Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
EJNMMI Res ; 14(1): 54, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38869684

ABSTRACT

BACKGROUND: Neuroblastoma is the most common extra-cranial pediatric solid tumor. 131I-metaiodobenzylguanidine (MIBG) is a targeted radiopharmaceutical highly specific for neuroblastoma tumors, providing potent radiotherapy to widely metastatic disease. Aurora kinase A (AURKA) plays a role in mitosis and stabilization of the MYCN protein in neuroblastoma. We aimed to study the impact of AURKA inhibitors on DNA damage and tumor cell death in combination with 131I-MIBG therapy in a pre-clinical model of high-risk neuroblastoma. RESULTS: Using an in vivo model of high-risk neuroblastoma, we demonstrated a marked combinatorial effect of 131I-MIBG and alisertib on tumor growth. In MYCN amplified cell lines, the combination of radiation and an AURKA A inhibitor increased DNA damage and apoptosis and decreased MYCN protein levels. CONCLUSION: The combination of AURKA inhibition with 131I-MIBG treatment is active in resistant neuroblastoma models.

2.
Res Sq ; 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38313265

ABSTRACT

Background: Neuroblastoma is the most common extra-cranial pediatric solid tumor. 131I-metaiodobenzylguanidine (MIBG) is a targeted radiopharmaceutical highly specific for neuroblastoma tumors, providing potent radiotherapy to widely metastatic disease. Aurora kinase A (AURKA) plays a role in mitosis and stabilization of the MYCN protein in neuroblastoma. Here we explore whether AURKA inhibition potentiates a response to MIBG therapy. Results: Using an in vivo model of high-risk neuroblastoma, we demonstrated a marked combinatorial effect of 131I-MIBG and alisertib on tumor growth. In MYCN amplified cell lines, the combination of radiation and an AURKA A inhibitor increased DNA damage and apoptosis and decreased MYCN protein levels. Conclusion: The combination of AURKA inhibition with 131I-MIBG treatment is active in resistant neuroblastoma models and is a promising clinical approach in high-risk neuroblastoma.

3.
Pediatr Blood Cancer ; 70(6): e30335, 2023 06.
Article in English | MEDLINE | ID: mdl-37036306

ABSTRACT

BACKGROUND: Venetoclax is frequently used as salvage treatment in pediatric, adolescent, and young adult (AYA) patients with advanced hematologic malignancies. However, more data are needed from real-world studies to guide the safe and appropriate use of venetoclax in this population. PROCEDURE: We retrospectively reviewed the medical records of all patients diagnosed with hematologic malignancies less than 30 years of age treated with venetoclax outside of clinical trials at the University of California San Francisco Benioff Children's Hospitals from 2016 to 2022. RESULTS: We identified 13 patients (acute myeloid leukemia, n = 8; B-acute lymphoblastic leukemia, n = 3; myelodysplastic syndrome, n = 2) aged 4 months to 27 years. A median of 3 prior lines of therapy weregiven (range 0-5). All patients received venetoclax in combination with either a hypomethylating agent or conventional chemotherapy. Three (23%) patients achieved complete remission (CR); two (15%) achieved partial remission (PR); 3 (23%) had stable disease (SD), and five (42%) had progressive disease. Median survival and time to progression from venetoclax initiation was 9 months (range 2.5-52 months) and 3 months (range 2 weeks to 7.5 months), respectively. Six patients (46%) developed grade 3 or higher infections while receiving venetoclax, including bacteremia due to atypical organisms, invasive pulmonary infections with Aspergillus, cytomegalovirus (CMV) viremia, skin infections, and encephalitis with bacterial brain abscesses. CONCLUSIONS: Venetoclax in combination with hypomethylating agents or cytotoxic chemotherapy was effective in a subset of pediatric/AYA patients with advanced hematologic malignancies, but multiple severe infections were observed, particularly among patients who received venetoclax in combination with chemotherapy. Prospective studies will be required to determine the optimal dose and duration of venetoclax in this population.


