CA2444471A1 - Synthesis of pancratistatin prodrugs - Google Patents

Synthesis of pancratistatin prodrugs Download PDF

Info

Publication number
CA2444471A1
CA2444471A1 CA002444471A CA2444471A CA2444471A1 CA 2444471 A1 CA2444471 A1 CA 2444471A1 CA 002444471 A CA002444471 A CA 002444471A CA 2444471 A CA2444471 A CA 2444471A CA 2444471 A1 CA2444471 A1 CA 2444471A1
Authority
CA
Canada
Prior art keywords
pancratistatin
phosphate
sodium methoxide
prodrug
dibenzyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002444471A
Other languages
French (fr)
Inventor
George R. Petit
Brian Orr
Sylvie Ducki
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Arizona Board of Regents of ASU
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2444471A1 publication Critical patent/CA2444471A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A new and efficient synthesis of the (+)-pancratistatin phosphate prodrug 2a has been accomplished. Selective protection (tetraacetate 4) of (+)-pancratistatin (1a) was followed by phosphorylation (to 5) with dibenzyl chlorophosphite (prepared in situ from dibenzyl phosphite). Cleavage of the acetate (with sodium methoxide) and benzyl (by hydrogenolysis) protecting groups followed by concomitant reaction with two equivalents of sodium methoxide afforded good yield of disodium (+)-pancratistatin phosphate (2a).
Further increases in yields of the prodrug (2a) were realized by avoiding heat in the final purification steps. Fourteen (2b-o) additional metal and ammonium derived phosphate prodrugs were also synthesized.

Description

Synthesis of Pancratistatin Prodrugs This application claims the benefit of United States provisional application filed on April 19, 2001.
Introduction This invention relates generally to the discovery of a new and efficient synthesis of the (+)-pancratistatin phosphate prodrug and fourteen (14) metal and ammonium cation derivatives thereof. The pancratistatin phosphate prodrug denominated 2a in the enclosed Scheme was found to have exceptional solubility in water and therefore readily administerable in the to treatment of human cancer.
This research was funded in part by Outstanding Investigator Grant CA
44344-O1-11 awarded by the National Cancer Institute, DHHS. The United States Government may have certain rights to this invention.
Background of the Invention The anticancer phenanthridone (+)-pancratistatin (la) was isolated from the bulbs of the Hawaiian Hymehocallis (formally Pa~rcratium) littoralis (Pettit et al., 1984a, Jout°faal of tlae Chemical Society, Chemical Communications, 1693; Pettit et al., 1984b, Jour°~zal ofNatural Ps°oducts, 47, 1018). The structure was determined by NMR spectroscopy and confirmed by 2o x-ray crystallographic analysis of its 7-methoxy derivative (1b).
Subsequently, pancratistatin (la) was found to exhibit strong ifi vitro cancer cell growth inhibitory activities including against the U.S. National Cancer Institute (NCI) panel of cancer cell lines (Pettit et al., 1986, Jou~~fial ofNatural Products, 49, 995; 1993 JoumZal of Natus~al P~°oducts, 56, 1682) and a series of i~c vivo experimental cancer systems. A related area of promise involves its activity against a range of RNA viruses (Gabrielsen et al., 1992, Journal of Natuf~al P~°oducts, 55, 1569). Due to the relatively low natural abundance (0.039% of dry bulb) and its increasing potential as a clinically useful antitumor agent, pancratistatin has become an important target for total 3o synthesis. The first total synthesis of (~)-pancratistatin was reported in (Danishefsky and Yon Lee, 1989, Jout~nal of the AmericasZ Chemical Society, 11I, 4829). Six stereospecific syntheses of natural (+)-pancratistatin (la) were subsequently completed (Tian et al., 1995, Jout°nal of tlae Atne~icatt Chemical Society, 117, 3643; Trost and Pulley, 1995, Jout°tZal of the Ante>ricata Chemical ,Society, 117, 10143; Hudlicky et al., 1996, Jotcrhal of the Atner~ica~a Chemical Society, 118, 10752; Doyle et al., 1997, Tett°ahedt°o>z., 53, 11153;
Magnus and Sebhat, 1998, Tett~ahedt°on, 54, 15509; Pettit et al., 2000, Jout~nal of OigatZic Chemistry, in preparation). Meanwhile, a number of partial syntheses have been described (Lopes et al., 1992, Tet>~ahedt~o>z Letters, 33, 6775; Angle and Louie, 1993, Tett~ahedf°on Letters, 34, 4751; Grubb et al., 1999, Tett°ahedt°on Letters, 40, 2691).
The clinical development of pancratistatin (la) was earlier hampered due to its poor solubility in water (<53 ~.ghnl). This problem was addressed (Pettit et al., 1995a, Artti-Cat~cer~ Dt~tsg Design, 10, 243) by developing a synthesis of the phosphate prodrug (2a) which displayed greatly improved solubility characteristics in water and was found to exhibit equal activity against the murine P388 lymphocytic leukemia cell. Presumably, attachment of a phosphate is also beneficial for in vivo systems since human non-specific phosphatases will hydrolyze the phosphate prodrug following administration 2o to release (+)-pancratistatin (la). Since the relatively unstable phosphorylating agent, dibenzyl-(N,N diisopropylamido)-phosphine, we employed in our initial synthesis of phosphate (2a), was less than desirable and yields for the phosphorylation reaction itself proved variable (0-91 yields of (5) with no recovery of valuable starting material), we thus investigated less astringent approaches. Herein, an alternate simpler synthesis of pancratistatin prodrug (2a) will be described. The new synthesis will be used to provide the pure drug (2a) in quantity for preclinical development. In addition, a series of phosphate metal and ammonium cation salt derivatives (2b-o) was prepared in order to evaluate effects on human cancer cell growth 30 and solubility properties (in water).

