Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cell ; 168(5): 817-829.e15, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28215705

ABSTRACT

Investigating therapeutic "outliers" that show exceptional responses to anti-cancer treatment can uncover biomarkers of drug sensitivity. We performed preclinical trials investigating primary murine acute myeloid leukemias (AMLs) generated by retroviral insertional mutagenesis in KrasG12D "knockin" mice with the MEK inhibitor PD0325901 (PD901). One outlier AML responded and exhibited intrinsic drug resistance at relapse. Loss of wild-type (WT) Kras enhanced the fitness of the dominant clone and rendered it sensitive to MEK inhibition. Similarly, human colorectal cancer cell lines with increased KRAS mutant allele frequency were more sensitive to MAP kinase inhibition, and CRISPR-Cas9-mediated replacement of WT KRAS with a mutant allele sensitized heterozygous mutant HCT116 cells to treatment. In a prospectively characterized cohort of patients with advanced cancer, 642 of 1,168 (55%) with KRAS mutations exhibited allelic imbalance. These studies demonstrate that serial genetic changes at the Kras/KRAS locus are frequent in cancer and modulate competitive fitness and MEK dependency.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Colorectal Neoplasms/genetics , Diphenylamine/analogs & derivatives , MAP Kinase Signaling System , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Line, Tumor , Clonal Evolution , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Drug Resistance, Neoplasm , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , MAP Kinase Signaling System/drug effects , Mice , Mutation , Retroviridae
2.
Ann Hematol ; 101(3): 557-569, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34981142

ABSTRACT

CC-90002 is an anti-CD47 antibody that inhibits CD47-SIRPα interaction and enables macrophage-mediated killing of tumor cells in hematological cancer cell lines. In this first clinical, phase 1, dose-escalation and -expansion study (CC-90002-AML-001; NCT02641002), we evaluated CC-90002 in patients with relapsed/refractory acute myeloid leukemia (AML) or high-risk myelodysplastic syndromes (MDS). CC-90002 was administered in escalating doses of 0.1-4.0 mg/kg, using a modified 3 + 3 design. Primary endpoints included dose-limiting toxicities (DLTs), non-tolerated dose (NTD), maximum tolerated dose (MTD), and recommended phase 2 dose. Secondary endpoints included preliminary efficacy, pharmacokinetics, and presence/frequency of anti-drug antibodies (ADAs). Between March 2016 and July 2018, 28 patients were enrolled (24 with AML and 4 with MDS) at 6 sites across the USA. As of July 18, 2018, all patients had discontinued, mainly due to death or progressive disease. The most common treatment-emergent adverse events were diarrhea (46.4%), thrombocytopenia (39.3%), febrile neutropenia (35.7%), and aspartate aminotransferase increase (35.7%). Four patients experienced DLTs (1 patient had grade 4 disseminated intravascular coagulation and grade 5 cerebral hemorrhage, 1 had grade 3 purpura, 1 had grade 4 congestive cardiac failure and grade 5 acute respiratory failure, and another had grade 5 sepsis). The NTD and MTD were not reached. No objective responses occurred. CC-90002 serum exposure was dose-dependent. ADAs were present across all doses, and the proportion of ADA-positive patients in cycle 1 increased over time. Despite no unexpected safety findings, the CC-90002-AML-001 study was discontinued in dose escalation for lack of monotherapy activity and evidence of ADAs. However, as other anti-CD47 agents in clinical trials are showing promising early results for AML and MDS, understanding preclinical and clinical differences between individual agents in this class will be of high importance.


Subject(s)
Antineoplastic Agents, Immunological , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Neoplasm Recurrence, Local , Adult , Aged , Aged, 80 and over , Animals , Female , Humans , Male , Mice , Middle Aged , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , CD47 Antigen/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Macaca fascicularis , Maximum Tolerated Dose , Mice, SCID , Myelodysplastic Syndromes/drug therapy , Neoplasm Recurrence, Local/drug therapy
3.
Blood ; 131(24): 2682-2697, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29592892

