<<

[CANCER RESEARCH 46, 519-523, February 1986] Potentiation of 2'- Cytotoxicity by a Novel Inhibitor of Furine Phosphorylase, 8-Amino-9-benzylguanine1

Donna S. Shewach,2 Ji-Wang Chern, Katherine E. Pillóte,Leroy B. Townsend, and Peter E. Daddona3

Departments of Internal Medicine [D.S.S., P.E.D.], Biological Chemistry [P.E.D.], and Medicinal Chemistry [J-W.C., K.E.P., L.B.T.], University ol Michigan, Ann Arbor, Michigan 48109

ABSTRACT to the ADA-deficient disease state (2). PNP is an essential enzyme of the salvage pathway, We have synthesized and evaluated a series of 9-substituted catalyzing the phosphorolysis of , , and their analogues of 8-aminoguanine, a known inhibitor of human purine 2'- derivatives to the respective purine nucleoside phosphorylase (PNP) activity. The ability of these bases. To date, several inhibitors of PNP have been identified, agents to inhibit PNP has been investigated. All compounds were and most of these compounds resemble purine bases or nucleo found to act as competitive (with inosine) inhibitors of PNP, with sides. The most potent inhibitors exhibit apparent K¡values in K¡values ranging from 0.2 to 290 /¿M.Themost potent of these the range of 10~6to 10~7 M (9-12). Using partially purified human analogues, 8-amino-9-benzylguanine, exhibited a K, value that erythrocyte PNP, the diphosphate derivative of acyclovir dis was 4-fold lower than that determined for the parent base, 8- played K¡values of 5.1 x 10~7 to 8.7 x 10~9 M, depending on aminoguanine. As a metabolically stable compound in human the concentration of inorganic phosphate (13). However, it is not blood, 8-amino-9-benzylguanine was more effective than 8-ami known whether acyclovir diphosphate can attain an intracellular noguanine at potentiating the toxicity of 2'-deoxyguanosine to concentration sufficient to inhibit PNP during i.v. administration MOLT-4 T-lymphoblasts in culture. 8-Amino-9-benzylguanine is of the parent drug to patients. the most potent base or nucleoside inhibitor of human PNP This laboratory has previously reported the PNP inhibitory reported to date, and it is a promising lead compound in the activity of 8-AGR, an analogue of guanosine, in cultured human development of more effective PNP inhibitors. T-lymphoblasts (9). In addition to its activity as a competitive inhibitor of PNP, 8-AGR is a substrate for this enzyme. In fact, its phosphorolytic cleavage product, 8-AG, has been reported to INTRODUCTION be a more potent inhibitor of PNP (10). On the basis of these The discovery of T-cell-related immunodeficiency diseases findings, 8-AGR is assumed to be a more soluble pro-drug form associated with a lack of ADA4 or PNP in patient erythrocytes of 8-AG. 8-AGR can potentiate the cytotoxic effects of dGuo and permit increased accumulation of the 5'-triphosphate of dGuo in has prompted considerable interest in developing inhibitors of these enzymes as a selective approach to cancer chemotherapy mammalian cells in culture (9), thereby mimicking the PNP- (1,2). In the ADA- and PNP-related immunodeficiency states, 2'- deficiency state. While it has been shown that 8-AG can decrease or dGuo accumulates, respectively, and these the rate of dGuo phosphorolysis in human red cells (10), its are phosphorylated preferentially by immature T- effects on dGuo cytotoxicity have not been reported. From the cells to their toxic 5'-triphosphate derivatives (3-6). Thus, inhi available data it appears that 8-AGR and 8-AG are not sufficiently bition of ADA or PNP in patients with T-cell lymphoproliferative potent to inhibit the high levels of PNP in human blood (10). diseases may result in the selective destruction of T-cells. The Hence, it would be useful to develop PNP inhibitors which could potent ADA inhibitor 2'-deoxycoformycin was developed with bind tightly to the active site of the enzyme without exhibiting this rationale as a selective antileukemic agent and resulted in substrate activity with PNP. We now report the development of T-lymphoblast lysis in vivo (7); however, use of this drug was a new PNP inhibitor, 8-ABG, which is an example of a novel also associated with severe toxicity (8). Administration of an class of PNP-resistant inhibitors. inhibitor of PNP in vivo may result in less toxicity since the effects on the immune system are more selective in the PNP- compared MATERIALS AND METHODS

