240 results on '"crenolanib"'
Search Results
2. Study of Crenolanib Combined With Chemotherapy in FLT3-mutated Acute Myeloid Leukemia Patients
- Published
- 2024
3. A Safety and Tolerability Study of Crenolanib in Combination With Chemotherapy in Newly Diagnosed Acute Myeloid Leukemia Patients With FLT3 Mutations
- Published
- 2024
4. Phase II Study of Crenolanib in Subjects With Relapsed/Refractory AML With FLT3 Activating Mutations
- Published
- 2024
5. Pilot Study of Crenolanib Combined With Standard Salvage Chemotherapy in Subjects With R/R AML
- Published
- 2023
6. Crenolanib Maintenance Following Allogeneic Stem Cell Transplantation in FLT3-positive Acute Myeloid Leukemia Patients
- Published
- 2023
7. A Phase II Study of Crenolanib in Relapsed/Refractory Acute Myeloid Leukemia Patients With FLT3 Activating Mutations
- Published
- 2023
8. Filamin C Deficiency Impairs Sarcomere Stability and Activates Focal Adhesion Kinase through PDGFRA Signaling in Induced Pluripotent Stem Cell-Derived Cardiomyocytes.
- Author
-
Gao, Shanshan, He, Lingaonan, Lam, Chi Keung, Taylor, Matthew R. G., Mestroni, Luisa, Lombardi, Raffaella, and Chen, Suet Nee
- Subjects
- *
FOCAL adhesion kinase , *CELL adhesion , *FOCAL adhesions , *CONNECTIN , *DILATED cardiomyopathy , *MEMBRANE proteins , *MICROFILAMENT proteins , *CYTOSKELETAL proteins - Abstract
Truncating mutations in filamin C (FLNC) are associated with dilated cardiomyopathy and arrhythmogenic cardiomyopathy. FLNC is an actin-binding protein and is known to interact with transmembrane and structural proteins; hence, the ablation of FLNC in cardiomyocytes is expected to dysregulate cell adhesion, cytoskeletal organization, sarcomere structural integrity, and likely nuclear function. Our previous study showed that the transcriptional profiles of FLNC homozygous deletions in human pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are highly comparable to the transcriptome profiles of hiPSC-CMs from patients with FLNC truncating mutations. Therefore, in this study, we used CRISPR-Cas-engineered hiPSC-derived FLNC knockout cardiac myocytes as a model of FLNC cardiomyopathy to determine pathogenic mechanisms and to examine structural changes caused by FLNC deficiency. RNA sequencing data indicated the significant upregulation of focal adhesion signaling and the dysregulation of thin filament genes in FLNC-knockout (FLNCKO) hiPSC-CMs compared to isogenic hiPSC-CMs. Furthermore, our findings suggest that the complete loss of FLNC in cardiomyocytes led to cytoskeletal defects and the activation of focal adhesion kinase. Pharmacological inhibition of PDGFRA signaling using crenolanib (an FDA-approved drug) reduced focal adhesion kinase activation and partially normalized the focal adhesion signaling pathway. The findings from this study suggest the opportunity in repurposing FDA-approved drug as a therapeutic strategy to treat FLNC cardiomyopathy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Study of Crenolanib in Combination With Chemotherapy in Patients With Relapsed or Refractory Acute Myeloid Leukemia and Activating FLT3 Mutations
- Author
-
Prof. Dr. Hartmut Doehner, Prof. Dr.
- Published
- 2021
10. Study Investigating the Efficacy of Crenolanib With Chemotherapy vs Chemotherapy Alone in R/R FLT3 Mutated AML
- Published
- 2021
11. Randomized Trial of Crenolanib in Subjects With D842V Mutated GIST
- Author
-
Centre Leon Berard and Fox Chase Cancer Center
- Published
- 2021
12. Filamin C Deficiency Impairs Sarcomere Stability and Activates Focal Adhesion Kinase through PDGFRA Signaling in Induced Pluripotent Stem Cell-Derived Cardiomyocytes
- Author
-
Shanshan Gao, Lingaonan He, Chi Keung Lam, Matthew R. G. Taylor, Luisa Mestroni, Raffaella Lombardi, and Suet Nee Chen
- Subjects
filamin C ,dilated cardiomyopathy ,PDGFRA ,crenolanib ,Cytology ,QH573-671 - Abstract
Truncating mutations in filamin C (FLNC) are associated with dilated cardiomyopathy and arrhythmogenic cardiomyopathy. FLNC is an actin-binding protein and is known to interact with transmembrane and structural proteins; hence, the ablation of FLNC in cardiomyocytes is expected to dysregulate cell adhesion, cytoskeletal organization, sarcomere structural integrity, and likely nuclear function. Our previous study showed that the transcriptional profiles of FLNC homozygous deletions in human pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are highly comparable to the transcriptome profiles of hiPSC-CMs from patients with FLNC truncating mutations. Therefore, in this study, we used CRISPR-Cas-engineered hiPSC-derived FLNC knockout cardiac myocytes as a model of FLNC cardiomyopathy to determine pathogenic mechanisms and to examine structural changes caused by FLNC deficiency. RNA sequencing data indicated the significant upregulation of focal adhesion signaling and the dysregulation of thin filament genes in FLNC-knockout (FLNCKO) hiPSC-CMs compared to isogenic hiPSC-CMs. Furthermore, our findings suggest that the complete loss of FLNC in cardiomyocytes led to cytoskeletal defects and the activation of focal adhesion kinase. Pharmacological inhibition of PDGFRA signaling using crenolanib (an FDA-approved drug) reduced focal adhesion kinase activation and partially normalized the focal adhesion signaling pathway. The findings from this study suggest the opportunity in repurposing FDA-approved drug as a therapeutic strategy to treat FLNC cardiomyopathy.
- Published
- 2024
- Full Text
- View/download PDF
13. Study of Crenolanib in Recurrent/Refractory Glioblastoma With PDGFRA Gene Amplification
- Published
- 2020
14. Study of Crenolanib With Ramucirumab and Paclitaxel for Advanced Esophagogastric Adenocarcinoma
- Published
- 2020
15. Study of Crenolanib vs Midostaurin Following Induction Chemotherapy and Consolidation Therapy in Newly Diagnosed FLT3 Mutated AML
- Published
- 2020
16. Drug-drug interactions associated with FLT3 inhibitors for acute myeloblastic leukemia: current landscape.
- Author
-
Solana-Altabella, Antonio, Megías-Vericat, Juan Eduardo, Ballesta-López, Octavio, Martínez-Cuadrón, David, and Montesinos, Pau
- Subjects
ACUTE myeloid leukemia ,DRUG interactions ,MEDICAL personnel ,CYTOCHROME P-450 ,PATIENT monitoring - Abstract
FLT3 inhibitors (FLT3i) are drugs in which there is limited experience and not yet enough information on the mechanisms of absorption, transport, and elimination; but especially on the potential drug-drug interactions (DDIs). There are therefore risks in the management of FLT3i DDIs (i.e. sorafenib, ponatinib, crenolanib, midostaurin, quizartinib, and gilteritinib) and ignoring them can compromise therapeutic success in acute myeloid leukemia (AML) treatment, in complex patients and secondary pathologies. This review summarizes the DDIs of FLT3i with P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), organic anion transporting (OAT), organic cationic transporting (OCT), cytochrome P450 (CYP) subunits, and other minor metabolic/transport pathways. EMBASE, PubMed, the Cochrane Central Register and the Web of Science were searched. The last literature search was performed on the 14 February 2022. FLT3i will be combined with other therapeutic agents (supportive care, doublet, or triplet therapy) and in different clinical settings, which means a greater chance of controlling and even eradicating the disease effectively, but also an increased risk to patients due to potential DDIs. Healthcare professionals should be aware of the potential interactions that may occur and be vigilant in monitoring those patients who are receiving any potentially interacting drug. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. Overcoming Resistance: FLT3 Inhibitors Past, Present, Future and the Challenge of Cure.
- Author
-
Capelli, Debora, Menotti, Diego, Fiorentini, Alessandro, Saraceni, Francesco, and Olivieri, Attilio
- Subjects
- *
PROTEIN metabolism , *HOMOGRAFTS , *CANCER relapse , *ALLELES , *PROTEIN-tyrosine kinase inhibitors , *TRANSFERASES , *DRUG resistance in cancer cells , *IMMUNOTHERAPY , *DISEASE remission - Abstract
Simple Summary: Despite the recent approval of some FLT3 inhibitors by drug regulatory agencies, treatment guidelines for FLT3-mutated AML still require allogeneic transplantation as a necessary procedure to treat the disease in first or second CR, due to the high relapse incidence related to the use of these drugs. The study of the heterogeneity of leukemogenesis and resistance mechanisms related to the use of FLT3 inhibitors, alone or in combination, represents one of the additional challenges in attempting to achieve the eradication of the mutated FLT3 leukemic clone. The analysis and knowledge of these pathways might drive future approach in this setting. FLT3 ITD and TKD mutations occur in 20% and 10% of Acute Myeloid Leukemia (AML), respectively, and they represent the target of the first approved anti-leukemic therapies in the 2000s. Type I and type II FLT3 inhibitors (FLT3i) are active against FLT3 TKD/ITD and FLT3 ITD mutations alone respectively, but they still fail remissions in 30–40% of patients due to primary and secondary mechanisms of resistance, with variable relapse rate of 30–50%, influenced by NPM status and FLT3 allelic ratio. Mechanisms of resistance to FLT3i have recently been analyzed through NGS and single cell assays that have identified and elucidated the polyclonal nature of relapse in clinical and preclinical studies, summarized here. Knowledge of tumor escape pathways has helped in the identification of new targeted drugs to overcome resistance. Immunotherapy and combination or sequential use of BCL2 inhibitors and experimental drugs including aurora kinases, menin and JAK2 inhibitors will be the goal of present and future clinical trials, especially in patients with FLT3-mutated (FLT3mut) AML who are not eligible for allogeneic transplantation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
18. A Study of Crenolanib With Fludarabine and Cytarabine in Pediatric Patients With Relapsed/Refractory FLT3-Mutated Acute Myeloid Leukemia
- Published
- 2019
19. "FLipping" the Story: FLT3-Mutated Acute Myeloid Leukemia and the Evolving Role of FLT3 Inhibitors.
- Author
-
Knight, Tristan E., Edwards, Holly, Meshinchi, Soheil, Taub, Jeffrey W., and Ge, Yubin
- Subjects
- *
THERAPEUTIC use of antineoplastic agents , *GENETIC mutation , *PROTEIN kinase inhibitors , *CELLULAR signal transduction , *PROTEIN-tyrosine kinases , *TRANSFERASES , *CHEMICAL inhibitors - Abstract
Simple Summary: Patients with acute myeloid leukemia (AML) may have a number of different mutations. Those with mutations in the FLT3 gene have a higher risk of relapse and death than those lacking these mutations. FLT3 is a key receptor on the surface of AML cells, which drives cell survival and growth. Although activation of this receptor is normally tightly controlled, in AML, FLT3 mutations allow it to activate itself, independent of external control. Over the past 5 years, a number of new drugs have been developed to specifically target these mutations. In this article, we discuss these drugs and their uses, as well as the mechanisms by which AML cells may gain resistance to them and how that resistance can be overcome. The treatment of many types of cancers, including acute myeloid leukemia (AML), has been revolutionized by the development of therapeutics targeted at crucial molecular drivers of oncogenesis. In contrast to broad, relatively indiscriminate conventional chemotherapy, these targeted agents precisely disrupt key pathways within cancer cells. FMS-like tyrosine kinase 3 (FLT3)—encoding a critical regulator of hematopoiesis—is the most frequently mutated gene in patients with AML, and these mutations herald reduced survival and increased relapse in these patients. Approximately 30% of newly diagnosed AML carries an FLT3 mutation; of these, approximately three-quarters are internal tandem duplication (ITD) mutations, and the remainder are tyrosine kinase domain (TKD) mutations. In contrast to its usual, tightly controlled expression, FLT3-ITD mutants allow constitutive, "run-away" activation of a large number of key downstream pathways which promote cellular proliferation and survival. Targeted inhibition of FLT3 is, therefore, a promising therapeutic avenue. In April 2017, midostaurin became both the first FLT3 inhibitor and the first targeted therapy of any kind in AML to be approved by the US FDA. The use of FLT3 inhibitors has continued to grow as clinical trials continue to demonstrate the efficacy of this class of agents, with an expanding number available for use as both experimental standard-of-care usage. This review examines the biology of FLT3 and its downstream pathways, the mechanism of FLT3 inhibition, the development of the FLT3 inhibitors as a class and uses of the agents currently available clinically, and the mechanisms by which resistance to FLT3 inhibition may both develop and be overcome. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
20. An overview of agents and treatments for PDGFRA-mutated gastrointestinal stromal tumors
- Author
-
Yingchao Sun, Lei Yue, Pengfu Xu, and Weiling Hu
- Subjects
PDGFRA mutation ,targeted therapy ,avapritinib ,ripretinib ,crenolanib ,gastrointestinal stromal tumors (GIST) ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Platelet-derived growth factor receptor A (PDGFRA) mutations occur in approximately 10–15% of gastrointestinal stromal tumors (GISTs). These tumors with PDGFRA mutations have a different pathogenesis, clinical characteristics, and treatment response compared to tumors with receptor tyrosine kinase protein (KIT) mutations (60–70%). Many clinical studies have investigated the use of tyrosine kinase inhibitors mainly in patients with KIT mutations; however, there is a lack of attention to the PDGFRA-mutated molecular subtype. The main effective inhibitors of PDGFRA are ripretinib, avapritinib, and crenolanib, and their mechanisms and efficacy in GIST (as confirmed in clinical trials) are described in this review. Some multi-targeted tyrosine kinase inhibitors with inhibitory effects on this molecular subtype are also introduced and summarized in this paper. This review focuses on PDGFRA-mutated GISTs, introduces their clinical characteristics, downstream molecular signaling pathways, and existing resistance mechanisms. We focus on the most recent literature that describes the development of PDGFRA inhibitors and their use in clinical trials, as well as the potential benefits from different combination therapy strategies.
- Published
- 2022
- Full Text
- View/download PDF
21. Study of Crenolanib, a Selective and Potent Inhibitor of PDGFR, for the Treatment of Adult Gliomas
- Published
- 2018
22. Study of Crenolanib for the Treatment of Patients With Advanced GIST With the D842-related Mutations and Deletions in the PDGFRA Gene
- Published
- 2018
23. Population pharmacokinetics of crenolanib in children and young adults with brain tumors.
- Author
-
Bisbee, Cora, Campagne, Olivia, Gajjar, Amar, Tinkle, Christopher L., and Stewart, Clinton F.
- Subjects
- *
YOUNG adults , *BRAIN tumors , *PLATELET-derived growth factor receptors , *PHARMACOKINETICS , *BODY surface area , *PROTON pump inhibitors - Abstract
Purpose: Crenolanib, an oral inhibitor of platelet-derived growth factor receptor, was evaluated to treat children and young adults with brain tumors. Crenolanib population pharmacokinetics and covariate influence were characterized in this patient population. Methods: Patients enrolled on this phase I study (NCT01393912) received oral crenolanib once daily. Serial single-dose and steady-state serum pharmacokinetic samples were collected and analyzed using a validated LC–ESI–MS/MS method. Population modeling and covariate analysis evaluating demographics, laboratory values, and comedications were performed. The impact of significant covariates on crenolanib exposure was further explored using model simulations. Results: Crenolanib serum concentrations were analyzed for 55 patients (2.1–19.2 years-old) and best fitted with a linear two-compartment model, with delayed absorption modeled with a lag time. A typical patient [8-year-old, body surface area (BSA) 1 m2] had an apparent central clearance, volume, and absorption rate of 41 L/h, 54.3 L, and 0.19 /h, respectively. Patients taking acid reducers (histamine H2 antagonists or proton pump inhibitors) concomitantly exhibited about 2- and 1.7-fold lower clearance and volume (p < 0.0001 and p = 0.018, respectively). Crenolanib clearance increased with BSA (p < 0.0001), and absorption rate decreased with age (p < 0.0001). Model simulations showed cotreatment with an acid reducer was the only covariate significantly altering crenolanib exposure and supported the use of BSA-based crenolanib dosages vs flat-dosages for this population. Conclusions: Crenolanib pharmacokinetics were adequately characterized in children and young adults with brain tumors. Despite marked increased drug exposure with acid reducer cotreatment, crenolanib therapy was well tolerated. No dosing adjustments are recommended for this population. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
24. Phase I/Ib study of crenolanib with ramucirumab and paclitaxel as second-line therapy for advanced esophagogastric adenocarcinoma.