ABSTRACT

Monosomy 7 (-7) and del(7q) are high-risk cytogenetic abnormalities common in myeloid malignancies. We previously reported that CUX1, a homeodomain-containing transcription factor encoded on 7q22, is frequently inactivated in myeloid neoplasms, and CUX1 myeloid tumor suppressor activity is conserved from humans to Drosophila. CUX1-inactivating mutations are recurrent in clonal hematopoiesis of indeterminate potential as well as myeloid malignancies, in which they independently carry a poor prognosis. To determine the role for CUX1 in hematopoiesis, we generated 2 short hairpin RNA-based mouse models with ∼54% (Cux1mid) or ∼12% (Cux1low) residual CUX1 protein. Cux1mid mice develop myelodysplastic syndrome (MDS) with anemia and trilineage dysplasia, whereas CUX1low mice developed MDS/myeloproliferative neoplasms and anemia. In diseased mice, restoration of CUX1 expression was sufficient to reverse the disease. CUX1 knockdown bone marrow transplant recipients exhibited a transient hematopoietic expansion, followed by a reduction of hematopoietic stem cells (HSCs), and fatal bone marrow failure, in a dose-dependent manner. RNA-sequencing after CUX1 knockdown in human CD34+ cells identified a -7/del(7q) MDS gene signature and altered differentiation, proliferative, and phosphatidylinositol 3-kinase (PI3K) signaling pathways. In functional assays, CUX1 maintained HSC quiescence and repressed proliferation. These homeostatic changes occurred in parallel with decreased expression of the PI3K inhibitor, Pik3ip1, and elevated PI3K/AKT signaling upon CUX1 knockdown. Our data support a model wherein CUX1 knockdown promotes PI3K signaling, drives HSC exit from quiescence and proliferation, and results in HSC exhaustion. Our results also demonstrate that reduction of a single 7q gene, Cux1, is sufficient to cause MDS in mice.


Subject(s)
Gene Dosage , Hematopoiesis , Hematopoietic Stem Cells/pathology , Homeodomain Proteins/genetics , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Nuclear Proteins/genetics , Repressor Proteins/genetics , Anemia/genetics , Anemia/pathology , Anemia/physiopathology , Animals , Cell Proliferation , Cellular Senescence , Disease Models, Animal , Gene Knockdown Techniques , Humans , Mice , Myelodysplastic Syndromes/physiopathology
4.
Proc Natl Acad Sci U S A ; 111(41): 14876-81, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25271328

ABSTRACT

MAGUK Inverted 2 (MAGI-2) is a PTEN-interacting scaffold protein implicated in cancer on the basis of rare, recurrent genomic translocations and deletions in various tumors. In the renal glomerulus, MAGI-2 is exclusively expressed in podocytes, specialized cells forming part of the glomerular filter, where it interacts with the slit diaphragm protein nephrin. To further explore MAGI-2 function, we generated Magi-2-KO mice through homologous recombination by targeting an exon common to all three alternative splice variants. Magi-2 null mice presented with progressive proteinuria as early as 2 wk postnatally, which coincided with loss of nephrin expression in the glomeruli. Magi-2-null kidneys revealed diffuse podocyte foot process effacement and focal podocyte hypertrophy by 3 wk of age, as well as progressive podocyte loss. By 5.5 wk, coinciding with a near-complete loss of podocytes, Magi-2-null mice developed diffuse glomerular extracapillary epithelial cell proliferations, and died of renal failure by 3 mo of age. As confirmed by immunohistochemical analysis, the proliferative cell populations in glomerular lesions were exclusively composed of activated parietal epithelial cells (PECs). Our results reveal that MAGI-2 is required for the integrity of the kidney filter and podocyte survival. Moreover, we demonstrate that PECs can be activated to form glomerular lesions resembling a noninflammatory glomerulopathy with extensive extracapillary proliferation, sometimes resembling crescents, following rapid and severe podocyte loss.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Guanylate Kinases/metabolism , Kidney/pathology , Animals , Cell Proliferation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Immunohistochemistry , Inflammation/pathology , Kidney/metabolism , Kidney Glomerulus/blood supply , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Membrane Proteins/metabolism , Mice, Knockout , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Podocytes/metabolism , Podocytes/pathology , Proteinuria/metabolism , Proteinuria/pathology , Survival Analysis , Up-Regulation
5.
Blood ; 124(26): 3947-55, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25361812

ABSTRACT

Oncogenic NRAS mutations are highly prevalent in acute myeloid leukemia (AML). Genetic analysis supports the hypothesis that NRAS mutations cooperate with antecedent molecular lesions in leukemogenesis, but have limited independent prognostic significance. Using short hairpin RNA-mediated knockdown in human cell lines and primary mouse leukemias, we show that AML cells with NRAS/Nras mutations are dependent on continued oncogene expression in vitro and in vivo. Using the Mx1-Cre transgene to inactivate a conditional mutant Nras allele, we analyzed hematopoiesis and hematopoietic stem and progenitor cells (HSPCs) under normal and stressed conditions and found that HSPCs lacking Nras expression are functionally equivalent to normal HSPCs in the adult mouse. Treating recipient mice transplanted with primary Nras(G12D) AMLs with 2 potent allosteric mitogen-activated protein kinase kinase (MEK) inhibitors (PD0325901 or trametinib/GlaxoSmithKline 1120212) significantly prolonged survival and reduced proliferation but did not induce apoptosis, promote differentiation, or drive clonal evolution. The phosphatidylinositol 3-kinase inhibitor GDC-0941 was ineffective as a single agent and did not augment the activity of PD0325901. All mice ultimately succumbed to progressive leukemia. Together, these data validate oncogenic N-Ras signaling as a therapeutic target in AML and support testing combination regimens that include MEK inhibitors.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , MAP Kinase Kinase Kinases/metabolism , Monomeric GTP-Binding Proteins/metabolism , Mutation , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Genes, ras , Hematopoietic Stem Cells/cytology , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Monomeric GTP-Binding Proteins/genetics , Protein Kinase Inhibitors/chemistry , RNA, Small Interfering/metabolism , Signal Transduction , Stem Cells/cytology , Transgenes
6.
Nat Med ; 23(3): 301-313, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28191887

ABSTRACT

Expression of the MECOM (also known as EVI1) proto-oncogene is deregulated by chromosomal translocations in some cases of acute myeloid leukemia (AML) and is associated with poor clinical outcome. Here, through transcriptomic and metabolomic profiling of hematopoietic cells, we reveal that EVI1 overexpression alters cellular metabolism. A screen using pooled short hairpin RNAs (shRNAs) identified the ATP-buffering, mitochondrial creatine kinase CKMT1 as necessary for survival of EVI1-expressing cells in subjects with EVI1-positive AML. EVI1 promotes CKMT1 expression by repressing the myeloid differentiation regulator RUNX1. Suppression of arginine-creatine metabolism by CKMT1-directed shRNAs or by the small molecule cyclocreatine selectively decreased the viability, promoted the cell cycle arrest and apoptosis of human EVI1-positive cell lines, and prolonged survival in both orthotopic xenograft models and mouse models of primary AML. CKMT1 inhibition altered mitochondrial respiration and ATP production, an effect that was abrogated by phosphocreatine-mediated reactivation of the arginine-creatine pathway. Targeting CKMT1 is thus a promising therapeutic strategy for this EVI1-driven AML subtype that is highly resistant to current treatment regimens.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Creatine Kinase/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Leukemia, Myeloid, Acute/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics , Adult , Aged , Aged, 80 and over , Blotting, Western , Computer Simulation , Core Binding Factor Alpha 2 Subunit/metabolism , Creatine Kinase/metabolism , Female , Flow Cytometry , Gene Expression Profiling , Genome-Wide Association Study , Humans , Leukemia, Myeloid, Acute/metabolism , MDS1 and EVI1 Complex Locus Protein , Male , Metabolic Networks and Pathways , Metabolomics , Middle Aged , Mitochondria , Proto-Oncogene Mas , RNA, Small Interfering
7.
ScientificWorldJournal ; 6: 918-30, 2006 Aug 11.
Article in English | MEDLINE | ID: mdl-16906325

ABSTRACT

Imatinib (Gleevec/STI-571/CGP57148B, Novartis) is a small-molecule, tyrosine kinase inhibitor developed to target BCR-ABL, c-Kit, and PDGF-R. Through inhibition of these oncogenic kinases, imatinib is effective in the treatment of BCR-ABL-positive leukemia, gastrointestinal stromal tumor, and hypereosinophilic syndrome, respectively. However, clinical success of imatinib is hampered by acquired resistance that may occur through several mechanisms including kinase domain mutation, target amplification, and activation of alternate signaling pathways. Strategies to overcome resistance have included targeting BCR-ABL stability and downstream signaling pathways important for tumor growth. Additional work has shown that new BCR-ABL kinase inhibitors with increased potency or alternate conformation-binding properties can target imatinib resistance. This review focuses on the mechanisms of imatinib resistance and the strategies currently being developed to overcome clinical resistance.