Received 8/15/85; revised 10/23/85; accepted 10/25/85. Synthesis of 9-Substituted Purine Analogues. Proton ' H-NMR spec The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in tra were obtained using a Brucker Wm 360 spectrometer (360 MHz) or accordance with 18 U.S.C. Section 1734 solely to indicate this fact. IBM WP-270SY NMR spectrometer (270 MHz) and are reported in &- 1This investigation was supported by Public Health Service grants CA-26284 units with tetramethylsilane (Me4Si) as the internal standard (Fig. 1). (P. E. D.) and CA-28381 (L. B. T.) awarded by the National Cancer Institute, Melting points are unconnected and were determined on a Thomas- Department of Health and Human Services, and aided by grant CH-299 (L. B. T.) from the American Cancer Society. Hoover melting point apparatus. IR spectra were recorded on a Hewlett- 2Arthritis Foundation Fellow. Packard UV8450 spectrometer, and elemental analyses were obtained 3 To whom requests for reprints should be addressed, at the Department of from M-H-W Laboratories, P. O. Box 15853, Phoenix, AZ 85018. Thin- Biological Chemistry, University of Michigan, Ann Arbor, Ml 48109. 4 The abbreviations used are: ADA, deaminase (EC 3.5.4.4); 8-ABG, layer chromatography was performed with Analtech silica gel GHLF thin- 8-amino-9-benzylguanine; 8-ABH, 8-amino-9-benzylhypoxanthine; 8-APEG, 8- layer chromatography plates (250 urn). amino-9-phenethylguanine; 8-APG, 8-amino-9-phenylguanine; 8-AG, 8-aminogua 2-Amino-4-phenylammo-5-[1-(3-methoxycarbonyl)thioureido]py- nine; 8-AGR, 8-aminoguanosine; dGuo, 2'-deoxyguanosine; HPLC, high-pressure rimidin-6-one (2a). A solution of sodium nitrite (6.5 g, 0.09 mol) in water liquid chromatography; ICsi, concentration of drug necessary to inhibit cell growth (20 ml) was added to a mixture of 2-amino-4-phenylaminopyrimidin-6- by 50%; PNP, purine nucleoside phosphorylase (EC 2.4.2.1); s, singlet; br s, broad singlet; d, doublet; t, triplet; q, quartet; m, multiplet; NMR, nuclear magnetic one (1a, 6.3 g, 0.03 mol) (14), glacial acetic acid (5 ml), and water (100 resonance; DMSO, dimethyl sulfoxide; DMF, dimethylformamide. ml) with stirring at room temperature. After 5 h, the yellow solid was

CANCER RESEARCH VOL. 46 FEBRUARY 1986 519

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 1986 American Association for Cancer Research. 8-ABG INHIBITION OF PNP

so tallized from a mixture of methanol and water (v:v, 1:1) to afford 3a (1.13 g, 90%). M.p. 230°C; white color changed to brown, >300°C; 'H-NMR (270 MHz, DMSO-de): ¿11.09(s, 1H, NH, D2O exchangeable); 9.40 (s, 1H, NH, D2O exchangeable); 7.88 (d, 2H, Ar—H); 7.26 (t, 2H, Ar—H); H,N 6.98 (t, 1H, Ar—H); 6.46 (s, 2H, NH2); and 3.71 (s, 3H, CH3).

