Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
JCI Insight ; 3(4)2018 02 22.
Article in English | MEDLINE | ID: mdl-29467321

ABSTRACT

BACKGROUND: Constitutive activation of ERK1/2 occurs in various cancers, and its reactivation is a well-described resistance mechanism to MAPK inhibitors. ERK inhibitors may overcome the limitations of MAPK inhibitor blockade. The dual mechanism inhibitor SCH772984 has shown promising preclinical activity across various BRAFV600/RAS-mutant cancer cell lines and human cancer xenografts. METHODS: We have developed an orally bioavailable ERK inhibitor, MK-8353; conducted preclinical studies to demonstrate activity, pharmacodynamic endpoints, dosing, and schedule; completed a study in healthy volunteers (P07652); and subsequently performed a phase I clinical trial in patients with advanced solid tumors (MK-8353-001). In the P07652 study, MK-8353 was administered as a single dose in 10- to 400-mg dose cohorts, whereas in the MK-8353-001 study, MK-8353 was administered in 100- to 800-mg dose cohorts orally twice daily. Safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity were analyzed. RESULTS: MK-8353 exhibited comparable potency with SCH772984 across various preclinical cancer models. Forty-eight patients were enrolled in the P07652 study, and twenty-six patients were enrolled in the MK-8353-001 study. Adverse events included diarrhea (44%), fatigue (40%), nausea (32%), and rash (28%). Dose-limiting toxicity was observed in the 400-mg and 800-mg dose cohorts. Sufficient exposure to MK-8353 was noted that correlated with biological activity in preclinical data. Three of fifteen patients evaluable for treatment response in the MK-8353-001 study had partial response, all with BRAFV600-mutant melanomas. CONCLUSION: MK-8353 was well tolerated up to 400 mg twice daily and exhibited antitumor activity in patients with BRAFV600-mutant melanoma. However, antitumor activity was not particularly correlated with pharmacodynamic parameters. TRIAL REGISTRATION: ClinicalTrials.gov NCT01358331. FUNDING: Merck Sharp & Dohme Corp., a subsidiary of Merck & Co. Inc., and NIH (P01 CA168585 and R35 CA197633).


Subject(s)
Indazoles/pharmacology , MAP Kinase Signaling System/drug effects , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrrolidines/pharmacology , Triazoles/pharmacology , Administration, Oral , Adult , Animals , Biological Availability , Cell Line, Tumor , Diarrhea/chemically induced , Diarrhea/epidemiology , Dogs , Dose-Response Relationship, Drug , Drug Eruptions/epidemiology , Drug Eruptions/etiology , Drug Evaluation, Preclinical , Fatigue/chemically induced , Fatigue/epidemiology , Female , Humans , Indazoles/therapeutic use , Male , Maximum Tolerated Dose , Mice , Middle Aged , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Nausea/chemically induced , Nausea/epidemiology , Neoplasm Staging , Neoplasms/genetics , Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Pyrrolidines/therapeutic use , Rats , Triazoles/therapeutic use , Xenograft Model Antitumor Assays , Young Adult
2.
ACS Med Chem Lett ; 7(3): 324-9, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26985323

ABSTRACT

A new subseries of substituted piperidines as p53-HDM2 inhibitors exemplified by 21 has been developed from the initial lead 1. Research focused on optimization of a crucial HDM2 Trp23-ligand interaction led to the identification of 2-(trifluoromethyl)thiophene as the preferred moiety. Further investigation of the Leu26 pocket resulted in potent, novel substituted piperidine inhibitors of the HDM2-p53 interaction that demonstrated tumor regression in several human cancer xenograft models in mice. The structure of HDM2 in complex with inhibitors 3, 10, and 21 is described.

3.
Cancer Discov ; 3(7): 742-50, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23614898

ABSTRACT

The high frequency of activating RAS or BRAF mutations in cancer provides strong rationale for targeting the mitogen-activated protein kinase (MAPK) pathway. Selective BRAF and MAP-ERK kinase (MEK) inhibitors have shown clinical efficacy in patients with melanoma. However, the majority of responses are transient, and resistance is often associated with pathway reactivation of the extracellular signal-regulated kinase (ERK) signaling pathway. Here, we describe the identification and characterization of SCH772984, a novel and selective inhibitor of ERK1/2 that displays behaviors of both type I and type II kinase inhibitors. SCH772984 has nanomolar cellular potency in tumor cells with mutations in BRAF, NRAS, or KRAS and induces tumor regressions in xenograft models at tolerated doses. Importantly, SCH772984 effectively inhibited MAPK signaling and cell proliferation in BRAF or MEK inhibitor-resistant models as well as in tumor cells resistant to concurrent treatment with BRAF and MEK inhibitors. These data support the clinical development of ERK inhibitors for tumors refractory to MAPK inhibitors.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/genetics , MAP Kinase Kinase Kinases/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mutation , Neoplasms/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Signal Transduction/drug effects
4.
Proc Natl Acad Sci U S A ; 109(41): 16666-71, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-23012407

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare, fatal, segmental premature aging syndrome caused by a mutation in LMNA that produces the farnesylated aberrant lamin A protein, progerin. This multisystem disorder causes failure to thrive and accelerated atherosclerosis leading to early death. Farnesyltransferase inhibitors have ameliorated disease phenotypes in preclinical studies. Twenty-five patients with HGPS received the farnesyltransferase inhibitor lonafarnib for a minimum of 2 y. Primary outcome success was predefined as a 50% increase over pretherapy in estimated annual rate of weight gain, or change from pretherapy weight loss to statistically significant on-study weight gain. Nine patients experienced a ≥50% increase, six experienced a ≥50% decrease, and 10 remained stable with respect to rate of weight gain. Secondary outcomes included decreases in arterial pulse wave velocity and carotid artery echodensity and increases in skeletal rigidity and sensorineural hearing within patient subgroups. All patients improved in one or more of these outcomes. Results from this clinical treatment trial for children with HGPS provide preliminary evidence that lonafarnib may improve vascular stiffness, bone structure, and audiological status.


Subject(s)
Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase/antagonists & inhibitors , Piperidines/therapeutic use , Progeria/drug therapy , Pyridines/therapeutic use , Adolescent , Carotid Arteries/drug effects , Carotid Arteries/pathology , Child , Child, Preschool , Diarrhea/chemically induced , Dose-Response Relationship, Drug , Drug Administration Schedule , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Farnesyltranstransferase/metabolism , Fatigue/chemically induced , Female , Humans , Male , Piperidines/adverse effects , Piperidines/pharmacokinetics , Progeria/pathology , Progeria/physiopathology , Pulse Wave Analysis , Pyridines/adverse effects , Pyridines/pharmacokinetics , Treatment Outcome , Vomiting/chemically induced , Weight Gain/drug effects
5.
Am J Med Genet C Semin Med Genet ; 157C(2): 136-46, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21495172

ABSTRACT

The RASopathies, one of the largest groups of multiple congenital anomaly syndromes known, are caused by germline mutations in various genes encoding components of the Ras/mitogen-activated protein kinase (MAPK) pathway. The RASopathies have many overlapping characteristics, including craniofacial manifestations, cardiac malformations, cutaneous, musculoskeletal, gastrointestinal, and ocular abnormalities, neurocognitive impairment, hypotonia, and an increased risk of developing cancer. Costello syndrome (CS) and cardio-facio-cutaneous (CFC) syndrome are two of the more rare RASopathies. CS is caused by activating mutations in HRAS, and CFC is caused by dysregulation of signaling in the Ras/MAPK pathway due to mutations in BRAF, MEK1, or MEK2. The Ras/MAPK pathway, which has been well-studied in cancer, is an attractive target for inhibition in the treatment of various malignancies utilizing small molecule therapeutics that specifically inhibit the pathway. With many inhibitors of the Ras/MAPK pathway in clinical trials, the notion of using these molecules to ameliorate developmental defects in CS and CFC is under consideration. CS and CFC, like other syndromes in their class, have a progressive phenotype and may be amenable to inhibition or normalization of signaling.


Subject(s)
Clinical Trials as Topic/methods , Costello Syndrome/genetics , Neurofibromatosis 1/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Research Design , Signal Transduction/genetics , Costello Syndrome/drug therapy , Ectodermal Dysplasia/drug therapy , Ectodermal Dysplasia/genetics , Facies , Failure to Thrive/drug therapy , Failure to Thrive/genetics , Farnesyltranstransferase/antagonists & inhibitors , Heart Defects, Congenital/drug therapy , Heart Defects, Congenital/genetics , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , Public-Private Sector Partnerships , raf Kinases/antagonists & inhibitors
6.
J Biol Chem ; 285(14): 10198-212, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20124408

ABSTRACT

Abrogation of p53 function occurs in almost all human cancers, with more than 50% of cancers harboring inactivating mutations in p53 itself. Mutation of p53 is indicative of highly aggressive cancers and poor prognosis. The vast majority of mutations in p53 occur in its core DNA binding domain (DBD) and result in inactivation of p53 by reducing its thermodynamic stability at physiological temperature. Here, we report a small molecule, SCH529074, that binds specifically to the p53 DBD in a saturable manner with an affinity of 1-2 microm. Binding restores wild type function to many oncogenic mutant forms of p53. This small molecule reactivates mutant p53 by acting as a chaperone, in a manner similar to that previously reported for the peptide CDB3. Binding of SCH529074 to the p53 DBD is specifically displaced by an oligonucleotide with a sequence derived from the p53-response element. In addition to reactivating mutant p53, SCH529074 binding inhibits ubiquitination of p53 by HDM2. We have also developed a novel variant of p53 by changing a single amino acid in the core domain of p53 (N268R), which abolishes binding of SCH529074. This amino acid change also inhibits HDM2-mediated ubiquitination of p53. Our novel findings indicate that through its interaction with p53 DBD, SCH529074 restores DNA binding activity to mutant p53 and inhibits HDM2-mediated ubiquitination.


Subject(s)
Cell Proliferation/drug effects , DNA/metabolism , Mutation/genetics , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/metabolism , Quinazolines/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitination , Animals , Apoptosis/drug effects , Binding Sites , Blotting, Western , Chromatin Immunoprecipitation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/prevention & control , DNA/chemistry , DNA/genetics , Female , Humans , Immunoprecipitation , Mice , Mice, Nude , Molecular Chaperones , Piperazines/isolation & purification , Protein Binding , Proto-Oncogene Proteins c-mdm2/genetics , Quinazolines/isolation & purification , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
7.
Mol Cancer Ther ; 9(2): 410-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20124453

ABSTRACT

The insulin-like growth factor-I receptor (IGF-IR) and its ligands (IGF-I and IGF-II) have been implicated in the growth, survival, and metastasis of a broad range of malignancies including pediatric tumors. Blocking the IGF-IR action is a potential cancer treatment. A fully human neutralizing monoclonal antibody, SCH 717454 (19D12, robatumumab), specific to IGF-IR, has shown potent antitumor effects in ovarian cancer in vitro and in vivo. In this study, SCH 717454 was evaluated in several pediatric solid tumors including neuroblastoma, osteosarcoma, and rhabdomyosarcoma. SCH 717454 is shown here to downregulate IGF-IR as well as inhibit IGF-IR and insulin receptor substrate-1 phosphorylation in pediatric tumor cells. IGF-IR and insulin receptor substrate-1 phosphorylation in the tumor cells. In vivo, SCH 717454 exhibits activity as a single agent and significantly inhibited growth of neuroblastoma, osteosarcoma, and rhabdomyosarcoma tumor xenografts. Combination of SCH 717454 with cisplatin or cyclophosphamide enhanced both the degree and the duration of the in vivo antitumor activity compared with single-agent treatments. Furthermore, SCH 717454 treatment markedly reduced Ki-67 expression and blood vessel formation in tumor xenografts, showing that the in vivo activity is derived from its inhibition of tumor cell proliferation and angiogenesis activity.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Receptor, IGF Type 1/immunology , Animals , Cell Line, Tumor , Female , Humans , Insulin-Like Growth Factor I/immunology , Ki-67 Antigen/biosynthesis , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic , Phosphorylation
8.
Bioorg Med Chem Lett ; 20(3): 1134-6, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20056542

ABSTRACT

The discovery of C-linked imidazole azaheptapyridine bridgehead FPT inhibitors is described. This novel class of compounds are sub nM FPT enzyme inhibitors with potent cellular inhibitory activities. This series also has reduced hERG activity versus previous N-linked imidazole series. X-ray of compound 10a bound to FTase revealed strong interaction between bridgehead imidazole 3N with catalytic zinc atom.


Subject(s)
Drug Discovery/methods , Farnesyltranstransferase/antagonists & inhibitors , Imidazoles/chemistry , Pyridines/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Farnesyltranstransferase/metabolism , Humans , Imidazoles/metabolism , Imidazoles/pharmacology , Pyridines/metabolism , Pyridines/pharmacology
9.
Int J Cancer ; 125(11): 2711-20, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19530253

ABSTRACT

Lonafarnib is a potent, selective farnesyltransferase inhibitor (FTI) undergoing clinical studies for the treatment of solid tumors and hematological malignancies. Preclinically, a number of FTIs, including lonafarnib, interact with taxanes to inhibit cancer cell growth in an additive/synergistic manner. These observations provided rationale for investigating the effects of combining lonafarnib and docetaxel on preclinical prostate cancer models. To date, docetaxel is the only chemotherapeutic agent in clinical use for hormone-refractory prostate cancer. In vitro experiments with 22Rv1, LNCaP, DU-145, PC3 and PC3-M prostate cancer cell lines showed significantly enhanced inhibition of cell proliferation and apoptosis when lonafarnib was added to docetaxel. In human tumor xenograft models, continuous coadministration of lonafarnib with docetaxel caused marked tumor regressions (24-47%) in tumors from all of the cell types as well as parental CWR22 xenografts. Intermittent dosing of lonafarnib (5 days on then 5 days off) coadministered with docetaxel produced similar regressions in hormone-refractory 22Rv1 tumors. 22Rv1 tumors progressing on docetaxel treatment also responded to treatment with intermittent lonafarnib (5 days on then 5 days off). Moreover, animals did not exhibit any signs of toxicity during coadministration of lonafarnib and docetaxel. In conclusion, coadministration of continuous and intermittent lonafarnib enhanced the antitumor activity of docetaxel in a panel of prostate cancer models. An intermittent dosing schedule of lonafarnib coadministered with docetaxel may allow enhanced efficacy to that of continuous dosing by improving the tolerability of higher doses of lonafarnib.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms, Hormone-Dependent/drug therapy , Piperidines/therapeutic use , Prostatic Neoplasms/drug therapy , Pyridines/therapeutic use , Taxoids/therapeutic use , Xenograft Model Antitumor Assays , Animals , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Docetaxel , Drug Synergism , Drug Therapy, Combination , Humans , Male , Mice , Mice, Nude , Mice, SCID , Neoplasms, Hormone-Dependent/blood , Neoplasms, Hormone-Dependent/pathology , Piperidines/blood , Piperidines/pharmacokinetics , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Pyridines/blood , Pyridines/pharmacokinetics
10.
Gynecol Oncol ; 109(1): 97-106, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18237771

ABSTRACT

OBJECTIVES: To determine the effects of combining lonafarnib with paclitaxel on the growth of human ovarian cancer cells and tumor xenografts as well as to monitor a pharmacodynamic marker of farnesyltransferase inhibition (HDJ-2) in peripheral blood mononuclear cells (PBMCs) isolated from tumor-bearing animals after treatment with this combination. METHODS: Proliferation of A2780, PA-1, IGROV-1, and TOV-112D cells was assessed after treatment with lonafarnib and paclitaxel. Cell cycle progression was determined by flow cytometry, and apoptosis was evaluated by assaying for caspase-3 and cleaved PARP. The effects of lonafarnib and paclitaxel on the tumor growth of each model were determined in immunocompromised mice. Proteins extracted from cells, tumors, and PBMCs were assayed for HDJ-2 mobility shifts by Western blotting as well as for farnesyl protein transferase (FTase) enzyme activity by biochemical analyses. RESULTS: In A2780, PA-1, IGROV-1, and TOV-112D cells lonafarnib potentiated the growth inhibitory effects of paclitaxel. In each of the models lonafarnib enhanced paclitaxel-induced mitotic arrest and apoptosis. The combination of lonafarnib plus paclitaxel resulted in marked tumor regressions in A2780, TOV-112D, PA-1, and IGROV-1 tumor xenografts. Western blotting demonstrated that in PBMCs isolated from the animals, paclitaxel treatment suppressed lonafarnib-induced HDJ-2 mobility shifts. Paclitaxel did not affect lonafarnib inhibition of FTase enzyme activity levels in these PBMCs. CONCLUSIONS: Lonafarnib enhances the antiproliferative effects of paclitaxel on ovarian cancer cells in vitro and ovarian tumor xenografts in vivo. Measuring FTase enzyme activity levels rather than HDJ-2 shifts in PBMCs may be a more accurate biomarker to predict levels of farnesyltransferase inhibition in patients who are also receiving paclitaxel chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Piperidines/pharmacology , Pyridines/pharmacology , Animals , Apoptosis/drug effects , Biomarkers, Tumor/blood , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Farnesyltranstransferase/blood , Farnesyltranstransferase/metabolism , Female , HSP40 Heat-Shock Proteins/metabolism , Humans , Leukocytes, Mononuclear/enzymology , Mice , Mice, Nude , Mice, SCID , Ovarian Neoplasms/blood , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Piperidines/administration & dosage , Pyridines/administration & dosage , Xenograft Model Antitumor Assays
11.
Bioorg Med Chem Lett ; 16(4): 984-8, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16298128

ABSTRACT

Benzocycloheptapyridine tricyclic compounds with piperazine or substituted piperidine moieties extending either from the 5- or 6-position of the tricyclic bridgehead exhibited enhanced FTase activity: this resulted from favorable binding of the ligand nitrogen with the catalytic zinc found in the FTase. A single isomer at C-11 with piperazine adduct extending from the 6-position, compound 24, exhibited excellent FTase activity with IC50 = 0.007 microM, soft agar IC50 = 72 nM, and Rat AUC(PO, 10 mpk) = 4.0 microM x h. X-ray of (-)-[8-chloro-6-(1-piperazinyl)-1H-benzo[5,6]]cyclohepta[1,2-b]pyridine-11-yl]-1-(methylsulfonyl)piperidine 24 bound to Ftase revealed favorable interaction between piperazine nitrogen and catalytic zinc atom.


Subject(s)
Alkyl and Aryl Transferases/chemistry , Organometallic Compounds/chemistry , Piperazines/chemistry , Zinc/chemistry , Alkyl and Aryl Transferases/antagonists & inhibitors , Binding Sites/drug effects , Catalysis/drug effects , Crystallography, X-Ray , Ligands , Models, Molecular , Molecular Structure , Organometallic Compounds/pharmacology , Piperazine , Stereoisomerism , Structure-Activity Relationship
12.
J Lipid Res ; 47(1): 15-31, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16278491

ABSTRACT

Some proteins undergo posttranslational modification by the addition of an isoprenyl lipid (farnesyl- or geranylgeranyl-isoprenoid) to a cysteine residue proximal to the C terminus. Protein isoprenylation promotes membrane association and contributes to protein-protein interactions. Farnesylated proteins include small GTPases, tyrosine phosphatases, nuclear lamina, cochaperones, and centromere-associated proteins. Prenylation is required for the transforming activity of Ras. Because of the high frequency of Ras mutations in cancer, farnesyl transferase inhibitors (FTIs) were investigated as a means to antagonize Ras function. Evaluation of FTIs led to the finding that both K- and N-Ras are alternatively modified by geranylgeranyl prenyltransferase-1 in FTI-treated cells. Geranylgeranylated forms of Ras retain the ability to associate with the plasma membrane and activate substrates. Despite this, FTIs are effective at inhibiting the growth of human tumor cells in vitro, suggesting that activity is dependent on blocking the farnesylation of other proteins. FTIs also inhibit the in vivo growth of human tumor xenografts and sensitize these models to chemotherapeutics, most notably taxanes. Several FTIs have entered clinical trials for various cancer indications. In some clinical settings, primarily hematologic malignancies, FTIs have displayed evidence of single-agent activity. Clinical studies in progress are exploring the antitumor activity of FTIs as single agents and in combination. This review will summarize the basic biology of FTIs, their antitumor activity in preclinical models, and the current status of clinical studies with these agents.


Subject(s)
Farnesyltranstransferase/antagonists & inhibitors , Lipid Metabolism , Protein Processing, Post-Translational , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Prenylation/drug effects , Protein Processing, Post-Translational/drug effects , ras Proteins/metabolism
13.
Mol Cancer Ther ; 4(8): 1214-21, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16093437

ABSTRACT

Insulin-like growth factor-I receptor (IGF-IR) plays an important role in tumor cell growth and survival. On ligand stimulation, IGF-IR, a receptor tyrosine kinase, phosphorylates tyrosine residues on two major substrates, IRS-1 and Shc, which subsequently signal through the Ras/mitogen-activated protein kinase and phosphatidylinositol 3-kinase/AKT pathways. Here, we describe the characterization of a fully human anti-IGF-IR monoclonal antibody 19D12 that inhibits IGF binding and autophosphorylation of both IGF-IR/IGF-IR homodimers and IGF-IR/insulin receptor heterodimers. 19D12 does not recognize insulin receptor homodimers. In addition to inhibiting IGF-IR autophosphorylation, 19D12 also inhibits IRS-1 phosphorylation and activation of the major downstream signaling molecules AKT and extracellular signal-regulated kinase 1/2. Furthermore, the antibody down-regulates the total IGF-IR protein level and can exhibit antibody-dependent cellular cytotoxicity activity against a non-small cell adenocarcinoma cell line in vitro in the presence of isolated human natural killer cells. 19D12 binds tightly to the receptor, with an affinity of 3.8 pmol/L as measured by KinExA. In cell culture, 19D12 inhibits proliferation and soft agar growth of various tumor cell lines. In vivo, 19D12 inhibits the tumor growth of a very aggressive human ovarian tumor xenograft model A2780. These data support the development of this anti-IGF-IR monoclonal antibody as a promising anticancer agent.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dimerization , Down-Regulation , Humans , Insulin Receptor Substrate Proteins , Insulin-Like Growth Factor I/metabolism , Mice , Neoplasms/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, IGF Type 1/immunology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
14.
J Biol Chem ; 280(35): 31101-8, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-16006564

ABSTRACT

Lonafarnib (SCH66336) is a farnesyl transferase inhibitor (FTI) that inhibits the post-translational lipid modification of H-Ras and other farnesylated proteins. K- and N-Ras are also substrates of farnesyl transferase; however, upon treatment with FTIs, they are alternatively prenylated by geranylgeranyl transferase-1. Despite the failure to abrogate prenylation of K- and N-Ras, growth of many tumors in preclinical models is inhibited by FTIs. This suggests that the anti-proliferative action of FTIs is dependent on blocking the farnesylation of other proteins. Rheb (Ras homologue enriched in brain) is a farnesylated small GTPase that positively regulates mTOR (mammalian target of rapamycin) signaling. We found that Rheb and Rheb2 mRNA were elevated in various tumor cell lines relative to normal cells. Peptides derived from the carboxyl termini of human Rheb and Rheb2 are in vitro substrates for farnesyl transferase but not geranylgeranyl transferase-1. Rheb prenylation in cell culture was completely inhibited by SCH66336, indicating a lack of alternative prenylation. SCH66336 treatment also inhibited the phosphorylation of S6 ribosomal protein, a downstream target of Rheb and mTOR signaling. SCH66336 did not inhibit S6 phosphorylation in cells expressing Rheb-CSVL, a mutant construct of Rheb designed to be geranylgeranylated. Importantly, expression of Rheb-CSVL also abrogated SCH66336 enhancement of tamoxifen- and docetaxel-induced apoptosis in MCF-7 breast cancer cells and ES-2 ovarian cancer cells, respectively. Further, inhibition of Rheb signaling by rapamycin treatment, small interfering RNA, or dominant negative Rheb enhanced tamoxifen- and docetaxel-induced apoptosis, similar to FTI treatment. These studies demonstrated that Rheb is modified by farnesylation, is not a substrate for alternative prenylation, and plays a role in SCH66336 enhancement of the anti-tumor response to other chemotherapeutics.


Subject(s)
Alkyl and Aryl Transferases , Bridged-Ring Compounds/therapeutic use , Monomeric GTP-Binding Proteins/metabolism , Neoplasms/drug therapy , Neuropeptides/metabolism , Piperidines/metabolism , Protein Kinases/metabolism , Pyridines/metabolism , Tamoxifen/therapeutic use , Taxoids/therapeutic use , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Animals , Antineoplastic Agents/therapeutic use , Caspases/metabolism , Cell Line, Tumor , Farnesyltranstransferase , Humans , Monomeric GTP-Binding Proteins/genetics , Neuropeptides/genetics , Phosphorylation , Protein Kinases/genetics , Protein Prenylation , RNA, Messenger/metabolism , Ras Homolog Enriched in Brain Protein , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases
15.
J Biol Chem ; 280(13): 12935-43, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15659406

ABSTRACT

In this study, differential gene expression between normal human mammary epithelial cells and their malignant counterparts (eight well established breast cancer cell lines) was studied using Incyte GeneAlbum 1-6, which contains 65,873 cDNA clones representing 33,515 individual genes. 3,152 cDNAs showed a > or =3.0-fold expression level change in at least one of the human breast cancer cell lines as compared with normal human mammary epithelial cells. Integration of breast tumor gene expression data with the genes in the tumor suppressor p53 signaling pathway yielded 128 genes whose expression is altered in breast tumor cell lines and in response to p53 expression. A hierarchical cluster analysis of the 128 genes revealed that a significant portion of genes demonstrate an opposing expression pattern, i.e. p53-activated genes are down-regulated in the breast tumor lines, whereas p53-repressed genes are up-regulated. Most of these genes are involved in cell cycle regulation and/or apoptosis, consistent with the tumor suppressor function of p53. Follow-up studies on one gene, RAI3, suggested that p53 interacts with the promoter of RAI3 and repressed its expression at the onset of apoptosis. The expression of RAI3 is elevated in most tumor cell lines expressing mutant p53, whereas RAI3 mRNA is relatively repressed in the tumor cell lines expressing wild-type p53. Furthermore, ectopic expression of RAI3 in 293 cells promotes anchorage-independent growth and small interfering RNA-mediated depletion of RAI3 in AsPc-1 pancreatic tumor cells induces cell morphological change. Taken together, these data suggest a role for RAI3 in tumor growth and demonstrate the predictive power of integrative genomics.


Subject(s)
Gene Expression Regulation, Neoplastic , Genome, Human , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Agar/chemistry , Apoptosis , Breast Neoplasms/metabolism , Cell Cycle , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Cluster Analysis , DNA, Complementary/metabolism , Down-Regulation , Epithelial Cells/metabolism , Genomics/methods , HeLa Cells , Humans , Mammary Glands, Human/metabolism , Models, Genetic , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
16.
Bioorg Med Chem Lett ; 14(23): 5877-80, 2004 Dec 06.
Article in English | MEDLINE | ID: mdl-15501060

ABSTRACT

Successful efforts to make farnesyl transferase (FT) inhibitors with appropriately tethered ligands designed to interact with a catalytic zinc that exist in the enzyme have been realized. Thus, by introducing either a pyridylmethylamino or propylaminolimidazole amide moieties off the 2-position of the piperidine ring, FT inhibitors with activities in the picomolar range have been achieved as exemplified by compounds 12a and 12b. An X-ray structure of 11b bound to FT shows the enhanced activity is a result of interacting with the active-site zinc.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Catalytic Domain/physiology , Drug Delivery Systems/methods , Enzyme Inhibitors/metabolism , Zinc/metabolism , Alkyl and Aryl Transferases/metabolism , Catalytic Domain/drug effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Protein Binding/physiology , Protein Conformation
17.
Curr Opin Drug Discov Devel ; 7(4): 478-86, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15338957

ABSTRACT

Farnesyltransferase (FT) inhibitors were originally designed as anticancer agents, and were thought to act by inhibiting the farnesylation of mutant Ras proteins. However, these compounds were subsequently demonstrated to have antitumor effects even in the absence of Ras mutations and it has now become clear that other protein targets are involved. This article discusses the preclinical and clinical development of FT inhibitors. To date, tipifarnib (Zarnestra; Janssen Pharmaceutica NV) and lonafarnib (Sarasar; Schering-Plough Research Institute) are the only two FT inhibitors to have been evaluated in phase III clinical trials. The clinical results of these two compounds are presented below, with emphasis on ways of enhancing the possibility of a successful FT inhibitor anticancer drug. Details of new FT inhibitors disclosed since the beginning of 2003 are also included.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/therapeutic use , Antineoplastic Agents/therapeutic use , Alkyl and Aryl Transferases/pharmacology , Animals , Antineoplastic Agents/pharmacology , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Drug Delivery Systems , Drug Screening Assays, Antitumor , Farnesyltranstransferase , Humans , Molecular Structure , Oncogene Proteins/biosynthesis , Oncogene Proteins/drug effects
18.
Clin Cancer Res ; 10(9): 2968-76, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15131032