Subject(s)
Hematologic Neoplasms , Leukemia, Myeloid, Acute , Adolescent , Young Adult , Humans , Child , Adult , Retrospective Studies , Prospective Studies , Bridged Bicyclo Compounds, Heterocyclic , Leukemia, Myeloid, Acute/drug therapy , Hematologic Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
4.
Haematologica ; 108(5): 1272-1283, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36700399

ABSTRACT

Hypodiploid acute lymphoblastic leukemia (ALL) is an aggressive blood cancer with a poor prognosis despite intensive chemotherapy or stem cell transplant. Children and adolescents with positive end-of-induction minimal residual disease have an overall survival lower than 30%. However, data regarding therapeutic alternatives for this disease is nearly nonexistent, emphasizing the critical need for new or adjunctive therapies that can improve outcomes. We previously reported on the therapeutic efficacy of venetoclax (ABT-199) in hypodiploid B-lineage ALL but with limitations as monotherapy. In this study, we set out to identify drugs enhancing the anti-leukemic effect of venetoclax in hypodiploid ALL. Using a highthroughput drug screen, we identified dinaciclib, a cyclin-dependent kinase inhibitor that worked synergistically with venetoclax to induce cell death in hypodiploid cell lines. This combination eradicated leukemic blasts within hypodiploid ALL patient-derived xenografts mice with low off-target toxicity. Our findings suggest that dual inhibition of BCL-2 (venetoclax) and CDK9/MCL-1 (dinaciclib) is a promising therapeutic approach in hypodiploid ALL, warranting further investigation to inform clinical trials in this high-risk patient population.


Subject(s)
Antineoplastic Agents , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Animals , Mice , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Cell Line, Tumor , Apoptosis , Proto-Oncogene Proteins c-bcl-2 , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Antineoplastic Agents/pharmacology
5.
Blood Adv ; 5(18): 3587-3591, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34464969

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is initiated in early childhood by somatic mutations that activate Ras signaling. Although some patients have only a single identifiable oncogenic mutation, others have 1 or more additional alterations. Such secondary mutations, as a group, are associated with an increased risk of relapse after hematopoietic stem cell transplantation or transformation to acute myeloid leukemia. These clinical observations suggest a cooperative effect between initiating and secondary mutations. However, the roles of specific genes in the prognosis or clinical presentation of JMML have not been described. In this study, we investigate the impact of secondary SH2B3 mutations in JMML. We find that patients with SH2B3 mutations have adverse outcomes, as well as higher white blood cell counts and hemoglobin F levels in the peripheral blood. We further demonstrate this interaction in genetically engineered mice. Deletion of Sh2b3 cooperates with conditional Nf1 deletion in a dose-dependent fashion. These studies illustrate that haploinsufficiency for Sh2b3 contributes to the severity of myeloproliferative disease and provide an experimental system for testing treatments for a high-risk cohort of JMML patients.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myelomonocytic, Juvenile , Animals , Child, Preschool , Humans , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/therapy , Mice , Mutation , Prognosis , Signal Transduction
7.
JCI Insight ; 5(21)2020 11 05.
Article in English | MEDLINE | ID: mdl-32990679

ABSTRACT

Somatic KRAS mutations are highly prevalent in many cancers. In addition, a distinct spectrum of germline KRAS mutations causes developmental disorders called RASopathies. The mutant proteins encoded by these germline KRAS mutations are less biochemically and functionally activated than those in cancer. We generated mice harboring conditional KrasLSL-P34Rand KrasLSL-T58I knock-in alleles and characterized the consequences of each mutation in vivo. Embryonic expression of KrasT58I resulted in craniofacial abnormalities reminiscent of those seen in RASopathy disorders, and these mice exhibited hyperplastic growth of multiple organs, modest alterations in cardiac valvulogenesis, myocardial hypertrophy, and myeloproliferation. By contrast, embryonic KrasP34R expression resulted in early perinatal lethality from respiratory failure due to defective lung sacculation, which was associated with aberrant ERK activity in lung epithelial cells. Somatic Mx1-Cre-mediated activation in the hematopoietic compartment showed that KrasP34R and KrasT58I expression had distinct signaling effects, despite causing a similar spectrum of hematologic diseases. These potentially novel strains are robust models for investigating the consequences of expressing endogenous levels of hyperactive K-Ras in different developing and adult tissues, for comparing how oncogenic and germline K-Ras proteins perturb signaling networks and cell fate decisions, and for performing preclinical therapeutic trials.


Subject(s)
Cardiomyopathies/pathology , Craniosynostoses/pathology , Hematologic Diseases/pathology , Lung Diseases/pathology , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Craniosynostoses/etiology , Craniosynostoses/metabolism , Female , Hematologic Diseases/etiology , Hematologic Diseases/metabolism , Lung Diseases/etiology , Lung Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Pregnancy
10.
Cancer Discov ; 9(6): 738-755, 2019 06.
Article in English | MEDLINE | ID: mdl-30952657

ABSTRACT

KRAS is the most frequently mutated oncogene. The incidence of specific KRAS alleles varies between cancers from different sites, but it is unclear whether allelic selection results from biological selection for specific mutant KRAS proteins. We used a cross-disciplinary approach to compare KRASG12D, a common mutant form, and KRASA146T, a mutant that occurs only in selected cancers. Biochemical and structural studies demonstrated that KRASA146T exhibits a marked extension of switch 1 away from the protein body and nucleotide binding site, which activates KRAS by promoting a high rate of intrinsic and guanine nucleotide exchange factor-induced nucleotide exchange. Using mice genetically engineered to express either allele, we found that KRASG12D and KRASA146T exhibit distinct tissue-specific effects on homeostasis that mirror mutational frequencies in human cancers. These tissue-specific phenotypes result from allele-specific signaling properties, demonstrating that context-dependent variations in signaling downstream of different KRAS mutants drive the KRAS mutational pattern seen in cancer. SIGNIFICANCE: Although epidemiologic and clinical studies have suggested allele-specific behaviors for KRAS, experimental evidence for allele-specific biological properties is limited. We combined structural biology, mass spectrometry, and mouse modeling to demonstrate that the selection for specific KRAS mutants in human cancers from different tissues is due to their distinct signaling properties.See related commentary by Hobbs and Der, p. 696.This article is highlighted in the In This Issue feature, p. 681.


Subject(s)
Alleles , Mutation , Oncogenes , Proto-Oncogene Proteins p21(ras)/genetics , Cell Transformation, Neoplastic/genetics , Humans , Models, Molecular , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Organ Specificity , Phenotype , Protein Conformation , Proteome , Proteomics/methods , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/metabolism , Structure-Activity Relationship
11.
Cancer Res ; 79(9): 2339-2351, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30862722

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the most common cancer in children. The highest rates of treatment failure occur in specific genetic subsets of ALL, including hypodiploid B-cell ALL (B-ALL), for which effective alternative therapies to current intensive chemotherapy treatments have yet to be developed. Here, we integrated biochemical and genomic profiling with functional drug assays to select effective agents with therapeutic potential against hypodiploid B-ALL. ABT-199, a selective Bcl-2 inhibitor, was effective in reducing leukemic burden in vitro and in vivo in patient-derived xenograft models of hypodiploid B-ALL. Daily oral treatment with ABT-199 significantly increased survival in xenografted mice. The unexpected efficacy of ABT-199 observed in hypodiploid leukemias lacking BIM expression (the major reported mediator of ABT-199-induced apoptosis) led us to investigate the mechanism of action of ABT-199 in the absence of BIM. Treatment with ABT-199 elicited responses in a dose-dependent manner, from cell-cycle arrest at low nanomolar concentrations to cell death at concentrations above 100 nmol/L. Collectively, these results demonstrate the efficacy of Bcl-2 inhibition and potential therapeutic strategy in hypodiploid B-ALL. SIGNIFICANCE: These results demonstrate the efficacy of ABT-199 in vivo and provide encouraging preclinical data of Bcl-2 as a potential target for the treatment of hypodiploid B-ALL.


Subject(s)
Antineoplastic Agents/pharmacology , Diploidy , Leukemia, Experimental/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Apoptosis , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Lineage , Cell Proliferation , Humans , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Nat Commun ; 8(1): 2127, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29259179

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is a myeloproliferative disorder of childhood caused by mutations in the Ras pathway. Outcomes in JMML vary markedly from spontaneous resolution to rapid relapse after hematopoietic stem cell transplantation. Here, we hypothesized that DNA methylation patterns would help predict disease outcome and therefore performed genome-wide DNA methylation profiling in a cohort of 39 patients. Unsupervised hierarchical clustering identifies three clusters of patients. Importantly, these clusters differ significantly in terms of 4-year event-free survival, with the lowest methylation cluster having the highest rates of survival. These findings were validated in an independent cohort of 40 patients. Notably, all but one of 14 patients experiencing spontaneous resolution cluster together and closer to 22 healthy controls than to other JMML cases. Thus, we show that DNA methylation patterns in JMML are predictive of outcome and can identify the patients most likely to experience spontaneous resolution.


Subject(s)
DNA Methylation , Genome, Human/genetics , Leukemia, Myelomonocytic, Juvenile/genetics , Neoplasm Regression, Spontaneous/genetics , Antineoplastic Agents/therapeutic use , Biopsy , Child , Child, Preschool , Disease-Free Survival , Female , Hematopoietic Stem Cell Transplantation , Humans , Infant , Kaplan-Meier Estimate , Leukemia, Myelomonocytic, Juvenile/blood , Leukemia, Myelomonocytic, Juvenile/mortality , Leukemia, Myelomonocytic, Juvenile/therapy , Male , Monocytes , Mutation , Prognosis , Prospective Studies
13.
Cancer Res ; 77(21): 5706-5711, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28993414

ABSTRACT

Preclinical studies using genetically engineered mouse models (GEMM) have the potential to expedite the development of effective new therapies; however, they are not routinely integrated into drug development pipelines. GEMMs may be particularly valuable for investigating treatments for less common cancers, which frequently lack alternative faithful models. Here, we describe a multicenter cooperative group that has successfully leveraged the expertise and resources from philanthropic foundations, academia, and industry to advance therapeutic discovery and translation using GEMMs as a preclinical platform. This effort, known as the Neurofibromatosis Preclinical Consortium (NFPC), was established to accelerate new treatments for tumors associated with neurofibromatosis type 1 (NF1). At its inception, there were no effective treatments for NF1 and few promising approaches on the horizon. Since 2008, participating laboratories have conducted 95 preclinical trials of 38 drugs or combinations through collaborations with 18 pharmaceutical companies. Importantly, these studies have identified 13 therapeutic targets, which have inspired 16 clinical trials. This review outlines the opportunities and challenges of building this type of consortium and highlights how it can accelerate clinical translation. We believe that this strategy of foundation-academic-industry partnering is generally applicable to many diseases and has the potential to markedly improve the success of therapeutic development. Cancer Res; 77(21); 5706-11. ©2017 AACR.


Subject(s)
Disease Models, Animal , Drug Discovery/methods , Neoplasms/drug therapy , Translational Research, Biomedical/methods , Animals , Humans , Mice , Molecular Targeted Therapy/methods , Neoplasms/complications , Neoplasms/diagnosis , Neurofibromatosis 1/complications , Neurofibromatosis 1/diagnosis , Neurofibromatosis 1/drug therapy
15.
Biotechniques ; 59(2): 82-6, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26260086

ABSTRACT

Time-lapse imaging can be used to quantify how cells move, divide, and die over time and under defined culture conditions. Open source software packages such as CellProfiler, Icy, and Fiji provide robust and convenient interfaces for performing such analyses. However, object tracking algorithms are imperfect, and validation of significant events is often required. This is challenging, as CellProfiler produces only tabular data for object tracking, and the graphical tools in Icy and Fiji are not optimal for manual review of these events. Here we describe Traxtile, a program that allows interactive graphical review and revision of object tracking assignments. Traxtile imports initial assignments and automatically identifies events needing review (i.e., apparent creation of new objects, splits, merges, and losses). For each such event, the object track is displayed on a montage of images centered on the event and spanning the preceding and subsequent frames. Links between cells in successive frames can be reviewed and edited, yielding validated tracks for the image series. Reports summarize events from the validated tracks. Traxtile is implemented in Python version 2.7 using standard distribution libraries (available at www.python.org) and is freely available at https://github.com/braunb/traxtile-public.


Subject(s)
Cell Tracking/methods , Image Processing, Computer-Assisted/methods , Software , Time-Lapse Imaging , Algorithms , Animals , Humans
16.
Blood ; 125(3): 516-24, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25395418

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is an aggressive myeloproliferative neoplasm of childhood associated with a poor prognosis. Recently, massively parallel sequencing has identified recurrent mutations in the SKI domain of SETBP1 in a variety of myeloid disorders. These lesions were detected in nearly 10% of patients with JMML and have been characterized as secondary events. We hypothesized that rare subclones with SETBP1 mutations are present at diagnosis in a large portion of patients who relapse, but are below the limits of detection for conventional deep sequencing platforms. Using droplet digital polymerase chain reaction, we identified SETBP1 mutations in 17/56 (30%) of patients who were treated in the Children's Oncology Group sponsored clinical trial, AAML0122. Five-year event-free survival in patients with SETBP1 mutations was 18% ± 9% compared with 51% ± 8% for those without mutations (P = .006).