- Author
-
Moy, Ryan H., Greally, Megan, Chou, Joanne F., Li, Jia, Desai, Avni M., Chalasani, Sree B., Won, Elizabeth, Kelsen, David P., Ilson, David H., Janjigian, Yelena Y., Capanu, Marinela, and Ku, Geoffrey Y.
- Subjects
- *
PACLITAXEL , *WITHDRAWAL of funds , *ADENOCARCINOMA , *PROGRESSION-free survival - Abstract
Purpose: Paclitaxel plus ramucirumab is a standard second-line regimen for patients with advanced gastric adenocarcinoma, but clinical benefit remains modest. One potential resistance mechanism to VEGFR2 inhibition is activation of the PDGF/PDGFR pathway, which can be blocked by the selective inhibitor crenolanib. Therefore, we performed a phase I/Ib study of crenolanib in combination with paclitaxel/ramucirumab. Methods: Patients with metastatic esophagogastric adenocarcinoma refractory to first-line therapy received escalating doses of crenolanib [60 mg twice daily (BID) to 100 mg three times daily (TID)] in combination with paclitaxel 80 mg/m2 intravenously on days 1, 8 and 15 and ramucirumab 8 mg/kg intravenously on days 1 and 15 of a 28-day cycle. The primary objective was to determine the maximally tolerated dose (MTD) of crenolanib. Additional patients were enrolled in the dose expansion cohort to assess 6-month progression-free survival (PFS) at the MTD. Results: We enrolled 19 patients in the dose escalation phase and 8 patients in the dose expansion phase at the MTD of crenolanib 100 mg BID. Common grade 3/4 treatment-emergent adverse events included leukopenia (19%), anemia (11%) and neutropenia (11%). In the 14 patients treated at the MTD, 6-month PFS was 43% [95% confidence interval (CI) 23–78%] and the objective response rate (ORR) was 42% (95% CI 15–72%). The trial was terminated early due to withdrawal of crenolanib by the sponsor. Conclusions: The addition of crenolanib to paclitaxel/ramucirumab is safe and well-tolerated at a dose level up to 100 mg BID. Clinical trial registration: NCT03193918. June 19, 2017. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
25. Crenolanib in Combination With Sorafenib in Patients With Refractory or Relapsed Hematologic Malignancies
- Author
-
Arog Pharmaceuticals, Inc. and Ohio State University
- Published
- 2017
26. PDGFR Inhibitor Crenolanib in Children/Young Adults With Diffuse Intrinsic Pontine Glioma or Recurrent High-Grade Glioma
- Author
-
Arog Pharmaceuticals, Inc. and The V Foundation for Cancer Research
- Published
- 2017
27. Activation PDGFR-α/AKT Mediated Signaling Pathways in Oral Squamous Cell Carcinoma by Mesenchymal Stem/Stromal Cells Promotes Anti-apoptosis and Decreased Sensitivity to Cisplatin
- Author
-
Jia Wang, Ruwen Cui, Cecila G. Clement, Ranjana Nawgiri, Don W. Powell, Irina V. Pinchuk, and Tammara L. Watts
- Subjects
PDGFR-α ,AKT ,oral cancer ,crenolanib ,apoptosis ,mesenchymal stem cells ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Desmoplasia, a hallmark of a head and neck cancer, has both biologic and physiologic effects on cancer progression and chemotherapeutic response. Mesenchymal stem/stromal cells (MSCs), also known as mesenchymal stromal progenitor cells, have been shown to play a role in cancer progression, alter apoptotic responses, and confer resistance to chemotherapy in various carcinomas. The pathophysiology of MSCs with respect to tumorigenesis is widely reported in other cancers and is sparsely reported in oral squamous cell carcinomas (OSCCs). We previously reported paracrine mediated PDGF-AA/PDGFR-α signaling to underlie MSCs chemotaxis in OSCC. Given the poor clinical response to primary chemotherapy, we hypothesized that MSCs may alter cancer cell sensitivity to cisplatin through activation of PDGFR-α mediated signaling pathways. Co-culture of MSCs with human derived OSCC cell lines, JHU-012 and −019, resulted in a significant increase in the production of PDGF-AA and MCP-1 compared to cancer cells grown alone (p < 0.005) and was accompanied by an increase in the phosphorylation state of PDGFR-α (p < 0.02) and downstream target AKT at S473 (p < 0.025) and T308 (p < 0.02). JHU-012 and −019 cancer cells grown in co-culture were significantly less apoptotic (p < 0.001), expressed significantly higher levels of Bcl-2 (p < 0.04) with a concomitant significant decrease in bid expression (p < 0.001) compared to cancer cells grown alone. There was a significant increase in the cisplatin dose response curve in cancer cell clones derived from JHU-012 and 019 cancer cells grown in co-culture with MSCs compared to clones derived from cancer cells grown alone (p < 0.001). Moreover clones derived from JHU-012 cells grown in co-culture with MSCs were significantly more susceptible to cisplatin following pretreatment with, crenolanib, a PDGFR inhibitor, compared to cancer cells grown alone or in co-culture with MSCs (p < 0.0001). These findings suggest that crosstalk between cancer cells and MSCs is mediated, at least in part, by activation of autocrine PDGF-AA/PDGFR-α loop driving AKT-mediated signaling pathways, resulting in reduced cancer cell sensitivity to cisplatin through alterations in apoptosis.
- Published
- 2020
- Full Text
- View/download PDF
28. Acute myeloid leukemia transformed to a targetable disease.
- Author
-
Saleh, Khalil, Khalifeh-Saleh, Nadine, and Kourie, Hampig Raphael
- Abstract
Acute myeloid leukemia (AML) is a heterogeneous neoplasm characterized by the monoclonal proliferation of immature progenitors. It is the most common acute leukemia in adults and its incidence increases with age. The standard traditional treatment in fit patients was the '3 + 7' regimen and cytarabine consolidation followed or not with allogeneic stem cell transplantation. Recently, several targeted therapies such as gemtuzumab ozogamicin targeting the CD33+ AML, midostaurin, gilteritinib and crenolanib inhibiting FLT3-positive AML and ivosidenib and enasidenib blocking IDH-mutated AML have been approved. These new drugs led to the change of the landscape of the treatment of AML and transforming this disease to a targetable one. We aimed in this paper to review the implications of each new target, the mechanisms of action of these new drugs and we discuss all the studies leading to the approval of these new drugs in their indications according to each target. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
29. Activation PDGFR-α/AKT Mediated Signaling Pathways in Oral Squamous Cell Carcinoma by Mesenchymal Stem/Stromal Cells Promotes Anti-apoptosis and Decreased Sensitivity to Cisplatin.
- Author
-
Wang, Jia, Cui, Ruwen, Clement, Cecila G., Nawgiri, Ranjana, Powell, Don W., Pinchuk, Irina V., and Watts, Tammara L.