ABSTRACT

PURPOSE: To establish the maximum tolerated dose of lonafarnib, a novel farnesyltransferase inhibitor, in combination with paclitaxel in patients with solid tumors and to characterize the safety, tolerability, dose-limiting toxicity, and pharmacokinetics of this combination regimen. EXPERIMENTAL DESIGN: In a Phase I trial, lonafarnib was administered p.o., twice daily (b.i.d.) on continuously scheduled doses of 100 mg, 125 mg, and 150 mg in combination with i.v. paclitaxel at doses of 135 mg/m(2) or 175 mg/m(2) administered over 3 h on day 8 of every 21-day cycle. Plasma paclitaxel and lonafarnib concentrations were collected at selected time points from each patient. RESULTS: Twenty-four patients were enrolled; 21 patients were evaluable. The principal grade 3/4 toxicity was diarrhea (5 of 21 patients), which was most likely due to lonafarnib. dose-limiting toxicities included grade 3 hyperbilirubinemia at dose level 3 (100 mg b.i.d. lonafarnib and 175 mg/m(2) paclitaxel); grade 4 diarrhea and grade 3 peripheral neuropathy at dose level 3A (125 mg b.i.d. lonafarnib and 175 mg/m(2) paclitaxel); and grade 4 neutropenia with fever and grade 4 diarrhea at level 4 (150 mg b.i.d. lonafarnib and 175 mg/m(2) paclitaxel). The maximum tolerated dose established by the continual reassessment method was lonafarnib 100 mg b.i.d. and paclitaxel 175 mg/m(2). Paclitaxel appeared to have no effect on the pharmacokinetics of lonafarnib. The median duration of therapy was eight cycles, including seven cycles with paclitaxel. Six of 15 previously treated patients had a durable partial response, including 3 patients who had previous taxane therapy. Notably, two of five patients with taxane-resistant metastatic non-small cell lung cancer had partial responses. CONCLUSIONS: When combined with paclitaxel, the recommended dose of lonafarnib for Phase II trials is 100 mg p.o. twice daily with 175 mg/m(2) of paclitaxel i.v. every 3 weeks. Additional studies of lonafarnib in combination regimens appear warranted, particularly in patients with non-small cell lung cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Alkyl and Aryl Transferases/antagonists & inhibitors , Anemia/chemically induced , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Area Under Curve , Dose-Response Relationship, Drug , Farnesyltranstransferase , Fatigue/chemically induced , Female , Heart Arrest/chemically induced , Humans , Leukopenia/chemically induced , Male , Middle Aged , Neoplasms/metabolism , Neoplasms/pathology , Neutropenia/chemically induced , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Paclitaxel/pharmacokinetics , Piperidines/administration & dosage , Piperidines/adverse effects , Piperidines/pharmacokinetics , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/pharmacokinetics , Treatment Outcome
19.
Drug Resist Updat ; 6(4): 183-95, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12962684

ABSTRACT

p21(WAF1/Cip1) was initially identified as a cell cycle regulatory protein that can cause cell cycle arrest. It is induced by both p53-dependent and p53-independent mechanisms. This mini-review briefly discusses its currently known functions in apoptosis and drug sensitivity. As an inhibitor of cell proliferation, p21(WAF1/Cip1) plays an important role in drug-induced tumor suppression. Nevertheless, a number of recent studies have shown that p21(WAF1/Cip1) can assume both pro- or anti-apoptotic functions in response to anti-tumor agents depending on cell type and cellular context. This dual role of p21(WAF1/Cip1) in cancer cells complicates using p21(WAF1/Cip1) status to predict response to anti-tumor agents. However, it is possible to develop p21(WAF1/Cip1)-targeted reagents or p21(WAF1/Cip1) gene transfer techniques to have a beneficial effect within a well-defined therapeutic context. Better understanding of the roles of p21(WAF1/Cip1) in tumors should enable a more rational approach to anti-tumor drug design and therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Cycle Proteins/physiology , Cell Cycle Proteins/pharmacokinetics , Cyclins/physiology , Cyclins/pharmacokinetics , Drug Resistance, Neoplasm , Apoptosis/physiology , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Gene Transfer Techniques , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/physiopathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...