These and still further objects as shall hereinafter appear are readily fulfilled by the present invention in a remarkably unexpected manner as will be readily discerned from the following detailed description of an exemplary embodiment thereof.
Brief Description Of Drawings Fig. 1 is a schematic showing of the step-by-step synthesis of the presentinvention.
Detailed Descriution Of Preferred Embodiment (+)-Pancratistatin (la) was isolated from Hymenocallis littof°alas as l0 previously described (Pettit et al., 1995b, Jouin2al ofNatural Products, 58, 37). Acetic anhydride, pyridine, dibenzyl phosphite, N ethyldiisopropylamine and 10% palladium over carbon catalyst were purchased from Lancaster Synthesis, Inc. Carbon tetrachloride, 4-dimethylaminopyridine, anhydrous magnesium sulfate (MgS04), potassium dihydrogenphosphate, phosphomolybdic acid, and zinc acetate dihydrate were from Sigma-Aldrich Chemical Co. Sodium methoxide, lithium hydroxide monohydrate, piperazine (anhydrous) and morpholine were from Acros Organics. Calcium acetate and manganese acetate were purchased from Fisher Scientific Co. and magnesium acetate quinine, quinidine and imidazole from J. T. Baker Chem. Rubidium 20 carbonate and cesium carbonate were supplied by Alfa Products, Inc. and potassium acetate from Mallinckrodt. All solvents were redistilled prior to use and dried when necessary. Solvent extracts of aqueous solutions were dried over magnesium sulfate. The 1.0 M solutions of the metal bases were prepared in distilled water and the 1.0 M solutions of the amines in dry methanol.
All reactions were carried out under an atmosphere of nitrogen unless otherwise specified and their progress was ascertained by thin-layer chromatography using Analtech silica gel GHLF Uniplates (visualized under long- and short-waves UV) and developed in an ethanolic solution of phosphomolybdic acid reagent. Column chromatography was performed with silica gel 60 (230-400 mesh) from E. Merck. The Sephadex~ G-10 was washed (prior to use) with 1 N sodium hydroxide, water, 1 N acetic acid and finally water until neutral pH.
All melting points were determined with an Electrotherinal digital melting point apparatus model IA9200 and are uncorrected. Optical rotation values were recorded using a Perkin Elmer 241 polarimeter. The IR spectra were from a Nicolet FTIR model MX-1 instrument. EIMS spectra were obtained with a MAT 312 mass spectrometer; high- and low-resolution FAB
l0 spectra were obtained with a Kratos MS-50 mass spectrometer (Midwest Center for Mass Spectrometry, University of Nebraska, Lincoln, NE) using a glycerol-triglycerol matrix. The ~H-, 13C- and 31P-NMR spectra were recorded with Varian Gemini 300, 400 or 500 MHz instruments. Elemental analyses were determined by Galbraith Laboratories, Inc. (Knoxville, TN).
1,2,3,4-O-Tetf°aacetoxy pane°atistatin (4) - The following procedure represents a useful improvement over our earlier method (Pettit et al., 1995x, A~zti-Ca~zcei-Dr~~g Designs, 10, 243). To a stirring solution of (+)-pancratistatin (la, 82% pure, 8.6 g, 21.7 mmol) in pyridine (50 ml) were added acetic anhydride (45 ml, 0.48 mol) and 4-dimethylaminopyridine (200 2o mg, 1.6 mmol) at room temperature. After stirring for 16 hours, ice (100 ml) was added to the mixture and stirring was continued for a further 1 hour. The resultant mixture was extracted with dichloromethane (2 x 100 ml) and the combined organic extracts were dried, filtered and evaporated in' vacuo.

Pyridine (80 ml) and water (40 ml) were added to the brown oily residue (14.3 g) and the mixture was heated under reflux for 1 hour. Ice (50 rnl) was added and the cooled mixture was extracted with dichloromethane (2 x 100 ml). The combined organic extract was dried, filtered and solvent evaporated ira vacuo.
The resulting residue was purified by column chromatography on silica gel eluting with 1.5% methanol in dichloromethane, affording a white amorphous powder, which was recrystallized from ethanol to give the title compound (4, 7.1 g, 66%) as colorless needles; m.p. 240°C [m.p. lit. (Pettit et al., 1995a, Ahti-Cancef° Dy~ug Design, 10, 243) 243-246°C]; [a]D2~
+31.5° (c 1.04, DCM) { [a]DSS lit. (Pettit et al., 1995a, Anti-Ca~ace~ Ds°ug Desigfz, 10, 243) +30.4° (c 0.72, DCM)~. The resulting IR, 1H-NMR and 13C-NMR spectra were as previously reported.
1,~,3,4-O-Tetf'aacetoxy-7-O-dibenzyloxyplaosphoryl pafacf°atistati~r (S) - To a solution of 1,2,3,4-O-tetraacetoxy-pancratistatin (4, 2.60 g, 5.27 mmol) in acetonitrile (30 ml), cooled to -30°C (ethylene glycol/dry ice), were added carbon tetrachloride (2.6 ml, 26.9 rmnol), N ethyldiisopropylamine (2.0 ml, 11.6 mmol) and 4-dimethylaminopyridine (71 mg, 0.58 xmnol). Following the slow dropwise addition of dibenzyl phosphite (1.80 ml, 8.15 mmol), the mixture was stirred for 3 hours. The reaction was terminated by addition of an 2o aqueous solution of potassium dihydrogenphosphate (0.5 M, 50 ml) and stirred at room temperature for 30 minutes. The resultant mixture was extracted with dichloromethane (2 x 50 ml) and the combined organic extract was dried, filtered and solvent evaporated (iia vacuo). The resulting yellow oil was purified, at low temperature (4°C), by column chromatography on silica gel and eluting with 0.8% methanol in dichloromethane. That procedure afforded recovered starting material, 1,2,3,4-O-tetraacetoxy-pancratistatin (4, 0.30 g, 11 %), closely followed by its dibenzyl phosphate derivative (S, 3.44 g, 87%) as a colorless crystalline solid; m.p. 119°C [m.p. lit. (Pettit et al., 1995x, Af~ti-Caracas°D~ugDesign, 10, 243) 119-121°C]; [a]D27 +
59.3° (c 1.33, DCM) f [a]D33 lit. (Pettit et al., 1995a, Anti-Cancer Ds°ug Desigfa, 10, 243) +69.1 ° (c 0.89, DCM)}; vnaX (KBr disc) 2910w, 1755s, 1674s, 1485s, 1373s, 1221s, l0 1037b, 871w, 738w, 493w cW 1; 8H/ppm (300 MHz, CDCl3) 2.04 (3H, s, OAc), 2.06 (3H, s, OAc), 2.07 (3H, s, OAc), 2.16 (3H, s, OAc), 3.40 (1H, dd, J3, 13 Hz, H-IOb), 4.26 (1H, dd, J l I, I3 Hz, H-4a), 5.13 (1H, dd, J3, I1 Hz, H-4), 5.20 ( 1 H, t, J 3 Hz, H-2), 5.23 ( 1 H, dd, J 7, 12 Hz, CH,~HBPh), 5.26 ( 1 H, dd, J 7, 12 Hz, CHAHBPh), 5.31 ( 1 H, dd, J 7, 12 Hz, CH~HDPh), 5.41 ( 1 H, dd, J 7, I 2 Hz, CHCHDPh), 5.43 ( 1 H, t, J 3 Hz, H-3 ), 5.54 ( 1 H, t, J 3 Hz, H-1 ), 5.92 (1H, d, J 1 Hz, OCH~HBO), 5.95 (1H, d, J 1 Hz, OCHAHBO), 6.10 (1H, bs, 5-NH), 6.43 (1H, s, H-10), 7.30-7.42 (10H, bm, 2 x Ph); 8o/ppm (125 MHz, CDC13) 170.1 (C=O at C-1), 169.7 (C=O at C-3), 169.0 (C=O at C-4), 168.2 (C=O at C-2), 162.6 (C-6), 152.4 (C-9), 139.3 (d, Jp~ 4 Hz, C-8), 136.0 20 (d, JPs 8 Hz, C of Ph), 135.9 (d, JPs 8 Hz, C of Ph), I34.1 (d, JPs 7 Hz, C-7)~, 133.0 (C-l0a), 128.42 (CH of Ph), 128.37 (CH of Ph), 128.3 (CH of Ph), 127.9 (CH of Ph), 127.7 (CH of Ph), 117.0 (C-6a), 102.7 (OCHZO), 101.5 (C-10), 71.6 (C-4), 70.1 (d, JP~ 5 Hz, CHaPh), 70.0 (d, JPs 6 Hz, CH2Ph), 67.6 (C-2), 66.7 (C-3), 66.4 (C-1), 47.6 (C-4a), 40.3 (C-lOb), 20.76 (CH3 at C-2), 20.73 (CH3 at C-3), 20.67 (CH3 at C-4), 20.5 (CH3 at C-1); ~p/ppm (200 MHz, CDC13, referenced to 85% H3P04) -6.61 (s, 1H decoupled); m/z (EI) 493 [M-P(O)(OBn)Z, 10%~, 271 (35), 91 (40), 61 (40), 44 (100).
Disodium 7-O plaosphosyl panes°atistatin (2a) - To a solution of dibenzyl pancratistatin phosphate (5a, 2.00 g, 2.66 mmol) in methanol (50 ml) was added sodium methoxide (87 mg, 1.61 mol). The mixture was stirred at room temperature for 2 hours and then water (50 ml) was added. The aqueous 1o mixture was extracted with dichloromethane (5 x 50 ml) and the combined organic extract was dried, filtered and evaporated i~z vacuo (NO HEAT) to give the crude deacetylated derivative as a white powder (1.8 g). The residue was promptly dissolved in ethanol (50 ml) and 10% Pd/C catalyst (201 mg) was added. The mixture was stirred under 1 atm. of hydrogen for 2 hours and filtered through fluted filter paper. The catalyst was washed thoroughly with methanol (50 ml) and the filtrate was evaporated ifa vacuo (NO HEAT) to give the crude debenzylated phosphoric acid (6) as a white powder (1.3 g); m.p.
195°C dec.; 8H/ppm (300 MHz, D20) 6.70 (1H, bs, H-10), 5.96 (1H, s, OCH~HBO), 5.93 (1H, s, OCHAHBO), 4.29 (1H, bs, H-1), 4.03 (1H, bs, H-2), 2o 3.87 (1H, bs, H-3), 3.74 (1H, m, H-4), 3.67 (1H, t, J 11 Hz, H-4a), 3.00 (1H, d, J 12 Hz, H-10b); m/z (FAB) 406 [(M + H)+, 20%], 405 [M~, 25], 154 (100);
HRFAB 405.0467 calculated for C14Hi6NOuP 405.0461. ' The phosphoric acid (6) was immediately redissolved in methanol (50 ml) and sodium methoxide (361 mg, 6.73 mmol) was added. The mixture was stirred overnight and then concentrated isa vacuo (NO HEAT) to give a white paste (1.6 g). The residue was purified on a Sephadex~ G-10 column eluting with water. The fluorescent fractions were combined and freeze dried to give di-sodium pancratistatin prodrug (2a) as a fluffy powder (1.l g, 92%); m.p.
