Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
JCI Insight ; 9(8)2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38646934

ABSTRACT

Acute myeloid leukemia (AML) is a fatal disease characterized by the accumulation of undifferentiated myeloblasts, and agents that promote differentiation have been effective in this disease but are not curative. Dihydroorotate dehydrogenase inhibitors (DHODHi) have the ability to promote AML differentiation and target aberrant malignant myelopoiesis. We introduce HOSU-53, a DHODHi with significant monotherapy activity, which is further enhanced when combined with other standard-of-care therapeutics. We further discovered that DHODHi modulated surface expression of CD38 and CD47, prompting the evaluation of HOSU-53 combined with anti-CD38 and anti-CD47 therapies, where we identified a compelling curative potential in an aggressive AML model with CD47 targeting. Finally, we explored using plasma dihydroorotate (DHO) levels to monitor HOSU-53 safety and found that the level of DHO accumulation could predict HOSU-53 intolerability, suggesting the clinical use of plasma DHO to determine safe DHODHi doses. Collectively, our data support the clinical translation of HOSU-53 in AML, particularly to augment immune therapies. Potent DHODHi to date have been limited by their therapeutic index; however, we introduce pharmacodynamic monitoring to predict tolerability while preserving antitumor activity. We additionally suggest that DHODHi is effective at lower doses with select immune therapies, widening the therapeutic index.


Subject(s)
Leukemia, Myeloid, Acute , Pyrimidines , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/immunology , Humans , Pyrimidines/therapeutic use , Mice , Animals , Dihydroorotate Dehydrogenase , Immunotherapy/methods , Cell Line, Tumor , Xenograft Model Antitumor Assays , Female
3.
Blood Adv ; 6(17): 5049-5060, 2022 09 13.
Article in English | MEDLINE | ID: mdl-35797240

ABSTRACT

Despite the clinical benefit associated with gilteritinib in relapsed/refractory acute myeloid leukemia (AML), most patients eventually develop resistance through unknown mechanisms. To delineate the mechanistic basis of resistance to gilteritinib, we performed targeted sequencing and scRNASeq on primary FLT3-ITD-mutated AML samples. Co-occurring mutations in RAS pathway genes were the most common genetic abnormalities, and unresponsiveness to gilteritinib was associated with increased expression of bone marrow-derived hematopoietic cytokines and chemokines. In particular, we found elevated expression of the TEK-family kinase, BMX, in gilteritinib-unresponsive patients pre- and post-treatment. BMX contributed to gilteritinib resistance in FLT3-mutant cell lines in a hypoxia-dependent manner by promoting pSTAT5 signaling, and these phenotypes could be reversed with pharmacological inhibition and genetic knockout. We also observed that inhibition of BMX in primary FLT3-mutated AML samples decreased chemokine secretion and enhanced the activity of gilteritinib. Collectively, these findings indicate a crucial role for microenvironment-mediated factors modulated by BMX in the escape from targeted therapy and have implications for the development of novel therapeutic interventions to restore sensitivity to gilteritinib.


Subject(s)
Aniline Compounds , Leukemia, Myeloid, Acute , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mutation , Protein-Tyrosine Kinases/genetics , Pyrazines/pharmacology , Pyrazines/therapeutic use , Tumor Microenvironment , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/therapeutic use
4.
JCI Insight ; 5(23)2020 12 03.
Article in English | MEDLINE | ID: mdl-33268594