- Subjects
MESENCHYMAL stem cells ,STROMAL cells ,SQUAMOUS cell carcinoma ,CISPLATIN ,PROGENITOR cells ,CANCER cells - Abstract
Desmoplasia, a hallmark of a head and neck cancer, has both biologic and physiologic effects on cancer progression and chemotherapeutic response. Mesenchymal stem/stromal cells (MSCs), also known as mesenchymal stromal progenitor cells, have been shown to play a role in cancer progression, alter apoptotic responses, and confer resistance to chemotherapy in various carcinomas. The pathophysiology of MSCs with respect to tumorigenesis is widely reported in other cancers and is sparsely reported in oral squamous cell carcinomas (OSCCs). We previously reported paracrine mediated PDGF-AA/PDGFR-α signaling to underlie MSCs chemotaxis in OSCC. Given the poor clinical response to primary chemotherapy, we hypothesized that MSCs may alter cancer cell sensitivity to cisplatin through activation of PDGFR-α mediated signaling pathways. Co-culture of MSCs with human derived OSCC cell lines, JHU-012 and −019, resulted in a significant increase in the production of PDGF-AA and MCP-1 compared to cancer cells grown alone (p < 0.005) and was accompanied by an increase in the phosphorylation state of PDGFR-α (p < 0.02) and downstream target AKT at S473 (p < 0.025) and T308 (p < 0.02). JHU-012 and −019 cancer cells grown in co-culture were significantly less apoptotic (p < 0.001), expressed significantly higher levels of Bcl-2 (p < 0.04) with a concomitant significant decrease in bid expression (p < 0.001) compared to cancer cells grown alone. There was a significant increase in the cisplatin dose response curve in cancer cell clones derived from JHU-012 and 019 cancer cells grown in co-culture with MSCs compared to clones derived from cancer cells grown alone (p < 0.001). Moreover clones derived from JHU-012 cells grown in co-culture with MSCs were significantly more susceptible to cisplatin following pretreatment with, crenolanib, a PDGFR inhibitor, compared to cancer cells grown alone or in co-culture with MSCs (p < 0.0001). These findings suggest that crosstalk between cancer cells and MSCs is mediated, at least in part, by activation of autocrine PDGF-AA/PDGFR-α loop driving AKT-mediated signaling pathways, resulting in reduced cancer cell sensitivity to cisplatin through alterations in apoptosis. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
30. Effects of the multi‐kinase inhibitor midostaurin in combination with chemotherapy in models of acute myeloid leukaemia.
- Author
-
Weisberg, Ellen, Meng, Chengcheng, Case, Abigail E., Tiv, Hong L., Gokhale, Prafulla C., Buhrlage, Sara J., Yang, Jing, Liu, Xiaoxi, Wang, Jinhua, Gray, Nathanael, Adamia, Sophia, Sattler, Martin, Stone, Richard, and Griffin, James D.
- Subjects
COMBINATION drug therapy ,LEUKEMIA ,CELL lines ,KINASE inhibitors ,EXTRACELLULAR signal-regulated kinases - Abstract
Recently, several targeted agents have been developed for specific subsets of patients with acute myeloid leukaemia (AML), including midostaurin, the first FDA‐approved FLT3 inhibitor for newly diagnosed patients with FLT3 mutations. However, in the initial Phase I/II clinical trials, some patients without FLT3 mutations had transient responses to midostaurin, suggesting that this multi‐targeted kinase inhibitor might benefit AML patients more broadly. Here, we demonstrate submicromolar efficacy of midostaurin in vitro and efficacy in vivo against wild‐type (wt) FLT3‐expressing AML cell lines and primary cells, and we compare its effectiveness with that of other FLT3 inhibitors currently in clinical trials. Midostaurin was found to synergize with standard chemotherapeutic drugs and some targeted agents against AML cells without mutations in FLT3. The mechanism may involve, in part, the unique kinase profile of midostaurin that includes proteins implicated in AML transformation, such as SYK or KIT, or inhibition of ERK pathway or proviability signalling. Our findings support further investigation of midostaurin as a chemosensitizing agent in AML patients without FLT3 mutations. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
31. Succinate dehydrogenase deficiency in a PDGFRA mutated GIST
- Author
-
Martin G. Belinsky, Kathy Q. Cai, Yan Zhou, Biao Luo, Jianming Pei, Lori Rink, and Margaret von Mehren
- Subjects
Gastrointestinal stromal tumor ,Kit ,Platelet derived growth factor receptor alpha ,Succinate dehydrogenase ,Imatinib mesylate ,Crenolanib ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Most gastrointestinal stromal tumors (GISTs) harbor mutually exclusive gain of function mutations in the receptor tyrosine kinase (RTK) KIT (70–80%) or in the related receptor PDGFRA (~10%). These GISTs generally respond well to therapy with the RTK inhibitor imatinib mesylate (IM), although initial response is genotype-dependent. An alternate mechanism leading to GIST oncogenesis is deficiency in the succinate dehydrogenase (SDH) enzyme complex resulting from genetic or epigenetic inactivation of one of the four SDH subunit genes (SDHA, SDHB, SDHC, SDHD, collectively referred to as SDHX). SDH loss of function is generally seen only in GIST lacking RTK mutations, and SDH-deficient GIST respond poorly to imatinib therapy. Methods Tumor and normal DNA from a GIST case carrying the IM-resistant PDGFRA D842V mutation was analyzed by whole exome sequencing (WES) to identify additional potential targets for therapy. The tumors analyzed were separate recurrences following progression on imatinib, sunitinib, and the experimental PDGFRA inhibitor crenolanib. Tumor sections from the GIST case and a panel of ~75 additional GISTs were subjected to immunohistochemistry (IHC) for the SDHB subunit. Results Surprisingly, a somatic, loss of function mutation in exon 4 of the SDHB subunit gene (c.291_292delCT, p.I97Mfs*21) was identified in both tumors. Sanger sequencing confirmed the presence of this inactivating mutation, and IHC for the SDHB subunit demonstrated that these tumors were SDH-deficient. IHC for the SDHB subunit across a panel of ~75 GIST cases failed to detect SDH deficiency in other GISTs with RTK mutations. Conclusions This is the first reported case of a PDGFRA mutant GIST exhibiting SDH-deficiency. A brief discussion of the relevant GIST literature is included.
- Published
- 2017
- Full Text
- View/download PDF
32. A Phase 2 Study in Patients With Advanced Non-Small Cell Lung Cancer Using New Agents With and Without Docetaxel.
- Published
- 2012
33. CP-868,596 And CP-868,596 Plus AG-013736 In Combination With Docetaxel In Advanced Solid Tumors
- Published
- 2012
34. Clinical considerations for the use of FLT3 inhibitors in acute myeloid leukemia.
- Author
-
Weis, Taylor M., Marini, Bernard L., Bixby, Dale L., and Perissinotti, Anthony J.
- Subjects
- *
ACUTE myeloid leukemia , *PROTEIN-tyrosine kinases , *COMBINATION drug therapy - Abstract
Internal tandem duplications and tyrosine kinase mutations in the fms-like tyrosine kinase 3 (FLT3) receptor can occur in acute myeloid leukemia (AML) and portend a poor prognosis. Midostaurin, a multikinase inhibitor that targets FLT3, demonstrated a survival benefit in FLT3-mutated AML in combination with front-line chemotherapy. Despite this advancement, the use of FLT3 inhibitors in clinical practice is complicated by significant drug-drug interactions and uncertainty about optimal timing, duration, and sequencing of therapy. As monotherapy, the utility of FLT3 inhibitors was initially limited by incomplete and transient clinical responses and the development of acquired resistance. This led to the development of more potent and selective FLT3 inhibitors designed to overcome common resistance mechanisms. One of these second generation FLT3 inhibitors, gilteritinib, is now FDA-approved for the treatment of relapsed or refractory AML. Now that multiple FLT3 inhibitors are commercially available, it is important to further delineate the role of these agents in the AML population. This review aims to provide a comprehensive overview of the role of FLT3 inhibitors in AML and apply the current literature to clinical practice. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
35. Incorporating FLT3 inhibitors in the frontline treatment of FLT3 mutant acute myeloid leukemia.
- Author
-
Wang, Eunice S.
