) approved for the treatment of patients with IDH2-mutant relapsed/refractory acute myeloid leukemia (AML). In this phase 2/1b Beat AML substudy, we applied a risk-adapted approach to assess the efficacy of ENA monotherapy for patients aged ≥60 years with newly diagnosed IDH2-mutant AML in whom genomic profiling demonstrated that mutant IDH2 was in the dominant leukemic clone. Patients for whom ENA monotherapy did not induce a complete remission (CR) or CR with incomplete blood count recovery (CRi) enrolled in a phase 1b cohort with the addition of azacitidine. The phase 2 portion assessing the overall response to ENA alone demonstrated efficacy, with a composite complete response (cCR) rate (CR/CRi) of 46% in 60 evaluable patients. Seventeen patients subsequently transitioned to phase 1b combination therapy, with a cCR rate of 41% and 1 dose-limiting toxicity. Correlative studies highlight mechanisms of clonal elimination with differentiation therapy as well as therapeutic resistance. This study demonstrates both efficacy of ENA monotherapy in the upfront setting and feasibility and applicability of a risk-adapted approach to the upfront treatment of IDH2-mutant AML. This trial is registered at www.clinicaltrials.gov as #NCT03013998., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
40. Mapping the proteogenomic landscape enables prediction of drug response in acute myeloid leukemia.
- Author
-
Pino JC, Posso C, Joshi SK, Nestor M, Moon J, Hansen JR, Hutchinson-Bunch C, Gritsenko MA, Weitz KK, Watanabe-Smith K, Long N, McDermott JE, Druker BJ, Liu T, Tyner JW, Agarwal A, Traer E, Piehowski PD, Tognon CE, Rodland KD, and Gosline SJC
- Subjects
- Humans, Proteomics methods, Genomics methods, Mutation, Proteogenomics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism
- Abstract
Acute myeloid leukemia is a poor-prognosis cancer commonly stratified by genetic aberrations, but these mutations are often heterogeneous and fail to consistently predict therapeutic response. Here, we combine transcriptomic, proteomic, and phosphoproteomic datasets with ex vivo drug sensitivity data to help understand the underlying pathophysiology of AML beyond mutations. We measure the proteome and phosphoproteome of 210 patients and combine them with genomic and transcriptomic measurements to identify four proteogenomic subtypes that complement existing genetic subtypes. We build a predictor to classify samples into subtypes and map them to a "landscape" that identifies specific drug response patterns. We then build a drug response prediction model to identify drugs that target distinct subtypes and validate our findings on cell lines representing various stages of quizartinib resistance. Our results show how multiomics data together with drug sensitivity data can inform therapy stratification and drug combinations in AML., Competing Interests: Declaration of interests J.W.T. has received research support from Agios, Aptose, Array, AstraZeneca, Constellation, Genentech, Gilead, Incyte, Janssen, Petra, Seattle Genetics, Syros, and Takeda and is on the Scientific Advisory Board of Recludix. C.E.T. has received support from Notable Labs. B.J.D. is/was on the Scientific Advisory Board of Adela Bio, Aileron Therapeutics (inactive), Celgene, Cepheid, DNA SEQ, Nemucore Medical Innovations, Novartis, RUNX1 Research Program, Therapy Architects/ALLCRON (inactive), and Vivid Biosciences (inactive); is/was on the Scientific Advisory Board of and owns/owned stock in Aptose Biosciences, Blueprint Medicines, Enliven Therapeutics, GRAIL, Iterion Therapeutics, and Recludix Pharma; is/was on the Board of Directors and owns/owned stock in Amgen and Vincerx Pharma; is/was on the Board of Directors of Burroughs Wellcome Fund, CureOne; is/was on the Joint Steering Committee of Beat AML (Leukemia & Lymphoma Society); is/was on the Advisory Committee of Multicancer Early Detection Consortium; is the Founder of VB Therapeutics; has/had sponsored research agreements with Enliven Therapeutics and Recludix Pharma; receives/received clinical trial funding from AstraZeneca and Novartis; receives/received royalties from Patent 6958335 (Novartis exclusive license), the Oregon Health & Science University, and the Dana-Farber Cancer Institute (one Merck exclusive license, one CytoImage, Inc. exclusive license, and one Sun Pharma Advanced Research Company nonexclusive license); and holds US Patents 4326534, 6958335, 7416873, 7592142, 10473667, 10664967, and 11049247., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
41. The FLT3 N701K mutation causes clinical AML resistance to gilteritinib and triggers TKI sensitivity switch to quizartinib.
