Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
ACS Appl Mater Interfaces ; 13(27): 31756-31765, 2021 Jul 14.
Article in English | MEDLINE | ID: mdl-34192458

ABSTRACT

Introduction of mesopore is critical for applications where mass-transport limitations within microporous networks, especially for zeolite with one-dimensional microporous network, hinder their performance. Generally, the creation of mesopore in zeolite through a direct synthesis route is strongly dependent on complex and expensive organic molecules, which limits their commercial application. Here, we successfully developed a facile synthesis route for preparing ZSM-48 zeolite (*MRE topology) with ultralarge mesoporosity in which typical 1,6-hexylenediamine worked as an organic structure-directing agent, innovatively assisted by a simple crystal growth modifier (tetraethylammonium bromide, TEABr). The working mechanism of TEABr during crystallization was revealed and proposed on the basis of TEM, thermal gravimetric mass spectrum, and 13C cross-polarization magic angle spinning NMR characterization results. In the process, TEA+ ions preferentially interacted with the solid during the induction period, which effectively suppressed the aggregation of ZSM-48 primary nanorods. As a result, ultralarge mesoporosity of 0.97 cm3·g-1 was constructed through the stacking of the nanorods. Interestingly, TEA+ ions only took part in the crystallization process and did not occlude in the pores of the final zeolites indicating its potential in recyclability. Moreover, similar synthesis strategy could be applied for the preparation of hierarchical ferrierite zeolites, implying the universality of this strategy. Compared with a conventional sample, ZSM-48 zeolite with ultralarge mesoporosity showed superior catalytic stability in the m-xylene isomerization reaction due to its significantly enhanced diffusion and mass transfer capability, which will greatly promote the practical application of ZSM-48 zeolite.

2.
Clin Cancer Res ; 27(2): 622-631, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33148666

ABSTRACT

PURPOSE: Mortality due to acute myeloid leukemia (AML) remains high, and the management of relapsed or refractory AML continues to be therapeutically challenging. The reapproval of Mylotarg, an anti-CD33-calicheamicin antibody-drug conjugate (ADC), has provided a proof of concept for an ADC-based therapeutic for AML. Several other ADCs have since entered clinical development of AML, but have met with limited success. We sought to develop a next-generation ADC for AML with a wide therapeutic index (TI) that overcomes the shortcomings of previous generations of ADCs. EXPERIMENTAL DESIGN: We compared the TI of our novel CD33-targeted ADC platform with other currently available CD33-targeted ADCs in preclinical models of AML. Next, using this next-generation ADC platform, we performed a head-to-head comparison of two attractive AML antigens, CD33 and CD123. RESULTS: Our novel ADC platform offered improved safety and TI when compared with certain currently available ADC platforms in preclinical models of AML. Differentiation between the CD33- and CD123-targeted ADCs was observed in safety studies conducted in cynomolgus monkeys. The CD33-targeted ADC produced severe hematologic toxicity, whereas minimal hematologic toxicity was observed with the CD123-targeted ADC at the same doses and exposures. The improved toxicity profile of an ADC targeting CD123 over CD33 was consistent with the more restricted expression of CD123 in normal tissues. CONCLUSIONS: We optimized all components of ADC design (i.e., leukemia antigen, antibody, and linker-payload) to develop an ADC that has the potential to translate into an effective new therapy against AML.


Subject(s)
Gemtuzumab/therapeutic use , Immunoconjugates/therapeutic use , Interleukin-3 Receptor alpha Subunit/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Area Under Curve , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Gemtuzumab/immunology , Gemtuzumab/pharmacokinetics , HL-60 Cells , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Macaca fascicularis , Mice , Sialic Acid Binding Ig-like Lectin 3/immunology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods
3.
Clin Cancer Res ; 26(9): 2188-2202, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31996389