Subject(s)
Antineoplastic Agents/therapeutic use , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia/drug therapy , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Benzamides , Drug Delivery Systems , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Leukemia/genetics , Mutation , Protein Structure, Tertiary/genetics
8.
Elife ; 42015 Jul 20.
Article in English | MEDLINE | ID: mdl-26193121

ABSTRACT

Chromosome 7 deletions are highly prevalent in myelodysplastic syndrome (MDS) and likely contribute to aberrant growth through haploinsufficiency. We generated mice with a heterozygous germ line deletion of a 2-Mb interval of chromosome band 5A3 syntenic to a commonly deleted segment of human 7q22 and show that mutant hematopoietic cells exhibit cardinal features of MDS. Specifically, the long-term hematopoietic stem cell (HSC) compartment is expanded in 5A3(+/del) mice, and the distribution of myeloid progenitors is altered. 5A3(+/del) HSCs are defective for lymphoid repopulating potential and show a myeloid lineage output bias. These cell autonomous abnormalities are exacerbated by physiologic aging and upon serial transplantation. The 5A3 deletion partially rescues defective repopulation in Gata2 mutant mice. 5A3(+/del) hematopoietic cells exhibit decreased expression of oxidative phosphorylation genes, increased levels of reactive oxygen species, and perturbed oxygen consumption. These studies provide the first functional data linking 7q22 deletions to MDS pathogenesis.


Subject(s)
Chromosomes, Human, Pair 7 , Genetic Loci , Haploinsufficiency , Myelodysplastic Syndromes/genetics , Animals , Disease Models, Animal , Humans , Mice, Inbred C57BL , Oxidative Phosphorylation , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Sequence Deletion
9.
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
10.
Proc Natl Acad Sci U S A ; 103(51): 19466-71, 2006 Dec 19.
Article in English | MEDLINE | ID: mdl-17164333

ABSTRACT

The success of targeting kinases in cancer with small molecule inhibitors has been tempered by the emergence of drug-resistant kinase domain mutations. In patients with chronic myeloid leukemia treated with ABL inhibitors, BCR-ABL kinase domain mutations are the principal mechanism of relapse. Certain mutations are occasionally detected before treatment, suggesting increased fitness relative to wild-type p210 BCR-ABL. We evaluated the oncogenicity of eight kinase inhibitor-resistant BCR-ABL mutants and found a spectrum of potencies greater or less than p210. Although most fitness alterations correlate with changes in kinase activity, this is not the case with the T315I BCR-ABL mutation that confers clinical resistance to all currently approved ABL kinase inhibitors. Through global phosphoproteome analysis, we identified a unique phosphosubstrate signature associated with each drug-resistant allele, including a shift in phosphorylation of two tyrosines (Tyr253 and Tyr257) in the ATP binding loop (P-loop) of BCR-ABL when Thr315 is Ile or Ala. Mutational analysis of these tyrosines in the context of Thr315 mutations demonstrates that the identity of the gatekeeper residue impacts oncogenicity by altered P-loop phosphorylation. Therefore, mutations that confer clinical resistance to kinase inhibitors can substantially alter kinase function and confer novel biological properties that may impact disease progression.


Subject(s)
Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Cell Line , DNA Mutational Analysis , Mass Spectrometry , Mice , Molecular Sequence Data , Mutation, Missense/genetics , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/metabolism , Proteomics
11.
Proc Natl Acad Sci U S A ; 102(9): 3395-400, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15705718

ABSTRACT

Structural studies suggest that most point mutations in the BCR-ABL kinase domain cause resistance to the ABL kinase inhibitor imatinib by impairing the flexibility of the kinase domain, restricting its ability to adopt the inactive conformation required for optimal imatinib binding, rather than by directly interfering with drug contact residues. BMS-354825, currently in clinical development for imatinib-resistant chronic myelogenous leukemia, is a dual SRC/ABL kinase inhibitor that binds ABL in both the active and inactive conformation. To examine the potential role of conformational binding properties in drug resistance, we mapped the mutations in BCR-ABL capable of conferring resistance to BMS-354825. Through saturation mutagenesis, we identified 10 such BCR-ABL mutations, 8 of which occurred at drug contact residues. Some mutants were unique to BMS-354825, whereas others also conferred imatinib resistance. Remarkably, the identity of the amino acid substitution at either of two contact residues differentially affects sensitivity to imatinib or BMS-354825. The combination of imatinib plus BMS-354825 greatly reduced the recovery of drug-resistant clones. Our findings provide further rationale for considering kinase conformation in the design of kinase inhibitors against cancer targets.


Subject(s)
Enzyme Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Base Sequence , Cell Line , DNA Primers , Fusion Proteins, bcr-abl , Humans , Models, Molecular , Protein Conformation , Protein-Tyrosine Kinases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL