Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Traffic Inj Prev ; 22(5): 366-371, 2021.
Article in English | MEDLINE | ID: mdl-33960857

ABSTRACT

OBJECTIVE: Sleep deprivation is known to affect driving behavior and may lead to serious car accidents similar to the effects from e.g., alcohol. In a previous study, we have demonstrated that the use of machine learning techniques allows adequate characterization of abnormal driving behavior after alprazolam and/or alcohol intake. In the present study, we extend this approach to sleep deprivation and test the model for characterization of new interventions. We aimed to classify abnormal driving behavior after sleep deprivation, and, by using a machine learning model, we tested if this model could also pick up abnormal driving behavior resulting from other interventions. METHODS: Data were collected during a previous study, in which 24 subjects were tested after being sleep-deprived and after a well-rested night. Features were calculated from several driving parameters, such as the lateral position, speed of the car, and steering speed. In the present study, we used a gradient boosting model to classify sleep deprivation. The model was validated using a 5-fold cross validation technique. Next, probability scores were used to identify the overlap of driving behavior after sleep deprivation and driving behavior affected by other interventions. In the current study alprazolam, alcohol, and placebo are used to test/validate the approach. RESULTS: The sleep deprivation model detected abnormal driving behavior in the simulator with an accuracy of 77 ± 9%. Abnormal driving behavior after alprazolam, and to a lesser extent also after alcohol intake, showed remarkably similar characteristics to sleep deprivation. The average probability score for alprazolam and alcohol measurements was 0.79, for alcohol 0.63, and for placebo only 0.27 and 0.30, matching the expected relative drowsiness. CONCLUSION: We developed a model detecting abnormal driving induced by sleep deprivation. The model shows the similarities in driving characteristics between sleep deprivation and other interventions, i.e., alcohol and alprazolam. Consequently, our model for sleep deprivation may serve as a next reference point for a driving test battery of newly developed drugs.


Subject(s)
Accidents, Traffic/prevention & control , Attention/physiology , Reaction Time/physiology , Sleep Deprivation/physiopathology , Adult , Alprazolam/therapeutic use , Automobile Driving , Computer Simulation/statistics & numerical data , GABA Agents/therapeutic use , Humans , Machine Learning , Male , Wakefulness/physiology
2.
Accid Anal Prev ; 148: 105822, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33125924

ABSTRACT

RATIONALE: Car-driving performance is negatively affected by the intake of alcohol, tranquillizers, sedatives and sleep deprivation. Although several studies have shown that the standard deviation of the lateral position on the road (SDLP) is sensitive to drug-induced changes in simulated and real driving performance tests, this parameter alone might not fully assess and quantify deviant or unsafe driving. OBJECTIVE: Using machine learning we investigated if including multiple simulator-derived parameters, rather than the SDLP alone would provide a more accurate assessment of the effect of substances affecting driving performance. We specifically analysed the effects of alcohol and alprazolam. METHODS: The data used in the present study were collected during a previous study on driving effects of alcohol and alprazolam in 24 healthy subjects (12 M, 12 F, mean age 26 years, range 20-43 years). Various driving features, such as speed and steering variations, were quantified and the influence of administration of alcohol or alprazolam was assessed to assist in designing a predictive model for abnormal driving behaviour. RESULTS: Adding additional features besides the SDLP increased the model performance for prediction of drug-induced abnormal driving behaviour (from an accuracy of 65 %-83 % after alprazolam intake and from 50 % to 76 % after alcohol ingestion). Driving behaviour influenced by alcohol and alprazolam was characterised by different feature importance, indicating that the two interventions influenced driving behaviour in a different way. CONCLUSION: Machine learning using multiple driving features in addition to the state-of-the-art SDLP improves the assessment of drug-induced abnormal driving behaviour. The created models may facilitate quantitative description of abnormal driving behaviour in the development and application of psychopharmacological medicines. Our models require further validation using similar and unknown interventions.


Subject(s)
Accidents, Traffic/prevention & control , Driving Under the Influence , Machine Learning , Adult , Computer Simulation , Female , Humans , Male , Psychomotor Performance/drug effects , Young Adult
3.
Pharmacol Rev ; 72(4): 899-909, 2020 10.
Article in English | MEDLINE | ID: mdl-32958524

ABSTRACT

Novel digital endpoints gathered via wearables, small devices, or algorithms hold great promise for clinical trials. However, implementation has been slow because of a lack of guidelines regarding the validation process of these new measurements. In this paper, we propose a pragmatic approach toward selection and fit-for-purpose validation of digital endpoints. Measurements should be value-based, meaning the measurements should directly measure or be associated with meaningful outcomes for patients. Devices should be assessed regarding technological validity. Most importantly, a rigorous clinical validation process should appraise the tolerability, difference between patients and controls, repeatability, detection of clinical events, and correlation with traditional endpoints. When technically and clinically fit-for-purpose, case building in interventional clinical trials starts to generate evidence regarding the response to new or existing health-care interventions. This process may lead to the digital endpoint replacing traditional endpoints, such as clinical rating scales or questionnaires in clinical trials. We recommend initiating more data-sharing collaborations to prevent unnecessary duplication of research and integration of value-based measurements in clinical care to enhance acceptance by health-care professionals. Finally, we invite researchers and regulators to adopt this approach to ensure a timely implementation of digital measurements and value-based thinking in clinical trial design and health care. SIGNIFICANCE STATEMENT: Novel digital endpoints are often cited as promising for the clinical trial of the future. However, clear validation guidelines are lacking in the literature. This paper contains pragmatic criteria for the selection, technical validation, and clinical validation of novel digital endpoints and provides recommendations for future work and collaboration.


Subject(s)
Clinical Trials as Topic/methods , Biomarkers/analysis , Biomarkers/metabolism , Endpoint Determination/methods , Humans , Reproducibility of Results
4.
Curr Med Chem ; 8(12): 1419-36, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11562275

ABSTRACT

Farnesyl protein transferase (FPT) inhibition is an interesting and promising approach to non-cytotoxic anticancer therapy. Research in this area has resulted in several orally active compounds that are currently in clinical evaluation. This review focuses on FPT inhibitors in clinical trials and concentrates on the benzocycloheptapyridine class, with details on the discovery and development of SCH 66336, currently in Phase II clinical trials.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Neoplasms/therapy , Piperidines/therapeutic use , Pyridines/therapeutic use , Animals , Binding Sites , Clinical Trials as Topic , Farnesyltranstransferase , Humans , Molecular Structure , Neoplasms/enzymology , Protein Conformation
5.
Exp Cell Res ; 262(1): 17-27, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11120601

ABSTRACT

SCH 66336 is a potent farnesyl transferase inhibitor (FTI) in clinical development. It efficiently prevents the membrane association of H-ras, but not K- or N-ras. Yet, in soft agar, it reverts the anchorage-independent growth of human tumor cell lines (hTCLs) harboring H-ras, K-ras, and N-ras mutations, implying that blocking farnesylation of proteins besides ras may be responsible for this effect. Experiments show that SCH 66336 altered the cell cycle distribution of sensitive human tumor cells in two distinct ways. Most sensitive hTCLs accumulated in the G(2)-->M phase after the FTI treatment, but those with an activated H-ras accumulated in G(1) phase, suggesting that the biological effects induced by FTIs in cells with an activated H-ras are distinct from other sensitive cells. A careful genotypic comparison of the hTCLs revealed that those cells with wild-type p53 are especially sensitive to the FTIs. In these cells p53 and its downstream target gene p21(Cip1) are induced after treatment with SCH 66336 for 24 h. These data suggest that cell cycle effects, either G(1) or G(2)-->M accumulation, and p53 status are important for mediating the effects of FTIs on tumor cells.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Cell Cycle/drug effects , 3T3 Cells , Animals , Cell Division/drug effects , Cell Membrane/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase , G1 Phase , G2 Phase , Humans , K562 Cells , Kinetics , Mice , Mitosis , Molecular Structure , Oncogene Protein p21(ras)/metabolism , Piperidines/chemistry , Piperidines/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
6.
IDrugs ; 4(12): 1382-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-15931570

ABSTRACT

The inhibition of farnesyl protein transferase (FPT) has been the subject of intense research over the last few years and offers a promising approach to non-cytotoxic, anticancer therapy. Several FPT inhibitors are currently in clinical trials for anticancer therapy. This paper reviews the patent applications that appeared from January to July 2001. A brief summary of the compounds in clinical evaluation is also given.

8.
Mol Carcinog ; 27(1): 24-33, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10642434

ABSTRACT

The Tg.AC mouse carries an activated v-Ha-ras oncogene fused to an embryonic zeta-globin promoter and develops cutaneous papillomas in response to specific chemicals, full thickness wounding, and ultraviolet radiation. Papilloma development in these mice has been suggested to be dependent upon activation of ras transgene expression, thus providing a potential model for studying ras-inhibitory compounds. Farnesyl transferase inhibitors (FTIs) prevent a critical posttranslational modification step necessary for activation of ras proteins. Our studies demonstrated that a tricyclic FTI (SCH 56582) applied directly to the skin of homozygous Tg.AC mice 1 h prior to administration of the tumor promoter TPA decreased tumor multiplicity compared to TPA-only controls. In addition, a reduction of TPA-induced tumor development was seen in similarly treated hemizygous Tg.AC mice either on an FVB/N strain background or 50% C57BL/6. Histological examination of skin from Tg. AC(+/-):FVB/N mice revealed no differences with respect to 12-O-tetradecamoylpharbol-13-acetate (TPA)-mediated hyperplasia. Keratinocytes isolated from treated and control skin were assayed for ras transgene expression by reverse transcription-polymerase chain reaction, and expression was detected in both TPA- and FTI+TPA-treated tissue, although the appearance of transgene positive pre-papillomas was observed only in histological sections taken 21 d after the first treatment. In summary, we have used a regimen of topical application of an FTI (SCH 56582) to suppress TPA-mediated papillomagenesis in v-Ha-ras transgenic Tg.AC mice. These studies demonstrate that TPA-induced epidermal hyperplasia is a ras-independent process, while papilloma development in response to TPA treatment requires the function of activated ras.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Benzazepines/pharmacology , Enzyme Inhibitors/pharmacology , Genes, ras , Papilloma/prevention & control , Skin Neoplasms/prevention & control , Skin/pathology , Tetradecanoylphorbol Acetate/toxicity , Animals , Crosses, Genetic , Farnesyltranstransferase , Female , Globins/genetics , Homozygote , Hyperplasia , Keratinocytes/drug effects , Keratinocytes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Papilloma/chemically induced , Papilloma/genetics , Promoter Regions, Genetic , Skin/drug effects , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Time Factors
9.
J Med Chem ; 42(14): 2651-61, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10411485

ABSTRACT

Farnesyl protein transferase (FPT) is a promising target for the development of cancer chemotherapeutics because it is responsible for the farnesylation of oncogenic p21 Ras proteins which are found in nearly 30% of all human cancers and necessary for cellular development and growth. The recent discovery and progression to phase II clinical trials of trihalobenzocycloheptapyridine Sch-66336 as a potent inhibitor of FPT with oral, in vivo efficacy in mice have spawned extensive structure-activity relationship studies (SAR) of this class of compounds. Of the many trihalobenzocycloheptapyridine analogues prepared, we have identified several which inhibit FPT and cellular proliferation at single-digit nanomolar concentrations and which have good pharmacokinetic properties in mice.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Piperidines/chemical synthesis , Pyridines/chemical synthesis , Sulfonamides/chemical synthesis , Sulfonylurea Compounds/chemical synthesis , Administration, Oral , Animals , Biological Availability , COS Cells , Cell Division/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Haplorhini , Mice , Mice, Nude , Piperidines/chemistry , Piperidines/pharmacokinetics , Protein Prenylation , Proto-Oncogene Proteins p21(ras)/metabolism , Pyridines/chemistry , Pyridines/pharmacokinetics , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Sulfonylurea Compounds/chemistry , Sulfonylurea Compounds/pharmacokinetics
10.
Bioorg Med Chem ; 7(5): 861-7, 1999 May.
Article in English | MEDLINE | ID: mdl-10400339

ABSTRACT

Introduction of bromine at the 10-position of 3-bromo-8-chloro-benzocycloheptapyridine analogues of type 3 results in formation of atropisomeric compounds of type (+/-)-1 and (+/-)-2 that are easily separable at room temperature on a ChiralPak AD column providing pure atropisomers, (+)-1, (-)-1, and (+)-2, (-)-2, respectively. Evaluation of the FPT activity of these atropisomers revealed that compounds (+)-1 and (+)-2 were more potent in the FPT enzyme and cellular assay than their (-)-isomer counterparts. Compounds (+)-1 and (+)-2 were found to inhibit FPT processing in COS cells at low micro molar range. They were also found to have excellent cellular antitumor activity. Evaluation of compound (+)-1 and (+)-2 in DLD-tumor model in nude mice revealed that they were efficacious, inhibiting tumor growth by 55 and 63% at 50 mpk, respectively.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Pyridines/chemical synthesis , Pyridines/pharmacology , Pyridines/pharmacokinetics , Animals , COS Cells , Dose-Response Relationship, Drug , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Nude , Models, Chemical , Time Factors , Tumor Cells, Cultured
11.
J Med Chem ; 42(12): 2125-35, 1999 Jun 17.
Article in English | MEDLINE | ID: mdl-10377218

ABSTRACT

Crystallographic and thermodynamic studies of farnesyl protein transferase (FPT) complexed with novel tricyclic inhibitors provide insights into the observed SAR for this unique class of nonpeptidic FPT inhibitors. The crystallographic structures reveal a binding pattern conserved across the mono-, di-, and trihalogen series. In the complexes, the tricycle spans the FPT active site cavity and interacts with both protein atoms and the isoprenoid portion of bound farnesyl diphosphate. An amide carbonyl, common to the tricyclic compounds described here, participates in a water-mediated hydrogen bond to the protein backbone. Ten high-resolution crystal structures of inhibitors complexed with FPT are reported. Included are crystallographic data for FPT complexed with SCH 66336, a compound currently undergoing clinical trials as an anticancer agent (SCH 66336, 4-[2-[4-(3,10-dibromo-8-chloro-6,11-dihydro-5H-benzo[5, 6]cyclohepta[1, 2-b]pyridin-11-yl)-1-piperidinyl]-2-oxoethyl]-1-piperidinecarbo xamide ). Thermodynamic binding parameters show favorable enthalpies of complex formation and small net entropic contributions as observed for 4-[2-[4-(3,10-dibromo-8-chloro-6,11-dihydro-11H-benzo[5, 6]cyclohepta[1, 2-b]pyridin-11-ylidene)-1-piperidinyl]-2-oxoethyl]pyridine N-oxide where DeltaH degrees bind = -12.5 kcal/mol and TDeltaS degrees bind = -1.5 kcal/mol.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/chemistry , Cyclic N-Oxides/chemistry , Enzyme Inhibitors/chemistry , Heterocyclic Compounds, 3-Ring/chemistry , Piperidines/chemistry , Protein Prenylation , Pyridines/chemistry , Binding Sites , Calorimetry , Crystallography, X-Ray , Hydrogen Bonding , Models, Molecular , Thermodynamics
12.
Cancer Chemother Pharmacol ; 43(1): 50-8, 1999.
Article in English | MEDLINE | ID: mdl-9923541

ABSTRACT

The products of the Ha-, Ki-, and N-ras proto-oncogenes comprise a family of 21 kDa guanine nucleotide-binding proteins which play a crucial role in growth factor signal transduction and in the control of cellular proliferation and differentiation. Activating mutations in the ras oncogenes occur in a wide variety of human tumors. Ras proteins undergo a series of posttranslational processing events. The first modification is addition of the 15-carbon isoprene, farnesyl, to a Cys residue near the carboxy-terminus of Ras. Prenylation allows the Ras oncoprotein to localize to the plasma membrane where it can initiate downstream signalling events leading to cellular transformation. Inhibitors of the enzyme which catalyzes this step, farnesyl protein transferase (FPT), are a potential class of novel anticancer drugs which interfere with Ras function. SCH 59228 is a tricyclic FPT inhibitor which inhibits the farnesylation of purified Ha-Ras with an IC50 of 95 nM and blocks the processing of Ha-Ras in Cos cells with an IC50 of 0.6 microM. SCH 59228 has favorable pharmacokinetic properties upon oral dosing in nude mice. The in vivo efficacy of SCH 59228 was evaluated using a panel of tumor models grown in nude mice. These included several rodent fibroblast lines expressing mutationally-activated (val12) forms of the Ha-Ras oncogene. In some cases, these proteins contain their native C-terminal sequence (CVLS) which directs farnesylation. In one model, the C-terminal sequence was altered to CVLL, making the expressed protein a substrate for a distinct prenyl transferase, geranylgeranyl protein transferase-1. When dosed orally at 10 and 50 mg/kg (four times a day, 7 days a week) SCH 59228 significantly inhibited tumor growth of cells expressing farnesylated Ha-Ras in a dose-dependent manner; over 90% growth inhibition was observed at the 50 mg/kg dose. Tumor growth of cells expressing the geranylgeranylated form of Ha-Ras was less potently inhibited. Growth of tumors derived from a rodent fibroblast line expressing activated Ki-Ras containing its native C-terminal sequence (CVIM), which preferentially directs farnesylation, was also inhibited by SCH 59228. Inhibition in the Ki-Ras model was less than that observed in the Ha-Ras model. In contrast, tumors derived from cells transformed with the mos oncogene were not significantly inhibited even at the highest dose level. SCH 59228 also significantly and dose-dependently inhibited the growth of human colon adenocarcinoma DLD-1 xenografts (which express activated Ki-ras). These results indicate that SCH 59228 possesses in vivo antitumor activity upon oral dosing in tumor models expressing activated ras oncogenes. This is the first report of oral antitumor activity with an FPT inhibitor. These results are discussed in light of recent observations on alternative prenylation of some Ras isoforms.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Colonic Neoplasms/pathology , Cyclic N-Oxides/pharmacology , Enzyme Inhibitors/pharmacology , Genes, ras , Piperazines/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Biological Availability , Cell Division/drug effects , Cell Line, Transformed , Colonic Neoplasms/drug therapy , Cyclic N-Oxides/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Fibroblasts , Genes, mos , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Piperazines/pharmacokinetics , Transfection
13.
Org Lett ; 1(9): 1371-3, 1999 Nov 04.
Article in English | MEDLINE | ID: mdl-10825985

ABSTRACT

[formula: see text] Synthesis of C-11 methyl-substituted benzocycloheptylpyridine tricyclic compounds has been achieved via two different methods. Methylation of C-11 has been effected by treatment of amine 4 with BuLi followed by Mel quenching. In a similar procedure, introduction of a C-11 substituent with concomitant rearrangement of the exocyclic double bond has been carried out. Potent farnesyl protein transferase inhibitors have been synthesized using the above methodologies.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Pyridines/chemical synthesis , Enzyme Inhibitors/pharmacology , Pyridines/pharmacology
14.
Biochemistry ; 37(45): 15631-7, 1998 Nov 10.
Article in English | MEDLINE | ID: mdl-9843367

ABSTRACT

Mutated, tumorigenic Ras is present in a variety of human tumors. Compounds that inhibit tumorigenic Ras function may be useful in the treatment of Ras-related tumors. The interaction of a novel GDP exchange inhibitor (SCH-54292) with the Ras-GDP protein was studied by NMR spectroscopy. The binding of the inhibitor to the Ras protein was enhanced at low Mg2+ concentrations, which enabled the preparation of a stable complex for NMR study. To understand the enhanced inhibitor binding and the increased GDP dissociation rates of the Ras protein, the conformational changes of the Ras protein at low Mg2+ concentrations was investigated using two-dimensional 1H-15N HSQC experiments. The Ras protein existed in two conformations in slow exchange on the NMR time scale under such conditions. The conformational changes mainly occurred in the GDP binding pocket, in the switch I and the switch II regions, and were reversible. The Ras protein resumed its regular conformation after an excess amount of Mg2+ was added. A model of the inhibitor in complex with the Ras-GDP protein was derived from intra- and intermolecular NOE distance constraints, and revealed that the inhibitor bound to the critical switch II region of the Ras protein.


Subject(s)
Glucosides/metabolism , Guanosine Diphosphate/metabolism , Proteins/antagonists & inhibitors , Sulfonamides/metabolism , ras Proteins/metabolism , Amino Acid Sequence , Binding Sites , Computer Simulation , Glucosides/chemistry , Guanine Nucleotide Exchange Factors , Humans , Macromolecular Substances , Magnesium/metabolism , Models, Molecular , Molecular Sequence Data , Monte Carlo Method , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Proteins/chemistry , Sulfonamides/chemistry , ras Guanine Nucleotide Exchange Factors
15.
Cancer Res ; 58(21): 4947-56, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9810004

ABSTRACT

We have been developing a series of nonpeptidic, small molecule farnesyl protein transferase inhibitors that share a common tricyclic nucleus and compete with peptide/protein substrates for binding to farnesyl protein transferase. Here, we report on pharmacological and in vivo studies with SCH 66336, a lead compound in this structural class. SCH 66336 potently inhibits Ha-Ras processing in whole cells and blocks the transformed growth properties of fibroblasts and human tumor cell lines expressing activated Ki-Ras proteins. The anchorage-independent growth of many human tumor lines that lack an activated ras oncogene is also blocked by treatment with SCH 66336. In mouse, rat, and monkey systems, SCH 66336 has excellent oral bioavailability and pharmacokinetic properties. In the nude mouse, SCH 66336 demonstrated potent oral activity in a wide array of human tumor xenograft models including tumors of colon, lung, pancreas, prostate, and urinary bladder origin. Enhanced in vivo efficacy was observed when SCH 66336 was combined with various cytotoxic agents (cyclophosphamide, 5-fluorouracil, and vincristine). In a Ha-Ras transgenic mouse model, prophylactic treatment with SCH 66336 delayed tumor onset, reduced the average number of tumors/mouse, and reduced the average tumor weight/animal. In a therapeutic mode in which gavage treatment was initiated after the transgenic mice had developed palpable tumors, significant tumor regression was induced by SCH 66336 in a dose-dependent fashion. This was associated with increased apoptosis and decreased DNA synthesis in tumors of animals treated with SCH 66336. Enhanced efficacy was also observed in this model when SCH 66336 was combined with cyclophosphamide. SCH 66336 is presently being evaluated in Phase I clinical trials.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Genes, ras/physiology , Neoplasms, Experimental/drug therapy , Piperidines/pharmacology , Pyridines/pharmacology , 3T3 Cells , Administration, Oral , Animals , Bromodeoxyuridine/metabolism , Cell Division/drug effects , Dose-Response Relationship, Drug , Female , Humans , Macaca fascicularis , Male , Mice , Neoplasm Transplantation , Rats , Transplantation, Heterologous
16.
J Med Chem ; 41(10): 1561-7, 1998 May 07.
Article in English | MEDLINE | ID: mdl-9572881

ABSTRACT

We previously reported compound 1 as a potent farnesyl protein transferase (FPT) inhibitor that exhibited reasonable pharmacokinetic stability and showed moderate in vivo activity against a variety of tumor cell lines. The analogous C-11 single compound, pyridylacetamide 2, was found to be more potent than 1 in FPT inhibition. Further studies showed that modification of the ethano bridge of the tricyclic ring system by conversion into a double bond with concomitant introduction of a single bond at C-11 piperidine resulted in compound 3 that had superior FPT activity and pharmacokinetic stability. Compound 4, a 5-bromo-substituted analogue of 3, showed improved FPT activity, had good cellular activity, and demonstrated a remarkably improved pharmacokinetic profile with AUC of 84.9 and t1/2 of 82 min. Compound4 inhibited the growth of solid tumor in DLD-1 model by 70% at 50 mpk and 52% at 10 mpk.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents , Cyclic N-Oxides , Enzyme Inhibitors , Pyridines , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biological Availability , Cyclic N-Oxides/administration & dosage , Cyclic N-Oxides/chemical synthesis , Cyclic N-Oxides/pharmacokinetics , Cyclic N-Oxides/pharmacology , Drug Screening Assays, Antitumor , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Oncogene Protein p21(ras)/antagonists & inhibitors , Pyridines/administration & dosage , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Pyridines/pharmacology , Tumor Cells, Cultured
17.
J Med Chem ; 41(6): 877-93, 1998 Mar 12.
Article in English | MEDLINE | ID: mdl-9526562

ABSTRACT

The synthesis of a variety of novel 4-amido, 4-carbamoyl and 4-carboxamido derivatives of 1-(8-chloro-6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridin-11-yl) piperazine to explore the SAR of this series of FPT inhibitors is described. This resulted in the synthesis of the 4- and 3-pyridylacetyl analogues 45a and 50a, respectively, both of which were orally active but were found to be rapidly metabolized in vivo. Identification of the principal metabolites led to the synthesis of a variety of new compounds that would be less readily metabolized, the most interesting of which were the 3- and 4-pyridylacetyl N-oxides 80a and 83a. Novel replacements for the pyridylacetyl moiety were also sought, and this resulted in the discovery of the 4-N-methyl and 4-N-carboxamidopiperidinylacetyl derivatives 135a and 160a, respectively. All of these derivatives exhibited greatly improved pharmacokinetics. The synthesis of the corresponding 3-bromo analogues resulted in the discovery of the 4-pyridylacetyl N-oxides 83b (+/-) and 85b [11S(-)] and the 4-carboxamidopiperidinylacetamido derivative 160b (+/-), all of which exhibited potent FPT inhibition in vitro. All three showed excellent oral bioavailability in vivo in nude mice and cynomolgus monkeys and exhibited excellent antitumor efficacy against a series of tumor cell lines when dosed orally in nude mice.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Cyclic N-Oxides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Piperazines/chemical synthesis , Piperidines/chemical synthesis , 3T3 Cells , Administration, Oral , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biological Availability , COS Cells , Cell Line, Transformed , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/metabolism , Cyclic N-Oxides/pharmacokinetics , Drug Screening Assays, Antitumor , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Genes, ras , Macaca fascicularis , Mice , Mice, Nude , Neoplasm Transplantation , Piperazines/chemistry , Piperazines/metabolism , Piperazines/pharmacokinetics , Piperidines/chemistry , Piperidines/metabolism , Piperidines/pharmacokinetics , Structure-Activity Relationship
18.
Bioorg Med Chem ; 5(5): 817-20, 1997 May.
Article in English | MEDLINE | ID: mdl-9208093

ABSTRACT

MS based methodology employing electrospray ionization (ESI) is described for the detection of ternary complexes in which SCH 54292 or SCH 54341 and GDP are noncovalently bound to oncogenic ras protein. The observed molecular weights of 19,816 and 19,570 Da confirmed the presence of noncovalent complexes of ras-GDP-SCH 54292 and ras-GDP-SCH 54341, respectively. We have also performed selective chemical modification of lysine residues of the ras protein complex followed by enzymatic digestion and on-line LC-ESI MS peptide mapping to determine protein-drug binding topography. There was a good correlation between nucleotide exchange inhibition as determined by the enzyme assay and evidence of complex formation as determined by MS.


Subject(s)
ras Proteins/antagonists & inhibitors , ras Proteins/chemistry , Amino Acid Sequence , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Drug Evaluation, Preclinical , Glucosides/chemistry , Glucosides/pharmacology , Guanine Nucleotide Exchange Factors , In Vitro Techniques , Macromolecular Substances , Mass Spectrometry/methods , Molecular Sequence Data , Molecular Weight , Peptide Mapping , Proteins/antagonists & inhibitors , Proteins/chemistry , Sulfonamides/chemistry , Sulfonamides/pharmacology , ras Guanine Nucleotide Exchange Factors , ras Proteins/genetics
19.
Bioorg Med Chem ; 5(1): 93-9, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9043661

ABSTRACT

Ras farnesylation by farnesyl protein transferase (FPT) is an intracellular event that facilitates the membrane association of the ras protein and is involved in the signal transduction process. FPT inhibition could be a novel, noncytotoxic method of treating ras dependent tumor growth. We report here three structural classes of 8-chlorobenzocycloheptapyridines as novel, nonpeptidic, nonsulfhydryl FPT inhibitors having antitumor activity in mice when dosed orally. We discuss structural and conformational aspects of these compounds in relation to biological activities as well as a comparison to the conformation of a bound tetrapeptide FPT inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Benzazepines/chemistry , Benzazepines/pharmacokinetics , Benzazepines/pharmacology , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Piperidines/chemistry , Piperidines/pharmacokinetics , Piperidines/pharmacology , Pyridines/chemistry , Pyridines/pharmacokinetics , Pyridines/pharmacology , Tumor Cells, Cultured
20.
Bioorg Med Chem ; 5(1): 101-13, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9043662

ABSTRACT

A comprehensive structure-activity relationship (SAR) study of novel tricyclic amides has been undertaken. The discovery of compounds that are potent FPT inhibitors in the nanomolar range has been achieved. These compounds are nonpeptidic and do not contain sulfhydryl groups. They selectively inhibit farnesyl protein transferase (FPT) and not geranylgeranyl protein transferase-1 (GGPT-1). They also inhibit H-Ras processing in Cos monkey kidney cells.


Subject(s)
Alkyl and Aryl Transferases , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Transferases/antagonists & inhibitors , Magnetic Resonance Spectroscopy , Spectrometry, Mass, Fast Atom Bombardment , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...