206°C (dec); [a]D28 +78.4° (c 1.02, HZO); v",~X (KBr disc) 3500-3000b, 2360w, 1670bs, 1480s, 1350w, 1090s, 980m, 720m crri l; 8H/ppm (400 MHz, DZO) 6.59 (1H, s, H-10), 5.96 (1H, s, OCHAHBO), 5.87 (1H, s, OCHAHBO), 4.39 to (1H, s, H-1), 4.11 (1H, bs, H-2), 3.93 (1H, bs, H-3), 3.81 (1H, bd, J9 Hz, H-4), 3.73 (1H, bt, J 12 Hz, H-4a), 3.00 (1H, bd, J 12 Hz, H-lOb); 8C/ppm (100 MHz, D20) 171.2 (C=O), 152.3 (C-9), 139.1 (C-8), 137.3 (C-7), 135.3 (C-l0a), 117.6 (C-6a), 102.4 (O-CHI-O), 101.0 (C-10), 72.7 (C-3), 70.7 (C-2), 70.3 (C-4), 68.8 (C-1), 49.1 (C-4a), 40.7 (C-lOb); 8p/ppm (162 MHz, D20, referenced to 85% H3P04) 0.90 (s, 1H decoupled); m/z (FAB) 449 (M+, 20%), 427 [(M-Na)+, 20), 154 (100).
Gehe~°al procedure for syratlzesis of tlae paf2cf°atistatin p~odr~ugs (2b-o): To an aqueous methanol solution of the phosphoric acid derivative of pancratistatin (6, 50 mg in 1 ml water or methanol) was added a 1.0 M solution (250 ~.1) of 2o the appropriate metal (carbonate or acetate) salt or amine free base. The solution became cloudy and the mixture was stirred for 6 hours. The mixture was freeze dried (or evaporated) to afford the desired prodrug salt (2b-o).
The salts were further purified by trituration with wet methanol and/or diethyl-ether to remove un-reacted starting materials.
Dilithiutn panct°atistatin 7-O phosphate (2b): Yellowish powder (61 mg); m.p.
240 °C dec.: 8H/ppm (300 MHz, Da0), 6.62 (1H, bs, H-10), 600 (1H, s, OCHAHBO), 5.90 (1H, s, OCHAHBO), 4.44 (1H, bs, H-1), 4.15 (IH, bs, H-2), 3.97 (1H, bs, H-3), 3.80 (2H, m, H-4, H-4a), 3.07 (1H, d, J 12 Hz, H-IOb).
Dipotassiuw panct°atistatin 7 O phosphate (2c): Colorless powder (66 mg);
m.p. 280°C dec.; 8H/ppm (300 MHz, D20) 6.68 (IH, bs, H-10), 6.00 (1H, s, OCHAHBO), 5.95 (1H, s, OCHAHBO), 4.45 (1H, bs, H-1), 4.16 (1H, bs, H-2), 3.97 (1H, bs, H-3), 3.89 (1H, m, H-4), 3.78 (1H, m, H-4a), 3.13 (1H, d, J 12 Hz, H-lOb).
Dii°ubidiunt pancs~atistatifa 7-O phosphate (2d): Colorless powder (0.1 I g);
m.p. 200°C (dec.); BHlppm (300 MHz, D20) 6.63 (IH, bs, H-10), 6.00 (1H, s, OCHAHBO), 5.90 (1H, s, OCHAHBO), 4.45 (1H, bs, H-1), 4.16 (1H, bs, H-2), 3.98 (1H, bs, H-3), 3.80 (2H, m, H-4, H-4a), 3.10 (1H, d, J 12 Hz, H-lOb);
ntlz (FAB) 573.9 [(M + H)+, 5%], 298.9 (35), 215.1 (25), 135.1 (100); HRFAB
573.8598 calculated for C14H1sNO11PRba 573.8619.
Dicesiutn panct°atistatin 7-O phosphate (2e): Yellow oil (0.22 g);
BHlppm (f00 MHz, D20) 6.63 (1H, bs, H-10), 6.01 (1H, s, OCH~HBO), 5.90 (1H, s, 2o OCHAHgO), 4.46 (1H, bs, H-1), 4.17 (1H, bs, H-2), 3.98 (1H, bs, H-3), 3.85 ( I H, m, H-4), 3 .79 ( 1 H, m, H-4a), 3 .08 ( 1 H, d, J 12 Hz, H-1 Ob); ntlz (FAB) 669.9 [(M + H)+, 50%], 225.0 (100); HRFAB 669.8468 calculated for C14H1sNOlIPCsa 669.8491.
Magnesium panc~atistatin 7 O phosphate (Z~: Colorless powder (64 mg);
m.p. 220°C dec.; ~H/ppm (300 MHz, D20) 6.66 (1H, bs, H-10), 6.01 (1H, s, OCHAHBO), 5.94 (1H, s, OCHAHBO), 4.44 (1H, bs, H-1), 4.16 (1H, bs, H-2), 3.98 (1H, bs, H-3), 3.80 (2H, m, H-4, H-4a), 3.07 (1H, d, J 12 Hz,~H-lOb).
Calcium paszcnatistatin-7-O phosphate (2g): Colorless powder (77 mg); m.p.
230 °C (dec.); 8H/ppm (300 MHz, D20) 6.64 (1H, bs, H-10), 5.99 (1H, s, OCHAHBO), 5.96 (1H, s, OCHAHaO), 4.41 (1H, bs, H-1), 4.14 (IH, bs, H-2), l0 3.97 (1H, bs, H-3), 3.76 (2H, m, H-4, H-4a), 3.41 (1H, d, J 11 Hz, H-lOb).
Zinc paracr°atistatin 7-O phosphate (21i): Colorless crystalline powder (76 mg);
m.p. I90°C (dec.); ~H/ppm (300 MHz, D20) 6.69 (1H, bs, H-10), 5.96 (2H, bs, OCHzO), 4.44 (1H, bs, H-1), 4.15 (1H, bs, H-2), 3.98 (1H, bs, H-3), 3.80 (2H, m, H-4, H-4a), 3.12 (1H, d, J 12 Hz, H-lOb).
Manganese pan.cratistatin 7-O phosphate (2i): Colorless powder (88 mg);
m.p. 240°C (dec.); 8H/ppm (300 MHz, D20) 6.65 (1H, bs, H-10), 6.00 (1H, s, OCHAHBO), 5.95 (1H, s, OCHAHBO), 4.44 (1H, bs, H-1), 4.15 (1H, bs, H-2), 3.99 (1H, bs, H-3), 3.79 (2H, m, H-4, H-4a), 3.08 (1H, d, J 12 Hz, H-lOb).
Pipes°azine pancratistatin 7-O phosphate (2j): Colorless powder (78 mg); m.p.
200°C (dec.); 8H/ppm (300 MHz, D20) 6.62 (IH, bs, H-10), 5.99 (1H, s, OCHAHBO), 5.88 (1H, s, OCHAHBO), 4.44 (1H, bs, H-1), 4.15 (1H, bs, H-2), 3.96 (1H, bs, H-3), 3.84 (1H, m, H-4), 3.77 (1H, m, H-4a), 3.10 (1H, d, J 12 Hz, H-l Ob), 2.99-3.01 (8H, m, 4 x CH2 piperazine); m/z (FAB) 492.2 [(M +
H)+, 30%], 460.1 (85), 154.1 (100); HRFAB~492.1373 calculated for CiaHz7NsOuP 492.1383.
Mofpholine pancratistatin 7-O phosphate (21t): Colorless powder (71 mg);
m.p. 180°C (dec.); 8H/ppm (300 MHz, DZO) 6.63 (1H, bs, H-10), 5.99 (1H, s, OCHAHBO), 5.89 (1H, s, OCHAHBO), 4.44 (1H, bs, H-1), 4.15 (1H, bs, H-2), 3.96 (1H, bs, H-3), 3.78-3.82 (6H, m, H-4, H-4a, 2 x CHzN morpholine), 3.10-3.14 (5H, m, H-l Ob, 2 x CH20 moipholine); m/z (FAB) 493.2 [(M + H)+, 15%], 406.1 (30), 154.1 (00); HRFAB 493.1215 calculated for C18H26N2012P
493.1222.
Pyridiyae pari.cf°atistatira 7-O phosphate (2Z): Colorless powder (63 mg); m.p.
270°C (dec.); 8H/ppm (300 MHz, D20) 8.66 (2H, d, J7 Hz, H-2 and H-6 pyridine), 8.49 ( 1 H, t, J 7 Hz, H-4 pyridine), 7.97 (2H, t, J 7 Hz, H-3 and pyridine), 6.45 (1H, bs, H-10), 5.95 (2H, s, OCH20), 4.41 (1H, bs, H-1), 4.15 (1H, bs, H-2), 3.97 (1H, bs, H-3), 3.89 (1H, m, H-4), 3.84 (1H, m, H-4a), 3.10 (1H, d, J 12 Hz, H-lOb).
Imidazole pahcratistatin 7-O phosphate (2m): Colorless powder (71 mg); m.p.
250°C (dec.); 8H/ppm (300 MHz, D20) 8.21 (1H, s, H-2 imidazole), 7.21 (2H, bs, H-4 and H-5 imidazole), 6.65 (1H, bs, H-10), 6.00 (1H, s, OCHAHBO), 5.95 (1H, s, OCHAHB O), 4.44 (1H, bs, H-1), 4.15 (1H, bs, H-2), 3.97 (1H, bs, H-3), 3.85 (1H, m, H-4), 3.75 (1H, m, H-4a), 3.38 (1H, d, J 12 Hz, H-lOb).