- Author
-
Joshi SK, Pittsenbarger J, Kennedy VE, Peretz CAC, Perl AE, Smith CC, Tyner JW, Druker BJ, and Traer E
- Subjects
- Humans, Mutation, Aniline Compounds therapeutic use, fms-Like Tyrosine Kinase 3 genetics, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Published
- 2023
- Full Text
- View/download PDF
42. Kinase Mutations and Imatinib Response in Patients With Metastatic Gastrointestinal Stromal Tumor.
- Author
-
Heinrich MC, Corless CL, Demetri GD, Blanke CD, von Mehren M, Joensuu H, McGreevey LS, Chen CJ, Van den Abbeele AD, Druker BJ, Kiese B, Eisenberg B, Roberts PJ, Singer S, Fletcher CDM, Silberman S, Dimitrijevic S, and Fletcher JA
- Abstract
Purpose: Most gastrointestinal stromal tumors (GISTs) express constitutively activated mutant isoforms of KIT or kinase platelet-derived growth factor receptor alpha (PDGFRA) that are potential therapeutic targets for imatinib mesylate. The relationship between mutations in these kinases and clinical response to imatinib was examined in a group of patients with advanced GIST., Patients and Methods: GISTs from 127 patients enrolled onto a phase II clinical study of imatinib were examined for mutations of KIT or PDGFRA . Mutation types were correlated with clinical outcome., Results: Activating mutations of KIT or PDGFRA were found in 112 (88.2%) and six (4.7%) GISTs, respectively. Most KIT mutations involved exon 9 (n = 23) or exon 11 (n = 85). All KIT mutant isoforms, but only a subset of PDGFRA mutant isoforms, were sensitive to imatinib, in vitro. In patients with GISTs harboring exon 11 KIT mutations, the partial response rate (PR) was 83.5%, whereas patients with tumors containing an exon 9 KIT mutation or no detectable mutation of KIT or PDGFRA had PR rates of 47.8% ( P = .0006) and 0.0% ( P < .0001), respectively. Patients whose tumors contained exon 11 KIT mutations had a longer event-free and overall survival than those whose tumors expressed either exon 9 KIT mutations or had no detectable kinase mutation., Conclusion: Activating mutations of KIT or PDGFRA are found in the vast majority of GISTs, and the mutational status of these oncoproteins is predictive of clinical response to imatinib. PDGFRA mutations can explain response and sensitivity to imatinib in some GISTs lacking KIT mutations.
- Published
- 2023
- Full Text
- View/download PDF
43. Clinical Correlates of Venetoclax-Based Combination Sensitivities to Augment Acute Myeloid Leukemia Therapy.
- Author
-
Eide CA, Kurtz SE, Kaempf A, Long N, Joshi SK, Nechiporuk T, Huang A, Dibb CA, Taylor A, Bottomly D, McWeeney SK, Minnier J, Lachowiez CA, Saultz JN, Swords RT, Agarwal A, Chang BH, Druker BJ, and Tyner JW
- Subjects
- Humans, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, Azacitidine pharmacology, Azacitidine therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use
- Abstract
The BCL2 inhibitor venetoclax combined with the hypomethylating agent azacytidine shows significant clinical benefit in a subset of patients with acute myeloid leukemia (AML); however, resistance limits response and durability. We prospectively profiled the ex vivo activity of 25 venetoclax-inclusive combinations on primary AML patient samples to identify those with improved potency and synergy compared with venetoclax + azacytidine (Ven + azacytidine). Combination sensitivities correlated with tumor cell state to discern three patterns: primitive selectivity resembling Ven + azacytidine, monocytic selectivity, and broad efficacy independent of cell state. Incorporation of immunophenotype, mutation, and cytogenetic features further stratified combination sensitivity for distinct patient subtypes. We dissect the biology underlying the broad, cell state-independent efficacy for the combination of venetoclax plus the JAK1/2 inhibitor ruxolitinib. Together, these findings support opportunities for expanding the impact of venetoclax-based drug combinations in AML by leveraging clinical and molecular biomarkers associated with ex vivo responses., Significance: By mapping drug sensitivity data to clinical features and tumor cell state, we identify novel venetoclax combinations targeting patient subtypes who lack sensitivity to Ven + azacytidine. This provides a framework for a taxonomy of AML informed by readily available sets of clinical and genetic features obtained as part of standard care. See related commentary by Becker, p. 437 . This article is featured in Selected Articles from This Issue, p. 419., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
44. Risk of progression in chronic phase-chronic myeloid leukemia patients eligible for tyrosine kinase inhibitor discontinuation: Final analysis of the TFR-PRO study.
- Author
-
Zambrotta GPM, Nicolini FE, Assouline S, Busque L, Pungolino E, Abruzzese E, Miggiano MC, Elena C, Alvarez-Larran A, Triguero A, Iurlo A, Bucelli C, Cerrano M, Capodanno I, Lunghi F, le Coutre P, Galimberti S, Caocci G, Maffioli M, Stagno F, Saussele S, Piazza R, Druker BJ, Fava C, Guglielmana V, Colombo F, Antolini L, and Gambacorti-Passerini C
- Abstract
Disease progression to accelerated/blast phase (AP/BP) in patients with chronic phase chronic myeloid leukemia (CP-CML) after treatment discontinuation (TD) has never been systematically reported in clinical trials. However, recent reports of several such cases has raised concern. To estimate the risk of AP/BP among TD-eligible patients, we conducted TFR-PRO, a cohort retro-prospective study: 870 CP-CML patients eligible for TD formed a discontinuation cohort (505 patients) and a reference one (365 patients). The primary objective was the time adjusted rate (TAR) of progression in relation to TD. Secondary endpoints included the TAR of molecular relapse, that is, loss of major molecular response (MMR). With a median follow up of 5.5 years and 5188.2 person-years available, no events occurred in the TD cohort. One event of progression was registered 55 months after the end of TD, when the patient was contributing to the reference cohort. The TAR of progression was 0.019/100 person-years (95% CI [0.003-0.138]) in the overall group; 0.0 (95% CI [0-0.163]) in the discontinuation cohort; and 0.030 (95% CI [0.004-0.215]) in the reference cohort. These differences are not statistically significant. Molecular relapses occurred in 172/505 (34.1%) patients after TD, and in 64/365 (17.5%) patients in the reference cohort, p < .0001. Similar rates were observed in TD patients in first, second or third line of treatment. CML progression in patients eligible for TD is rare and not related to TD. Fears about the risk of disease progression among patients attempting TD should be dissipated., (© 2023 The Authors. American Journal of Hematology published by Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
45. Patient-Specific Targeting of the T-Cell Receptor Variable Region as a Therapeutic Strategy in Clonal T-Cell Diseases.
- Author
-
Lucero OM, Lee JA, Bowman J, Johnson K, Sapparapu G, Thomas JK, Fan G, Chang BH, Thiel-Klare K, Eide CA, Okada C, Palazzolo M, Lind E, Kosaka Y, Druker BJ, Lydon N, and Bowers PM
- Subjects
- Humans, Mice, Animals, Rituximab, T-Lymphocytes immunology, Receptors, Antigen, T-Cell, alpha-beta genetics, Receptors, Antigen, T-Cell, alpha-beta immunology, Receptors, Antigen, T-Cell genetics, Lymphoma, T-Cell
- Abstract
Purpose: Targeted therapeutics are a goal of medicine. Methods for targeting T-cell lymphoma lack specificity for the malignant cell, leading to elimination of healthy cells. The T-cell receptor (TCR) is designed for antigen recognition. T-cell malignancies expand from a single clone that expresses one of 48 TCR variable beta (Vβ) genes, providing a distinct therapeutic target. We hypothesized that a mAb that is exclusive to a specific Vβ would eliminate the malignant clone while having minimal effects on healthy T cells., Experimental Design: We identified a patient with large granular T-cell leukemia and sequenced his circulating T-cell population, 95% of which expressed Vβ13.3. We developed a panel of anti-Vβ13.3 antibodies to test for binding and elimination of the malignant T-cell clone., Results: Therapeutic antibody candidates bound the malignant clone with high affinity. Antibodies killed engineered cell lines expressing the patient TCR Vβ13.3 by antibody-dependent cellular cytotoxicity and TCR-mediated activation-induced cell death, and exhibited specific killing of patient malignant T cells in combination with exogenous natural killer cells. EL4 cells expressing the patient's TCR Vβ13.3 were also killed by antibody administration in an in vivo murine model., Conclusions: This approach serves as an outline for development of therapeutics that can treat clonal T-cell-based malignancies and potentially other T-cell-mediated diseases. See related commentary by Varma and Diefenbach, p. 4024., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
46. Entospletinib with decitabine in acute myeloid leukemia with mutant TP53 or complex karyotype: A phase 2 substudy of the Beat AML Master Trial.
- Author
-
Duong VH, Ruppert AS, Mims AS, Borate U, Stein EM, Baer MR, Stock W, Kovacsovics T, Blum W, Arellano ML, Schiller GJ, Olin RL, Foran JM, Litzow MR, Lin TL, Patel PA, Foster MC, Redner RL, Al-Mansour Z, Cogle CR, Swords RT, Collins RH, Vergilio JA, Heerema NA, Rosenberg L, Yocum AO, Marcus S, Chen T, Druggan F, Stefanos M, Gana TJ, Shoben AB, Druker BJ, Burd A, Byrd JC, Levine RL, and Boyiadzis MM
- Subjects
- Humans, Decitabine, Karyotype, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols adverse effects, Tumor Suppressor Protein p53 genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology
- Abstract
Background: Patients with acute myeloid leukemia (AML) who have tumor protein p53 (TP53) mutations or a complex karyotype have a poor prognosis, and hypomethylating agents are often used. The authors evaluated the efficacy of entospletinib, an oral inhibitor of spleen tyrosine kinase, combined with decitabine in this patient population., Methods: This was a multicenter, open-label, phase 2 substudy of the Beat AML Master Trial (ClinicalTrials.gov identifier NCT03013998) using a Simon two-stage design. Eligible patients aged 60 years or older who had newly diagnosed AML with mutations in TP53 with or without a complex karyotype (cohort A; n = 45) or had a complex karyotype without TP53 mutation (cohort B; n = 13) received entospletinib 400 mg twice daily with decitabine 20 mg/m
2 on days 1-10 every 28 days for up to three induction cycles, followed by up to 11 consolidation cycles, in which decitabine was reduced to days 1-5. Entospletinib maintenance was given for up to 2 years. The primary end point was complete remission (CR) and CR with hematologic improvement by up to six cycles of therapy., Results: The composite CR rates for cohorts A and B were 13.3% (95% confidence interval, 5.1%-26.8%) and 30.8% (95% confidence interval, 9.1%-61.4%), respectively. The median duration of response was 7.6 and 8.2 months, respectively, and the median overall survival was 6.5 and 11.5 months, respectively. The study was stopped because the futility boundary was crossed in both cohorts., Conclusions: The combination of entospletinib and decitabine demonstrated activity and was acceptably tolerated in this patient population; however, the CR rates were low, and overall survival was short. Novel treatment strategies for older patients with TP53 mutations and complex karyotype remain an urgent need., (© 2023 American Cancer Society.)- Published
- 2023
- Full Text
- View/download PDF
47. Disruption of the MYC Superenhancer Complex by Dual Targeting of FLT3 and LSD1 in Acute Myeloid Leukemia.
- Author
-
Yashar WM, Curtiss BM, Coleman DJ, VanCampen J, Kong G, Macaraeg J, Estabrook J, Demir E, Long N, Bottomly D, McWeeney SK, Tyner JW, Druker BJ, Maxson JE, and Braun TP
- Subjects
- Humans, Apoptosis, Histone Demethylases genetics, Histone Demethylases metabolism, Mutation, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, STAT5 Transcription Factor metabolism, fms-Like Tyrosine Kinase 3 genetics, fms-Like Tyrosine Kinase 3 metabolism, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism
- Abstract
Mutations in Fms-like tyrosine kinase 3 (FLT3) are common drivers in acute myeloid leukemia (AML) yet FLT3 inhibitors only provide modest clinical benefit. Prior work has shown that inhibitors of lysine-specific demethylase 1 (LSD1) enhance kinase inhibitor activity in AML. Here we show that combined LSD1 and FLT3 inhibition induces synergistic cell death in FLT3-mutant AML. Multi-omic profiling revealed that the drug combination disrupts STAT5, LSD1, and GFI1 binding at the MYC blood superenhancer, suppressing superenhancer accessibility as well as MYC expression and activity. The drug combination simultaneously results in the accumulation of repressive H3K9me1 methylation, an LSD1 substrate, at MYC target genes. We validated these findings in 72 primary AML samples with the nearly every sample demonstrating synergistic responses to the drug combination. Collectively, these studies reveal how epigenetic therapies augment the activity of kinase inhibitors in FLT3-ITD (internal tandem duplication) AML., Implications: This work establishes the synergistic efficacy of combined FLT3 and LSD1 inhibition in FLT3-ITD AML by disrupting STAT5 and GFI1 binding at the MYC blood-specific superenhancer complex., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
48. Pevonedistat with azacitidine in older patients with TP53-mutated AML: a phase 2 study with laboratory correlates.
- Author
-
Saliba AN, Kaufmann SH, Stein EM, Patel PA, Baer MR, Stock W, Deininger M, Blum W, Schiller GJ, Olin RL, Litzow MR, Lin TL, Ball BJ, Boyiadzis MM, Traer E, Odenike O, Arellano ML, Walker A, Duong VH, Kovacsovics T, Collins RH, Shoben AB, Heerema NA, Foster MC, Peterson KL, Schneider PA, Martycz M, Gana TJ, Rosenberg L, Marcus S, Yocum AO, Chen T, Stefanos M, Mims AS, Borate U, Burd A, Druker BJ, Levine RL, Byrd JC, and Foran JM
- Subjects
- Humans, Aged, Cyclopentanes, Pyrimidines, Tumor Suppressor Protein p53 genetics, Azacitidine therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Published
- 2023
- Full Text
- View/download PDF
49. Molecular response in newly diagnosed chronic-phase chronic myeloid leukemia: prediction modeling and pathway analysis.
- Author
-
Radich JP, Wall M, Branford S, Campbell CD, Chaturvedi S, DeAngelo DJ, Deininger M, Guinney J, Hochhaus A, Hughes TP, Kantarjian HM, Larson RA, Li S, Maegawa R, Mishra K, Obourn V, Pinilla-Ibarz J, Purkayastha D, Sadek I, Saglio G, Shrestha A, White BS, and Druker BJ
- Subjects
- Humans, Fusion Proteins, bcr-abl genetics, Protein Kinase Inhibitors adverse effects, Treatment Outcome, Antineoplastic Agents pharmacology, Leukemia, Myeloid, Chronic-Phase, Leukemia, Myelogenous, Chronic, BCR-ABL Positive diagnosis, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics
- Abstract
Tyrosine kinase inhibitor therapy revolutionized chronic myeloid leukemia treatment and showed how targeted therapy and molecular monitoring could be used to substantially improve survival outcomes. We used chronic myeloid leukemia as a model to understand a critical question: why do some patients have an excellent response to therapy, while others have a poor response? We studied gene expression in whole blood samples from 112 patients from a large phase III randomized trial (clinicaltrials gov. Identifier: NCT00471497), dichotomizing cases into good responders (BCR::ABL1 ≤10% on the International Scale by 3 and 6 months and ≤0.1% by 12 months) and poor responders (failure to meet these criteria). Predictive models based on gene expression demonstrated the best performance (area under the curve =0.76, standard deviation =0.07). All of the top 20 pathways overexpressed in good responders involved immune regulation, a finding validated in an independent data set. This study emphasizes the importance of pretreatment adaptive immune response in treatment efficacy and suggests biological pathways that can be targeted to improve response.
- Published
- 2023
- Full Text
- View/download PDF
50. Comparison and validation of the 2022 European LeukemiaNet guidelines in acute myeloid leukemia.
- Author
-
Lachowiez CA, Long N, Saultz J, Gandhi A, Newell LF, Hayes-Lattin B, Maziarz RT, Leonard J, Bottomly D, McWeeney S, Dunlap J, Press R, Meyers G, Swords R, Cook RJ, Tyner JW, Druker BJ, and Traer E
- Subjects
- Humans, Risk Factors, Mutation, Cytogenetics, Induction Chemotherapy, Leukemia, Myeloid, Acute genetics
- Abstract
Risk stratification in acute myeloid leukemia (AML) remains principle in survival prognostication and treatment selection. The 2022 European LeukemiaNet (ELN) recommendations were recently published, with notable updates to risk group assignment. The complexity of risk stratification and comparative outcomes between the 2022 and 2017 ELN guidelines remains unknown. This comparative analysis evaluated outcomes between the 2017 and 2022 ELN criteria in patients enrolled within the multicenter Beat AML cohort. Five hundred thirteen patients were included. Most patients had 1 or 2 ELN risk-defining abnormalities. In patients with ≥2 ELN risk-defining mutations, 44% (n = 132) had mutations spanning multiple ELN risk categories. Compared with ELN 2017 criteria, the updated ELN 2022 guidelines changed the assigned risk group in 15% of patients, including 10%, 26%, and 6% of patients categorized as being at ELN 2017 favorable-, intermediate-, and adverse-risk, respectively. The median overall survival across ELN 2022 favorable-, intermediate-, and adverse-risk groups was not reached, 16.8, and 9.7 months, respectively. The ELN 2022 guidelines more accurately stratified survival between patients with intermediate- or adverse-risk AML treated with induction chemotherapy compared with ELN 2017 guidelines. The updated ELN 2022 guidelines better stratify survival between patients with intermediate- or adverse-risk AML treated with induction chemotherapy. The increased complexity of risk stratification with inclusion of additional cytogenetic and molecular aberrations necessitates clinical workflows simplifying risk stratification., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.