AU2589900A - Synthesis of dihydrohonokiol compositions - Google Patents

Synthesis of dihydrohonokiol compositions Download PDF

Info

Publication number
AU2589900A
AU2589900A AU25899/00A AU2589900A AU2589900A AU 2589900 A AU2589900 A AU 2589900A AU 25899/00 A AU25899/00 A AU 25899/00A AU 2589900 A AU2589900 A AU 2589900A AU 2589900 A AU2589900 A AU 2589900A
Authority
AU
Australia
Prior art keywords
compound
och
diazepam
cooh
dihydrohonokiol
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
AU25899/00A
Inventor
Neera Satsangi
Rajiv K. Satsangi
William B. Stavinoha
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.)
University of Texas System
Original Assignee
University of Texas System
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 University of Texas System filed Critical University of Texas System
Publication of AU2589900A publication Critical patent/AU2589900A/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C39/00Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring
    • C07C39/205Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring polycyclic, containing only six-membered aromatic rings as cyclic parts with unsaturation outside the rings
    • C07C39/21Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring polycyclic, containing only six-membered aromatic rings as cyclic parts with unsaturation outside the rings with at least one hydroxy group on a non-condensed ring

Description

WO 00/40532 PCTIUS99/30168 1 DESCRIPTION SYNTHESIS OF DIHYDROHONOKIOL COMPOSITIONS BACKGROUND OF THE INVENTION 5 1.1 Field of the Invention The present invention relates to the synthesis of dihydrohonokiol, its derivatives, analogs and homologs, and to methods of use for the dihydrohonokiol compounds. Also included are compositions particularly useful for treatment of anxiety disorders. 10 1.2 Description of Related Art Anxiety and anxiety-related disorders are extremely common. Anxiety-related conditions can be relatively mild or can be sufficiently severe as to be disabling. Also noteworthy is that anxiety, while infrequently a "disease" in itself, is an almost 15 inevitable and often exacerbating consequence of many other medical and surgical conditions. Estimates of the number of patients suffering from various anxiety disorders range between 12 and 35 million persons in seven major industrialized nations. Saiboku-to, a Chinese herbal medicine, has long been used to treat anxiety and 20 neurotic disorders (Hosaya and Yamamura, 1988; Nartia, 1990). A disadvantage of Saiboku-to is that it requires approximately seven days or more of daily administration before an anxiolytic effect is observed (Maruyama et al., 1998). Fractionation of Saiboku-to has identified the two putative principal active anxiolytic components as magnolol (5,5'-di-(2-propenyl)-1,1'-biphenyl-2,2'-diol) and its positional isomer, 25 honokiol (3,5'-di-(2-propenyl)-1,1'-biphenyl-2',4diol) (Maruyama et al., 1998). Behavioral tests indicate that honokiol is at least 5000 times more potent than Saiboku-to. But honokiol still requires several days of administration to elicit an WO 00/40532 PCT/US99/30168 2 anxiolytic effect (Maruyama et al., 1998). The delay in the onset of anxiolytic activity of honokiol is thought to be due to either changes in receptors or a slow build-up of honokiol metabolites within the body. The metabolic pathway of honokiol has not been fully established. The 5 positional isomer magnolol is known to be metabolized to a number of compounds including the hydrogenated products dihydromagnolol (5-(2-propenyl)-5'-n-propyl 1,1'biphenyl-2,2'diol) and tetrahydromagnolol (5,5'-6t-n-propyl-1,1'biphenyl-2,2' diol) (Hattori et al., 1984a). The excretion of the reduced metabolites in feces and urine increased significantly in amount after repeated daily administration of 10 magnolol. This pattern was possibly the result of induction of the enzymes responsible for the metabolism. Benzodiazapines, such as diazepam and alprazolam, represent the most commonly used class of anxiolytic agents administered for the treatment of anxiety. Benzodiazapines can act to counteract anxiety by depressing the electrical 15 afterdischarge in the limbic system, and appear to potentiate neural inhibition that is mediated by gamma-aminobutyrate (GABA) (Baldessarini, 1990). These compounds have proven to be effective at reducing anxiety, but they also have significant side effects including sedation, ataxia, amnesia, dependence, tolerance, and behavioral disturbances and act as skeletal muscle relaxers. These side effects can render these 20 compounds unsuitable for many patients, particularly those whose anxiety is coupled to another form of illness. In addition to benzodiazepines, other drugs used to treat anxiety include barbiturates, certain anticholinergic agents, antihistamines, and azaspirodecanediones (Baldessarini, 1990). These drugs are sedatives, or at least have many properties in 25 common with traditional sedatives. The barbiturates are general neuronal depressants. The use of certain anticholinergic agents and antihistamines for treating anxiety appears to be based on their sedative properties. The azaspirodecanediones, buspirone in particular, are used in treating anxiety but require weeks of administration before anxiolytic activity is noted. The mechanism of action of the azaspirodecanediones is 30 unknown.
WO 00/40532 PCT/US99/30168 3 2.0 SUMMARY OF THE INVENTION The present invention addresses problems associated with commonly-used anxiolytic drugs, with Saiboku-to therapy in general, and with honokiol in particular, for treatment of anxiety. The present invention provides compounds which exhibit 5 anxiolytic activity within hours without many of the negative side effects associated with well-known anxiolytic compounds, such as the benzodiazepines. The invention also provides a method of treating anxiety disorders. Further, the invention provides methods of synthesis of novel anxiolytic compounds. 10 2.1 Novel Anxiolytic Compounds The present invention provides new anxiolytic compounds that do not have the adverse side effects of the benzodiazepines. In particular embodiments, the invention relates to the use of dihydrohonokiol, its derivatives, analogs and homologs, as anxiolytic agents. The inventors have demonstrated that dihydrohonokiol, for 15 example, exerts an anxiolytic effect more rapidly than honokiol and exhibits greater potency and fewer side effects than diazepam, a benzodiazepine anxiolytic. An aspect of the present invention encompasses novel compositions of matter comprising compounds of the formula: H H 6H 20 R R In a preferred embodiment, R is CH 2
-CH
2
-CH
3 and X is 5'-CH 2
-CH=CH
2 and X' is 3-CH 2
-CH=CH
2 . In other embodiments, the group R represents -CH 2
-CH=CH
2 , CH=CH-CH 3 , 25 or -CH 2
-CH
2
CH
3 . The group X represents from one to two substituents in any of the 3-, 4-, 5-, or 6-positions and is separately and independently fluorine, hydroxy, methoxy, (1-adamantyl), C1-Cs alkyl, C 2
-C
3 alkenyl, C 2
-C
3 alkylcarbonyl or C 1
-C
4 WO 00/40532 PCT/US99/30168 4 carboxyalkyl. The CI-C 5 alkyl group may be substituted with one or more fluorine or hydroxyl. Excepted from this list are the compounds where R is -CH 2
-CH=CH
2 and X is 5'-CH 2
-CH
3 , 5'-CH 2
-CH=CH
2 , 5'-OfH, 3'-OH, 5'-CH 2
-CH-CH
3 , and 3'-OCH 3 , 5'-CH 2
-CH=CH
2 , and where R is -CH 2
-CH
2
-CH
3 .and X is 3-CH 2 -CHr-CH 3 . 5 In particular, X may represent: 5'-CH=CH 2 ; 5'-CH 2
-CH
3 ; 5'-CH=CH 2 , 3'-OH; 5'-CH 2
-CH=CH
2 ; 5'-CH-CH=CH 2 , 3'-OH; 5'-CH 2
-CH
2
-CH
3 , 3'-OH; 5'-CH 2 CH=CH 2 , 3'-OCH 3 ; 5'-CHr-CH 2
-CH
3 , 3'-OCH 3 ; 5'-CH=CH-CH 3 , 3'-OH; 5'-CH=CH
CH
3 , 3'-OCH 3 ; 5'-CH 3 ; 5'-CH(CH 3
)
2 ; 5'-CH 2
CH(CH
3
)
2 ; 5'-C(CH 3
)
3 ; 5'
CH(CH
3
)
2
C
2
H
5 ; 5'-(l-adamantyl); 5'-CH(CH 3
)
2 , 6 -CH 3 ; 5'-CH(CH 3
)
2 , 4 -CH 3 ; 5' 10 C(=O)-CH 3 ; 5'-CH(-OH)-CH 3 ; 5'-CH 2
-C(=O)-CH
3 ; 5'-CH 2
-CH(-OH)-CH
3 ; 5'-CH 2 COOH; 5'-CH 2
CH
2 -COOH; 5'-CH 2
CH
2
CH
2 -COOH; 5'-CH 2 -COOH, 3-OH; 5'-OH; 5'-OCH 3 ; 3'-F; 4'-F; 5'-F; 3'-F, 5'-CH 3 ; 3'-F, 5'-CH 2
-CH
3 ; 3'-F, 5'-CH 2 -OH; 3'-F, 5'
CH
2
-CH
2 -OH; 3'-F, 5'-COOH; 3'-F, 5'-CH-COOH; 3'-F, 5'-CH=CH-CH 3 ; 3'-F, 5'
CH
2
-CH
2
-CH
3 ; 3'-F, 5'-CH 2 -CH=CH2; 3'-F, 5'-CH 2
-CHF-CH
3 ; 3'-F, 5'-CH 2
-CHF
15 CH 2 F; 4'-F, 5'-CH 3 ; 4'-F, 5'-CH 2 -OH; 3'-F, 5'-CH 2
-CH
3 ; 6'-F, 5'-CH 2
-CH
3 ; 6'-F, 5'
CH
2 -OH; and 6'-F, 5'-COOH. In other embodiments, the group X' represents from one to two substituents in any of the 3-, 4-, 5-, or 6-positions and is separately and independently fluorine, hydroxy, methoxy, Ci-C 4 alkyl, C 3 alkenyl, Ci-C 3 alkylcarbonyl, C1 -C 3 carboxyalkyl 20 or C3 carboxyalkenyl. The alkyl group may be substituted with one or more fluorine or hydroxyl. Excepted from this list are where R = -CH 2
-CH=CH
2 and X' = 3-CH 2 CH 3 , 3'-CH 2
-CH=CH
2 , and where R = -CH 2
-CH
2
-CH
3 and X' = 3-CH 2
-CH
2
-CH
3 . In particular, X' may represent: 3-CH 3 ; 3-CH 2
-CH
3 ; 3-CH 2
-CH=CH
2 ; 3-CH 2 CH=CH 2 , 5-OH; 3-CH 2
-CH
2
-CH
3 , 5-OH; 3-CH 2
-CH=CH
2 , 5-OCH 3 ; 3-CH 2
CH
2
CH
3 , 25 5-OCH 3 ; 3-CH=CH-CH 3 ; 3-CH 3 , 6-CH(CH 3
)
2 ; 3,5 di-CH 3 ; 2,6 di-CH 3 ; 3-CH(CH 3
)
2 ; 3-CH(CH 3
)
2 , 6-CH 3 ; 3-CH 2
CH(CH
3
)
2 ; 3-C(=O)-CH 3 ; 3-CH(-OH)-CH 3 ; 3-CH 2 C(=0)
CH
3 ; 3-CH 2
CH(-OH)-CH
3 ; 3-COOH, 6-OH; 2-COOH; 3-OCH 3 ; 3-CH 2 COOH; 3-CH 2
CH
2 COOH; 3-CH=CHCOOH; 3-CHO, 2-OH; 3-CH 2 OH, 2-OH; 3-CHO, 5
OCH
3 ; 3-CH 2 OH, 5-OCH 3 ; 3-CHO, 5-CH 3 ; 3-CH 2 OH, 5-CH 3 , 3-F; 2F; 2-F, 3-CH 3 ; 30 6-F, 3-CH 3 ; 5-F, 3-CH=CH-CH 3 ; 5-F,3-CH 2
-CH
2
-CH
3 ; 5-F, 3-CH 2 -CH=CH2; 5-F, 3
CH
2
-CHF-CH
3 ; or 5-F, 3-CH 2
-CHF-CH
2
F.
WO 00/40532 PCT/US99/30168 5 2.1.2 Pharmaceutical Compositions Another aspect of the present invention includes novel compositions comprising dihydrohonokiol, its derivatives, analogs and homologs. It will, of course, 5 be understood that one or more dihydrohonokiol, dihydrohonokiol derivative, analogs and homologs may be used in the methods and compositions of the invention. The maximum number of novel anxiolytic compounds of the present invention that may be administered is limited only by practical considerations, such as the possibility of eliciting an adverse effect. 10 Compositions employing the novel anxiolytic compounds will contain a biologically effective amount of the anxiolytic compound. As used herein, a "biologically effective" amount of an anxiolytic compound refers to an amount effective to alter, modulate, or reduce anxiety or related conditions. As disclosed herein, different amounts of the novel anxiolytic compounds may be effective. 15 Clinical doses will of course be determined by the nutritional status, age, weight and health of the patient. The quantity and volume of the composition administered will depend on the subject and the route of administration. The precise amounts of active anxiolytic compounds required will depend on the judgment of the practitioner and may be peculiar to each individual. However, in light of the data 20 presented here, the determination of a suitable dosage range for use in different mammals will be straightforward. Pharmaceutical compositions prepared in accordance with the present invention find use in several applications, including inhibition or reduction of anxiety. Such methods generally involve administering to a mammal a pharmaceutical 25 composition comprising an anxiolytically effective amount of dihydrohonokiol, its derivatives, analogs or homologs. Therapeutic kits comprising dihydrohonokiol, its derivatives, analogs and homologs comprise another aspect of the present invention. Such kits will generally contain, in suitable container means, a pharmaceutically acceptable formulation of 30 dihydrohonokiol, its derivatives, analogs and homologs. The kit may have a single container means that contains the novel composition(s) or it may have distinct WO 00/40532 PCT/US99/30168 6 container means for the novel composition(s) and other reagents which may be included within such kits. 2.2 Method of Treating Anxiety Disorders 5 The invention also includes a method of treating anxiety disorders with dihydrohonokiol, its derivatives, analogs and homologs. This method comprises administration of an anxiolytic amount of a suitable composition containing dihydrohonokiol, its derivatives, analogs and homologs, to a subject in need thereof. Administration is preferably by oral dosage, but any route of administration capable 10 of delivering an effective dose my be used. The choice of the composition to be used for treatment, the amount of the composition to be administered, and the duration of treatment will depend on the judgment of the practitioner and may be peculiar to each individual, as disclosed above. The treatment may be maintained as long as necessary and may be used in conjunction with other forms of treatment. 15 2.3 Methods of Synthesis of Anxiolytic Compounds In yet another aspect, the invention relates to synthetic methods for producing dihydrohonokiol and its various derivatives, analogs and homologs. The compounds may be synthesized from honokiol derived from plant sources. This reaction is 20 described herein as a hemi-synthesis. Alternatively, the total synthesis may be accomplished from the asymmetric coupling of 4-allylphenylalkyl ether with 4-alkoxy haloaryls, followed by dealkylation of the dialkoxyl biaryl formed. In the hemi-synthesis of dihydrohonokiol, honokiol is partially hydrogenated by reacting hydrogen atoms in the presence of a tris-(triphenylphosphine) rhodium(I) 25 chloride catalyst with honokiol dissolved in a non-polar solvent. In preferred embodiments, the non-polar solvent is selected from toluene, benzene or hexane. Under conditions of a limited supply of hydrogen, the predominant reaction product has only one of the two similar allylic groups of the biphenyl molecule reduced. Tetrahydrohonkiol is formed as a minor component, thus enabling unreacted honokiol 30 to be recovered and reused. The partially hydrogenated honokiol products are collected and the two dihydrohonokiol isomers are isolated and purified.
WO 00/40532 PCT/US99/30168 7 The total synthesis of 3-n-propyl-5'-(2-propenyl)-1,1'-biphenyl-2',4-diol, one of the two isomers of dihydrohonokiol, designated herein as I, utilizes 4-allylanisole as the starting chemical compound. This starting chemical compound is reacted with tert-butyllithium and anhydrous zinc chloride. The intermediate compound so formed 5 is coupled with 2-propyl-4-iodo-anisole in the presence of a catalyst, resulting in 2',4-dimethoxy-3-n-propyl-5'-(2-propenyl)-1,1'-biphenyl, which after demethylation generates 3-n-propyl-5'-(2-propenyl)-1,1'-biphenyl-2',4-diol (I) as one of the reaction products. The synthesis of the other isomer of dihydrohonokiol, 5'-n-propyl-3-(2 propenyl)-1,1'-biphenyl-2',4-diol, hereinafter designated as II, can be accomplished in 10 a similar manner, using 4-propylanisole as the starting chemical compound and reacting it with tert-butyllithium and anhydrous zinc chloride. This intermediate compound is coupled with 2-allyl-4-iodo-anisole in the presence of a catalyst, resulting in 2',4-dimethoxy-5'-n-propyl-3-(2-propenyl)-1,1'-biphenyl, which after demethylation generates 5'-n-propyl-3-(2-propenyl)-1,1'-biphenyl-2',4-diol (II) as one 15 of the reaction products. The two structural isomers of dihydrohonokiol can be isolated from the reaction mixtures by procedures known in the art, such as preparative reverse phase high pressure liquid chromatography or preparative thin layer chromatography. Analogs and homologs of 3-n-propyl-5'-(2-propenyl)-1,1'-biphenyl-2',4-diol 20 (I) and 5'-n-propyl-3-(2-propenyl)-1,1'-biphenyl-2',4-diol (II) are synthesized from substituted 4-allyl or alkylphenyl alkyl ethers which are converted to the corresponding 2-alkoxy-5-allylphenyl metal halide or 2-alkoxy-5-alkylphenyl metal halide by a directed ortho metalation reaction. The products of such reactions are, in turn, reacted with various 4-alkoxyhaloaryls in-situ to generate the required homologs 25 or analogs of compounds I and II. Carboxylic groups may be protected as esters (e.g., ethyl ester) during the synthetic process. The dihydrohonokiol analog formed after the coupling reaction and demethylation can be deprotected by base catalyzed hydrolysis of the ester group. Alcoholic groups may be protected as acyl esters, e.g., by base catalyzed acetylation with acetic anhydride. The dihydrohonokiol analog formed after 30 the coupling and demethylation can be deprotected by base catalyzed removal of acyl groups.
WO 00/40532 PCT/US99/30168 8 3.0 DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS The anxiolytic compounds of the present invention, including dihydrohonokiol, its derivatives, analogs and homologs, in contrast to the 5 benzodiazepines, can be administered without significant side effects. The benzodiazepines have side effects including sedation, ataxia, amnesia, dependence, tolerance, and behavioral disturbances. These side effects have not been observed with therapeutic doses of the dihydrohonokiol isomers I and II. In tests in mice maximum anxiolytic activity of dihydrohonokiol was observed 10 three hours after oral administration. This contrasts with up to at least seven days for anxiolytic activity to manifest in mice treated with honokiol. The novel dihydrohonokiols shown below have been synthesized: )H H 2' 6 2' 6 1 l' 1. 43 6' 2 5 4 6' 20 5 3 OH 53 OH 15 The nomenclature adopted for the dihydrohonokiols, 3-n-propyl-5'-(2 propenyl)-1,1'-biphenyl-2',4-diol (I) and 5'-n-propyl-3-(2-propenyl)-1,1'-biphenyl 2',4-diol (II), is based on the original nomenclature for honokiol and tetrahydrohonokiol as used by Takeya et al, (1986). 20 The dihydrohonokiol compounds I and II can be synthesized by partial hydrogenation of honokiol as described in Example 1, § 5.1.2. This methods allows for the hydrogenation of only one of the two very similar allylic groups on the biphenyl moiety. In a typical reaction of the exemplified synthesis, 55 % of the reaction products was dihydrohonokiol, 35.5% unreacted honokiol, and 9.5% WO 00/40532 PCT/US99/30168 9 tetrahydrohonokiol. Thus dihydrohonkiol represents almost 86% of the consumed honokiol. This method allows for the recovery of unreacted honokiol, the reuse of which in the synthesis of the dihydrohonokiols isomers markedly increases the potential efficiency of this synthesis. 5 Limiting the supply of hydrogen was important in preventing conversion to tetrahydrohonkiol. Thus increasing the total amount of hydrogen in the reaction mixture, e.g., by increasing the volume of solvent and associated dissolved hydrogen, or increasing the time hydrogen is passed through the reaction mixture after the addition of honokiol, increases the amount of tetrahydrohonokiol produced. Preferred 10 reaction conditions were those which resulted in the reaction of 0.6 to 1.2 mol of hydrogen per mol of honokiol. Despite use of a variety of conditions, dihydrohonkiol could not be prepared by hydrogenation using either 5% or 10% palladium on activated carbon as catalyst. With palladium/carbon, either unreacted honokiol or fully reduced tetrahydrohonokiol was recovered from the reaction mixture. 15 Alternatively, compounds I and II can be synthesized by a total synthesis, details of which are given in Example 1, § 5.1.3. Briefly, 4-allyl or alkyl phenyl ethers are converted to the respective 2-alkoxy 5-allyl or 5-alkyl phenyl metal halides by a directed ortho metalation reaction. Such intermediates are reacted with 4-alkoxy haloaryls in the presence of a palladium catalyst to yield unsymetrical biphenyl 20 coupling products. The removal of alkyl protecting groups by boron tribromide under mild conditions, followed by preparative high pressure liquid chromatographic purification, produces purified compounds I and II. In like fashion, homologs and analogs of 3-n-propyl-5'-(2-propenyl)-1,1' biphenyl-2',4-diol (I) may be generated by converting 4-allylphenyl alkyl ethers to 25 2-alkoxy-5-allylphenyl metal halides by a directed ortho metalation reaction, which in turn will be reacted with various 4-alkoxyhaloaryls in-situ. Similarly, various substituted-phenyl alkyl ethers will be converted to 2-alkoxy-substituted phenyl metal halides and be reacted in-situ with various 3-allyl-4-alkoxyhalophenyls to generate the homologs and analogs of 5'-n-propyl-3-(2-propenyl)-1,1'-biphenyl-2',4-diol (II). 30 WO 00/40532 PCT/US99/30168 10 3.1 Anxiety As used herein, the term "anxiety" is intended to refer to a condition of apprehension, uncertainty, dread, or fear unattached to a clearly defined stimulus accompanied by numerous physiological and psychological symptoms such as 5 tachycardia, dyspnia, tension, restlessness, inattentiveness, and loss of appetite, skeletal motor function, initiative, cognitive logic, short- and long-term memory, and the like. Practice of the method of the present invention can combat, i.e., reduce or alleviate, some, most, or all of these physiological symptoms. A suitable subject to be treated by the present method is an animal, such as a 10 human or other mammal (e.g., house pets such as dogs and cats, or other commercially valuable or domestic animals), which experience anxiety-related symptoms due to some external or internal stimulus that are desirably combated. Preferably, the subject is human. As used herein the term "treating" includes prophylaxis of a physical and/or 15 mental condition or amelioration or elimination of the developed physical and/or mental condition once it has been established or alleviation of the characteristic symptoms of such condition. The term "antianxiety dose", as used herein, represents an amount of compound necessary to prevent or treat a human susceptible to or suffering from 20 anxiety following administration to such human. The active compounds are effective over a wide dosage range. For example, dosages per day will normally fall within the range of about 0.005 to about 500 mg/kg of body weight. In the treatment of adult humans, the range of about 0.05 to about 100 mg/kg, in single or divided doses, is preferred. However, it will be understood that the amount of the compound actually 25 administered will be determined by a physician, in the light of the relevant circumstances including the condition to be treated, the choice of compound to be administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the chosen route of administration, and therefore the above dosage ranges are not intended to limit the scope of the invention in any way. 30 While the present compounds are preferably administered orally to humans susceptible to or suffering from anxiety, the compounds may also be administered by a variety of other routes such as the transdermal, parenteraly, subcutaneous, intranasal, WO 00/40532 PCT/US99/30168 11 intramuscular and intravenous routes. Such formulations may be designed to provide delayed or controlled release using formulation techniques which are known in the art. Examples of anxiety disorders which may preferably be treated using an effective amount of a named compound or pharmaceutically acceptable salt thereof 5 include, but are not limited to: Panic Attack; Agoraphobia; Acute Stress Disorder; Specific Phobia; Panic Disorder; Psychoactive Substance Anxiety Disorder; Organic Anxiety Disorder; Obsessive-Compulsive Anxiety Disorder; Posttraumatic Stress Disorder; Generalized Anxiety Disorder; and Anxiety Disorder NOS. The named anxiety disorders have been characterized in the DSM-IV-R. 10 Diagnostic and Statistical Manual of Mental Disorders, Revised, 4th Ed. (1994). The DSM-IV-R was prepared by the Task Force on Nomenclature and Statistics of the American Psychiatric Association, and provides clear descriptions of diagnostic categories. The skilled artisan will recognize that there are alternative nomenclatures, nosologies, and classification systems for pathologic psychological conditions and that 15 these systems evolve with medical scientific progress. Social anxiety disorder may also be preferably treated using an effective amount of this compound or pharmacologically acceptable salt thereof. 3.2 Antidepressant Activity 20 The compounds of the present invention are also contemplated to be useful for the treatment of depression. There is considerable support the involvement of GABA in mood disorders. Somatic treatment for depression and mania upregulate the GABA B receptor similar to the effect of GABA agonists. Decreased GABA function accompanies depressed or manic mood states. Low GABA levels are found in brain, 25 cerebrospinal fluid and plasma in patients with depression and in plasma of patients with mania. Low GABA function is proposed to be an inherited biological marker of vulnerability for development of mood disorders. Benzodiazepines are GABA A receptor agonists and augment the activity of GABA while generally not affecting biogenic amine uptake or metabolism. A 30 metaanalysis of the use of benzodiazepines in the treatment of depression found that alprazolam had activity comparable to standard antidepressants.
WO 00/40532 PCT/US99/30168 12 The anxiolytic activity of the mixture of compound I and II and the benzodiazepine diazepam in the elevated plus maize model was blocked by the GABA antagonist bicuculline. But the anxiolytic activity of diazepam but not the Ill mixture was blocked by flumazenil, a benzodiazepine GABA site antagonist. While 5 these results differentiate the mechanism of action of compounds I and II from that of benzodiazepines, they also indicate a possible augmenting action of dihydrohonokiol compounds on the GABA system, an action indicative of antidepressant activity. 4.2 Pharmaceutical Compositions 10 Another aspect of the present invention includes novel compositions comprising the novel dihydrohonokiol, its derivatives, analogs and homologs. In a preferred embodiment the composition will comprise dihydrohonokiol, either 3-n propyl-5'-(2-propenyl)-1,1'-biphenyl-2',4-diol (I) or 5'-n-propyl-3-(2-propenyl)-1,1' biphenyl-2',4-diol (II). Alternatively, compositions may comprise any ratio of the two 15 isomers such as 10-90% of I and 90-10% of II. The composition may further comprise derivatives, analogs or homologs of dihydrohonokiol, or one or more other pharmacologically-active compounds, and particularly one or more anxiety-altering compounds. The methods of the invention may thus entail the administration of one, two, three, or more, of the new dihydrohonokiol, its derivatives, analogs and 20 homologs. The maximum number of species that may be administered is limited only by practical considerations, such as the particular effects of each compound. Of course, a composition of derivatives of I or II may be preferable, depending on their particular physical properties and physiological effects. The compositions of the invention may include a dihydrohonokiol, its 25 derivatives, analogs and homologs, modified to render it biologically protected. Biologically protected compounds have certain advantages over unprotected compounds when administered to human subjects and, may exhibit increased pharmacological activity. Compositions employing the novel compounds will contain a biologically 30 effective amount of the compounds. As used herein a "biologically effective amount" of a compound or composition refers to an amount effective to alter, modulate or reduce anxiety or related conditions. For oral administration, a satisfactory result may WO 00/40532 PCT/US99/30168 13 be obtained employing the compounds in an amount within the range of from about 0.001 mg/kg to about 50 mg/kg, preferably from about 0.005 mg/kg to about 35 mg/kg and more preferably from about 0.01 mg/kg to about 20 mg/kg alone or in combination with one or more additional anti-anxiety compounds in an amount within 5 the range from about 0.001 mg/kg to about 50 mg/kg, preferably from about 0.005 mg/kg to about 35 mg/kg and more preferably from about 0.01 mg/kg to about 20 mg/kg both being employed together in the same oral dosage form or in separate oral dosage forms taken at the same time. For parenteral administration, e.g., subcutaneously or intramuscularly, the dihydrohonokiol compound(s) will be 10 employed in an amount within the range of from about 0.005 mg/kg to about 10 mg/kg and preferably from about 0.01 mg/kg to about 1 mg/kg, alone or with the additional anti-anxiety compounds in an amount within the range of from about 0.005 mg/kg to about 20 mg/kg and preferably from about 0.01 mg/kg to about 2 mg/kg. It will also be understood that, if desired, the disclosed dihydrohonokiol, its 15 derivatives, analogs and homologs, may be administered in combination with additional agents, such as, e.g., proteins or polypeptides or various pharmaceutically active agents. So long as the composition comprises a dihydrohonokiol, its derivatives, analogs and homologs, there is virtually no limit to other components which may also be included, given that the additional agents do not cause a significant 20 adverse effect upon contact with the target cells or host tissues. The dihydrohonokiol, its derivatives, analogs and homologs, may thus be delivered along with various other agents as required in the particular instance. In carrying out the method of the present invention, the pharmaceutical compound(s) alone or in combination with one or more anxiety-modulating 25 compounds may be administered to mammalian species, such as monkeys, dogs, cats, rats and humans, and as such may be incorporated in a conventional systemic dosage form, such as a tablet, capsule, elixir or injectable. The above dosage forms will also include the necessary carrier material, excipient, lubricant, buffer, antibacterial, bulking agent (such as mannitol), anti-oxidants (ascorbic acid of sodium bisulfite) or 30 the like. Oral dosage forms are preferred, although parenteral forms such as intramuscular, intraperitoneal, or intravenous are quite satisfactory as well. The dose administered must be carefully adjusted according to age, weight and condition of the WO 00/40532 PCT/US99/30168 14 patient, as well as the route of administration, dosage form and regimen and the desired result. The pharmaceutical compositions disclosed herein may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or they may be 5 enclosed in hard or soft shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet. For oral therapeutic administration, the active compounds may be incorporated with excipients and used in the form of solutions, suspensions, elixirs, troches, tablets, pills, capsules, sustained release formulations, powders, syrups, wafers and the like, and contain about 0.1% to 10 about 95% of active ingredient, preferably about 1% to about 70%. Normally employed excipients may include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. Such compositions and preparations should contain at least 0.1% of active compound. The percentage of the compositions and preparations may, of course, be varied and 15 may conveniently be between about 2 to about 60% of the weight of the unit. The amount of active compounds in such therapeutically useful compositions is such that a suitable dosage will be obtained. The tablets, troches, pills, capsules and the like may also contain the following: a binder, as gum tragacanth, acacia, cornstarch, or gelatin; excipients, such 20 as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin may be added or a flavoring agent, such as peppermint, oil of wintergreen, or cherry flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. 25 Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar or both. A syrup of elixir may contain the active compounds sucrose as a sweetening agent methyl and propylparabens as preservatives, a dye and flavoring, such as cherry or orange flavor. Of course, any 30 material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active compounds may be incorporated into sustained-release preparation and formulations.
WO 00/40532 PCTIUS99/30168 15 Tablets of various sizes can be prepared, e.g., of about 30 to 900 mg in total weight, containing one or more of the active substances in the ranges described above, with the remainder being a physiologically acceptable carrier of other materials according to accepted pharmaceutical practice. These tablets can, of course, be scored 5 to provide for fractional doses. Gelatin capsules can be similarly formulated. Liquid formulations can also be prepared by dissolving or suspending one or the combination of active substances in a conventional liquid vehicle acceptable for pharmaceutical administration so as to provide the desired dosage in one to four teaspoonfuls. Such dosage forms can be administered to the patient on a regimen of one to four doses per 10 day. According to another modification, in order to more finely regulate the dosage schedule, the active substances may be administered separately in individual dosage units at the same time or carefully coordinated times. Since blood levels are built up and maintained by a regulated schedule of administration, the same result is achieved 15 by the simultaneous presence of the two substances. The respective substances can be individually formulated in separate unit dosage forms in a manner similar to that described above. In formulating the compositions, the active substances, in the amounts described above, are compounded according to accepted pharmaceutical practice with 20 a physiologically acceptable vehicle, carrier, excipient, binder, preservative, stabilizer, flavor, etc., in the particular type of unit dosage form. The active compounds may also be administered parenterally or intraperitoneally. Solutions of the active compounds as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a 25 surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. The pharmaceutical forms suitable for injectable use include sterile aqueous 30 solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the WO 00/40532 PCT/US99/30168 16 conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), 5 suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, 10 sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. Sterile injectable solutions are prepared by incorporating the active compounds 15 in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the 20 preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The dihydrohonokiol compound(s) described above may be administered in 25 the dosage forms as described above in single or divided doses of one to four times daily. It may be advisable to start a patient on a low dose combination and work up gradually to a high dose combination. The formulations as described above may be administered for a prolonged period, that is, for as long as the potential for onset of anxiety remains or the 30 symptoms of anxiety continue. Sustained release forms of such formulations which may provide such amounts biweekly, weekly, monthly and the like may also be WO 00/40532 PCTIUS99/30168 17 employed. A dosing period of at least one to two weeks are required to achieve minimal benefit. Formulations suitable for rectal administration are preferably presented as unit dose suppositories. These may be prepared by admixing the active compound with 5 one or more conventional solid carriers, for example, polyalkylene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient in the range of about 0.5% to about 10%, preferably about 1 to about 2%. Formulations suitable for transdermal administration may be presented as discrete patches adapted to remain in intimate contact with the epidermis of the 10 recipient for a prolonged period of time. Such patches suitably contain the active compound as an optionally buffered aqueous solution of, for example, 0.1 to 0.2M concentration with respect to the said active compound. Formulations suitable for transdermal administration may also be delivered by iontophoresis (see, for example, Pharmaceutical Research, 3(6):318, 1986) and 15 typically take the form of an optionally buffered aqueous solution of the active compound. Suitable formulations comprise citrate or bis tris buffer (pH 6) or ethanol/water and contain from 0.1 to 0.2M active ingredient. The compositions are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically effective. The quantity to be 20 administered depends on the subject to be treated, including, e.g., age, physical condition and degree of symptoms presented. Precise amounts of active ingredient required to be administered depend on the judgment of the practitioner. However, suitable dosage ranges are of the order of several hundred micrograms active ingredient per dose. Suitable regimes for initial administration and booster doses are also variable, 25 but are typified by an initial administration followed by subsequent administrations. Clinical doses will of course be determined by the nutritional status, age, weight and health of the patient. The quantity and volume of the composition administered will depend on the subject and the route of administration. The precise amounts of active compound required will depend on the judgment of the practitioner 30 and may be peculiar to each individual. However, in light of the data presented herein, the determination of a suitable dosage range for use in humans will be straightforward.
WO 00/40532 PCT/US99/30168 18 As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any 5 conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. 4.2.1 Liposomes and Nanocapsules 10 In certain embodiments, the inventors contemplate the use of liposomes and/or nanocapsules for the introduction of one or more of the disclosed pharmaceutical composition into a host cell. Such formulations may be preferred for the introduction of pharmaceutically-acceptable formulations of the honokiol derivatives and/or analogs disclosed herein. 15 The formation and use of liposomes is generally known to those of skill in the art (see for example, Couvreur et al., 1977 which describes the use of liposomes and nanocapsules in the targeted antibiotic therapy of intracellular bacterial infections and diseases). More recently, liposomes were developed with improved serum stability and circulation half-times (Gabizon and Papahadjopoulos, 1988; Allen and Choun, 20 1987). In one instance, the disclosed composition may be entrapped in a liposome. Liposomes are vesicular structures characterized by a phospholipid bilayer membrane and an inner aqueous medium. Multilamellar liposomes have multiple lipid layers separated by aqueous medium. The term "liposome" is intended to mean a 25 composition arising spontaneously when phospholipids are suspended in an excess of aqueous solution. The lipid components undergo self-rearrangement before the formation of closed structures and entrap water and dissolved solutes between the lipid bilayers (Ghosh and Bachhawat, 1991). Nanocapsules can generally entrap compounds in a stable and reproducible 30 way (Henry-Michelland et al., 1987). To avoid side effects due to intracellular polymeric overloading, such ultrafine particles (sized around 0.1 tm) should be designed using polymers able to be degraded in vivo. Biodegradable polyalkyl-cyano- WO 00/40532 PCT/US99/30168 19 acrylate nanoparticles that meet these requirements are contemplated for use in the present invention, and such particles may be are easily made, as described (Couvreur et al., 1977; 1988). Methods of preparing polyalkyl-cyano-acrylate nanoparticles containing biologically active substances and their use are described in U.S. Patent 5 4,329,332, U.S. Patent 4,489,055, and U.S. Patent 4,913,908. Pharmaceutical compositions containing nanocapsules for the oral delivery of active agents are described in U.S. Patent 5,500,224 and U.S. Patent 5,620,708. U.S. Patent 5,500,224 describes a pharmaceutical composition in the form of a colloidal suspension of nanocapsules comprising an oily phase consisting essentially 10 of an oil containing dissolved therein a surfactant and suspended therein a plurality of nanocapsules having a diameter of less than 500 nanometers. U.S. Patent 5,620,708 describes compositions and methods for the oral administration of drugs and other active agents. The compositions comprise an active agent carrier particle attached to a binding moiety which binds specifically to a target molecule present on the surface of 15 a mammalian enterocyte. The binding moiety binds to the target molecule with a binding affinity or avidity sufficient to initiate endocytosis or phagocytosis of the particulate active agent carrier so that the carrier will be absorbed by the enterocyte. The active agent will then be released from the carrier to the host's systemic circulation. In this way, degradation of the disclosed pharmaceutical compounds in 20 the intestines can be avoided while absorption of compound form the intestinal tract is increased. An excellent review of nanoparticles and nanocapsular carriers is provided by Arshady 1996. Arshady notes that one of the major obstacles to the targeted delivery of colloidal carriers, or nanocapsules, is the body's own defense mechanism in 25 capturing foreign particles by the reticuloendothelial system (RES). This means that following intravenous administration, practically all nanometer size particles are captured by the RES (mainly the liver). The review describes recent initiatives on the design of macromolecular homing devices which seem to disguise nanoparticles from the RES and, hence, are of potential interest to the targeted delivery of nanocapsular 30 carriers. The idea is based on a graft copolymer model embodying a link site for attachment to the carrier, a floating pad for maintaining the particles afloat in the WO 00/40532 PCT/US99/30168 20 blood stream, an affinity ligand for site-specific delivery and a structural tune for balancing the overall structure of the homing device. Yu and Chang, 1996 describe the use of nanocapsules containing hemoglobin as potential blood substitutes. They use different polymers including polylactic acid 5 and polyisobutyl-cyanoacrylate and modify the surface of the nanocapsules with polyethylene glycol (PEG) or with PEG 2000 PE. The surface modified nanocapsules containing hemoglobin survive longer in the circulation. U.S. Patent 5,451,410 describes the use of modified amino acid for the encapsulation of active agents. Modified amino acids and methods for the preparation 10 and used as oral delivery systems for pharmaceutical agents are described. The modified amino acids are preparable by reacting single amino acids or mixtures of two or more kinds of amino acids with an amino modifying agent such as benzene sulfonyl chloride, benzoyl chloride, and hippuryl chloride. The modified amino acids form encapsulating microspheres in the presence of the active agent under sphere-forming 15 conditions. Alternatively, the modified amino acids may be used as a carrier by simply mixing the amino acids with the active agent. The modified amino acids are particularly useful in delivering peptides or other agents which are sensitive to the denaturing conditions of the gastrointestinal tract. 20 4.3 Diagnostic and Therapeutic Kits Therapeutic kits comprising dihydrohonokiol, its derivatives, analogs and homologs, comprise another aspect of the present invention. Such kits will generally contain, in suitable container means, a therapeutically-effective amount of a pharmaceutically acceptable composition of dihydrohonokiol, its derivatives, analogs 25 or homologs, and a pharmaceutically acceptable excipient. The diagnostic/therapeutic kits comprising the pharmaceutical compositions disclosed herein will generally contain, in suitable container means, a therapeutically-effective amount of dihydrohonokiol, its derivatives, analogs and homologs, in a pharmaceutically acceptable excipient. The kit may have a single container means that contains the 30 dihydrohonokiol, its derivatives, analogs and homologs, and a suitable excipient or it may have distinct container means for each compound.
WO 00/40532 PCT/US99/30168 21 The components of the kit may be provided as liquid solution(s), or as dried powder(s). When the components are provided in a liquid solution, the liquid solution is an aqueous solution, with a sterile aqueous solution being particularly preferred. When reagents or components are provided as a dry powder, the powder can be 5 reconstituted by the addition of a suitable solvent. It is envisioned that the solvent may also be provided in another container means. When the components of the kit are provided in one or more liquid solutions, the liquid solution is an aqueous solution, with a sterile aqueous solution being particularly preferred. The dihydrohonokiol, its derivatives, analogs and homologs, 10 may also be formulated into a syringeable composition. In which case, the container means may itself be a syringe, or other such like apparatus, from which the formulation may be administered into the body, preferably by injection or even mixed with the other components of the kit prior to injection. The dihydrohonokiol, its derivatives, analogs and homologs, to be administered may be a single compound, or 15 a composition comprising two or more of dihydrohonokiol, its derivatives, analogs and homologs, in a single or multiple dose for administration. Alternatively, one or more dihydrohonokiol, its derivatives, analogs and homologs, may be administered consecutively or concurrently with other agents as deemed appropriate by the clinician. Dosage of each of the compositions will vary from subject to subject 20 depending upon severity of conditions, size, body weight, etc. The calculation and adjustment of dosages of pharmaceutical compositions is well-known to those of skill in the art. In an alternate embodiment, components of the kit may be provided as dried powder(s). When reagents or components are provided as a dry powder, the powder 25 can be reconstituted by the addition of a suitable solvent. It is envisioned that the solvent may also be provided in another container means. The container means will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which the dihydrohonokiol, its derivatives, analogs and homologs, may be placed, preferably, suitably allocated. 30 Where two or more of dihydrohonokiol, its derivatives, analogs and homologs, are provided, the kit will also generally contain a second vial or other container into which this additional compound may be formulated. The kits may also comprise a WO 00/40532 PCTIUS99/30168 22 second/third container means for containing a sterile, pharmaceutically acceptable buffer or other diluent. The kits of the present invention will also typically include a means for containing the vials in close confinement for commercial sale, such as, e.g., injection 5 or blow-molded plastic containers into which the desired vials are retained. Alternatively, the vials may be prepared in such a way as to permit direct introduction of the composition into an intravenous drug delivery system. Irrespective of the number or type of containers, the kits of the invention may also comprise, or be packaged with, an instrument for assisting with the 10 injection/administration or placement of the ultimate dihydrohonokiol, its derivatives, analogs and homologs, composition within the body of an animal. Such an instrument may be a syringe, pipette, forceps, measured spoon, eye dropper or any such medically approved delivery vehicle. 15 5.0 EXAMPLES 5.1 Example 1. Synthesis of Dihydrohonokiols 5.1.1 Materials and Methods: Materials 4-Allyl anisole, t-butyllithium (1.7 M solution in anhydrous pentane), 20 anhydrous tetrahydrofuran, anhydrous dichloromethane, anhydrous zinc chloride (1.0 M solution in anhydrous diethyl ether), bis(triphenylphosphine) palladium(II)chloride, diisobutylaluminum hydride (1.0 M solution in hexane), hexane, tris-(tiphenylphosphine) rhodium(I) chloride, toluene, deuterated chloroform, tetramethylsilane and florisil were purchased from Aldrich Chemical Co. (Milwaukee, 25 Wisconsin). Honokiol was obtained from Wako Pure Chemical Industries Ltd. (Osaka, Japan). Alternatively, honokiol may be extracted by supercritical carbon dioxide extraction from suitable portions of Magnolia trees (Chandra and Nair, 1995). Tetrahydrohonokiol was prepared by the hydrogenation in the presence of 5% 30 Palladium on active carbon catalyst as described by Fujita et al. (1972).
WO 00/40532 PCT/US99/30168 23 2-Propyl-4-iodo anisole was obtained from Rann Research Laboratory (San Antonio, Texas). Ammonium chloride, sodium chloride, anhydrous diethyl ether and anhydrous sodium sulfate were purchased from Fisher Scientific (Fair Lawn, New Jersey). 5 HPLC grade Ethyl acetate, acetone, water and acetonitrile were obtained from Burdick and Jackson Inc. (Muskegon, Michigan). Analytical and preparative TLC plates (20 x 20 cm glass plates coated with 250 micron and 1 mm thickness of silica gel (G) were purchased from Analtech Inc. (Newark, Delaware). 10 HPLC The analytical and preparative high pressure liquid chromatography were performed on a Milton Roy HPLC equipment consisting of a CM 4000 solvent delivery system, the autoinjector A1000 and the spectro-monitor 3100 variable 15 wavelength detector from LCD Analytical (Riviera Beach, Florida). A reverse phase HPLC column (Econosil C18, 10p.i, 250 x 10 mm, Catalog #C231) was purchased from Alltech Associates, Inc. (Deerfield, Illinois). The solvent for elution consisted of a mixture of acetonitrile and water (6:4). The elution of the honokiol and its analogs was detected at a wavelength of 280 nm. The chart speed was 0.5 cm/min, solvent 20 flow rate was 5 ml/min and the detection was performed at the sensitivity of 0.05 and 2.0 absorbance units full scale (for analytical and preparative use, respectively). The proton magnetic resonance spectra were recorded on a 300 MHz GE QE 300 NMR spectrometer. The samples were dissolved in deuterated chloroform and tetramethylsilane was used as the internal standard. 25 5.1.2 Synthesis of 3-n-Propyl-5'-(2-propenyl)-1,1'-biphenyl-2',4-dio (I) and 5' n-Propyl-3-(2-propenyl)-1,1'-biphenyl-2',4-diol (II) by the Partial Hydrogenation of Honokiol: Hydrogen gas was passed for two hours with stirring in a solution of 30 tris-(triphenylphosphine) rhodium(I) chloride (10 mg) in toluene (4 ml) at room temperature. Then, honokiol (20 mg) dissolved in toluene (1 ml) was added slowly into the reaction flask with stirring and the hydrogen gas was passed for another 10 WO 00/40532 PCT/US99/30168 24 min after the complete addition of honokiol solution. The reaction mixture was stirred overnight at room temperature. The reaction mixture was passed through a column of florisil (5 gm), which was finally washed with 50 ml of dry diethyl ether. The solvents were evaporated to obtain crude residue which was resuspended in 5 aforesaid HPLC eluant to be injected in the HPLC system for analysis. The HPLC analysis indicated the presence of three peaks: (a) peak 1: Retention time- 9 min, honokiol. (b) peak 2 (86.4% based on honokiol consumed): Retention time- 11 min. (c) peak 3 (13.6% based on honokiol consumed): Retention time- 14.2 10 min, m/z 270, confirmed to be tetrahydrohonokiol when compared with standard tetrahydrohonokiol. The compounds eluting as peak 2 were identified as the two isomers of dihydrohonokiol. The first component of the peak (up to the begining of the shoulder) was 92% of the total peak, [m/z 268; 1 H NMR(CDCl 3 ): 6: 0.994 (t,3H), 1.663 15 (dt,3H), 2.629 (t,2H), 3.347 (d,2H), 5.03-5.11 (m,2H), 5.90-6.04 (m,1H), 6.88-7.20 (in, 6H) with a small shoulder (approximately 8% of total peak 2) ['H NMR(CDC1 3 ): 8: 0.994 (t,3H), 1.663 (dt,3H), 2.55(t,2H), 3.45(d,2H), 5.13(m,2H), 5.90-6.04 (m,1H), 6.88-7.20 (m,6H). The dihydrohonokiol isomers in peak 2 were isolated by preparative HPLC purification. The major component (approximately 92%) was 20 assigned the structure as 3-n-propyl-5'-(2-propenyl)-1,1'-biphenyl-2',4-diol (I), while the minor component (approximately 8%). was assigned the structure as 5'-n-propyl-3-(2-propenyl)-1,1'-biphenyl-2',4-diol, (II). The HPLC retention times for the purified isomers was 11 min for isomer I and 12 min for isomer II. A typical reaction resulted in a product mixture consisting of 55% dihydrohonokiols-, 35.5% 25 unreacted honokiol and 9.5% tetrahydrohonokiol. The dihydrohonokiol isomers represented approximately 86% of the dihydrohonokiol consumed in the reaction.
WO 00/40532 PCT/US99/30168 25 H H H 00 H2 0 OH (Ph 3
P)
3 RhCl OH OH Honokiol Synthesis of 3-n-propyl-5'-(2-propenyl)-1,1'-biphenyl-2',4-diol (I) and 5 5'-n-propyl-3-(2-propenyl)-1,l'-biphenyl-2',4-diol (II) from Honokiol by Partial Hydrogenation 5.1.3 Total Synthesis of 3-n-Propyl-5'-(2-propenyl)-1,1'-biphenyl-2',4-diol (I) and 5'-n-Propyl-3-(2-propenyl)-1,1'biphenyl-2',4-diol (II) 10 5.1.3.1 3-n-Propyl-5'-(2-propenyl)-1,1'-biphenyl-2',4-diol (I): To a solution of 4-allylanisole (148 mg, 1.0 mmol) in 1 ml anhydrous tetrahydrofuran was added a solution of tert-butyllithium in pentane (1.5 mmol; as 0.88 ml of 1.7 M solution in anhydrous pentane) with stirring under the nitrogen atmosphere at -78'C. After two hours, the mixture was warmed to -10'C and the 15 solution was reacted with anhydrous zinc chloride (1 ml, 1.0 mmol; as 1.0 M solution in anhydrous diethyl ether). Then, the reaction mixture was stirred at room temperature for 1 h. The palladium catalyst was prepared separately. Thus, an anhydrous hexane solution of diisolbutyl aluminum hydride (0.066 ml, 0.066 mmol; 1.0 M solution in hexane) was added to a solution of 20 bis-(triphenylphosphine)palladium(H) chloride (22 mg; 0.033 mmol) in 1 ml anhydrous tetrahydrofuran with stirring under nitrogen atmosphere. This organic catalyst solution was then reacted with 2-propyl-4-iodoanisole (193 mg, 0.70 mmol) in 2 ml of anhydrous tetrahydrofuran and a solution of the aryl zinc chloride, prepared as above. The mixture was stirred for 2 h at room temperature and then quenched 25 with 5 ml saturated ammonium chloride solution. The aqueous layer was extracted with three 10 ml portions of ethyl acetate. The combined extract was washed once with brine and dried over anhydrous sodium sulphate. After the removal of solvent in vacuo, the crude product was chromatographed on a 1 mm thickness 20 x 20 cm silica WO 00/40532 PCT/US99/30168 26 gel plate (silica gel G; eluent: hexane, ethyl acetate; 9:1) to get 2',4-dimethoxy 3-n-propyl-5'-(2-propenyl)-1,1'-biphenyl (95 mg, 45.9%; based on the amount of 2-propyl-4-iodoanisole): m/z: 296; 'H NMR (CDCl 3 ): 8: 0.90 (t, 3H, 3-CH 2
.CH
2 .CH), 1.45 (qt,3H, 3-CH 2
.CH
2
.CH
3 ), 2.53 (t,2H, 3-CH 2
.CH
2
.CH
3 ), 3.38 5 (d,2H, 5'-CH 2
.HC=CH
2 ), 3.71 (s, 3H, 4-OCH), 3.77 (s, 3H, 2'-OCH 3 ), 4.99 (dd,2H, 5'-CH 2
.HC=CH
2 ), 5.85-5.98 (m, 1H, 5'-CH 2
.HC=CH
2 ), 6.79-7.28 (m, 6H, aromatic H). To a solution of 2',4-dimethoxy-3-n-propyl-5'-(2-propenyl)-1,1'-biphenyl (10 mg: 0.03 mmol) in 1 ml of anhydrous dichloromethane was slowly added a 10 dichloromethane solution of boron tribromide (75 microliters, 1M solution in dichlormethane) at -78'C with magnetic stirring for one h. Then, the reaction mixture was stirred at room temperature overnight. Next day, the reaction was cooled and subjected to an aqueous quenching. The aqueous phase was extracted three times with 2 ml portions of dichloromethane. The combined organic layer was washed once 15 with brine (1 ml) and dried over anhydrous sodium sulfate. The solid was filtered off and the solvent was evaporated under nitrogen. The crude product was chromatographed on a 250 micro thickness 20 x 20 cm silica gel plate (silica gel G; eluent: hexane, ethylacetate, 80:20) to get 3-n propyl-5'-(2-propenyl)-1,1'-biphenyl-2',4-diol (6.5 mg; 72%). In a HPLC analysis, the 20 retention time of total synthetic 3-n-propyl-5'-(2-propenyl)-1,1'- biphenyl-2',4-diol was comparable to that of the hemi-synthetic sample made earlier; and in a mixed co-injection both co-eluted together. The structure was further confirmed by the proton magnetic resonance analysis on a HPLC pure sample. 1 H NMR (CDCl 3 ): 8: 0.994 (t, 3H, 3-CH 2
.CH
2
.CH
3 ), 1.663 (qt,3H, 3-CH 2
.CH
2
.CH
3 ), 2.629 (t,2H, 25 3-CH 2
.CH
2 .CH3), 3.347 (d,2H, 5'-CH 2
.HC=CH
2 ), 5.03-5.11 (dd,2H, 5'CH 2
.HC=CH
2 ), 5.90-6.04 (m,1H, 5'-CH 2
.HC=CH
2 ), 6.88-7.20 (m, 6H, aromatic H). 5.1.3.2 5'-n-Propyl-3-(2-propenyl)-1,1'biphenyl-2',4-dio (II): The same method as described above in § 5.1.3.1 for the synthesis 30 2',4-dimethoxy-3-n-propyl-5'-(2-propenyl)-1,1'-biphenyl can be used to synthesize 2'4-dimethoxy-5'-n-propyl-3-(2-propenyl)- 1,1 '-biphenyl. Thus, 4-propylanisole is WO 00/40532 PCT/US99/30168 27 reacted with tert-butyllithium and anhydrous zinc chloride to form an aryl zinc chloride which is then coupled with 2-allyl-4-iodo-anisole, in the presence of the palladium catalyst made in a separate reaction of bis (triphenylphosphine)palladium(II) chloride and diisolbutyl aluminum hydride. The 5 product, 2'4-dimethoxy-5'-n-propyl-3-(2-propenyl)-1,1'-biphenyl, is purified by extraction and thin layer chromatography as described above. The methyl protecting groups are removed from 2'4-dimethoxy-5'-n-propyl-3 (2-propenyl)-1,1'-biphenyl as described above in § 5.1.3.1 for 3-n-propyl-5'-(2-propenyl)- 1,1'- biphenyl-2',4-diol. The product, 10 5'-n-propyl-3-(2-propenyl)-1,1'biphenyl-2',4-diol (II), is purified by extraction and thin layer chromatography, as described above.
WO 00/40532 PCT/US99/30168 28
OCH
3
OCH
3
OCH
3 OCH 3 ZnCl t.bu.Li 0 01 0
OCH
3 (Ph 3
P)
2 PdCI 2 DIBALH BBr 3 H OH
OCH
3
OCH
3
OCH
3 OCH 3 ZnC1 t.bu.Li 0nC0200
OCH
3 (Ph 3
P)
2 PdCl 2 DIBALH BBr 3 H OH Total synthesis of 3-n-propyl-5'-(2-propenyl)-1,1'-biphenyl-2',4-diol (I) and 5' 5 n-propyl-3-(2-propenyl)-1,1'-biphenyl-2',4-diol (II) 5.2 Example 2. Synthesis of Analogs and Homologs of Dihydrohonokiol For the synthesis of analogs and homologs of dihydrohonokiol, 4-allylphenyl alkyl ethers can be converted to 2-alkoxy, 5-allylphenyl metal halides by a directed WO 00/40532 PCT/US99/30168 29 ortho metalation reaction, which in turn is reacted with various 4-alkoxyhaloaryls in situ to generate the homologs and analogs of 3-n-propyl, 5'-(2-propenyl)-1,1' biphenyl-2',4-diol. Similarly, various substituted-phenyl metal halides are reacted in situ with various 5'-n-propyl, 3-(2-propenyl)- 1,1 '-biphenyl-2',4-diol. 5 Carboxylic groups may be protected as esters (e.g., ethyl ester) during the synthetic process. The dihydrohonokiol analog formed after the coupling reaction and demethylation can be deprotected by based catalyzed hydrolysis of the ester group. Thus, for example, for the synthesis of 3-n-propyl-5'-(3-carboxy propyl)-1,1' biphenyl-2',4-diol, the starting compound 3-(4-methoxyphenyl)-propionic acid, 10 available from Aldrich (Milwaukee, WI) can be converted to its acid chloride and then to the ethyl ester, i.e. 3-(4-methoxyphenyl)-propionate. This compound may be treated with tert-butyllithium followed by anhydrous zinc chloride to form the intermediate 2-methoxy, 5-(3-ethoxy carbonyl propyl)-phenyl zinc chloride. This intermediate can be coupled with 2-n-propyl-4-iodo anisole in the presence of the 15 palladium catalyst (prepared in situ by the reaction of bis-(triphenyl phosphine) palladium (II) chloride and diisobutyl aluminum hydride) to synthesize 2',4 dimethoxy-3-n-propyl-5'-(3-ethoxycarbonyl propyl)-1,1'-biphenyl. The demethylation of this compound with borontribromide will give 3-n-propyl-5'-(3-ethoxycarbonyl propyl)-1,1'-biphenyl-2',4-diol. Finally, the base catalyzed hydrolysis of the ester 20 group, e.g., with potassium hydroxide, will generate 3-n-propyl-5'-(3-carboxy propyl) 1,1 '-biphenyl-2',4-diol. Alcoholic groups may be protected as acyl esters, e.g., by base catalyzed acetylation with acetic anhydride. The dihydrohonokiol analog formed after the coupling and demethylation can be deprotected by base catalyzed removal of acyl 25 groups. Thus, for example, for the synthesis of 3-n-propyl-5'-(2-hydroxy propyl)-1,1' biphenyl-2',4-diol, the intermediate 1-(4-methoxyphenyl)-2-propanol is made by sodium borohydride reduction of 4-methoxyphenyl acetone, available from Aldrich (Milwaukee, WI). The 1-(4-methoxyphenyl)-2-propanol can be reacted with acetic anhydride in basic medium to get 1-(4-methoxyphenyl)-2-propyl acetate. This 30 compound may be treated with tert-butyllithium followed by anhydrous zinc chloride to get 2-methoxy, 5-(2-acetoxypropyl)-phenyl zinc chloride as an intermediate. This WO 00/40532 PCT/US99/30168 30 intermediate can be coupled with 2-n-propyl-4-iodo anisole in the presence of the palladium catalyst (prepared in situ by the reaction of bis-(triphenyl phosphine) palladium (II) chloride and diisobutyl aluminum hydride) to synthesize 2',4 dimethoxy-3-n-propyl-5'-(2-acetoxypropyl)-1,1'-biphenyl. The demethylation of this 5 compound with borontribromide will yield 2',4-dimethoxy-3-n-propyl-5'-(2 acetoxypropyl)-1,1'-biphenyl-2',4-diol. Finally the base catalyzed hydrolysis of the acetyl group will generate 3-n-propyl-5-(2-hydroxy propyl)-1,1-biphenyl-2,4-diol. 5.3 Example 3. Anxiolytic Effect of a Mixture of Dihydrohonokiol Isomers in 10 the Elevated Plus-Maze Test in Mice 5.3.1 Materials and Methods 5.3.1.1 Animals Male mice of the Balb/C strain (Harlan, Indianapolis, IN) were used at about 6 wk of age and weighing of 22-27 g. Groups of 5 mice each were housed in standard 15 polycarbonate cages (15W x 25Lx12H cm) with woodchip bedding with free access to a standard solid diet and tap water. The environment of the animal room was well controlled with a temperature of 23±1'C, humidity 40-55% and a 12:12-h light-dark cycle; lights on between 0600-1800h). All the experimental procedures were carried out according to "The Guide for 20 the Care and Use of Laboratory Animals and Animal Welfare Act, and the experimental protocol (97031-34-01-A) was approved by the Institutional Animal Care and Use Committee of the University of Texas Health Science Center at San Antonio. 25 5.3.1.2 Drugs For oral administration, the mixture of dihydrohonokiol isomers I and II prepared by the partial hydrogenation of honokiol (designated 111 mixture) as described in § 5.1.2, was first dissolved in a very small amount of ethanol, and the solution was diluted with physiological saline containing Tween-80 (0.1%) (vehicle). 30 Diazepam was suspended in the Tween-80/physiological saline. The concentration of WO 00/40532 PCT/US99/30168 31 each drug solution or suspension was adjusted so that each volume administered was constant at 0.1 ml/10 g body weight of the mouse. 5.3.1.3 Apparatus and Experimental Procedures 5 The elevated plus-maze test: The elevated plus-maze used was the same as that used in previous studies (Kuribara et al., 1996; Maruyama et al., 1997), and was an improvement of the original apparatus for rats (Pellow et al., 1985) and for mice (Lister, 1987). Briefly, the plus-maze consisted of two closed-arms and two open-arms (6W x 30L cm). The arms extended from a center platform (8 x 8 cm). 10 The closed arms had side-walls (10H cm), and they were made of non-transparent polyvinyl chloride of gray color. The open arms were made of transparent acrylic fiber, and had no side-walls. The platform was made of non-transparent polyvinyl chloride of gray color. This plus-maze was set 40 cm above the base. Each mouse was placed at the center platform facing one of the closed arms, and the cumulative 15 time of entering into the open arms during the 5 min observation period was recorded. The criterion of the mouse's entering into the open arms was crossing with all four paws the borderline separating the open arm and the center platform. Activity test: Mouse activity was measured with a tilting-type ambulometer which had a bucket-like Plexiglas activity cage 20 cm in diameter (SMA-1: O'Hara 20 and Co., Tokyo). Only a horizontal movement (ambulation) of the mouse caused a slight tilt of the activity cage, and was detected by microswitches attached to the cage. The duration of measurement was 5 min for each mouse. This activity test was carried out immediately after the end of the plus-maze test. Traction test: The traction test was carried out to assess muscle strength. This 25 experimental procedure was described elsewhere (Kuribara et al., 1977). Briefly, a wire (1.6 mm in diameter, and 30 cm in length) was set horizontally 30 cm above the base. The mouse was first allowed to grip the wire with the fore paws, and the duration of clinging to the wire was measured up to 60 sec. The trial was held twice for each mouse, and the longer duration of clinging was used in the calculation of 30 mean value. When the duration of clinging was over 60 sec, the mouse was released from the wire, and the clinging time was recorded as 60 sec. The traction test was carried out immediately after the end of the activity test.
WO 00/40532 PCT/US99/30168 32 5.3.1.4 Statistical Analysis The time in open arms in the elevated plus-maze test, the activity counts in the activity test, and the durations of clinging in the traction test were compared by the 5 Fisher's protected least significant difference statistical test. Values of P less than 0.05 were considered significant. 5.3.2 Dose Response of Dihydrohonokiol Isomers I/I Mixture 10 TABLE 1 Dose Effect in the Mouse of I/II Mixture Assessed Using the Elevated Plus-Maze and Motor Activity Treatment, P.O. Time (sec) spent in open arm Motor Activity mg/kg 1 h post 3 h post adm. 1 h post 3 h post adm. adm. adm. Vehicle 13.2 ±2.7 13.1 ± 4.7 30.6± 1.7 31.2± 1.5 I/iI 0.1 26.0± 8.4 30.5± 1.5 1111 0.2 27.6 ±6.2 37.9± 6.2* 36.1 ±2.3 21.9± 3.3 I/II 0.5 33.8 ± 4.9 87.3 ± 12.8* 32.7 ± 2.7 26.0 ± 1.8 I/Il 1.0 53.8 ± 7.7 90.0 ± 9.2* 32.0 ± 1.7 30.8 ± 1.5 1111 2.0 50.2 ± 5.71 95.3 ± 8.7* 34.6 ± 2.0 27.0 ± 1.7 The data for the plus-maze are mean times ± SEM spent in the open arm 15 during the observation period of 5 min. The data for the activity test are mean counts ± SEM during the observation period of 5 min. The group size was 10. *p<0.00 2 . Increasing doses of the Ill isomer mixture cause an increasing anxiolytic effect- a dose/effect relationship. A 0.2 mg/kg oral dose of the I/ll mixture shows significant anxiolytic effects at 3 h post administration, while a higher dose of 1 mg/kg is 20 significantly anxiolytic at only one hour. Since the inventors used the 0.2 mg/kg dose orally in this series the tests were performed 3 h after dosing of the Ill mixture.
WO 00/40532 PCT/US99/30168 33 Motor activity was unchanged by dose or time except for the 0.2 mg/kg dose at 3 h post administration. 5.3.3 Duration of III Mixture Induced Anxiolytic Activity 5 TABLE 2 Duration of Pharmacological Action Following 0.5 mg/kg I/II Mixture Given Orally Treatment Hours post treatment Time spent in Motor activity mglkg open arm, sec Vehicle 0 12.9 ±3.8 26.9 ±2.1 e I/II 0.5 1/2 15.5 ±3.7 28.6 ±1.7 1/11 0.5 1 33.8 ± 4.9 32.7 ± 2.7 1/1I 0.5 2 43.0 ±7.5* 25.8 ±1.1 I/II 0.5 3 87.3 ± 12.8* 26.0 ± 1.8 I/II 0.5 4 77.7 ± 13.2* 26.9 ± 2.0 111 0.5 6 37.4± 11.0 22.3 ±1.8 I/II 0.5 8 46.2± 9.1* 27.5 ±2.1 I/I 0.5 12 47.0± 13.1* 24.0± 2.0 1/11 0.5 24 14.0± 5.0 17.3 ±3.1 I/II 0.5 48 11.5 ± 4.5 27.3 ± 1.5 10 The data for the plus-maze test are mean times ± SEM spent in the open arm during the observation period of 5 min. The data for the activity test are the mean counts ± SEM during the test period of 5 min. The group size was 10 mice. *p<0.02 vs control vehicle. The duration of anxiolytic activity of the I/II mixture following an oral dose of 15 0.5 mg/kg extends for at least 12 h post administration, ending between 12 h and 24 h after oral administration. The anxiolytic activity was not measured between 12 and 24 h. This long duration of action indicates the possibility of a one single dose per WO 00/40532 PCT/US99/30168 34 day schedule. Single dosage greatly improves patient compliance. The motor activity was unchanged. 5.3.4 Duration of Diazepam Anxiolytic Activity 5 TABLE 3 Duration of Pharmacological Action Following 1.0 mg/kg Diazepam Given Orally Treatment, Hours post Time spent in Motor Traction dose in mg/kg treatment open arm, sec. activity Vehicle 0 12.3 ± 2.6 24.9 ± 1.7 60.0 ± 0.0 Diazepam 1.0 0.17 43.5 ± 6.1* 38.9 ± 3.2* 42.6 ± 6.3* Diazepam 1.0 0.5 41.2 ± 5.2* 23.2 ± 3.8 53.3 ± 3.7 Diazepam 1.0 1 29.7 ± 6.9* 32.1 ± 2.3 60.0 ± 0.0 Diazepam 1.0 2 27.0 ± 8.0 32.2 ± 4.1 60.0 ± 0.0 Diazepam 1.0 4 21.7 ± 6.3 31.6 ± 3.1 60.0 ± 0.0 Diazepam 1.0 8 13.2 ± 5.9 27.0 ± 2.4 60.0 ± 0.0 Diazepam 1.0 24 12.2 ± 2.7 29.5 ± 3.4 60.0 ± 0.0 10 The data for the plus-maze test are mean times SEM spent in the open arm during the observation period of 5 min. The data for the activity test are the mean counts ± SEM during the test period of 5 min. The group size was 10 mice. *p<0.04 vs control. The benzodiazepine compound diazepam was used as a positive control for 15 anxiolytic activity in this series of tests because of wide use and its acceptance as representative of the benzodiazepine series of compounds. The time of peak anxiolytic action of diazepam is 0.17 h (10 min.). The anxiolytic tests in the subsequent series were done on diazepam 10 min after oral dosing to obtain the peak response. The duration of diazepam following the effective dose of 1 mg/kg orally is 20 shown to be 1 h for its anxiolytic activity and 0.17 h for its effect on motor activity and its detrimental affect on traction (ataxia production).
WO 00/40532 PCTIUS99/30168 35 5.3.5 Effect of Multiple Doses of I/II Mixture and Diazepam TABLE 4 5 Effect of seven days treatment with 0.2 mg/kg po 1I Mixture and 1.0 mg/kg po diazepam Treatment Time in open arm (sec) Motor activity Traction vehicle 15.6±5.0 25.9±3.8 60.0±0.0 I/II 44.9±7.7* 31.9±4.1 60.0±0.0 vehicle 12.3±2.6 24.9±1.7 60.0±0.0 Diazepam 43.5±6.1* 38.9±3.2* 42.6±6.2* *p<0.016 vs vehicle control The data for the plus-maze are mean times ± SEM spent in the open arm 10 during the observation period of 5 min. The data for the activity test are mean counts ± SEM during the test period of 5 min. Traction was evaluated by the ability of the mouse to hold on to a bar for 60 sec. The group size was 10 mice. Tolerance following chronic use, that is diminished anxiolytic activity requiring increasing doses in long term use, is evaluated in this data. The 1/II mixture 15 and diazepam were given orally each day for 7 days and then the anxiolytic activity was tested. There was no decrease in the effectiveness of either agent. The dose of each compound produced the same anxiolytic activity after 7 days of dosing as occurred after one dose. The I/II mixture still did not change motor activity. Diazepam still retained its change of motor activity and diminution of traction. 20 WO 00/40532 PCTIUS99/30168 36 5.3.6 Synergistic Effect of Combination of I/II Mixture and Diazepam TABLE Synergy in the anxiolytic activity of I/II Mixture and Diazepam Treatment mg/kg Time in open arm, Motor Activity Traction po insect Vehicle 13.1 ±4.7 31.2±1.5 60.0±0.0 I/II 0.2 37.9 ± 6.2* 21.9±3.1 60.0±0.0 Diazepam 1.0 43.5 ±6.1* 38.9±3.2# 42.6±6.3* I/II+ Diazepam 137.0 ±11.0** 27.1±4.5 44.0±4.8* 5 *p<0.02 vs vehicle control. **p<0.0001 vs I/II mixture or diazepam alone. #p<0.02 vs 1111 mixture + diazepam. The I/II mixture was given 3 h before testing and diazepam was given 10 min 10 before testing. The data for the plus-maze are mean times ± SEM spent in the open arm during the observation period of 5 min. The data for the motor activity are mean counts ± SEM during the test period of 5 min. Traction was evaluated by the ability of the mouse to hold on to a bar for 60 sec. The group size was 10 mice. To establish whether the I/II mixture and diazepam interact, an effective dose 15 of the I/11 mixture and diazepam were given to separate mice to show the anxiolytic activity of each compound when given alone. The effective dose of each compound given using appropriate timing was then combined mice. The combination of I/II mixture and diazepam resulted in a 3 fold increase in anxiolytic activity over either agent given separately. The compounds act synergistically as anxiolytics. The effect 20 of diazepam on motor activity is abolished by combining with the 1III mixture UT 2 but the ill mixture has no effect on the ataxia produced by diazepam.
WO 00/40532 PCT/US99/30168 37 5.3.7 Effect of Coadministration of Flumazenil TABLE6 The Effect of the Benzodiazepine Receptor Antagonist Flumazenil 5 on the Anxiolytic Effect of the I/II Mixture and Diazepam Treatment Challenge Time in Open Arms Vehicle saline 13.0 ± 3.4 Vehicle flumazenil 6.7 ± 2.9 I/11 saline 37.9 ± 6.2 1/11 flumazenil 31.2 ±3.8 Diazepam saline 43.5 ± 6.1 Diazepam flumazenil 13.5 ± 3.8* *p<0.0001 vs the mice pretreated only with diazepam. The time in the open arm of the elevated plus maze was observed during a 5 min period. The 1/II mixture (0.2 mg/kg po) was given 3 h before testing. Diazepam 10 (1 mg/kg po) was given 10 min before testing. Flumazenil (0.3 mg/kg sc) was given 10 min before testing. Flumazenil is a benzodiazepine receptor blocking agent which when given blocks the anxiolytic effects of benzodiazepines. Flumazenil blocks the anxiolytic activity of diazepam but does not block the anxiolytic activity of the il mixture. This 15 indicates that the I/ll mixture does not physiologically act at the same site as the benzodiazepine anxiolytics.
WO 00/40532 PCT/US99/30168 38 TABLE7 The Effects of the Benzodiazepine Receptor Antagonist Flumazenil on Motor Activity and Traction Measured Following Administration of Diazepam or I/II mixture Treatment Challenge Motor Activity Traction Vehicle saline 29.0 ± 2.4 60.0 ± 0.0 Vehicle flumazenil 25.6 ± 3.0 60.0 ± 0.0 I/II saline 21.9 ±3.1 60.0 ±0.0 YII flumazenil 28.6 ± 2.7 60.0 ± 0.0 Diazepam saline 38.9 ±3.2* 42.6± 6.3# Diazepam flumazenil 27.9 ± 1.8** 54.5 ± 2.9## 5 *p< 0.01 vs control **p< 0.007 vs diazepam alone #p<0.0001 vs control ##p<0.004 vs diazepam alone 10 The data for the activity test are for an observation period of 5 min. The data for the traction test are the mean duration of clinging. The I/ll mixture (0.2 mg/kg po) was given three hours before testing. Diazepam (1 mg/kg po) was given ten min before testing as was flumazenil (0.3 mg/kg sc). Motor activity and traction are not changed by the Ill mixture or flumazenil. 15 Flumazenil blocks the motor activity effect of diazepam and significantly decreases the diazepam alteration of traction. These serious side effects of the benzodiazepine anxiolytics appear to reside in that receptor interaction.
WO 00/40532 PCT/US99/30168 39 5.3.8 Effect of Coadministration of Bicuculline TABLE8 The Effect of the GABA Antagonist Bicuculline Administered 5 with the I/II mixture and Diazepam Treatment Bicuculline, Time spent in mg/kg po mg/kg open arm, Motor Traction sec. Activity Vehicle saline 13.0±3.5 29.0±2.4 60.0±0.0 Vehicle 0.1 7.1±2.8 21.5±2.7 60.0±0.0 I/II 0.2 saline 37.9±6.2* 21.9±3.1 60.0±0.0 I/II 0.2 0.1 16.7±5.3 36.1±2.7 60.0±0.0 Diazepam 1.0 saline 43.5±6.1* 38.9±3.2* 42.6±6.3* Diazepam 1.0 0.1 17.4±4.9 37.9±2.3* 46.6±6.9* *p<0.5 vs control vehicle. The I/II mixture was given 3 h before testing and diazepam and bicuculline were given 10 min before testing. The data for the plus-maze are mean times ± SEM 10 spent in the open arms during the observation period of 5 min. The data for the motor activity are mean counts ± SEM during the test period of 5 min. Traction was evaluated by the ability of the mouse to hold on to a bar for 60 sec. The group size was 10 mice. The GABAergic antagonist bicuculline abolished the anxiolytic activity of 15 both the 1111 mixture and diazepam identifying a common effect on the GABAergic system. Bicuculline did not block the effect of diazepam on motor activity or traction.
WO 00/40532 PCT/US99/30168 40 5.3.9 Effect of Coadministration of Caffeine TABLE9 Effect of Caffeine on the Pharmacological Activity of Diazepam 5 and I/II Mixture Treatment Challenge Time in Open Motor Traction Activity Vehicle saline 14.0±3.8 28.9±2.4 60.0±0.0 Vehicle Caffeine 13.9±9.0 60.5±7.4*** 60.0±0.0 1/11 saline 37.9±6.2 21.9±3.1 60.0±0.0 I/II Caffeine 6.1±2.2* 61.1±5.0# 60.0±0.0 Diazepam saline 43.5±6.1 38.9±3.2 42.6±6.3+ Diazepam Caffeine 93.8±11.3** 55.1±7.1## 50.4±5.1+ *p<0.003 vs ill mixture **p<0.0001 vs diazepam ***p<0.0001 vs control 10 #p<0.0001 vs I/II mixture ##p<0.03 vs diazepam + p<0.05 vs vehicle control I/II mixture, 0.2 mg/kg, was given po 3 hr before testing. Diazepam 1.0 mg/kg was given po 10 min before testing. Caffeine 30 mg/kg was given ip 15 min before 15 testing. The time in sec in the open arms is reported as means ± SEM during a 5 min observation period. The motor activity is reported as mean ± SEM for the five min test period. Traction reports the ability of the mouse to hold to a bar for 60 sec mean SEM. Group size was 10 mice. Caffeine is an axiogenic agent in high doses and increases motor activity. 20 Given at a high dose with the il mixture, caffeine abolished the anxiolytic activity of the Ill mixture. A series of doses would be needed to establish a dose effect curve. Diazepam combined with caffeine resulted in increased anxiolytic activity and motor activity but did not change the ataxia production.
WO 00/40532 PCTIUS99/30168 41 5.3.10 Effect of Coadministration of CCK TABLE 10 5 The Effect of CCK on the Pharmacological Activity of the I/II Mixture and Diazepam Treatment Challenge Time in Open Motor Traction Activity Vehicle saline 13.7 ± 4.9 27.1 ± 1.9 60.0 ± 0.0 Vehicle CCK 2.2 ± 1.1 29.2 ± 1.9 60.0 ± 0.0 I/II saline 37.9 ±6.2 21.9 ±3.1 60.0 ±0.0 I/II CCK 11.1 ±3.6* 20.2 ±3.7 60.0 ±0.0 Diazepam saline 43.5 ± 6.1 38.9 ± 3.2 42.6 ± 6.3 Diazepam CCK 57.3 ± 13.0 34.1 ± 5.1 41.3 ± 7.7 *p<0.008 vs I/II mixture alone. The I/II mixture was given 0.2 mg/kg po 3 h before the test. Diazepam 1.0 10 mg/kg po and CCK 50 ug/kg ip were given 10 min before the test. The data for the plus-maze test are mean ± SEM time spent in the open arms during the 5 min observation period. The data for the motor activity are mean counts ± SEM during the 5 min test period. Traction was evaluated by the ability of the mouse to hold on to a bar for 60 sec. The group size was 10 mice. CCK is cholecystokinin Ac-fragment 26 15 29 amide non-sulfated. Cholecystokinin (CCK) is anxiogenic and its administration is used as a screen for discovery of new anxiolytic agents. CCK in this initial test shows it anxiogenic effect and effectively abolished the anxiolytic effect of I/II mixture. It had no effect on any of the three effects of diazepam. 20 WO 00/40532 PCT/US99/30168 42 TABLE 11 The Effect of a Constant Dose of CCK on Three Doses of the I/II Mixture Treatment Dose mg/kg Challenge Time in Open Motor Activity Vehicle 0 saline 13.7 ±4.9 27.1 ± 1.9 Saline 0 CCK 2.2 ± 1.1 29.2 ± 2.2 1111 0.2 saline 37.9 ± 6.3 21.9 ± 3.1 I/II 0.2 CCK 11.1 ±3.5 20.2 ±3.7 I/II 0.5 saline 87.3 ± 12.8 26.0± 1.8 I/II 0.5 CCK 28.5 ± 5.4* 35.5± 4.7# I11 2.0 saline 95.3 ± 8.7 27.0 ± 1.7 II 2.0 CCK 48.3 ± 15.0*# 32.3 ± 2.1 *p<0.05 vs CCK 5 # p<0.05 vs control vehicle. The I/II mixture was given orally 3 h before the test. CCK, 50 ug/kg, was given ip 10 min before the test. The data for the plus-maze are mean times ± SEM spent in the open arms during the 5 min observation period. The data for the motor activity are mean counts ± SEM during the 5 min test interval. The group size was 10 10 mice. CCK is cholecystokinin Ac-fragment 26-29 amide non-sulfated. CCK at a constant dose of 50 micrograms/kg abolishes the anxiolytic activity of the I/II mixture at a dose of 0.2 mg/kg po. CCK is antagonized in a dose dependent manner by 0.5 mg/kg and 2 mg/kg doses of the I/II mixture.
WO 00/40532 PCT/US99/30168 43 5.4 Example 4. Evaluation of Dependence Liability of the Dihydrohonokiol Isomer Mixture TABLE 12 5 Number of Mice in Groups of Ten Showing Symptoms When Treated with Flumazenil Following 12 Days Administration of Diazepam or I/II Mixture Drug Flumaz. Hr Tr Cc Tc Tf Rf Saline 0 1 0 0 0 0 0 Saline 10 mg/kg 3 0 0 0 1 0 Diazepam 10 7 0 0 0 0 0 0.5 mg/kg 1.0 10 10 0 0 0 0 3 2.0 10 10 5 0 0 2 5 5.0 10 10 7 0 0 1 8 10.0 10 10 5 3 1 1 10 1111 10 4 0 0 0 0 0 0.2 mg/kg 0.5 10 0 0 0 0 0 0 2.0 10 3 0 0 0 0 0 Saline and Tween 80, /111 mixture and Diazepam were given orally once a day for twelve days. The challenge with 10 mg/kg I p Flumazenil followed 24 h after the 10 last treatment. The number represents the number of mice of the ten treated mice showing symptoms during the 20 min observation period following Flumazenil treatment. Hr: hyper-reactivity indicated by vocalization caused by light pressure on the back 15 Tr: tremor Cc: clonic convulsions Tc: tonic convulsions Tf: tail flick WO 00/40532 PCT/US99/30168 44 Rf: running fit evoked by auditory stimulus To evaluate dependence liability, I/I mixture and diazepam were given for 12 days. This was followed by administration of flumazenil the benzodiazepine antagonist. No dihydrohonokiol (I/II mixture) antagonist is known. Six signs of 5 withdrawal following flumazenil were observed and recorded. Diazepam showed extensive withdrawal signs following all doses 0.5 mg/kg to 10 mg/kg. The I/IL mixture showed no withdrawal response at the tested doses of 0.2 mg/Kg, 0.5 mg/Kg or 2.0 mg/Kg. The 2.0 mg/Kg dose is 10 times the effective dose. The hyper reactivity response of the I/II mixture given flumazenil was no different from 10 flumazenil given alone. The 0.5 mg/kg dose of the I/I mixture followed by flumazenil produced no hyper-reactivity response at all.
