Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
2.
J Clin Oncol ; 30(19): 2307-13, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22547604

ABSTRACT

PURPOSE: Aberrant Notch signaling has been implicated in the pathogenesis of many human cancers. MK-0752 is a potent, oral inhibitor of γ-secretase, an enzyme required for Notch pathway activation. Safety, maximum-tolerated dose, pharmacokinetics (PKs), pharmacodynamics, and preliminary antitumor efficacy were assessed in a phase I study of MK-0752. PATIENTS AND METHODS: MK-0752 was administered in three different schedules to patients with advanced solid tumors. Hair follicles were collected at higher dose levels to assess a gene signature of Notch inhibition. RESULTS: Of 103 patients who received MK-0752, 21 patients received a continuous once-daily dosing at 450 and 600 mg; 17 were dosed on an intermittent schedule of 3 of 7 days at 450 and 600 mg; and 65 were dosed once per week at 600, 900, 1,200, 1,500, 1,800, 2,400, 3,200, and 4,200 mg. The most common drug-related toxicities were diarrhea, nausea, vomiting, and fatigue. PKs (area under the concentration-time curve and maximum measured plasma concentration) increased in a less than dose proportional manner, with a half-life of approximately 15 hours. Significant inhibition of Notch signaling was observed with the 1,800- to 4,200-mg weekly dose levels, confirming target engagement at those doses. One objective complete response and an additional 10 patients with stable disease longer than 4 months were observed among patients with high-grade gliomas. CONCLUSION: MK-0752 toxicity was schedule dependent. Weekly dosing was generally well tolerated and resulted in strong modulation of a Notch gene signature. Clinical benefit was observed, and rational combination trials are currently ongoing to maximize clinical benefit with this novel agent.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/metabolism , Propionates/pharmacology , Propionates/pharmacokinetics , Sulfones/pharmacology , Sulfones/pharmacokinetics , Administration, Oral , Adult , Aged , Aged, 80 and over , Benzene Derivatives , Cohort Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Neoplasms/pathology , Propionates/adverse effects , Sulfones/adverse effects , Treatment Outcome
3.
Cancer Chemother Pharmacol ; 67(2): 305-14, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20386909

ABSTRACT

PURPOSE: To assess the maximum-tolerated dose (MTD), dose-limiting toxicity (DLT), safety, and tolerability of the 24-h continuous intravenous (CIV) infusion of MK-0457, a novel pan-Aurora kinase inhibitor, in patients with advanced solid tumors and to determine the bioavailability of an oral dose of 100 mg MK-0457. STUDY DESIGN: MK-0457 was administered as a 24-h CIV infusion every 21 days. Dose escalation proceeded per toxicity criteria. A 100-mg oral dose was administered to seven patients 48 h prior to the CIV infusion dose of 64 mg/m(2)/h. RESULTS: Twenty-seven patients received a total of 86 infusions of MK-0457. Dose-limiting toxicity at 96 mg/m(2)/h included grade 4 neutropenia and grade 3 herpes zoster. The MTD was identified as 64 mg/m(2)/h. The most common adverse events were nausea, vomiting, diarrhea, and fatigue. Pharmacokinetic analyses revealed that CIV infusion MK-0457 had an estimated mean terminal half-life of approximately 6.6-10.2 h and that end-of-infusion concentrations and mean AUCs were approximately dose proportional. The estimated mean oral bioavailability of MK-0457 was 7.9%. One patient with advanced ovarian cancer attained prolonged stable disease for 11 months. CONCLUSIONS: MK-0457 was well tolerated in this schedule. Almost half the patients attained stable disease. Further development of this class of agents will likely occur in combination with other anti-cancer treatments.


Subject(s)
Neoplasms/drug therapy , Piperazines/pharmacology , Piperazines/toxicity , Piperazines/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Adult , Aged , Aged, 80 and over , Area Under Curve , Aurora Kinases , Biological Availability , Female , Hematologic Diseases/chemically induced , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Treatment Outcome , Young Adult
4.
Blood ; 109(2): 500-2, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-16990603

