US20040229938A1 - Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents - Google Patents

Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents Download PDF

Info

Publication number
US20040229938A1
US20040229938A1 US10/872,616 US87261604A US2004229938A1 US 20040229938 A1 US20040229938 A1 US 20040229938A1 US 87261604 A US87261604 A US 87261604A US 2004229938 A1 US2004229938 A1 US 2004229938A1
Authority
US
United States
Prior art keywords
compound
glycol
purification
followed
reaction mixture
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
US10/872,616
Inventor
Mahmoud Elsohly
Samir Ross
Ahmed Galal
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.)
Individual
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
Priority to US10/872,616 priority Critical patent/US20040229938A1/en
Publication of US20040229938A1 publication Critical patent/US20040229938A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • 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
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to dihydroartemisinin and dihydroartemisitene dimers and their use in the treatment of cancer and as antiprotzoal agents.
  • Natural products have historically been a rich source of new, successful prototype classes of lead compounds from which analogs have been developed. According to a recent review, 60% of the anti-infective and anti-cancer drugs that have successfully advanced to the clinic are derived from natural products (2). Examples of these among currently used anti-cancer agents include the anthracycline class (e.g., doxorubicin), the Catharanthus (Vinca) alkaloids, paclitaxel, and derivatives of podophyllotoxin and camptothecin.
  • a recently published tabulation of natural product-based anti-tumor drugs shows more than 25 agents currently in Phase I or II (3). This and other recent reviews are important reminders of the critical role of natural products as a resource for the discovery of new anti-tumor agents (4,5).
  • the natural product artemisinin (1) is a sesquiterpene endoperoxide first isolated in 1971 from the Chinese plant Artemisia annua (6).
  • the compounds as numbered herein are depicted in FIG. 1.
  • the compound was shown to have anti-malarial activity against both chloroquine-sensitive and chloroquine-resistant strains of Plasmodium falciparum.
  • This invention comprises compositions containing dihydroartemisinin and dihydroartemisitene dimers with activity as anticancer agents and anti-protozal, including anti-malarial and anti-leishmanial properties.
  • This invention also describes methods of preparation of these compositions and methods of use of such compositions for the treatment of cancer, and protozoal infections, including malaria, or leishmaniasis.
  • the compositions of this invention have not been previously described.
  • the compounds of this invention represent a potential new class of anti-tumor agents, one that has shown promising activity against solid tumors, and with a pattern of selectivity that suggests a possible new mechanism of action.
  • artemisinin dimers were prepared in this invention by condensation of DHA with a variety of vicinal and non-vicinal glycols. These dimers have been evaluated in the NCI anti-tumor screening program, and all passed to the 60-cell line screen (4-9). Several of these compounds have been advanced into further testing by the NCI into the Hollow Fiber Assay (HFA) protocol (Compounds 4, 5, and 6). Additional supplies of Compounds 5 and 6 have been recently prepared and provided to the NCI for testing in xenograft tumor models.
  • HFA Hollow Fiber Assay
  • the present invention relates to a method of treating cancer comprising administering to a subject suffering from cancer an effective amount of at least one compound of the formula:
  • R is selected from one of the substituents described above or a simple (C 2 -C 4 ) alkyl residue.
  • the invention encompasses a method of treating a protozoal infection comprising administering to a subject suffering from an infection an effective amount of at least one compound of the formulas given hereinabove.
  • R is selected from one of the substituents described above or a simple (C 2 -C 4 ) alkyl residue.
  • a pharmaceutical composition can be prepared which comprise at least of one compound of this invention and pharmaceutically acceptable carrier and/or excipient.
  • Compounds of the invention can be prepared by reacting dihydroartemisin or dihydroartemistene with an appropriate optionally substituted 1, 2 or 1, 3 or 1, 4 glycol under acidic conditions such as borontrifluoride etherate followed by additional functionallization of the resulting dimer as necessary.
  • reaction comprises reaction of dihydroartemisinin with glycerol in the presence of an acid catalyst such as boron trifluoride etherate followed by purification of the reaction mixture.
  • an acid catalyst such as boron trifluoride etherate
  • R is a cyclohexane diol residue
  • the reaction can conprise of dihydroartemisinin with cis- or trans-cycloheane diols or a mixture thereof in the presence of an acid catalyst such as boron trifluoride etherate followed by purification of the reaction mixture and separation of the appropriate isomer.
  • an acid catalyst such as boron trifluoride etherate
  • R is a dihydroxy acetone residue
  • the reaction can be carried out in the presence of an acid such as boron trifluoride etherate followed by purification of the reaction mixture followed by sodium borohydride reduction and purification of the reaction mixture.
  • the hemisuccinate esters can be prepared by reacting the appropriate precursor with succinic anhydride in the presence of a base catalyst such as a mixture of dimethylaminopyridine and triethylamine followed by the purification of the reaction mixture.
  • a base catalyst such as a mixture of dimethylaminopyridine and triethylamine followed by the purification of the reaction mixture.
  • Dihydroartemisitene dimers can be prepared with the appropriate 1, 2 or 1, 3 or 1, 4 glycol in the presence of an acid catalyst such as borontrifluride etherate followed by the purification of the reaction mixture.
  • an acid catalyst such as borontrifluride etherate followed by the purification of the reaction mixture.
  • Illustrative glycols include, for example, ethylene glycol, 1, 2 propane-diol, glycerol, dihydroxy acetone, or 1, 4-butane-diol, 1, 4-cis-cyclohexanediol, 1, 4-trans-cyclohexanediol or a mixture thereof.
  • Administration of the instant dimers may be by any of the conventional routes of administration, for example, oral, subcutaneous, intraperitoneal, intramuscular, intravenous or rectally.
  • the compound is administered in combination with a pharmaceutically-acceptable carrier which may be solid or liquid, dependent upon choice and route of administration.
  • acceptable carriers include, but are not limited to, starch, dextrose, sucrose, lactose, gelatin, agar, stearic acid, magnesium stearate, acacia, and similar carriers.
  • liquids include saline, water, edible oils, e.g. peanut and corn.
  • the compound and diluent carrier When administered in solid form, may be in the form of tablets, capsules, powders, lozenges, suppositories prepared by any of the well known methods.
  • the mixture of active compound and liquid diluent carrier When given as a liquid preparation, may be in the form of a suspension administered as such.
  • the compound is administered in a non-toxic dosage concentration sufficient to inhibit the growth and/or destroy cancer or to destroy protozoal organisms such as malaria and leishmania.
  • the actual dosage unit will be determined by the well recognized factors as body weight of the patient and/or severity and type of pathological condition the patient might be suffering with. With these considerations in mind, the dosage unit for a particular patient can be readily determined by the medical practitioner in accordance with the techniques known in the medical arts.
  • the compounds of this invention have been prepared by reaction of dihydroartemisinin or dihydroartemistene with a variety of optionally substituted 1,2-, 1-3- or 1,4 glycols under acidic conditions (borontrifluoride etherate) in dry ether followed by chromatography of the reaction mixture to isolate the desired product.
  • Optional substitutients include, for example, alkoxy or acyloxy groups.
  • the dimers of the present invention can also be prepared by the reaction of dihydroxy ketones such as, for example, dihydroxyacetone, with DHA or dihydroartemistene followed by reduction of the keto-group and reaction of the hydroxy group formed in the reduction of the ketone with hydroxy reactive compounds such as mono or dicarboxylic acids as their acid halides and acid anhydrides.
  • the starting material (dihydroartemisinin) is prepared by sodium borohydrite reduction of the natural product artemisinin (1).
  • the latter compound is isolated from the leaves of Artemisia annua following the procedures previously described (15, 16).
  • the compounds of the invention were tested in the NCI anti-tumor screen and in the anti-malarial and anti-Leishmanial screens. The activities are shown in Tables 1-8 as shown in FIGS. 2A, 2B, 3A, 3B, 4A, 4B and FIGS. 5 to 9.
  • Mp's were recorded on an Electrothermal 9100 instrument.
  • IR spectra were obtained using AATI Mattson Genesis Series FTIR.
  • Optical rotations were recorded at ambient temperature using JASCO, DIP-370, digital polarimeter.
  • 1D and 2D NMR spectra were obtained on Bruker Avance DRX 500 spectrometers at 500 MHz ( 1 H) and 125 MHz ( 13 C) or Bruker DRX 400 spectrometer using the solvent peak as the internal standard.
  • HRESIFTMS were obtained using a Bruker Bioapex FT-MS in ESI mode. Low resolution MS were measured on a ThermoQuest aQa LC/MS.
  • the preferred method of preparing Compound 4 was to first prepare the ketone precursor through condensation of dihydroxyacetone with dihydroartemisinin in the presence of boron trifluoride-etherate followed by sodium borohydride reduction of the resulting ketone to give Compound 4. This is detailed in the following examples.
  • a mixture of dihydroartemisinin (372 mg 1.31 mmol) and 1,4-cyclohexanediol (cis and trans mixture) (74.4 mg, 0.64 mmol) were suspended in 10 mL dry ether and 0.25 mL of BF 3 .Et 2 O (280 mg, 1.97 mmol) was added at 0° C. The mixture was then stirred at room temperature for 80 minutes followed by workup by shaking with a sodium bicarbonate solution and separation of the ether layer. The aqueous layer was washed with ether (2 ⁇ 10 mL). The ether layers were combined and washed with water and brine and the ether layer was dried over anhydrous sodium sulfate.
  • a mixture of dihydroartemisinin (10 g, 35.2 mmol) and 1,4-cyclohexanediol (cis and trans mixture) (2 g, 17.2 mmol) were suspended in 260 mL dry ether and 1.11 mL of BF 3 .Et 2 O was added at 0° C. under argon. Two additional portions of BF 3 .Et 2 O (1.11 mL each) were added after 1 hr intervals.
  • Example 12 The reaction of Example 12 was repeated on a larger scale (starting with 550 mg of Compound 4) where all reactants were scaled up proportionally. Purification of the reaction product in the same manner produced 355 mg of Compound 8 as amorphous white powder with the following spectral characteristics:
  • Compounds of this invention were subjected to anti-cancer activity screen carried out by the National Cancer Institute (NCI) following their standard protocol against 60 different cancer cell lines.
  • NCI National Cancer Institute
  • the activity of the compounds of this invention against selected cell lines fro leukemia, non-small cell lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, and breast cancer were determined in terms of GI50 (Table 1), TGI (Table 2), and LC50 (Table 3) concentrations.
  • G150 is the concentration which inhibits 50% of the growth of the cells
  • TGI is the concentration causing total growth inhibition
  • LC50 is the concentration which kills 50% of the cells.
  • Compounds of this invention were subjected to in vitro assays to assess their anti-angiogenic activity. These assays are carried out by the NCI according to their standard protocol for HUVEC assays for initial in vitro testing. The three assays are the Growth Inhibition Assay, the Cord Formation Assay, and the Cell Migration Assay. Compounds are considered for further testing if activity is shown in at least one of the above assays. Table 5 (FIG. 6) shows the activity of compounds of this invention in these assays.
  • Table 6 shows the activity of compounds of this invention against two strains of the malaria parasite ( Plasmodium falciparum ), one is chloroquine sensitive (D6 clone) and one is chloroquine resistant (W2 clone). The cytotoxicity of the compounds was also assessed using Vero cells. The data show that compounds of this invention are more active than chloroquine or artemisinin as anti-malarial drugs.
  • Table 7 shows the activity of a selected group of compounds of this invention against the malaria parasite. These are from different synthetic lots than those tested in Table 6 (FIG. 7). This confirms the activity of compounds of this invention as anti-malarial agents.
  • Table 8 shows the activity of compounds of this invention against the leishmania parasite with activity comparable to that of pentamidine.