WO 00/40532 PCT/US99/30168 45 5.5 Example 5. Evaluation of Sedative Potential of the Dihydrohonokiol Isomer Mixture TABLE 13 5 Effect of 1/11 Mixture, Diazepam and I/II mixture plus Diazepam on the Duration of Hexobarbital Induced Sleep Treatment, dose mg/kg po Additional drug, dose Duration of sleep following hexobarbital mg/kg po in sec Vehicle 2552 ±209 I/II 0.1 2822 ±138 I/II 0.2 2580 255 I/II 0.5 2753 ±296 1/11 1.0 2440 ±37 /111 2.0 2870± 191 Vehicle 2902 ± 84 Diazepam 1.0 4476 ±336* Diazepam 1.0 111 0.1 4342 ± 195* Diazepam 1.0 IIl 0.2 4248 ± 66* Diazepam 1.0 I/I 0.5 4526 ±467* Diazepam 1.0 1/11 1.0 4052 ±217* Diazepam 1.0 I/I 2.0 4758 ±299* *p<0.0003 vs vehicle containing hexobarbital control. All mice received 100 mg/kg hexobarbital ip. The I/11 mixture was given 10 orally 3 h prior to hexobarbital to obtain the maximum anxiolytic effect of the I/ll mixture. Diazepam was given 10 min before hexobarbital to obtain the maximum effect. The time from loss of righting reflex in each mouse to the return of the righting reflex was recorded as the sleep duration. To evaluate the sedative effects of the I/II mixture and diazepam all mice 15 received a sedative dose of hexobarbital. The duration of the loss of righting reflex WO 00/40532 PCTIUS99/30168 46 (ability of the mouse to change from lying on the side to upright posture) was recorded. Doses of the I/II mixture up to 2 mg/kg which is 10 times the effective anxiolytic doses caused no increase in sleep time. Diazepam at the effective anxiolytic dose of 1 mg/kg increased sleep time significantly. The I/I mixture when 5 combined with diazepam, and hexobarbital, caused no increase in sleep time over that of diazepam plus hexobarbital. 5.6 Example 6. Evaluation of Effect of Dihydrohonokiol Isomer Mixture on Cognitive Function 10 TABLE 14 Effect of Diazepam and I/II Mixture on Learning and Memory Treatment Dose, po mg/kg Training Retention Latency Latency Vehicle saline 59.1 ± 10.1 32.4 ± 9.4 Diazepam 1.0 41.8 ± 9.3 109.2 ± 16.9* Vehicle saline 71.7 ± 10.4 31.9 ± 9.9 Diazepam 1.0 85.3 ± 13.2 37.1 ± 5.8 Vehicle saline 59.1 ± 9.8 25.6 ± 3.3 I/II 0.2 59.3 ± 8.8 26.2 ± 3.1 I/II 2.0 121.0 46.0 ±6.6* 12.7* Vehicle saline 62.8 ± 12.1 25.2 ± 2.8 I/II 0.2 60.1 ±9.1 24.3 ±2.8 III 2.0 71.9 ± 13.1 29.9 ± 5.3 *p<0004 vs control vehicle and saline 15 The I/II mixture was given 3 h before testing and Diazepam was given 10 min before testing. Group size was 10 mice.
WO 00/40532 PCT/US99/30168 47 To test the cognitive effects of the Il mixture and diazepam, training latency and retention latency were evaluated using the elevated plus-maze. On one of two tests diazepam show significant retention latency at the effective anxiolytic doses of 1 mg/kg po. In one of two tests the J/II mixture showed only at 10 times the effective 5 anxiolytic dose a training and retention latency. 5.7 Example 7. Evaluation of Effect of Dihydrohonokiol Isomer Mixture on Conditioned Place Preference 10 TABLE 15 The Effect of the I/I mixture and Diazepam on Conditioned Place Preference Treatment mg/kg orally for six Time in sec in light compartment days after conditioning Saline 11 2
.
7 ± 10.l Diazepam 1.0 182.9±16.8** Saline 105.
4
±
6
.
9 ' I/II 0.2 105.2±10.8' *p<0.002 vs saline tn=10 mice. 15 The elevated plus-maze test was used to evaluate the ability of the drugs to engender a desire to continue the drug, time spent at the site of six days prior administration of the drug was observed. The time spent at the site of administration following diazepam was significantly increased over saline administration. The time spent at the site following the 111 mixture administration was no different from that of 20 saline administration.
WO 00/40532 PCT/US99/30168 48 5.8 Example 8. Anxiolytic Activity of 3-n-Propyl-5'-(2-propenyl)-1,1' biphenyl-2',4-diol (I) and 5'-n Propyl-3-(2-propenyl)-1,1'biphenyl-2',4-diol (II) The I/II mixture consists of the two dihydrohonokiol isomers in the ratio of 5 92:08. These isomers were separated by preparative HPLC as described in § 5.1.1 and their anxiolytic activity compared using a modified elevated plus-maze. The major modification change from the maze used in the previous studies was the use of a camcorder placed above and to the side of the maze so that no human observer was visible to the mouse running the maze. This change resulted in an increase in the time 10 that the mouse remained on the transparent arm. The control time increased from 13 sec to 41 sec and the treated times also increased. The small amount of the II isomer (5'-n-propyl-3-(2-propenyl)-1,1'-biphenyl-2',4-diol) separated limited the inventors' group size to 5 instead of 10 mice. The oral doses were diazepam 1 mg/kg and I isomer (3-n-propyl-5'-(2-propenyl)-1,1'-biphenyl-2',4-diol) and II isomer each at 0.2 15 mg/kg. As shown in Table 16 and Table 17, both isomers were active but the I isomer was more active than the II isomer. TABLE 16 Anxiolytic Activity of Isomer I and Isomer II in the Modified Elevated 20 Plus Maze Test Mean Std. Dev. Std. Error Count Maze, Total 93.195 69.503 15.541 20 Maze, CONTROL 41.220 18.693 8.360 5 Maze, Group A 134.860 43.782 19.580 5 Maze, Group B 113.280 62.607 27.999 5 Maze, Group C 83.420 102.905 46.020 5 Group A: Diazepam Group B: Isomer I Group C: Isomer II WO 00/40532 PCT/US99/30168 49 TABLE 17 Statistical Analysis of Anxiolytic Activity of Isomer I and Isomer II in the Modified Elevated Plus Maze Test Mean Dif. Crit. Diff. P-Value CONTROL, Group A -93.6400 86.8267 0.0362 CONTROL, Group B -72.0600 86.8267 0.0976 CONTROL, Group C -42.2000 86.8267 0.3182 Group A, Group B 21.5800 86.8267 0.6055 Group A, Group C 51.4400 86.8267 0.2272 Group B, Group C 29.8600 86.8267 0.4765 5 Fisher's PLSD for Maze. Descriptive statistics split by Group.
WO 00/40532 PCT/US99/30168 50 5. 8.1 Further Studies on the Anxiolytic Activity of Isomer I The I isomer was further tested as shown in Tables 17 and 18. The anxiolytic activity of isomer I at an oral dose of 0.2 mg/kg was compared to diazepam at an oral dose of 1 mg/kg using the new elevated plus maze system with camcorder. Both 5 compounds showed significant anxiolytic effect. TABLE 18 Anxiolytic Activity of Isomer I and Isomer II in the Modified Elevated Plus Maze Test Mean Std. Dev. Std. Error Count Maze, Total 83.585 57.096 10.988 27 Maze, CONTROL 41.167 26.178 8.726 9 Maze, Group A 118.089 57.168 19.056 9 Maze, Group B 91.500 56.633 18.878 9 10 Group A: Diazepam Group B: Isomer I TABLE 19 15 Statistical Analysis of Anxiolytic Activity of Isomer I in the Modified Elevated Plus Maize Test Mean Difference Crit. Difference P-Value CONTROL, Group A -76.9222 47.5334 0.0027 S CONTROL, Group B -50.3333 47.5334 0.0388 S Group A, Group B 26.5889 47.5334 0.2597 Fisher's PLSD for Maze. Descriptive statistics split by Group.
WO 00/40532 PCT/US99/30168 51 5.8.2 Evaluation of Effect of Isomer I on Ultrasonic Vocalization TABLE20 The Effect of Isomer I, Gepirone and the Anxiogenic Compound 5 WAY on Conditioned Ultrasonic Distress Vocalizations in Adult Male Rats Treatment Number of Ultrasonic Vocalizations, 10 min. Control 114.4± 10.7 Vehicle 154.8 ± 11.7 Vehicle 94.6 ± 10.8 Isomer I 1.0 mg/kg po 48.1 ± 4.7* Vehicle 62.5 ± 8.4 Gepirone 0.5 mg/kg sc 20.0 ± 14.6* Vehicle 30.0 ±7.4 WAY 0.25 mg/kg sc 107.1 ± 19.2* *p<0.0001 vs vehicle. The rats were divided into four groups depending on their level of ultrasonic vocalization. Each of these groups was divided into an ultrasonic vocalization 10 matched control and drug test group consisting of three rats each. The conditioned ultrasonic distress vocalization test for anxiolytic activity in rats was used to compare isomer I to Gepirone, an anxiolytic of the azapirone class of compounds. The azapirone class of anxiolytics differ structurally and pharmacologically from the benzodiazepines. Their exact mechanism is unknown. 15 The primary action appears to be binding to serotonin receptors in the brain. Only Buspirone is marketed and it is not prescribed, often the benzodiazepines being preferred. In this test, single pure isomer I reduced vocalization effectively indicating it is effective in aversive conditioning test for anxiolytic activity.
WO 00/40532 PCT/US99/30168 52 5.9 Example 9- Methods for Assessing Antianxiety Activity of the Disclosed Compounds This example describes protocols to facilitate preclinical and clinical studies of the compounds of the present invention. The various elements of conducting 5 preclinical and clinical trials, including animal and patient treatment and monitoring, will be known to those of skill in the art in light of the present disclosure. The following information is being presented as a general guideline for use in preclinical and clinical antianxiety studies. 10 5.9.1 Punished Responding This procedure has been used to establish antianxiety activity in clinically established compounds. Response of rats or pigeons is maintained by a multiple schedule of food presentation. In one component of the schedule, responding produces food pellet presentation only. In a second component, responding produces both food 15 pellet presentation and is also punished by presentation of a brief electric shock. Each component of the multiple schedule is approximately 4 min in duration, and the shock duration is approximately 0.3 sec. The shock intensity is adjusted for each individual animal so that the rate of punished responding is approximately 15 to 30% of the rate in the unpunished component of the multiple schedule. Sessions are conducted each 20 weekday and are approximately 60 min in duration. Vehicle or a dose of compound are administered 30 min to 6 hr before the start of the test session by the subcutaneous or oral route. Compound effects for each dose for each animal are calculated as a percent of the vehicle control data for that animal. The data are expressed as the mean +- the standard error of the mean. 25 5.9.2 Monkey Taming Model The antianxiety activity of a compound may be established by demonstrating that the compounds are effective in the monkey taming model. Plotnikoff, (1973) described the response of rhesus monkeys to pole prodding as a method of evaluating 30 the antiaggressive activity of a test compound. In this method, the antiaggressive activity of a compound was considered to be indicative of its antianxiety activity. Hypoactivity and ataxia were considered to be indicative of a sedative component of WO 00/40532 PCT/US99/30168 53 the compound. In one study, the pole prod response-inhibition induced by a compound of this invention may be analyzed and compared with that of a standard antianxiety compound such as diazepam as a measure of antiaggressive potential, and to obtain an indication of the duration of action of the compound. 5 Male and female rhesus, cynomolgus or squirrel monkeys, selected for their aggressiveness toward a pole, are housed individually in a primate colony room. Compounds or appropriate vehicle are administered orally or subcutaneously and the animals are observed by a trained observer at varying times after drug administration. A minimum of three days (usually a week or more) elapses between treatments. 10 Treatments are assigned in random fashion except that no monkey receives the same compound two times consecutively. Aggressiveness and motor impairment are graded by response to a pole being introduced into the cage. The individuals responsible for grading the responses are unaware of the dose levels received by the monkeys. 15 5.9.3 Human Clinical Trials Antianxiety activity may be demonstrated by human clinical trials. A study may be designed as a double-blind, parallel, placebo-controlled multicenter trial. Patients are randomized into four groups, placebo and three different appropriate 20 doses of the test compound either once, twice or three times per day depending on the pharmacokinetics of the drug. The dosages may be administered orally with food. Patients are then observed during four visits to provide baseline measurements, and then visits 5 and beyond may be used as the treatment phase for the study. During the visits, patients and their caregivers may be questioned and 25 observed for signs of agitation, mood swings, vocal outbursts, suspiciousness, and fearfulness. Each of these behaviors are indicative of the effect of the test compound on an anxiety disorder.
WO 00/40532 PCTIUS99/30168 54 6.0 REFERENCES The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated 5 herein by reference. U. S. Patent 4,329,332, issued May 11, 1982. U. S. Patent 4,489,055, issued Dec. 18, 1984. U. S. Patent 5,451,410, issued Sept. 19, 1995. 10 U. S. Patent 5,500,224, issued Mar. 19, 1996. U. S. Patent 5,556,617, issued Sept. 17, 1996. U. S. Patent 5,620,708, issued April 15, 1997. U. S. Patent 5,641,515, issued June 24, 1997. U. S. Patent 5,698,515, issued Dec. 16, 1997. 15 Allen and Choun, "Large unilamellar liposomes with low uptake into the reticuloendothelial system," FEBS Lett., 223:42-46, 1987. Arshady, "In vivo targeting of colloidal carriers by novel graft copolymers," J. Mol. Recognit., 9(5-6):536-542, 1996. Baldessarini, In: Goodman and Gilman's the Pharmacological Basis of Therapeutics, 20 Goodman, Rall, Nies, Taylor (Eds.), Pergamon Press pp383-435, 1990. Calvo, Vila-Jato, Alonso, "Effect of lysozyme on the stability of polyester nanocapsules and nanoparticles: stabilization approaches," Biomaterials, 18(19):1305-1310, 1997. Calvo, Vila-Jato, Alonso, "Improved ocular bioavailability of indomethacin by novel 25 ocular drug carriers," J. Pharm. Pharmacol., 48(11):1147-1152, 1996. Chandra and Nair, "Supercritical carbon dioxide extraction and quanitifaction of bioactive neolignans from Magnolia virginiana flowers," Planta Med., 61:192 195, 1995. Couvreur et al., "Nanocapsules, a new lysosomotropic carrier," FEBS Lett., 84:323 30 326, 1977.
WO 00/40532 PCT/US99/30168 55 Damge, Vonderscher, Marback, Pinget, "Poly(alkyl cyanoacrylate) nanocapsules as a delivery system in the rate for octreotide, a long-acting somatostatin analogue," J. Pharm. Pharmacol., 49(10):949-954, 1997. Fujita, Itokawa, Sashida, Honkiol, "A new phenolic compound isolated from the bark 5 of Magnolia obvata," Chem. Pharmacol. Bull., 20:212-213, 1972. Gabizon and Papahadjopoulos, "Liposomes formulations with prolonged circulation time in blood and enhanced uptake by tumors," Proc. Natl. Acad. Sci. USA, 85:6949-6953, 1988. Gardner, Tully, Hedgecock, "The rapidly expanding range of neuronal benzodiazepine 10 receptor ligands," Prog. Neurobiol., 40:1-61, 1993. Ghosh and Bachhawat, "Targeting of liposomes to hepatocytes," In: Liver diseases, Targeted Diagnosis and Therapy Using Specific Receptors and Ligands, Wu and Wu (Ed.), Marcel Dekker, New York, pp 87-104, 1991. Hattori, Sakamoto, Endo, Kakiuchi, Kobashi, Mizuno, Namba, "Metabolism of 15 magnolol from Magnoliae cortex. I. Application of liquid chromatography mass spectrometry to the analysis of metabolites of magnolol in rats," Chem. Pharm. Bull., 32:5010-5017, 1984. Hattori, Sakamoto, Endo, Kakiuchi, Kobashi, Mizuno, Namba, "Metabolism of magnolol from Magnoliae cortex. II. Absorption, metabolism and excretion of 20 [ring-14C]magnolol in rats," Chem. Pharm. Bull., 34:158-167, 1986. Henry-Michelland et al., "Attachment of antibiotics to nanoparticles; Preparation, drug-release and antimicrobial activity in vitro," Int. J. Pharm., 35:121-127, 1987. Hosaya and Yamamura (Eds.), Recent Advances in the Pharmacology of Kampo 25 (Japanese Herbal) Medicines, International Congress Series 854, Excerpta Medica, Tokyo, 1988. Kuribara, Morita, Ishige, Hayashi, Maruyama, "Investigation of the anxiolytic effect of the extracts derived from Saiboku-To, an oriental herbal medicine, by an improved plus-maze test in mice," Jpn. J. Neuropsychopharmacol., 18:179 30 190 (Abstract in English), 1996.
WO 00/40532 PCT/US99/30168 56 Maruyama, Kuribara, Morita, Yuzurihara, Weintraub, "Identification of magnolol and honokiol as anxiolytic agents in extracts of saiboku-to, an oriental herbal medicine," J. Nat. Prod., 61(1):135-138, 1998. Narita H, "Use of Kampo medicine in psychiatry," Jpn. J. Neuropsychopharmacol., 5 12:165-172 (In Japanese), 1990. Plotnikoff, "Clorazepate dipotassium, tranxene: anti-anxiety and antidepresant activity," Res. Comm. Chem. Path. Pharmacol., 5:128-134, 1973. Watanabe, Watanabe, Goto, Yamaguichi, Yamamoto, Hagine, "Pharmacological properties of magnolol and honokiol extracted from Magnolia officinalis: 10 central depressant effects," Planta Med., 49:103-108, 1983. Yu and Chang, "Submicron polymer membrane hemoglobin nanocapsules as potential blood substitutes: preparation and characterization," Artif Cells Blood Substit. Immobil. Biotechnol., 24(3):169-183, 1996.