Quinifze pa~zc~atistatih 7-O phosphate (2n): White fluffy powder (0.11 g);
m.p. 173-175°C; 8H/ppm (300 MHz, DZO) 8.59 (1H, d, J4.5 Hz, H-2' quinine),,7.86 (1H, d, J9 Hz, H-8' quinine), 7.57 (1H, d, J4.5 Hz, H-3' quinine), 7.3 6 ( 1 H, dd, J 2, 9 Hz, H-5' quinine), 7.23 ( 1 H, m, H-7' quinine), 6.34 (1H, s, H-10), 5.91 (1H, s, OCHAH$O), 5.87 (1H, s, OCHAHBO), 5.66 ( I H, m, H-10 quinine), 5. 59 ( 1 H, m, H-9 quinine), 4.98 ( 1 H, d, J 17 Hz, H-11 a quinine), 4. 93 ( 1 H, d, J 11 Hz, H-11 b quinine), 4. 3 5 ( 1 H, b s, H-1 ), 4. I 2 ( 1 H, bs, H-2), 3.95 (1H, bs, H-3), 3.83-3.87 (5H, m, OMe quinine, H-4, H-4a), 3.80 (1H, m, H-6a quinine), 3.22 (2H, m, H-8 quinine, H-2a quinine), 2.93 (1H, d, J 12 Hz, H-lOb), 2.77 (2H, m, H-2b quinine, H-6b quinine), 2.04 (1H, m, H-3 quinine), 1.93 (2H, m, H-Sa quinine, H-7a quinine), 1.86 (1H, m, H-4 quinine), 1.62 ( 1 H, m, H-Sb quinine), 1.27 ( 1 H, m, H-7b quinine); n2/z (FAB) 730.2 [(M + H)+, 60%], 325,2 (100); HRFAB 730.2372 calculated for C3aH4iNsOi3P 730.2377.
QuiYaidine paizcf°atistatin 7-O phosphate (20): Colorless powder (0.14 g); m.p.
183-185°C; 8~/ppm (300 MHz, D20) 8.62 (1H, d, J4.5 Hz, H-2' quinidine), 7.88 (1H, d, J9 Hz, H-8' quinidine), 7.61 (1H, d, J4.5 Hz, H-3' quinidine), 7.41 (1H, dd, J2, 9 Hz, H-5' quinidine), 7.25 (1H, m, H-7' quinidine), 6.34 (1H, s, H-10), 5.89 (2H, s, OCH20), 5.66 (1H, m, H-10 quinidine), 5.59 (1H, 2o m, H-9 quinidine), 5.09 (1H, d, J 17 Hz, H-l la quinidine), 4.95 (1H, d, J

Hz, H-l 1b quinidine), 4.35 (1H, bs, H-1), 4.14 (1H, bs, H-2), 3.95 (1H, bs, H-3), 3.85-3.89 (5H, m, OMe quinidine, H-4, H-4a), 3.79 (1H, m, H-6a quinidine), 3.23 (2H, m, H-8 and H-2a quinidine), 2.93 (1H, d, J 12 Hz, H-10b), 2.65 (2H, m, H-2b and H-6b quinidine), 2.04 (1H, m, H-3 quinidine), 1.88 (2H, m, H-Sa and H-7a quinidine), 1.86 (1H, m, H-4 quinidine), 1.69 (1H, m, H-Sb quinidine), 1.44 (1H, m, H-7b quinidine); ni/z (FAB)730 [(M +
H)~, 5%], 325 (65), 154 (100); HRFAB 730.2384 calculated for C34H41N3W sP
730.2377.
Results and discussion From the outset, a more direct approach to the protected phosphate intermediate (5) was sought. In this respect, utilization of dibenzyl phosphite techniques (Silverberg et al., 1996, Tet~alaed~on Letters, 37, 771) by our group (Pettit et al., 1998, Arati-Cafzcer Drug Design, I3, 981 ) has proven to be an invaluable alternative to the alkylamidophosphine method (Perrich and Johns, 1988. Sysathesis, 142). Due to its poor solubility in organic solvents and the presence of four secondary alcohol groups in the C-ring, upon applying our carefully investigated dibenzyl phosphite methods, direct phosphorylation of (+)-pancratistatin (la) gave a very complex mixture of products as well as some unreacted starting material. Our attention was therefore focused on selective acetylation of the pancratistatin C-ring hydroxyl groups to give a more soluble and partially protected starting material for the phosphorylation.
That objective was easily realized when pancratistatin (la) was simply 2o allowed to react with acetic anhydride and 4-dimethylarninopyridine in pyridine to give the 1,2,3,4,7-O-pentaacetoxy derivative (3) and then inunediately converted to the desired 1,2,3,4-O-tetraacetate (4) by heating in refluxing water-pyridine (66% yield). Reaction of the latter with dibenzyl phosphate in the presence of carbon tetrachloride, N ethyldiisopropylamine and 4-dimethylaminopyridine afforded the heat sensitive pancratistatin 7-O-dibenzylphosphate (5) in 87% yield. Prolonged contact of (5) with heat and/or silica gel gave phosphate cleavage back to (4). The benzyl phosphate (5) was next deacetylated in the presence of sodium methoxide. Catalytic hydrogenation was used to cleave the benzyl ester groups, affording the corresponding phosphoric acid derivative (6). The acid was immediately treated with two equivalents of sodium methoxide to yield (92%) the disodium phosphate prodrug (2a).
to Once this more practical synthesis of (+)-pancratistatin prodrug (2a) was in hand, we proceeded to synthesize and evaluate a variety of canon derivatives of the parent phosphate. By treating the prodrug phosphoric acid precursor (6) with the appropriate base, the required salts (2b-o) were formed.
By replacing the sodium cations in prodrug (2a) with different cations, we were able to evaluate water solubility characteristics (mg/ml) as shown in Table II. Some of the ammonium cations were also explored with the goal of obtaining a stable, water-soluble drug with the ability to reverse multidrug resistance through interference with thep-glycoprotein mechanism (Sato et al., 1995, Cayacer Chemotlaes°apy a~zd PlaafrnZacology, 35, 271).
20 All of the synthetic products were evaluated against the murine P388 lymphocytic leukemia cell line and a minipanel of human cancer cell lines.
The results are summarized in Table I. Most of the prodrug derivatives exhibited activities analogous to that of (+)-pancratistatin (la) and the sodium prodrug (2a). Although the manganese (2i) and morpholine (2k) derivatives showed a 10- to 20-fold increase in activity, their poor water solubility made questionable their use for further preclinical development as potential prodrugs. Consequentially, the sodium derivative (2a) continues to be the preferred choice owing to its high activity, adequate water solubility, and the efficient synthesis described herein.
From the foregoing it becomes readily apparent new and useful antineoplastic preparations have been herein described and illustrated which fulfill all of the aforestated objectives. It is of course understood that such to modifications, alterations and adaptations as will readily occur to the artisan confronted with this disclosure are intended within the scope of the invention.

Claims (12)

Claims
1. A method of synthesizing phosphate prodrug comprising selectively protecting (+)-pancratistatin with a tetra acetate and thereafter phospharlating said protected pancratistatin with dibenzyl chloro phosphate; clearing the acetate and benzyl protecting groups with sodium methoxide while reacting with two equivalents of sodium methoxide to yield disodium (+)-pancratistatin phosphate.
2. The method of claim 1 in which the acetate and benzyl protecting groups are cleaned at room temperature.
3. The method of claim 1 in which is the sodium methoxide is replaced by a methoxide having and anion selected from the group consisting of lithium, potassium, risbridium, cesium, magnesium, calcium, zinc, manganese, piperazine, morpholine, pyridine, imidazoles, quinine, and quinidine.
4. The method of claim 3 in which the acetate and benzyl protective groups are cleaned at room temperature.
5. An improved method of synthesizing pancratistatin prodrug comprising, selecting pancratistatin as a starting material and obtaining a protected phosphate intermediate by means of utilizing dibenzyl phosphate techniques.
6. A method according to claim 5 containing the additional steps of:
selectively acetylating the C ring hydroxyl groups of said pancratistatin to obtain a pentaacetoxy derivative.
7. A method according to claim 6 containing the additional step of converting said pentaacetoxy derivative to a tetraacetate derivative.
8. A method according to claim 7 containing the additional step of reacting said tetraacetate with said dibenzyl phosphate in the presence of carbon tetrachloride to obtain a dibenzylphosphate derivative.
9. A method according to claim 8 wherein said dibenzyl phosphate was deacetylated in the presence of sodium methoxide.
10. A method according to claim 9 containing the additional step of catalytic hydrogenation of said deacetylated dibenzyl phosphate.
11. A method according to claim 9 containing the additional step of treating said phosphoric acid derivative with sodium methoxide to obtain a disodium phosphate pancratistatin prodrug.
12. A method according to claim 9 wherein said phosphoric acid derivative is treated with an appropriate base so as to replace the Phosphorous atom with an ion or molecule selected from the group of structures set forth in Figure 1 denoted as 2a - 2o inclusive.
CA002444471A 2001-04-18 2002-04-17 Synthesis of pancratistatin prodrugs Abandoned CA2444471A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US28471701P 2001-04-18 2001-04-18
US60/284,717 2001-04-18
PCT/US2002/011964 WO2002085848A2 (en) 2001-04-18 2002-04-17 Synthesis of pancratistatin prodrugs

Publications (1)

Publication Number Publication Date
CA2444471A1 true CA2444471A1 (en) 2002-10-31

Family

ID=23091255

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002444471A Abandoned CA2444471A1 (en) 2001-04-18 2002-04-17 Synthesis of pancratistatin prodrugs

Country Status (3)

Country Link
EP (1) EP1385846A4 (en)
CA (1) CA2444471A1 (en)
WO (1) WO2002085848A2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016130969A1 (en) 2015-02-13 2016-08-18 George Robert Pettit Silstatin compounds

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5529989A (en) * 1995-01-09 1996-06-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Pancratistatin prodrug

Also Published As

Publication number Publication date
WO2002085848A8 (en) 2003-05-30
EP1385846A2 (en) 2004-02-04
EP1385846A4 (en) 2005-11-30
WO2002085848A3 (en) 2003-04-24
WO2002085848A2 (en) 2002-10-31

Similar Documents

Publication Publication Date Title
EP2058320B1 (en) Process for preparing water-soluble phosphonooxymethyl derivatives of alcohol and phenol
EP1504014B1 (en) Process for preparation of cyclic prodrugs of pmea and pmpa
HU202547B (en) Process for producing epipodophyllotoxin-glycoside-4&#39;-phosphate derivatives and pharmaceutical compositions comprising such active ingredient
US20020119951A1 (en) Efficient method of synthesizing combretastatin A-4 prodrugs
Stawinski et al. Nucleoside H-phosphonates. 12. Synthesis of nucleoside 3'-(hydrogen phosphonothioate) monoesters via phosphinate intermediates
US20090082569A1 (en) Narcistatin prodrugs
US20100179108A1 (en) Synthesis of Sodium Narcistatin and Related Compounds
US20040127467A1 (en) Synthesis of pancratistatin prodrugs
CA2444471A1 (en) Synthesis of pancratistatin prodrugs
Wada et al. Nonoxidative chlorination of dialkyl phosphonates to dialkyl phosphorochloridites. A new approach to oligonucleotide synthesis
AU719277B2 (en) Non-nucleosidic coumarin derivatives as polynucleotide-crosslinking agents
Rakhit et al. Phospholipids. Part I. Synthesis of phosphatidyl ethanolamines
Legrand et al. Totally Regio‐and Stereoselective P–O‐to‐P–C Rearrangement in the Synthesis of Chiral P‐(o‐Hydroxyaryl) diazaphospholidine P‐Oxides
JP2005515958A (en) Synthesis of panclastatin prodrug
Pettit et al. Anticancer, pancratistatin, phosphate prodrugs, phosphorylation
Scheigetz et al. Synthesis of fluorescein phosphates and sulfates
EP0998518A1 (en) Process for the preparation of polyether phosphates
AU653788B2 (en) Process for the preparation of 4&#39;-demethylepipodophyllotoxin glucoside 4&#39;-phosphates
Chapman et al. Activated phosphate triesters. Synthesis and reactivity of N-hydroxysuccinimide and N-mercaptosuccinimide esters
US20040138182A1 (en) Colchicine derivatives, process for preparing them, products obtained therefrom and use thereof
IL107141A (en) PHOSPHOLIPID DERIVATIVES CONTAINING HIGHER ELEMENTS OF THE Vth MAIN GROUP, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
KR20000009216A (en) New manufacturing method of acechlorophenac
HU222122B1 (en) Process for producing intermediates for the synthesis of etoposide phosphate and etoposide
WO2006023445A2 (en) Psorospermin and analogues
JPS60120890A (en) Novel phosphorylation agent

Legal Events

Date Code Title Description
FZDE Dead