Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Anal Biochem ; 275(2): 141-7, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10552897

ABSTRACT

A sensitive fluorescence resonance energy transfer method was developed for the direct measurement of the dissociation constants of stromelysin inhibitors. The method is applied to the thiadiazole class of stromelysin inhibitors and it takes advantage of the fact that, upon binding to the active site of enzyme, the thiadiazole ring, with its absorbance centered at 320 nm, is able to quench the fluorescence of the tryptophan residues surrounding the catalytic site. The changes in fluorescence are proportional to the occupancy of the active site: Analysis of the fluorescence versus inhibitor concentration data yields dissociation constants that are in agreement with the corresponding competitive inhibitory constants measured by a catalytic rate assay. The affinity of nonthiadiazole inhibitors of stromelysin-such as hydroxamic acids and others-can be determined from the concentration-dependent displacement of a thiadiazole of known affinity. Using this displacement method, we determined the affinities of a number of structurally diverse inhibitors toward stromelysin. Since the three tryptophan residues located in the vicinity of the active site of stromelysin are conserved in gelatinase and collagenase, the method should also be applicable to inhibitors of other matrix metalloproteinases.


Subject(s)
Enzyme Inhibitors/metabolism , Matrix Metalloproteinase 3/metabolism , Catalytic Domain , Kinetics , Matrix Metalloproteinase 3/chemistry , Protein Binding , Spectrometry, Fluorescence
2.
J Biomol NMR ; 15(1): 55-64, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10549133

ABSTRACT

This report describes the backbone amide dynamics of the uniformly 15N labeled catalytic domain of human stromelysin complexed to PNU-99533, a hydroxamate-containing ligand that binds to the S'1-S'3 region (right side) of the stromelysin active site, and to PNU-107859 and PNU-142372, both thiadiazole-containing ligands that bind to the S1-S3 region (left side) of the stromelysin active site. 15N R1, R2 and NOE NMR relaxation measurements were recorded and analyzed for each complex. Different dynamic behaviors were observed for stromelysin complexed to the two classes of ligands, indicating that it may be possible to use protein dynamics to distinguish between different binding orientations. In the absence of bound ligand at the S1-S3 subsites, the S1-S3 residues were found to be relatively rigid. In contrast, the S'1-S'3 subsites were found to be flexible in the absence of interactions with ligand. The relative rigidness of the S1-S3 subsites may be responsible for MMP binding specificity by discriminating between ligands of different shapes. By contrast, the inherent flexibility of the S'1-S'3 subsites allows structural rearrangement to accommodate a broad range of incoming substrates or inhibitors. Similarities and differences in dynamics observed for each complex provide insights into the interactions responsible for protein-ligand recognition. The relevance of protein dynamics to structure-based drug design is discussed.


Subject(s)
Matrix Metalloproteinase 3/chemistry , Matrix Metalloproteinase Inhibitors , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , In Vitro Techniques , Ligands , Magnetic Resonance Spectroscopy , Matrix Metalloproteinase 3/metabolism , Models, Molecular , Nitrogen Isotopes , Protein Conformation , Thiadiazoles/chemistry , Thiadiazoles/metabolism , Thiadiazoles/pharmacology , Urea/analogs & derivatives , Urea/chemistry , Urea/metabolism , Urea/pharmacology
3.
Biochim Biophys Acta ; 1434(2): 304-16, 1999 Oct 12.
Article in English | MEDLINE | ID: mdl-10525149

ABSTRACT

Interactions of stromelysin with a series of inhibitors representative of three chemical templates with distinct binding modes were examined. Unfolding temperatures for inhibitor complexes were 10 degrees C to 15 degrees C greater than for apo stromelysin. Minor changes in ellipticity in the far-UV CD spectra of complexes indicated that ligand-induced conformational changes were localized to the binding site and did not involve gross changes in protein folding. Isothermal titrating calorimetry of thiadiazole-containing inhibitors, which bind in the S(1)-S(3) subsites of stromelysin, indicated that the binding interaction was exothermic and only slightly favorable entropically. Near-UV CD spectra showed large positive ellipticity increases from 250 to 300 nm, consistent with an interaction between the benzene ring of the inhibitor and stromelysin residues Tyr155 and Tyr168. Interactions between stromelysin and amide-hydroxamate ligands, which bind in the S(')(1)-S(')(3) subsites, were found to be both enthalpically and entropically driven. Binding of this class of ligands resulted in modest negative ellipticity changes at 260-285 nm and positive increases at 292 nm. Stromelysin complexed to a lactam-hydroxamate inhibitor with structure extending into both the S(1)-S(3) and S(')(1)-S(')(3) subsites showed increased ellipticity at 245 nm and negative changes at 260-285 and 295 nm.


