51
|
Lee-Sherick AB, Zhang W, Menachof KK, Hill AA, Rinella S, Kirkpatrick G, Page LS, Stashko MA, Jordan CT, Wei Q, Liu J, Zhang D, DeRyckere D, Wang X, Frye S, Earp HS, Graham DK. Efficacy of a Mer and Flt3 tyrosine kinase small molecule inhibitor, UNC1666, in acute myeloid leukemia. Oncotarget 2015; 6:6722-36. [PMID: 25762638 PMCID: PMC4466645 DOI: 10.18632/oncotarget.3156] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/15/2015] [Indexed: 01/28/2023] Open
Abstract
Mer and Flt3 receptor tyrosine kinases have been implicated as therapeutic targets in acute myeloid leukemia (AML). In this manuscript we describe UNC1666, a novel ATP-competitive small molecule tyrosine kinase inhibitor, which potently diminishes Mer and Flt3 phosphorylation in AML. Treatment with UNC1666 mediated biochemical and functional effects in AML cell lines expressing Mer or Flt3 internal tandem duplication (ITD), including decreased phosphorylation of Mer, Flt3 and downstream effectors Stat, Akt and Erk, induction of apoptosis in up to 98% of cells, and reduction of colony formation by greater than 90%, compared to treatment with vehicle. These effects were dose-dependent, with inhibition of downstream signaling and functional effects correlating with the degree of Mer or Flt3 kinase inhibition. Treatment of primary AML patient samples expressing Mer and/or Flt3-ITD with UNC1666 also inhibited Mer and Flt3 intracellular signaling, induced apoptosis, and inhibited colony formation. In summary, UNC1666 is a novel potent small molecule tyrosine kinase inhibitor that decreases oncogenic signaling and myeloblast survival, thereby validating dual Mer/Flt3 inhibition as an attractive treatment strategy for AML.
Collapse
Affiliation(s)
| | - Weihe Zhang
- University of North Carolina, Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | | | - Amanda A. Hill
- University of Colorado, Department of Pediatrics, Aurora, CO, USA
| | - Sean Rinella
- University of Colorado, Department of Pediatrics, Aurora, CO, USA
| | | | - Lauren S. Page
- University of Colorado, Department of Pediatrics, Aurora, CO, USA
| | - Michael A. Stashko
- University of North Carolina, Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Craig T. Jordan
- University of Colorado, Department of Medicine, Aurora, CO, USA
| | - Qi Wei
- Children's Hospital Colorado, Department of Pathology, Aurora, CO, USA
| | - Jing Liu
- University of North Carolina, Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Dehui Zhang
- University of North Carolina, Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | | | - Xiaodong Wang
- University of North Carolina, Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - Stephen Frye
- University of North Carolina, Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | - H. Shelton Earp
- University of North Carolina, Department of Medicine, Chapel Hill, NC, USA
| | | |
Collapse
|
52
|
Jiang Y, Ludwig J, Janku F. Targeted therapies for advanced Ewing sarcoma family of tumors. Cancer Treat Rev 2015; 41:391-400. [PMID: 25869102 DOI: 10.1016/j.ctrv.2015.03.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/17/2015] [Accepted: 03/20/2015] [Indexed: 12/30/2022]
Abstract
The prognosis of adolescent and young adult patients battling metastatic Ewing sarcoma family of tumors (ESFT) remains less than 30% despite the development of systemic therapies. In the era of personalized medicine, novel molecular targets have been tested in preclinical or clinical settings in ESFT. In this review, we focus on early clinical and translational research that identified multiple molecular targets, including IGF-1R; mTOR; tyrosine kinase inhibitors; EWS-FLI1-related targets, and others. Overall, novel targeted therapies demonstrated modest efficacy; however pronounced and durable antineoplastic responses have been observed in small subsets of treated patients, for example with IGF-1R antibodies. Identifying outcome-predicting biomarkers and overcoming treatment resistance remain major challenges. Due to the rarity of ESFT, multi-institutional collaboration efforts of clinicians, basic and translational scientists are needed in order to understand biology of therapeutic response or resistance, which can lead to development of novel therapeutic methods and improved patient outcomes.
Collapse
Affiliation(s)
- Yunyun Jiang
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph Ludwig
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Filip Janku
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
53
|
Secondary mutations as mediators of resistance to targeted therapy in leukemia. Blood 2015; 125:3236-45. [PMID: 25795921 DOI: 10.1182/blood-2014-10-605808] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 03/12/2015] [Indexed: 02/07/2023] Open
Abstract
The advent of small molecule-based targeted therapy has improved the treatment of both acute and chronic leukemias. Resistance to small molecule inhibitors has emerged as a common theme. The most frequent mode of acquired resistance is the acquisition of point mutations in the kinase domain. FLT3 inhibitors have improved response rates in FLT3-mutated acute myeloid leukemia (AML). The occurrence of the ATP-binding site and activation loop mutations confers varying degrees of resistance to the individual FLT3 inhibitors. Second-generation FLT3 inhibitors such as crenolanib may overcome the resistance of these mutations. Furthermore, nonmutational mechanisms of resistance such as prosurvival pathways and bone marrow signaling may be upregulated in FLT3 inhibitor-resistant AML with secondary kinase domain mutations. More recently, point mutations conferring resistance to the Bruton tyrosine kinase inhibitor ibrutinib in chronic lymphocytic leukemia, arsenic trioxide in acute promyelocytic leukemia, and the BH3-mimetic ABT199 in lymphoma have been identified. In chronic myeloid leukemia, the emergence of tyrosine kinase domain mutations has historically been the dominant mechanism of resistance. The early identification of secondary point mutations and their downstream effects along with the development of second- or third-generation inhibitors and rationally designed small molecule combinations are potential strategies to overcome mutation-mediated resistance.
Collapse
|
54
|
Andersson AK, Ma J, Wang J, Chen X, Gedman AL, Dang J, Nakitandwe J, Holmfeldt L, Parker M, Easton J, Huether R, Kriwacki R, Rusch M, Wu G, Li Y, Mulder H, Raimondi S, Pounds S, Kang G, Shi L, Becksfort J, Gupta P, Payne-Turner D, Vadodaria B, Boggs K, Yergeau D, Manne J, Song G, Edmonson M, Nagahawatte P, Wei L, Cheng C, Pei D, Sutton R, Venn NC, Chetcuti A, Rush A, Catchpoole D, Heldrup J, Fioretos T, Lu C, Ding L, Pui CH, Shurtleff S, Mullighan CG, Mardis ER, Wilson RK, Gruber TA, Zhang J, Downing JR. The landscape of somatic mutations in infant MLL-rearranged acute lymphoblastic leukemias. Nat Genet 2015; 47:330-7. [PMID: 25730765 PMCID: PMC4553269 DOI: 10.1038/ng.3230] [Citation(s) in RCA: 377] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 02/02/2015] [Indexed: 12/13/2022]
Abstract
Infant acute lymphoblastic leukemia (ALL) with MLL rearrangements (MLL-R) represents a distinct leukemia with a poor prognosis. To define its mutational landscape, we performed whole genome, exome, RNA and targeted DNA sequencing on 65 infants (47 MLL-R and 18 non-MLL-R) and 20 older children (MLL-R cases) with leukemia. Our data demonstrated infant MLL-R ALL to have one of the lowest frequencies of somatic mutations of any sequenced cancer, with the predominant leukemic clone carrying a mean of 1.3 non-silent mutations. Despite the paucity of mutations, activating mutations in kinase/PI3K/RAS signaling pathways were detected in 47%. Surprisingly, however, these mutations were often sub-clonal and frequently lost at relapse. In contrast to infant cases, MLL-R leukemia in older children had more somatic mutations (a mean of 6.5/case versus 1.3/case, P=7.15×10−5) and contained frequent mutations (45%) in epigenetic regulators, a category of genes that with the exception of MLL was rarely mutated in infant MLL-R ALL.