ABSTRACT

MK-0457 (VX-680) is a small-molecule aurora kinase (AK) inhibitor with preclinical antileukemia activity. The T315I BCR-ABL mutation mediates resistance to imatinib, nilotinib, and dasatinib. MK-0457 has in vitro activity against cells expressing wild-type or mutated BCR-ABL, including the T315I BCR-ABL mutation. Three patients with T315I abl-mutated chronic myeloid leukemia (CML) or Philadelphia chromosome (Ph)-positive acute lymphocytic leukemia (ALL) have achieved clinical responses to doses of MK-04547 that are not associated with adverse events. Higher MK-0457 dose levels were associated with clinical responses and down-regulation of CrkL phosphorylation in leukemia cells. The possible role of AK inhibition in these clinical responses requires further investigation. The currently reported cases are the first observed clinical activity of a kinase inhibitor against the T315I phenotype. The observation of responses in 3 patients with T315I phenotype-refractory CML or Ph-positive ALL, at doses of MK-0457 associated with no significant extramedullary toxicity, is very encouraging.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Piperazines/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Kinase Inhibitors/therapeutic use , Adaptor Proteins, Signal Transducing/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Adult , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Female , Fusion Proteins, bcr-abl/drug effects , Humans , Infusions, Intravenous , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Male , Maximum Tolerated Dose , Middle Aged , Mutation , Nuclear Proteins/drug effects , Nuclear Proteins/metabolism , Phenotype , Phosphorylation , Piperazines/administration & dosage , Piperazines/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Treatment Outcome
5.
Cancer Chemother Pharmacol ; 59(3): 407-12, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17051369

ABSTRACT

PURPOSE: Aprepitant, a selective neurokinin-1 (NK-1) receptor antagonist approved for the treatment and prevention of emesis caused by moderately and highly emetogenic chemotherapy, is an inhibitor, inducer, and substrate of the cytochrome P450 3194 pathway. The CYP3A4 pathway is the major pathway of the metabolism of vinorelbine, a vinca alkaloid frequently used in combination with cisplatin. Therefore, we studied the potential interaction of the aprepitant 3-day antiemetic regimen on the pharmacokinetics of vinorelbine. PATIENTS AND METHODS: Fourteen patients with metastatic solid tumors were included in this open-label, balanced, 2-period crossover study. In treatment arm A, vinorelbine (25 mg/m2 weekly) was administered alone, while in treatment arm B the same dose of vinorelbine was administered following the administration of the aprepitant antiemetic regimen on day 1 and alone on day 8. The antiemetic regimen of aprepitant was comprised of the following; on day 1: 125 mg aprepitant, 12 mg dexamethasone, and 32 mg ondansetron; on days 2 and 3: 80 mg aprepitant and 8 mg dexamethasone and on day 4: 8 mg dexamethasone. Blood samples for vinorelbine pharmacokinetic analysis were collected over 96 h. RESULTS: Two patients discontinued the study due to adverse events that were judged not to be drug-related. Complete pharmacokinetic data of vinorelbine administered alone and with the aprepitant antiemetic regimen were obtained in 12 patients. The mean plasma concentration profile of vinorelbine administered with aprepitant was identical to that following vinorelbine administered alone, with geometric mean vinorelbine plasma AUC ratios of treatment B day 1/treatment A day 1 and of treatment B day 8/treatment A day of 1.01 (0.93, 1.10) and 1.00 (0.92, 1.08), respectively. CONCLUSION: As the aprepitant antiemetic regimen has no detectable inhibitory or inductive effect on the pharmacokinetics of vinorelbine, aprepitant when added to a standard antiemetic regimen consisting of ondansetron and dexamethasone can be safely combined with vinorelbine at clinically recommended doses.


Subject(s)
Antiemetics/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacokinetics , Dexamethasone/therapeutic use , Morpholines/therapeutic use , Neoplasms/metabolism , Ondansetron/therapeutic use , Vinblastine/analogs & derivatives , Aprepitant , Area Under Curve , Cross-Over Studies , Drug Therapy, Combination , Female , Half-Life , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/drug therapy , Vinblastine/pharmacokinetics , Vinorelbine
6.
Cancer Cell ; 6(1): 33-43, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15261140

ABSTRACT

Homeostasis under hypoxic conditions is maintained through a coordinated transcriptional response mediated by the hypoxia-inducible factor (HIF) pathway and requires coactivation by the CBP and p300 transcriptional coactivators. Through a target-based high-throughput screen, we identified chetomin as a disrupter of HIF binding to p300. At a molecular level, chetomin disrupts the structure of the CH1 domain of p300 and precludes its interaction with HIF, thereby attenuating hypoxia-inducible transcription. Systemic administration of chetomin inhibited hypoxia-inducible transcription within tumors and inhibited tumor growth. These results demonstrate a therapeutic window for pharmacological attenuation of HIF activity and further establish the feasibility of disrupting a signal transduction pathway by targeting the function of a transcriptional coactivator with a small molecule.


Subject(s)
Anti-Bacterial Agents/pharmacology , DNA-Binding Proteins , Nuclear Proteins/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Carcinoma, Hepatocellular/pathology , Cell Hypoxia/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Disulfides , E1A-Associated p300 Protein , Erythropoietin/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Indole Alkaloids , Liver Neoplasms/pathology , Luciferases/metabolism , Male , Mice , Mice, Nude , Nuclear Proteins/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Protein Binding/drug effects , Receptors, Aryl Hydrocarbon/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Trans-Activators/genetics , Transcription Factors/genetics , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/metabolism
7.
Biochemistry ; 42(33): 9937-45, 2003 Aug 26.
Article in English | MEDLINE | ID: mdl-12924942

ABSTRACT

Numerous transcription factors interact with the basal transcriptional machinery through the transcriptional co-activators p300 and CREB-binding protein (CBP). The Zn(2+)-binding cysteine/histidine-rich 1 (CH1) domain of p300/CBP binds many of these transcription factors, including hypoxia-inducible factor (HIF). We studied the structural and biophysical properties of the p300 CH1 domain alone and bound to the HIF-1 alpha C-terminal transactivation domain (TAD) to understand the diverse binding properties of CH1. The Zn(2+)-bound CH1 domain (CH1-Zn(2+)) and the HIF-1 alpha TAD-CH1 complex (CH1-Zn(2+)-HIF-1 alpha) are similarly helical, whereas metal-free CH1 is mostly random coil. CH1-Zn(2+) undergoes noncooperative thermal denaturation, does not have a near-UV elliptical signal, and binds the hydrophobic fluorophore ANS. In contrast, the CH1-Zn(2+)-HIF-1 alpha complex undergoes cooperative thermal denaturation, does produce a near-UV signal, and does not bind ANS. Addition of Zn(2+) ions to metal-free CH1 produced one conformational change, and subsequent addition of a HIF-1 alpha TAD peptide induced a second conformational change as detected by intrinsic tryptophan fluorescence spectroscopy. The NMR (1)H-(15)N HSQC spectrum of CH1-Zn(2+) exhibits few poorly dispersed peaks with broad line widths. Removal of metal ions produces more poorly dispersed peaks with sharper line widths. Addition of a HIF-1 alpha TAD peptide to CH1-Zn(2+) produces many well-dispersed peaks with sharp line widths. Taken together, these data support three conformational states for CH1, including an unstructured metal-free domain, a partially structured Zn(2+)-bound domain with molten globule characteristics, and a stable, well-ordered HIF-1 alpha TAD-CH1 complex.


Subject(s)
Nuclear Proteins/chemistry , Trans-Activators/chemistry , Amino Acid Sequence , Escherichia coli/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation , Protein Folding , Sequence Homology, Amino Acid , Spectrometry, Fluorescence , Trans-Activators/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Zinc/chemistry , Zinc/metabolism
8.
Nat Struct Biol ; 10(7): 504-12, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12778114

ABSTRACT

Expression of hypoxia-responsive genes is mediated by the heterodimeric transcription factor hypoxia-inducible factor-1 (HIF-1) in complex with the p300/CREB-binding protein (p300/CBP) transcriptional coactivator. The protein CITED2, which binds p300/CBP, is thought to be a negative regulator of HIF-1 transactivation. We show that the CITED2 transactivation domain (TAD) disrupts a complex of the HIF-1alpha C-terminal TAD (C-TAD) and the cysteine-histidine-rich 1 (CH1) domain of p300/CBP by binding CH1 with high affinity. The high-resolution solution structure of the CITED2 TAD-p300 CH1 complex shows that the CITED2 TAD, like the HIF-1alpha C-TAD, folds on a helical, Zn2+-containing CH1 scaffold. The CITED2 TAD binds a different, more extensive surface of CH1 than does the HIF-1alpha C-TAD. However, a conserved 'LPXL' sequence motif in CITED2 and HIF-1alpha interacts with an overlapping binding site on CH1. Mutation of the LPEL sequence in full-length CITED2 abolishes p300 binding in vivo. These findings reveal that CITED2 regulates HIF-1 by competing for a hot spot on the p300 CH1 domain.