Subject(s)
Enzyme Inhibitors/chemistry , Matrix Metalloproteinase Inhibitors , Binding Sites , Calorimetry, Differential Scanning , Circular Dichroism , Drug Design , Molecular Structure , Regression Analysis , Thermodynamics , Thiadiazoles/chemistry , Urea/analogs & derivatives , Urea/chemistry
4.
J Med Chem ; 42(9): 1525-36, 1999 May 06.
Article in English | MEDLINE | ID: mdl-10229623

ABSTRACT

The synthesis and enzyme inhibition data for a series of thiadiazole urea matrix metalloproteinase (MMP) inhibitors are described. A broad screening effort was utilized to identify several thiadiazoles which were weak inhibitors of stromelysin. Optimization of the thiadiazole leads to include an alpha-amino acid side chain with variable terminal amide substituents provided a series of ureas which were moderately effective stromelysin inhibitors, with Ki's between 0.3 and 1.0 microM. The most effective analogues utilized an L-phenylalanine as the amino acid component. In particular, unsubstituted 46 had a Ki of 710 nM, while the p-fluoro analogue 52 displayed increased potency (100 nM). Stromelysin inhibition was further improved using a pentafluorophenylalanine substituent which resulted in 70, a 14 nM inhibitor. While gelatinase inhibition was generally poor, the use of 1-(2-pyridyl)piperazine as the amide component usually provided for enhanced activity, with 71 inhibiting gelatinase with a Ki of 770 nM. The combination of this heterocycle with a p-fluorophenylalanine substituent provided the only analogue, 69, with collagenase activity (13 microM). The SAR for analogues described within this series can be rationalized through consideration of the X-ray structure recently attained for70 complexed to stromelysin. Uniquely, this structure showed the inhibitor to be completely orientated on the left side of the enzyme cleft. These results suggest that thiadiazole urea heterocycles which incorporate a substituted phenylalanine can provide selective inhibitors of stromelysin. Careful selection of the amide substituent can also provide for analogues with modest gelatinase inhibition.


Subject(s)
Matrix Metalloproteinase Inhibitors , Protease Inhibitors/chemical synthesis , Thiadiazoles/chemical synthesis , Urea/analogs & derivatives , Urea/chemical synthesis , Binding Sites , Fluorescence , Humans , Models, Molecular , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Structure-Activity Relationship , Thiadiazoles/chemistry , Thiadiazoles/pharmacology , Urea/chemistry , Urea/pharmacology
5.
J Protein Chem ; 17(7): 699-712, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9853686

ABSTRACT

The active site of the catalytic domain of stromelysin-1 (matrix metalloproteinase-3, MMP-3) was probed by fluorescence quenching, lifetime, and polarization of its three intrinsic tryptophans and by the environmentally sensitive fluorescent reporter molecule bisANS. Wavelength-dependent acrylamide quenching identified three distinct emitting tryptophan species, only one of which changes its emission and fluorescence lifetime upon binding of the competitive inhibitor Batimastat. Significant changes in the tryptophan fluorescence polarization occur upon binding by any of the three hydroxamate inhibitors Batimastat, CAS108383-58-0, and Celltech CT1418, all of which bind in the P2'-P3' region of the active site. In contrast, the inhibitor CGS27023A, which is thought to bind in the P1-P1' region, does not induce any change in tryptophan fluorescence polarization. The use of the fluorescent probe bisANS revealed the existence of an auxiliary binding site extrinsic to the catalytic cleft. BisANS acts as a competitive inhibitor of stromelysin with a dissociation constant of Ki=22 microM. In addition to this binding to the active site, it also binds to the auxiliary site with a dissociation constant of 3.40+/-0.17 microM. The auxiliary site is open, hydrophobic, and near the fluorescing tryptophans. The binding of bisANS to the auxiliary site is greatly enhanced by Batimastat, but not by the other competitive inhibitors tested.