Abstract

This invention comprises compositions containing dihydroartemisinin and dihydroartemisitene dimers with activity as anticancer agents and anti-protozal, including anti-malarial and anti-leishmanial properties. This invention also describes methods of preparation of these compositions and methods of use of such compositions for the treatment of cancer, and protozoal infections, including malaria, or leishmaniasis.
The compounds of this invention represent a potential new class of anti-tumor agents, one that has shown promising activity against solid tumors, and with a pattern of selectivity that suggests a possible new mechanism of action.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application is a divisional application of U.S. Ser. No. 10/271,960, filed on Oct. 15, 2002. The disclosure of that application is expressly incorporated herein by reference.[0001]
  • FIELD OF INVENTION
  • The present invention relates to dihydroartemisinin and dihydroartemisitene dimers and their use in the treatment of cancer and as antiprotzoal agents. [0002]
  • BACKGROUND OF THE INVENTION
  • Cancer deaths in the U.S. alone were over 500,000 in 2001, and in spite of many advances, cancer remains one of the leading killers (1). There is a critical need for the development of new anti-cancer agents, especially those with novel and selective mechanisms of action. Although some of the promise of non-cytotoxic therapies is beginning to be realized (e.g. immunostimulants, growth factor antagonists, anti-sense therapy), the mainstay of the treatment of most cancers remains with cytotoxic drugs. In view of the limited success rates, incidence of toxicities, and development of resistance to such agents, there is a dire need for new classes of these drugs, especially those that may act by new mechanisms or exhibit exploitable selectivity. There is also a need for a better understanding of dosing, scheduling, and concomitant therapies in order to optimize treatment protocols. [0003]
  • Natural products have historically been a rich source of new, successful prototype classes of lead compounds from which analogs have been developed. According to a recent review, 60% of the anti-infective and anti-cancer drugs that have successfully advanced to the clinic are derived from natural products (2). Examples of these among currently used anti-cancer agents include the anthracycline class (e.g., doxorubicin), the Catharanthus (Vinca) alkaloids, paclitaxel, and derivatives of podophyllotoxin and camptothecin. A recently published tabulation of natural product-based anti-tumor drugs shows more than 25 agents currently in Phase I or II (3). This and other recent reviews are important reminders of the critical role of natural products as a resource for the discovery of new anti-tumor agents (4,5). [0004]
  • The natural product artemisinin (1) is a sesquiterpene endoperoxide first isolated in 1971 from the Chinese plant [0005] Artemisia annua (6). The compounds as numbered herein are depicted in FIG. 1. The compound was shown to have anti-malarial activity against both chloroquine-sensitive and chloroquine-resistant strains of Plasmodium falciparum.
  • Because of the importance of the clinical effects of artemisinin in treating malaria, many derivatives were prepared in order to develop the most effective and least toxic anti-malarial agent. Initially, simple derivatives were prepared—e.g., dihydroartemisinin (DHA, in which the lactone carbonyl is reduced resulting in a hemiacetal), artemether (the methyl ether of DHA) and several other ether and ester analogs. The sodium salt of the hemisuccinate ester (sodium artesunate) was one of these derivatives that showed more activity and less toxicity than artemether, the latter being more active than artemisinin itself. Continued interest in the activity of artemisinin and DHA analogs later resulted in the preparation of artemisinin acetal dimers through reaction of dihydroartemisinin with borontrifluoride-etherate. [0006]
  • In addition to its anti-malarial activity, artemisinin had been reported to have cytotoxic effects against EN-2 tumor cells (7), P-388, A549, HT-29, MCF-7, and KB-tumor cells (8). As more analogs were evaluated for anti-tumor activity, it was reported that the unsymmetrical dimer (2) showed strong cytotoxic activity and was more potent than cisplatin (9). The symmetrical dimer (3) also showed pronounced cytotoxic activity (10). [0007]
  • This finding stimulated interest in other types of DHA dimers. Posner et al. (11) prepared dimers linked with a polyethylene glycol spacer (3 units of ethylene glycol), an eight carbon glycol, and a dithio-derivative. The authors also prepared simpler trioxane dimers. Posner et al. also prepared several dimers of DHA where the linking units between the two molecules of dihydroartemisinin were dicarboxylic acids of different types (12). Zhang and Darbie (13,14) also proposed several dihydroartemisinin dimers to be linked via different coupling agents. Some of these artemisinin dimers and some of the simpler trioxanes had anti-malarial effects, anti-cancer activity, and anti-proliferative effects with some compounds being as active as calcitriol in an anti-proliferative assay in murine keratinocytes. [0008]
  • SUMMARY OF THE INVENTION
  • This invention comprises compositions containing dihydroartemisinin and dihydroartemisitene dimers with activity as anticancer agents and anti-protozal, including anti-malarial and anti-leishmanial properties. This invention also describes methods of preparation of these compositions and methods of use of such compositions for the treatment of cancer, and protozoal infections, including malaria, or leishmaniasis. The compositions of this invention have not been previously described. [0009]
  • The compounds of this invention represent a potential new class of anti-tumor agents, one that has shown promising activity against solid tumors, and with a pattern of selectivity that suggests a possible new mechanism of action.[0010]
  • DESCRIPTION OF THE INVENTION
  • In the interest of development of new chemotherapeutic agents, artemisinin dimers were prepared in this invention by condensation of DHA with a variety of vicinal and non-vicinal glycols. These dimers have been evaluated in the NCI anti-tumor screening program, and all passed to the 60-cell line screen (4-9). Several of these compounds have been advanced into further testing by the NCI into the Hollow Fiber Assay (HFA) protocol (Compounds 4, 5, and 6). Additional supplies of Compounds 5 and 6 have been recently prepared and provided to the NCI for testing in xenograft tumor models. [0011]
  • The present invention relates to a method of treating cancer comprising administering to a subject suffering from cancer an effective amount of at least one compound of the formula: [0012]
    Figure US20040229938A1-20041118-C00001
  • where R is [0013]
    Figure US20040229938A1-20041118-C00002
  • or a compound of the formula [0014]
    Figure US20040229938A1-20041118-C00003
  • where R is selected from one of the substituents described above or a simple (C[0015] 2-C4) alkyl residue.
  • Furthermore the invention encompasses a method of treating a protozoal infection comprising administering to a subject suffering from an infection an effective amount of at least one compound of the formulas given hereinabove. [0016]
  • Compounds within the scope of the invention are compounds of the formula: [0017]
    Figure US20040229938A1-20041118-C00004
  • where R is [0018]
    Figure US20040229938A1-20041118-C00005
  • or a compound of the formula [0019]
    Figure US20040229938A1-20041118-C00006
  • where R is selected from one of the substituents described above or a simple (C[0020] 2-C4) alkyl residue.
  • A pharmaceutical composition can be prepared which comprise at least of one compound of this invention and pharmaceutically acceptable carrier and/or excipient. [0021]
  • Compounds of the invention can be prepared by reacting dihydroartemisin or dihydroartemistene with an appropriate optionally substituted 1, 2 or 1, 3 or 1, 4 glycol under acidic conditions such as borontrifluoride etherate followed by additional functionallization of the resulting dimer as necessary. [0022]
  • In the case where R is a glycerol residue the reaction comprises reaction of dihydroartemisinin with glycerol in the presence of an acid catalyst such as boron trifluoride etherate followed by purification of the reaction mixture. [0023]
  • Where R is a cyclohexane diol residue the reaction can conprise of dihydroartemisinin with cis- or trans-cycloheane diols or a mixture thereof in the presence of an acid catalyst such as boron trifluoride etherate followed by purification of the reaction mixture and separation of the appropriate isomer. [0024]
  • Where R is a dihydroxy acetone residue the reaction can be carried out in the presence of an acid such as boron trifluoride etherate followed by purification of the reaction mixture followed by sodium borohydride reduction and purification of the reaction mixture. [0025]
  • The hemisuccinate esters can be prepared by reacting the appropriate precursor with succinic anhydride in the presence of a base catalyst such as a mixture of dimethylaminopyridine and triethylamine followed by the purification of the reaction mixture. [0026]
  • Dihydroartemisitene dimers can be prepared with the appropriate 1, 2 or 1, 3 or 1, 4 glycol in the presence of an acid catalyst such as borontrifluride etherate followed by the purification of the reaction mixture. [0027]
  • Illustrative glycols include, for example, ethylene glycol, 1, 2 propane-diol, glycerol, dihydroxy acetone, or 1, 4-butane-diol, 1, 4-cis-cyclohexanediol, 1, 4-trans-cyclohexanediol or a mixture thereof. [0028]
  • Although the mechanism of action of these DHA dimers remains to be determined, some clues regarding possible molecular targets are suggested. Use of the NCI COMPARE analysis revealed that the cell sensitivity profile of these compounds in the 60-cell line assay was similar to platinum compounds. These compounds inhibit cell replication by forming DNA intrastrand cross-links. Correlations on micro-array data for the 60 cell lines also indicate that cells most sensitive to these dimers contain lower levels of the mRNAs encoding proteins involved in integrin and hypoxia signaling. Lower levels of expression of these proteins may result in enhanced sensitivity either because these proteins are direct targets, or because their reduced expression reflects a condition within the cell (e.g., redox potential) that augments sensitivity. [0029]
  • Administration of the instant dimers may be by any of the conventional routes of administration, for example, oral, subcutaneous, intraperitoneal, intramuscular, intravenous or rectally. In the preferred embodiment, the compound is administered in combination with a pharmaceutically-acceptable carrier which may be solid or liquid, dependent upon choice and route of administration. Examples of acceptable carriers include, but are not limited to, starch, dextrose, sucrose, lactose, gelatin, agar, stearic acid, magnesium stearate, acacia, and similar carriers. Examples of liquids include saline, water, edible oils, e.g. peanut and corn. [0030]
  • When administered in solid form, the compound and diluent carrier may be in the form of tablets, capsules, powders, lozenges, suppositories prepared by any of the well known methods. When given as a liquid preparation, the mixture of active compound and liquid diluent carrier may be in the form of a suspension administered as such. The compound is administered in a non-toxic dosage concentration sufficient to inhibit the growth and/or destroy cancer or to destroy protozoal organisms such as malaria and leishmania. The actual dosage unit will be determined by the well recognized factors as body weight of the patient and/or severity and type of pathological condition the patient might be suffering with. With these considerations in mind, the dosage unit for a particular patient can be readily determined by the medical practitioner in accordance with the techniques known in the medical arts. [0031]
  • The compounds of this invention have been prepared by reaction of dihydroartemisinin or dihydroartemistene with a variety of optionally substituted 1,2-, 1-3- or 1,4 glycols under acidic conditions (borontrifluoride etherate) in dry ether followed by chromatography of the reaction mixture to isolate the desired product. Optional substitutients include, for example, alkoxy or acyloxy groups. The dimers of the present invention can also be prepared by the reaction of dihydroxy ketones such as, for example, dihydroxyacetone, with DHA or dihydroartemistene followed by reduction of the keto-group and reaction of the hydroxy group formed in the reduction of the ketone with hydroxy reactive compounds such as mono or dicarboxylic acids as their acid halides and acid anhydrides. The starting material (dihydroartemisinin) is prepared by sodium borohydrite reduction of the natural product artemisinin (1). The latter compound is isolated from the leaves of [0032] Artemisia annua following the procedures previously described (15, 16). The compounds of the invention were tested in the NCI anti-tumor screen and in the anti-malarial and anti-Leishmanial screens. The activities are shown in Tables 1-8 as shown in FIGS. 2A, 2B, 3A, 3B, 4A, 4B and FIGS. 5 to 9.
  • EXAMPLES
  • Reactions were run in oven dried round-bottomed flasks. Diethyl ether (ether) was distilled from sodium benzophenone ketyl prior to use under an atmosphere of argon. All chemicals were purchased from Sigma-Aldrich and used without further purification, except the diols, which were dried over grade I alumina prior to use. Artemisinin (1) was isolated from locally grown [0033] Artemisia annua L. plants, using a literature procedure (15,16), and was reduced to dihydroartemisinin as previously reported (17).
  • Column chromatography was performed using flash chromatography, using silica gel purchased from Merck (particle size 230-400 mesh). Analytical thin-layer chromatography (TLC) was performed with silica gel 60 F[0034] 254 plates (250 μm thickness; Merck), using n-hexane-EtOAc mixtures as solvent systems. Visualization was accomplished by spraying with p-anisaldehyde spray reagent followed by heating using a hot-air gun (18).
  • Mp's were recorded on an Electrothermal 9100 instrument. IR spectra were obtained using AATI Mattson Genesis Series FTIR. Optical rotations were recorded at ambient temperature using JASCO, DIP-370, digital polarimeter. 1D and 2D NMR spectra were obtained on Bruker Avance DRX 500 spectrometers at 500 MHz ([0035] 1H) and 125 MHz (13C) or Bruker DRX 400 spectrometer using the solvent peak as the internal standard. HRESIFTMS were obtained using a Bruker Bioapex FT-MS in ESI mode. Low resolution MS were measured on a ThermoQuest aQa LC/MS.
  • Preparation of the 1,3-β,β-Dihyroartemisinin Dimer with Glycerol (Compound 4) [0036]
  • Example 1
  • To a stirred solution of dihydroartemisinin (160 mg, 0.56 mmol) in a round bottomed flask (50 mL) in dry ether (10 mL), was added dry glycerol (26 mg) and BF[0037] 3.OEt2 (267 μL) using a hypodermic syringe. The mixture was stirred under argon for 70 min. then quenched and worked up as usual to leave a gummy residue (199 mg). Upon crystallization from ether, it yielded 4 (52 mg) as cubic crystals, 28.7%); [α]D+173° (c 0.022, CHCl3); IR (film) νmax: 352 (OH), 2953, 2933, 2881, 1449, 1376, 1194, 1176, 1134, 1107, 1027, 991 cm−1; 1H NMR (CDCl3, 500 MHz, for one of the two identical monomeric units): δ 5.40 (1H, s, H-5,), 4.79 (1H, d, J J=3.9, H-12), 4.78 (1H, d, J=3.5 Hz, H-12′), 3.87 (1H, m, H-16, H-16′, H-17), 3.49 (1H, dd, J=5.9, 4.3 Hz, H-18), 3.42 (1H, q, J=5.4 Hz, H-18′), 2.63 (1H, m, H-11), 2.34 (1H, ddd, J=14.0, 4.0, 3.9 Hz, H-3), 2.00 (1H, m, H-3′), 1.85 (1H, m, H-2), 1.68 (3H, m, H-2′, H-8, H-9), 1.46 (2H, m, H-7, H-8′), 1.39 (3H, s, Me-15), 1.34 (1H, m, H-10), 1.21 (1H, m, H-1), 0.92 (3H, d, J=6.4 Hz, Me-14), 0.90 (3H, d, J=7.4 Hz, Me-13), 0.89 (3H, d, J=7.3 Hz, H-13′), 0.87 (1H, m, H-9′); 13C NMR (CDCl3, 125 MHz): δ 104.5 (s, C-4), 103.13 (d, C-12), 103.06 (s, C-12′), 88.3 (d, C-5), 81.4 (s, C-6), 70.3 (t, C-16), 70.1 (t, C-18), 70.0 (d, C-17), 52.9 (d, C-1), 44.7 (d, C-7, C-7′), 37.7 (d, C-10), 36.8 (t, C-3), 35.0 (t, C-9), 31.27 (d, C-11), 31.25 (d, C-11′), 26.5 (q, C-15), 25.0 (t, C-2), 25.0 (t, C-8), 20.7 (q, C-14), 13.4 (q, C-13); HRESIFTMS [m/z] 625.351 [M+H]+ (calcd for C33H53O11, 625.3582).
  • Preferred Procedure for Preparation of Compound 4. [0038]
  • Example 2
  • The preferred method of preparing Compound 4 was to first prepare the ketone precursor through condensation of dihydroxyacetone with dihydroartemisinin in the presence of boron trifluoride-etherate followed by sodium borohydride reduction of the resulting ketone to give Compound 4. This is detailed in the following examples. [0039]
  • Preparation of the β,β-Dihydroartemisinin Dimer with Dihydroxyacetone (Compound 7) [0040]
  • Example 3
  • Dihydroartemisinin (284 mg, 1 mmol) and 1,3-dihydroxyacetone dimer (45.05 mg, 0.25 mmol) were suspended in diethylether (10 mL). To the mixture (cooled to 5° C. under argon) was then added 35.5 mg BF[0041] 3.Et2O (0.25 mmol, 31 μL) and the mixture stirred at 5° C. for 20 minutes then at room temperature for 1 hr. Workup as usual gave 319 mg of residue.
  • The residue was chromatographed on silica gel column (30 g) and eluted with hexane:EtOAC (95:5) with polarity increasing to 80:20. Fractions were collected and pooled according to TLC similarities to give four major fractions. The most polar fraction (140.2 mg) was identified as Compound 7 (converts to Compound 4 upon NaBH[0042] 4 reduction):
  • [0043] 1H-NMR in CDCl3 at 500 MHz: δ 5.44 (2H, s, H-5 & H-5′), 4.805 (2H, d, J=3.39 Hz, H-12 & H-12′), two broad doublets (2H each J=17.59) centered at δ 4.46 & 4.285 (H-16 & H-18), 2.665 (2H, m, H-11 & H-11′), 2.355 (2H, ddd, H-3), 2.025 (2H, m, H-3′), 1.88 (2H, m, H-2), 1.81 (4H, m, H-9 & H-9′), 1.675 (2H, m, H-8), 1.475 (4H, H-7, H-7′, H-10 & H-10′), 1.41 (6H, s, Me-15 & Me-15′), 1.255 (2H, m, H-1 & H-1′), 0.99-0.95 (12H, Me-14, Me-14′, Me-13 & Me-13′).
  • [0044] 13C-NMR in CDCl3 at 124 MHz: δ 204.8 (s, C=0), 104.53 (s, C-4), 102.87 (d, C-12), 88.51 (d, C-5), 81.36 (s, C-6), 72.06 (t, C-16 & C-17), 52.89 (d, C-1), 44.67 (d, C-7), 37.75 (d, C-10), 36.77 (t, C-3), 34.99 (5, C-9), 31.13 (d, C-11), 26.47 (q, C-15), 25.03 (t, C-2), 24.74 (t, C-8), 20.69 (q, C-14), 13.43 (q, C-13).
  • Example 4
  • To a suspension of 2.84 g dihydroartemisinin (10 mmol) and 450 mg (2.4 mmol) of 1,3-dihydroxyacetone dimer in ether (100 mL) was added 127 μL of BF[0045] 3.Et2O (142 mg, 1 mmol) at 5° C. The mixture was stirred at room temperature for 30 minutes, then a second portion (127 μL) of BF3.Et2O was added. A third portion and a fourth portion (254 μL) of BF3.Et2O were added at 15-minute intervals making up a total of 4 mmols. Stirring was continued for 1.5 hr. Workup as usual provided an oily residue which was chromatographed in a manner similar to that described under Example 3 and fractions were combined based on their TLC similarities.
  • The fractions with R[0046] f values corresponding to the dimer prepared in Example 3 were combined and the solvent evaporated to produce 2.05 g of an oily residue which foamed in vacuum. This material was identical to that prepared under Example 3 (Compound 7).
  • Example 5
  • To a suspension of 3.3 g dihydroartemisinin (11.6 mmols) and 522 mg, 2.9 mmols) of 1,3-dihydroxyacetone dimer (0.25 equivalent) in ether (100 mL) was added 0.88 mL of BF[0047] 3.Et2O (0.986 g, 6.9 mmol, 0.6 equivalent) at 5° C. The mixture was then stirred at room temperature for 3 hr, then worked up as usual to provide an oily residue. The residue was chromatographed over silica gel column (130 g) and eluted with mixtures of hexane-EtOAC ranging from 95:5 to 85:15 to give several fractions which were combined according to TLC similarities. Fractions containing the desired product (identical to that prepared under Example 3) were combined to give 1.628 g of Compound 7.
  • Preparation of the 1,3-β,β-Dihydroartemisinin Dimer of Glycerol (Compound 4) Starting from Compound 7. [0048]
  • Example 6
  • The ketone intermediate (Compound 7), (1.94 g, 312 mmols) was dissolved in 225 mL of a mixture of THF and water (2:1). The solution was stirred and NBH[0049] 4 (474 mg, 4 molar equivalent) was then added in portions at room temperature over a 15 minute period. The mixture was then neutralized with 2N HCl. The THF was then evaporated under vacuum. The precipitate was filtered and washed with water and air dried to give 1.8 g (92.5% yield) of Compound 4, identical to that prepared under Example 1).
  • Example 7
  • The same procedure described under Example 6 was repeated using 0.778 g of Compound 7 to yield 0.73 g of Compound 4. [0050]
  • Preparation of the β,β-Dihydroartemisinin Dimer with Cyclohexanediol (Compounds 5 and 6). [0051]
  • Example 8
  • In a round-bottomed flask (100 mL) was introduced dihydroartemisinin (850 mg, 3.0 mmol) and dry ether (25 mL) then the mixture was stirred at room temperature with cyclohexane-1,4-diol (mixture of cis and trans) (170 mg). To the stirred solution, BF[0052] 3.OEt2 (570 μL) was then added using a hypodermic syringe. The stirring was continued for 80 min., then the reaction was quenched and worked up as usual to leave a gummy residue (1.13 g). The residue was loaded on Si gel column (170 g) and eluted with increasing amounts of EtOAc in n-hexane (15→50%). Fractions of 5 mL were collected and similar fractions were pooled by guidance of TLC to afford Compound 5 (238 mg, oil). Earlier fractions were pooled and re-chromatographed on a silica gel column to yield 6 (70 mg, white amorphous solid). Compound 5; [α]D+142°(c 0.036, MeOH); IR (film) νmax: 2938, 2872, 1448, 1375, 1227, 1194, 1122, 1099, 1029 cm−1; 1H NMR (CDCl3, 500 MHz, for one of the identical monomereric units): δ 5.42 (1H, s, H-5), 4.90 (1H, d, J=3.3, H-12), 3.78 (1H, brs, H-16), 2.62 (1H, m, H-11), 2.36 (1H, ddd, J=14.0, 12.4, 3.8 Hz, H-3), 2.04 (1H, m, H-3′), 1.88 (2H, m, H-2, H-8), 1.77 (2H, m, H-2′, H-8′), 1.71 (2H, m, H-17a, H-17b), 1.65-1.56 (4H, m, H-9, H-9′, H-17′a, H-17′b), 1.48 (1H, m, H-7), 1.43 (3H, s, Me-15), 1.32 (1H, m, H-10), 1.25 (1H, m, H-1), 0.95 (3H, d, J=6.2 Hz, Me-14), 0.90 (3H, d, J=7.2 Hz, Me-13); 13C NMR (CDCl3, 125 MHz): δ 104.4 (s, C-4), 100.0 (d, C-12), 88.4 (d, C-5), 81.5 (s, C-6), 72.7 (d, C-16), 53.0 (d, C-1), 44.9 (d, C-7), 37.9 (d, C-10), 36.9 (t, C-3), 35.2 (t, C-9), 31.2 (d, C-11), 26.6 (q, C-15), 24.9 (t, C-2), 25.1 (t, C-8), 20.7 (q, C-14), 13.5 (q, C-13); HRESIFTMS [m/z] 647.3445[M−1](calcd for C36H55O10, 647.3510).
  • Compound 6; R[0053] f 0.42 (n-hexane:EtOAc, 8:2), [α]D+114° (c. 0.042, CHCl3); IR (film) no OH absorption; 1H NMR (CDCl3, 400 MHz): δ 5.41 (1H, s, H-5), 4.88(1H, d, J=3.3 Hz, H-12), 3.75 (1H, brs, H-16), 1.47 (1H, m, H-7), 1.43 (3H, s, Me-15), 1.26 (2H, m, H-1, H-10), 0.95 (3H, d, J=6.0 Hz, Me-14), 0.87 (3H, d, J=7.3 Hz, Me-13); 13C NMR (CDCl3, 100 MHz): δ 104.4 (s, C-4), 100.4 (d, C-12), 88.4 (d, C-5), 81.6 (s, C-6), 73.9 (t, C-16), 53.0 (d, C-1), 44.9 (m, C-7), 37.9 (d, C-10), 36.9 (t, C-3), 35.1 (t, C-9), 31.2 (d, C-11), 30.4 (t, C-17′), 27.5 (t, C-17), 26.6 (q, C-15), 24.9 (t, C-2), 25.1 (t, C-8), 20.8 (q, C-14), 13.5 (q, C-13); HRESIFTMS [m/z] 671.3772 [M+Na]+(calcd for C36H56O10Na, 671.3765).
  • Example 9
  • A mixture of dihydroartemisinin (372 mg 1.31 mmol) and 1,4-cyclohexanediol (cis and trans mixture) (74.4 mg, 0.64 mmol) were suspended in 10 mL dry ether and 0.25 mL of BF[0054] 3.Et2O (280 mg, 1.97 mmol) was added at 0° C. The mixture was then stirred at room temperature for 80 minutes followed by workup by shaking with a sodium bicarbonate solution and separation of the ether layer. The aqueous layer was washed with ether (2×10 mL). The ether layers were combined and washed with water and brine and the ether layer was dried over anhydrous sodium sulfate. Evaporation of the ether resulted in an oily residue (429 mg) which was chromatographed on silica gel column (30 g) and eluted with hexane ether mixtures ranging from 97:3 to 80:20 and fractions were pooled together according to their TLC similarities. Compound 6 (56 mg) was isolated as white solid and Compound 5 (24 mg) was isolated as an oil which foamed under vacuum. These were found to be identical to Compounds 5 and 6 prepared under Example 8.
  • Example 10
  • A mixture of dihydroartemisinin (744 mg, 2.62 mmol) and 1,4-cyclohexanediol (149 mg, 1.28 mmol) of the cis and trans mixture were stirred in 20 mL dry ether at 5° C. To the mixture was added BF[0055] 3.Et2O (83 μL, 0.655 mmol) and stirring was continued at room temperature for 1 hr. A second portion of BF3.Et2O (83 μL) was added, and the mixture continued to stir for 1 hr. The reaction mixture was then worked up as usual to give 890 mg of an oily residue. Column chromatography of the residue using silica gel (32 g) and elution with hexane:ether 92:2, 96:4, 94:6, and then 90:10 (200 mL each) yielded several fractions pooled according to TLC similarities. Compound 6 was isolated as cubic crystals (238 mg, melting point 146-148° C. Compound 5 was isolated from later fractions as an amorphous foam (184 mg, melting point 93-97° C.). These were found to be identical to those previously prepared under Examples 8 and 9.
  • Example 11
  • A mixture of dihydroartemisinin (10 g, 35.2 mmol) and 1,4-cyclohexanediol (cis and trans mixture) (2 g, 17.2 mmol) were suspended in 260 mL dry ether and 1.11 mL of BF[0056] 3.Et2O was added at 0° C. under argon. Two additional portions of BF3.Et2O (1.11 mL each) were added after 1 hr intervals. The mixture was then stirred at room temperature for 1 hr after the last addition of BF3.Et2O followed by workup as usual to give 12 g of an amorphous residue which was chromatographed on silica gel column to produce 2.69 g of Compound 5 and 2.78 g of Compound 6. These were found to be identical to Compounds 5 and 6 prepared under Examples 8-10.
  • It must be mentioned that should only Compound 5 or only Compound 6 be desired that only the cis or trans 1,4-Cyclohexanediol be used as the starting material to produce the desired product. [0057]
  • Preparation of the Hemisuccinate Ester of Compound 4 (Compound 8) [0058]
  • Example 12
  • To a stirred solution of Compound 4 (200 mg, 0.32 mmol) in dry methylene chloride (4 mL) were added triethylamine (0.14 mL, 1.3 equiv), dimethylaminopyridine (16 mg, 0.4 equiv) and succinic anhydride (92 mg, 3 equiv). The resulting solution was slowly stirred at room temperature for 16 hr. Following evaporation of the solvent under reduced pressure, the residue was purified over a silica gel column using hexane:acetone (6:4) as the eluent. The product of the reaction was isolated as white amorphous solid (156 mg) with R[0059] f value of 0.68 (hexane:acetone, 1:1) identified as the hemisuccinate ester of Compound 4 (Compound 8), based on spectral data (see details under Example 13).
  • Example 13
  • The reaction of Example 12 was repeated on a larger scale (starting with 550 mg of Compound 4) where all reactants were scaled up proportionally. Purification of the reaction product in the same manner produced 355 mg of Compound 8 as amorphous white powder with the following spectral characteristics: [0060]
  • [0061] 1H-NMR (acetone-d6, 500 MHz): δ 5.442 (1H, s, H-5), 5.396 (IH, s, H-5′), 5.181 (IH, t, J=4.9 Hz, H-17), 4.755 (IH, d, J=3.4 Hz, H-12), 4.740 (IH, d, J=3.4 Hz, H-12′), 3.990 (2H, m, H-16), 3.608 (IH, dd, J=4.5 Hz & 4.5 Hz, H-18), 3.560 (IH, q, J=5.19 Hz, H-18), 2.652 (4H, m, CO—CH2—CH2—CO—), 2.548 (2H, m, H-11 & H-11′), 2.304 (2H, ddd, J=3.0, 3.89 & 3.0 Hz, H-3), 2.065 (2H, m, H-3′), 1.886 (2H, m, H-2), 1.786 (2H, m, H-2), 1.786 (2H, m, H-9), 1.686 (2H, m, H-8), 1.534 (2H, m, H-2′), 1.489 (2H, m, H-7 & H-10), 1.332 (6H, s, Me-15 & Me-15′), 1.213 (2H, m, H-1 & H-1′), 0.980-0.937 (12H, Me-14, Me-13, Me-14′ & Me-13′).
  • [0062] 13CNMR (acetone-d6, 125 MHz): δ 172.99 (s, CO—OH), 171.7 (s, —CO—O—), 103.9 (s, C-4), 102.46 (d, C-12), 102.32 (d, C-12′), 88.02 (d, C-5), 87.99 (d, C-5′), 80.97 (s, C-6), 71.92 (d, C-17), 66.74 (t, C-16), 66.59 (t, C-18), 53.09 (d, C-1), 44.89 (d, C-7), 37.63 (d, C-10), 36.70 (t, C-3), 34.98 (t, C-8), 31.33 (d, C-11), 31.29 (d, C-11′), 29.48 & 28.78 (t, methylenes of CO—CH2—CH2—CO), 25.72 (q, C-15), 24.98 (t, C-2), 24.7 (t, C-9), 20.23 (q, C-14), 12.81 (q, C-13). HRESIFTMS m/z) 723.362 [M−H]+, (Calcd. for C37H55O14).
  • Preparation of the β,β-Dihydroartemisitene Dimer with Ethylene Glycol (Compound 9) [0063]
  • Example 14
  • To a stirred solution of dihydroartemisitene (prepared from artemisinin as previously described (19) (75 mg) in dry ether (15 mL) and ethylene glycol (52 mg), was added BF[0064] 3.OEt2 (18 μL) and the reaction mixture was allowed to stand for 24 hr, then quenched and worked up as usual. Column chromatography of the crude reaction mixture using a gradient of EtOAc in n-hexane (20%→50%) afforded Compound 9 as a gum (7 mg), [α]D+181° (c 0.022, MeOH); IR (film) νmax: 2937, 2875, 1681, 1449, 1376, 1191, 1102, 987 cm−1; 1H NMR (CDCl3, 400 MHz): δ 5.89 (1H, s, H-5), 5.38 (1H, s, H-12), 5.08 (1H, s, H-13a), 4.94 (1H, s, H-13b), 3.89 (1H, d, J=7.5 Hz, H-16a), 3.57 (1H, d, J=7.6 Hz, H-16b), 2.31 (1H, m, H-7), 1.44 (3H, s, Me-15), 1.25 (2H, m, H-1, H-10), 0.7 (3H, d, J=6.2 Hz, Me-14); 13C NMR (CDCl3, 100 MHz): δ 143.1 (s, C-11), 114.5 (t, C-13), 103.5 (s, C-4), 101.2 (d, C-12), 88.1 (d, C-5), 80.8 (s, C-6), 66.6 (t, C-16), 51.9 (d, C-1), 48.3 (d, C-7), 37.0 (d, C-10), 36.5 (t, C-3), 34.4 (t, C-9), 31.4 (t, C-8), 24.6 (t, C-2), 25.8 (q, C-15), 20.0 (q, C-13). HRESIFTMS [m/z] 613.2943 [M+Na]+ (calcd for C32H46O10Na, 613.2983).
  • Example 15
  • Compounds of this invention were subjected to anti-cancer activity screen carried out by the National Cancer Institute (NCI) following their standard protocol against 60 different cancer cell lines. The activity of the compounds of this invention against selected cell lines fro leukemia, non-small cell lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, and breast cancer were determined in terms of GI50 (Table 1), TGI (Table 2), and LC50 (Table 3) concentrations. G150 is the concentration which inhibits 50% of the growth of the cells, TGI is the concentration causing total growth inhibition, and LC50 is the concentration which kills 50% of the cells. [0065]
  • Example 16
  • Compounds of this invention were further tested under the NCI's Hollow Fiber Assay Standard Protocol which assesses the in vivo activity. Compounds are considered to have enough activity to progress into further testing if the combined IP and SC sores were ≧20 or if the SC score was ≧8 or if there was a net cell kill of one or more cell lines. Table 4 (FIG. 5) shows the results of the testing of compounds of this inversion in this assay. [0066]
  • Example 17
  • Compounds of this invention were subjected to in vitro assays to assess their anti-angiogenic activity. These assays are carried out by the NCI according to their standard protocol for HUVEC assays for initial in vitro testing. The three assays are the Growth Inhibition Assay, the Cord Formation Assay, and the Cell Migration Assay. Compounds are considered for further testing if activity is shown in at least one of the above assays. Table 5 (FIG. 6) shows the activity of compounds of this invention in these assays. [0067]
  • Example 18
  • Compounds of this invention were subjected to anti-protozoal screens at the National Center for Natural Products Research (NCNPR) at the University of Mississippi following standard protocols for assessing anti-malarial and anti-Leishmanial activity. Compounds' activities against these two organisms were compared to the activity of standard medications for each organism. [0068]
  • Table 6 (FIG. 7) shows the activity of compounds of this invention against two strains of the malaria parasite ([0069] Plasmodium falciparum), one is chloroquine sensitive (D6 clone) and one is chloroquine resistant (W2 clone). The cytotoxicity of the compounds was also assessed using Vero cells. The data show that compounds of this invention are more active than chloroquine or artemisinin as anti-malarial drugs.
  • Table 7 (FIG. 8) shows the activity of a selected group of compounds of this invention against the malaria parasite. These are from different synthetic lots than those tested in Table 6 (FIG. 7). This confirms the activity of compounds of this invention as anti-malarial agents. [0070]
  • Table 8 (FIG. 9) shows the activity of compounds of this invention against the leishmania parasite with activity comparable to that of pentamidine. [0071]

Claims (17)

1. A method of treating cancer comprising administering to a subject suffering from cancer an effective amount of at least one compound of the formula:
Figure US20040229938A1-20041118-C00007
where R is
Figure US20040229938A1-20041118-C00008
or a compound of the formula
Figure US20040229938A1-20041118-C00009
where R is selected from one of the substituents described above or a simple (C2-C4) alkyl residue.
2. A method of treating a protozoal infection comprising administering to a subject suffering from an infection an effective amount of at least one compound of the formula:
Figure US20040229938A1-20041118-C00010
where R is
Figure US20040229938A1-20041118-C00011
or a compound of the formula
Figure US20040229938A1-20041118-C00012
where R is selected from one of the substituents described above or a simple (C2-C4) alkyl residue.
3. A compound of the formula:
Figure US20040229938A1-20041118-C00013
where R is
Figure US20040229938A1-20041118-C00014
or a compound of the formula
Figure US20040229938A1-20041118-C00015
where R is selected from one of the substituents described above or a simple (C2-C4) alkyl residue.
4. A pharmaceutical composition comprising at least of one compound according to claim 3 and pharmaceutically acceptable carrier and/or excipient.
5. A method of preparing compounds of the formulas:
Figure US20040229938A1-20041118-C00016
where R is
Figure US20040229938A1-20041118-C00017
or a compound of the formula
Figure US20040229938A1-20041118-C00018
where R is selected from one of the substituents described above or a simple (C2-C4) alkyl residue;
comprising reacting dihydroartemisin or dihydroartemistene with an appropriate optionally substituted 1, 2 or 1, 3 or 1, 4 glycol under acidic conditions such as borontrifluoride etherate followed by additional functionallization of the resulting dimer as necessary.
6. The method of claim 5 where R is a glycerol residue comprising reaction of dihydroartemisinin with glycerol in the presence of an acid catalyst such as boron trifluoride etherate followed by purification of the reaction mixture.
7. The method of claim 5 where R is a cyclohexane diol residue comprising the reaction of dihydroartemisinin with cis- or trans-cycloheane diols or a mixture thereof in the presence of an acid catalyst such as boron trifluoride etherate followed by purification of the reaction mixture and separation of the appropriate isomer.
8. The method of claim 5 where R is a dihydroxy acetone residue in the presence of an acid such as boron trifluoride etherate followed by purification of the reaction mixture.
9. The method of claim 6 by sodium borohydride reduction of the compound of claim 8 followed by purification of the reaction mixture.
10. The method of preparing the hemisuccinate ester of the compound of claim 6 by reacting the compound of claim 6 with succinic anhydride in the presence of a base catalyst such as a mixture of dimethylaminopyridine and triethylamine followed by the purification of the reaction mixture.
11. The method of preparing compounds of claim 5 by reacting dihydroartemisitene with the appropriate 1, 2 or 1, 3 or 1, 4 glycol in the presence of an acid catalyst such as borontrifluride etherate followed by the purification of the reaction mixture.
12. The method of claim 11 where the 1, 2 glycol is ethylene glycol.
13. The method of claim 11 where the 1, 2 glycol is 1, 2 propane-diol.
14. The method of claim 11 where the 1, 3 glycol is glycerol.
15. The method of claim 11 where the 1, 3 glycol is dihydroxy acetone.
16. The method of claim 11 where the 1,4 glycol is 1, 4-butane-diol.
17. The method of preparing a compound of claim 11 where the 1, 4 glycol is selected from 1, 4-cis-cyclohexanediol, 1, 4-trans-cyclohexanediol or a mixture thereof, followed by the purification of the reaction mixture and separation of the desired product.
US10/872,616 2002-10-15 2004-06-21 Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents Abandoned US20040229938A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/872,616 US20040229938A1 (en) 2002-10-15 2004-06-21 Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10/271,960 US6790863B2 (en) 2002-10-15 2002-10-15 Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents
US10/872,616 US20040229938A1 (en) 2002-10-15 2004-06-21 Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/271,960 Division US6790863B2 (en) 2002-10-15 2002-10-15 Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents

Publications (1)

Publication Number Publication Date
US20040229938A1 true US20040229938A1 (en) 2004-11-18

Family

ID=32069214

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/271,960 Expired - Lifetime US6790863B2 (en) 2002-10-15 2002-10-15 Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents
US10/872,616 Abandoned US20040229938A1 (en) 2002-10-15 2004-06-21 Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents
US10/896,192 Expired - Lifetime US7098242B2 (en) 2002-10-15 2004-07-21 Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/271,960 Expired - Lifetime US6790863B2 (en) 2002-10-15 2002-10-15 Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/896,192 Expired - Lifetime US7098242B2 (en) 2002-10-15 2004-07-21 Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents

Country Status (7)

Country Link
US (3) US6790863B2 (en)
EP (1) EP1551389A4 (en)
JP (1) JP2006504753A (en)
AU (1) AU2003282543B2 (en)
CA (1) CA2501942A1 (en)
MX (1) MXPA05003920A (en)
WO (1) WO2004034976A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1311832C (en) * 2005-01-12 2007-04-25 四川科伦药业股份有限公司 Medicinal use of bromodihydroarteannuin
US20080275106A1 (en) * 2004-06-21 2008-11-06 The University Of Mississippi Anticancer and Antiprotozoal Dihydroartemisinene and Dihydroartemisitene Dimers with Desirable Chemical Functionalities
US20140243402A1 (en) * 2006-12-21 2014-08-28 The United States Of America As Represented By The Secretary Of The Army Artemisinins in the Clinical and Veterinary Management of Kinetoplastid Infextions
CN110305017A (en) * 2019-08-09 2019-10-08 大理大学 Sesquiterpene eudesmanolide class compound and its pharmaceutical composition and preparation method and application are dropped

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6790863B2 (en) * 2002-10-15 2004-09-14 University Of Mississippi Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents
FR2899586B1 (en) * 2006-04-11 2008-06-20 Sanofi Aventis Sa DIMERS OF ARTEMISININE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
FR2934593B1 (en) * 2008-07-29 2010-09-10 Pf Medicament ARTEMISININE DIMERIC DERIVATIVES AND ANTICANCER THERAPY APPLICATION
WO2011089507A1 (en) 2010-01-22 2011-07-28 Council Of Scientific & Industrial Research A new series of artemisinin derivatives and process for preparation thereof
WO2012111025A2 (en) 2011-02-14 2012-08-23 Council Of Scientific & Industrial Research 1,2,3-triazole containing artemisinin compounds and process for preparation thereof
US9358261B2 (en) 2011-10-25 2016-06-07 U.S. Phytotherapy, Inc. Additional artemisinin and berberine compositions and methods of making
US9675582B2 (en) 2011-10-25 2017-06-13 U.S. Phytotherapy, Inc. Alternative ACT with natural botanical active GRAS ingredients for treatment and prevention of the Zika virus
US9011892B2 (en) 2011-10-25 2015-04-21 U.S. Phytotherapy, Inc. Artemisinin with berberine compositions and methods of making
WO2014116642A1 (en) * 2013-01-22 2014-07-31 The Johns Hopkins University Two-carbon linked artemisinin-derived trioxane dimers
WO2015041723A1 (en) 2013-09-17 2015-03-26 Kryptonite Group, Ltd Artemisinin-based combination therapy for treating viral mediated disease
CN106928274B (en) * 2017-02-28 2019-09-10 东南大学 A kind of dihydroartemisinine diploid derivative, its pharmaceutical composition and application
CN109467565B (en) * 2018-08-16 2020-07-10 云白药征武科技(上海)有限公司 Dihydroartemisinin trimer and preparation method and application thereof
CN110448551A (en) * 2019-08-23 2019-11-15 西南大学 Dihydroqinghaosu is preparing the application in anti-angiogenic medicaments
CN110354270B (en) * 2019-08-30 2022-04-15 中国中医科学院中药研究所 Artesunate polyethylene glycol derivative and preparation method and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6790863B2 (en) * 2002-10-15 2004-09-14 University Of Mississippi Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4647674A (en) * 1985-10-07 1987-03-03 Pennwalt Corporation Bis-(dibenzo[b,d]pyranyloxy) propanes
US4952603A (en) 1988-06-20 1990-08-28 Elferaly Farouk S Method for the isolation of artemisinin from Artemisia annua
IT1247476B (en) * 1991-06-14 1994-12-17 Fidia Spa DERIVATIVES OF CUMARINA.
US5677468A (en) 1995-06-29 1997-10-14 Hauser, Inc. Artemisinin dimer compounds having anticancer activity
US6160004A (en) * 1997-12-30 2000-12-12 Hauser, Inc. C-10 carbon-substituted artemisinin-like trioxane compounds having antimalarial, antiproliferative and antitumor activities

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6790863B2 (en) * 2002-10-15 2004-09-14 University Of Mississippi Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080275106A1 (en) * 2004-06-21 2008-11-06 The University Of Mississippi Anticancer and Antiprotozoal Dihydroartemisinene and Dihydroartemisitene Dimers with Desirable Chemical Functionalities
US7842720B2 (en) 2004-06-21 2010-11-30 Elsohly Laboratories, Incorporated Anticancer and antiprotozoal dihydroartemisinene and dihydroartemisitene dimers with desirable chemical functionalities
CN1311832C (en) * 2005-01-12 2007-04-25 四川科伦药业股份有限公司 Medicinal use of bromodihydroarteannuin
US20140243402A1 (en) * 2006-12-21 2014-08-28 The United States Of America As Represented By The Secretary Of The Army Artemisinins in the Clinical and Veterinary Management of Kinetoplastid Infextions
CN110305017A (en) * 2019-08-09 2019-10-08 大理大学 Sesquiterpene eudesmanolide class compound and its pharmaceutical composition and preparation method and application are dropped

Also Published As

Publication number Publication date
AU2003282543B2 (en) 2009-02-26
WO2004034976A3 (en) 2004-08-12
AU2003282543A1 (en) 2004-05-04
US7098242B2 (en) 2006-08-29
MXPA05003920A (en) 2005-06-17
US20040072896A1 (en) 2004-04-15
US20040266860A1 (en) 2004-12-30
EP1551389A4 (en) 2008-08-27
EP1551389A2 (en) 2005-07-13
CA2501942A1 (en) 2004-04-29
WO2004034976A2 (en) 2004-04-29
JP2006504753A (en) 2006-02-09
US6790863B2 (en) 2004-09-14

Similar Documents

Publication Publication Date Title
US7098242B2 (en) Dihydroartemisinin and dihydroartemisitene dimers as anti-cancer and anti-infective agents
Chaturvedi et al. Artemisinin and its derivatives: a novel class of anti-malarial and anti-cancer agents
Galal et al. Synthesis and evaluation of dihydroartemisinin and dihydroartemisitene acetal dimers showing anticancer and antiprotozoal activity
AU739687B2 (en) C-10 carbon-substituted artemisinin-like trioxane compounds having antimalarial, antiproliferative and antitumour activities
CN101481401B (en) Tripterygium wilfordii diterpenoid lactone derivative, pharmaceutical composition thereof and use in genital system tumor resistance
EP2223929A1 (en) Gambogic glycoside derivatives and analogs, the preparation and the application thereof
WO2011017998A1 (en) Gambogic acid cyclization analogues, preparation method and application thereof
US6156790A (en) C-10 carbon-substituted artemisinin-like trioxane compounds having antimalarial, antiproliferative and antitumor activities
EP1753419B1 (en) Anticancer and antiprotozoal dihydroartemisinin and dihydroartemisitene dimers with desirable chemical functionalities
El-Feraly et al. A new method for the preparation of arteether and its C-9 epimer
ElSohly et al. New orally active artemisinin dimer antimalarials
Truong et al. Novel Thioethers of Dihydroartemisinin Exhibiting Their Biological Activities
EP1758904B1 (en) Flavopereirine derivatives for cancer therapy
CA2787722C (en) A series of artemisinin derivatives and process for preparation thereof
CN1429203A (en) Sponge antitumor compounds

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION