Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 48(4): 1079-87, 2005 Feb 24.
Article in English | MEDLINE | ID: mdl-15715475

ABSTRACT

The extravascular diffusion of antitumor agents is a key determinant of their therapeutic activity, but the relationships between physicochemical properties of drugs and their extravascular transport are poorly understood. It is well-known that drug lipophilicity plays an important role in transport across biological membranes, but the net effect of lipophilicity on transport through multiple layers of tumor cells is less clear. This study examines the influence of lipophilicity (measured as the octanol-water partition coefficient P) on the extravascular transport properties of the hypoxic cytotoxin tirapazamine (TPZ, 1) and a series of 13 neutral analogues, using multicellular layers (MCLs) of HT29 human colon carcinoma cells as an in vitro model for the extravascular compartment of tumors. Flux of drugs across MCLs was determined using diffusion chambers, with the concentration-time profile on both sides of the MCL measured by HPLC. Diffusion coefficients in the MCLs (D(MCL)) were inversely proportional to M(r)(0.5) (M(r), relative molecular weight), although this was a minor contributor to differences between compounds over the narrow M(r) range investigated. Differences in lipophilicity had a larger effect, with a sigmoidal dependence of D(MCL) on log P. Correcting for M(r) differences, lipophilic compounds (log P > 1.5) had ca. 15-fold higher D(MCL) than hydrophilic compounds (log P < -1). Using a pharmacokinetic/pharmacodynamic (PK/PD) model in which diffusion in the extravascular compartment of tumors is considered explicitly, we demonstrated that hypoxic cell kill is very sensitive to changes in extravascular diffusion coefficient of TPZ analogues within this range. This study shows that simple monosubstitution of TPZ can alter log P enough to markedly improve extravascular transport and activity against target cells, especially if rates of metabolic activation are also optimized.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Triazines/pharmacokinetics , Antineoplastic Agents/chemistry , Biological Transport , Diffusion , HT29 Cells , Humans , Models, Biological , Solubility , Tirapazamine , Triazines/chemistry
2.
Cancer Res ; 64(2): 736-42, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14744792

ABSTRACT

Tirapazamine (TPZ), a bioreductive drug with selective toxicity for hypoxic cells in tumors, is currently in Phase III clinical trials. It has been suggested to have a dual mechanism of action, both generating DNA radicals and oxidizing these radicals to form DNA breaks; whether the second (radical oxidation) step is rate-limiting in cells is not known. In this study we exploit the DNA radical oxidizing ability of the 1-N-oxide metabolite of TPZ, SR 4317, to address this question. SR 4317 at high, but nontoxic, concentrations potentiated the hypoxic (but not aerobic) cytotoxicity of TPZ in all four of the human tumor cell lines tested (HT29, SiHa, FaDu, and A549), thus providing a 2-3-fold increase in the hypoxic cytotoxicity ratio. In potentiating TPZ, SR 4317 was 20-fold more potent than the hypoxic cell radiosensitizers misonidazole and metronidazole but was less potent than misonidazole as a radiosensitizer, suggesting that the initial DNA radicals from TPZ and radiation are different. SR 4317 had favorable pharmacokinetic properties in CD-1 nude mice; coadministration with TPZ provided a large increase in the SR 4317 plasma concentrations relative to that for endogenous SR 4317 from TPZ. It also showed excellent extravascular transport properties in oxic and anoxic HT29 multicellular layers (diffusion coefficient 3 x 10(-6) cm(2)s(-1), with no metabolic consumption). Coadministration of SR 4317 (1 mmol/kg) with TPZ at a subtherapeutic dose (0.133 mmol/kg) significantly enhanced hypoxic cell killing in HT29 tumor xenografts without causing oxic cell killing, and the combination at its maximum tolerated dose was less toxic to hypoxic cells in the retina than was TPZ alone at its maximum tolerated dose. This study demonstrates that benzotriazine mono-N-oxides have potential use for improving the therapeutic utility of TPZ as a hypoxic cytotoxin in cancer treatment.


Subject(s)
Antineoplastic Agents/toxicity , Cell Hypoxia/drug effects , Cell Survival/drug effects , Triazines/toxicity , Adenocarcinoma , Animals , Cell Line, Tumor , Colonic Neoplasms , Drug Synergism , Female , Humans , Lung Neoplasms , Mice , Pharyngeal Neoplasms , Tirapazamine , Triazines/chemical synthesis , Triazines/pharmacokinetics , Uterine Cervical Neoplasms
3.
Cancer Res ; 63(18): 5970-7, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-14522924

ABSTRACT

In common with other bioreductive drugs, metabolic reduction is required for activation of the benzotriazine-di-N-oxide tirapazamine (TPZ) in hypoxic regions of tumors. This same metabolism also consumes the drug as it diffuses, impeding its penetration into hypoxic tissue. In this study, we develop a pharmacokinetic (PK)/pharmacodynamic (PD) model for TPZ that explicitly includes its diffusion characteristics as measured in multicellular layer (MCL) cultures of HT29 colon carcinoma cells. The kinetics of TPZ metabolism to its mono-N-oxide derivative SR 4317, determined by high-performance liquid chromatography using anoxic HT29 single cell suspensions, demonstrated both a first order and saturable (K(m) = 3.6 micro M) component. Cell killing, assessed by clonogenic assay under the same conditions, demonstrated an approximately quadratic concentration dependence and linear time dependence. TPZ transport through MCLs, determined under hyperoxic conditions (95% O(2)) to suppress reductive metabolism, provided a concentration-independent diffusion coefficient of 0.40 x 10(-6) cm(2)s(-1). Under anoxia, this transport was strongly suppressed and was well predicted by the single cell metabolism parameters (scaled to the cell density in MCLs). These PK (transport) and PD (cytotoxicity) parameters were used to calculate cell killing as a function of distance in anoxic HT29 MCLs after the addition of TPZ to both sides of the MCL. The predicted average cell kill was in good agreement with measured values, which showed much less killing than for single cell suspensions under the same conditions. The success of this PK/PD model in predicting response in MCL shows that inefficient transport, rather than changes in intrinsic sensitivity, is responsible for TPZ resistance in these three-dimensional cell cultures and suggests that optimization of transport properties is a high priority in developing second-generation TPZ analogues.


Subject(s)
Antineoplastic Agents/pharmacology , Triazines/pharmacology , Antineoplastic Agents/pharmacokinetics , Biological Transport , Chromatography, High Pressure Liquid , Drug Resistance, Neoplasm , HT29 Cells , Humans , Models, Biological , Spheroids, Cellular , Tirapazamine , Triazines/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...