Subject(s)
Anilino Naphthalenesulfonates , Fluorescent Dyes , Matrix Metalloproteinase 3/chemistry , Pyrazines , Tryptophan , Acrylamides , Binding Sites , Catalytic Domain , Fluorescence Polarization , Hydroxamic Acids/pharmacology , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase Inhibitors , Models, Chemical , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Protease Inhibitors/pharmacology , Protein Binding , Spectrometry, Fluorescence , Sulfonamides/pharmacology , Thiophenes/pharmacology
6.
Protein Sci ; 7(11): 2281-6, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9827994

ABSTRACT

Unregulated or overexpressed matrix metalloproteinases (MMPs), including stromelysin, collagenase, and gelatinase. have been implicated in several pathological conditions including arthritis and cancer. Small-molecule MMP inhibitors may have therapeutic value in the treatment of these diseases. In this regard, the solution structures of two stromelysin/ inhibitor complexes have been investigated using 1H, 13C, and 15N NMR spectroscopy. Both-inhibitors are members of a novel class of matrix metalloproteinase inhibitor that contain a thiadiazole group and that interact with stromelysin in a manner distinct from other classes of inhibitors. The inhibitors coordinate the catalytic zinc atom through their exocyclic sulfur atom, with the remainder of the ligand extending into the S1-S3 side of the active site. The binding of inhibitor containing a protonated or fluorinated aromatic ring was investigated using 1H and 19F NMR spectroscopy. The fluorinated ring was found to have a reduced ring-flip rate compared to the protonated version. A strong, coplanar interaction between the fluorinated ring of the inhibitor and the aromatic ring of Tyr155 is proposed to account for the reduced ring-flip rate and for the increase in binding affinity observed for the fluorinated inhibitor compared to the protonated inhibitor. Binding interactions observed for the thiadiazole class of ligands have implications for the design of matrix metalloproteinase inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Matrix Metalloproteinase 3/chemistry , Matrix Metalloproteinase Inhibitors , Thiadiazoles/chemistry , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Protein Conformation , Solutions , Thiadiazoles/metabolism , Urea/analogs & derivatives , Urea/chemistry , Urea/metabolism , Zinc/chemistry
7.
Protein Sci ; 7(10): 2118-26, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9792098

ABSTRACT

The binding of two 5-substituted-1,3,4-thiadiazole-2-thione inhibitors to the matrix metalloproteinase stromelysin (MMP-3) have been characterized by protein crystallography. Both inhibitors coordinate to the catalytic zinc cation via an exocyclic sulfur and lay in an unusual position across the unprimed (P1-P3) side of the proteinase active site. Nitrogen atoms in the thiadiazole moiety make specific hydrogen bond interactions with enzyme structural elements that are conserved across all enzymes in the matrix metalloproteinase class. Strong hydrophobic interactions between the inhibitors and the side chain of tyrosine-155 appear to be responsible for the very high selectivity of these inhibitors for stromelysin. In these enzyme/inhibitor complexes, the S1' enzyme subsite is unoccupied. A conformational rearrangement of the catalytic domain occurs that reveals an inherent flexibility of the substrate binding region leading to speculation about a possible mechanism for modulation of stromelysin activity and selectivity.


Subject(s)
Matrix Metalloproteinase Inhibitors , Thiadiazoles/chemistry , Urea/analogs & derivatives , Animals , Binding Sites/physiology , Collagenases/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hydrogen Bonding , Models, Molecular , Molecular Structure , Protein Binding/physiology , Protein Conformation , Thiadiazoles/pharmacology , Urea/chemistry , Urea/pharmacology , Vertebrates , Zinc/chemistry
8.
Cancer Res ; 58(17): 4001-7, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9731514

ABSTRACT

The solubilization of plasma membrane receptors through proteolytic cleavage of the ligand binding domain at the cell surface is an important mechanism for regulating cytokine function and receptor signaling. The inhibition of the shedding of a variety of receptors by synthetic inhibitors of the matrix metalloproteinases (MMPs) implicates metalloproteinases in this regulatory event. We examined the effects of two naturally occurring tissue inhibitors of metalloproteinases, TIMP-1 and TIMP-2, and several synthetic MMP inhibitors (MMPIs) on the shedding of both tumor necrosis factor alpha receptor type I (TNFalpha-RI; Mr 55,000) and TNFalpha-RII (Mr 75,000) by the Colo 205 human colon adenocarcinoma cell line. Culture of Colo 205 cells for 48 h resulted in the shedding of both TNFalpha-RI and TNFalpha-RII, as determined by ELISA. The shedding of TNFalpha receptors was not affected by TIMP-1 or protease inhibitors aprotinin, pepstatin, or leupeptin but was inhibited in a dose-dependent manner by the following synthetic MMPIs: batimastat and marimastat (BB-94 and BB-2516, respectively, British Biotech, Inc.); CT1418 (Celltech Therapeutics); CGS27023A (Novartis Pharmaceuticals); and RO31-9790 (Roche), with IC50s ranging from 3.2 to 38.0 microM. Similarly, TIMP-2 from two different sources reproducibly inhibited the shedding of both TNFalpha-RI and TNFalpha-RII in a dose-dependent manner (IC50 = 286 +/- 33 nM for TNFalpha-RI shedding and 462 +/- 52 nM for shedding of TNFalpha-RII). The inhibition of TNFalpha-RI shedding was confirmed in the SW626 human ovarian adenocarcinoma cell line. The synthetic MMPIs and TIMP-2, but not TIMP-1, also caused a dose-dependent increase in the number of TNFalpha receptors retained on the surface of Colo 205 cells, as determined by flow cytometry. Inhibition of TNFalpha receptor shedding with TIMP-2 occurs at molar concentrations 10-100 times less than those required with low molecular weight, synthetic MMPIs but at concentrations greater than those required to inhibit collagen degradation. Modulation of TNFalpha receptor shedding by TIMP-2 could have important implications for the pleiotropic effects of TNFalpha in both normal and malignant cells and for the pharmacological activity of synthetic MMPIs.


Subject(s)
Adenocarcinoma/metabolism , Colonic Neoplasms/metabolism , Metalloendopeptidases/antagonists & inhibitors , Protease Inhibitors/pharmacology , Receptors, Tumor Necrosis Factor/drug effects , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Binding Sites , Humans , Receptors, Tumor Necrosis Factor/metabolism , Tumor Cells, Cultured
9.
Cancer Res ; 52(20): 5687-92, 1992 Oct 15.
Article in English | MEDLINE | ID: mdl-1394193

ABSTRACT

Adozelesin (U-73975) is an extremely potent cytotoxic agent which causes 90% lethality, after 2 h exposure in vitro, of Chinese hamster ovary and lung (CHO and V79), mouse melanoma (B16), and human ovarian carcinoma (A2780) cells at 0.33, 0.19, 0.2, and 0.025 ng/ml, respectively. Under similar conditions, Adriamycin and cisplatin had 90% lethality values in CHO cells of 150 ng/ml (= 249 nM) and 6800 ng/ml (= 2266 nM), respectively. The relative drug sensitivity of the cell lines (A2780 > V79, B16, CHO) was correlated to the relative amounts of [3H]adozelesin alkylated to DNA. The greater sensitivity of A2780 was due to (a) greater DNA alkylation at different drug doses and (b) greater intrinsic sensitivity of A2780 which resulted in greater cell kill at comparable DNA alkylation. Phase specific toxicity studies show that adozelesin was least lethal to CHO cells in mitosis and very early G1. Lethality increased as cells progressed through G1 and was maximal in late G1 and early S. Mitotic cells had lower drug uptake and correspondingly less drug binding to DNA than G1 or S-phase cells. However, based on the amount of drug alkylated per micrograms of DNA, cells in M, G1, and S were equally sensitive. Therefore, the lower sensitivity of M-phase cells was due to lower drug uptake. Adozelesin had three different effects on progression of CHO, V79, B16, and A2780 through the cell cycle: (a) slowed progression through S which resulted in significantly increasing the percentage of S-phase cells. This effect was transient; (b) cell progression was blocked in G2 for a long time period; (c) the response of the cell lines to the G2 block differed. CHO and V79 cells escaped G2 block by dividing and entered the diploid DNA cycle or did not undergo cytokinesis and became tetraploid. On the contrary, B16 and A2780 cells remained blocked in G2 and did not become tetraploid. Cell progression was inhibited in a similar manner when a synchronized population of M, G1, or S-phase cells were exposed to adozelesin.


Subject(s)
Antineoplastic Agents/toxicity , Cell Cycle/drug effects , Cell Survival/drug effects , Cyclohexanecarboxylic Acids/toxicity , DNA/drug effects , Growth Inhibitors/toxicity , Indoles , Alkylation/drug effects , Animals , Benzofurans , CHO Cells/cytology , CHO Cells/drug effects , Cell Death/drug effects , Cricetinae , Cyclohexenes , Duocarmycins , Female , G1 Phase/drug effects , G2 Phase/drug effects , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Mitosis/drug effects , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , S Phase/drug effects , Time Factors , Tumor Cells, Cultured
10.
Cancer Res ; 52(18): 4904-13, 1992 Sep 15.
Article in English | MEDLINE | ID: mdl-1516047

ABSTRACT

The cyclopropylpyrroloindole analogues are DNA minor-groove binders containing a cyclopropyl group, which mediates N3-adenine covalent adduct formation in a sequence-selective fashion. Carzelesin (U-80244) is a cyclopropylpyrroloindole prodrug containing a relatively nonreactive chloromethyl precursor to the cyclopropyl function. Activation of carzelesin requires two steps, (a) hydrolysis of a phenylurethane substituent to form U-76073, followed by (b) ring closure to form the cyclopropyl-containing DNA-reactive U-76074. The formation of the DNA-reactive U-76074, via U-76073, from carzelesin was shown to proceed very slowly in phosphate-buffered saline (t1/2 greater than 24 h) but to occur rapidly in plasma from mouse, rat, dog, and human (initial t1/2 values ranging from 18 min for mouse to 52 min for rat) and in cell culture medium (t1/2 approximately 40 min). Although carzelesin was less potent in terms of in vitro cytotoxicity and in vivo optimal dosage and showed low affinity for binding to DNA, it was therapeutically more efficacious against mouse L1210 leukemia than was U-76074 or adozelesin (U-73975), another cyclopropylpyrroloindole analogue which is currently in phase I clinical trials. Carzelesin also proved to be more efficacious than U-76074 or adozelesin against mouse pancreatic ductal 02 adenocarcinoma, a system reported to be resistant to every agent tested. Carzelesin was highly effective against this tumor and produced 97% tumor growth inhibition. In addition, i.v. administered carzelesin showed significant activity (National Cancer Institute criteria) against i.v. or s.c. implanted Lewis lung carcinoma, i.p. or s.c. implanted B16 melanoma, s.c. implanted colon 38 carcinoma, and five s.c. implanted human tumor xenografts, including clear cell Caki-1 carcinoma, colon CX-1 adenocarcinoma, lung LX-1 tumor, ovarian 2780 carcinoma, and prostatic DU-145 carcinoma. Carzelesin treatment produced 100% complete remissions (no palpable tumor mass at the termination of the experiment) in mice bearing early-stage human ovarian 2780. Pharmacologically, carzelesin proved to be relatively schedule and route independent and was highly active against i.p. implanted L1210 leukemia, regardless of whether the analogue was given i.v., i.p., s.c., or p.o. These results, collectively, suggest that carzelesin is absorbed and distributed well. Both carzelesin and adozelesin caused marked tumor shrinkage in mice bearing human lung LX-1 or advanced-stage human ovarian 2780 carcinoma; however, tumor regrowth occurred shortly after the treatment with adozelesin was stopped. Little or no apparent tumor regrowth occurred after treatment with carzelesin.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Antineoplastic Agents , Benzofurans , Indoles/toxicity , Prodrugs/toxicity , Adenocarcinoma/drug therapy , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Culture Media , Duocarmycins , Indoles/pharmacokinetics , Leukemia L1210/drug therapy , Metabolic Clearance Rate , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/drug therapy , Plasma/metabolism , Prodrugs/pharmacokinetics , Transplantation, Heterologous
11.
Cancer Res ; 50(8): 2485-92, 1990 Apr 15.
Article in English | MEDLINE | ID: mdl-2317831

ABSTRACT

U-71,184 is a CC-1065 analogue which is highly cytotoxic in vitro and has a broad spectrum of antitumor activity in vivo. Against B16 cells, U-71,184 was 8-fold and 253-fold more potent than Actinomycin D and Adriamycin, respectively. U-71,184 killed 90% of B16 cells at 0.01 ng/ml levels of drug in the medium, which was equivalent to an intracellular concentration of about 8 pg/10(6) cell (= 2 x 10(-8) pmol/cell). A B16 cell line resistant to U-71,184 developed after 3 months of in vitro exposure to gradually increasing concentrations of the drug. The sensitive and resistant cell lines were cloned and a B16/R clone was selected which was 60 to 100 times more resistant to U-71,184 than the cloned sensitive parent (B16/S). Cells grown in the absence of U-71,184 for 2 months retained resistance to the drug. B16/R was slightly cross-resistant only to Adriamycin but not to Actinomycin D, vinblastine, or colchicine. Among alkylating agents, it was slightly cross-resistant to Melphalan but not to 1,3-bis(2-chloroethyl)-1-nitrosourea or cisplatin. B16/R did not overexpress mdr mRNA. Therefore, this cell line does not exhibit the multidrug-resistant phenotype. Most karyotypes of B16/R had a marker chromosome which carried an aberrantly staining region apparently containing repetitive replication of the same segment. Resistance can be partly accounted for by the approximately 10-fold lesser uptake of [3H]-U-71,184 in B16/R, as compared to B16/S. B16/R was cross-resistant in varying degrees to several other CC-1065 analogues. The ratio of the 50% lethal dose of U-71,184 for B16/R, as compared to B16/S, was about 60 (i.e., R/S = 60). In comparison, the following compounds had an R/S ratio of less than 20 (i.e., modest level of cross-resistance to U-71,184): U-68,819, U-73,975, U-75,500, U-75,559, and CC-1065. In contrast, the following compounds had an R/S ratio greater than 20 (i.e., highly cross-resistant to U-71,184): U-71,184 analogues U-71,185, U-73,903, and U-75,012; U-73,975 analogues U-75,613, U-75,032, and U-73,896; and CC-1065 enantiomer U-76,915. We cannot yet explain the difference in the level of cross-resistance between these compounds in vitro. B16/S and B16/R cells were tumorigenic in mice and B16/R was resistant to U-71,184 in vivo. There was no clear indication of cross-resistance of B16/R in vivo to Adriamycin, Actinomycin D, cisplatin, or Melphalan. However, U-73,975, a compound with modest cross-resistance in vitro, was significantly cross-resistant in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance/genetics , Indoles/pharmacology , Melanoma, Experimental/genetics , Animals , Biological Transport , Cell Division/drug effects , Cell Line , Cell Survival/drug effects , Indoles/metabolism , Karyotyping , Kinetics , Mice , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
12.
Chem Biol Interact ; 67(3-4): 199-213, 1988.
Article in English | MEDLINE | ID: mdl-3191534

ABSTRACT

The factors influencing the binding of CC-1065 to DNA were examined using racemic analogs with varying chain lengths. The ability of these agents to bind DNA appeared to be related to cytotoxic potency, however this did not appear to be a direct quantitative correlation. Two enantiomers of a bis-indole analog of CC-1065 were studied for DNA binding and cytotoxic activity. The agent with the same stereochemical configuration as CC-1065 was a potent cytotoxin, but its enantiomer was essentially inactive. Both enantiomers showed significant binding to DNA, but the biologically less active isomer showed less overall binding. In all cases, the agents preferred AT-rich DNA, and all bound to similar regions in DNA as evidenced by positions of drug-initiated thermal breaks in single end-labelled fragments of phi X 174RF DNA. The overall similarity in site specificity for binding of the structurally diverse agents suggests that much of the specificity observed in binding of the agent to DNA lies in the DNA itself. Thus, it may be difficult to change minor groove specificity for agents of this type simply by designing structures that can encompass guanine or cytosine residues. Other modifications, such as changing the specificity of the alkylating moiety, may be required to achieve this goal.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage , DNA/metabolism , Leucomycins/pharmacology , Animals , Antineoplastic Agents/toxicity , Base Sequence , Cell Survival/drug effects , Circular Dichroism , DNA, Superhelical/drug effects , Duocarmycins , In Vitro Techniques , Indoles/pharmacology , Leucomycins/toxicity , Leukemia L1210 , Mercaptoethanol/pharmacology , Piperidines/pharmacology , Stereoisomerism , Structure-Activity Relationship
13.
Mutagenesis ; 1(6): 407-10, 1986 Nov.
Article in English | MEDLINE | ID: mdl-3331678

ABSTRACT

CC-1065, a very potent antitumor antibiotic, is active against several animal tumors, and against human tumors in the cloning assay at doses 50-1000 times lower than other agents such as adriamycin. It binds and alkylates DNA, and inhibits DNA synthesis, suggesting a potential for genotoxicity. Therefore, the genotoxic effects of CC-1065 were tested in several assay systems. CC-1065 was weakly mutagenic in the Ames Salmonella mutation assay (strain TA100) without S9 activation, but lacked mutagenic activity in TA98 with or without activation. CC-1065 was a very potent mutagen in the Salmonella forward mutation assay (induction of 8-azaguanine resistance), increasing the mutation frequency 19-fold over background at 0.1 ng/ml without activation. In mammalian (V79) cells it was a very potent mutagen without activation, increasing the mutation frequency 20-fold over background a 0.5 ng/ml. CC-1065 induced chromosome aberrations in V79 cells at very low (less than 0.1 ng/ml) doses, making this assay the most sensitive. CC-1065 increased the induction of micronuclei in rats 10- to 20-fold over the background at 200 and 400 micrograms/kg, but not at 100 micrograms/kg. CC-1065 failed to cause DNA breaks or DNA--protein cross-links as measured by the DNA damage/alkaline elution assay.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Indoles , Leucomycins/toxicity , Mutagens , Animals , Cell Line , Cell Nucleus/drug effects , Cell Survival/drug effects , Chromosome Aberrations , DNA/drug effects , Drug Resistance/genetics , Duocarmycins , Male , Mutagenicity Tests/methods , Rats , Rats, Inbred Strains , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Sister Chromatid Exchange/drug effects , Thioguanine/pharmacology
14.
Cancer Lett ; 33(1): 75-81, 1986 Oct.
Article in English | MEDLINE | ID: mdl-3768862

ABSTRACT

To investigate the hypothesis that the similarity of dose-response curves for induction of thymic lymphoma in C57BL mice was due to similar DNA alkylation profiles for 1-ethyl-1-nitrosourea (ENU) and 1-(2-hydroxyethyl)-1-nitrosourea (HNU), we measured the reaction of the two agents with DNA in vitro and in target tissues in vivo. At equimolar doses, alkylation of DNA by HNU was about 20% greater than that by ENU in vitro. As a percentage of total DNA-bound alkyl groups, relative reaction at a minor groove site (3 of adenine) was similar for the two agents, but HNU caused greater relative alkylation at the major groove sites, O6 and N-7 of guanine. At equi-oncogenic doses, alkylation at the O6 of guanine in liver and thymus was similar for both agents, but O6-alkylguanine formation in bone marrow by HNU was almost twice that by ENU. Because alkylation at O6 of guanine has previously been shown to be a key procarcinogenic lesion in this system, these findings suggest the thymus, rather than the marrow as a primary target for tumor induction by these agents, although involvement of marrow alkylation cannot be ruled out.


Subject(s)
DNA Damage , DNA/metabolism , Ethylnitrosourea/analogs & derivatives , Alkylation , Animals , Bone Marrow/drug effects , Ethylnitrosourea/metabolism , Ethylnitrosourea/pharmacology , Guanine/metabolism , Mice , Thymus Gland/drug effects
15.
Chem Biol Interact ; 59(1): 55-72, 1986.
Article in English | MEDLINE | ID: mdl-3019583

ABSTRACT

In this work, we report on the binding of the novel antitumor agent CC-1065 to poly(dA).poly(dT) and to mixtures of dA and dT oligomers as determined by electronic absorption and circular dichroism (CD) methods. In addition, the DNA binding properties of CC-1065 and its binding mechanism are compared to those of netropsin. CC-1065 binds to the polymer by at least three mechanisms to produce one irreversibly and two reversibly bound species. One reversibly bound species is moderately stable, but in time (days), it converts to the irreversibly bound species. Both of these species bind within the minor groove of the polymer and exhibit intense CC-1065 induced CD spectra. The other reversibly bound species does not acquire an induced CD. CC-1065 forces B-form duplex formation between mixtures of single strand dA and dT oligomers and binds irreversibly to the duplexes without showing the presence of an intermediate, reversibly bound species. The induced CD increases with increasing length of the oligomer, from the 5-mer (barely detectable CD) to the 14-mer (intense CD). The 7-, 10- and 14-mer mixtures bind about 1, between 1 and 2, and between 2 and 3 CC-1065 molecules, respectively. Computer graphic models of the CC-1065-DNA complex show that the covalent adduct of CC-1065 and unreacted CC-1065 can attain the same close van der Waals contacts between adenine C2 hydrogens and antibiotic CH groups that were observed in the crystal structure of the netropsin-DNA complex. These contacts may account for the dA-dT base pair binding specificity of CC-1065 and for the stability of the reversibly bound CC-1065 species.


Subject(s)
Deoxyadenine Nucleotides , Indoles , Leucomycins , Poly dA-dT , Polydeoxyribonucleotides , Thymidine , Binding Sites , Chemical Phenomena , Chemistry , Chloroform , Circular Dichroism , Duocarmycins , Hydrogen Bonding , Netropsin , Nucleic Acid Conformation , Oligodeoxyribonucleotides , Phenols , Structure-Activity Relationship
16.
Science ; 226(4676): 843-4, 1984 Nov 16.
Article in English | MEDLINE | ID: mdl-6494915

ABSTRACT

Sequence-dependent variations in DNA revealed by x-ray crystallographic studies have suggested that certain DNA-reactive drugs may react preferentially with defined sequences in DNA. Drugs that wind around the helix and reside within one of the grooves of DNA have perhaps the greatest chance of recognizing sequence-dependent features of DNA. The antitumor antibiotic CC-1065 covalently binds through N-3 of adenine and resides within the minor groove of DNA. This drug overlaps with five base pairs for which a high sequence specificity exists.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Base Sequence , DNA/metabolism , Indoles , Leucomycins/metabolism , Binding Sites , Chemical Phenomena , Chemistry , Duocarmycins , Molecular Conformation , X-Ray Diffraction
17.
Cancer Res ; 42(7): 2821-8, 1982 Jul.
Article in English | MEDLINE | ID: mdl-7083173

ABSTRACT

CC-1065 (NSC 298223), a potent new antitumor antibiotic produced by Streptomyces zelensis, interacts strongly with double-stranded DNA and appears to exert its cytotoxic effects through disruption of DNA synthesis. We undertook this study to elucidate the sites and mechanisms of CC-1065 interaction with DNA. The binding of CC-1065 to synthetic and native DNA was examined by differential circular dichroism or by Sephadex chromatography with photometric detection. The binding of CC-1065 with calf thymus DNA was rapid, being complete within 2 hr, and saturated at 1 drug per 7 to 11 base pairs. The interaction of CC-1065 with synthetic DNA polymers indicated a specificity for adenine- and thymine-rich sites. Agarose gel electrophoresis of CC-1065-treated supercoiled DNA showed that CC-1065 did not intercalate. Site exclusion studies using substitutions in the DNA grooves showed CC-1065 to bind primarily in the minor groove. CC-1065 did not cause DNA breaks; it inhibited susceptibility of DNA to nuclease S1 digestion. It raised the thermal melting temperature of DNA, and it inhibited the ethidium-induced unwinding of DNA. Thus, in contrast to many antitumor agents, CC-1065 stabilized the DNA helix. DNA helix overstabilization may be relevant to the mechanism of action of CC-1065.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , DNA , Indoles , Leucomycins/pharmacology , Animals , Anthramycin/metabolism , Cattle , Circular Dichroism , DNA/metabolism , Deoxyribonucleases/pharmacology , Duocarmycins , Electrophoresis, Agar Gel , Hot Temperature , Leucomycins/metabolism , Thymus Gland/metabolism
18.
Mutat Res ; 58(1): 79-86, 1978 Sep.
Article in English | MEDLINE | ID: mdl-362195

ABSTRACT

Sixteen carcinogens were evaluated in rats for their ability to induce micronuclei. The direct acting agent, ethyl methanesulfonate and the procarcinogens/promutagens, cyclophosphamide and 4-nitroquinoline-1-oxide, induced dose-related increases in micronucleated polychromatophilic erythrocytes. Aflatoxin B1 also significantly increased the number of micronucleated polychromatophillic erythrocytes for 2 doses although no dose-response could be detected. Dimethylnitrosamine produced variable results. The remaining 11 compounds, 2-acetylaminofluorene, 4-aminobiphenyl, benzidine, diethylnitrosamine, dimethylbenzanthracene, 1,2-dimethylhydrazine, ethionine, ethyl carbamate, hexametapol, metronidazole, and beta-naphthylamine, failed to induce significantly increased numbers of micronuclei. The large number of false negative results obtained in the present investigations using the micronucleus test suggests that in vivo cytogenetic assays utilizing bone marrow may also lack the sensitivity needed to detect clastogenic effects of procarcinogens/promutagens which require tissue specific metabolic activation.


Subject(s)
Carcinogens/pharmacology , Cell Nucleus , Chromosome Aberrations , Mutagens , Animals , Bone Marrow/ultrastructure , Dose-Response Relationship, Drug , Genetic Techniques , Male , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...