- Abstract
FLT3 mutations occur in up to a third of newly diagnosed patients with acute myeloid leukemia (AML) and confer poor prognosis. Clinical development of FLT3 tyrosine kinase inhibitors for AML initially involved broad-spectrum inhibitors (midostaurin, sorafenib) targeting multiple kinases. Addition of midostaurin to upfront intensive chemotherapy for younger patients with FLT3 mutant AML significantly improved overall survival and validated FLT3 as a therapeutic target. Other regimens such as sorafenib and hypomethylating agents (azacitidine, decitabine) have expanded the use of FLT3 inhibitors to other populations with FLT3 mutant AML. However, emerging data on new highly potent and specific FLT3 inhibitors such as quizartinib, gilteritinib, and crenolanib suggest that these agents may soon supplant midostaurin and sorafenib in the upfront setting. Using case presentations, this review provides guidelines and practical management strategies for frontline therapy of patients with newly diagnosed FLT3 mutant AML in the current era. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
36. FMS-Like Tyrosine Kinase 3 Inhibitors for the Treatment of Acute Myeloid Leukemia
- Author
-
Elli D. Novatcheva, James K. Mangan, Ila M Saunders, Yasmine Anouty, and Aaron M. Goodman
- Subjects
Cancer Research ,NPM1 ,Antineoplastic Agents ,Receptor tyrosine kinase ,chemistry.chemical_compound ,hemic and lymphatic diseases ,Humans ,Medicine ,Midostaurin ,Protein Kinase Inhibitors ,neoplasms ,Quizartinib ,Acute leukemia ,biology ,business.industry ,Myeloid leukemia ,Hematology ,Sorafenib ,Leukemia, Myeloid, Acute ,fms-Like Tyrosine Kinase 3 ,Oncology ,chemistry ,Mutation ,embryonic structures ,Fms-Like Tyrosine Kinase 3 ,biology.protein ,Cancer research ,business ,Crenolanib - Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia of adults, with a five-year survival that remains poor (approximately 25%). Knowledge and understanding of AML genomics have expanded tremendously over the past decade and are now included in AML prognostication and treatment decisions. FMS-like tyrosine kinase 3 (FLT3) is a Class III receptor tyrosine kinase (RTK) expressed primarily in the cell membranes of early hematopoietic progenitor cells, found in 28% of all patients with AML. FLT3 is the second most frequent mutation in adult AML following Nuclear-cytoplasmic shuttling phosphoprotein (NPM1), which is found in 50% of cases.1 FLT3 inhibitors are promising new molecular therapeutics increasingly becoming standard of care for both newly diagnosed and relapsed/refractory FLT3 positive AML. This review will focus on the clinical trials/evidence, similarities, differences, clinical toxicities, and drug interactions relevant to treating clinicians as pertains to 5 FLT3-inhibitors: midostaurin, sorafenib, gilteritinib, crenolanib, and quizartinib.
- Published
- 2022
37. Emerging FLT3 inhibitors for the treatment of acute myeloid leukemia
- Author
-
Antonio Solana-Altabella, Octavio Ballesta-López, Juan Eduardo Megías-Vericat, David Martínez-Cuadrón, and Pau Montesinos
- Subjects
Pharmacology ,hypomethylating agents ,venetoclax ,Antineoplastic Agents ,acute myeloid leukemia ,crenolanib ,quizartinib ,Leukemia, Myeloid, Acute ,midostaurin ,fms-Like Tyrosine Kinase 3 ,FLT3 inhibitors ,Mutation ,Humans ,Pharmacology (medical) ,gilteritinib ,Protein Kinase Inhibitors - Abstract
INTRODUCTION: The FMS-like tyrosine kinase 3 (FLT3) gene is mutated in one-third of patients with Acute Myeloid Leukemia (AML). Midostaurin, quizartinib, and gilteritinib have been approved in the last years for the treatment of AML, and more Tyrosine Kinase Inhibitors (TKIs) targeting FLT3 are being developed such as crenolanib. AREAS COVERED: In this systematic review, we will analyze the available clinical data on FLT3 inhibitors in development and describe the potential role that these FLT3-TKIs may play in the future management of FLT3-mutated (FLT3mut) AML. EXPERT OPINION: Although several aspects may challenge the use of FLT3 inhibitors in AML (resistance mechanisms, on- and off-target toxicities or drug-drug interactions), these drugs are generally well tolerated, particularly if we compare their safety profile with classical chemotherapy agents or even with newer immunotherapies, thus enabling their use in fit and unfit AML patients, alone or combined. As AML is a polyclonal disease and FLT3 mutations are a late leukemogenic event, combinations of these FLT3 inhibitors with other antileukemic agents (like venetoclax or hypomethylating agents) seem a necessary research pathway.
- Published
- 2022
38. Wu-5, a novel USP10 inhibitor, enhances crenolanib-induced FLT3-ITD-positive AML cell death via inhibiting FLT3 and AMPK pathways
- Author
-
Yu, Miao, Fang, Zhi-xiao, Wang, Wei-wei, Zhang, Ying, Bu, Zhi-lei, Liu, Meng, Xiao, Xin-hua, Zhang, Zi-lu, Zhang, Xing-ming, Cao, Yang, Wang, Ying-ying, Lei, Hu, Xu, Han-zhang, Wu, Yun-zhao, Liu, Wei, and Wu, Ying-li
- Published
- 2021
- Full Text
- View/download PDF
39. Insight into the inhibitor discrimination by FLT3 F691L.
- Author
-
Verma, Sharad, Singh, Aditi, Kumari, Anchala, Pandey, Bharati, Jamal, Salma, Goyal, Sukriti, Sinha, Siddharth, and Grover, Abhinav
- Subjects
- *
PROTEIN-tyrosine kinases , *HEMATOPOIETIC stem cells , *GENETIC mutation , *BINDING energy , *FREE energy (Thermodynamics) - Abstract
Fms‐like tyrosine kinase 3 (FLT3) belongs to the receptor tyrosine kinase family and expressed in hematopoietic progenitor cells. FLT3 gene mutations are reported in ~30% of acute myeloid leukemia cases. FLT3 kinase domain mutation F691L is one of the common causes of acquired resistance to the FLT3 inhibitors including quizartinib. MZH29 and crenolanib were previously reported to inhibit FLT3 F691L. However, crenolanib was reported for the moderate inhibition. We found that Glu661and Asp829 were the most significant residues to target the FLT3 F691L which contribute most significantly to the binding energy with MZH29 and crenolanib. These interactions were found absent with quizartinib. Further free energy landscape analysis revealed that FLT3 F691L bound to MZH29 and crenolanib was more stable as compared to quizartinib. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
40. Integrated Molecular Characterization of Gastrointestinal Stromal Tumors (GIST) Harboring the Rare D842V Mutation in PDGFRA Gene.
- Author
-
Indio, Valentina, Astolfi, Annalisa, Tarantino, Giuseppe, Urbini, Milena, Patterson, Janice, Nannini, Margherita, Saponara, Maristella, Gatto, Lidia, Santini, Donatella, do Valle, Italo F., Castellani, Gastone, Remondini, Daniel, Fiorentino, Michelangelo, von Mehren, Margaret, Brandi, Giovanni, Biasco, Guido, Heinrich, Michael C., and Pantaleo, Maria Abbondanza
- Subjects
- *
GASTROINTESTINAL stromal tumors , *GENETIC mutation , *PLATELET-derived growth factor , *MUTANT proteins , *CELL cycle - Abstract
Gastrointestinal stromal tumors (GIST) carrying the D842V activating mutation in the platelet-derived growth factor receptor alpha (PDGFRA) gene are a very rare subgroup of GIST (about 10%) known to be resistant to conventional tyrosine kinase inhibitors (TKIs) and to show an indolent behavior. In this study, we performed an integrated molecular characterization of D842V mutant GIST by whole-transcriptome and whole-exome sequencing coupled with protein-ligand interaction modelling to identify the molecular signature and any additional recurrent genomic event related to their clinical course. We found a very specific gene expression profile of D842V mutant tumors showing the activation of G-protein-coupled receptor (GPCR) signaling and a relative downregulation of cell cycle processes. Beyond D842V, no recurrently mutated genes were found in our cohort. Nevertheless, many private, clinically relevant alterations were found in each tumor (TP53, IDH1, FBXW7, SDH-complex). Molecular modeling of PDGFRA D842V suggests that the mutant protein binds imatinib with lower affinity with respect to wild-type structure, showing higher stability during the interaction with other type I TKIs (like crenolanib). D842V mutant GIST do not show any actionable recurrent molecular events of therapeutic significance, therefore this study supports the rationale of novel TKIs development that are currently being evaluated in clinical studies for the treatment of D842V mutant GIST. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
41. Stromal Platelet–Derived Growth Factor Receptor-β Signaling Promotes Breast Cancer Metastasis in the Brain
- Author
-
Luke Russell, Sarah A. Steck, Bhuvaneswari Ramaswamy, Daniel G. Stover, Jose Otero, Zaibo Li, Robert Pilarski, Jonathan M. Spehar, Jonathan P. Godbout, Raleigh D. Kladney, Michael C. Ostrowski, Gustavo Leone, Gina M. Sizemore, Steven T. Sizemore, Arnab Chakravarti, Maria C. Cuitiño, Blake E. Hildreth, Christopher Koivisto, Anisha M. Hammer, Lynn Schoenfield, Manjusri Das, Katie A. Thies, Anthony J. Trimboli, Jennifer A. Geisler, Chelsea Bolyard, Matthew D. Ringel, Balveen Kaur, and Jerome F. Bey
- Subjects
0301 basic medicine ,Cancer Research ,Stromal cell ,Breast Neoplasms ,Metastasis ,Receptor, Platelet-Derived Growth Factor beta ,Mice ,03 medical and health sciences ,Paracrine signalling ,chemistry.chemical_compound ,0302 clinical medicine ,Animals ,Humans ,Medicine ,Mammary tumor ,PDGFB ,biology ,business.industry ,Brain ,Endothelial Cells ,medicine.disease ,Primary tumor ,MicroRNAs ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,business ,Platelet-derived growth factor receptor ,Crenolanib - Abstract
Platelet-derived growth factor receptor-beta (PDGFRβ) is a receptor tyrosine kinase found in cells of mesenchymal origin such as fibroblasts and pericytes. Activation of this receptor is dependent on paracrine ligand induction, and its preferred ligand PDGFB is released by neighboring epithelial and endothelial cells. While expression of both PDGFRβ and PDGFB has been noted in patient breast tumors for decades, how PDGFB-to-PDGFRβ tumor–stroma signaling mediates breast cancer initiation, progression, and metastasis remains unclear. Here we demonstrate this paracrine signaling pathway that mediates both primary tumor growth and metastasis, specifically, metastasis to the brain. Elevated levels of PDGFB accelerated orthotopic tumor growth and intracranial growth of mammary tumor cells, while mesenchymal-specific expression of an activating mutant PDGFRβ (PDGFRβD849V) exerted proproliferative signals on adjacent mammary tumor cells. Stromal expression of PDGFRβD849V also promoted brain metastases of mammary tumor cells expressing high PDGFB when injected intravenously. In the brain, expression of PDGFRβD849V was observed within a subset of astrocytes, and aged mice expressing PDGFRβD849V exhibited reactive gliosis. Importantly, the PDGFR-specific inhibitor crenolanib significantly reduced intracranial growth of mammary tumor cells. In a tissue microarray comprised of 363 primary human breast tumors, high PDGFB protein expression was prognostic for brain metastases, but not metastases to other sites. Our results advocate the use of mice expressing PDGFRβD849V in their stromal cells as a preclinical model of breast cancer–associated brain metastases and support continued investigation into the clinical prognostic and therapeutic use of PDGFB-to-PDGFRβ signaling in women with breast cancer. Significance: These studies reveal a previously unknown role for PDGFB-to-PDGFRβ paracrine signaling in the promotion of breast cancer brain metastases and support the prognostic and therapeutic clinical utility of this pathway for patients. See related article by Wyss and colleagues, p. 594
- Published
- 2021
42. A genome-wide CRISPR screen identifies regulators of MAPK and MTOR pathways that mediate resistance to sorafenib in acute myeloid leukemia
- Author
-
Christopher A. Eide, Stephen E. Kurtz, Shannon K. McWeeney, Tamilla Nechiporuk, Daniel Bottomly, Alisa Damnernsawad, and Jeffrey W. Tyner
- Subjects
0301 basic medicine ,Sorafenib ,MAPK/ERK pathway ,Drug resistance ,mTORC1 ,03 medical and health sciences ,chemistry.chemical_compound ,fluids and secretions ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,neoplasms ,PI3K/AKT/mTOR pathway ,Quizartinib ,business.industry ,Myeloid leukemia ,hemic and immune systems ,Hematology ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,embryonic structures ,Cancer research ,business ,medicine.drug ,Crenolanib - Abstract
Drug resistance impedes the long-term effect of targeted therapies in acute myeloid leukemia (AML), necessitating the identification of mechanisms underlying resistance. Approximately 25% of AML patients carry FLT3 mutations and develop post-treatment insensitivity to FLT3 inhibitors, including sorafenib. Using a genomewide CRISPR screen, we identified LZTR1, NF1, TSC1 and TSC2, negative regulators of the MAPK and MTOR pathways, as mediators of resistance to sorafenib. Analyses of ex vivo drug sensitivity assays in samples from patients with FLT3-ITD AML revealed that lower expression of LZTR1, NF1, and TSC2 correlated with sensitivity to sorafenib. Importantly, MAPK and/or MTOR complex 1 (MTORC1) activity was upregulated in AML cells made resistant to several FLT3 inhibitors, including crenolanib, quizartinib, and sorafenib. These cells were sensitive to MEK inhibitors, and the combination of FLT3 and MEK inhibitors showed enhanced efficacy, suggesting the effectiveness of such treatment in AML patients with FLT3 mutations and those with resistance to FLT3 inhibitors.
- Published
- 2020
43. Succinate dehydrogenase deficiency in a PDGFRA mutated GIST.
- Author
-
Belinsky, Martin G., Cai, Kathy Q., Yan Zhou, Biao Luo, Jianming Pei, Rink, Lori, von Mehren, Margaret, Zhou, Yan, Luo, Biao, and Pei, Jianming
- Subjects
- *
SUCCINATE dehydrogenase , *GASTROINTESTINAL stromal tumors , *PLATELET-derived growth factor receptors , *IMATINIB , *ANTINEOPLASTIC agents , *PROTEIN-tyrosine kinases , *IMMUNOHISTOCHEMISTRY - Abstract
Background: Most gastrointestinal stromal tumors (GISTs) harbor mutually exclusive gain of function mutations in the receptor tyrosine kinase (RTK) KIT (70-80%) or in the related receptor PDGFRA (~10%). These GISTs generally respond well to therapy with the RTK inhibitor imatinib mesylate (IM), although initial response is genotype-dependent. An alternate mechanism leading to GIST oncogenesis is deficiency in the succinate dehydrogenase (SDH) enzyme complex resulting from genetic or epigenetic inactivation of one of the four SDH subunit genes (SDHA, SDHB, SDHC, SDHD, collectively referred to as SDHX). SDH loss of function is generally seen only in GIST lacking RTK mutations, and SDH-deficient GIST respond poorly to imatinib therapy.Methods: Tumor and normal DNA from a GIST case carrying the IM-resistant PDGFRA D842V mutation was analyzed by whole exome sequencing (WES) to identify additional potential targets for therapy. The tumors analyzed were separate recurrences following progression on imatinib, sunitinib, and the experimental PDGFRA inhibitor crenolanib. Tumor sections from the GIST case and a panel of ~75 additional GISTs were subjected to immunohistochemistry (IHC) for the SDHB subunit.Results: Surprisingly, a somatic, loss of function mutation in exon 4 of the SDHB subunit gene (c.291_292delCT, p.I97Mfs*21) was identified in both tumors. Sanger sequencing confirmed the presence of this inactivating mutation, and IHC for the SDHB subunit demonstrated that these tumors were SDH-deficient. IHC for the SDHB subunit across a panel of ~75 GIST cases failed to detect SDH deficiency in other GISTs with RTK mutations.Conclusions: This is the first reported case of a PDGFRA mutant GIST exhibiting SDH-deficiency. A brief discussion of the relevant GIST literature is included. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
44. Acute myeloid leukemia transformed to a targetable disease
- Author
-
Hampig Raphael Kourie, Khalil Saleh, and Nadine Khalifeh-Saleh
- Subjects
0301 basic medicine ,Cancer Research ,Gemtuzumab ozogamicin ,CD33 ,Enasidenib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Midostaurin ,Clinical Trials as Topic ,Acute leukemia ,business.industry ,Disease Management ,Myeloid leukemia ,General Medicine ,Leukemia, Myeloid, Acute ,Treatment Outcome ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cytarabine ,Cancer research ,Disease Susceptibility ,business ,Crenolanib ,medicine.drug - Abstract
Acute myeloid leukemia (AML) is a heterogeneous neoplasm characterized by the monoclonal proliferation of immature progenitors. It is the most common acute leukemia in adults and its incidence increases with age. The standard traditional treatment in fit patients was the ‘3 + 7’ regimen and cytarabine consolidation followed or not with allogeneic stem cell transplantation. Recently, several targeted therapies such as gemtuzumab ozogamicin targeting the CD33+ AML, midostaurin, gilteritinib and crenolanib inhibiting FLT3-positive AML and ivosidenib and enasidenib blocking IDH-mutated AML have been approved. These new drugs led to the change of the landscape of the treatment of AML and transforming this disease to a targetable one. We aimed in this paper to review the implications of each new target, the mechanisms of action of these new drugs and we discuss all the studies leading to the approval of these new drugs in their indications according to each target.
- Published
- 2020
45. Effects of the multi‐kinase inhibitor midostaurin in combination with chemotherapy in models of acute myeloid leukaemia
- Author
-
Sara J. Buhrlage, Xiaoxi Liu, Jing Yang, Jinhua Wang, Abigail E Case, Sophia Adamia, Richard Stone, Chengcheng Meng, Prafulla C. Gokhale, Nathanael S. Gray, Ellen Weisberg, James D. Griffin, Hong L. Tiv, and Martin Sattler
- Subjects
0301 basic medicine ,medicine.medical_treatment ,synergy ,Syk ,Apoptosis ,Mice ,chemistry.chemical_compound ,fluids and secretions ,0302 clinical medicine ,Piperidines ,hemic and lymphatic diseases ,SYK ,Midostaurin ,Aniline Compounds ,Drug Synergism ,hemic and immune systems ,Sorafenib ,crenolanib ,3. Good health ,Gene Expression Regulation, Neoplastic ,Leukemia, Myeloid, Acute ,midostaurin ,Pyrazines ,030220 oncology & carcinogenesis ,embryonic structures ,Molecular Medicine ,Original Article ,gilteritinib ,FLT3 Inhibitor ,medicine.drug ,Crenolanib ,Antineoplastic Agents ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Syk Kinase ,Chemosensitizing agent ,acute myeloid leukaemia ,Benzothiazoles ,Protein Kinase Inhibitors ,Cell Proliferation ,Quizartinib ,Chemotherapy ,non‐mutant FLT3 ,business.industry ,Phenylurea Compounds ,Original Articles ,Cell Biology ,Staurosporine ,quizartinib ,030104 developmental biology ,fms-Like Tyrosine Kinase 3 ,chemistry ,Mutation ,Cancer research ,Benzimidazoles ,business - Abstract
Recently, several targeted agents have been developed for specific subsets of patients with acute myeloid leukaemia (AML), including midostaurin, the first FDA‐approved FLT3 inhibitor for newly diagnosed patients with FLT3 mutations. However, in the initial Phase I/II clinical trials, some patients without FLT3 mutations had transient responses to midostaurin, suggesting that this multi‐targeted kinase inhibitor might benefit AML patients more broadly. Here, we demonstrate submicromolar efficacy of midostaurin in vitro and efficacy in vivo against wild‐type (wt) FLT3‐expressing AML cell lines and primary cells, and we compare its effectiveness with that of other FLT3 inhibitors currently in clinical trials. Midostaurin was found to synergize with standard chemotherapeutic drugs and some targeted agents against AML cells without mutations in FLT3. The mechanism may involve, in part, the unique kinase profile of midostaurin that includes proteins implicated in AML transformation, such as SYK or KIT, or inhibition of ERK pathway or proviability signalling. Our findings support further investigation of midostaurin as a chemosensitizing agent in AML patients without FLT3 mutations.
- Published
- 2020
46. FLT3 inhibitors in acute myeloid leukemia: ten frequently asked questions
- Author
-
Ahmad Antar, Ali Bazarbachi, Zaher K Otrock, Elias Jabbour, and Mohamad Mohty
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,chemistry.chemical_compound ,fluids and secretions ,0302 clinical medicine ,Piperidines ,hemic and lymphatic diseases ,Midostaurin ,Enzyme Inhibitors ,Randomized Controlled Trials as Topic ,Aniline Compounds ,Lestaurtinib ,Myeloid leukemia ,hemic and immune systems ,Hematology ,Sorafenib ,Prognosis ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,Pyrazines ,030220 oncology & carcinogenesis ,embryonic structures ,medicine.drug ,Crenolanib ,medicine.medical_specialty ,Carbazoles ,Antineoplastic Agents ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,Benzothiazoles ,Furans ,Quizartinib ,business.industry ,Phenylurea Compounds ,DNA Methylation ,Staurosporine ,medicine.disease ,Transplantation ,030104 developmental biology ,fms-Like Tyrosine Kinase 3 ,chemistry ,Mutation ,Benzimidazoles ,Neoplasm Recurrence, Local ,business - Abstract
The FMS-like tyrosine kinase 3 (FLT3) gene is mutated in approximately one third of patients with acute myeloid leukemia (AML), either by internal tandem duplications (FLT3-ITD), or by a point mutation mainly involving the tyrosine kinase domain (FLT3-TKD). Patients with FLT3-ITD have a high risk of relapse and low cure rates. Several FLT3 tyrosine kinase inhibitors have been developed in the last few years with variable kinase inhibitory properties, pharmacokinetics, and toxicity profiles. FLT3 inhibitors are divided into first generation multi-kinase inhibitors (such as sorafenib, lestaurtinib, midostaurin) and next generation inhibitors (such as quizartinib, crenolanib, gilteritinib) based on their potency and specificity of FLT3 inhibition. These diverse FLT3 inhibitors have been evaluated in myriad clinical trials as monotherapy or in combination with conventional chemotherapy or hypomethylating agents and in various settings, including front-line, relapsed or refractory disease, and maintenance therapy after consolidation chemotherapy or allogeneic stem cell transplantation. In this practical question-and-answer-based review, the main issues faced by the leukemia specialists on the use of FLT3 inhibitors in AML are addressed.
- Published
- 2020
47. Phase I study using crenolanib to target PDGFR kinase in children and young adults with newly diagnosed DIPG or recurrent high-grade glioma, including DIPG
- Author
-
Christopher L Tinkle, Alberto Broniscer, Jason Chiang, Olivia Campagne, Jie Huang, Brent A Orr, Xiaoyu Li, Zoltan Patay, Jinghui Zhang, Suzanne J Baker, Thomas E Merchant, Vinay Jain, Arzu Onar-Thomas, Clinton F Stewart, Cynthia Wetmore, and Amar Gajjar
- Subjects
diffuse midline glioma ,Oncology ,H3 K27M ,Clinical Investigations ,pediatric high-grade glioma ,AcademicSubjects/MED00300 ,diffuse intrinsic pontine glioma ,Surgery ,AcademicSubjects/MED00310 ,Neurology (clinical) ,crenolanib ,phase I clinical trial - Abstract
BackgroundPlatelet-derived growth factor receptor (PDGFR) signaling has been directly implicated in pediatric high-grade gliomagenesis. This study evaluated the safety and tolerability of crenolanib, a potent, selective inhibitor of PDGFR-mediated phosphorylation, in pediatric patients with high-grade glioma (HGG).MethodsWe used a rolling-6 design to study the maximum tolerated dose (MTD) of once-daily crenolanib administered during and after focal radiation therapy in children with newly diagnosed diffuse intrinsic pontine glioma (DIPG) (stratum A) or with recurrent/progressive HGG (stratum B). Pharmacokinetics were studied during the first cycle at the first dose and at steady state (day 28). Alterations in PDGFRA were assessed by Sanger or exome sequencing and interphase fluorescence in situ hybridization or single nucleotide polymorphism arrays.ResultsFifty evaluable patients were enrolled in the 2 strata, and an MTD of 170 mg/m2 was established for both. Dose-limiting toxicities were primarily liver enzyme elevations and hematologic count suppression in both strata. Crenolanib AUC0–48h and CMAX did not differ significantly for crushed versus whole-tablet administration. Overall, PDGFRA alterations were observed in 25% and 30% of patients in stratum A and B, respectively. Neither crenolanib therapy duration nor survival outcomes differed significantly by PDGFRA status, and overall survival of stratum A was similar to that of historical controls.ConclusionsChildren tolerate crenolanib well at doses slightly higher than the established MTD in adults, with a toxicity spectrum generally similar to that in adults. Studies evaluating intratumoral PDGFR pathway inhibition in biomarker-enriched patients are needed to evaluate further the clinical utility of crenolanib in this population.
- Published
- 2021
48. Integrated Molecular Characterization of Gastrointestinal Stromal Tumors (GIST) Harboring the Rare D842V Mutation in PDGFRA Gene
- Author
-
Valentina Indio, Annalisa Astolfi, Giuseppe Tarantino, Milena Urbini, Janice Patterson, Margherita Nannini, Maristella Saponara, Lidia Gatto, Donatella Santini, Italo F. do Valle, Gastone Castellani, Daniel Remondini, Michelangelo Fiorentino, Margaret von Mehren, Giovanni Brandi, Guido Biasco, Michael C. Heinrich, and Maria Abbondanza Pantaleo
- Subjects
gastrointestinal stromal tumors ,D842V ,GIST ,KIT ,PDGFRA ,crenolanib ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Gastrointestinal stromal tumors (GIST) carrying the D842V activating mutation in the platelet-derived growth factor receptor alpha (PDGFRA) gene are a very rare subgroup of GIST (about 10%) known to be resistant to conventional tyrosine kinase inhibitors (TKIs) and to show an indolent behavior. In this study, we performed an integrated molecular characterization of D842V mutant GIST by whole-transcriptome and whole-exome sequencing coupled with protein–ligand interaction modelling to identify the molecular signature and any additional recurrent genomic event related to their clinical course. We found a very specific gene expression profile of D842V mutant tumors showing the activation of G-protein-coupled receptor (GPCR) signaling and a relative downregulation of cell cycle processes. Beyond D842V, no recurrently mutated genes were found in our cohort. Nevertheless, many private, clinically relevant alterations were found in each tumor (TP53, IDH1, FBXW7, SDH-complex). Molecular modeling of PDGFRA D842V suggests that the mutant protein binds imatinib with lower affinity with respect to wild-type structure, showing higher stability during the interaction with other type I TKIs (like crenolanib). D842V mutant GIST do not show any actionable recurrent molecular events of therapeutic significance, therefore this study supports the rationale of novel TKIs development that are currently being evaluated in clinical studies for the treatment of D842V mutant GIST.
- Published
- 2018
- Full Text
- View/download PDF
49. RhoGDI1-Cdc42 Signaling Is Required for PDGF-BB-Induced Phenotypic Transformation of Vascular Smooth Muscle Cells and Neointima Formation
- Author
-
Jingwen Sun, Haijing Guan, Xiuying Liang, Yan Qi, and Wenjuan Yao
- Subjects
Neointima ,Vascular smooth muscle ,QH301-705.5 ,Medicine (miscellaneous) ,macromolecular substances ,CDC42 ,Article ,General Biochemistry, Genetics and Molecular Biology ,VSMC phenotypic transformation ,chemistry.chemical_compound ,MG132 ,Rho GTPases ,Biology (General) ,Gene knockdown ,biology ,Endoplasmic reticulum ,musculoskeletal system ,RhoGDI ,Cell biology ,chemistry ,biology.protein ,cardiovascular system ,tissues ,intimal hyperplasia ,Platelet-derived growth factor receptor ,Crenolanib - Abstract
RhoGTPase is involved in PDGF-BB-mediated VSMC phenotypic modulation. RhoGDIs are key factors in regulating RhoGTPase activation. In the present study, we investigated the regulatory effect of RhoGDI1 on the activation of RhoGTPase in VSMC transformation and neointima formation. Western blot and co-immunoprecipitation assays showed that the PDGF receptor inhibition by crenolanib promoted RhoGDI1 polyubiquitination and degradation. Inhibition of RhoGDI1 degradation via MG132 reversed the decrease in VSMC phenotypic transformation. In addition, RhoGDI1 knockdown significantly inhibited VSMC phenotypic transformation and neointima formation in vitro and in vivo. These results suggest that PDGF-BB promotes RhoGDI1 stability via the PDGF receptor and induces the VSMC synthetic phenotype. The co-immunoprecipitation assay showed that PDGF-BB enhanced the interaction of RhoGDI1 with Cdc42 and promoted the activation of Cdc42, these enhancements were blocked by crenolanib and RhoGDI1 knockdown. Moreover, RhoGDI1 knockdown and crenolanib pretreatment prevented the localization of Cdc42 to the plasma membrane (PM) to activate and improve the accumulation of Cdc42 on endoplasmic reticulum (ER). Furthermore, Cdc42 inhibition or suppression significantly reduced VSMC phenotypic transformation and neointima formation in vitro and in vivo. This study revealed the novel mechanism by which RhoGDI1 stability promotes the RhoGDI1-Cdc42 interaction and Cdc42 activation, thereby affecting VSMC phenotypic transformation and neointima formation.
- Published
- 2021
- Full Text
- View/download PDF
50. How acute myeloid leukemia (AML) escapes from FMS-related tyrosine kinase 3 (FLT3) inhibitors? Still an overrated complication?
- Author
-
Perrone S, Ottone T, Zhdanovskaya N, and Molica M
- Abstract
FMS-related tyrosine kinase 3 (FLT3) mutations, present in about 25%-30% of acute myeloid leukemia (AML) patients, constitute one of the most frequently detected mutations in these patients. The binding of FLT3L to FLT3 activates the phosphatidylinositol 3-kinase (PI3K) and RAS pathways, producing increased cell proliferation and the inhibition of apoptosis. Two types of FLT3 mutations exist: FLT3-ITD and FLT3-TKD (point mutations in D835 and I836 or deletion of codon I836). A class of drugs, tyrosine-kinase inhibitors (TKI), targeting mutated FLT3, is already available with 1
st and 2nd generation molecules, but only midostaurin and gilteritinib are currently approved. However, the emergence of resistance or the selection of clones not responding to FLT3 inhibitors has become an important clinical dilemma, as the duration of clinical responses is generally limited to a few months. This review analyzes the insights into mechanisms of resistance to TKI and poses a particular view on the clinical relevance of this phenomenon. Has resistance been overlooked? Indeed, FLT3 inhibitors have significantly contributed to reducing the negative impact of FLT3 mutations on the prognosis of AML patients who are no longer considered at high risk by the European LeukemiaNet (ELN) 2022. Finally, several ongoing efforts to overcome resistance to FLT3-inhibitors will be presented: new generation FLT3 inhibitors in monotherapy or combined with standard chemotherapy, hypomethylating drugs, or IDH1/2 inhibitors, Bcl2 inhibitors; novel anti-human FLT3 monoclonal antibodies (e.g., FLT3/CD3 bispecific antibodies); FLT3-CAR T-cells; CDK4/6 kinase inhibitor (e.g., palbociclib)., Competing Interests: All authors declared that there are no conflicts of interest., (© The Author(s) 2023.)- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.