C,3H12N603 (300.278)

Calculated: C 52.00, H 4.03, N 27.99 Found: C 52.18, H 4.18, N 28.09 8-Amino-9-phenylguanine (5a). A mixture of methyl 6-amino-4-phen- O ylamino-oxazolo[5,4-d]-2-carbamate (3a, 0.8 g, 2.66 mmol) II NHCOCH, and potassium carbonate (0.74 g, 5.36 mmol) in anhydrous methanol (30 ml) was heated at reflux in an oil bath for 5 h. The mixture was evaporated to dryness in vacuo (oil pump) at 60°C,and the residual solid HN was dissolved in water (30 ml). The resulting solution was heated at reflux for 24 h and cooled to room temperature, and the pH was adjusted to 5 with glacial acetic acid. The solid was collected by filtration and washed with water (50 ml). The crude product was dissolved in 0.1 N sodium hydroxide solution and precipitated with glacial acetic acid to give pure 5a (0.51 g, 79%). M.p. >300°C; UV Xm«nm (t x 104): MeOH:DMF, v:v, 9:1:262 (1.3), 298 (0.7); pH 1:253 (1.4), 288 (1.0); and pH 11:259 (1.3); and 'H-NMR (270 MHz, DMSO-d6): ¿10.39(s, 1H, NH, D20 exchangeable); 7.54-7.36 (m, 5H, Ar—H); 6.14 (s, 2H, NH2, D20 exchangeable); and 5.57 (s, 2H, NH2, D2O exchangeable).

C,,H10N6O (242.242)