ABSTRACT

PURPOSE: Gastrointestinal cancers remain areas of high unmet need despite advances in targeted and immunotherapies. Here, we demonstrate potent, tumor-selective efficacy with PF-07062119, a T-cell engaging CD3 bispecific targeting tumors expressing Guanylyl Cyclase C (GUCY2C), which is expressed widely across colorectal cancer and other gastrointestinal malignancies. In addition, to address immune evasion mechanisms, we explore combinations with immune checkpoint blockade agents and with antiangiogenesis therapy. EXPERIMENTAL DESIGN: PF-07062119 activity was evaluated in vitro in multiple tumor cell lines, and in vivo in established subcutaneous and orthotopic human colorectal cancer xenograft tumors with adoptive transfer of human T cells. Efficacy was also evaluated in mouse syngeneic tumors using human CD3ε transgenic mice. IHC and mass cytometry were performed to demonstrate drug biodistribution, recruitment of activated T cells, and to identify markers of immune evasion. Combination studies were performed with anti-PD-1/PD-L1 and anti-VEGF antibodies. Toxicity and pharmacokinetic studies were done in cynomolgus macaque. RESULTS: We demonstrate that GUCY2C-positive tumors can be targeted with an anti-GUCY2C/anti-CD3ε bispecific, with selective drug biodistribution to tumors. PF-07062119 showed potent T-cell-mediated in vitro activity and in vivo efficacy in multiple colorectal cancer human xenograft tumor models, including KRAS- and BRAF-mutant tumors, as well as in the immunocompetent mouse syngeneic tumor model. PF-07062119 activity was further enhanced when combined with anti-PD-1/PD-L1 treatment or in combination with antiangiogenic therapy. Toxicity studies in cynomolgus indicated a monitorable and manageable toxicity profile. CONCLUSIONS: These data highlight the potential for PF-07062119 to demonstrate efficacy and improve patient outcomes in colorectal cancer and other gastrointestinal malignancies.


Subject(s)
Antibodies, Bispecific/administration & dosage , CD3 Complex/immunology , Colorectal Neoplasms/therapy , Gastrointestinal Neoplasms/therapy , Immunotherapy/methods , Receptors, Enterotoxin/immunology , T-Lymphocytes/immunology , Adoptive Transfer/methods , Animals , Antibodies, Bispecific/pharmacokinetics , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Disease Models, Animal , Female , Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Tissue Distribution
4.
Chem Commun (Camb) ; 55(92): 13804-13807, 2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31633709

ABSTRACT

Methyl carboxylate esters can be used as additives to promote the zeolite catalysed formation of dimethly ether (DME) from methanol. By taking advantage of the well-known confinement effect in combination with further functionalisation the potency of the promoter can be markedly enhanced, giving significant increases in DME yield at promoter concentrations as low as 10 ppm relative to methanol. The promotion is readily reversible and promoter concentration can be used to fine tune the zeolite productivity to DME.

5.
Mol Cancer Ther ; 17(1): 243-253, 2018 01.
Article in English | MEDLINE | ID: mdl-29054985

ABSTRACT

Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate (ADC) that has demonstrated clinical benefit for patients with HER2+ metastatic breast cancer; however, its clinical activity is limited by inherent or acquired drug resistance. The molecular mechanisms that drive clinical resistance to T-DM1, especially in HER2+ tumors, are not well understood. We used HER2+ cell lines to develop models of T-DM1 resistance using a cyclical dosing schema in which cells received T-DM1 in an "on-off" routine until a T-DM1-resistant population was generated. T-DM1-resistant N87 cells (N87-TM) were cross-resistant to a panel of trastuzumab-ADCs (T-ADCs) with non-cleavable-linked auristatins. N87-TM cells do not have a decrease in HER2 protein levels or an increase in drug transporter protein (e.g., MDR1) expression compared with parental N87 cells. Intriguingly, T-ADCs using auristatin payloads attached via an enzymatically cleavable linker overcome T-DM1 resistance in N87-TM cells. Importantly, N87-TM cells implanted into athymic mice formed T-DM1 refractory tumors that remain sensitive to T-ADCs with cleavable-linked auristatin payloads. Comparative proteomic profiling suggested enrichment in proteins that mediate caveolae formation and endocytosis in the N87-TM cells. Indeed, N87-TM cells internalize T-ADCs into intracellular caveolin-1 (CAV1)-positive puncta and alter their trafficking to the lysosome compared with N87 cells. T-DM1 colocalization into intracellular CAV1-positive puncta correlated with reduced response to T-DM1 in a panel of HER2+ cell lines. Together, these data suggest that caveolae-mediated endocytosis of T-DM1 may serve as a novel predictive biomarker for patient response to T-DM1. Mol Cancer Ther; 17(1); 243-53. ©2017 AACR.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Endocytosis/drug effects , Trastuzumab/therapeutic use , Animals , Antineoplastic Agents, Immunological/pharmacology , Caveolae , Drug Resistance, Neoplasm , Female , Humans , Male , Mice , Trastuzumab/pharmacology
6.
PLoS One ; 12(5): e0178452, 2017.
Article in English | MEDLINE | ID: mdl-28558059

ABSTRACT

Antibody drug conjugates (ADCs) are no longer an unknown entity in the field of cancer therapy with the success of marketed ADCs like ADCETRIS and KADCYLA and numerous others advancing through clinical trials. The pursuit of novel cytotoxic payloads beyond the mictotubule inhibitors and DNA damaging agents has led us to the recent discovery of an mRNA splicing inhibitor, thailanstatin, as a potent ADC payload. In our previous work, we observed that the potency of this payload was uniquely tied to the method of conjugation, with lysine conjugates showing much superior potency as compared to cysteine conjugates. However, the ADC field is rapidly shifting towards site-specific ADCs due to their advantages in manufacturability, characterization and safety. In this work we report the identification of a highly efficacious site-specific thailanstatin ADC. The site of conjugation played a critical role on both the in vitro and in vivo potency of these ADCs. During the course of this study, we developed a novel methodology of loading a single site with multiple payloads using an in situ generated multi-drug carrying peptidic linker that allowed us to rapidly screen for optimal conjugation sites. Using this methodology, we were able to identify a double-cysteine mutant ADC delivering four-loaded thailanstatin that was very efficacious in a gastric cancer xenograft model at 3mg/kg and was also shown to be efficacious against T-DM1 resistant and MDR1 overexpressing tumor cell lines.


Subject(s)
Antineoplastic Agents/chemistry , Immunoconjugates/chemistry , Peptides/chemistry , Pyrans/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Carriers , Drug Screening Assays, Antitumor , Humans , Pyrans/chemistry
7.
Mol Cancer Ther ; 14(4): 952-63, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25646013

ABSTRACT

Antibody-drug conjugates (ADC) are emerging as clinically effective therapy. We hypothesized that cancers treated with ADCs would acquire resistance mechanisms unique to immunoconjugate therapy and that changing ADC components may overcome resistance. Breast cancer cell lines were exposed to multiple cycles of anti-Her2 trastuzumab-maytansinoid ADC (TM-ADC) at IC80 concentrations followed by recovery. The resistant cells, 361-TM and JIMT1-TM, were characterized by cytotoxicity, proteomic, transcriptional, and other profiling. Approximately 250-fold resistance to TM-ADC developed in 361-TM cells, and cross-resistance was observed to other non-cleavable-linked ADCs. Strikingly, these 361-TM cells retained sensitivity to ADCs containing cleavable mcValCitPABC-linked auristatins. In JIMT1-TM cells, 16-fold resistance to TM-ADC developed, with cross-resistance to other trastuzumab-ADCs. Both 361-TM and JIMT1-TM cells showed minimal resistance to unconjugated mertansine (DM1) and other chemotherapeutics. Proteomics and immunoblots detected increased ABCC1 (MRP1) drug efflux protein in 361-TM cells, and decreased Her2 (ErbB2) in JIMT1-TM cells. Proteomics also showed alterations in various pathways upon chronic exposure to the drug in both cell models. Tumors derived from 361-TM cells grew in mice and were refractory to TM-ADC compared with parental cells. Hence, acquired resistance to trastuzumab-maytansinoid ADC was generated in cultured cancer cells by chronic drug treatment, and either increased ABCC1 protein or reduced Her2 antigen were primary mediators of resistance. These ADC-resistant cell models retain sensitivity to other ADCs or standard-of-care chemotherapeutics, suggesting that alternate therapies may overcome acquired ADC resistance. Mol Cancer Ther; 14(4); 952-63. ©2015 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Immunoconjugates/pharmacology , Trastuzumab/pharmacology , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Immunoconjugates/administration & dosage , Inhibitory Concentration 50 , Mice , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Protein Transport , Proteome , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Signal Transduction , Transcriptome , Trastuzumab/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
Bioorg Med Chem ; 15(11): 3635-48, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17416531

ABSTRACT

A series of 4-dimethylamino-but-2-enoic acid [4-(3,6-dioxo-cyclohexa-1,4-dienylamino)-7-ethoxy-quinazolin-6-yl]-amide derivatives were prepared. These compounds have two independent reactive centers and were designed to function as dual irreversible inhibitors of the kinase domains of both Epidermal Growth Factor Receptor (EGFR) and Vascular Endothelial Growth Factor Receptor-2 (VEGFR-2) where each reactive center targets a different, non-conserved, cysteine residue located in the ATP binding pocket of these enzymes. The compounds contain a 6-(4-(dimethylamino) crotonamide) Michael acceptor group that targets Cys-773 in EGFR and a 4-(amino-[1,4]benzoquinone) moiety that targets Cys-1045 in VEGFR-2. In vitro studies indicated that most of these compounds are relatively potent inhibitors of each enzyme. These inhibitors were compared with reference compounds that lack one or both of the reactive centers. The relative dependence of the IC(50) values on the concentration of ATP used in the assays suggests that these compounds appear to function as irreversible inhibitors of each kinase.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Quinazolines/chemistry , Quinazolines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Binding Sites , Biological Assay , Cells, Cultured , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Protein Conformation/drug effects , Protein Kinase Inhibitors/chemical synthesis , Quinazolines/chemical synthesis , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/metabolism
9.
Mol Cancer Ther ; 5(2): 270-8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16505100

ABSTRACT

Resistance to paclitaxel-based therapy is frequently encountered in the clinic. The mechanisms of intrinsic or acquired paclitaxel resistance are not well understood. We sought to characterize the resistance mechanisms that develop upon chronic exposure of a cancer cell line to paclitaxel in the presence of the P-glycoprotein reversal agent, CL-347099. The epidermoid tumor line KB-3-1 was exposed to increasing concentrations of paclitaxel and 5 micromol/L CL-347099 for up to 1 year. Cells grown in 15 nmol/L paclitaxel plus CL-347099 (KB-15-PTX/099) developed 18-fold resistance to paclitaxel and were dependent upon paclitaxel for maximal growth. They grew well and retained resistance to paclitaxel when grown in athymic mice. Cross-resistance (3- to 5-fold) was observed in tissue culture to docetaxel, the novel taxane MAC-321, and epothilone B. Collateral sensitivity (approximately 3-fold) was observed to the depolymerizing agents vinblastine, dolastatin-10, and HTI-286. KB-15-PTX/099-resistant cells did not overexpress P-glycoprotein nor did they have an alteration of [14C]paclitaxel accumulation compared with parental cells. However, a novel point mutation (T to A) resulting in Asp26 to glutamate substitution in class I (M40) beta-tubulin was found. Based on an electron crystallography structure of Zn-stabilized tubulin sheets, the phenyl ring of C-3' NHCO-C6H5 of paclitaxel makes contact with Asp26 of beta-tubulin, suggesting a ligand-induced mutation. Optimized model complexes of paclitaxel, docetaxel, and MAC-321 in beta-tubulin show a novel hydrogen bonding pattern for the glutamate mutant and rationalize the observed resistance profiles. However, a mutation in the paclitaxel binding pocket does not explain the phenotype completely. KB-15-PTX/099 cells have impaired microtubule stability as determined by a reduced percentage of tubulin in microtubules and reflected by less acetylated tubulin. These results suggest that a mutation in tubulin might affect microtubule stability as well as drug binding and contribute to the observed resistance profile.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Drug Resistance, Neoplasm/genetics , Paclitaxel/therapeutic use , Tubulin/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Amino Acid Substitution/genetics , Animals , Antineoplastic Agents, Phytogenic/chemistry , Aspartic Acid/chemistry , Aspartic Acid/genetics , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Docetaxel , Epothilones/chemistry , Epothilones/therapeutic use , Glutamic Acid/chemistry , Glutamic Acid/genetics , Humans , Mice , Mice, Nude , Microtubules/genetics , Microtubules/metabolism , Paclitaxel/analogs & derivatives , Paclitaxel/chemistry , Point Mutation , Protein Conformation , Taxoids/chemistry , Taxoids/therapeutic use , Tubulin/chemistry , Verapamil/analogs & derivatives , Verapamil/pharmacology
10.
J Med Chem ; 48(24): 7560-81, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16302797

ABSTRACT

A series of 2-(quinazolin-4-ylamino)-[1,4] benzoquinone derivatives that function as potent covalent-binding, irreversible inhibitors of the kinase domain of vascular endothelial growth factor receptor-2 (VEGFR-2) has been prepared by ceric ammonium nitrate oxidation of substituted (2,5-dimethoxyphenyl)(6,7-disubstituted-quinazolin-4-yl)amines and by displacement of the chlorine atom of substituted 2-chloro-5-(6,7-disubstituted-quinazolin-4-ylamino)-[1,4]benzoquinones with various amines, anilines, phenols, and alcohols. Enzyme studies were conducted in the absence and presence of glutathione and plasma. Several of the compounds inhibit VEGF-stimulated autophosphorylation in intact cells. Kinetic experiments were performed to study the reactivity of selected inhibitors toward glutathione. Reactivities correlated with LUMO energies calculated as averages of those of individual conformers weighted by the Boltzmann distribution. These results and molecular modeling were used to rationalize the biological observations. The compounds behave as non-ATP-competitive inhibitors. Unequivocal evidence, from mass spectral studies, indicates that these inhibitors form a covalent interaction with Cys-1045. One member of this series displays antitumor activity in an in vivo model.


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Benzoquinones/chemical synthesis , Quinazolines/chemical synthesis , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Benzoquinones/chemistry , Benzoquinones/pharmacology , Binding Sites , Cell Line , Female , Glutathione/chemistry , Humans , Kinetics , Mice , Mice, Nude , Models, Molecular , Molecular Conformation , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Quantum Theory , Quinazolines/chemistry , Quinazolines/pharmacology , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Structure-Activity Relationship , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
11.
Chem Biol Interact ; 152(2-3): 131-8, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15840386

ABSTRACT

2-Acetylaminonaphthalene (2-AAN) has been recognized as a urinary bladder carcinogen in humans. The deacetylated form, 2-aminonaphthalene (2-AN), is metabolized in vivo and reacts primarily with guanine residues in DNA, resulting in the formation of dG-N(2)-aminonaphthalene (dG-N(2)-AN) adduct. Phosphoramidite chemical procedure has recently been established in our laboratory to prepare oligodeoxynucleotides containing a single dG-N(2)-acetylaminonaphthalene (dG-N(2)-AAN) adduct. Oligodeoxynucleotides ((5')TCCTCCTNXCCTCTC, where X is dG or dG-N(2)-AAN and N is C, A, T or G) with different bases 5' flanking to the lesion were prepared and were inserted into a single-strand shuttle vectors and used to establish the mutational frequency and specificity of dG-N(2)-AAN adduct in simian kidney cells. dG-N(2)-AAN adduct promoted preferential incorporation of dCMP, the correct base, opposite the lesion. When the 5' flanking base to the lesion was C, A or T, the mutational frequency was under 2.1%. When G flanked to the lesion, the mutational frequency was slightly increased to 4.2%. Misincorporation of dAMP, dTMP, and/or dGMP varied depending on the 5' flanking base. When dG-N(2)-AAN was positioned at codon 61 of noncoding strand of human c-Ha-ras1 gene ((5')TCCTCCTXGCCTCTC, where X is dG-N(2)-AAN), the mutational frequency was 6.7%; G-->T transversions (4.7%), followed by G-->A transition (2.0%), were observed. These results demonstrated that dG-N(2)-AAN is a weak mutagenic lesion in mammalian cells. The influence of 5' flanking sequence context was observed on the mutational frequency and specificity of this adduct.


Subject(s)
2-Naphthylamine/analogs & derivatives , 2-Naphthylamine/toxicity , Carcinogens/toxicity , DNA Adducts/metabolism , 2-Naphthylamine/metabolism , Animals , Base Sequence , COS Cells , Carcinogens/metabolism , Chlorocebus aethiops , DNA Adducts/genetics , Deoxyguanosine/genetics , Deoxyguanosine/metabolism , Genetic Vectors , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides/chemical synthesis , Oligodeoxyribonucleotides/metabolism
12.
Mol Cancer Ther ; 3(10): 1319-27, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15486199

ABSTRACT

HTI-286, a synthetic analogue of hemiasterlin, depolymerizes microtubules and is proposed to bind at the Vinca peptide site in tubulin. It has excellent in vivo antitumor activity in human xenograft models, including tumors that express P-glycoprotein, and is in phase II clinical evaluation. To identify potential mechanisms of resistance induced by HTI-286, KB-3-1 epidermoid carcinoma cells were exposed to increasing drug concentrations. When maintained in 4.0 nmol/L HTI-286, cells had 12-fold resistance to HTI-286. Cross-resistance was observed to other Vinca peptide-binding agents, including hemiasterlin A, dolastatin-10, and vinblastine (7- to 28-fold), and DNA-damaging drugs, including Adriamycin and mitoxantrone (16- to 57-fold), but minimal resistance was seen to taxanes, epothilones, or colchicine (1- to 4-fold). Resistance to HTI-286 was retained when KB-HTI-resistant cells were grown in athymic mice. Accumulation of [(3)H]HTI-286 was lower in cells selected in intermediate (2.5 nmol/L) and high (4.0 nmol/L) concentrations of HTI-286 compared with parental cells, whereas accumulation of [(14)C]paclitaxel was unchanged. Sodium azide treatment partially reversed low HTI-286 accumulation, suggesting involvement of an ATP-dependent drug pump. KB-HTI-resistant cells did not overexpress P-glycoprotein, breast cancer resistance protein (BCRP/ABCG2/MXR), MRP1, or MRP3. No mutations were found in the major beta-tubulin isoform. However, 4.0 nmol/L HTI-286-selected cells had a point mutation in alpha-tubulin that substitutes Ser for Ala(12) near the nonexchangeable GTP-binding site of alpha-tubulin. KB-HTI-resistant cells removed from drug became less resistant to HTI-286, no longer had low HTI-286 accumulation, and retained the Ala(12) mutation. These data suggest that HTI-286 resistance may be partially mediated by mutation of alpha-tubulin and by an ATP-binding cassette drug pump distinct from P-glycoprotein, ABCG2, MRP1, or MRP3.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Oligopeptides/pharmacology , Point Mutation , Tubulin/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , Adenosine Triphosphate/chemistry , Alanine/chemistry , Animals , Cell Line, Tumor , Cell Proliferation , Codon , DNA Damage , DNA, Complementary/metabolism , Depsipeptides , Dimerization , Doxorubicin/pharmacology , Humans , Mice , Mice, Nude , Mitoxantrone/pharmacology , Models, Molecular , Multidrug Resistance-Associated Proteins/biosynthesis , Mutation , Neoplasm Proteins/biosynthesis , Neoplasm Transplantation , Protein Conformation , Sequence Analysis, DNA , Sodium Azide/pharmacology , Time Factors , Tubulin/chemistry , Vinblastine/pharmacology
13.
Mol Cancer Ther ; 2(9): 873-84, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14555706

ABSTRACT

The taxanes, paclitaxel (PTX) and docetaxel (DTX), belong to a novel class of anticancer drugs that stabilize microtubules and lead to tumor cell death. While both agents are widely used for the treatment of lung, breast, and ovarian cancer, many tumor types are refractory or develop resistance to these drugs. We describe here a novel analogue of DTX, designated MAC-321 [Microtubule/Apoptosis/Cytotoxic: 5beta, 20-epoxy-1, 2alpha-, 4-, 7beta-, 10beta-, 13alpha-hexahydroxytax-11-en-9-one 4 acetate 2 benzoate 7-propionate 13-ester with (2R,3S)-N-tertbutoxycarbonyl-3-(2-furyl)isoserine], that overcomes P-glycoprotein-mediated resistance to PTX and DTX in preclinical model systems. Similar to PTX or DTX, MAC-321 enhanced the rate of tubulin polymerization in vitro and caused the bundling of microtubules in cells. MAC-321 inhibited proliferation of a panel of 14 tumor cell lines with minimal variation in potency (IC(50) = 2.2 +/- 1.4 nM; range = 0.6-5.3 nM). Unlike PTX or DTX, the IC(50) of MAC-321 did not vary in cells that expressed low to moderate levels of P-glycoprotein. Even under extraordinary conditions in KB-V1 cells, which highly overexpress P-glycoprotein, resistance to MAC-321 was 80-fold compared with that of PTX (1400-fold) and DTX (670-fold). In addition, equivalent or less resistance to MAC-321 compared with PTX or DTX was observed in four cell lines that contain distinct point mutations within the taxane-binding site of beta-tubulin. Most importantly, MAC-321 displayed superior in vivo efficacy because: (a) MAC-321 either partially or completely inhibited tumor growth in three tumor models that overexpressed P-glycoprotein and were resistant to PTX; and (b) unlike PTX or DTX, MAC-321 was highly effective when given orally. MAC-321 was also highly effective when given as single i.v. dose. Our findings suggest that MAC-321, which is currently under clinical evaluation, may have broad therapeutic value.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Neoplasms, Experimental/pathology , Paclitaxel/pharmacology , Taxoids/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Administration, Oral , Animals , Cell Division/drug effects , Docetaxel , Female , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Microtubules/drug effects , Neoplasms, Experimental/metabolism , Paclitaxel/analogs & derivatives , Tubulin/biosynthesis , Tubulin/drug effects , Tubulin/isolation & purification , Tumor Cells, Cultured
14.
Cancer Res ; 63(8): 1838-45, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12702571