Subject(s)
DNA-Binding Proteins , Repressor Proteins , Trans-Activators/physiology , Transcription Factors/chemistry , Transcription Factors/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , Circular Dichroism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Trans-Activators/metabolism , Transcription Factors/metabolism
9.
Brain Pathol ; 13(2): 235-6, 239, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12744478

ABSTRACT

The November 2002 COM. A 72-year-old immunocompetent woman presented with recent confusion, memory loss, visual and a gait disturbance. MRI scans demonstrated a T2 hypointense, gadolinium enhancing mass in her pineal region. A whole body CT scan identified a large retroperitoneal mass and significant lymphadenopathy. She underwent a biopsy of the pineal lesion. The intraoperative smear contained discohesive, basophilic malignant cells of moderate size that had only scant cytoplasm, an increased nuclear-to-cytoplasmic ratio and irregular, hyperchromatic nuclei containing prominent nucleoli. Mitotic figures, apoptotic and lymphoglandular bodies were frequent. Histologic examination showed malignant cells infiltrating and almost completely replacing the residual pineal gland. The tumor cells immunoreacted with LCA and CD-20. Scattered CD3 and UCHL-1 positive cells were also present. The diagnosis was stage IV, high grade, large B-cell lymphoma involving the pineal gland and retroperitoneum. Four cycles of systemic chemotherapy significantly reduced the patient's retroperitoneal mass. However, a repeat head MRI scan continued to show gadolinium-enhancement in the pineal region, now with extension into contiguous brain parenchyma and the subependymal region. The patient died 4 months after presentation. At autopsy, the lymphoma encased the pineal region and disseminated along the ventricular walls and into the subarachnoid spread. Pineal-region tumors are rare in the elderly population, and although unusual, lymphomas should be considered in this context. The behavior of this circumventricular organ lymphoma was comparable to both peripheral and primary central nervous system lymphomas.


Subject(s)
Brain Neoplasms/pathology , Lymphoma, B-Cell/pathology , Pineal Gland/pathology , Pinealoma/pathology , Aged , Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Diagnosis, Differential , Fatal Outcome , Female , Humans , Lymphoma, B-Cell/diagnosis , Lymphoma, B-Cell/therapy , Pinealoma/diagnosis , Pinealoma/therapy
10.
J Biol Chem ; 278(15): 13462-7, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12556468

ABSTRACT

SNARE proteins mediate intracellular membrane fusion by forming a coiled-coil complex to merge opposing membranes. A "fusion-active" neuronal SNARE complex is a parallel four-helix bundle containing two coiled-coil domains from SNAP-25 and one coiled-coil domain each from syntaxin-1a and VAMP-2. "Prefusion" assembly intermediate complexes can also form from these SNAREs. We studied the N-terminal coiled-coil domain of SNAP-23 (SNAP-23N), a non-neuronal homologue of SNAP-25, and its interaction with other coiled-coil domains. SNAP-23N can assemble spontaneously with the coiled-coil domains from SNAP-23C, syntaxin-4, and VAMP-3 to form a heterotetrameric complex. Unexpectedly, pure SNAP-23N crystallizes as a coiled-coil homotetrameric complex. The four helices have a parallel orientation and are symmetrical about the long axis. The complex is stabilized through the interaction of conserved hydrophobic residues comprising the a and d positions of the coiled-coil heptad repeats. In addition, a central, highly conserved glutamine residue (Gln-48) is buried within the interface by hydrogen bonding between glutamine side chains derived from adjacent subunits and to solvent molecules. A comparison of the SNAP-23N structure to other SNARE complex structures reveals how a simple coiled-coil motif can form diverse SNARE complexes.


Subject(s)
Carrier Proteins/chemistry , Amino Acid Sequence , Animals , Carrier Proteins/ultrastructure , Crystallography, X-Ray , Humans , Macromolecular Substances , Models, Molecular , Molecular Sequence Data , Polymerase Chain Reaction , Protein Conformation , Protein Structure, Secondary , Qb-SNARE Proteins , Qc-SNARE Proteins , Recombinant Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid
11.
Proc Natl Acad Sci U S A ; 99(8): 5367-72, 2002 Apr 16.
Article in English | MEDLINE | ID: mdl-11959990

ABSTRACT

Adaptation to hypoxia is mediated by transactivation of hypoxia-responsive genes by hypoxia-inducible factor-1 (HIF-1) in complex with the CBP and p300 transcriptional coactivators. We report the solution structure of the cysteine/histidine-rich 1 (CH1) domain of p300 bound to the C-terminal transactivation domain of HIF-1 alpha. CH1 has a triangular geometry composed of four alpha-helices with three intervening Zn(2+)-coordinating centers. CH1 serves as a scaffold for folding of the HIF-1 alpha C-terminal transactivation domain, which forms a vise-like clamp on the CH1 domain that is stabilized by extensive hydrophobic and polar interactions. The structure reveals the mechanism of specific recognition of p300 by HIF-1 alpha, and shows how HIF-1 alpha transactivation is regulated by asparagine hydroxylation.


Subject(s)
Nuclear Proteins/chemistry , Trans-Activators/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Animals , Asparagine/chemistry , Escherichia coli/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Transcription Factors/metabolism , Transcriptional Activation , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...