Collapse
Affiliation(s)
- Anna K Andersson
- 1] Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA. [2] Department of Clinical Genetics, Lund University, Lund, Sweden
| | - Jing Ma
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jianmin Wang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Amanda Larson Gedman
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jinjun Dang
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Joy Nakitandwe
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Linda Holmfeldt
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Matthew Parker
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - John Easton
- Pediatric Cancer Genome Project Laboratory, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Robert Huether
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Richard Kriwacki
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Michael Rusch
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Gang Wu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Yongjin Li
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Heather Mulder
- Pediatric Cancer Genome Project Laboratory, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Susana Raimondi
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stanley Pounds
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Guolian Kang
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Lei Shi
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jared Becksfort
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Pankaj Gupta
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Debbie Payne-Turner
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Bhavin Vadodaria
- Pediatric Cancer Genome Project Laboratory, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Kristy Boggs
- Pediatric Cancer Genome Project Laboratory, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Donald Yergeau
- Pediatric Cancer Genome Project Laboratory, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jayanthi Manne
- Pediatric Cancer Genome Project Laboratory, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Guangchun Song
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Michael Edmonson
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Panduka Nagahawatte
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Lei Wei
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Deqing Pei
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Rosemary Sutton
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Nicola C Venn
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Albert Chetcuti
- Tumor Bank, Children's Cancer Research Unit, Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Amanda Rush
- Tumor Bank, Children's Cancer Research Unit, Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Daniel Catchpoole
- Tumor Bank, Children's Cancer Research Unit, Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Jesper Heldrup
- Department of Pediatrics, Skåne University Hospital, Lund, Sweden
| | - Thoas Fioretos
- Department of Clinical Genetics, Lund University, Lund, Sweden
| | - Charles Lu
- 1] Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA. [2] Genome Institute, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Li Ding
- 1] Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA. [2] Genome Institute, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Ching-Hon Pui
- 1] Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA. [2] Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Sheila Shurtleff
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Elaine R Mardis
- 1] Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA. [2] Genome Institute, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Richard K Wilson
- 1] Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA. [2] Genome Institute, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Tanja A Gruber
- 1] Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA. [2] Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - James R Downing
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | |
Collapse
|
55
|
Moreira RB, Peixoto RD, de Sousa Cruz MR. Clinical Response to Sorafenib in a Patient with Metastatic Colorectal Cancer and FLT3 Amplification. Case Rep Oncol 2015; 8:83-7. [PMID: 25848357 PMCID: PMC4361904 DOI: 10.1159/000375483] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background A considerable number of patients with metastatic colorectal cancer progress after exhausting all approved standard therapies but maintain an adequate performance status and could be candidates for further treatment. We aim at reviewing our experience with sorafenib treatment of a patient with FLT3 mutation in refractory metastatic colorectal cancer. Methods Treatment with sorafenib of a patient with metastatic colorectal cancer and FLT3 translocation who had previously been heavily treated. Results The patient with metastatic colorectal cancer, aged 51 years, showed significant symptomatic and laboratory improvement with sorafenib treatment (400 mg twice daily). Conclusion The presented case illustrates how an aggressive and refractory colorectal tumor may respond well to targeted therapy.
Collapse
|
56
|
Li C, Liu L, Liang L, Xia Z, Li Z, Wang X, McGee LR, Newhall K, Sinclair A, Kamb A, Wickramasinghe D, Dai K. AMG 925 is a dual FLT3/CDK4 inhibitor with the potential to overcome FLT3 inhibitor resistance in acute myeloid leukemia. Mol Cancer Ther 2014; 14:375-83. [PMID: 25487917 DOI: 10.1158/1535-7163.mct-14-0388] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Resistance to FLT3 inhibitors is a serious clinical issue in treating acute myelogenous leukemia (AML). AMG 925, a dual FLT3/CDK4 inhibitor, has been developed to overcome this resistance. It is hypothesized that the combined inhibition of FLT3 and CDK4 may reduce occurrence of the FLT3 resistance mutations, and thereby prolong clinical responses. To test this hypothesis, we attempted to isolate AML cell clones resistant to AMG 925 or to FLT3 inhibitors. After a selection of over 8 months with AMG 925, we could only isolate partially resistant clones. No new mutations in FLT3 were found, but a 2- to 3-fold increase in total FLT3 protein was detected and believed to contribute to the partial resistance. In contrast, selection with the FLT3 inhibitors sorafenib or AC220 (Quizartinib), led to a resistance and the appearance of a number of mutations in FLT3 kinase domains, including the known hot spot sites D835 and F691. However, when AC220 was combined with the CDK4 inhibitor PD0332991 (palbociclib) at 0.1 μmol/L or higher, no resistance mutations were obtained, indicating that the CDK4-inhibiting activity of AMG 925 contributed to the failure to develop drug resistance. AMG 925 was shown to potently inhibit the FLT3 inhibitor-resistant mutation D835Y/V. This feature of AMG 925 was also considered to contribute to the lack of resistance mutations to the compound. Together, our data suggest that AMG 925 has the potential to reduce resistance mutations in FLT3 and may prolong clinical responses.
Collapse
Affiliation(s)
- Cong Li
- Amgen Discovery Research, Amgen Inc., South San Francisco, California.
| | - Liqin Liu
- Amgen Discovery Research, Amgen Inc., South San Francisco, California
| | - Lingming Liang
- Amgen Discovery Research, Amgen Inc., South San Francisco, California
| | - Zhen Xia
- Amgen Discovery Research, Amgen Inc., South San Francisco, California
| | - Zhihong Li
- Amgen Discovery Research, Amgen Inc., South San Francisco, California
| | - Xianghong Wang
- Amgen Discovery Research, Amgen Inc., South San Francisco, California
| | - Lawrence R McGee
- Amgen Discovery Research, Amgen Inc., South San Francisco, California
| | - Katie Newhall
- Amgen Discovery Research, Amgen Inc., South San Francisco, California
| | - Angus Sinclair
- Amgen Discovery Research, Amgen Inc., South San Francisco, California
| | - Alexander Kamb
- Amgen Discovery Research, Amgen Inc., South San Francisco, California
| | | | - Kang Dai
- Amgen Discovery Research, Amgen Inc., South San Francisco, California
| |
Collapse
|
57
|
Abstract
PURPOSE OF REVIEW 'FMS'-like tyrosine kinase 3 (FLT3) mutations in acute myeloid leukemia (AML) have been brought from discovery in the early 1990s to clinical targeting in the past 10 years. Despite several promising leads in preclinical models, no agent has yet been approved for clinical use. Here we will review the development of novel therapies for AML with FLT3 mutations. RECENT FINDINGS Initial clinical development focused on broad kinase inhibitors which were found to have limited clinical activity due to insufficient kinase inhibitory activity and high toxicity. Subsequent development has brought forth narrow-spectrum inhibitors with potent in-vivo activity and reasonable clinical tolerance, but many patients still progress with prolonged use. SUMMARY The optimal role for targeting FLT3 may depend on multimodality therapy and will likely require hematopoietic transplant. The incorporation of ABL kinase inhibitors into acute lymphoblastic leukemia management should serve as a model for incorporation of FLT3-targeted agents into clinical care. Strategies incorporating FLT3-targeted agents into AML therapy are ongoing, but challenges in trial design, clinical heterogeneity and need for long-term follow-up make these investigations complicated in design and implementation.
Collapse
|
58
|
Annesley CE, Brown P. The Biology and Targeting of FLT3 in Pediatric Leukemia. Front Oncol 2014; 4:263. [PMID: 25295230 PMCID: PMC4172015 DOI: 10.3389/fonc.2014.00263] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 09/08/2014] [Indexed: 12/22/2022] Open
Abstract
Despite remarkable improvement in treatment outcomes in pediatric leukemia over the past several decades, the prognosis for high-risk groups of acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL), as well as for relapsed leukemia, remains poor. Intensification of chemotherapy regimens for those at highest risk has improved success rates, but at the cost of significantly increased morbidity and long-term adverse effects. With the success of imatinib in Philadelphia-chromosome-positive leukemia and all-trans retinoic acid in acute promyelocytic leukemia, the quest to find additional molecularly targeted therapies has generated much excitement over recent years. Another such possible target in pediatric acute leukemia is FMS-like tyrosine kinase 3 (FLT3). FLT3 aberrations are among the most frequently identified transforming events in AML, and have significant clinical implications in both high-risk pediatric AML and in certain high-risk groups of pediatric ALL. Therefore, the successful targeting of FLT3 has tremendous potential to improve outcomes in these subsets of patients. This article will give an overview of the molecular function and signaling of the FLT3 receptor, as well as its pathogenic role in leukemia. We review the discovery of targeting FLT3, discuss currently available FLT3 inhibitors in pediatric leukemia and results of clinical trials to date, and finally, consider the future promise and challenges of FLT3 inhibitor therapy.
Collapse
Affiliation(s)
- Colleen E. Annesley
- Oncology and Pediatrics, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patrick Brown
- Oncology and Pediatrics, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
59
|
Wander SA, Levis MJ, Fathi AT. The evolving role of FLT3 inhibitors in acute myeloid leukemia: quizartinib and beyond. Ther Adv Hematol 2014; 5:65-77. [PMID: 24883179 DOI: 10.1177/2040620714532123] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Acute myeloid leukemia remains associated with poor outcomes despite advances in our understanding of the complicated molecular events driving leukemogenesis and malignant progression. Those patients harboring mutations in the FLT3 receptor tyrosine kinase have a particularly poor prognosis; however, significant excitement has been generated by the emergence of a variety of targeted inhibitors capable of suppressing FLT3 signaling in vivo. Here we will review results from preclinical studies and early clinical trials evaluating both first- and second-generation FLT3 inhibitors. Early FLT3 inhibitors (including sunitinib, midostaurin, and lestaurtinib) demonstrated significant promise in preclinical models of FLT3 mutant AML. Unfortunately, many of these compounds failed to achieve robust and sustained FLT3 inhibition in early clinical trials, at best resulting in only transient decreases in peripheral blast counts. These results have prompted the development of second-generation FLT3 inhibitors, epitomized by the novel agent quizartinib. These second-generation inhibitors have demonstrated enhanced FLT3 specificity and have been generally well tolerated in early clinical trials. Several FLT3 inhibitors have reached phase III clinical trials, and a variety of phase I/II trials exploring a role for these novel compounds in conjunction with conventional chemotherapy or hematopoietic stem cell transplantation are ongoing. Finally, molecular insights provided by FLT3 inhibitors have shed light upon the variety of mechanisms underlying the acquisition of resistance and have provided a rationale supporting the use of combinatorial regimens with other emerging targeted therapies.
Collapse
Affiliation(s)
- Seth A Wander
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Mark J Levis
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Amir T Fathi
- Massachusetts General Hospital, Harvard Medical School, Zero Emerson Place, Suite 118, Boston, MA 02114, USA
| |
Collapse
|
60
|
Yu C, Kancha RK, Duyster J. Targeting oncoprotein stability overcomes drug resistance caused by FLT3 kinase domain mutations. PLoS One 2014; 9:e97116. [PMID: 24849514 PMCID: PMC4029991 DOI: 10.1371/journal.pone.0097116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 04/14/2014] [Indexed: 12/22/2022] Open
Abstract
FLT3 is the most frequently mutated kinase in acute myeloid leukemia (AML). Internal tandem duplications (ITDs) in the juxta-membrane region constitute the majority of activating FLT3 mutations. Several FLT3 kinase inhibitors were developed and tested in the clinic with significant success. However, recent studies have reported the development of secondary drug resistance in patients treated with FLT3 inhibitors. Since FLT3-ITD is an HSP90 client kinase, we here explored if targeting the stability of drug-resistant FLT3 mutant protein could be a potential therapeutic option. We observed that HSP90 inhibitor treatment resulted in the degradation of inhibitor-resistant FLT3-ITD mutants and selectively induced toxicity in cells expressing FLT3-ITD mutants. Thus, HSP90 inhibitors provide a potential therapeutic choice to overcome secondary drug resistance following TKI treatment in FLT3-ITD positive AML.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Benzoquinones/pharmacology
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Leukemic
- HSP90 Heat-Shock Proteins/antagonists & inhibitors
- HSP90 Heat-Shock Proteins/genetics
- HSP90 Heat-Shock Proteins/metabolism
- Humans
- Lactams, Macrocyclic/pharmacology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Mice
- Molecular Sequence Data
- Mutation
- Protein Binding
- Protein Kinase Inhibitors/pharmacology
- Protein Stability/drug effects
- Protein Structure, Tertiary
- Proteolysis
- Sequence Alignment
- Signal Transduction
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
Collapse
Affiliation(s)
- Chuanjiang Yu
- Department Medicine I, University Medical Center Freiburg, Freiburg, Germany
| | - Rama Krishna Kancha
- Department Medicine I, University Medical Center Freiburg, Freiburg, Germany
| | - Justus Duyster
- Department Medicine I, University Medical Center Freiburg, Freiburg, Germany
- * E-mail:
| |
Collapse
|
61
|
Alvarado Y, Kantarjian HM, Luthra R, Ravandi F, Borthakur G, Garcia-Manero G, Konopleva M, Estrov Z, Andreeff M, Cortes JE. Treatment with FLT3 inhibitor in patients with FLT3-mutated acute myeloid leukemia is associated with development of secondary FLT3-tyrosine kinase domain mutations. Cancer 2014; 120:2142-9. [PMID: 24737502 DOI: 10.1002/cncr.28705] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/23/2013] [Accepted: 01/08/2014] [Indexed: 11/08/2022]
Abstract
BACKGROUND FLT3-internal tandem duplication (ITD) mutations are found in approximately 30% of patients with acute myeloid leukemia (AML). FLT3 inhibitors have shown clinical activity in AML with FLT3-ITD, but responses are usually short-lived. METHODS This study examined 69 FLT3-mutated patients with AML, who were treated with different FLT3 inhibitors to analyze emergence of new mutations. RESULTS At baseline, 87% of patients had an ITD mutation, 7% had a D835/I836 mutation, and 6% had combined ITD and D835/I836 mutations. Responses occurred in 32% of patients, all with FLT3-ITD; none of the patients with D835/I836 or ITD+D835/I836 responded. Mutational assessment at the time of FLT3 inhibitor discontinuation showed that 68% of patients were unchanged, 10% had become undetectable, and 22% of patients progressed from a single ITD to have combined ITD+D835/I836 mutations. In those patients with unchanged FLT3 mutation at progression, the median survival was 5 months, whereas in those with undetectable and with combined ITD+D835/I836 mutations, the median survival was 7 months, respectively. CONCLUSIONS These data confirm in vitro observations that a secondary tyrosine kinase domain mutation may arise after the use of FLT3 inhibitors in patients with single FLT3-ITD mutated AML, a phenomenon that is associated with resistance and a poor prognosis.
Collapse
Affiliation(s)
- Yesid Alvarado
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Zhang W, Gao C, Konopleva M, Chen Y, Jacamo RO, Borthakur G, Cortes JE, Ravandi F, Ramachandran A, Andreeff M. Reversal of acquired drug resistance in FLT3-mutated acute myeloid leukemia cells via distinct drug combination strategies. Clin Cancer Res 2014; 20:2363-74. [PMID: 24619500 DOI: 10.1158/1078-0432.ccr-13-2052] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE FMS-like tyrosine kinase-3 (FLT3) internal tandem duplication (FLT3-ITD) mutations are common in patients with acute myeloid leukemia (AML). These patients regularly develop resistance to FLT3 inhibitors suggesting that targeted combination drug strategies are needed to enhance AML therapy efficacy. EXPERIMENTAL DESIGN Acquired point mutations of FLT3-ITD gene were screened using cDNA-based sequencing approach in vitro sorafenib-resistant cells, which were developed by long-term exposure of Ba/F3-ITD to increasing doses of sorafenib, and in FLT3-ITD mutated AML patients, who developed relapse following sorafenib therapy. Drug effects (e.g., proliferation inhibition, apoptosis induction, and changes in signal transduction protein expression) were assessed in AML cells harboring the point mutations in vitro and in FLT3-ITD-mutated AML patient samples. RESULTS We identified several acquired point mutations in the tyrosine kinase domains (TKD) of the FLT3 gene in sorafenib-resistant murine leukemia cell line carrying human FLT3-ITD mutations, which were also detected in two of four sorafenib-resistant patient samples. Engineering these point mutations into Ba/F3-ITD cells generated sublines that demonstrated varying degrees of sorafenib [a type II tyrosine kinase inhibitor (TKI)] resistance. A similar pattern of resistance could be observed by exposing these sublines to the other type II TKIs AC220 and MLN518. However, these sublines retained sensitivity to the type I TKIs PKC412 or crenolanib. The combination of crenolanib with sorafenib demonstrated marked cytotoxic effects in all of the sorafenib-resistant sublines. CONCLUSIONS These combination strategies could be clinically important in reversing acquired resistance to FLT3 inhibition in AML.
Collapse
Affiliation(s)
- Weiguo Zhang
- Authors' Affiliations: Section of Molecular Hematology and Therapy, Department of Leukemia, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston; and AROG Pharmaceuticals LLC, Dallas, Texas
| | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
The N550K/H mutations in FGFR2 confer differential resistance to PD173074, dovitinib, and ponatinib ATP-competitive inhibitors. Neoplasia 2014; 15:975-88. [PMID: 23908597 DOI: 10.1593/neo.121106] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 04/30/2013] [Accepted: 05/03/2013] [Indexed: 01/26/2023] Open
Abstract
We sought to identify fibroblast growth factor receptor 2 (FGFR2) kinase domain mutations that confer resistance to the pan-FGFR inhibitor, dovitinib, and explore the mechanism of action of the drug-resistant mutations. We cultured BaF3 cells overexpressing FGFR2 in high concentrations of dovitinib and identified 14 dovitinib-resistant mutations, including the N550K mutation observed in 25% of FGFR2(mutant) endometrial cancers (ECs). Structural and biochemical in vitro kinase analyses, together with BaF3 proliferation assays, showed that the resistance mutations elevate the intrinsic kinase activity of FGFR2. BaF3 lines were used to assess the ability of each mutation to confer cross-resistance to PD173074 and ponatinib. Unlike PD173074, ponatinib effectively inhibited all the dovitinib-resistant FGFR2 mutants except the V565I gatekeeper mutation, suggesting ponatinib but not dovitinib targets the active conformation of FGFR2 kinase. EC cell lines expressing wild-type FGFR2 were relatively resistant to all inhibitors, whereas EC cell lines expressing mutated FGFR2 showed differential sensitivity. Within the FGFR2(mutant) cell lines, three of seven showed marked resistance to PD173074 and relative resistance to dovitinib and ponatinib. This suggests that alternative mechanisms distinct from kinase domain mutations are responsible for intrinsic resistance in these three EC lines. Finally, overexpression of FGFR2(N550K) in JHUEM-2 cells (FGFR2(C383R)) conferred resistance (about five-fold) to PD173074, providing independent data that FGFR2(N550K) can be associated with drug resistance. Biochemical in vitro kinase analyses also show that ponatinib is more effective than dovitinib at inhibiting FGFR2(N550K). We propose that tumors harboring mutationally activated FGFRs should be treated with FGFR inhibitors that specifically bind the active kinase.
Collapse
|
64
|
Keegan K, Li C, Li Z, Ma J, Ragains M, Coberly S, Hollenback D, Eksterowicz J, Liang L, Weidner M, Huard J, Wang X, Alba G, Orf J, Lo MC, Zhao S, Ngo R, Chen A, Liu L, Carlson T, Quéva C, McGee LR, Medina J, Kamb A, Wickramasinghe D, Dai K. Preclinical evaluation of AMG 925, a FLT3/CDK4 dual kinase inhibitor for treating acute myeloid leukemia. Mol Cancer Ther 2014; 13:880-9. [PMID: 24526162 DOI: 10.1158/1535-7163.mct-13-0858] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) remains a serious unmet medical need. Despite high remission rates with chemotherapy standard-of-care treatment, the disease eventually relapses in a major proportion of patients. Activating Fms-like tyrosine kinase 3 (FLT3) mutations are found in approximately 30% of patients with AML. Targeting FLT3 receptor tyrosine kinase has shown encouraging results in treating FLT3-mutated AML. Responses, however, are not sustained and acquired resistance has been a clinical challenge. Treatment options to overcome resistance are currently the focus of research. We report here the preclinical evaluation of AMG 925, a potent, selective, and bioavailable FLT3/cyclin-dependent kinase 4 (CDK4) dual kinase inhibitor. AMG 925 inhibited AML xenograft tumor growth by 96% to 99% without significant body weight loss. The antitumor activity of AMG 925 correlated with the inhibition of STAT5 and RB phosphorylation, the pharmacodynamic markers for inhibition of FLT3 and CDK4, respectively. In addition, AMG 925 was also found to inhibit FLT3 mutants (e.g., D835Y) that are resistant to the current FLT3 inhibitors (e.g., AC220 and sorafenib). CDK4 is a cyclin D-dependent kinase that plays an essential central role in regulating cell proliferation in response to external growth signals. A critical role of the CDK4-RB pathway in cancer development has been well established. CDK4-specific inhibitors are being developed for treating RB-positive cancer. AMG 925, which combines inhibition of two kinases essential for proliferation and survival of FLT3-mutated AML cells, may improve and prolong clinical responses.
Collapse
Affiliation(s)
- Kathleen Keegan
- Authors' Affiliation: Amgen Discovery Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Abstract
In this issue of Blood, Zimmerman and colleagues demonstrate that the tyrosine kinase inhibitor (TKI) crenolanib effectively suppresses growth of leukemic cells harboring both FLT3-ITD and FLT3-TKD mutations, the latter of which are increasingly seen to emerge as resistant mutations after FMS-like tyrosine kinase 3 (FLT3) inhibitor therapy.
Collapse
|
66
|
Serve H, Krug U, Wagner R, Sauerland MC, Heinecke A, Brunnberg U, Schaich M, Ottmann O, Duyster J, Wandt H, Fischer T, Giagounidis A, Neubauer A, Reichle A, Aulitzky W, Noppeney R, Blau I, Kunzmann V, Stuhlmann R, Krämer A, Kreuzer KA, Brandts C, Steffen B, Thiede C, Müller-Tidow C, Ehninger G, Berdel WE. Sorafenib in Combination With Intensive Chemotherapy in Elderly Patients With Acute Myeloid Leukemia: Results From a Randomized, Placebo-Controlled Trial. J Clin Oncol 2013; 31:3110-8. [DOI: 10.1200/jco.2012.46.4990] [Citation(s) in RCA: 258] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Purpose The prognosis of elderly patients with acute myeloid leukemia (AML) is still dismal even with intensive chemotherapy. In this trial, we compared the antileukemic activity of standard induction and consolidation therapy with or without the addition of the kinase inhibitor sorafenib in elderly patients with AML. Patients and Methods All patients received standard cytarabine and daunorubicin induction (7+3 regimen) and up to two cycles of intermediate-dose cytarabine consolidation. Two hundred one patients were equally randomly assigned to receive either sorafenib or placebo between the chemotherapy cycles and subsequently for up to 1 year after the beginning of therapy. The primary objective was to test for an improvement in event-free survival (EFS). Overall survival (OS), complete remission (CR) rate, tolerability, and several predefined subgroup analyses were among the secondary objectives. Results Age, sex, CR and early death (ED) probability, and prognostic factors were balanced between both study arms. Treatment in the sorafenib arm did not result in significant improvement in EFS or OS. This was also true for subgroup analyses, including the subgroup positive for FLT3 internal tandem duplications. Results of induction therapy were worse in the sorafenib arm, with higher treatment-related mortality and lower CR rates. More adverse effects occurred during induction therapy in the sorafenib arm, and patients in this arm received less consolidation chemotherapy as a result of higher induction toxicity. Conclusion In conclusion, combination of standard induction and consolidation therapy with sorafenib in the schedule investigated in our trial is not beneficial for elderly patients with AML.
Collapse
Affiliation(s)
- Hubert Serve
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Utz Krug
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Ruth Wagner
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - M. Cristina Sauerland
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Achim Heinecke
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Uta Brunnberg
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Markus Schaich
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Oliver Ottmann
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Justus Duyster
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Hannes Wandt
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Thomas Fischer
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Aristoteles Giagounidis
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Andreas Neubauer
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Albrecht Reichle
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Walter Aulitzky
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Richard Noppeney
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Igor Blau
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Volker Kunzmann
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Reingard Stuhlmann
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Alwin Krämer
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Karl-Anton Kreuzer
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Christian Brandts
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Björn Steffen
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Christian Thiede
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Carsten Müller-Tidow
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Gerhard Ehninger
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| | - Wolfgang E. Berdel
- Hubert Serve, Uta Brunnberg, Oliver Ottmann, Christian Brandts, Björn Steffen, Goethe-University, Frankfurt; Utz Krug, Ruth Wagner, Carsten Müller-Tidow, and Wolfgang E. Berdel, University Hospital; Ruth Wagner, M. Cristina Sauerland, Achim Heinecke, University of Muenster, Muenster; Markus Schaich, Christian Thiede, and Gerhard Ehninger, University Hospital, Dresden; Justus Duyster, Technische Universität München, München; Hannes Wandt, Klinikum Nürnberg, Nürnberg; Thomas Fischer, University Hospital,
| |
Collapse
|
67
|
Baker SD, Zimmerman EI, Wang YD, Orwick S, Zatechka DS, Buaboonnam J, Neale GA, Olsen SR, Enemark EJ, Shurtleff S, Rubnitz JE, Mullighan CG, Inaba H. Emergence of polyclonal FLT3 tyrosine kinase domain mutations during sequential therapy with sorafenib and sunitinib in FLT3-ITD-positive acute myeloid leukemia. Clin Cancer Res 2013; 19:5758-68. [PMID: 23969938 DOI: 10.1158/1078-0432.ccr-13-1323] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE To evaluate the clinical activity of sequential therapy with sorafenib and sunitinib in FMS-like tyrosine kinase 3 (FLT3)-internal tandem duplication (ITD)-positive acute myelogenous leukemia (AML) and monitor the emergence of secondary FLT3 tyrosine kinase domain (TKD) mutations during treatment. EXPERIMENTAL DESIGN Six children with relapsed/refractory AML were treated with sorafenib in combination with clofarabine and cytarabine, followed by single-agent sorafenib if not a candidate for transplantation. Sunitinib was initiated after sorafenib relapse. Bone marrow samples were obtained for assessment of FLT3 TKD mutations by deep amplicon sequencing. The phase of secondary mutations with ITD alleles was assessed by cloning and sequencing of FLT3 exons 14 through 20. Identified mutations were modeled in Ba/F3 cells, and the effect of kinase inhibitors on FLT3 signaling and cell viability was assessed. RESULTS Four patients achieved complete remission, but 3 receiving maintenance therapy with sorafenib relapsed after 14 to 37 weeks. Sunitinib reduced circulating blasts in two patients and marrow blasts in one. Two patients did not respond to sorafenib combination therapy or sunitinib. FLT3 mutations at residues D835 and F691 were observed in sorafenib resistance samples on both ITD-positive and -negative alleles. Deep sequencing revealed low-level mutations and their evolution during sorafenib treatment. Sunitinib suppressed leukemic clones with D835H and F691L mutations, but not D835Y. Cells expressing sorafenib-resistant FLT3 mutations were sensitive to sunitinib in vitro. CONCLUSIONS Sunitinib has activity in patients that are resistant to sorafenib and harbor secondary FLT3 TKD mutations. The use of sensitive methods to monitor FLT3 mutations during therapy may allow individualized treatment with the currently available kinase inhibitors.
Collapse
Affiliation(s)
- Sharyn D Baker
- Authors' Affiliations: Departments of Pharmaceutical Sciences, Structural Biology, Pathology, and Oncology, Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children's Research Hospital; and Department of Pediatrics, University of Tennessee, Memphis, Tennessee
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Kayser S, Levis MJ. FLT3 tyrosine kinase inhibitors in acute myeloid leukemia: clinical implications and limitations. Leuk Lymphoma 2013; 55:243-55. [PMID: 23631653 DOI: 10.3109/10428194.2013.800198] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Internal tandem duplications of the FMS-like tyrosine kinase 3 (FLT3) gene are one of the most frequent gene mutations in acute myeloid leukemia (AML) and are associated with poor clinical outcome. The remission rate is high with intensive chemotherapy, but most patients eventually relapse. During the last decade, FLT3 mutations have emerged as an attractive target for a molecularly specific treatment strategy. Targeting FLT3 receptor tyrosine kinases in AML has shown encouraging results in the treatment of FLT3 mutated AML, but in most patients responses are incomplete and not sustained. Newer, more specific compounds seem to have a higher potency and selectivity against FLT3. During therapy with FLT3 tyrosine kinase inhibitors (TKIs) the induction of acquired resistance has emerged as a clinical problem. Therefore, optimization of the targeted therapy and potential treatment options to overcome resistance is currently the focus of clinical research. In this review we discuss the use and limitations of TKIs as a therapeutic strategy for the treatment of FLT3 mutated AML, including mechanisms of resistance to TKIs as well as possible novel strategies to improve FLT3 inhibitor therapy.
Collapse
Affiliation(s)
- Sabine Kayser
- Department of Internal Medicine III, University Hospital of Ulm , Germany
| | | |
Collapse
|
69
|
Grunwald MR, Levis MJ. FLT3 inhibitors for acute myeloid leukemia: a review of their efficacy and mechanisms of resistance. Int J Hematol 2013; 97:683-94. [PMID: 23613268 DOI: 10.1007/s12185-013-1334-8] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 04/08/2013] [Accepted: 04/10/2013] [Indexed: 12/11/2022]
Abstract
Since the Food and Drug Administration approval of imatinib for treatment of chronic myeloid leukemia in 2001, tyrosine kinase inhibitors (TKIs) have become a mainstay in the care of many malignancies. In acute myeloid leukemia (AML), activating mutations in the FMS-like tyrosine kinase 3 (FLT3) gene result in survival and proliferation of leukemic blasts and are associated with adverse prognosis. Therefore, the FLT3 receptor is an appealing target for inhibition. Multiple small molecule TKIs are currently in development for FLT3-mutated AML, and agents are beginning to show promising efficacy. In other malignancies, the development of resistance to TKIs during the course of therapy has proven to be a challenge, and thus far, in clinical trials of FLT3 TKIs, resistance to inhibition represents a significant barrier to successful FLT3 inhibition. Understanding the mechanisms of resistance and overcoming these obstacles to target inhibition will be central to the success of these agents.
Collapse
Affiliation(s)
- Michael R Grunwald
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, 1650 Orleans Street, Baltimore, MD 21287, USA.
| | | |
Collapse
|
70
|
Activity of ponatinib against clinically-relevant AC220-resistant kinase domain mutants of FLT3-ITD. Blood 2013; 121:3165-71. [PMID: 23430109 DOI: 10.1182/blood-2012-07-442871] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Secondary point mutations in the Fms-like tyrosine kinase 3 (FLT3) tyrosine kinase domain (KD) are common causes of acquired clinical resistance to the FLT3 inhibitors AC220 (quizartinib) and sorafenib. Ponatinib (AP24534) is a multikinase inhibitor with in vitro and clinical activity in tyrosine kinase inhibitor (TKI)-resistant chronic myeloid leukemia, irrespective of BCR-ABL KD mutation. Ponatinib has demonstrated early clinical efficacy in chemotherapy-resistant acute myeloid leukemia (AML) patients with internal tandem duplication (ITD) mutations in FLT3. We assessed the in vitro activity of ponatinib against clinically relevant FLT3-ITD mutant isoforms that confer resistance to AC220 or sorafenib. Substitution of the FLT3 "gatekeeper" phenylalanine with leucine (F691L) conferred mild resistance to ponatinib, but substitutions at the FLT3 activation loop (AL) residue D835 conferred a high degree of resistance. Saturation mutagenesis of FLT3-ITD exclusively identified FLT3 AL mutations at positions D835, D839, and Y842. The switch control inhibitor DCC-2036 was similarly inactive against FLT3 AL mutations. On the basis of its in vitro activity against FLT3 TKI-resistant F691 substitutions, further clinical evaluation of ponatinib in TKI-naïve and select TKI-resistant FLT3-ITD+ AML patients is warranted. Alternative strategies will be required for patients with TKI-resistant FLT3-ITD D835 mutations.
Collapse
|
71
|
Albers C, Leischner H, Verbeek M, Yu C, Illert AL, Peschel C, von Bubnoff N, Duyster J. The secondary FLT3-ITD F691L mutation induces resistance to AC220 in FLT3-ITD+ AML but retains in vitro sensitivity to PKC412 and Sunitinib. Leukemia 2013; 27:1416-8. [PMID: 23392356 DOI: 10.1038/leu.2013.14] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
72
|
Williams AB, Nguyen B, Li L, Brown P, Levis M, Leahy D, Small D. Mutations of FLT3/ITD confer resistance to multiple tyrosine kinase inhibitors. Leukemia 2012; 27:48-55. [PMID: 22858906 DOI: 10.1038/leu.2012.191] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
FMS-like tyrosine kinase 3 (FLT3) normally functions in the survival/proliferation of hematopoietic stem/progenitor cells, but its constitutive activation by internal tandem duplication (ITD) mutations correlates with a poor prognosis in AML. The development of FLT3 tyrosine kinase inhibitors (TKI) is a promising strategy, but resistance that arises during the course of treatment caused by secondary mutations within the mutated gene itself poses a significant challenge. In an effort to predict FLT3 resistance mutations that might develop in patients, we used saturation mutagenesis of FLT3/ITD followed by selection of transfected cells in FLT3 TKI. We identified F621L, A627P, F691L and Y842C mutations in FLT3/ITD that confer varying levels of resistance to FLT3 TKI. Western blotting confirmed that some FLT3 TKI were ineffective at inhibiting FLT3 autophosphorylation and signaling through MAP kinase, STAT5 and AKT in some mutants. Balb/c mice transplanted with the FLT3/ITD Y842C mutation confirmed resistance to sorafenib in vivo but not to lestaurtinib. These results indicate a growing number of FLT3 mutations that are likely to be encountered in patients. Such knowledge, combined with known remaining sensitivity to other FLT3 TKI, will be important to establish as secondary drug treatments that can be substituted when these mutants are encountered.
Collapse
Affiliation(s)
- A B Williams
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | | | | | |
Collapse
|
73
|
Zirm E, Spies-Weisshart B, Heidel F, Schnetzke U, Böhmer FD, Hochhaus A, Fischer T, Scholl S. Ponatinib may overcome resistance of FLT3-ITD harbouring additional point mutations, notably the previously refractory F691I mutation. Br J Haematol 2012; 157:483-92. [DOI: 10.1111/j.1365-2141.2012.09085.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 02/03/2012] [Indexed: 12/16/2022]
Affiliation(s)
- Elisabeth Zirm
- Department of Haematology/Oncology; Clinic for Internal Medicine II; Jena University Hospital; Jena
| | - Bärbel Spies-Weisshart
- Department of Haematology/Oncology; Clinic for Internal Medicine II; Jena University Hospital; Jena
| | - Florian Heidel
- Department of Haematology/Oncology; Otto-von-Guericke-University; Magdeburg; Germany
| | - Ulf Schnetzke
- Department of Haematology/Oncology; Clinic for Internal Medicine II; Jena University Hospital; Jena
| | | | - Andreas Hochhaus
- Department of Haematology/Oncology; Clinic for Internal Medicine II; Jena University Hospital; Jena
| | - Thomas Fischer
- Department of Haematology/Oncology; Otto-von-Guericke-University; Magdeburg; Germany
| | - Sebastian Scholl
- Department of Haematology/Oncology; Clinic for Internal Medicine II; Jena University Hospital; Jena
| |
Collapse
|
74
|
Moore AS, Faisal A, Gonzalez de Castro D, Bavetsias V, Sun C, Atrash B, Valenti M, de Haven Brandon A, Avery S, Mair D, Mirabella F, Swansbury J, Pearson ADJ, Workman P, Blagg J, Raynaud FI, Eccles SA, Linardopoulos S. Selective FLT3 inhibition of FLT3-ITD+ acute myeloid leukaemia resulting in secondary D835Y mutation: a model for emerging clinical resistance patterns. Leukemia 2012; 26:1462-70. [PMID: 22354205 PMCID: PMC3523391 DOI: 10.1038/leu.2012.52] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Acquired resistance to selective FLT3 inhibitors, is an emerging clinical problem in the treatment of FLT3-ITD+ acute myeloid leukaemia (AML). The paucity of valid pre-clinical models has limited investigations to determine the mechanism of acquired therapeutic resistance, thereby limiting the development of effective treatments. We generated selective FLT3 inhibitor-resistant cells by treating the FLT3-ITD+ human AML cell line MOLM-13 in vitro with the FLT3-selective inhibitor MLN518, and validated the resistant phenotype in vivo and in vitro. The resistant cells, MOLM-13-RES, harboured a new D835Y tyrosine kinase domain (TKD) mutation on the FLT3-ITD+ allele. Acquired TKD mutations, including D835Y, have recently been identified in FLT3-ITD+ patients relapsing after treatment with the novel FLT3 inhibitor, AC220. Consistent with this clinical pattern of resistance, MOLM-13- RES cells displayed high relative resistance to AC220 and Sorafenib. Furthermore, treatment of MOLM-13-RES cells with AC220 lead to loss of the FLT3 wild type allele and duplication of the FLT3-ITD-D835Y allele. Our FLT3-Aurora kinase inhibitor, CCT137690, successfully inhibited growth of FLT3-ITD-D835Y cells in vitro and in vivo, suggesting that dual FLT3-Aurora inhibition may overcome selective FLT3 inhibitor resistance, in part due to inhibition of Aurora kinase, and may benefit patients with FLT3-mutated AML.
Collapse
Affiliation(s)
- A S Moore
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
|
76
|
Weisberg E, Ray A, Nelson E, Adamia S, Barrett R, Sattler M, Zhang C, Daley JF, Frank D, Fox E, Griffin JD. Reversible resistance induced by FLT3 inhibition: a novel resistance mechanism in mutant FLT3-expressing cells. PLoS One 2011; 6:e25351. [PMID: 21980431 PMCID: PMC3182213 DOI: 10.1371/journal.pone.0025351] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 09/01/2011] [Indexed: 12/12/2022] Open
Abstract
Objectives Clinical responses achieved with FLT3 kinase inhibitors in acute myeloid leukemia (AML) are typically transient and partial. Thus, there is a need for identification of molecular mechanisms of clinical resistance to these drugs. In response, we characterized MOLM13 AML cell lines made resistant to two structurally-independent FLT3 inhibitors. Methods MOLM13 cells were made drug resistant via prolonged exposure to midostaurin and HG-7-85-01, respectively. Cell proliferation was determined by Trypan blue exclusion. Protein expression was assessed by immunoblotting, immunoprecipitation, and flow cytometry. Cycloheximide was used to determine protein half-life. RT-PCR was performed to determine FLT3 mRNA levels, and FISH analysis was performed to determine FLT3 gene expression. Results and Conclusions We found that MOLM13 cells readily developed cross-resistance when exposed to either midostaurin or HG-7-85-01. Resistance in both lines was associated with dramatically elevated levels of cell surface FLT3 and elevated levels of phosphor-MAPK, but not phospho-STAT5. The increase in FLT3-ITD expression was at least in part due to reduced turnover of the receptor, with prolonged half-life. Importantly, the drug-resistant phenotype could be rapidly reversed upon withdrawal of either inhibitor. Consistent with this phenotype, no significant evidence of FLT3 gene amplification, kinase domain mutations, or elevated levels of mRNA was observed, suggesting that protein turnover may be part of an auto-regulatory pathway initiated by FLT3 kinase activity. Interestingly, FLT3 inhibitor resistance also correlated with resistance to cytosine arabinoside. Over-expression of FLT3 protein in response to kinase inhibitors may be part of a novel mechanism that could contribute to clinical resistance.
Collapse
MESH Headings
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Enzyme Stability/drug effects
- Enzyme Stability/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Half-Life
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Mutation
- Phosphorylation/drug effects
- Phosphorylation/genetics
- Piperazines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Staurosporine/analogs & derivatives
- Staurosporine/pharmacology
- Thiazoles/pharmacology
- Tyrosine/metabolism
- Up-Regulation/drug effects
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- fms-Like Tyrosine Kinase 3/chemistry
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
Collapse
Affiliation(s)
- Ellen Weisberg
- Department of Medical Oncology/Hematologic Neoplasia, Dana Farber Cancer Institute, Boston, Massachusetts, United States of America.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Abstract
INTRODUCTION because of their important roles in disease and excellent 'druggability', kinases have become the second largest drug target family. The great success of the BCR-ABL inhibitor imatinib in treating chronic myelogenous leukemia illustrates the high potential of kinase inhibitor (KI) therapeutics, but also unveils a major limitation: the development of drug resistance. This is a significant concern as KIs reach large patient populations for an expanding array of indications. AREAS COVERED we provide an up-to-date understanding of the mechanisms through which KIs function and through which cells can become KI-resistant. We review current and future approaches to overcome KI resistance, focusing on currently approved KIs and KIs in clinical trials. We then discuss approaches to improve KI efficacy and overcome drug resistance and novel approaches to develop less drug resistance-prone KI therapeutics. EXPERT OPINION although drug resistance is a concern for current KI therapeutics, recent progress in our understanding of the underlying mechanisms and promising technological advances may overcome this limitation and provide powerful new therapeutics.
Collapse
Affiliation(s)
- Rina Barouch-Bentov
- Stanford University School of Medicine, Division of Infectious Disease and Geographic Medicine, Department of Medicine, Stanford, California 94305, USA
| | | |
Collapse
|
78
|
A potential therapeutic target for FLT3-ITD AML: PIM1 kinase. Leuk Res 2011; 36:224-31. [PMID: 21802138 DOI: 10.1016/j.leukres.2011.07.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 06/24/2011] [Accepted: 07/03/2011] [Indexed: 12/18/2022]
Abstract
Patients with acute myeloid leukemia (AML) and a FLT3 internal tandem duplication (ITD) mutation have a poor prognosis, and FLT3 inhibitors are now under clinical investigation. PIM1, a serine/threonine kinase, is up-regulated in FLT3-ITD AML and may be involved in FLT3-mediated leukemogenesis. We employed a PIM1 inhibitor, AR00459339 (Array Biopharma Inc.), to investigate the effect of PIM1 inhibition in FLT3-mutant AML. Like FLT3 inhibitors, AR00459339 was preferentially cytotoxic to FLT3-ITD cells, as demonstrated in the MV4-11, Molm-14, and TF/ITD cell lines, as well as 12 FLT3-ITD primary samples. Unlike FLT3 inhibitors, AR00459339 did not suppress phosphorylation of FLT3, but did promote the de-phosphorylation of downstream FLT3 targets, STAT5, AKT, and BAD. Combining AR00459339 with a FLT3 inhibitor resulted in additive to mildly synergistic cytotoxic effects. AR00459339 was cytotoxic to FLT3-ITD samples from patients with secondary resistance to FLT3 inhibitors, suggesting a novel benefit to combining these agents. We conclude that PIM1 appears to be closely associated with FLT3 signaling, and that inhibition of PIM1 may hold therapeutic promise, either as monotherapy, or by overcoming resistance to FLT3 inhibitors.
Collapse
|
79
|
Parmar A, Marz S, Rushton S, Holzwarth C, Lind K, Kayser S, Döhner K, Peschel C, Oostendorp RA, Götze KS. Stromal Niche Cells Protect Early Leukemic FLT3-ITD+ Progenitor Cells against First-Generation FLT3 Tyrosine Kinase Inhibitors. Cancer Res 2011; 71:4696-706. [DOI: 10.1158/0008-5472.can-10-4136] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
80
|
Lee J, Paek SM, Han SY. FMS-like tyrosine kinase 3 inhibitors: a patent review. Expert Opin Ther Pat 2011; 21:483-503. [DOI: 10.1517/13543776.2011.560115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
81
|
Borthakur G, Kantarjian H, Ravandi F, Zhang W, Konopleva M, Wright JJ, Faderl S, Verstovsek S, Mathews S, Andreeff M, Cortes JE. Phase I study of sorafenib in patients with refractory or relapsed acute leukemias. Haematologica 2011; 96:62-8. [PMID: 20952518 PMCID: PMC3012766 DOI: 10.3324/haematol.2010.030452] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
UNLABELLED Background Sorafenib is a multi-kinase inhibitor with activity against fms-like tyrosine kinase 3 with internal tandem duplication mutation and Raf kinase among others. A phase I dose escalation study of sorafenib was conducted in patients with advanced myelodysplastic syndrome and relapsed or refractory acute leukemias. DESIGN AND METHODS Fifty patients received one of two different schedules; Schedule "A": once or twice daily, five days per week, every week for a 21 day cycle, and Schedule "B": once or twice daily, for 14 days every 21 days. Dose limiting toxicities were grade 3/4 hypertension, hyperbilirubinemia, and amylase elevation. The recommended phase II dose in hematologic malignancies is 400 mg twice daily for both schedules. RESULTS Complete remissions or complete remissions with incomplete recovery of platelets were achieved in 5 (10%) patients (all with fms-like tyrosine kinase 3-internal tandem duplication). Significant reduction in bone marrow and/or peripheral blood blasts was seen in an additional 17 (34%) patients (all with fms-like tyrosine kinase 3-internal tandem duplication). Eleven of these responses (including 3 complete remissions/complete remissions with incomplete recovery) lasted for 2 cycles or beyond. In conclusion, sorafenib is active and well tolerated in acute myelogenous leukemia with fms-like tyrosine kinase 3 internal tandem duplication mutation. Conclusions Additional studies of sorafenib in patients with acute myelogenous leukemia, particularly those with fms-like tyrosine kinase 3 internal tandem duplication, are warranted, including sorafenib-based combinations. (ClinicalTrials.gov Identifier: NCT00217646).
Collapse
Affiliation(s)
| | | | | | - Weiguo Zhang
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M. D. Anderson Cancer Center
| | - Marina Konopleva
- Leukemia Department and, Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M. D. Anderson Cancer Center
| | - John J. Wright
- Cancer Therapy Evaluation Program, National Cancer Institute
| | | | | | | | - Michael Andreeff
- Leukemia Department and, Department of Stem Cell Transplantation and Cellular Therapy, University of Texas M. D. Anderson Cancer Center
| | - Jorge E. Cortes
- Leukemia Department and,Correspondence: Jorge E. Cortes, M.D., Leukemia Department 1515 Holcombe Blvd., Unit 428 Houston, TX 77030, USA. Phone: +1.713.7945783. Fax: +1.713.7944297. E-mail:
| |
Collapse
|
82
|
Cai J, Han S, Qing R, Liao D, Law B, Boulton ME. In pursuit of new anti-angiogenic therapies for cancer treatment. FRONT BIOSCI-LANDMRK 2011; 16:803-814. [PMID: 21196204 PMCID: PMC3627482 DOI: 10.2741/3721] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite advances in surgery, radiation therapy, and chemotherapy, patients with cancer have a poor prognosis. Sustained aberrant tumor angiogenesis and metastasis is a major obstacle for effective cancer treatment. Just a few years ago, few would argue that one of the key success stories of the modern cancer medicine were the anti-angiogenic drugs targeting the vascular endothelial growth factor (VEGF) signaling pathway approved by FDA. This initial success inspired many researchers to search for new anti-angiogenic targets and drugs with the hope that one day, anti-angiogenic therapy might really become the panacea for cancer patients. Unfortunately, the limited clinical benefits achieved with anti-angiogenic drugs conflicts with the widely accepted notion that angiogenesis is a key event in tumor progression. Emerging data indicate that unique characteristics of the tumor vasculature within the tumor microenvironment may hold the key for success of anti-angiogenic therapy. In particular, the molecular and cellular alterations that sustain aberrant tumor angiogenesis in the face of angiogenic inhibitors represents novel targets for rationally designing and improving current anti-angiogenic strategies.
Collapse
Affiliation(s)
- Jun Cai
- Department of Anatomy and Cell Biology University of Florida, Gainesville, FL 32610, USA
| | - Song Han
- Department of Surgery University of Florida, Gainesville, FL 32610, USA
| | - Ruan Qing
- Department of Anatomy and Cell Biology University of Florida, Gainesville, FL 32610, USA
| | - Daiqing Liao
- Department of Anatomy and Cell Biology University of Florida, Gainesville, FL 32610, USA
| | - Brian Law
- Department of Pharmacology and Therapeutics; University of Florida, Gainesville, FL 32610, USA
| | - Michael E. Boulton
- Department of Anatomy and Cell Biology University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
83
|
Nguewa PA, Calvo A, Pullamsetti SS, Banat GA, Grimminger F, Savai R. Tyrosine kinase inhibitors with antiangiogenic properties for the treatment of non-small cell lung cancer. Expert Opin Investig Drugs 2010; 20:61-74. [PMID: 21142806 DOI: 10.1517/13543784.2011.541153] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD The 5-year survival rate in NSCLC remains < 15% in spite of new chemotherapeutic regimens and targeted therapies. Tyrosine kinase inhibitors (TKIs) with antiangiogenic properties show promise as a new therapeutic approach for NSCLC in recent studies. AREAS COVERED IN THIS REVIEW This article reviews significant preclinical and clinical studies related to TKI therapy. Many drugs that target tyrosine kinases involved in tumor angiogenesis have recently been developed. Results of preclinical experiments and clinical trials for NSCLC are quite promising. However, response rates are low and the duration of therapeutic response is short. Mechanisms of resistance may reduce the efficacy of TKI therapy, and biomarkers of response are needed to select patients who are more likely to benefit from the therapy. Studies in mice have shown that antiangiogenic TKIs may increase metastasis, although no clear clinical evidence supports these results. WHAT THE READER WILL GAIN An understanding of the mechanisms of action, clinical trial results, biomarkers of response, adverse effects and possible mechanisms of resistance associated with novel TKI therapy in NSCLC. TAKE HOME MESSAGE More preclinical and clinical research on the efficacy of TKIs in treating NSCLC is needed, but present results offer great hope for patients.
Collapse
Affiliation(s)
- Paul A Nguewa
- Center for Applied Medical Research (CIMA), Division of Oncology, University of Navarra, Pamplona, 31008, Spain
| | | | | | | | | | | |
Collapse
|
84
|
VEGF targeted therapy in acute myeloid leukemia. Crit Rev Oncol Hematol 2010; 80:241-56. [PMID: 21035354 DOI: 10.1016/j.critrevonc.2010.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 09/24/2010] [Accepted: 09/28/2010] [Indexed: 01/07/2023] Open
Abstract
The cooperation of two classes of mutations in hematopoietic cells is hypothesized in a multistep pathogenesis model of acute myeloid leukemia (AML). Class I mutations confer a proliferative and/or survival advantage, whereas Class II mutations block hematopoietic differentiation and impair apoptosis in AML cells. In addition to these two classes of mutations, a relevant role for angiogenesis in the pathophysiology of AML has been recently proposed. The recognition that the vascular endothelial growth factor (VEGF) pathway is a key regulator of angiogenesis has led to the development of several VEGF-targeted approaches. These include neutralizing antibodies, VEGF traps or selective tyrosine kinase inhibitors for VEGFRs. Other drugs that indirectly affect VEGF pathway, such as statins or arsenic trioxide, also have been shown to possess antiangiogenic activity in leukemias. The benefits of these VEGF targeted agents and their current stage of development as novel anti-antiangiogenic therapies in AML are discussed in this review.
Collapse
|
85
|
von Bubnoff N, Gorantla SP, Engh RA, Oliveira TM, Thöne S, Aberg E, Peschel C, Duyster J. The low frequency of clinical resistance to PDGFR inhibitors in myeloid neoplasms with abnormalities of PDGFRA might be related to the limited repertoire of possible PDGFRA kinase domain mutations in vitro. Oncogene 2010; 30:933-43. [PMID: 20972453 DOI: 10.1038/onc.2010.476] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Myeloproliferation with prominent eosinophilia is associated with rearrangements of PDGFR-A or -B. The most common rearrangement is FIP1L1-PDGFRA (FP). The majority of patients with PDGFR-rearranged myeloproliferation respond to treatment with imatinib. In contrast to BCR-ABL-positive chronic myelogenous leukemia, only few cases of imatinib resistance and mutations of the FP kinase domain have been described so far. We hypothesized that the number of critical residues mediating imatinib resistance in FP in contrast to BCR-ABL might be limited. We performed an established systematic and comprehensive in vitro resistance screen to determine the pattern and frequency of possible TKI resistance mutations in FP. We identified 27 different FP kinase domain mutations including 25 novel variants, which attenuated response to imatinib, nilotinib or sorafenib. However, the majority of these exchanges did not confer complete inhibitor resistance. At clinically achievable drug concentrations, FP/T674I predominated with imatinib, whereas with nilotinib and sorafenib, FP/D842V and the compound mutation T674I+T874I became prevalent. Our results suggest that the PDGFR kinase domain contains a limited number of residues where exchanges critically interfere with binding of and inhibition by available PDGFR kinase inhibitors at achievable concentrations, which might explain the low frequency of imatinib resistance in this patient population. In addition, these findings would help to select the appropriate second-line drug in cases of imatinib-resistant disease and may be translated to other neoplasms driven by activated forms of PDGFR-A or -B.
Collapse
Affiliation(s)
- N von Bubnoff
- III. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, München, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Abstract
Mutations within the FMS-like tyrosine kinase 3 (FLT3) gene on chromosome 13q12 have been detected in up to 35% of acute myeloid leukemia (AML) patients and represent one of the most frequently identified genetic alterations in AML. Over the last years, FLT3 has emerged as a promising molecular target in therapy of AML. Here, we review results of clinical trials and of correlative laboratory studies using small molecule FLT3 tyrosine kinase inhibitors (TKIs) in AML patients. We also review mechanisms of primary and secondary drug resistance to FLT3-TKI, and from the data currently available we summarize lessons learned from FLT3-TKI monotherapy. Finally, for using FLT3 as a molecular target, we discuss novel strategies to overcome treatment failure and to improve FLT3 inhibitor therapy.
Collapse
|
87
|
Secondary resistance to sorafenib in two patients with acute myeloid leukemia (AML) harboring FLT3-ITD mutations. Ann Hematol 2010; 90:473-5. [PMID: 20652568 DOI: 10.1007/s00277-010-1027-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 06/30/2010] [Indexed: 01/27/2023]
|
88
|
Weisberg E, Sattler M, Ray A, Griffin JD. Drug resistance in mutant FLT3-positive AML. Oncogene 2010; 29:5120-34. [DOI: 10.1038/onc.2010.273] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
89
|
von Bubnoff N, Rummelt C, Menzel H, Sigl M, Peschel C, Duyster J. Identification of a secondary FLT3/A848P mutation in a patient with FLT3-ITD-positive blast phase CMML and response to sunitinib and sorafenib. Leukemia 2010; 24:1523-5. [DOI: 10.1038/leu.2010.122] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
90
|
Sierra JR, Cepero V, Giordano S. Molecular mechanisms of acquired resistance to tyrosine kinase targeted therapy. Mol Cancer 2010; 9:75. [PMID: 20385023 PMCID: PMC2864216 DOI: 10.1186/1476-4598-9-75] [Citation(s) in RCA: 172] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 04/12/2010] [Indexed: 02/07/2023] Open
Abstract
In recent years, tyrosine kinases (TKs) have been recognized as central players and regulators of cancer cell proliferation, apoptosis, and angiogenesis, and are therefore considered suitable potential targets for anti-cancer therapies. Several strategies for targeting TKs have been developed, the most successful being monoclonal antibodies and small molecule tyrosine kinase inhibitors. However, increasing evidence of acquired resistance to these drugs has been documented, and extensive preclinical studies are ongoing to try to understand the molecular mechanisms by which cancer cells are able to bypass their inhibitory activity.This review intends to present the most recently identified molecular mechanisms that mediate acquired resistance to tyrosine kinase inhibitors, identified through the use of in vitro models or the analysis of patient samples. The knowledge obtained from these studies will help to design better therapies that prevent and overcome resistance to treatment in cancer patients.
Collapse
Affiliation(s)
- J Rafael Sierra
- Institute for Cancer Research and Treatment, University of Torino Medical School, 10060 Candiolo (Torino), Italy
| | | | | |
Collapse
|
91
|
Abstract
The heterogeneity of acute myeloid leukaemia (AML) and myelodysplastic syndromes (MDS) has led to a multiplicity of treatments, from cytotoxic agents to signal transduction modulators, cell-cycle inhibitors and epigenetic therapies. While some have shown promising initial results, the outlook for AML patients, particularly older and relapsed patients, as well as patients whose cells exhibit certain adverse chromosomal abnormalities or mutant oncoproteins, continues to be grim. Combination chemotherapy using new agents that act at a number of different levels may provide the greatest potential for successful future therapies. A select number of new agents, approaches and combinations are reviewed here.
Collapse
Affiliation(s)
- Steven Grant
- Department of Medicine, Virginia Commonwealth University and Massey Cancer Center, Richmond, VA 23298-0035, USA.
| |
Collapse
|
92
|
Weisberg E, Barrett R, Liu Q, Stone R, Gray N, Griffin JD. FLT3 inhibition and mechanisms of drug resistance in mutant FLT3-positive AML. Drug Resist Updat 2009; 12:81-9. [PMID: 19467916 DOI: 10.1016/j.drup.2009.04.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 04/16/2009] [Accepted: 04/16/2009] [Indexed: 10/20/2022]
Abstract
An appealing therapeutic target in AML is constitutively activated, mutant FLT3, which is expressed in a subpopulation of AML patients and is generally a poor prognostic indicator in patients under the age of 65. There are currently several FLT3 inhibitors that are undergoing clinical investigation. However, the discovery of drug-resistant leukemic blast cells in FLT3 inhibitor-treated AML patients has prompted the search for novel, structurally diverse FLT3 inhibitors that could be alternatively used to circumvent drug resistance. Here, we provide an overview of FLT3 inhibitors under preclinical and clinical investigation, and we discuss mechanisms whereby AML cells develop resistance to FLT3 inhibitors, and the ways in which combination therapy could potentially be utilized to override drug resistance. We discuss how the cross-talk between major downstream signaling pathways, such as PI3K/PTEN/Akt/mTOR, RAS/Raf/MEK/ERK, and Jak/STAT, can be exploited for therapeutic purposes by targeting key signaling molecules with selective inhibitors, such as mTOR inhibitors, HSP90 inhibitors, or farnesyltransferase inhibitors, and identifying those agents with the ability to positively combine with inhibitors of FLT3, such as PKC412 and sunitinib. With the widespread onset of drug resistance associated with tyrosine kinase inhibitors, due to mechanisms involving development of point mutations or gene amplification of target proteins, the use of a multi-targeted therapeutic approach is of potential clinical benefit.
Collapse
Affiliation(s)
- Ellen Weisberg
- Department of Medical Oncology/Hematologic Neoplasia, Dana Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA.
| | | | | | | | | | | |
Collapse
|