Subject(s)
Carrier Proteins/genetics , Leukemia, Myelomonocytic, Juvenile/genetics , Mutation/genetics , Nuclear Proteins/genetics , Child, Preschool , Female , Follow-Up Studies , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Leukemia, Myelomonocytic, Juvenile/pathology , Male , Neoplasm Staging , Prognosis , Survival Rate
17.
Sci Signal ; 6(304): ra105, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24300897

ABSTRACT

Oncogenic K-Ras proteins, such as K-Ras(G12D), accumulate in the active, guanosine triphosphate (GTP)-bound conformation and stimulate signaling through effector kinases. The presence of the K-Ras(G12D) oncoprotein at a similar abundance to that of endogenous wild-type K-Ras results in only minimal phosphorylation and activation of the canonical Raf-mitogen-activated or extracellular signal-regulated protein kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) signaling cascades in primary hematopoietic cells, and these pathways remain dependent on growth factors for efficient activation. We showed that phospholipase C-γ (PLC-γ), PI3K, and their generated second messengers link activated cytokine receptors to Ras and ERK signaling in differentiated bone marrow cells and in a cell population enriched for leukemia stem cells. Cells expressing endogenous oncogenic K-Ras(G12D) remained dependent on the second messenger diacylglycerol for the efficient activation of Ras-ERK signaling. These data raise the unexpected possibility of therapeutically targeting proteins that function upstream of oncogenic Ras in cancer.


Subject(s)
Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hematopoietic Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Amino Acid Substitution , Animals , Cells, Cultured , Cytokines/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Hematopoietic Stem Cells/pathology , Leukemia/genetics , Leukemia/metabolism , Leukemia/pathology , MAP Kinase Signaling System/genetics , Mice , Mutation, Missense , Neoplastic Stem Cells/pathology , Phosphatidylinositol 3-Kinases/genetics , Phospholipase C gamma/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Second Messenger Systems/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
18.
J Clin Invest ; 123(1): 335-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23221337

ABSTRACT

Children with neurofibromatosis type 1 (NF1) are predisposed to juvenile myelomonocytic leukemia (JMML), an aggressive myeloproliferative neoplasm (MPN) that is refractory to conventional chemotherapy. Conditional inactivation of the Nf1 tumor suppressor in hematopoietic cells of mice causes a progressive MPN that accurately models JMML and chronic myelomonocytic leukemia (CMML). We characterized the effects of Nf1 loss on immature hematopoietic populations and investigated treatment with the MEK inhibitor PD0325901 (hereafter called 901). Somatic Nf1 inactivation resulted in a marked expansion of immature and lineage-committed myelo-erythroid progenitors and ineffective erythropoiesis. Treatment with 901 induced a durable drop in leukocyte counts, enhanced erythropoietic function, and markedly reduced spleen sizes in mice with MPN. MEK inhibition also restored a normal pattern of erythroid differentiation and greatly reduced extramedullary hematopoiesis. Remarkably, genetic analysis revealed the persistence of Nf1-deficient hematopoietic cells, indicating that MEK inhibition modulates the proliferation and differentiation of Nf1 mutant cells in vivo rather than eliminating them. These data provide a rationale for performing clinical trials of MEK inhibitors in patients with JMML and CMML.


Subject(s)
Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Erythropoiesis/drug effects , Hematopoiesis, Extramedullary/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelomonocytic, Juvenile/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neurofibromin 1 , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Child , Child, Preschool , Diphenylamine/pharmacology , Disease Models, Animal , Erythropoiesis/genetics , Hematopoiesis, Extramedullary/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelomonocytic, Juvenile/etiology , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/metabolism , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurofibromatosis 1/complications , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/genetics
20.
Blood ; 120(17): 3397-406, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22898602

ABSTRACT

Ras proteins are critical nodes in cellular signaling that integrate inputs from activated cell surface receptors and other stimuli to modulate cell fate through a complex network of effector pathways. Oncogenic RAS mutations are found in ∼25% of human cancers and are highly prevalent in hematopoietic malignancies. Because of their structural and biochemical properties, oncogenic Ras proteins are exceedingly difficult targets for rational drug discovery, and no mechanism-based therapies exist for cancers with RAS mutations. This article reviews the properties of normal and oncogenic Ras proteins, the prevalence and likely pathogenic role of NRAS, KRAS, and NF1 mutations in hematopoietic malignancies, relevant animal models of these cancers, and implications for drug discovery. Because hematologic malignancies are experimentally tractable, they are especially valuable platforms for addressing the fundamental question of how to reverse the adverse biochemical output of oncogenic Ras in cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Hematologic Neoplasms/genetics , ras Proteins/metabolism , Animals , Clinical Trials as Topic , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Humans , Mice , Molecular Targeted Therapy , Mutation , Neurofibromin 1/antagonists & inhibitors , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction/drug effects , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism , ras Proteins/antagonists & inhibitors , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...