Calculated: C 54.54, H 4.16, N 34.69 Found: C 54.59, H 4.24, N 34.56 8-Amino-9-benzylguanine (5b). A solution of methyl 9-benzylgua- nine-8-carbamate (4b, 0.3 g, 0.95 mmol) (16) and potassium hydroxide -CH2CH,- (0.145 g, 2.5 mmol) in water (15 ml) was heated at reflux for 20 h. The mixture was cooled to room temperature, and the pH was adjusted to 5 with glacial acetic acid. The solid was collected by filtration and washed with water (100 ml). The crude product was reprecipitated with 0.1 N sodium hydroxide and glacial acetic acid to afford pure 5b (0.21 g, 86%). Fig. 1. Scheme for the synthesis of 9-substituted analogues of 8-AG. The compound was identical in all respects with 8-amino-9-benzylgua- nine prepared by a previous method.5 M.p. >300°C; 'H-NMR (360 MHz, collected by filtration and washed with water (100 ml). The solid was DMSO-de): ¿5.03(s, 2H, CH2); 5.90 (s, 2H, NH2, D2O exchangeable); suspended in water (200 ml) and the suspension was heated with stirring 6.17 (s, 2H, NH2, D2O exchangeable); 7.15-7.32 (m, 5H, Ar—H); and to a boil on a hot plate. Sodium dithionite (10 g) was added in small 10.42 (s, 1H, NH, exchangeable). portions to the hot stirred mixture over a period of 1 h until the color had 2-Amino-4-phenethylaminopynmidin-6-one (1c). A mixture of 2- changed to light yellow. The hot mixture was filtered, and two equivalents amino-4-chloropyrimidin-6-one (14) (10.13 g, 70 mmol), phenethylamine of freshly prepared methoxycarbonyl isothiocyanate (15) in acetonitrile (26.0 ml, 0.21 mol), and anhydrous ethanol (200 ml) was heated at reflux (30 ml) were added to the filtrate. The mixture was heated at reflux in for 24 h. The solvent was then removed in vacuo (water pump) at 70°C, an oil bath for 6 h. The solid was collected from the hot mixture by and the residue (brown oil) was dissolved in acetone. The solid that had filtration and washed first with water (100 ml) and then methanol (10 ml). formed was collected by filtration to afford 1c (4.32 g, 26.8%). An The crude product was reprecipitated from a mixture of DMF and water analytical sample was prepared by recrystallization from a mixture of (v:v, 1:1) to afford 2a (1.4 g, 14%). M.p. 248-250°C; 'H-NMR (270 MHz, acetone and water (v:v, 1:1). M.p. 250-254°C; 'H-NMR (270 MHz, DMSO-de): ¿11.19(s, 1H, NH, D2O exchangeable); 10.35 (s, 1H, NH, DMSO-de): ¿9.66(s, 1H, NH, D2O exchangeable); 7.31-7.18 (m, 5H, D2O exchangeable); 10.22 (s, 1H, NH, D2O exchangeable); 8.02 (s, 1H, Ar—H); 6.34 (br s, 1H, NH, D2O exchangeable); 6.11 (s, 2H, NH2, D2O NH, D2O exchangeable); 7.50 (d, 2H, Ar—H); 7.24 (t, 2H, Ar—H); 6.94 exchangeable); 4.44 (s, 1H, =C—H); 3.25 (m, 2H, CH2), and 2.75 (t, 2H, (t, 1H, Ar—H); 6.44 (s, 2H, NH2, D2O exchangeable); and 3.72 (s, 3H, CH2). CH3). C12H14N4O(230.71) CiaHuNeOaS (334.358) Calculated: C 62.59, H 6.13, N 24.33 Calculated: C 46.70, H 4.22, N 25.14 Found: C 62.78, H 6.18, N 24.47 Found: C 46.46, H 4.41, N 24.90 2-Amino-4-phenethylamino-5-[1-(3-methoxycarbonyl)thioureido]- Methyl 6-Amino-4-phenylaminooxazolo[5,4-d]pyrimidine-2-carba- pyrimidin-6-one (2c). Compound 2c was prepared in 77% yield using a mate (3a). Dicyclohexylcarbodiimide (2.6 g, 12.6 mmol) was added to a procedure similar to that which afforded 2a. An analytical sample was mixture of 2-amino-4-phenylamino-5-[1 -(3-methoxycarbonyl)thioureido]- prepared by recrystallization from a mixture of DMF and water (v:v, 1:1). pyrimidin-6-one (2a, 1.4 g, 4.1 mmol) and anhydrous DMF (50 ml). The M.p. 218-219°C; 1H-NMR(270 MHz, DMSO-de): ¿11.12(s, 1H, NH, D2O mixture was allowed to stir at room temperature for 6 h, and the solvent exchangeable); 7.24 (m, 5H, Ar—H); 6.30 (s, 2H, NH2, D20 exchangea was then evaporated to dryness at 60°C in vacuo (oil pump). A solid ble); 6.27 (br s, 1H, NH, D2O exchangeable); 3.70 (s, 3H, CH3); 3.43 (t, was formed by the addition of boiling toluene to the residue. This solid was collected by filtration, washed with diethyl ether (10 ml), and recrys- 5P. E. Daddona,J-W. Chern, and L. B. Townsend, unpublishedresults.

CANCER RESEARCH VOL. 46 FEBRUARY 1986 520

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 1986 American Association for Cancer Research. 8-ABG INHIBITION OF PNP

2H, CH2); and 2.73 (t, 2H, CH2); IR (KBr): 1780 cm-1. containing blood was removed from each culture and centrifuged. The separated serum was removed and acidified with perchloric acid to 0.4 dsHisNeSOa (362.412) N, and the sample was then centrifuged at 8000 x g for 10 min at 4°C. Calculated: C 49.74, H 4.97, N 23.20 The resulting supernatant was neutralized with 10 N KOH and stored at Found: C 49.52, H 5.14, N 22.99 -20°C. All samples were analyzed by HPLC using a Waters Associates (Milford, MA) gradient high-pressure liquid Chromatograph. Samples were Methyl 6-Amino-4-phenethylaminooxazolo[5,4-d]pyrimidin-2-car- loaded onto a /^Bondapak Ci8 column (Waters Associates) and eluted at bamate (3c). Compound 3c was prepared in 90.5% yield using a 1 ml/min with a gradient of 5 to 41% HPLC-grade MeOH (Mallinckrodt, procedure similar to that which afforded 3a. An analytical sample was Inc., Paris, KY) in 0.05 M KH2PO4, pH 5.4, over a 20-min period. Eluted prepared by recrystallization from a mixture of methanol and water (v:v, 1:1). M.p. 283-284°C; 'H-NMR (270 MHz, DMSO-d6): «10.99(S, 1H, compounds were detected at 254 nm and quantitated by comparison with authentic standards. NH, D2O exchangeable); 7.30 (s, 1H, NH, D2O exchangeable); 7.28 (m, 5H, Ar—H); 6.20 (s, 2H, NH2, D2O exchangeable); 3.71 (s, 3H, CH3); 3.62 (br s, 2H, CH2); and 2.85 (t, 2H, CH2); IR (KBr): 1679 crrr1. RESULTS AND DISCUSSION C15H15N6O3(327.324) The compounds resulting from the addition of an NH2-group Calculated: C 54.70, H 5.20, N 25.52 at the C-8 position of or guanosine (20, 21), 8-AG and Found: C 54.81, H 5.06, N 25.73 8-AGR, are effective PNP inhibitors (9, 10). It has been shown 8-Amino-9-phenethylguanine (Se). Compound 5c was prepared in that 8-AG is approximately 10-fold more potent an inhibitor than 82% yield using a procedure similar to that which afforded 5a. M.p. is 8-AGR (10); however, 8-AG is also a substrate for PNP (10) >300°C; 'H-NMR (270 MHz, DMSO-cfe): 510.39 (s, 1H, NH, D2O ex which may limit its inhibitory activity. This suggested that it would changeable); 7.31 -7.18 (m, 5H, Ar—H); 6.16 (s, 2H, NH2, D2O exchange be desirable to develop analogues of 8-AG that would not be able); 5.83 (s, 2H, NH2, D2O exchangeable); 3.98 (t, 2H, CH2); and 2.90 (t, 2H, CH2); IR (KBr): 1680 cm-1; UV X™nm (e x 104): MeOH:DMF, v:v, substrates for PNP but would retain their ability to inhibit PNP. We have now synthesized various 9-substituted 8-AG analogues 8:2: 262 (1.64), 291 (1.07); and pH 11: 260 (1.54). which should not be susceptible to phosphorolysis by PNP, since C13H,6N6O (272.312) they lack a glycosidic bond. As illustrated in Fig. 2, we have Calculated: C 57.72, H 5.22, N 31.09 synthesized 8-APG as well as analogues in which the distance Found: C 57.69, H 5.36, N 30.89 between the purine base and the benzene ring was varied by the addition of a méthylèneorethylene group. In addition 8-ABH5 Determination of Kinetic Constants for Human PNP. PNP activity was assayed in dialyzed T-lymphoblast (MOLT-4) cell extract or human erythrocyte lysate using a radiochemical assay described previously (9, 17). Substrate, [8-14C]inosine or [8-14C]guanosine, was used at a specific activity of 22.5 mCi/mmol. For Km determinations, a fixed amount of diluted cell extract was incubated with variable amounts of radiolabeled substrate (12.5 to 100 UM) and inorganic phosphate (50 mw). K, deter minations were performed with variable radiolabeled inosine concentra tions (12.5 to 100 MM),fixed inorganic phosphate (50 ITIM),and variable inhibitor concentrations (0.03 MMto 1.0 mw). All reactions were incubated for 10 min at 37°C.Substrate and product (inosine and or guanosine and guanine, respectively) were separated by high-voltage Compound RÌ Bi paper electrophoresis. The radiolabeled product of the reaction was visualized by UV light (290 nm), cut from the paper, and counted in a 8-AG H NH, toluene-based scintillation fluid in a Packard Tri-carb liquid scintillation spectrometer. Enzyme-free blank reactions were used as controls for all reactions. In all initial velocity determinations with or without inhibitor, not more than 15% of the substrate was converted to product. Double 8-AGR R ibose NH, reciprocal plots of the initial velocity values versus the substrate concen trations were linear. Kinetic data were fit to a simple hyperbola by a modification of Cleland's program (18) using the reciprocal variance of the velocity as a weighting factor on a Hewlett-Packard 9825A calculator. 8-APG NH, Standard errors of the slopes and intercepts of each Lineweaver-Burk plot were less than 10%. Secondary replots were used to determine Km and K¡values using least squares regression analysis for linear replots. Drug Cytotoxicity and Metabolism. MOLT-4 T-lymphoblasts were 8-ABG grown at 37°C in RPMI 1640 medium supplemented with 10% horse NH2 serum (Grand Island Biological Co., Grand Island, NY). The effects of the various drugs on cell growth were determined as described previously (19) during a 72-h incubation period. Cell number was determined with 8-ABH the aid of a Model ZF Coulter electronic particle counter (Coulter Elec -CH2- H tronics, Inc., Hialeah, FL). Drug metabolism in human blood was determined as follows. Freshly isolated heparinized blood from a normal donor was diluted with an equal volume of buffer (50 mw KH2PO4:75 mw NaCI:10 mw glucose, pH 7.4); 8-APEG (( )>-CH2CH2- NH2 dGuo, 8-AG, 8-AGR, or 8-ABG was then added to a final concentration of 50 /¿M.At0 and 4 h following drug addition, 1 ml of control or drug- Fig. 2. Structures of PNP inhibitors.

CANCER RESEARCH VOL. 46 FEBRUARY 1986 521

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 1986 American Association for Cancer Research. 8-ABG INHIBITION OF PNP was prepared for comparison with 8-ABG. The 8-AG analogues were studied for inhibitory activity with human PNP. The kinetic constants for the various substrates and inhibitors of PNP are listed in Table 1. Inosine and guanosine exhibited similar Km values of approximately 40 MMin this assay system. With inosine as the substrate for PNP, all purine ana logues tested showed competitive inhibition. 8-AGR exhibited a Kj value of 7 MM, while 8-AG was approximately 10-fold more potent (K, = 0.8 MM) as a PNP inhibitor. 8-APG showed poor inhibitory activity, with a K

CANCER RESEARCH VOL. 46 FEBRUARY 1986 522

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 1986 American Association for Cancer Research. .','••'".**?•

8-ABG INHIBITION OF PNP

ABG compared to 8-AG for the inhibition of inosine phospho- Deoxyguanosine triphosphate as a possible toxic metabolite in the immuno deficiency associated with purine nucleoside phosphorylase deficiency. J. Clin rolysis. While 8-ABG is the most potent PNP inhibitor of all the Invest., 67:1405-1409,1978. nucleosides and bases reported to date, it is still necessary to 6. Mitchell, B. S., Mejias, E., Daddona, P. E., and Kelley, W. N. Purinogenic immunodeficiency diseases: selective toxicity of for T- develop a more tight binding compound for in vivo use in order cells. Proc. Nati. Acad. Sci. USA, 75: 5011-5014,1978. to inhibit the overwhelming amount of PNP activity in human 7. Mitchell, B. S., Koller, C. A., and Heyn, R. Inhibition of erythrocytes (10). We are presently investigating whether the activity results in cytotoxicity to T-lymphoblasts in vivo. Blood, 56: 556-559 inhibitory activity of 8-ABG with PNP may be increased by steric, 1980. 8. Koller, C. A., and Mitchell, B. S. Alterations in erythrocyte electronic, or hydrophobic modifications on the benzyl group, pools resulting from 2'-deoxycoformycin therapy. Cancer Res., 43: 1409- according to the method of Topliss (23). 1414, 1983. 9. Kazmers, I. S., Mitchell, B. S., Daddona, P. E., Wotring, L. L., Townsend, L. Developing a potent PNP inhibitor to produce selective lymph- B., and Kelley, W. N. Inhibition of purine nucleoside phosphorylase by 8- oid toxicity in vivo is an attractive approach to chemotherapy of aminoguanosine: selective toxicity for T-lymphoblasts. Science (Wash. DC), 274:1137-1139,1981. lymphoid malignancies. It is expected that, in comparison to the 10. Stoeckler, J. D., Cambor, C., Kuhns, V., Chu, S-H„and Parks, R. E., Jr. results of in vivo inhibition of ADA, the inhibition of PNP will be Inhibitors of purine nucleoside phosphorylase. C(8) and C(5') substitutions. more specifically toxic to immature T-lymphoblasts, based on Biochem. Pharmacol., 37:163-171,1982. the clinical manifestation of the PNP- and ADA-related immuno 11. Willis, R. C., Robins, R. K., and Seegmiller, J. E. An in vivo and in vitro evaluation of 1-/3-o-ribofuranosyl-1,2,4-triazole-3-carboxamidine: an inhibitor deficiency diseases. In addition, the PNP substrate dGuo is of human lymphoblast purine nucleoside phosphorylase. Mol. Pharmacol., 78: apparently phosphorylated to dGMP, the precursor of the cyto- 287-295,1980. 12. Stoeckler, J. D. Purine nucleoside pnosphorylase: a target for chemotherapy. toxic metabolite dGTP, via only one nucleoside kinase (presum In: R.I. Glazer (ed.), Developments in Cancer Chemotherapy, pp. 35-60. Boca ably 2'- kinase) (24, 25), while the ADA substrate Raton, FL: CRC Press, Inc., 1984. 2'-deoxyadenosine can be phosphorylated by either adenosine 13. Tuttle, J. V., and Krenitsky, T. A. Effects of acyclovir and its metabolites on kinase or 2'- (24, 26). The relative abun purine nucleoside phosphorylase. J. Biol. Chem., 259: 4065-4069, 1984. 14. Noell, C. W., and Robins, R. K. Potential purine antagonists XXXI. The dance of adenosine kinase compared to 2'-deoxycytidine kinase preparation of certain 9-alkyl-2-amino-6-purinethiols and related derivatives as antitumor agents. J. Med. Chem., 5: 558-588,1962. in human tissues suggests that the toxic effects of dGuo may be more limited than those of 2'-deoxyadenosine (27). Despite 15. Lamon, R. W. A new synthesis of 4-thiouracils. J. Heterocycl. Chem., 5: 837- 844,1968. intensive efforts in several laboratories to develop a tight-binding 16. Chem, J. W., Wise, D. S., and Townsend, L. B. A convenient synthesis of 2- W-methoxy-carbonylaminooxazolo[5,4-d]. J. Heterocycl. Chem., PNP inhibitor, this goal has not yet been achieved. Certainly the 27:1245-1246,1984. recent elucidation6 of the three-dimensional structure of PNP 17. Fox, I. H., Andres, C. M., Gelfand, E. W., and Biggar, D. Purine nucleoside phosphorylase deficiency: altered kinetic properties of a mutant enzyme. should lead to the rapid development of more potent inhibitors Science (Wash. DC), 797: 1084-1086,1977. of this enzyme. 18. Cleland, W. W. The statistical analysis of enzyme kinetic data. Adv. Enzymol, 29: 1-32, 1967. 19. Shewach, D. S., Daddona, P. E., Ashcraft, E., and Mitchell, B. S. Metabolism ACKNOWLEDGMENTS and selective cytotoxicity of 9-/3-o-arabinofuranosylguanine in human lympho- blasts. Cancer Res., 45:1008-1014, 1985. We are grateful to Sue Grech for her excellent typing of this manuscript. We 20. Cavalieri, R. F., and Bendich, A. The ultraviolet absorption spectra of pyrimi- thank Dr. P. Arjunan for the preparation of 8-amino-9-benzyladenine. dines and . J. Am. Chem. Soc., 72: 2587-2594, 1950. 21. Long, R. A., Robins, R. K., and Townsend, L. B. Purine nucleosides. XV. The synthesis of 8-amino and 8-substituted aminopurine nucleosides. J. Org. Chem., 32: 2751 -2756,1967. REFERENCES 22. Osbome, W. R. A. Human red cell purine nucleoside phosphorylase: purification by biospecific affinity chromatography and physical properties. J. Biol. Chem., 1. Giblett, E. Ft., Andersen, J. E., Cohen, F., Pollara, B., and Meuwissen, H. J. 255:7089-7092,1980. Adenosine deaminase deficiency in two patients with severely impaired cellular 23. Topliss, J. G. Utilization of operational schemes for analog synthesis in drug immunity. Lancet, ii: 1067-1069,1972. design. J. Med. Chem., 75: 1006-1011,1972. 2. Giblett, E. R., Ammann, A. J., Wara, D. W., Sandman, R., and Diamond, L. K. 24. Hershfield, M. S., Fetter, J. E., Small, W. C., Bagnara, A. S., Williams, S. R., Nucleoside-phosphorylase deficiency in a child with severely defective T-cell Ullman, B., Martin, D. W., Jr., Wasson, D. B., and Carson, D. A. Effects of immunity and normal B-cell immunity. Lancet, /: 1010-1013,1975. mutational loss of adenosine kinase and deoxycytidine kinase on deoxyATP 3. Coleman, M. S., Donofrio, J., Hutton, J. J., Hahn, L., Daoud, A., Lampkin, B., accumulation and deoxyadenosine toxicity in cultured CEM human T-lympho- and Dyminski, J. Identification and quantitation of adenine deoxynucleotides blastoid cells. J. Biol. Chem., 257: 6380-6386, 1982. in erythrocytes of a patient with adenosine deaminase deficiency and severe 25. Hurley, M. C., Patella, T. D., and Fox, I. H. Human placental deoxyadenosine combined immunodeficiency. J. Biol. Chem., 253:1619-1626,1978. and deoxyguanosine phosphorylating activity. J. Biol. Chem., 258: 15021- 4. Gudas, L. J., Ullman, B., Cohen, A., and Martin, D. W., Jr. Deoxyguanosine 15027, 1983. toxicity in a mouse T-lymphoma: relationship to purine nucleoside phosphoryl- 26. Ullman, B., Levinson, B. B., Hershfield, M. S., and Martin, D. W., Jr. A ase-associated immune dysfunction. Cell, 14: 531-538,1978. biochemical genetic study of the role of specific nucleoside kinases in deoxy 5. Cohen, A., Gudas, L. J., Ammann, A. J., Stool, G. E. J., and Martin, D. W., Jr. adenosine phosphorylation by cultured human cells. J. Biol. Chem., 256: 848- 852,1981. 6 C. E. Bugg, presented in a seminar at the University of Michigan College of 27. Carson, D. A., Kaye, J., and Wasson, D. B. Differences in deoxyadenosine Pharmacy, January 31,1985. metabolism in human and mouse lymphocytes. J. Immunol., 724:8-12,1980.

CANCER RESEARCH VOL 46 FEBRUARY 1986

523

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 1986 American Association for Cancer Research. Potentiation of 2′-Deoxyguanosine Cytotoxicity by a Novel Inhibitor of Purine Nucleoside Phosphorylase, 8-Amino-9-benzylguanine

Donna S. Shewach, Ji-Wang Chern, Katherine E. Pillote, et al.

Cancer Res 1986;46:519-523.

Updated version Access the most recent version of this article at: http://cancerres.aacrjournals.org/content/46/2/519

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/46/2/519. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 1986 American Association for Cancer Research.