ABSTRACT

Hemiasterlin is a natural product derived from marine sponges that, like other structurally diverse peptide-like molecules, binds to the Vinca-peptide site in tubulin, disrupts normal microtubule dynamics, and, at stoichiometric amounts, depolymerizes microtubules. Total synthesis of hemiasterlin and its analogues has been accomplished, and optimal pharmacological features of the series have been explored. The biological profile of one analogue, HTI-286, was studied here. HTI-286 inhibited the polymerization of purified tubulin, disrupted microtubule organization in cells, and induced mitotic arrest, as well as apoptosis. HTI-286 was a potent inhibitor of proliferation (mean IC(50) = 2.5 +/- 2.1 nM in 18 human tumor cell lines) and had substantially less interaction with multidrug resistance protein (P-glycoprotein) than currently used antimicrotubule agents, including paclitaxel, docetaxel, vinorelbine, or vinblastine. Resistance to HTI-286 was not detected in cells overexpressing the drug transporters MRP1 or MXR. In athymic mice implanted with human tumor xenografts, HTI-286 administered i.v. in saline inhibited the growth of numerous human tumors derived from carcinoma of the skin, breast, prostate, brain, and colon. Marked tumor regression was observed when used on established tumors that were >1 gram in size. Moreover, HTI-286 inhibited the growth of human tumor xenografts (e.g., HCT-15, DLD-1, MX-1W, and KB-8-5) where paclitaxel and vincristine were ineffective because of inherent or acquired resistance associated with P-glycoprotein. Efficacy was also achieved with p.o. administration of HTI-286. These data suggest that HTI-286 has excellent preclinical properties that may translate into superior clinical activity, as well as provide a useful synthetic reagent to probe the drug contact sites of peptide-like molecules that interact with tubulin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Agents/pharmacology , Microtubules/drug effects , Oligopeptides/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Animals , Cattle , Cell Cycle/drug effects , Cell Division/drug effects , Drug Resistance, Neoplasm , Female , Humans , KB Cells , Mice , Mice, Nude , Microtubules/metabolism , Tubulin/metabolism , Xenograft Model Antitumor Assays
15.
Biochemistry ; 41(48): 14255-62, 2002 Dec 03.
Article in English | MEDLINE | ID: mdl-12450390

ABSTRACT

Comparative mutagenesis studies of N-(2'-deoxyguanosin-8-yl)-2-acetylaminofluorene (dG-AAF) and N-(2'-deoxyguanosin-8-yl)-2-aminofluorene (dG-AF) adducts positioned in the Nar I restriction enzyme site were performed using Escherichia coli (E. coli) and simian kidney (COS-7) cells. Oligodeoxynucleotides ((5)(')TCCTCG(1)G(2)CG(3)CCTCTC) containing a recognition sequence for the Nar I restriction enzyme were modified site-specifically with dG-AAF or dG-AF. Modified and unmodified oligomers inserted into single-stranded phagemid shuttle vectors were used to transform E. coli or to transfect COS-7 cells. Following replication in host cells, progeny plasmids were recovered and analyzed for mutations. In SOS-induced E. coli, dG-AAF primarily induced one- and two-base deletions. The mutational frequency varied, depending on the position modified in the Nar I site; 91% two-base deletions were observed at G(3), while 8.4% and 2.8% deletions were detected at G(2) and G(1), respectively. In contrast, dG-AF at any position in the Nar I site failed to produce deletions, generating primarily G --> T transversions (mutational frequency, 7.6-8.4%). In COS-7 cells, both dG-AAF and dG-AF primarily induced G --> T transversions. Mutation frequencies for dG-AAF were 9.4-24%, the highest values being at G(1) and G(3). Mutation frequencies for dG-AF were 9.3-21%, the higher value at G(2). We conclude from this study that the mutation potential of dG-AAF and dG-AF depends on the structure of the adduct, the sequence context of the lesion, and the host cell used for the experiment.


Subject(s)
2-Acetylaminofluorene/analogs & derivatives , 2-Acetylaminofluorene/pharmacology , DNA Adducts/pharmacology , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/pharmacology , Deoxyribonucleases, Type II Site-Specific/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Fluorenes/pharmacology , Mutagens/chemistry , 2-Acetylaminofluorene/chemistry , Animals , COS Cells/drug effects , COS Cells/metabolism , Chlorocebus aethiops , DNA Adducts/chemical synthesis , DNA Adducts/chemistry , DNA Mutational Analysis , DNA, Single-Stranded/chemical synthesis , Deoxyguanosine/chemistry , Deoxyribonucleases, Type II Site-Specific/chemistry , Fluorenes/chemistry , Frameshift Mutation , Mutagens/chemical synthesis , Mutagens/pharmacology , Oligodeoxyribonucleotides/chemical synthesis , Plasmids/chemical synthesis , Point Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...