ABSTRACT

Effective treatment for AML is challenging due to the presence of clonal heterogeneity and the evolution of polyclonal drug resistance. Here, we report that TP-0903 has potent activity against protein kinases related to STAT, AKT, and ERK signaling, as well as cell cycle regulators in biochemical and cellular assays. In vitro and in vivo, TP-0903 was active in multiple models of drug-resistant FLT3 mutant AML, including those involving the F691L gatekeeper mutation and bone marrow microenvironment-mediated factors. Furthermore, TP-0903 demonstrated preclinical activity in AML models with FLT3-ITD and common co-occurring mutations in IDH2 and NRAS genes. We also showed that TP-0903 had ex vivo activity in primary AML cells with recurrent mutations including MLL-PTD, ASXL1, SRSF2, and WT1, which are associated with poor prognosis or promote clinical resistance to AML-directed therapies. Our preclinical studies demonstrate that TP-0903 is a multikinase inhibitor with potent activity against multiple drug-resistant models of AML that will have an immediate clinical impact in a heterogeneous disease like AML.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Gene Duplication/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Male , Mice , Mice, Nude , Mutation/drug effects , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Pyrimidines/metabolism , Sulfonamides/metabolism , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
5.
J Hematol Oncol ; 13(1): 8, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31992353

ABSTRACT

BACKGROUND: Acute myeloid leukemia (AML) is the most common type of adult leukemia. Several studies have demonstrated that oncogenesis in AML is enhanced by kinase signaling pathways such as Src family kinases (SFK) including Src and Lyn, spleen tyrosine kinase (SYK), and bruton's tyrosine kinase (BTK). Recently, the multi-kinase inhibitor ArQule 531 (ARQ 531) has demonstrated potent inhibition of SFK and BTK that translated to improved pre-clinical in vivo activity as compared with the irreversible BTK inhibitor ibrutinib in chronic lymphocytic leukemia (CLL) models. Given the superior activity of ARQ 531 in CLL, and recognition that this molecule has a broad kinase inhibition profile, we pursued its application in pre-clinical models of AML. METHODS: The potency of ARQ 531 was examined in vitro using FLT3 wild type and mutated (ITD) AML cell lines and primary samples. The modulation of pro-survival kinases following ARQ 531 treatment was determined using AML cell lines. The effect of SYK expression on ARQ 531 potency was evaluated using a SYK overexpressing cell line (Ba/F3 murine cells) constitutively expressing FLT3-ITD. Finally, the in vivo activity of ARQ 531 was evaluated using MOLM-13 disseminated xenograft model. RESULTS: Our data demonstrate that ARQ 531 treatment has anti-proliferative activity in vitro and impairs colony formation in AML cell lines and primary AML cells independent of the presence of a FLT3 ITD mutation. We demonstrate decreased phosphorylation of oncogenic kinases targeted by ARQ 531, including SFK (Tyr416), BTK, and fms-related tyrosine kinase 3 (FLT3), ultimately leading to changes in down-stream targets including SYK, STAT5a, and ERK1/2. Based upon in vitro drug synergy data, we examined ARQ 531 in the MOLM-13 AML xenograft model alone and in combination with venetoclax. Despite ARQ 531 having a less favorable pharmacokinetics profile in rodents, we demonstrate modest single agent in vivo activity and synergy with venetoclax. CONCLUSIONS: Our data support consideration of the application of ARQ 531 in combination trials for AML targeting higher drug concentrations in vivo.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism
6.
Clin Transl Sci ; 12(6): 641-647, 2019 11.
Article in English | MEDLINE | ID: mdl-31350825

ABSTRACT

Fms-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD) mutations, common in pediatric acute myeloid leukemia (AML), associate with early relapse and poor prognosis. Past studies have suggested additional cooperative mutations are required for leukemogenesis in FLT3-ITD+ AML. Using RNA sequencing and a next-generation targeted gene panel, we broadly characterize the co-occurring genomic alterations in pediatric cytogenetically normal (CN) FLT3-ITD+ AML to gain a deeper understanding of the clonal patterns and heterogeneity at diagnosis and relapse. We show that chimeric transcripts were present in 21 of 34 (62%) of de novo samples, 2 (6%) of these samples included a rare reoccurring fusion partner BCL11B. At diagnosis, the median number of mutations other than FLT3 per patient was 1 (range 0-3), which involved 8 gene pathways; WT1 and NPM1 mutations were frequently observed (35% and 24%, respectively). Fusion transcripts and high variant allele frequency (VAF) mutants, which included WT1, NPM1, SMARCA2, RAD21, and TYK2, were retained from diagnosis to relapse. We did observe reduction in VAF of simple or single mutation clones, but VAFs were preserved or expanded in more complex clones with multiple mutations. Our data provide the first insight into the genomic complexity of pediatric CN FLT3-ITD+ AML and could help stratify future targeted treatment strategies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Genetic Heterogeneity , Leukemia, Myeloid, Acute/genetics , Neoplasm Recurrence, Local/genetics , fms-Like Tyrosine Kinase 3/genetics , Adolescent , Child , Child, Preschool , Cytogenetic Analysis , Disease-Free Survival , Female , Gene Duplication , High-Throughput Nucleotide Sequencing , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Nucleophosmin , Precision Medicine , Prognosis , Randomized Controlled Trials as Topic , Remission Induction/methods , Sequence Analysis, RNA , Tandem Repeat Sequences/genetics , Young Adult
7.
Nat Commun ; 10(1): 2189, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31097698

ABSTRACT

Improvement in survival has been achieved for children and adolescents with AML but is largely attributed to enhanced supportive care as opposed to the development of better treatment regimens. High risk subtypes continue to have poor outcomes with event free survival rates <40% despite the use of high intensity chemotherapy in combination with hematopoietic stem cell transplant. Here we combine high-throughput screening, intracellular accumulation assays, and in vivo efficacy studies to identify therapeutic strategies for pediatric AML. We report therapeutics not currently used to treat AML, gemcitabine and cabazitaxel, have broad anti-leukemic activity across subtypes and are more effective relative to the AML standard of care, cytarabine, both in vitro and in vivo. JAK inhibitors are selective for acute megakaryoblastic leukemia and significantly prolong survival in multiple preclinical models. Our approach provides advances in the development of treatment strategies for pediatric AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , Janus Kinase Inhibitors/pharmacology , Leukemia, Experimental/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Adult , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/pathology , Bone Marrow/radiation effects , Bone Marrow Transplantation , Cell Line, Tumor , Child , Child, Preschool , Cytarabine/pharmacology , Cytarabine/therapeutic use , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Disease-Free Survival , Female , High-Throughput Screening Assays/methods , Humans , Infant , Janus Kinase Inhibitors/therapeutic use , Leukemia, Experimental/etiology , Leukemia, Experimental/mortality , Leukemia, Experimental/pathology , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Taxoids/pharmacology , Taxoids/therapeutic use , Whole-Body Irradiation/adverse effects , Xenograft Model Antitumor Assays , Young Adult , Gemcitabine
8.
Haematologica ; 102(8): 1391-1400, 2017 08.
Article in English | MEDLINE | ID: mdl-28473620

ABSTRACT

Long non-coding ribonucleic acids (RNAs) are a novel class of RNA molecules, which are increasingly recognized as important molecular players in solid and hematologic malignancies. Herein we investigated whether long non-coding RNA expression is associated with clinical and molecular features, as well as outcome of younger adults (aged <60 years) with de novo cytogenetically normal acute myeloid leukemia. Whole transcriptome profiling was performed in a training (n=263) and a validation set (n=114). Using the training set, we identified 24 long non-coding RNAs associated with event-free survival. Linear combination of the weighted expression values of these transcripts yielded a prognostic score. In the validation set, patients with high scores had shorter disease-free (P<0.001), overall (P=0.002) and event-free survival (P<0.001) than patients with low scores. In multivariable analyses, long non-coding RNA score status was an independent prognostic marker for disease-free (P=0.01) and event-free survival (P=0.002), and showed a trend for overall survival (P=0.06). Among multiple molecular alterations tested, which are prognostic in cytogenetically normal acute myeloid leukemia, only double CEBPA mutations, NPM1 mutations and FLT3-ITD associated with distinct long non-coding RNA signatures. Correlation of the long non-coding RNA scores with messenger RNA and microRNA expression identified enrichment of genes involved in lymphocyte/leukocyte activation, inflammation and apoptosis in patients with high scores. We conclude that long non-coding RNA profiling provides meaningful prognostic information in younger adults with cytogenetically normal acute myeloid leukemia. In addition, expression of prognostic long non-coding RNAs associates with oncogenic molecular pathways in this disease. clinicaltrials.gov Identifier: 00048958 (CALGB-8461), 00899223 (CALGB-9665), and 00900224 (CALGB-20202).


Subject(s)
Leukemia, Myeloid, Acute/genetics , RNA, Long Noncoding/analysis , Adult , Cytogenetic Analysis , Disease-Free Survival , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Middle Aged , Nucleophosmin , Prognosis , Supervised Machine Learning , Young Adult
9.
Cancer Chemother Pharmacol ; 77(6): 1231-43, 2016 06.
Article in English | MEDLINE | ID: mdl-27125973

ABSTRACT

PURPOSE: Investigate antileukemic activity of artemisinins, artesunate (ART), and dihydroartemisinin (DHA), in combination with cytarabine, a key component of acute myeloid leukemia (AML) chemotherapy using in vitro and in vivo models. METHODS: Using ten human AML cell lines, we conducted a high-throughput screen to identify antimalarial agents with antileukemic activity. We evaluated effects of ART and DHA on cell viability, cytotoxicity, apoptosis, lysosomal integrity, and combination effects with cytarabine in cell lines and primary patient blasts. In vivo pharmacokinetic studies and efficacy of single-agent ART or combination with cytarabine were evaluated in three xenograft models. RESULTS: ART and DHA had the most potent activity in a panel of AML cell lines, with selectivity toward samples harboring MLL rearrangements and FLT3-ITD mutations. Combination of ART or DHA was synergistic with cytarabine. Single-dose ART (120 mg/kg) produced human equivalent exposures, but multiple dose daily administration required for in vivo efficacy was not tolerated. Combination treatment produced initial regression, but did not prolong survival in vivo. CONCLUSIONS: The pharmacology of artemisinins is problematic and should be considered in designing AML treatment strategies with currently available agents. Artemisinins with improved pharmacokinetic properties may offer therapeutic benefit in combination with conventional therapeutic strategies in AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Artemisinins/therapeutic use , Cytarabine/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Apoptosis/drug effects , Artemisinins/administration & dosage , Artemisinins/adverse effects , Artemisinins/pharmacokinetics , Artesunate , Cell Line, Tumor , Cell Survival/drug effects , Cytarabine/administration & dosage , Cytarabine/adverse effects , Cytarabine/pharmacokinetics , High-Throughput Screening Assays , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Lysosomes/drug effects , Lysosomes/pathology , Mice, Inbred Strains , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
10.
Cancer Res ; 76(1): 117-26, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26677977

ABSTRACT

The use of multikinase inhibitors (MKI) in oncology, such as sorafenib, is associated with a cutaneous adverse event called hand-foot skin reaction (HFSR), in which sites of pressure or friction become inflamed and painful, thus significantly impacting quality of life. The pathogenesis of MKI-induced HFSR is unknown, and the only available treatment options involve dose reduction or discontinuation of therapy, which have negative effects on primary disease management. To investigate the underlying mechanisms by which sorafenib promotes keratinocyte cytotoxicity and subsequent HFSR induction, we performed a transporter-directed RNAi screen in human epidermal keratinocytes and identified SLC22A20 (OAT6) as an uptake carrier of sorafenib. Further investigations into the intracellular mechanism of sorafenib activity through in situ kinome profiling identified the mitogen-activated protein kinase MAP3K7 (TAK1) as a target of sorafenib that induces cell death. Finally, we demonstrate that sorafenib induced keratinocyte injury in vivo and that this effect could be reversed by cotreatment with the OAT6 inhibitor probenecid. Collectively, our findings reveal a novel pathway that regulates the entry of some MKIs into keratinocytes and explains the basis underlying sorafenib-induced skin toxicity, with important implications for the therapeutic management of HFSR.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Niacinamide/analogs & derivatives , Organic Anion Transporters/metabolism , Phenylurea Compounds/toxicity , Protein Kinase Inhibitors/toxicity , Skin Diseases/chemically induced , Animals , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Female , Hep G2 Cells , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Inbred C57BL , Niacinamide/pharmacokinetics , Niacinamide/toxicity , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , Organic Anion Transporters/genetics , Phenylurea Compounds/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Random Allocation , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin Diseases/metabolism , Skin Diseases/pathology , Sorafenib , Transfection
11.
Clin Cancer Res ; 19(6): 1458-66, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23340295

ABSTRACT

PURPOSE: Many tyrosine kinase inhibitors (TKI) undergo extensive hepatic metabolism, but mechanisms of their hepatocellular uptake remain poorly understood. We hypothesized that liver uptake of TKIs is mediated by the solute carriers OATP1B1 and OATP1B3. EXPERIMENTAL DESIGN: Transport of crizotinib, dasatinib, gefitinib, imatinib, nilotinib, pazopanib, sorafenib, sunitinib, vandetanib, and vemurafenib was studied in vitro using artificial membranes (PAMPA) and HEK293 cell lines stably transfected with OATP1B1, OATP1B3, or the ortholog mouse transporter, Oatp1b2. Pharmacokinetic studies were conducted with Oatp1b2-knockout mice and humanized OATP1B1- or OATP1B3-transgenic mice. RESULTS: All 10 TKIs were identified as substrates of OATP1B1, OATP1B3, or both. Transport of sorafenib was investigated further, as its diffusion was particularly low in the PAMPA assay (<4%) than other TKIs that were transported by both OATP1B1 and OATP1B3. Whereas Oatp1b2 deficiency in vivo had minimal influence on parent and active metabolite N-oxide drug exposure, plasma levels of the glucuronic acid metabolite of sorafenib (sorafenib-glucuronide) were increased more than 8-fold in Oatp1b2-knockout mice. This finding was unrelated to possible changes in intrinsic metabolic capacity for sorafenib-glucuronide formation in hepatic or intestinal microsomes ex vivo. Ensuing experiments revealed that sorafenib-glucuronide was itself a transported substrate of Oatp1b2 (17.5-fold vs. control), OATP1B1 (10.6-fold), and OATP1B3 (6.4-fold), and introduction of the human transporters in Oatp1b2-knockout mice provided partial restoration of function. CONCLUSIONS: These findings signify a unique role for OATP1B1 and OATP1B3 in the elimination of sorafenib-glucuronide and suggest a role for these transporters in the in vivo handling of glucuronic acid conjugates of drugs.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters/genetics , Phenylurea Compounds/pharmacokinetics , Animals , Glucuronic Acid/blood , HEK293 Cells , Humans , Liver-Specific Organic Anion Transporter 1 , Mice , Mice, Transgenic , Neoplasms/blood , Neoplasms/genetics , Niacinamide/administration & dosage , Niacinamide/pharmacokinetics , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Phenylurea Compounds/administration & dosage , Solute Carrier Organic Anion Transporter Family Member 1B3 , Sorafenib
12.
Clin Cancer Res ; 16(19): 4789-99, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20858838

ABSTRACT

PURPOSE: Carnitine is an essential cofactor for mitochondrial fatty acid oxidation that is actively reabsorbed by the luminal transporter Octn2 (Slc22a5). Because the nephrotoxic agent cisplatin causes urinary loss of carnitine in humans, we hypothesized that cisplatin may affect Octn2 function. EXPERIMENTAL DESIGN: Excretion of carnitine and acetylcarnitine was measured in urine collected from mice with or without cisplatin administration. The transport of carnitine was assessed in cells that were transfected with OCT1 or OCT2. The effect of cisplatin treatment on gene expression was analyzed using a mouse GeneChip array and validated using quantitative reverse transcriptase-PCR. RESULTS: In wild-type mice, urinary carnitine excretion at baseline was ∼3-fold higher than in mice lacking the basolateral cisplatin transporters Oct1 and Oct2 [Oct1/2(-/-) mice], indicating that carnitine itself undergoes basolateral uptake into the kidney. Transport of carnitine by OCT2, but not OCT1, was confirmed in transfected cells. We also found that cisplatin caused an increase in the urinary excretion of carnitine and acetylcarnitine in wild-type mice but not in Oct1/2(-/-) mice, suggesting that tubular transport of cisplatin is a prerequisite for this phenomenon. Cisplatin did not directly inhibit the transport of carnitine by Octn2 but downregulated multiple target genes of the transcription factor peroxisome proliferator activated receptor α, including Slc22a5, in the kidney of wild-type mice that were absent in Oct1/2(-/-) mice. CONCLUSION: Our study shows a pivotal role of Oct1 and Oct2 in cisplatin-related disturbances in carnitine homeostasis. We postulate that this phenomenon is triggered by deactivation of peroxisome proliferator activated receptor α and leads to deregulation of carnitine-shuttle genes.


Subject(s)
Carnitine/urine , Cisplatin/pharmacology , Down-Regulation/drug effects , Organic Cation Transport Proteins/antagonists & inhibitors , Acetylcarnitine/urine , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Line , Gene Expression Profiling , Humans , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout , Organic Cation Transport Proteins/deficiency , Organic Cation Transport Proteins/metabolism , PPAR alpha/antagonists & inhibitors , PPAR alpha/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Solute Carrier Family 22 Member 5 , Symporters
13.
Clin Cancer Res ; 14(10): 3141-8, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18483382

ABSTRACT

PURPOSE: The activity of imatinib in leukemia has recently been linked with expression of the organic cation transporter 1 (OCT1) gene SLC22A1. Here, we characterized the contribution of solute carriers to imatinib transport in an effort to further understand mechanisms involved in the intracellular uptake and retention (IUR) of the drug. EXPERIMENTAL DESIGN: IUR of [3H]imatinib was studied in Xenopus laevis oocytes and HEK293 cells expressing OATP1A2, OATP1B1, OATP1B3, OCT1-3, OCTN1-2, or OAT1-3. Gene expression was determined in nine leukemia cell lines using the Affymetrix U133 array. RESULTS: Imatinib was not found to be a substrate for OCT1 in oocytes (P = 0.21), whereas in HEK293 cells IUR was increased by only 1.20-fold relative to control cells (P = 0.002). Furthermore, in 74 cancer patients, the oral clearance of imatinib was not significantly altered in individuals carrying reduced-function variants in SLC22A1 (P = 0.99). Microarray analysis indicated that SLC22A1 was interrelated with gene expression of various transporters, including ABCB1, ABCC4, ABCG2 (negative), and OATP1A2 (positive). Imatinib was confirmed to be a substrate for the three efflux transporters (P < 0.05) as well as for OATP1A2 (P = 0.0001). CONCLUSIONS: This study suggests that SLC22A1 expression is a composite surrogate for expression of various transporters relevant to imatinib IUR. This observation provides a mechanistic explanation for previous studies that have linked SLC22A1 with the antitumor activity of imatinib. Because of its high expression in the intestine, ciliary body, gliomas, and leukemia cells, OATP1A2 may play a key role in imatinib pharmacokinetics-pharmacodynamics.


Subject(s)
Antineoplastic Agents/metabolism , Drug Resistance, Neoplasm/genetics , Gastrointestinal Stromal Tumors/genetics , Organic Cation Transporter 1/genetics , Piperazines/metabolism , Pyrimidines/metabolism , Animals , Benzamides , Cell Line, Tumor , Gastrointestinal Stromal Tumors/drug therapy , Gene Expression , Humans , Imatinib Mesylate , Organic Cation Transporter 1/metabolism , Polymerase Chain Reaction , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...