73 results on '"Roy M Pollock"'
Search Results
2. Preclinical Evidence of Anti-Tumor Activity Induced by EZH2 Inhibition in Human Models of Synovial Sarcoma.
- Author
-
Satoshi Kawano, Alexandra R Grassian, Masumi Tsuda, Sarah K Knutson, Natalie M Warholic, Galina Kuznetsov, Shanqin Xu, Yonghong Xiao, Roy M Pollock, Jesse S Smith, Kevin K Kuntz, Scott Ribich, Yukinori Minoshima, Junji Matsui, Robert A Copeland, Shinya Tanaka, and Heike Keilhack
- Subjects
Medicine ,Science - Abstract
The catalytic activities of covalent and ATP-dependent chromatin remodeling are central to regulating the conformational state of chromatin and the resultant transcriptional output. The enzymes that catalyze these activities are often contained within multiprotein complexes in nature. Two such multiprotein complexes, the polycomb repressive complex 2 (PRC2) methyltransferase and the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeler have been reported to act in opposition to each other during development and homeostasis. An imbalance in their activities induced by mutations/deletions in complex members (e.g. SMARCB1) has been suggested to be a pathogenic mechanism in certain human cancers. Here we show that preclinical models of synovial sarcoma-a cancer characterized by functional SMARCB1 loss via its displacement from the SWI/SNF complex through the pathognomonic SS18-SSX fusion protein-display sensitivity to pharmacologic inhibition of EZH2, the catalytic subunit of PRC2. Treatment with tazemetostat, a clinical-stage, selective and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity reverses a subset of synovial sarcoma gene expression and results in concentration-dependent cell growth inhibition and cell death specifically in SS18-SSX fusion-positive cells in vitro. Treatment of mice bearing either a cell line or two patient-derived xenograft models of synovial sarcoma leads to dose-dependent tumor growth inhibition with correlative inhibition of trimethylation levels of the EZH2-specific substrate, lysine 27 on histone H3. These data demonstrate a dependency of SS18-SSX-positive, SMARCB1-deficient synovial sarcomas on EZH2 enzymatic activity and suggests the potential utility of EZH2-targeted drugs in these genetically defined cancers.
- Published
- 2016
- Full Text
- View/download PDF
3. Synergistic Anti-Tumor Activity of EZH2 Inhibitors and Glucocorticoid Receptor Agonists in Models of Germinal Center Non-Hodgkin Lymphomas.
- Author
-
Sarah K Knutson, Natalie M Warholic, L Danielle Johnston, Christine R Klaus, Tim J Wigle, Dorothy Iwanowicz, Bruce A Littlefield, Margaret Porter-Scott, Jesse J Smith, Mikel P Moyer, Robert A Copeland, Roy M Pollock, Kevin W Kuntz, Alejandra Raimondi, and Heike Keilhack
- Subjects
Medicine ,Science - Abstract
Patients with non-Hodgkin lymphoma (NHL) are treated today with a cocktail of drugs referred to as CHOP (Cyclophosphamide, Hydroxyldaunorubicin, Oncovin, and Prednisone). Subsets of patients with NHL of germinal center origin bear oncogenic mutations in the EZH2 histone methyltransferase. Clinical testing of the EZH2 inhibitor EPZ-6438 has recently begun in patients. We report here that combining EPZ-6438 with CHOP in preclinical cell culture and mouse models results in dramatic synergy for cell killing in EZH2 mutant germinal center NHL cells. Surprisingly, we observe that much of this synergy is due to Prednisolone - a glucocorticoid receptor agonist (GRag) component of CHOP. Dramatic synergy was observed when EPZ-6438 is combined with Prednisolone alone, and a similar effect was observed with Dexamethasone, another GRag. Remarkably, the anti-proliferative effect of the EPZ-6438+GRag combination extends beyond EZH2 mutant-bearing cells to more generally impact germinal center NHL. These preclinical data reveal an unanticipated biological intersection between GR-mediated gene regulation and EZH2-mediated chromatin remodeling. The data also suggest the possibility of a significant and practical benefit of combining EZH2 inhibitors and GRag that warrants further investigation in a clinical setting.
- Published
- 2014
- Full Text
- View/download PDF
4. Supplementary Table 2 from Selective Inhibition of EZH2 by EPZ-6438 Leads to Potent Antitumor Activity in EZH2-Mutant Non-Hodgkin Lymphoma
- Author
-
Heike Keilhack, Akira Yokoi, Kevin W. Kuntz, Roy M. Pollock, Toshimitsu Uenaka, Victoria M. Richon, Robert A. Copeland, Mikel P. Moyer, Richard Chesworth, Margaret Porter-Scott, Jesse J. Smith, Nigel J. Waters, Alejandra Raimondi, Christina J. Allain, Christine R. Klaus, Tim J. Wigle, Namita Kumar, Galina Kuznetsov, Mai Uesugi, Tadashi Kadowaki, Yonghong Xiao, Kuan-Chun Huang, Natalie M. Warholic, Yukinori Minoshima, Satoshi Kawano, and Sarah K. Knutson
- Abstract
XLSX file - 132K, Table S2: Effects of EPZ-6438 on Gene Expression in EZH2 Mutant Lymphoma Cells.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figures 1 - 6 from Selective Inhibition of EZH2 by EPZ-6438 Leads to Potent Antitumor Activity in EZH2-Mutant Non-Hodgkin Lymphoma
- Author
-
Heike Keilhack, Akira Yokoi, Kevin W. Kuntz, Roy M. Pollock, Toshimitsu Uenaka, Victoria M. Richon, Robert A. Copeland, Mikel P. Moyer, Richard Chesworth, Margaret Porter-Scott, Jesse J. Smith, Nigel J. Waters, Alejandra Raimondi, Christina J. Allain, Christine R. Klaus, Tim J. Wigle, Namita Kumar, Galina Kuznetsov, Mai Uesugi, Tadashi Kadowaki, Yonghong Xiao, Kuan-Chun Huang, Natalie M. Warholic, Yukinori Minoshima, Satoshi Kawano, and Sarah K. Knutson
- Abstract
PDF file - 811K, Fig. S1. Effects of EPZ-6438 on Gene Promoter H3K27 Methylation, Recruitment of PRC2 Components to Gene Promoters, and Cell Cycle, in WSU-DLCL2 Cells. Fig. S2: Effects of EPZ-6438 on Gene Expression in EZH2 Mutant Lymphoma Cell Lines. Fig. S3: Pharmacokinetic Profiles of EPZ-6438 in Rats and Mice. Fig. S4: EPZ-6438 Compound Levels in Plasma and WSU-DLCL2 Xenograft Tumor Homogenates from Mice Dosed for 7 or 28 Days. Fig. S5: In vivo Effects of EPZ-6438 in Lymphoma Xenograft Models. Fig. S6: Body Weights of Mice during Xenograft Efficacy Studies.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Table 1 from Selective Inhibition of EZH2 by EPZ-6438 Leads to Potent Antitumor Activity in EZH2-Mutant Non-Hodgkin Lymphoma
- Author
-
Heike Keilhack, Akira Yokoi, Kevin W. Kuntz, Roy M. Pollock, Toshimitsu Uenaka, Victoria M. Richon, Robert A. Copeland, Mikel P. Moyer, Richard Chesworth, Margaret Porter-Scott, Jesse J. Smith, Nigel J. Waters, Alejandra Raimondi, Christina J. Allain, Christine R. Klaus, Tim J. Wigle, Namita Kumar, Galina Kuznetsov, Mai Uesugi, Tadashi Kadowaki, Yonghong Xiao, Kuan-Chun Huang, Natalie M. Warholic, Yukinori Minoshima, Satoshi Kawano, and Sarah K. Knutson
- Abstract
PDF file - 25K, Table S1: LCC Values for EPZ-6438 for WSU-DLCL2 Human Lymphoma Cells Dosed Either Continuously or After Compound Washout.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplemental Table S4 from Mechanisms of Pinometostat (EPZ-5676) Treatment–Emergent Resistance in MLL-Rearranged Leukemia
- Author
-
Scott R. Daigle, Kathrin M. Bernt, Stephen J. Blakemore, Robert A. Copeland, Jesse J. Smith, Roy M. Pollock, Edward J. Olhava, Nigel J. Waters, Taylor Yamauchi, Michael J. Maria, Ty M. Thomson, David A. Drubin, Jessica N. Haladyna, and Carly T. Campbell
- Abstract
Contains the mechanism footprint gene list referenced by supplemental Table S3.
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Selective Inhibition of EZH2 by EPZ-6438 Leads to Potent Antitumor Activity in EZH2-Mutant Non-Hodgkin Lymphoma
- Author
-
Heike Keilhack, Akira Yokoi, Kevin W. Kuntz, Roy M. Pollock, Toshimitsu Uenaka, Victoria M. Richon, Robert A. Copeland, Mikel P. Moyer, Richard Chesworth, Margaret Porter-Scott, Jesse J. Smith, Nigel J. Waters, Alejandra Raimondi, Christina J. Allain, Christine R. Klaus, Tim J. Wigle, Namita Kumar, Galina Kuznetsov, Mai Uesugi, Tadashi Kadowaki, Yonghong Xiao, Kuan-Chun Huang, Natalie M. Warholic, Yukinori Minoshima, Satoshi Kawano, and Sarah K. Knutson
- Abstract
Mutations within the catalytic domain of the histone methyltransferase EZH2 have been identified in subsets of patients with non-Hodgkin lymphoma (NHL). These genetic alterations are hypothesized to confer an oncogenic dependency on EZH2 enzymatic activity in these cancers. We have previously reported the discovery of EPZ005678 and EPZ-6438, potent and selective S-adenosyl-methionine-competitive small molecule inhibitors of EZH2. Although both compounds are similar with respect to their mechanism of action and selectivity, EPZ-6438 possesses superior potency and drug-like properties, including good oral bioavailability in animals. Here, we characterize the activity of EPZ-6438 in preclinical models of NHL. EPZ-6438 selectively inhibits intracellular lysine 27 of histone H3 (H3K27) methylation in a concentration- and time-dependent manner in both EZH2 wild-type and mutant lymphoma cells. Inhibition of H3K27 trimethylation (H3K27Me3) leads to selective cell killing of human lymphoma cell lines bearing EZH2 catalytic domain point mutations. Treatment of EZH2-mutant NHL xenograft-bearing mice with EPZ-6438 causes dose-dependent tumor growth inhibition, including complete and sustained tumor regressions with correlative diminution of H3K27Me3 levels in tumors and selected normal tissues. Mice dosed orally with EPZ-6438 for 28 days remained tumor free for up to 63 days after stopping compound treatment in two EZH2-mutant xenograft models. These data confirm the dependency of EZH2-mutant NHL on EZH2 activity and portend the utility of EPZ-6438 as a potential treatment for these genetically defined cancers. Mol Cancer Ther; 13(4); 842–54. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplemental Methods, Figures, and Tables from Mechanisms of Pinometostat (EPZ-5676) Treatment–Emergent Resistance in MLL-Rearranged Leukemia
- Author
-
Scott R. Daigle, Kathrin M. Bernt, Stephen J. Blakemore, Robert A. Copeland, Jesse J. Smith, Roy M. Pollock, Edward J. Olhava, Nigel J. Waters, Taylor Yamauchi, Michael J. Maria, Ty M. Thomson, David A. Drubin, Jessica N. Haladyna, and Carly T. Campbell
- Abstract
Contains supplemental methods, figures, and tables 1, 2, 3, and 5
- Published
- 2023
- Full Text
- View/download PDF
10. Abstract 3425: The discovery and characterization of CFT1946: A potent, selective, and orally bioavailable degrader of mutant BRAF for the treatment of BRAF-driven cancers
- Author
-
Yanke Liang, Mathew E. Sowa, Katrina L. Jackson, Jeffrey R. Simard, Bridget Kreger, Ping Li, Laura Poling, Joelle Baddour, Andrew Good, Hongwei Huang, Scott Eron, Christopher G. Nasveschuk, Robert Yu, Mark Fitzgerald, Victoria Garza, Morgan W. O’Shea, Gesine Veits, Jeremy Y. Yap, Moses Moustakim, Ashley Hart, Roman V. Agafonov, Grace Sarkissian, Joe S. Patel, Richard Deibler, Kyle S. Cole, David Cocozziello, Fazlur Rahman, Andrew J. Phillips, Elizabeth Norton, Adam S. Crystal, Roy M. Pollock, and Stewart L. Fisher
- Subjects
Cancer Research ,Oncology - Abstract
The BRAF kinase plays a critical role in the MAPK signaling pathway and is mutated in ~8% of all human cancers including melanoma (~60%), thyroid (~60%), and lung adenocarcinoma (~10%). The most common mutation in BRAF is V600E (Class I), which is found in >70% in these cancers. Despite the clinical success of approved small molecule inhibitors of BRAF V600E (vemurafenib, dabrafenib and encorafenib), this remains an area of unmet medical need because nearly all patients progress, due to either primary or acquired resistance. A bifunctional degradation activating compound (or BiDACTM degrader) may address the liabilities of approved drugs by overcoming, or preventing the emergence of, resistance to BRAF inhibitors. Here we describe CFT1946, an orally bioavailable cereblon-based BiDAC degrader of BRAF V600 mutant proteins, and provide an overview of the medicinal chemistry path leading to its discovery. CFT1946 degrades BRAF V600 mutant proteins, while maintaining exquisite selectivity against the proteome, sparing wild type BRAF (BRAF-WT), ARAF, and CRAF. In A375 cells, CFT1946 potently degraded BRAF V600E and inhibited ERK phosphorylation and cell growth while having no effect in the mutant KRAS, BRAF-WT driven cell line HCT116. In the A375 xenograft model, oral delivery of CFT1946 at 10 mg/kg PO BID resulted in deeper and more durable tumor regression compared to a clinically relevant dose of encorafenib. Further evaluation of CFT1946 in an engineered, clinically relevant BRAFi-resistant A375 cell line (endogenous BRAF V600E + engineered expression of NRAS Q61K) demonstrated that CFT1946 both degraded BRAF V600E and caused a loss of viability in these cells, while treatment with encorafenib had no effect. In xenografts derived from this BRAFi-resistant cell line, oral dosing of CFT1946 as a single agent led to tumor growth inhibition, while treatment with a clinically relevant dose of encorafenib had no effect on tumor growth. Furthermore, dosing CFT1946 in combination with the MEK inhibitor, trametinib, resulted in significant tumor regression, whereas combining encorafenib with the same dose of trametinib had no effect. The medicinal chemistry campaign resulting in CFT1946 focused on the improvement of in vivo pharmacokinetics and rational linker design to achieve high oral bioavailability in a beyond Rule of 5 heterobifunctional degrader. The preclinical data presented herein support the planned Phase 1/2 clinical trial of CFT1946 for the treatment of BRAF-V600 mutant solid tumors. Citation Format: Yanke Liang, Mathew E. Sowa, Katrina L. Jackson, Jeffrey R. Simard, Bridget Kreger, Ping Li, Laura Poling, Joelle Baddour, Andrew Good, Hongwei Huang, Scott Eron, Christopher G. Nasveschuk, Robert Yu, Mark Fitzgerald, Victoria Garza, Morgan W. O’Shea, Gesine Veits, Jeremy Y. Yap, Moses Moustakim, Ashley Hart, Roman V. Agafonov, Grace Sarkissian, Joe S. Patel, Richard Deibler, Kyle S. Cole, David Cocozziello, Fazlur Rahman, Andrew J. Phillips, Elizabeth Norton, Adam S. Crystal, Roy M. Pollock, Stewart L. Fisher. The discovery and characterization of CFT1946: A potent, selective, and orally bioavailable degrader of mutant BRAF for the treatment of BRAF-driven cancers [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 3425.
- Published
- 2023
- Full Text
- View/download PDF
11. MLL partial tandem duplication leukemia cells are sensitive to small molecule DOT1L inhibition
- Author
-
Michael W.M. Kühn, Michael J. Hadler, Scott R. Daigle, Richard P. Koche, Andrei V. Krivtsov, Edward J. Olhava, Michael A. Caligiuri, Gang Huang, James E. Bradner, Roy M. Pollock, and Scott A. Armstrong
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2015
- Full Text
- View/download PDF
12. CFT7455: A NOVEL, IKZF1/3 DEGRADER THAT DEMONSTRATES POTENT TUMOR REGRESSION IN A SPECTRUM OF NHL XENOGRAFT MODELS
- Author
-
Ashley A. Hart, Stewart L. Fisher, Christina S. Henderson, Scott J. Eron, David A. Proia, Roy M. Pollock, Prasoon Chaturvedi, James A. Henderson, S Perino, B. T Kreger, Christopher G. Nasveschuk, Adam S. Crystal, Andrew C. Good, Roman V. Agafonov, Marta Isasa, B Class, Michelle Mahler, and R. J Kirby
- Subjects
Cancer Research ,Oncology ,business.industry ,Cancer research ,Tumor regression ,Medicine ,Hematology ,General Medicine ,business - Published
- 2021
- Full Text
- View/download PDF
13. Abstract ND09: The discovery and characterization of CFT8634: A potent and selective degrader of BRD9 for the treatment of SMARCB1-perturbed cancers
- Author
-
Katrina L. Jackson, Roman V. Agafonov, Mark W. Carlson, Prasoon Chaturvedi, David Cocozziello, Kyle Cole, Richard Deibler, Scott J. Eron, Andrew Good, Ashley A. Hart, Minsheng He, Christina S. Henderson, Hongwei Huang, Marta Isasa, R. Jason Kirby, Linda Lee, Michelle Mahler, Moses Moustakim, Christopher G. Nasveschuk, Michael Palmer, Laura L. Poling, Roy M. Pollock, Matthew Schnaderbeck, Stan Spence, Gesine K. Veits, Jeremy L. Yap, Ning Yin, Rhamy Zeid, Adam S. Crystal, Andrew J. Phillips, and Stewart L. Fisher
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: The chromatin factor BRD9 is a genetic dependency in some cancers, often referred to as SMARCB1-perturbed cancers. Two types of genetic alterations result in SMARCB1 perturbation: SS18-SSX gene fusion and SMARCB1 loss-of-function mutations. In synovial sarcoma, a rare and aggressive soft tissue malignancy comprising approximately 10% of all soft tissue sarcomas, the presence of the SS18-SSX fusion gene drives the disruption of SMARCB1 function and leads to a synthetic lethal dependence on BRD9. In SMARCB1-null solid tumors, for example malignant rhabdoid tumors (MRT), poorly differentiated chordomas, and epithelioid sarcomas, the absence of SMARCB1 protein results in a similar BRD9 dependence. Thus, in SMARCB1-perturbed cancers, including synovial sarcoma and SMARCB1-null cancers, degradation of BRD9 is hypothesized to result in an anticancer effect. CFT8634 is an orally bioavailable heterobifunctional degrader that induces ternary complex formation with BRD9 and an E3 ligase, leading to the ubiquitination of BRD9 and its subsequent degradation by the proteasome. Results: Here we describe the chemical structure of CFT8634 and an overview of the medicinal chemistry path leading to its discovery. In vitro, CFT8634 promotes rapid, potent, deep, and selective degradation of BRD9 with a half-maximal degradation concentration (DC50) of 2 nM in a synovial sarcoma cell line. In long-term growth assays, CFT8634 is effective at impairing cell growth in a concentration-dependent manner specifically in SMARCB1-perturbed contexts. In vivo, oral dosing of CFT8634 in xenograft models of SMARCB1-perturbed cancers leads to robust and dose-dependent degradation of BRD9, which translates to significant and dose-dependent inhibition of tumor growth in preclinical xenograft models. Conclusion: The preclinical data presented herein support the clinical development of CFT8634 for the treatment of synovial sarcoma and SMARCB1-null tumors. Citation Format: Katrina L. Jackson, Roman V. Agafonov, Mark W. Carlson, Prasoon Chaturvedi, David Cocozziello, Kyle Cole, Richard Deibler, Scott J. Eron, Andrew Good, Ashley A. Hart, Minsheng He, Christina S. Henderson, Hongwei Huang, Marta Isasa, R. Jason Kirby, Linda Lee, Michelle Mahler, Moses Moustakim, Christopher G. Nasveschuk, Michael Palmer, Laura L. Poling, Roy M. Pollock, Matthew Schnaderbeck, Stan Spence, Gesine K. Veits, Jeremy L. Yap, Ning Yin, Rhamy Zeid, Adam S. Crystal, Andrew J. Phillips, Stewart L. Fisher. The discovery and characterization of CFT8634: A potent and selective degrader of BRD9 for the treatment of SMARCB1-perturbed cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr ND09.
- Published
- 2022
- Full Text
- View/download PDF
14. Abstract CT186: Pharmacokinetic (PK) profile of a novel IKZF1/3 degrader, CFT7455, enables significant potency advantage over other IKZF1/3 degraders in models of multiple myeloma (MM) and the results of the initial treatment cohort from a first-in-human (FIH) phase 1/2 study of CFT7455 in MM
- Author
-
Sagar Lonial, Shambavi Richard, Jeffrey V Matous, Andrew J. Yee, Urvi Shah, Neha Mehta-Shah, Thomas Martin, Eli Muchtar, Sikander Ailawadhi, Paul G. Richardson, Manisha Bhutani, Samantha Perino, Jason Kirby, Roman V. Agafonov, Prasoon Chaturvedi, Bradley Class, Matthew Schnaderbeck, Michael R. Palmer, Cathleen Gorman, Oliver Schoenborn-Kellenberger, Amanda Hoerres, Stewart L. Fisher, Roy M. Pollock, Adam Crystal, Michelle Mahler, and Jesus Bardeja
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: CFT7455 is a highly potent and novel Ikaros Family Zinc Finger Protein 1/3 (IKZF1/3) degrader. In xenograft models, CFT7455 has more potent IKZF1/3 degradation compared to other degraders. Early observations from the FIH clinical trial (NCT04756726) along with supporting translational studies are presented here. Methods: Pre-clinical studies comparing CFT7455 and CC-92480 in both in vitro and xenograft models were performed. The pre-clinical studies’ results coincided with observations from the on-going clinical trial. The clinical trial is an open-label, Phase 1/2, multi-center, FIH study in heavily pretreated relapsed/refractory (R/R) MM and non-Hodgkin’s lymphoma (NHL) patients evaluating safety, tolerability, and PK of CFT7455. Eligible MM patients are R/R to therapy and are not candidates for regimens known to provide clinical benefit. A starting dose of 50 μg QD 21 days on/7 days off (21/7) in 28-day cycles was administered. Results: CFT7455 and CC-92480 showed similar cereblon binding profiles and in vitro IKZF1/3 degradation kinetics, translating into sub-nanomolar GI50 values in proliferation assays across a panel of MM cell lines. In the NCI-H929 xenograft model, 100 μg/kg/day of CFT7455 resulted in durable tumor regressions, while 1000 μg/kg/day of CC-92480 gave tumor stasis. Similar results were seen in a systemic model of MM, MM1.S. Both compounds achieved >95% IKZF3 degradation in tumors 4h post dose. At 48h post dose, CFT7455 was more effective than CC-92480 in maintaining IKZF3 degradation (65% vs. 6% respectively). When levels of CFT7455 and CC-92480 in plasma and tumor were compared, CFT7455 concentrations were > DC80 in tumor 48h post dose, while CC-92480 levels were undetectable in tumor and plasma, demonstrating CFT7455 has longer exposure resulting in sustained IKZF1/3 degradation in pre-clinical models. In cohort A, 5 heavily pre-treated MM patients (pts) received single agent CFT7455. 4 pts have received at least 3 cycles, with 2 pts receiving 5 cycles. Neutropenia (grade 4) was observed in 3/5 pts without coincident fever or infection. Additionally, a 2-4 fold accumulation in plasma CFT7455 exposure at steady state was observed. Early pharmacodynamic (PD) data demonstrates deep persistent degradation of IKZF3 (~100%) and serum free light chain reduction (up to 72%) in response to treatment. Stable disease has been observed in 34 pts, suggesting clinical benefit. Conclusions: While CFT7455 showed clinical benefit at 50 ug with deep target degradation, neutropenia was dose limiting. PK/PD modeling suggests alternative dosing regimens may result in increased tolerability with preserved efficacy, and evaluation of them is underway. Updated results will be presented at the meeting. Citation Format: Sagar Lonial, Shambavi Richard, Jeffrey V Matous, Andrew J. Yee, Urvi Shah, Neha Mehta-Shah, Thomas Martin, Eli Muchtar, Sikander Ailawadhi, Paul G. Richardson, Manisha Bhutani, Samantha Perino, Jason Kirby, Roman V. Agafonov, Prasoon Chaturvedi, Bradley Class, Matthew Schnaderbeck, Michael R. Palmer, Cathleen Gorman, Oliver Schoenborn-Kellenberger, Amanda Hoerres, Stewart L. Fisher, Roy M. Pollock, Adam Crystal, Michelle Mahler, Jesus Bardeja. Pharmacokinetic (PK) profile of a novel IKZF1/3 degrader, CFT7455, enables significant potency advantage over other IKZF1/3 degraders in models of multiple myeloma (MM) and the results of the initial treatment cohort from a first-in-human (FIH) phase 1/2 study of CFT7455 in MM [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr CT186.
- Published
- 2022
- Full Text
- View/download PDF
15. Abstract 2158: Preclinical evaluation of CFT1946 as a selective degrader of mutant BRAF for the treatment of BRAF driven cancers
- Author
-
Mathew E. Sowa, Bridget Kreger, Joelle Baddour, Yanke Liang, Jeffrey R. Simard, Laura Poling, Ping Li, Robert Yu, Ashley Hart, Roman V. Agafonov, Grace Sarkissian, Joe Sahil Patel, Richard Deibler, Kyle S. Cole, Scott Eron, David Cocozziello, Fazlur Rahman, Moses Moustakim, Christopher G. Nasveschuk, Katrina L. Jackson, Mark Fitzgerald, Victoria Garza, Morgan O’Shea, Gesine Veits, Jeremy L. Yap, Andrew J. Phillips, Elizabeth Norton, Adam S. Crystal, Stewart L. Fisher, and Roy M. Pollock
- Subjects
Cancer Research ,Oncology - Abstract
The BRAF kinase is a critical node in the MAPK signaling pathway and is mutated in approximately 8% of human cancers including melanoma (~60%), thyroid (~60%), and lung adenocarcinoma (~10%). The most common mutation in BRAF is V600E (Class I), occurring in half of malignant melanomas. This mutation hyperactivates ERK and signals as a RAF inhibitor-sensitive monomer. BRAF inhibitors including vemurafenib, dabrafenib and encorafenib have produced impressive responses in V600X patients, however resistance usually emerges within a year, including RAS mutation, BRAFV600E amplification, and BRAFV600E intragenic deletion or splice variants. These inhibitors are also ineffective against non-V600 BRAF mutants (Class II & III). To address some of these limitations we have developed CFT1946, a bifunctional degradation activating compound (BiDAC™) degrader comprising a BRAF kinase domain targeting ligand linked to a cereblon ligand. CFT1946 is capable of degrading BRAF V600E (Class I), G469A (Class II), G466V (Class III) mutations, and the p61-BRAFV600E splice variant while maintaining exquisite selectivity against the proteome including WT BRAF and CRAF. In A375 cells, CFT1946 potently degraded BRAFV600E (Emax = 26%; DC50 = 14nM at 24hr) and, inhibited ERK phosphorylation (IC50 = 11nM at 24hr) and cell growth (GI50 = 94nM at 96hr) while having no effect in the mutant KRAS driven cell line HCT116. In A375 xenografts, oral delivery of CFT1946 resulted in deeper tumor regressions when dosed at 10 mg/kg PO BID and compared favorably to a clinically relevant dose of encorafenib. We further evaluated CFT1946 in an engineered A375-BRAFV600E/NRASQ61K double mutant model of BRAF inhibitor resistance. CFT1946 was able to degrade BRAFV600E in these cells and was much more effective than encorafenib at inhibiting viability in vitro. In this model, in vivo dosing of single agent CFT1946 caused robust tumor growth inhibition and combination with the MEK inhibitor, trametinib, resulted in tumor regressions. The combination of encorafenib and trametinib showed no activity in the same model. Next, we demonstrated that CFT1946 was able to degrade additional BRAF mutant proteins including G469A (Class II), G466V (Class III), and the p61-BRAFV600E splice variant using heterologous expression in HEK293T cells. Additionally, we also showed that CFT1946, but not encorafenib, inhibited proliferation of the BRAFG466V heterozygous lung tumor cell line H1666. Based on its activity in preclinical models, including models of BRAF inhibitor resistance, and its drug-like properties we are progressing CFT1946 as a candidate for clinical development in patients with solid tumors bearing BRAF V600X mutations. Further, given CFT1946’s activity on non-V600 BRAF mutations, we are continuing to explore CFT1946 and related BiDAC degraders as therapeutic options for patients bearing Class II or Class III BRAF mutations. Citation Format: Mathew E. Sowa, Bridget Kreger, Joelle Baddour, Yanke Liang, Jeffrey R. Simard, Laura Poling, Ping Li, Robert Yu, Ashley Hart, Roman V. Agafonov, Grace Sarkissian, Joe Sahil Patel, Richard Deibler, Kyle S. Cole, Scott Eron, David Cocozziello, Fazlur Rahman, Moses Moustakim, Christopher G. Nasveschuk, Katrina L. Jackson, Mark Fitzgerald, Victoria Garza, Morgan O’Shea, Gesine Veits, Jeremy L. Yap, Andrew J. Phillips, Elizabeth Norton, Adam S. Crystal, Stewart L. Fisher, Roy M. Pollock. Preclinical evaluation of CFT1946 as a selective degrader of mutant BRAF for the treatment of BRAF driven cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2158.
- Published
- 2022
- Full Text
- View/download PDF
16. Abstract ND13: The discovery and characterization of CFT7455: A potent and selective degrader of IKZF1/3 for the treatment of relapsed/refractory multiple myeloma
- Author
-
James A. Henderson, Scott J. Eron, Andrew Good, R Jason Kirby, Samantha Perino, Roman V. Agafonov, Prasoon Chaturvedi, Bradley Class, David Cocozziello, Ashley A. Hart, Christina S. Henderson, Marta Isasa, Brendon Ladd, Matt Schnaderbeck, Michelle Mahler, Adam S. Crystal, Roy M. Pollock, Christopher G. Nasveschuk, Andrew J. Phillips, Stewart L. Fisher, and David A. Proia
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Ikaros family zinc finger protein 1 and 3 (IKZF1/3) are essential transcription factors (TF) for terminal differentiation of B and T cells. Depletion of IKZF1/3 inhibits the growth of multiple myeloma (MM) cells, confirming their dependency on IKZF1/3. IMiDs (lenalidomide, pomalidomide) are effective therapies for treatment of MM and promote degradation of IKZF1/3 via their interaction with CRL4-CRBN E3 ligase. However, most patients treated with lenalidomide or pomalidomide eventually develop progressive disease due to acquired resistance, underscoring the unmet medical need. CFT7455 is a novel IKZF1/3 degrader optimized for high binding affinity to cereblon (CRBN), rapid and deep IKZF1/3 degradation, and potent dose-dependent efficacy in vivo. Results: A series of novel benzoimidazolone-based CRBN ligands with potent binding affinity were discovered and their binding modes were informed by CRBN co-crystal structures. Although the benzoimidazolone-based CRBN binders did not exhibit IKZF1/3 degradation activity, structural insights into their unique binding modes and knowledge of the IKZF1/3 degradation pharmacophore were combined to enable identification of a novel benzoisoindolone-based ligand that exhibited a 10-fold potency increase in biochemical CRBN binding and a 30-fold potency increase in H929 MM cell growth inhibition when compared to lenalidomide. Additional rounds of structure-based drug design, degradation and phenotypic profiling led to the discovery of CFT7455, a highly potent, selective and orally bioavailable degrader of IKZF1/3. CFT7455 demonstrated an 800 and 1600-fold improvement in CRBN binding compared to pomalidomide in biochemical and cellular NanoBRET assays, respectively. In H929 MM cells expressing HiBiT-tagged IKZF1, CFT7455 induced >75% degradation of IKZF1 within 1.5 hrs. The high binding affinity and degradation catalysis shown with CFT7455 enabled potent antiproliferative activity across a panel of MM cell lines, as well as H929 cells made resistant to IMiDs. In vivo, CFT7455 catalyzed deep and durable degradation of IKZF3, translating into potent antitumor activity in multiple myeloma xenograft models. CFT7455 also retained its activity in models resistant or insensitive to clinically approved IMiDs as single agent or in combination with standard of care agent dexamethasone. Conclusion: Overall, CFT7455 is a next generation IKZF1/3 degrader, with improved potency and anticancer efficacy in preclinical models compared to existing IMiDs. These features make CFT7455 an exciting drug candidate, as a single agent or for use in combination. CFT7455 is currently being studied in a Ph1 clinical trial. Citation Format: James A. Henderson, Scott J. Eron, Andrew Good, R Jason Kirby, Samantha Perino, Roman V. Agafonov, Prasoon Chaturvedi, Bradley Class, David Cocozziello, Ashley A. Hart, Christina S. Henderson, Marta Isasa, Brendon Ladd, Matt Schnaderbeck, Michelle Mahler, Adam S. Crystal, Roy M. Pollock, Christopher G. Nasveschuk, Andrew J. Phillips, Stewart L. Fisher, David A. Proia. The discovery and characterization of CFT7455: A potent and selective degrader of IKZF1/3 for the treatment of relapsed/refractory multiple myeloma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr ND13.
- Published
- 2022
- Full Text
- View/download PDF
17. High-Throughput Quantitative Assay Technologies for Accelerating the Discovery and Optimization of Targeted Protein Degradation Therapeutics
- Author
-
Jeffrey R Simard, Trang N. Tieu, Eunice Park, Ellen F. Vieux, Andrew J. K. Phillips, Reina Improgo, Stewart L. Fisher, Linda Lee, and Roy M. Pollock
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,Recombinant Fusion Proteins ,Ubiquitin-Protein Ligases ,Protein degradation ,Ligands ,Biochemistry ,Analytical Chemistry ,Small Molecule Libraries ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Drug Discovery ,medicine ,Humans ,Luciferase ,Molecular Targeted Therapy ,biology ,Staining and Labeling ,Chemistry ,Drug discovery ,Neurodegeneration ,Ubiquitination ,medicine.disease ,Flow Cytometry ,Small molecule ,Cell biology ,High-Throughput Screening Assays ,030104 developmental biology ,Eukaryotic Cells ,Spectrometry, Fluorescence ,Proteasome ,030220 oncology & carcinogenesis ,Proteolysis ,biology.protein ,Molecular Medicine ,Protein Processing, Post-Translational ,Function (biology) ,Biotechnology ,Protein Binding - Abstract
The aberrant regulation of protein expression and function can drastically alter cellular physiology and lead to numerous pathophysiological conditions such as cancer, inflammatory diseases, and neurodegeneration. The steady-state expression levels of endogenous proteins are controlled by a balance of de novo synthesis rates and degradation rates. Moreover, the levels of activated proteins in signaling cascades can be further modulated by a variety of posttranslational modifications and protein-protein interactions. The field of targeted protein degradation is an emerging area for drug discovery in which small molecules are used to recruit E3 ubiquitin ligases to catalyze the ubiquitination and subsequent degradation of disease-causing target proteins by the proteasome in both a dose- and time-dependent manner. Traditional approaches for quantifying protein level changes in cells, such as Western blots, are typically low throughput with limited quantification, making it hard to drive the rapid development of therapeutics that induce selective, rapid, and sustained protein degradation. In the last decade, a number of techniques and technologies have emerged that have helped to accelerate targeted protein degradation drug discovery efforts, including the use of fluorescent protein fusions and reporter tags, flow cytometry, time-resolved fluorescence energy transfer (TR-FRET), and split luciferase systems. Here we discuss the advantages and disadvantages associated with these technologies and their application to the development and optimization of degraders as therapeutics.
- Published
- 2021
18. Genomic discovery of an evolutionarily programmed modality for small-molecule targeting of an intractable protein surface
- Author
-
Dylan T. Stiles, Keith Robison, Alan S. Mann, Brian R. Bowman, Tara Hardy, Michelle L. Stewart, Siavash Mostafavi, Gregory L. Verdine, Seung-Joo Lee, Morgenstern Jay P, Zhigang Weng, Mathew E. Sowa, Sukrat Arya, Andrew M. Fry, Kyle Kenyon, Ende Pan, Richard D. Klausner, Khian Hong Pua, Roy M. Pollock, Sharon A. Townson, Minyun Zhou, Uddhav Kumar Shigdel, Andrew T Rajczewski, Joshua A. V. Blodgett, Daniel W. Udwary, and Daniel C. Gray
- Subjects
Models, Molecular ,Evolution ,Protein Conformation ,natural products ,Druggability ,Sequence Homology ,Cell Cycle Proteins ,Computational biology ,Tacrolimus Binding Protein 1A ,FK506-binding protein ,Microbiology ,Autoantigens ,Antiviral Agents ,Evolution, Molecular ,Small Molecule Libraries ,Protein structure ,Models ,genome mining ,Humans ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Structural motif ,Coiled coil ,Sirolimus ,Multidisciplinary ,Genome ,Chemistry ,Calcineurin ,TOR Serine-Threonine Kinases ,Bacterial ,Molecular ,Biological Sciences ,Small molecule ,Anti-Bacterial Agents ,CEP250 ,Actinobacteria ,HEK293 Cells ,Macrolides ,Genome, Bacterial - Abstract
Significance This manuscript reports on a member of the FK506/rapamycin family, WDB002, and the realization that FKBP-mediated recognition is a genetically programmable modality that enables engagement of topologically flat targets. FKBP-mediated recognition is thus nature’s strategy for drugging the “undruggable.” The surface of FKBP engages three completely unrelated targets—calcineurin, MTOR, and CEP250—with high-target affinity and specificity, using different constellations of amino acid residues. Target specificity is determined solely by the “variable domain” of the bound small molecule alone, suggesting the modality might be generalizable to other undruggable targets through variable domain engineering. Finally, since WDB002 targets CEP250, it may be a promising starting point for developing a treatment for COVID-19., The vast majority of intracellular protein targets are refractory toward small-molecule therapeutic engagement, and additional therapeutic modalities are needed to overcome this deficiency. Here, the identification and characterization of a natural product, WDB002, reveals a therapeutic modality that dramatically expands the currently accepted limits of druggability. WDB002, in complex with the FK506-binding protein (FKBP12), potently and selectively binds the human centrosomal protein 250 (CEP250), resulting in disruption of CEP250 function in cells. The recognition mode is unprecedented in that the targeted domain of CEP250 is a coiled coil and is topologically featureless, embodying both a structural motif and surface topology previously considered on the extreme limits of “undruggability” for an intracellular target. Structural studies reveal extensive protein–WDB002 and protein–protein contacts, with the latter being distinct from those seen in FKBP12 ternary complexes formed by FK506 and rapamycin. Outward-facing structural changes in a bound small molecule can thus reprogram FKBP12 to engage diverse, otherwise “undruggable” targets. The flat-targeting modality demonstrated here has the potential to expand the druggable target range of small-molecule therapeutics. As CEP250 was recently found to be an interaction partner with the Nsp13 protein of the SARS-CoV-2 virus that causes COVID-19 disease, it is possible that WDB002 or an analog may exert useful antiviral activity through its ability to form high-affinity ternary complexes containing CEP250 and FKBP12.
- Published
- 2020
19. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia
- Author
-
Stephen J. Blakemore, Alicia Clawson, Guillermo Garcia-Manero, Michael R. Savona, Jorge DiMartino, Nigel J. Waters, Scott A. Armstrong, David A. Rizzieri, Roy M. Pollock, Martin S. Tallman, Jesus G. Berdeja, Eric Hedrick, Jessica K. Altman, Mojca Jongen-Lavrenic, A. Benjamin Suttle, Blythe Thomson, Scott R. Daigle, Raoul Tibes, Bob Löwenberg, Eytan M. Stein, Andrei V. Krivtsov, and Hematology
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Clinical Trials and Observations ,Nausea ,Immunology ,Antineoplastic Agents ,Methylation ,Biochemistry ,Histones ,Young Adult ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,Adverse effect ,Aged ,Aged, 80 and over ,Acute leukemia ,business.industry ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,Cell Biology ,Hematology ,DOT1L ,Middle Aged ,medicine.disease ,Clinical trial ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,Pharmacodynamics ,Benzimidazoles ,Female ,medicine.symptom ,business ,Febrile neutropenia - Abstract
Pinometostat (EPZ-5676) is a first-in-class small-molecule inhibitor of the histone methyltransferase disrupter of telomeric silencing 1-like (DOT1L). In this phase 1 study, pinometostat was evaluated for safety and efficacy in adult patients with advanced acute leukemias, particularly those involving mixed lineage leukemia (MLL) gene rearrangements (MLL-r) resulting from 11q23 translocations. Fifty-one patients were enrolled into 6 dose-escalation cohorts (n = 26) and 2 expansion cohorts (n = 25) at pinometostat doses of 54 and 90 mg/m(2) per day by continuous intravenous infusion in 28-day cycles. Because a maximum tolerated dose was not established in the dose-escalation phase, the expansion doses were selected based on safety and clinical response data combined with pharmacodynamic evidence of reduction in H3K79 methylation during dose escalation. Across all dose levels, plasma pinometostat concentrations increased in an approximately dose-proportional fashion, reaching an apparent steady-state by 4-8 hours after infusion, and rapidly decreased following treatment cessation. The most common adverse events, of any cause, were fatigue (39%), nausea (39%), constipation (35%), and febrile neutropenia (35%). Overall, 2 patients, both with t(11;19), experienced complete remission at 54 mg/m(2) per day by continuous intravenous infusion, demonstrating proof of concept for delivering clinically meaningful responses through targeting DOT1L using the single agent pinometostat in MLL-r leukemia patients. Administration of pinometostat was generally safe, with the maximum tolerated dose not being reached, although efficacy as a single agent was modest. This study demonstrates the therapeutic potential for targeting DOT1L in MLL-r leukemia and lays the groundwork for future combination approaches in this patient population. This clinical trial is registered at www.clinicaltrials.gov as NCT01684150.
- Published
- 2018
- Full Text
- View/download PDF
20. Abstract LB007: CFT7455: A novel, IKZF1/3 degrader that demonstrates potent tumor regression in IMiD-resistant multiple myeloma (MM) xenograft models
- Author
-
Scott J. Eron, Roy M. Pollock, Adam S. Crystal, James A. Henderson, Prasoon Chaturvedi, Christina S. Henderson, Samantha Perino, Stewart L. Fisher, Matt Schnaderbeck, Marta Isasa, Ashley A. Hart, Christopher G. Nasveschuk, Andrew C. Good, Roman V. Agafonov, R. Jason Kirby, Bradley Class, David Cocozziello, Michelle Mahler, David A. Proia, Andrew J. K. Phillips, and Brendon Ladd
- Subjects
Cancer Research ,Chemistry ,Cereblon ,Cancer ,Pharmacology ,medicine.disease ,Pomalidomide ,Oncology ,In vivo ,medicine ,Progressive disease ,Multiple myeloma ,Dexamethasone ,medicine.drug ,Lenalidomide - Abstract
Introduction Ikaros family zinc finger protein 1 and 3 (IKZF1/3) are essential transcription factors (TF) for terminal differentiation of B and T cells. Depletion of IKZF1/3 in MM cells inhibits growth, confirming their dependency on IKZF1/3. IMiDs (lenalidomide[len], pomalidomide[pom]) are effective therapies for treatment of MM and promote degradation of IKZF1/3 via their interaction with CRL4-CRBN E3 ligase. Most patients treated with len or pom eventually develop progressive disease due to acquired resistance, underscoring an unmet medical need. CFT7455 is a novel IKZF1/3 degrader optimized for high affinity cereblon (CRBN) binding, rapid IKZF1/3 degradation, and potent in vivo efficacy. Results CFT7455 demonstrated an 800 to 1600-fold improvement in CRBN binding compared to pom in biochemical and cellular NanoBRET assays, respectively. In H929 MM cells expressing HiBiT-tagged IKZF1, CFT7455 induced >75% degradation of IKZF1 within 1.5 hr. The high binding affinity and degradation catalysis shown with CFT7455 enabled potent antiproliferative activity across a panel of MM cell lines as well as H929 cells made resistant to IMiDs. In RPMI-8226 MM mice xenografts, CFT7455 (0.1 mg/kg/day) resulted in deep, durable degradation of IKZF3 (21% and 9.5% of vehicle levels at 4 and 24 hr, respectively). Protein levels for IRF4, an essential TF in MM, declined over 7 days with daily CFT7455 treatment to 8% of vehicle levels. Dose dependent efficacy ranged from 0.003-0.1 mg/kg/day, with tumor regression evident at doses ≥0.01 mg/kg/day. Pom was inactive in this model at a human equivalent dose (3 mg/kg/day), with no observed tumor shrinkage in these mice following 17 days of dosing. Switching pom treatment to CFT7455 (0.1 mg/kg/day) on Day 18 led to tumor regression in 67% of animals on Day 28 and 100% tumor regression on Day 35, demonstrating that CFT7455 penetrates large tissues and is efficacious in rapidly growing, IMiD resistant tumors. In the H929 tumor xenograft model, administration of CFT7455 (0.1 mg/kg/day) promoted tumor regression (95% tumor growth inhibition by 7 days); dosing was stopped after 21 days. On Day 63, half the tumors remained below their starting tumor volume. Additionally, CFT7455 demonstrated durable tumor regression in the aggressive MM1.S systemic MM tumor model. In mice bearing RPMI-8226 xenograft tumors, the combination of CFT7455 (QD) and dexamethasone (QW) was more active, and demonstrated a significant survival improvement, compared to either agent alone. Conclusions CFT7455 is a highly potent, catalytic degrader of IKZF1/3, with marked antitumor activity as a single agent and in combination with dexamethasone. Importantly, CFT7455 retains activity in models resistant or insensitive to IMiDs. These results warrant investigation of CFT7455 as a therapeutic approach for MM and a clinical study is planned. Citation Format: James A. Henderson, R. Jason Kirby, Samantha Perino, Roman V. Agafonov, Prasoon Chaturvedi, Bradley Class, David Cocozziello, Scott J. Eron, Andrew Good, Ashley A. Hart, Christina Henderson, Marta Isasa, Brendon Ladd, Matt Schnaderbeck, Michelle Mahler, Roy M. Pollock, Adam S. Crystal, Christopher G. Nasveschuk, Andrew J. Phillips, Stewart L. Fisher, David A. Proia. CFT7455: A novel, IKZF1/3 degrader that demonstrates potent tumor regression in IMiD-resistant multiple myeloma (MM) xenograft models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr LB007.
- Published
- 2021
- Full Text
- View/download PDF
21. The Importance of Being Me: Magic Methyls, Methyltransferase Inhibitors, and the Discovery of Tazemetostat
- Author
-
Tim J. Wigle, Roy M. Pollock, Margaret Porter-Scott, Christina J. Allain, Mikel P. Moyer, John Campbell, Chris J. Sneeringer, Kevin Wayne Kuntz, Sarah K. Knutson, Robert A. Copeland, Richard Chesworth, Heike Keilhack, Christina R. Majer, and Victoria M. Richon
- Subjects
0301 basic medicine ,Methyltransferase ,Methylation ,Histones ,Small Molecule Libraries ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,Drug Discovery ,Histone methylation ,Animals ,Humans ,Structure–activity relationship ,Enhancer of Zeste Homolog 2 Protein ,Enzyme Inhibitors ,Regulation of gene expression ,Genetics ,biology ,Chemistry ,EZH2 ,Polycomb Repressive Complex 2 ,030104 developmental biology ,Histone ,Histone methyltransferase ,biology.protein ,Cancer research ,Molecular Medicine ,Protein Processing, Post-Translational - Abstract
Posttranslational methylation of histones plays a critical role in gene regulation. Misregulation of histone methylation can lead to oncogenic transformation. Enhancer of Zeste homologue 2 (EZH2) methylates histone 3 at lysine 27 (H3K27) and abnormal methylation of this site is found in many cancers. Tazemetostat, an EHZ2 inhibitor in clinical development, has shown activity in both preclinical models of cancer as well as in patients with lymphoma or INI1-deficient solid tumors. Herein we report the structure-activity relationships from identification of an initial hit in a high-throughput screen through selection of tazemetostat for clinical development. The importance of several methyl groups to the potency of the inhibitors is highlighted as well as the importance of balancing pharmacokinetic properties with potency.
- Published
- 2016
- Full Text
- View/download PDF
22. Exploring drug delivery for the DOT1L inhibitor pinometostat (EPZ-5676): Subcutaneous administration as an alternative to continuous IV infusion, in the pursuit of an epigenetic target
- Author
-
Nigel J. Waters, Angelos Dovletoglou, Tyler B. Jensen, Scott R. Daigle, Edward J. Olhava, Brett Franklin Truitt, Aravind Basavapathruni, Carly T. Campbell, Kim Stickland, Roy M. Pollock, and Bruce N. Rehlaender
- Subjects
Male ,Chemistry, Pharmaceutical ,Injections, Subcutaneous ,Administration, Oral ,Biological Availability ,Pharmaceutical Science ,Phases of clinical research ,Antineoplastic Agents ,Pharmacology ,Drug Administration Schedule ,Epigenesis, Genetic ,Rats, Sprague-Dawley ,Mice ,Dogs ,Oral administration ,medicine ,Animals ,Humans ,Dosing ,Enzyme Inhibitors ,Infusions, Intravenous ,Drug Carriers ,Acute leukemia ,Gene Expression Regulation, Leukemic ,business.industry ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,DOT1L ,DNA Methylation ,medicine.disease ,Xenograft Model Antitumor Assays ,Leukemia, Biphenotypic, Acute ,Tumor Burden ,Leukemia ,Delayed-Action Preparations ,Histone methyltransferase ,Drug delivery ,Benzimidazoles ,business - Abstract
Protein methyltransferases are emerging as promising drug targets for therapeutic intervention in human cancers. Pinometostat (EPZ-5676) is a small molecule inhibitor of the DOT1L enzyme, a histone methyltransferase that methylates lysine 79 of histone H3. DOT1L activity is dysregulated in the pathophysiology of rearranged mixed lineage leukemia (MLL-r). Pinometostat is currently in Phase 1 clinical trials in relapsed refractory acute leukemia patients and is administered as a continuous IV infusion (CIV). The studies herein investigated alternatives to CIV administration of pinometostat to improve patient convenience. Various sustained release technologies were considered, and based on the required dose size as well as practical considerations, subcutaneous (SC) bolus administration of a solution formulation was selected for further evaluation in preclinical studies. SC administration offered improved exposure and complete bioavailability of pinometostat relative to CIV and oral administration. These findings warranted further evaluation in rat xenograft models of MLL-r leukemia. SC dosing in xenograft models demonstrated inhibition of MLL-r tumor growth and inhibition of pharmacodynamic markers of DOT1L activity. However, a dosing frequency of thrice daily (t.i.d) was required in these studies to elicit optimal inhibition of DOT1L target genes and tumor growth inhibition. Development of an extended release formulation may prove useful in the further optimization of the SC delivery of pinometostat, moving towards a more convenient dosing paradigm for patients.
- Published
- 2015
- Full Text
- View/download PDF
23. Abstract B37: Development of inhibitors of the activated form of KRAS G12C
- Author
-
Meizhong Jin, Linlong Xue, Sharon A. Townson, Alan C. Rigby, Siminia Grigoriu, Kathryn M. Luly, Earl W. May, Alex Yuzhakov, Cindy C. Benod, Michelle L. Stewart, Alan S. Mann, Mark J. Mulvihill, Anna Kohlmann, Nicholas Perl, Jason T. Lowe, Jonah Simon, Alec D Silver, Gregory L. Verdine, Roy M. Pollock, Minyun Zhou, and Seung-Joo Lee
- Subjects
Cancer Research ,biology ,Effector ,Cell growth ,Growth factor ,medicine.medical_treatment ,Mutant ,Context (language use) ,Cypa ,medicine.disease_cause ,biology.organism_classification ,digestive system diseases ,chemistry.chemical_compound ,Oncology ,chemistry ,medicine ,Cancer research ,KRAS ,Growth inhibition ,Molecular Biology - Abstract
Activating mutations in RAS proteins occur in ~1/3 of human cancers. These mutations impair the ability of the protein to hydrolyze GTP to GDP. As a result, mutant RAS proteins exist predominantly in the GTP-bound state, which directly activates aberrant downstream signaling via interaction with effectors such as RAF. Most RAS mutations occur at glycine 12 of the KRAS isoform. One such mutation, KRAS G12C, is particularly common in non-small cell lung cancer where it is found in ~15% of lung adenocarcinomas. Recent efforts have targeted KRAS G12C in the GDP-bound state; however, direct pharmacologic inhibition of active, GTP-bound KRAS G12C has proved challenging. Here, we deployed a novel SMARTTM (Small Molecule Assisted Receptor Targeting) platform to advance covalent compounds that selectively inhibit GTP-bound KRAS G12C. Using a mechanism reminiscent of the natural products rapamycin and cyclosporine, these compounds promote formation of a novel inhibitory ternary complex consisting of cyclophilin A (CypA, an abundant immunophilin present in all human cells), the SMART inhibitor, and GTP-KRAS G12C. Structure-based design of the SMART inhibitor yielded potent covalent inhibitors of GTP-KRAS G12C that exhibit >100-fold selectivity for mutant KRAS G12C over WT KRAS. Structural analysis of the ternary complex revealed that the covalent linkage between the SMART inhibitor and the mutant cysteine of KRAS occurred in the context of extensive interactions between CypA, the SMART inhibitor, and GTP-KRAS G12C that provide significant binding affinity (KI = 2.5 μM). The GTP-KRAS G12C|Inhibitor|CypA complex directly occluded effector binding, and as such, the compounds disrupted the KRAS-RAF interaction in biochemical assays. This activity was dependent on CypA, underlining the importance of the KRAS G12C| CypA protein-protein interaction in driving target engagement. In cell-based studies, SMART inhibitors crosslinked KRAS G12C and potently inhibited ERK phosphorylation and cell growth in G12C mutant tumor cell lines but had no effect on non-G12C bearing tumor cells. CRISPR knockout of cellular CypA confirmed that these activities were dependent on the presence of endogenous CypA. Importantly, SMART inhibitors bind directly to active, GTP-KRAS G12C and thus, their activity does not rely on trapping KRAS G12C in the inactive GDP-bound state. As a result, the cellular potency of SMART inhibitors with respect to crosslinking, pERK inhibition, and growth inhibition was maintained in the presence of growth factor treatments that reduce the cellular GDP-KRAS G12C pool. In contrast, we found that the activity of a previously described GDP-KRAS G12C targeting inhibitor was attenuated by growth factor treatment. To our knowledge, these are the first examples of mutant-selective KRAS inhibitors that target the active, GTP-bound state of KRAS G12C. We are currently optimizing the drug-like properties of these SMART inhibitors and evaluating their activity in in vivo models. Citation Format: Michelle L. Stewart, Nicholas R. Perl, Seung-Joo Lee, Linlong Xue, Minyun Zhou, Jonah Simon, Kathryn M. Luly, Siminia Grigoriu, Alex Yuzhakov, Alec Silver, Jason T. Lowe, Cindy C. Benod, Alan S. Mann, Gregory L. Verdine, Alan C. Rigby, Mark J. Mulvihill, Earl W. May, Anna Kohlmann, Sharon A. Townson, Roy M. Pollock, Meizhong Jin. Development of inhibitors of the activated form of KRAS G12C [abstract]. In: Proceedings of the AACR Special Conference on Targeting RAS-Driven Cancers; 2018 Dec 9-12; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2020;18(5_Suppl):Abstract nr B37.
- Published
- 2020
- Full Text
- View/download PDF
24. Mechanisms of pinometostat (EPZ-5676) treatment–emergent resistance in MLL-rearranged leukemia
- Author
-
Edward J. Olhava, Nigel J. Waters, J. Joshua Smith, David A. Drubin, Kathrin M. Bernt, Jessica Haladyna, Taylor Yamauchi, Roy M. Pollock, Carly T. Campbell, Timothy M. Thomson, Stephen J. Blakemore, Robert A. Copeland, Michael J. Maria, and Scott R. Daigle
- Subjects
0301 basic medicine ,Cancer Research ,Acute leukemia ,ATP-binding cassette transporter ,Gene rearrangement ,Drug resistance ,DOT1L ,Biology ,medicine.disease ,Molecular biology ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,Oncology ,Downregulation and upregulation ,Cancer research ,medicine ,Efflux - Abstract
DOT1L is a protein methyltransferase involved in the development and maintenance of MLL-rearranged (MLL-r) leukemia through its ectopic methylation of histones associated with well-characterized leukemic genes. Pinometostat (EPZ-5676), a selective inhibitor of DOT1L, is in clinical development in relapsed/refractory acute leukemia patients harboring rearrangements of the MLL gene. The observation of responses and subsequent relapses in the adult trial treating MLL-r patients motivated preclinical investigations into potential mechanisms of pinometostat treatment-emergent resistance (TER) in cell lines confirmed to have MLL-r. TER was achieved in five MLL-r cell lines, KOPN-8, MOLM-13, MV4-11, NOMO-1, and SEM. Two of the cell lines, KOPN-8 and NOMO-1, were thoroughly characterized to understand the mechanisms involved in pinometostat resistance. Unlike many other targeted therapies, resistance does not appear to be achieved through drug-induced selection of mutations of the target itself. Instead, we identified both drug efflux transporter dependent and independent mechanisms of resistance to pinometostat. In KOPN-8 TER cells, increased expression of the drug efflux transporter ABCB1 (P-glycoprotein, MDR1) was the primary mechanism of drug resistance. In contrast, resistance in NOMO-1 cells occurs through a mechanism other than upregulation of a specific efflux pump. RNA-seq analysis performed on both parental and resistant KOPN-8 and NOMO-1 cell lines supported two unique candidate pathway mechanisms that may explain the pinometostat resistance observed in these cell lines. These results are the first demonstration of TER models of the DOT1L inhibitor pinometostat and may provide useful tools for investigating clinical resistance. Mol Cancer Ther; 16(8); 1669–79, The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
- Published
- 2017
25. Reaction Coupling between Wild-Type and Disease-Associated Mutant EZH2
- Author
-
Tim J. Wigle, Roy M. Pollock, Sarah K. Knutson, Heike Keilhack, Margaret Porter Scott, Lei Jin, J. Joshua Smith, Robert A. Copeland, Mikel P. Moyer, Kevin Wayne Kuntz, Natalie Warholic, and Brooke M. Swalm
- Subjects
chemistry.chemical_classification ,biology ,Point mutation ,EZH2 ,Mutant ,Polycomb Repressive Complex 2 ,Wild type ,macromolecular substances ,General Medicine ,Methylation ,Biochemistry ,Molecular biology ,Histone H3 ,Enzyme ,chemistry ,Neoplasms ,hemic and lymphatic diseases ,Mutation ,biology.protein ,Humans ,Molecular Medicine ,Enhancer of Zeste Homolog 2 Protein ,PRC2 - Abstract
EZH2 and EZH1 are protein methyltransferases (PMTs) responsible for histone H3, lysine 27 (H3K27) methylation. Trimethylation of H3K27 (H3K27me3) is a hallmark of many cancers, including non-Hodgkin lymphoma (NHL). Heterozygous EZH2 point mutations at Tyr641, Ala677, and Ala687 have been observed in NHL. The Tyr641 mutations enhance activity on H3K27me2 but have weak or no activity on unmethylated H3K27, whereas the Ala677 and Ala687 mutations use substrates of all methylation states effectively. It has been proposed that enzymatic coupling of the wild-type and mutant enzymes leads to the oncogenic H3K27me3 mark in mutant-bearing NHL. We show that coupling with the wild-type enzyme is needed to achieve H3K27me3 for several mutants, but that others are capable of achieving H3K27me3 on their own. All forms of PRC2 (wild-type and mutants) display kinetic signatures that are consistent with a distributive mechanism of catalysis.
- Published
- 2014
- Full Text
- View/download PDF
26. DOT1L Inhibitor EPZ-5676 Displays Synergistic Antiproliferative Activity in Combination with Standard of Care Drugs and Hypomethylating Agents inMLL-Rearranged Leukemia Cells
- Author
-
J. Joshua Smith, L. Danielle Johnston, Edward J. Olhava, Carly A Campbell, Robert A. Copeland, Mikel P. Moyer, Margaret Porter Scott, Scott R. Daigle, Dorothy Iwanowicz, Alejandra Raimondi, Roy M. Pollock, and Christine Klaus
- Subjects
Myeloid ,Methyltransferase ,Daunorubicin ,Antineoplastic Agents ,Pharmacology ,Biology ,Methylation ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Humans ,neoplasms ,Cell Proliferation ,Acute leukemia ,Myeloid leukemia ,Drug Synergism ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,medicine.disease ,Growth Inhibitors ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Cytarabine ,Molecular Medicine ,Benzimidazoles ,DOT1L Inhibitor EPZ-5676 ,medicine.drug - Abstract
EPZ-5676 [(2R,3R,4S,5R)-2-(6-amino-9H-purin-9-yl)-5-((((1r,3S)-3-(2-(5-(tert-butyl)-1H-benzo[d]imidazol-2-yl)ethyl)cyclobutyl)(isopropyl)amino)methyl)tetrahydrofuran-3,4-diol], a small-molecule inhibitor of the protein methyltransferase DOT1L, is currently under clinical investigation for acute leukemias bearing MLL-rearrangements (MLL-r). In this study, we evaluated EPZ-5676 in combination with standard of care (SOC) agents for acute leukemias as well as other chromatin-modifying drugs in cellular assays with three human acute leukemia cell lines: MOLM-13 (MLL-AF9), MV4-11 (MLL-AF4), and SKM-1 (non-MLL-r). Studies were performed to evaluate the antiproliferative effects of EPZ-5676 combinations in a cotreatment model in which the second agent was added simultaneously with EPZ-5676 at the beginning of the assay, or in a pretreatment model in which cells were incubated for several days in the presence of EPZ-5676 prior to the addition of the second agent. EPZ-5676 was found to act synergistically with the acute myeloid leukemia (AML) SOC agents cytarabine or daunorubicin in MOLM-13 and MV4-11 MLL-r cell lines. EPZ-5676 is selective for MLL-r cell lines as demonstrated by its lack of effect either alone or in combination in the nonrearranged SKM-1 cell line. In MLL-r cells, the combination benefit was observed even when EPZ-5676 was washed out prior to the addition of the chemotherapeutic agents, suggesting that EPZ-5676 sets up a durable, altered chromatin state that enhances the chemotherapeutic effects. Our evaluation of EPZ-5676 in conjunction with other chromatin-modifying drugs also revealed a consistent combination benefit, including synergy with DNA hypomethylating agents. These results indicate that EPZ-5676 is highly efficacious as a single agent and synergistically acts with other chemotherapeutics, including AML SOC drugs and DNA hypomethylating agents in MLL-r cells.
- Published
- 2014
- Full Text
- View/download PDF
27. Nonclinical pharmacokinetics and metabolism of EPZ-5676, a novel DOT1L histone methyltransferase inhibitor
- Author
-
Edward J. Olhava, Angelos Dovletoglou, Christina J. Allain, Nigel J. Waters, Richard Chesworth, Aravind Basavapathruni, Alejandra Raimondi, Roy M. Pollock, Carly A. Therkelsen, Paul G. Pearson, Lei Jin, Victoria M. Richon, Robert A. Copeland, Mikel P. Moyer, P. Ann Boriack-Sjodin, Christine Klaus, Margaret Porter Scott, and Scott R. Daigle
- Subjects
Pharmacology ,Volume of distribution ,Chemistry ,Pharmaceutical Science ,Renal function ,General Medicine ,DOT1L ,Bioavailability ,Pharmacokinetics ,In vivo ,Histone methyltransferase ,Pharmacology (medical) ,ADME - Abstract
(2R,3R,4S,5R)-2-(6-Amino-9H-purin-9-yl)-5-((((1r,3S)-3-(2-(5-(tert-butyl)-1H-benzo[d]imidazol-2-yl)ethyl)cyclobutyl)(isopropyl)amino)methyl)tetrahydrofuran-3,4-diol (EPZ-5676) is a novel DOT1L histone methyltransferase inhibitor currently in clinical development for the treatment of MLL-rearranged leukemias. This report describes the preclinical pharmacokinetics and metabolism of EPZ-5676, an aminonucleoside analog with exquisite target potency and selectivity that has shown robust and durable tumor growth inhibition in preclinical models. The in vivo pharmacokinetics in mouse, rat and dog were characterized following i.v. and p.o. administration; EPZ-5676 had moderate to high clearance, low oral bioavailability with a steady-state volume of distribution 2-3 fold higher than total body water. EPZ-5676 showed biexponential kinetics following i.v. administration, giving rise to a terminal elimination half-life (t1/2 ) of 1.1, 3.7 and 13.6 h in mouse, rat and dog, respectively. The corresponding in vitro ADME parameters were also studied and utilized for in vitro-in vivo extrapolation purposes. There was good agreement between the microsomal clearance and the in vivo clearance implicating hepatic oxidative metabolism as the predominant elimination route in preclinical species. Furthermore, low renal clearance was observed in mouse, approximating to fu -corrected glomerular filtration rate (GFR) and thus passive glomerular filtration. The metabolic pathways across species were studied in liver microsomes in which EPZ-5676 was metabolized to three monohydroxylated metabolites (M1, M3 and M5), one N-dealkylated product (M4) as well as an N-oxide (M6).
- Published
- 2014
- Full Text
- View/download PDF
28. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia
- Author
-
Carly A. Therkelsen, Alejandra Raimondi, Roy M. Pollock, Margaret Porter Scott, Scott R. Daigle, Robert A. Copeland, Mikel P. Moyer, Aravind Basavapathruni, Edward J. Olhava, Christina J. Allain, Lei Jin, Christine Klaus, Richard Chesworth, Nigel J. Waters, Victoria M. Richon, and P. Ann Boriack-Sjodin
- Subjects
Histone methyltransferase activity ,Methyltransferase ,Protein Conformation ,Immunology ,Antineoplastic Agents ,Biology ,Biochemistry ,Histones ,Rats, Nude ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,neoplasms ,Cell Proliferation ,Acute leukemia ,Myeloid Neoplasia ,Leukemia ,Dose-Response Relationship, Drug ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,Cell Biology ,Hematology ,Methylation ,DOT1L ,DNA Methylation ,medicine.disease ,Rats ,Cell killing ,Histone methyltransferase ,Histone Methyltransferases ,Cancer research ,Benzimidazoles ,Female ,Myeloid-Lymphoid Leukemia Protein ,Neoplasm Transplantation - Abstract
Rearrangements of the MLL gene define a genetically distinct subset of acute leukemias with poor prognosis. Current treatment options are of limited effectiveness; thus, there is a pressing need for new therapies for this disease. Genetic and small molecule inhibitor studies have demonstrated that the histone methyltransferase DOT1L is required for the development and maintenance of MLL-rearranged leukemia in model systems. Here we describe the characterization of EPZ-5676, a potent and selective aminonucleoside inhibitor of DOT1L histone methyltransferase activity. The compound has an inhibition constant value of 80 pM, and demonstrates 37 000-fold selectivity over all other methyltransferases tested. In cellular studies, EPZ-5676 inhibited H3K79 methylation and MLL-fusion target gene expression and demonstrated potent cell killing that was selective for acute leukemia lines bearing MLL translocations. Continuous IV infusion of EPZ-5676 in a rat xenograft model of MLL-rearranged leukemia caused complete tumor regressions that were sustained well beyond the compound infusion period with no significant weight loss or signs of toxicity. EPZ-5676 is therefore a potential treatment of MLL-rearranged leukemia and is under clinical investigation.
- Published
- 2013
- Full Text
- View/download PDF
29. Mechanisms of Pinometostat (EPZ-5676) Treatment-Emergent Resistance in
- Author
-
Carly T, Campbell, Jessica N, Haladyna, David A, Drubin, Ty M, Thomson, Michael J, Maria, Taylor, Yamauchi, Nigel J, Waters, Edward J, Olhava, Roy M, Pollock, Jesse J, Smith, Robert A, Copeland, Stephen J, Blakemore, Kathrin M, Bernt, and Scott R, Daigle
- Subjects
Gene Rearrangement ,ATP Binding Cassette Transporter, Subfamily B ,Leukemia ,Gene Expression Regulation, Leukemic ,Lysine ,Histone-Lysine N-Methyltransferase ,Methylation ,Models, Biological ,Histones ,Drug Resistance, Neoplasm ,Cell Line, Tumor ,Biomarkers, Tumor ,Humans ,Benzimidazoles ,RNA, Messenger ,Myeloid-Lymphoid Leukemia Protein - Abstract
DOT1L is a protein methyltransferase involved in the development and maintenance of
- Published
- 2016
30. Preclinical Evidence of Anti-Tumor Activity Induced by EZH2 Inhibition in Human Models of Synovial Sarcoma
- Author
-
Jesse S. Smith, Sarah K. Knutson, Heike Keilhack, Alexandra R. Grassian, Scott Ribich, Roy M. Pollock, Galina Kuznetsov, Yukinori Minoshima, Satoshi Kawano, Robert A. Copeland, Masumi Tsuda, Kevin K. Kuntz, Shanqin Xu, Shinya Tanaka, Yonghong Xiao, Junji Matsui, and Natalie Warholic
- Subjects
0301 basic medicine ,Cancer Treatment ,Gene Expression ,lcsh:Medicine ,Apoptosis ,Biochemistry ,Histones ,0302 clinical medicine ,Medicine and Health Sciences ,lcsh:Science ,Cultured Tumor Cells ,Multidisciplinary ,Cell Death ,biology ,Chromosome Biology ,Sarcomas ,EZH2 ,Sarcoma Cells ,Synovial sarcoma ,Chromosomal Aberrations ,Chromatin ,Histone ,Oncology ,Cell Processes ,030220 oncology & carcinogenesis ,Translocations ,Biological Cultures ,Karyotypes ,PRC2 ,Research Article ,macromolecular substances ,Research and Analysis Methods ,Chromatin remodeling ,Cytogenetics ,03 medical and health sciences ,Histone H3 ,DNA-binding proteins ,Genetics ,medicine ,Biology and life sciences ,Cell growth ,lcsh:R ,Cancers and Neoplasms ,Proteins ,Cell Biology ,Cell Cultures ,medicine.disease ,030104 developmental biology ,biology.protein ,Cancer research ,lcsh:Q - Abstract
The catalytic activities of covalent and ATP-dependent chromatin remodeling are central to regulating the conformational state of chromatin and the resultant transcriptional output. The enzymes that catalyze these activities are often contained within multiprotein complexes in nature. Two such multiprotein complexes, the polycomb repressive complex 2 (PRC2) methyltransferase and the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeler have been reported to act in opposition to each other during development and homeostasis. An imbalance in their activities induced by mutations/deletions in complex members (e.g. SMARCB1) has been suggested to be a pathogenic mechanism in certain human cancers. Here we show that preclinical models of synovial sarcoma—a cancer characterized by functional SMARCB1 loss via its displacement from the SWI/SNF complex through the pathognomonic SS18-SSX fusion protein—display sensitivity to pharmacologic inhibition of EZH2, the catalytic subunit of PRC2. Treatment with tazemetostat, a clinical-stage, selective and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity reverses a subset of synovial sarcoma gene expression and results in concentration-dependent cell growth inhibition and cell death specifically in SS18-SSX fusion-positive cells in vitro. Treatment of mice bearing either a cell line or two patient-derived xenograft models of synovial sarcoma leads to dose-dependent tumor growth inhibition with correlative inhibition of trimethylation levels of the EZH2-specific substrate, lysine 27 on histone H3. These data demonstrate a dependency of SS18-SSX-positive, SMARCB1-deficient synovial sarcomas on EZH2 enzymatic activity and suggests the potential utility of EZH2-targeted drugs in these genetically defined cancers.
- Published
- 2016
31. A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells
- Author
-
Natalie Warholic, Christina R. Majer, Christopher J. Sneeringer, Christina J. Allain, Edward J. Olhava, Mikel P. Moyer, Sarah K. Knutson, Heike Keilhack, J. Joshua Smith, Lei Jin, Joelle D. Sacks, Robert A. Copeland, Tim J. Wigle, Richard Chesworth, Margaret Porter Scott, Jeffrey Song, Alejandra Raimondi, Kevin Wayne Kuntz, Roy M. Pollock, Victoria M. Richon, and Christine Klaus
- Subjects
Indazoles ,Methyltransferase ,Lymphoma ,Pyridones ,Mutant ,Antineoplastic Agents ,macromolecular substances ,Methylation ,Histones ,Structure-Activity Relationship ,medicine ,Humans ,Point Mutation ,Enhancer of Zeste Homolog 2 Protein ,Epigenetics ,Enzyme Inhibitors ,Molecular Biology ,Cell Proliferation ,Cell Death ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,Lysine ,Point mutation ,Cell Cycle ,fungi ,EZH2 ,Polycomb Repressive Complex 2 ,Cell Biology ,medicine.disease ,Chromatin ,Cancer research ,biology.protein ,PRC2 - Abstract
EZH2 catalyzes trimethylation of histone H3 lysine 27 (H3K27). Point mutations of EZH2 at Tyr641 and Ala677 occur in subpopulations of non-Hodgkin's lymphoma, where they drive H3K27 hypertrimethylation. Here we report the discovery of EPZ005687, a potent inhibitor of EZH2 (K(i) of 24 nM). EPZ005687 has greater than 500-fold selectivity against 15 other protein methyltransferases and has 50-fold selectivity against the closely related enzyme EZH1. The compound reduces H3K27 methylation in various lymphoma cells; this translates into apoptotic cell killing in heterozygous Tyr641 or Ala677 mutant cells, with minimal effects on the proliferation of wild-type cells. These data suggest that genetic alteration of EZH2 (for example, mutations at Tyr641 or Ala677) results in a critical dependency on enzymatic activity for proliferation (that is, the equivalent of oncogene addiction), thus portending the clinical use of EZH2 inhibitors for cancers in which EZH2 is genetically altered.
- Published
- 2012
- Full Text
- View/download PDF
32. MLL-Rearranged Leukemia Is Dependent on Aberrant H3K79 Methylation by DOT1L
- Author
-
Joerg Faber, Nan Zhu, Sridhar Vempati, Andrei V. Krivtsov, Lars Bullinger, Kathrin M. Bernt, Amit U. Sinha, Andrew L. Kung, Roy M. Pollock, Zhaohui Feng, Amanda Daigle, Scott A. Armstrong, Natalie Punt, and Victoria M. Richon
- Subjects
Cancer Research ,Oncogene Proteins, Fusion ,Cellular differentiation ,Apoptosis ,Biology ,Methylation ,Article ,Histones ,Mice ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Epigenetics ,Myeloid Ecotropic Viral Integration Site 1 Protein ,neoplasms ,Myeloid Progenitor Cells ,030304 developmental biology ,Gene Rearrangement ,Homeodomain Proteins ,0303 health sciences ,Lysine ,Myelodysplastic syndromes ,Cell Cycle ,Cell Differentiation ,Cell Biology ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,DOT1L ,medicine.disease ,Molecular biology ,Hematopoiesis ,Neoplasm Proteins ,3. Good health ,Leukemia ,Cell Transformation, Neoplastic ,Oncology ,Genetic Loci ,030220 oncology & carcinogenesis ,Histone methyltransferase ,Cancer research ,H3K4me3 ,Protein Processing, Post-Translational ,Myeloid-Lymphoid Leukemia Protein - Abstract
SummaryThe histone 3 lysine 79 (H3K79) methyltransferase Dot1l has been implicated in the development of leukemias bearing translocations of the Mixed Lineage Leukemia (MLL) gene. We identified the MLL-fusion targets in an MLL-AF9 leukemia model, and conducted epigenetic profiling for H3K79me2, H3K4me3, H3K27me3, and H3K36me3 in hematopoietic progenitor and leukemia stem cells (LSCs). We found abnormal profiles only for H3K79me2 on MLL-AF9 fusion target loci in LSCs. Inactivation of Dot1l led to downregulation of direct MLL-AF9 targets and an MLL translocation-associated gene expression signature, whereas global gene expression remained largely unaffected. Suppression of MLL translocation-associated gene expression corresponded with dependence of MLL-AF9 leukemia on Dot1l in vivo. These data point to DOT1L as a potential therapeutic target in MLL-rearranged leukemia.
- Published
- 2011
- Full Text
- View/download PDF
33. Ridaforolimus (AP23573; MK-8669), a Potent mTOR Inhibitor, Has Broad Antitumor Activity and Can Be Optimally Administered Using Intermittent Dosing Regimens
- Author
-
Victor M. Rivera, Roy M. Pollock, Lori Berk, Frank Wang, Narayana I. Narasimhan, Tim Clackson, Rachel M. Squillace, Scott Wardwell, David Miller, John Iuliucci, and Yaoyu Ning
- Subjects
Male ,Vascular Endothelial Growth Factor A ,Cancer Research ,Angiogenesis ,Cell Growth Processes ,Pharmacology ,Ridaforolimus ,Mice ,chemistry.chemical_compound ,Cell Line, Tumor ,Animals ,Humans ,PTEN ,Dosing ,Phosphorylation ,Mode of action ,PI3K/AKT/mTOR pathway ,Sirolimus ,Mice, Inbred BALB C ,Antibiotics, Antineoplastic ,Neovascularization, Pathologic ,biology ,Cell growth ,TOR Serine-Threonine Kinases ,PTEN Phosphohydrolase ,Endothelial Cells ,HCT116 Cells ,Xenograft Model Antitumor Assays ,Mice, Inbred C57BL ,Oncogene Protein v-akt ,Vascular endothelial growth factor ,Glucose ,Oncology ,chemistry ,biology.protein ,Female - Abstract
The mTOR pathway is hyperactivated through oncogenic transformation in many human malignancies. Ridaforolimus (AP23573; MK-8669) is a novel rapamycin analogue that selectively targets mTOR and is currently under clinical evaluation. In this study, we investigated the mechanistic basis for the antitumor activity of ridaforolimus in a range of human tumor types, exploring potential markers of response, and determining optimal dosing regimens to guide clinical studies. Administration of ridaforolimus to tumor cells in vitro elicited dose-dependent inhibition of mTOR activity with concomitant effects on cell growth and division. We showed that ridaforolimus exhibits a predominantly cytostatic mode of action, consistent with the findings for other mTOR inhibitors. Potent inhibitory effects on vascular endothelial growth factor secretion, endothelial cell growth, and glucose metabolism were also observed. Although PTEN and/or phosphorylated AKT status have been proposed as potential mTOR pathway biomarkers, neither was predictive for ridaforolimus responsiveness in the heterogeneous panel of cancer cell lines examined. In mouse models, robust antitumor activity was observed in human tumor xenografts using a series of intermittent dosing schedules, consistent with pharmacodynamic observations of mTOR pathway inhibition for at least 72 hours following dosing. Parallel skin-graft rejection studies established that intermittent dosing schedules lack the immunosuppressive effects seen with daily dosing. Overall these findings show the broad inhibitory effects of ridaforolimus on cell growth, division, metabolism, and angiogenesis, and support the use of intermittent dosing as a means to optimize antitumor activity while minimizing systemic effects. Mol Cancer Ther; 10(6); 1059–71. ©2011 AACR.
- Published
- 2011
- Full Text
- View/download PDF
34. Genetic and Pharmacological Inhibition of PDK1 in Cancer Cells
- Author
-
Youyuan Xu, Cloud P. Paweletz, Maciej Wiznerowicz, Victoria M. Richon, Sanjeev Munshi, Brian Dolinski, Kumiko Nagashima, Anke Klippel, Sujal V. Deshmukh, Peter Blume-Jensen, Bo-Sheng Pan, Jannik N. Andersen, Alan B. Northrup, Timothy Allison, Alexander A. Szewczak, Zangwei Xu, Manfred Kraus, Heike Keilhack, Sriram Sathyanarayanan, Youwei Yan, Albert H. Y. Chen, Uwe Mueller, Lixia Li, An Chi, Thi D.T. Nguyen, Ekaterina V. Bobkova, Roy M. Pollock, Bart Lutterbach, and Stuart D. Shumway
- Subjects
Gene knockdown ,animal structures ,Kinase ,Allosteric regulation ,Cell Biology ,Biology ,medicine.disease_cause ,Biochemistry ,Cell biology ,Cell culture ,Cancer cell ,medicine ,Phosphorylation ,Carcinogenesis ,Molecular Biology ,Protein kinase B - Abstract
Phosphoinositide-dependent kinase 1 (PDK1) is a critical activator of multiple prosurvival and oncogenic protein kinases and has garnered considerable interest as an oncology drug target. Despite progress characterizing PDK1 as a therapeutic target, pharmacological support is lacking due to the prevalence of nonspecific inhibitors. Here, we benchmark literature and newly developed inhibitors and conduct parallel genetic and pharmacological queries into PDK1 function in cancer cells. Through kinase selectivity profiling and x-ray crystallographic studies, we identify an exquisitely selective PDK1 inhibitor (compound 7) that uniquely binds to the inactive kinase conformation (DFG-out). In contrast to compounds 1–5, which are classical ATP-competitive kinase inhibitors (DFG-in), compound 7 specifically inhibits cellular PDK1 T-loop phosphorylation (Ser-241), supporting its unique binding mode. Interfering with PDK1 activity has minimal antiproliferative effect on cells growing as plastic-attached monolayer cultures (i.e. standard tissue culture conditions) despite reduced phosphorylation of AKT, RSK, and S6RP. However, selective PDK1 inhibition impairs anchorage-independent growth, invasion, and cancer cell migration. Compound 7 inhibits colony formation in a subset of cancer cell lines (four of 10) and primary xenograft tumor lines (nine of 57). RNAi-mediated knockdown corroborates the PDK1 dependence in cell lines and identifies candidate biomarkers of drug response. In summary, our profiling studies define a uniquely selective and cell-potent PDK1 inhibitor, and the convergence of genetic and pharmacological phenotypes supports a role of PDK1 in tumorigenesis in the context of three-dimensional in vitro culture systems.
- Published
- 2011
- Full Text
- View/download PDF
35. Coordinated activities of wild-type plus mutant EZH2 drive tumor-associated hypertrimethylation of lysine 27 on histone H3 (H3K27) in human B-cell lymphomas
- Author
-
Roy M. Pollock, Victoria M. Richon, Robert A. Copeland, Margaret Porter Scott, Christopher J. Sneeringer, Sarah K. Knutson, and Kevin Wayne Kuntz
- Subjects
Lymphoma, B-Cell ,Mutant ,macromolecular substances ,Methylation ,Catalysis ,Histones ,Histone H3 ,Humans ,Point Mutation ,Enhancer of Zeste Homolog 2 Protein ,Multidisciplinary ,biology ,Lysine ,EZH2 ,Polycomb Repressive Complex 2 ,Wild type ,Biological Sciences ,Molecular biology ,DNA-Binding Proteins ,Kinetics ,Histone ,Histone methyltransferase ,biology.protein ,PRC2 ,Transcription Factors - Abstract
EZH2, the catalytic subunit of the PRC2 complex, catalyzes the mono- through trimethylation of lysine 27 on histone H3 (H3K27). Histone H3K27 trimethylation is a mechanism for suppressing transcription of specific genes that are proximal to the site of histone modification. Point mutations of the EZH2 gene (Tyr641) have been reported to be linked to subsets of human B-cell lymphoma. The mutant allele is always found associated with a wild-type allele (heterozygous) in disease cells, and the mutations were reported to ablate the enzymatic activity of the PRC2 complex for methylating an unmodified peptide substrate. Here we demonstrate that the WT enzyme displays greatest catalytic efficiency ( k cat / K ) for the zero to monomethylation reaction of H3K27 and diminished efficiency for subsequent (mono- to di- and di- to trimethylation) reactions. In stark contrast, the disease-associated Y641 mutations display very limited ability to perform the first methylation reaction, but have enhanced catalytic efficiency for the subsequent reactions, relative to the WT enzyme. These results imply that the malignant phenotype of disease requires the combined activities of a H3K27 monomethylating enzyme (PRC2 containing WT EZH2 or EZH1) together with the mutant PRC2s for augmented conversion of H3K27 to the trimethylated form. To our knowledge, this is the first example of a human disease that is dependent on the coordinated activities of normal and disease-associated mutant enzymatic function.
- Published
- 2010
- Full Text
- View/download PDF
36. Epigenetic approaches to cancer therapy
- Author
-
Victoria M. Richon and Roy M. Pollock
- Subjects
Pharmacology ,Methyltransferase ,biology ,Cancer ,medicine.disease ,Chromatin remodeling ,Chromatin ,Histone ,Histone methyltransferase ,Drug Discovery ,biology.protein ,Cancer research ,medicine ,Molecular Medicine ,Epigenetics ,Cancer epigenetics - Abstract
Epigenetic abnormalities are common in human cancer and play a key role in tumor progression through dysregulation of gene expression and chromatin function. The recent identification of the enzymes regulating epigenetic modifications and their altered function in cancer supports the possibility that these abnormalities can be reversed by inhibiting these enzymes. Indeed, drugs targeting such enzymes including DNA methyltransferases (DNMTs) and histone deacetylases (HDACs) have demonstrated antitumor activity in the clinic and inhibitors of DNMTs and HDACs are now approved as anticancer agents. As our understanding of cancer epigenetics grows, members of additional enzyme classes such as histone methyltransferases and demethylases are emerging as targets for future epigenetic cancer therapies.
- Published
- 2009
- Full Text
- View/download PDF
37. Quantitative Analysis of Histone Deacetylase-1 Selective Histone Modifications by Differential Mass Spectrometry
- Author
-
Fanyu Meng, Jonathan C. Cruz, Astrid M. Kral, Robert E. Settlage, Victoria M. Richon, Cloud P. Paweletz, Ronald C. Hendrickson, J. Paul Secrist, Roy M. Pollock, Thomas A. Miller, Nathan A. Yates, Anita Y. H. Lee, Nicole Ozerova, and Matthew G. Stanton
- Subjects
Proteomics ,Spectrometry, Mass, Electrospray Ionization ,animal structures ,Proteome ,Antineoplastic Agents ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Histone Deacetylases ,Mass Spectrometry ,Histones ,Histone H3 ,In vivo ,Cell Line, Tumor ,Histone H2B ,Humans ,Protein Isoforms ,False Positive Reactions ,Regulation of gene expression ,biology ,Chemistry ,General Chemistry ,HDAC1 ,enzymes and coenzymes (carbohydrates) ,Histone ,ROC Curve ,embryonic structures ,biology.protein ,Histone deacetylase ,biological phenomena, cell phenomena, and immunity ,Peptides ,Chromatography, Liquid - Abstract
Inhibitors of class 1 and class 2 histone deacetylase (HDAC) enzymes have shown antitumor activity in human clinical trials. More recently, there has been interest in developing subtype-selective HDAC inhibitors designed to retain anticancer activity while reducing potential side effects. Efforts have been initiated to selectively target HDAC1 given its role in tumor proliferation and survival. The development of HDAC1-specific inhibitors will require the identification of HDAC1-selective pharmacodynamic markers that correlate closely with HDAC1-inhibition in vitro and in vivo. Existing histone markers of HDAC target engagement were developed using pan-HDAC inhibitors and do not necessarily represent robust readouts for isoform-specific inhibitors. Therefore, we have initiated a proteomic approach to identify readouts for HDAC1 inhibition. This approach involves the use of differential mass spectrometry (dMS) to identify post-translational changes in histones by profiling histone-enriched cellular fractions treated with various HDAC inhibitors. In this study, we profiled histones isolated from the HCT116 human colon cancer cell line that have been treated with compounds from multiple chemical classes that are specific for HDAC1; HDAC1 and 3; and HDAC1, 3, and 6 enzymes. In two independent experiments, we identified 24 features that correlated with HDAC1-inhibition. Among the peptides modulated by HDAC1-selective inhibitors were Ac-H2B-K5 from histone H2B, and Ac-H3-K18 from histone H3. Commercially available antibodies to specific histone acetyl-lysine residues were used to confirm that these peptides also provide pharmacodynamic readouts for HDAC1-selective inhibitors in vivo and in vitro. These results show the utility of dMS in guiding the identification of specific readouts to aid in the development of HDAC-selective inhibitors.
- Published
- 2008
- Full Text
- View/download PDF
38. Metabolism and disposition of the DOT1L inhibitor, pinometostat (EPZ-5676), in rat, dog and human
- Author
-
Edward J. Olhava, James O’Neill, Nigel J. Waters, Mikel P. Moyer, Sherri Smith, Marie-Eve Rodrigue, Richard D. Burton, Kenneth W. Duncan, Roy M. Pollock, and Richard Chesworth
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Metabolite ,Antineoplastic Agents ,Biology ,Pharmacology ,Gut flora ,Toxicology ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,Feces ,0302 clinical medicine ,Dogs ,Pharmacokinetics ,Species Specificity ,Tandem Mass Spectrometry ,Internal medicine ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Large intestine ,Rats, Long-Evans ,Tissue Distribution ,Infusions, Intravenous ,Metabolism ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,biology.organism_classification ,Rats ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Oncology ,chemistry ,Excretory system ,030220 oncology & carcinogenesis ,Renal physiology ,Autoradiography ,Benzimidazoles ,Female ,Drug metabolism ,Chromatography, Liquid - Abstract
The metabolism and disposition of the first-in-class DOT1L inhibitor, EPZ-5676 (pinometostat), was investigated in rat and dog. Metabolite profiles were compared with those from adult patients in the first-in-man phase 1 study as well as the cross-species metabolism observed in vitro. EPZ-5676 was administered to rat and dog as a 24-h IV infusion of [14C]-EPZ-5676 for determination of pharmacokinetics, mass balance, metabolite profiling and biodistribution by quantitative whole-body autoradiography (QWBA). Metabolite profiling and identification was performed by radiometric and LC–MS/MS analysis. Fecal excretion was the major route of elimination, representing 79 and 81 % of the total dose in and rat and dog, respectively. QWBA in rats showed that the radioactivity was well distributed in the body, except for the central nervous system, and the majority of radioactivity was eliminated from most tissues by 168 h. Fecal recovery of dose-related material in bile duct-cannulated animals as well as higher radioactivity concentrations in the wall of the large intestine relative to liver implicated intestinal secretion as well as biliary elimination. EPZ-5676 underwent extensive oxidative metabolism with the major metabolic pathways being hydroxylation of the t-butyl group (EPZ007769) and N-dealkylation of the central nitrogen. Loss of adenine from parent EPZ-5676 (M7) was observed only in rat and dog feces, suggesting the involvement of gut microbiota. In rat and dog, steady-state plasma levels of total radioactivity and parent EPZ-5676 were attained rapidly and maintained through the infusion period before declining rapidly on cessation of dosing. Unchanged EPZ-5676 was the predominant circulating species in rat, dog and man. The excretory and metabolic pathways for EPZ-5676 were very similar across species. Renal excretion of both parent EPZ-5676 and EPZ-5676-related material was low, and in preclinical species fecal excretion of parent EPZ-5676 and EPZ007769 accounted for the majority of drug-related elimination.
- Published
- 2015
39. Structure and Property Guided Design in the Identification of PRMT5 Tool Compound EPZ015666
- Author
-
J. Joshua Smith, Christina R. Majer, Michael John Munchhof, Roy M. Pollock, Nigel J. Waters, Kristy G Kuplast, Lei Jin, L. Danielle Johnston, Lawrence A. Reiter, Nathalie Rioux, Richard Chesworth, Margaret Porter Scott, Elayne Chan-Penebre, P. Ann Boriack-Sjodin, Robert A. Copeland, Mikel P. Moyer, and Kenneth W. Duncan
- Subjects
0301 basic medicine ,03 medical and health sciences ,030104 developmental biology ,In vivo ,Chemistry ,Stereochemistry ,Protein arginine methyltransferase 5 ,Organic Chemistry ,Drug Discovery ,Computational biology ,Biochemistry - Abstract
The recent publication of a potent and selective inhibitor of protein methyltransferase 5 (PRMT5) provides the scientific community with in vivo-active tool compound EPZ015666 (GSK3235025) to probe the underlying pharmacology of this key enzyme. Herein, we report the design and optimization strategies employed on an initial hit compound with poor in vitro clearance to yield in vivo tool compound EPZ015666 and an additional potent in vitro tool molecule EPZ015866 (GSK3203591).
- Published
- 2015
40. MLL partial tandem duplication leukemia cells are sensitive to small molecule DOT1L inhibition
- Author
-
James E. Bradner, Michael Hadler, Scott A. Armstrong, Scott R. Daigle, Gang Huang, Michael A. Caligiuri, Edward J. Olhava, Andrei V. Krivtsov, Roy M. Pollock, Michael W.M. Kühn, and Richard Koche
- Subjects
Methyltransferase ,Cell Survival ,MLL Partial Tandem Duplication ,Antineoplastic Agents ,Apoptosis ,Biology ,Histones ,hemic and lymphatic diseases ,Cell Line, Tumor ,Gene Duplication ,Gene duplication ,medicine ,Humans ,Enzyme Inhibitors ,Online Only Articles ,neoplasms ,Cell Proliferation ,Leukemia ,Myeloid leukemia ,Hematology ,DOT1L ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,medicine.disease ,Drug Resistance, Neoplasm ,Tandem Repeat Sequences ,Cancer research ,Myeloid-Lymphoid Leukemia Protein ,Tandem exon duplication - Abstract
Genetic alterations of the mixed-lineage leukemia (MLL) gene are commonly implicated in the development of acute leukemias. In acute myeloid leukemia (AML) a partial tandem duplication (PTD) of MLL occurs in about 5%–11%1–4 of patients and has been linked to poor treatment outcome.1,5 Although recent studies show that MLL-PTD does not have a prognostic impact in CN-AML patients treated with intensive therapy,2,3 many patients still succumb to their disease,2,3,5–7 and the course of disease for patients not eligible for intensive chemotherapy is even more dismal. Thus, novel and innovative treatment approaches are needed. In acute leukemias, the MLL gene is also commonly affected by chromosomal rearrangements. Several MLL-rearranged leukemias were recently shown to be dependent on the only known histone 3 lysine 79 (H3K79) methyltransferase DOT1L.8,9 Genetic deletion of DOT1L impairs initiation and maintenance of MLL-rearranged leukemias in mouse models8–12 and pharmacological inhibition of DOT1L with the specific small molecules EPZ004777 or EPZ-5676 selectively kills MLL-rearranged leukemia cells in vitro13 and in vivo.14 Furthermore, DOT1L inhibition leads to a reversal of a characteristic gene expression signature, including downregulation of HOXA-cluster genes in MLL-rearranged leukemias.8–10 Based on these pre-clinical findings, EPZ-5676 is currently under investigation in a clinical phase I trial (clinicaltrials.gov identifier: 01684150). In the current study, we assessed whether MLL-PTD-positive leukemias that share critical biological features with MLL-rearranged leukemias, such as high expression of HOXA-cluster genes, similarly require DOT1L.
- Published
- 2015
41. MLL1 and DOT1L cooperate with meningioma-1 to induce acute myeloid leukemia
- Author
-
Matthew Bezzant, Craig T. Jordan, Kathrin M. Bernt, Brett M. Stevens, Qi Wei, Scott A. Armstrong, Patricia Ernst, Jessica Haladyna, Scott R. Daigle, Daniel A. Pollyea, Roy M. Pollock, Amit U. Sinha, Tobias Neff, and Simone S. Riedel
- Subjects
0301 basic medicine ,Male ,Methyltransferase ,Biology ,03 medical and health sciences ,Mice ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Epigenetics ,Myeloid Ecotropic Viral Integration Site 1 Protein ,Homeodomain Proteins ,Mice, Knockout ,Oncogene Proteins ,Tumor Suppressor Proteins ,Myeloid leukemia ,General Medicine ,DOT1L ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,medicine.disease ,Neoplasm Proteins ,Leukemia ,Haematopoiesis ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Histone methyltransferase ,Immunology ,Cancer research ,Trans-Activators ,Myeloid-Lymphoid Leukemia Protein ,Female ,Research Article - Abstract
Meningioma-1 (MN1) overexpression is frequently observed in patients with acute myeloid leukemia (AML) and is predictive of poor prognosis. In murine models, forced expression of MN1 in hematopoietic progenitors induces an aggressive myeloid leukemia that is strictly dependent on a defined gene expression program in the cell of origin, which includes the homeobox genes Hoxa9 and Meis1 as key components. Here, we have shown that this program is controlled by two histone methyltransferases, MLL1 and DOT1L, as deletion of either Mll1 or Dot1l in MN1-expressing cells abrogated the cell of origin-derived gene expression program, including the expression of Hoxa cluster genes. In murine models, genetic inactivation of either Mll1 or Dot1l impaired MN1-mediated leukemogenesis. We determined that HOXA9 and MEIS1 are coexpressed with MN1 in a subset of clinical MN1hi leukemia, and human MN1hi/HOXA9hi leukemias were sensitive to pharmacologic inhibition of DOT1L. Together, these data point to DOT1L as a potential therapeutic target in MN1hi AML. In addition, our findings suggest that epigenetic modulation of the interplay between an oncogenic lesion and its cooperating developmental program has therapeutic potential in AML.
- Published
- 2015
42. Inhibition of wild-type and mutant Bcr-Abl by AP23464, a potent ATP-based oncogenic protein kinase inhibitor: implications for CML
- Author
-
Victor M. Rivera, Hao Tang, Chester A. Metcalf, Michael W. Deininger, Tomi K. Sawyer, Brian J. Druker, Thomas O'Hare, Regine S. Bohacek, William C. Shakespeare, David C. Dalgarno, Erika M. Moseson, Raji Sundaramoorthi, Eric P. Stoffregen, Jeffrey A. Keats, Omar M. Abdullah, Yihan Wang, Tim Clackson, and Roy M. Pollock
- Subjects
Models, Molecular ,Fusion Proteins, bcr-abl ,Apoptosis ,Biochemistry ,Piperazines ,chemistry.chemical_compound ,Adenosine Triphosphate ,hemic and lymphatic diseases ,STAT5 Transcription Factor ,Amino Acids ,Enzyme Inhibitors ,Phosphorylation ,ABL ,Cell Cycle ,Nuclear Proteins ,Hematology ,Protein kinase inhibitor ,Milk Proteins ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Benzamides ,Imatinib Mesylate ,Tyrosine kinase ,Cell Division ,Proto-oncogene tyrosine-protein kinase Src ,medicine.drug ,Pyridones ,medicine.drug_class ,Immunology ,HL-60 Cells ,Biology ,Philadelphia chromosome ,Inhibitory Concentration 50 ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Humans ,Phosphotyrosine ,neoplasms ,Adaptor Proteins, Signal Transducing ,Tyrosine phosphorylation ,Imatinib ,Cell Biology ,medicine.disease ,Protein Structure, Tertiary ,Pyrimidines ,Imatinib mesylate ,chemistry ,Mutation ,Trans-Activators ,Cancer research ,K562 Cells - Abstract
The deregulated, oncogenic tyrosine kinase Bcr-Abl causes chronic myeloid leukemia (CML). Imatinib mesylate (Gleevec, STI571), a Bcr-Abl kinase inhibitor, selectively inhibits proliferation and promotes apoptosis of CML cells. Despite the success of imatinib mesylate in the treatment of CML, resistance is observed, particularly in advanced disease. The most common imatinib mesylate resistance mechanism involves Bcr-Abl kinase domain mutations that impart varying degrees of drug insensitivity. AP23464, a potent adenosine 5′-triphosphate (ATP)–based inhibitor of Src and Abl kinases, displays antiproliferative activity against a human CML cell line and Bcr-Abl–transduced Ba/F3 cells (IC50 = 14 nM; imatinib mesylate IC50 = 350 nM). AP23464 ablates Bcr-Abl tyrosine phosphorylation, blocks cell cycle progression, and promotes apoptosis of Bcr-Abl–expressing cells. Biochemical assays with purified glutathione S transferase (GST)–Abl kinase domain confirmed that AP23464 directly inhibits Abl activity. Importantly, the low nanomolar cellular and biochemical inhibitory properties of AP23464 extend to frequently observed imatinib mesylate–resistant Bcr-Abl mutants, including nucleotide binding P-loop mutants Q252H, Y253F, E255K, C-terminal loop mutant M351T, and activation loop mutant H396P. AP23464 was ineffective against mutant T315I, an imatinib mesylate contact residue. The potency of AP23464 against imatinib mesylate–refractory Bcr-Abl and its distinct binding mode relative to imatinib mesylate warrant further investigation of AP23464 for the treatment of CML.
- Published
- 2004
- Full Text
- View/download PDF
43. Dimerizer-regulated gene expression
- Author
-
Roy M. Pollock and Tim Clackson
- Subjects
Transcription, Genetic ,Recombinant Fusion Proteins ,Genetic enhancement ,Genetic Vectors ,Biomedical Engineering ,Chemical biology ,Bioengineering ,Computational biology ,Biology ,Transfection ,Bioinformatics ,Dexamethasone ,Mice ,Transcription (biology) ,Gene expression ,Animals ,Humans ,Gene ,Sirolimus ,Regulation of gene expression ,TOR Serine-Threonine Kinases ,DNA ,Dependovirus ,Phosphotransferases (Alcohol Group Acceptor) ,Methotrexate ,Gene Expression Regulation ,Carrier Proteins ,Dimerization ,Biotechnology - Abstract
Control of gene expression using small molecules is a powerful research tool and has clinical utility in the context of regulated gene therapy. Use of chemical inducers of dimerization, or dimerizers, for this purpose has several advantages, including tight regulation, modularity to facilitate iterative improvements, and assembly from human proteins to minimize immune responses in clinical applications. Recent developments include the use of the rapamycin-based dimerizer system to regulate the expression of endogenous genes, the generation of new chemical dimerizers based on FK506, dexamethasone and methotrexate, and progress towards the clinical use of adeno-associated virus and adenovirus vectors regulated by rapamycin analogs.
- Published
- 2002
- Full Text
- View/download PDF
44. Regulation of endogenous gene expression with a small-molecule dimerizer
- Author
-
Tim Clackson, Roy M. Pollock, Katja Linher, and Maryann Giel
- Subjects
Vascular Endothelial Growth Factor A ,Transcription, Genetic ,Biomedical Engineering ,Bioengineering ,Endogeny ,Endothelial Growth Factors ,Biology ,Kidney ,Applied Microbiology and Biotechnology ,Cell Line ,Transcription (biology) ,Gene expression ,Humans ,Gene ,Transcription factor ,Sirolimus ,Lymphokines ,Dose-Response Relationship, Drug ,Models, Genetic ,Vascular Endothelial Growth Factors ,Activator (genetics) ,Zinc Fingers ,Molecular biology ,Small molecule ,Fusion protein ,Gene Expression Regulation ,Intercellular Signaling Peptides and Proteins ,Molecular Medicine ,Dimerization ,Plasmids ,Transcription Factors ,Biotechnology - Abstract
Artificial transcription factors containing designer zinc-finger DNA-binding domains (DBDs) have been used to activate or repress expression of a growing number of endogenous genes. We have combined targeted zinc-finger DBD technology with a dimerizer-regulated gene expression system to permit the small-molecule control of endogenous gene transcription. We constructed a dimerizer-responsive transcription factor that incorporates an artificial zinc-finger DBD targeted to the promoter of the human VEGF gene. Introduction of this activator into human cells allowed expression of the chromosomal VEGF gene to be induced by a small-molecule dimerizer compound consisting of a nonimmunosuppressive rapamycin analog. We found that by directly regulating zinc-finger protein (ZFP) activity, we could circumvent difficulties encountered in the generation of cell lines stably expressing conventional unregulated activators. Dimerizer-dependent VEGF induction was rapid, tight, and dose dependent, and resulted in VEGF protein expression levels several-fold greater than those produced by the natural hypoxic response.
- Published
- 2002
- Full Text
- View/download PDF
45. Preclinical Evidence of Anti-Tumor Activity Induced by EZH2 Inhibition in Human Models of Synovial Sarcoma
- Author
-
Satoshi Kawano, Alexandra R Grassian, Masumi Tsuda, Sarah K Knutson, Natalie M Warholic, Galina Kuznetsov, Shanqin Xu, Yonghong Xiao, Roy M Pollock, Jesse J Smith, Kevin W Kuntz, Scott Ribich, Yukinori Minoshima, Junji Matsui, Robert A Copeland, Shinya Tanaka, and Heike Keilhack
- Subjects
Mice, Inbred BALB C ,Multidisciplinary ,Oncogene Proteins, Fusion ,lcsh:R ,Polycomb Repressive Complex 2 ,lcsh:Medicine ,Correction ,Mice, Nude ,Antineoplastic Agents ,SMARCB1 Protein ,Xenograft Model Antitumor Assays ,Mice ,Sarcoma, Synovial ,Cell Line, Tumor ,Animals ,Humans ,lcsh:Q ,Enhancer of Zeste Homolog 2 Protein ,lcsh:Science - Abstract
The catalytic activities of covalent and ATP-dependent chromatin remodeling are central to regulating the conformational state of chromatin and the resultant transcriptional output. The enzymes that catalyze these activities are often contained within multiprotein complexes in nature. Two such multiprotein complexes, the polycomb repressive complex 2 (PRC2) methyltransferase and the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeler have been reported to act in opposition to each other during development and homeostasis. An imbalance in their activities induced by mutations/deletions in complex members (e.g. SMARCB1) has been suggested to be a pathogenic mechanism in certain human cancers. Here we show that preclinical models of synovial sarcoma-a cancer characterized by functional SMARCB1 loss via its displacement from the SWI/SNF complex through the pathognomonic SS18-SSX fusion protein-display sensitivity to pharmacologic inhibition of EZH2, the catalytic subunit of PRC2. Treatment with tazemetostat, a clinical-stage, selective and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity reverses a subset of synovial sarcoma gene expression and results in concentration-dependent cell growth inhibition and cell death specifically in SS18-SSX fusion-positive cells in vitro. Treatment of mice bearing either a cell line or two patient-derived xenograft models of synovial sarcoma leads to dose-dependent tumor growth inhibition with correlative inhibition of trimethylation levels of the EZH2-specific substrate, lysine 27 on histone H3. These data demonstrate a dependency of SS18-SSX-positive, SMARCB1-deficient synovial sarcomas on EZH2 enzymatic activity and suggests the potential utility of EZH2-targeted drugs in these genetically defined cancers.
- Published
- 2017
- Full Text
- View/download PDF
46. Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-Hodgkin lymphoma
- Author
-
Kuan-Chun Huang, Mikel P. Moyer, Tim J. Wigle, Tadashi Kadowaki, Nigel J. Waters, Kevin Wayne Kuntz, J. Joshua Smith, Alejandra Raimondi, Roy M. Pollock, Mai Uesugi, Natalie Warholic, Margaret Porter-Scott, Akira Yokoi, Victoria M. Richon, Namita Kumar, Yonghong Xiao, Christina J. Allain, Galina Kuznetsov, Sarah K. Knutson, Heike Keilhack, Robert A. Copeland, Richard Chesworth, Toshimitsu Uenaka, Yukinori Minoshima, Satoshi Kawano, and Christine Klaus
- Subjects
Male ,Cancer Research ,Pyridones ,Morpholines ,Molecular Sequence Data ,Antineoplastic Agents ,Apoptosis ,macromolecular substances ,Mice, SCID ,Pharmacology ,Biology ,Rats, Sprague-Dawley ,Histone H3 ,Mice ,Catalytic Domain ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Point Mutation ,Lymphoma, Non-Hodgkin ,EZH2 ,Biphenyl Compounds ,Cell Cycle ,Polycomb Repressive Complex 2 ,Cancer ,Methylation ,medicine.disease ,Xenograft Model Antitumor Assays ,Lymphoma ,Rats ,Gene Expression Regulation, Neoplastic ,Cell killing ,Oncology ,Mechanism of action ,Cell culture ,Benzamides ,Female ,medicine.symptom - Abstract
Mutations within the catalytic domain of the histone methyltransferase EZH2 have been identified in subsets of patients with non-Hodgkin lymphoma (NHL). These genetic alterations are hypothesized to confer an oncogenic dependency on EZH2 enzymatic activity in these cancers. We have previously reported the discovery of EPZ005678 and EPZ-6438, potent and selective S-adenosyl-methionine-competitive small molecule inhibitors of EZH2. Although both compounds are similar with respect to their mechanism of action and selectivity, EPZ-6438 possesses superior potency and drug-like properties, including good oral bioavailability in animals. Here, we characterize the activity of EPZ-6438 in preclinical models of NHL. EPZ-6438 selectively inhibits intracellular lysine 27 of histone H3 (H3K27) methylation in a concentration- and time-dependent manner in both EZH2 wild-type and mutant lymphoma cells. Inhibition of H3K27 trimethylation (H3K27Me3) leads to selective cell killing of human lymphoma cell lines bearing EZH2 catalytic domain point mutations. Treatment of EZH2-mutant NHL xenograft-bearing mice with EPZ-6438 causes dose-dependent tumor growth inhibition, including complete and sustained tumor regressions with correlative diminution of H3K27Me3 levels in tumors and selected normal tissues. Mice dosed orally with EPZ-6438 for 28 days remained tumor free for up to 63 days after stopping compound treatment in two EZH2-mutant xenograft models. These data confirm the dependency of EZH2-mutant NHL on EZH2 activity and portend the utility of EPZ-6438 as a potential treatment for these genetically defined cancers. Mol Cancer Ther; 13(4); 842–54. ©2014 AACR.
- Published
- 2014
47. Delivery of a stringent dimerizer-regulated gene expression system in a single retroviral vector
- Author
-
Sridaran Natesan, Roy M. Pollock, Robbyn Issner, Tim Clackson, Karen Zoller, and Victor M. Rivera
- Subjects
Transcription, Genetic ,Recombinant Fusion Proteins ,Genetic Vectors ,Regulatory Sequences, Nucleic Acid ,Biology ,Transfection ,Cell Line ,Viral vector ,Mice ,Retrovirus ,Gene expression ,Tumor Cells, Cultured ,Animals ,Humans ,Immunophilins ,Transcription factor ,Sirolimus ,Regulation of gene expression ,Multidisciplinary ,TOR Serine-Threonine Kinases ,3T3 Cells ,Biological Sciences ,biology.organism_classification ,Molecular biology ,Long terminal repeat ,Kinetics ,Phosphotransferases (Alcohol Group Acceptor) ,Retroviridae ,Gene Expression Regulation ,Carrier Proteins ,Dimerization - Abstract
Small molecule-regulated transcription has broad utility and would benefit from an easily delivered self-contained regulatory cassette capable of robust, tightly controlled target gene expression. We describe the delivery of a modified dimerizer-regulated gene expression system to cells on a single retrovirus. A transcription factor cassette responsive to the natural product dimerizer rapamycin was optimized for retroviral delivery by fusing a highly potent chimeric activation domain to the rapamycin-binding domain of FKBP-rapamycin-associated protein (FRAP). This improvement led to an increase in both the potency and maximal levels of gene expression induced by rapamycin, or nonimmunosuppressive rapamycin analogs. The modified transcription factor cassette was incorporated along with a target gene into a single rapamycin-responsive retrovirus. Cell pools stably transduced with the single virus system displayed negligible basal expression and gave induction ratios of at least three orders of magnitude in the presence of rapamycin or a nonimmunosuppressive rapamycin analog. Levels of induced gene expression were comparable to those obtained with the constitutive retroviral long terminal repeat and the single virus system performed well in four different mammalian cell lines. Regulation with the dimerizer-responsive retrovirus was tight enough to allow the generation of cell lines displaying inducible expression of the highly toxic diphtheria toxin A chain gene. The ability to deliver the tightly inducible rapamycin system in a single retrovirus should facilitate its use in the study of gene function in a broad range of cell types.
- Published
- 2000
- Full Text
- View/download PDF
48. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2
- Author
-
Mikel P. Moyer, Tim J. Wigle, Christina J. Allain, Alejandra Raimondi, Roy M. Pollock, Natalie Warholic, Robert A. Copeland, Christine Klaus, Kevin Wayne Kuntz, Sarah K. Knutson, Heike Keilhack, Margaret Porter Scott, Richard Chesworth, and Victoria M. Richon
- Subjects
Methyltransferase ,Pyridines ,Lysine ,Antineoplastic Agents ,Apoptosis ,Epigenesis, Genetic ,Histones ,Histone H3 ,Mice ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Rhabdoid Tumor ,Cell Proliferation ,Multidisciplinary ,biology ,Cell growth ,Gene Expression Profiling ,EZH2 ,Biphenyl Compounds ,Polycomb Repressive Complex 2 ,Biological Sciences ,Molecular biology ,Biphenyl compound ,Histone ,HEK293 Cells ,Drug Design ,biology.protein ,Neoplasm Transplantation - Abstract
Inactivation of the switch/sucrose nonfermentable complex component SMARCB1 is extremely prevalent in pediatric malignant rhabdoid tumors (MRTs) or atypical teratoid rhabdoid tumors. This alteration is hypothesized to confer oncogenic dependency on EZH2 in these cancers. We report the discovery of a potent, selective, and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity, (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(morpholinomethyl)-[1,1′-biphenyl]-3-carboxamide). The compound induces apoptosis and differentiation specifically in SMARCB1 -deleted MRT cells. Treatment of xenograft-bearing mice with (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(morpholinomethyl)-[1,1′-biphenyl]-3-carboxamide) leads to dose-dependent regression of MRTs with correlative diminution of intratumoral trimethylation levels of lysine 27 on histone H3, and prevention of tumor regrowth after dosing cessation. These data demonstrate the dependency of SMARCB1 mutant MRTs on EZH2 enzymatic activity and portend the utility of EZH2-targeted drugs for the treatment of these genetically defined cancers.
- Published
- 2013
49. Leukemic transformation by the MLL-AF6 fusion oncogene requires the H3K79 methyltransferase Dot1l
- Author
-
Kathrin M. Bernt, Liying Chen, Amit U. Sinha, Stuart Dias, Victoria M. Richon, Scott A. Armstrong, Scott R. Daigle, Jenny Chang, Maurizio Fazio, Edward J. Olhava, Aniruddha J. Deshpande, Roy M. Pollock, and Deepti Banka
- Subjects
Adenosine ,Oncogene Proteins ,Oncogene Proteins, Fusion ,Immunology ,Kinesins ,Chromosomal translocation ,Biology ,Myosins ,Biochemistry ,Models, Biological ,Mice ,hemic and lymphatic diseases ,Conditional gene knockout ,medicine ,Animals ,Enzyme Inhibitors ,neoplasms ,Cells, Cultured ,Cell Proliferation ,Mice, Knockout ,Myeloid Neoplasia ,Oncogene ,Lysine ,Phenylurea Compounds ,Cell Biology ,Hematology ,DOT1L ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,medicine.disease ,Molecular biology ,Mice, Inbred C57BL ,Leukemia ,Cell Transformation, Neoplastic ,Histone methyltransferase ,Histone Methyltransferases ,Chromatin immunoprecipitation ,Myeloid-Lymphoid Leukemia Protein - Abstract
The t(6;11)(q27;q23) is a recurrent chromosomal rearrangement that encodes the MLLAF6 fusion oncoprotein and is observed in patients with diverse hematologic malignancies. The presence of the t(6;11)(q27;q23) has been linked to poor overall survival in patients with AML. In this study, we demonstrate that MLL-AF6 requires continued activity of the histone-methyltransferase DOT1L to maintain expression of the MLL-AF6-driven oncogenic gene-expression program. Using gene-expression analysis and genome-wide chromatin immunoprecipitation studies followed by next generation sequencing, we found that MLL-fusion target genes display markedly high levels of histone 3 at lysine 79 (H3K79) dimethylation in murine MLL-AF6 leukemias as well as in ML2, a human myelomonocytic leukemia cell line bearing the t(6;11)(q27;q23) translocation. Targeted disruption of Dot1l using a conditional knockout mouse model inhibited leukemogenesis mediated by the MLL-AF6 fusion oncogene. Moreover, both murine MLL-AF6–transformed cells as well as the human MLL-AF6–positive ML2 leukemia cell line displayed specific sensitivity to EPZ0004777, a recently described, selective, small-molecule inhibitor of Dot1l. Dot1l inhibition resulted in significantly decreased proliferation, decreased expression of MLL-AF6 target genes, and cell cycle arrest of MLL-AF6–transformed cells. These results indicate that patients bearing the t(6;11)(q27;q23) translocation may benefit from therapeutic agents targeting aberrant H3K79 methylation.
- Published
- 2013
50. Abrogation of MLL-AF10 and CALM-AF10-mediated transformation through genetic inactivation or pharmacological inhibition of the H3K79 methyltransferase Dot1l
- Author
-
Kathrin M. Bernt, Edward J. Olhava, Stuart Dias, Victoria M. Richon, Deepti Banka, Christian Buske, Roy M. Pollock, Liying Chen, Aniruddha J. Deshpande, Scott R. Daigle, and Scott A. Armstrong
- Subjects
Cancer Research ,Oncogene Proteins, Fusion ,Blotting, Western ,Fluorescent Antibody Technique ,Chromosomal translocation ,Apoptosis ,Biology ,Real-Time Polymerase Chain Reaction ,Leukemogenic ,Article ,Mice ,hemic and lymphatic diseases ,Conditional gene knockout ,medicine ,Gene silencing ,Animals ,Humans ,Gene Silencing ,Enzyme Inhibitors ,neoplasms ,Cell Proliferation ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Cycle ,Myeloid leukemia ,Hematology ,DOT1L ,Methyltransferases ,medicine.disease ,Molecular biology ,Mice, Mutant Strains ,Leukemia ,medicine.anatomical_structure ,Oncology ,Cancer research ,Bone marrow ,Myeloid-Lymphoid Leukemia Protein - Abstract
The t(10;11)(p12;q23) translocation and the t(10;11)(p12;q14) translocation, which encode the MLL (mixed lineage leukemia)-AF10 and CALM (clathrin assembly lymphoid myeloid leukemia)-AF10 fusion oncoproteins, respectively, are two recurrent chromosomal rearrangements observed in patients with acute myeloid leukemia and acute lymphoblastic leukemia. Here, we demonstrate that MLL-AF10 and CALM-AF10-mediated transformation is dependent on the H3K79 methyltransferase Dot1l using genetic and pharmacological approaches in mouse models. Targeted disruption of Dot1l using a conditional knockout mouse model abolished in vitro transformation of murine bone marrow cells and in vivo initiation and maintenance of MLL-AF10 or CALM-AF10 leukemia. The treatment of MLL-AF10 and CALM-AF10 transformed cells with EPZ004777, a specific small-molecule inhibitor of Dot1l, suppressed expression of leukemogenic genes such as Hoxa cluster genes and Meis1, and selectively impaired proliferation of MLL-AF10 and CALM-AF10 transformed cells. Pretreatment with EPZ004777 profoundly decreased the in vivo spleen-colony-forming ability of MLL-AF10 or CALM-AF10 transformed bone marrow cells. These results show that patients with leukemia-bearing chromosomal translocations that involve the AF10 gene may benefit from small-molecule therapeutics that inhibit H3K79 methylation.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.