Claims (25)

1. A compound of the formula: H O OH 5 or a salt thereof.
2. A compound of the formula: H t OH or a salt thereof. 10
3. A compound of the formula: H 2, 5 6 5 5' 2 3 OH R WO 00/40532 PCT/US99/30168 58 wherein R is -CH 2 -CH=CH 2 , -CH=CH-CH 3 , and -CH 2 -CH 2 -CH 3 ; and X is from one to two substituents in any of the 3'-, 4-', 5'-, or 6'-positions and is separately and independently fluorine, hydroxy, methoxy, (1-adamantyl), C 1 -C 5 alkyl, 5 C 2 -C 3 alkenyl, C 2 -C 3 alkylcarbonyl, and C 1 -C 4 carboxyalkyl, wherein the C 1 -C 5 alkyl group may be substituted with one or more fluorine or hydroxyl; or a salt thereof; providing, wherein R is -CH2-CH=CH 2 , X is not 5'-CH 2 -CH 3 ; 5'-CH 2 -CH=CH 2 ; 5' OH; 3'-OH, 5'-CH 2 -CH-CH 3 ; and 3'-OCH 3 , 5'-CH 2 -CH=CH 2 ; or wherein R is -CH 2 -CH 2 -CH 3 , X is not 5'-CH 2 -CH 2 -CH 3 . 10
4. The compound according to claim 3, wherein R is -CH 2 -CH=CH 2 .
5. The compound according to claim 3, wherein R is -CH 2 -CH 2 -CH 3 . 15
6. The compound according to claim 3, wherein R is -CH 2 =CH-CH 3 .
7. The compound according to claim 3, wherein X is from one to two substituents in any of the 3'-, 4-', 5'- or 6'-positions and is independently 5'-CH=CH 2 ; 5'-CH 2 -CH 3 ; 5'-CH=CH 2 , 3'-OH; 5'-CH 2 -CH=CH 2 ; 5'-CH 2 -CH=CH 2 , 3'-OH; 5' 20 CH 2 -CH 2 -CH 3 , 3'-OH; 5'-CH 2 -CH=CH 2 , 3'-OCH 3 ; 5'-CH 2 -CH2-CH 3 , 3'-OCH 3 ; 5' CH=CH-CH 3 , 3'-OH; 5'-CH=CH-CH 3 , 3'-OCH 3 ; 5'-CH 3 ; 5'-CH(CH 3 ) 2 ; 5' CH 2 CH(CH 3 ) 2 ; 5'-C(CH 3 ) 3 ; 5'-CH(CH 3 ) 2 C 2 H 5 ; 5'-(1-adamantyl); 5'-CH(CH 3 ) 2 , 6 CH 3 ; 5'-CH(CH 3 ) 2 , 4 -CH 3 ; 5'-CH(-OH)-CH 3 ; 5'-CH 2 -CH(-OH)-CH 3 ; 5'-CH 2 -COOH; 5'-CH 2 CH 2 -COOH; 5'--CH 2 CH 2 CH 2 -COOH; 5'-CH 2 -COOH, 3-OH; 5'-OH; 5'-OCH 3 ; 25 3'-F; 4'-F; 5'-F; 3'-F, 5'-CH 3 ; 3'-F, 5'-CH2-CH 3 ; 3'-F, 5'-CH 2 -OH; 3'-F, 5'-CH 2 -CH 2 OH; 3'-F, 5'-COOH; 3'-F, 5'-CH 2 -COOH; 3'-F, 5'-CH=CH-CH 3 ; 3'-F, 5'-CH 2 -CH 2 CH 3 ; 3'-F, 5'-CH 2 -CH=CH 2 ; 3'-F, 5'-CH 2 -CHF-CH 3 ; 3'-F, 5'-CH 2 -CHF-CH 2 F; 4'-F, 5'-CH 3 ; 4'-F, 5'-CH 2 -OH; 3'-F, 5'-CH 2 -CH 3 ; 6'-F, 5'-CH 2 -CH 3 ; 6'-F, 5'-CH 2 -OH; and 6'-F, 5'-COOH. 30 WO 00/40532 PCT/US99/30168 59
8. The compound according to claim 3, wherein X is independently 5'-C(=0) CH 3 and 5'-CH 2 -C(=O)-CH 3
9. A compound of the formula: H 2' 6 3' 5 4- 6' X ' 4' 2 OH 55 5 R wherein R is -CH 2 -CH=CH 2 , -CH=CH-CH 3 , and -CH 2 -CH 2 -CH 3 ; and X' is from one to two substituents in any of the 2-, 3-, 5-, or 6-positions and is separately and independently fluorine, hydroxy, methoxy, C-C 4 alkyl, C 3 alkenyl, C C 3 alkylcarbonyl, Cr-C 3 carboxyalkyl, and C 3 carboxyalkenyl, wherein the CI-C 4 alkyl 10 group may be substituted with one or more fluorine or hydroxyl; or a salt thereof; providing, wherein R = -CH 2 -CH=CH 2 , X' is not 3-CH2-CH 3 and 3'-CH 2 -CH=CH 2 ; or wherein R = -CH 2 -CH 2 -CH 3 , X' is not 3-CH 2 -CH 2 -CH 3 .
10. The compound according to claim 9, wherein R is -CH 2 -CH=CH 2 . 15
11. The compound according to claim 9, wherein R is -CH 2 -CH 2 -CH 3 .
12. The compound according to claim 9, wherein R is -CH 2 =CH-CH 3 . 20
13. The compound according to claim 9 wherein X' is from one to two substituents in any of the 2-, 3-, 5- or 6-positions and is independently 3-CH 3 ; 3-CH 2 CH 3 ; 3-CH 2 -CH=CH 2 ;3-CH 2 -CH=CH 2 , 5-OH; 3-CH 2 -CH 2 -CH 3 , 5-OH; 3-CH 2 CH=CH 2 , 5-OCH 3 ; 3-CH 2 CH 2 CH 3 , 5-OCH 3 ; 3-CH=CH-CH 3 ; 3-CH 3 , 6-CH(CH 3 ) 2 ; 3,5 di-CH 3 ; 2,6 di-CH 3 ; 3-CH(CH 3 ) 2 ; 3-CH(CH 3 ) 2 , 6-CH 3 ; 3-CH 2 CH(CH 3 ) 2 ; 3-CH( 25 OH)-CH 3 ; 3-CH 2 -CH(-OH)-CH 3 ; 3-COOH, 6-OH; 2-COOH; 3-OCH 3 ; 3-CH 2 COOH; 3-CH 2 CH 2 COOH; 3-CH=CHCOOH; 3-CH2OH, 2-OH; 3-CH 2 OH, 5-OCH 3 ; 3- WO 00/40532 PCT/US99/30168 60 CH 2 OH, 5-CH 3 , 3-F; 2F; 2-F, 3-CH 3 ; 6-F, 3-CH 3 ; 5-F, 3-CH=CH-CH 3 ; 5-F,3-CH 2 CH 2 -CH 3 ; 5-F, 3-CH 2 -CH=CH 2 ; 5-F, 3-CH 2 -CHF-CH 3 ; and 5-F, 3-CH 2 -CHF-CH 2 F.
14. The compound according to claim 9 wherein X' is from one to two 5 substituents in any of the 2-, 3- or 5-positions and is independently 3-C(=O)-CH 3 ; 3 CH2-C(=O)-CH 3 ; 3-CHO, 2-OH; 3-CHO, 5-OCH 3 ; and 3-CHO, 5-CH 3 .
15. A pharmaceutical composition comprising a suitable carrier and the compound of claim 1 or the compound of claim 2 or the compound of claim 1 and the compound 10 of claim 2.
16. The composition of claim 15, wherein said compound of claim 1 or said compound of claim 2 or said compound of claim 1 and said compound of claim 2 are present in an anxiolytically effective amount. 15
17. A pharmaceutical composition comprising a suitable carrier and one or more compounds of claim 3 or claim 9.
18. The composition of claim 17, wherein said one or more compounds of claim 3 20 or claim 9 are present in an anxiolytically effective amount.
19. A method of reducing or preventing anxiety in mammals, which comprises administering to a mammal in need of such treatment an effective amount of the compound of claim 1 or the compound of claim 2 or the compound of claim 1 and the 25 compound of claim 2.
20. A method of reducing or preventing anxiety in mammals, which comprises administering to a mammal in need of such treatment an effective amount of one or more compounds of claim 3 or claim 9. 30
21. A method of alleviating an anxiety-related disorder in a patient comprising administering to a patient in need of such treatment an amount of a compound of WO 00/40532 PCT/US99/30168 61 claim 3 or claim 9 or a plurality of compounds of claim 3 or claim 9 effective in reducing or preventing anxiety and the symptoms associated with such disorder.
22. A method according to claim 21, wherein said anxiety-related disorder is 5 selected from the group consisting of panic disorder, generalized anxiety disorder, agoraphobia, simple phobias, social phobia, posttraumatic stress disorder, obsessive compulsive disorder, and avoidant personality disorder.
23. A process for preparing dihydrohonokiol comprising the steps of: 10 a) admixing tris-(triphenylphosphine) rhodium(I) chloride catalyst with non-polar organic solvent; b) admixing honokiol with said catalyst and said non-polar solvent to form a mixture; and c) saturating said mixture with hydrogen to form dihydrohonokiol such 15 that 0.6 to 1.2 mol of hydrogen reacts per mol of honokiol.
24. The process of claim 22, wherein said non-polar organic solvent is selected from the group consisting of toluene, benzene, pentane, hexane, heptane and octane. 20
25. The process of claim 22, further comprising chromatographic separation and purification of said dihydrohonokiol into the two positional isomers 3-n-propyl-5'-(2 propenyl)- 1,1 '-biphenyl-2',4-diol and 5'-n-propyl-3-(2-propenyl)- 1,1 '-biphenyl-2',4 diol.
AU25899/00A 1998-12-31 1999-12-17 Synthesis of dihydrohonokiol compositions Abandoned AU2589900A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US22418898A 1998-12-31 1998-12-31
US09224188 1998-12-31
PCT/US1999/030168 WO2000040532A2 (en) 1998-12-31 1999-12-17 Synthesis of dihydrohonokiol compositions

Publications (1)

Publication Number Publication Date
AU2589900A true AU2589900A (en) 2000-07-24

Family

ID=22839629

Family Applications (1)

Application Number Title Priority Date Filing Date
AU25899/00A Abandoned AU2589900A (en) 1998-12-31 1999-12-17 Synthesis of dihydrohonokiol compositions

Country Status (4)

Country Link
EP (1) EP1140756A2 (en)
AU (1) AU2589900A (en)
CA (1) CA2358554A1 (en)
WO (1) WO2000040532A2 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2423181A1 (en) 2010-07-28 2012-02-29 Prous Institute For Biomedical Research S.A. Multitarget substituted biphenyl diol derivatives
WO2014170902A1 (en) * 2013-04-17 2014-10-23 Ariel - University Research And Development Company, Ltd. Cb2 receptor ligands for the treatment of psychiatric disorders
CN108883082B (en) * 2015-06-11 2021-10-26 威斯康星州医药大学股份有限公司 Silo-honokiol compounds and methods of synthesis and use thereof
US11897910B2 (en) 2015-06-11 2024-02-13 The Medical College Of Wisconsin, Inc. Mito-honokiol compounds and methods of synthesis and use thereof
EP3364957B1 (en) 2015-10-23 2020-12-09 Colgate-Palmolive Company Improved synthesis of honokiol
MX2022001429A (en) * 2019-08-06 2022-04-06 Justin Kirkland Formulations including dihydrohonokiol.
WO2022160065A1 (en) * 2021-01-31 2022-08-04 Betterlife Pharma Inc. Formulations including dihydrohonokiol

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS59139335A (en) * 1983-01-31 1984-08-10 Tsumura Juntendo Inc Novel hydroxybiphenyl compound
KR900012875A (en) * 1989-02-08 1990-09-03 오스카 아끼히꼬 A biphenyl derivative, a neuronal cell-denatured restoration or protection agent, and a method for producing a cuneyl derivative used therein
JPH072655A (en) * 1993-06-18 1995-01-06 Otsuka Pharmaceut Co Ltd Agent for repairing or protecting denaturation of peripheral nerve
AU8168998A (en) * 1997-06-26 1999-01-19 Board Of Regents, The University Of Texas System Synthesis of dihydrohonokiol compositions

Also Published As

Publication number Publication date
WO2000040532A2 (en) 2000-07-13
CA2358554A1 (en) 2000-07-13
WO2000040532A3 (en) 2000-11-16
EP1140756A2 (en) 2001-10-10

Similar Documents

Publication Publication Date Title
US7279501B2 (en) Pharmaceutical composition useful in prevention and treatment of beta-amyloid protein-induced disease
EP2275097B1 (en) Compositions comprising cannabinoids for treatment of nausea, vomiting, emesis, motion sickness or like conditions
JP2002511084A (en) Selective thyroid hormone analogs
JP2009528330A (en) Hydroxylated long chain resveratrol derivatives useful as neurotrophic agents
AU768712B2 (en) Antiprotozoal saponins
Ng et al. Functional properties of guinea pig eosinophil leukotriene B4 receptor.
KR101270841B1 (en) Hydroquinone long-chain derivatives and/or phenoxy long-chain derivatives, and pharmaceuticals comprising the same
EP0991614A1 (en) Synthesis of dihydrohonokiol compositions
WO2001030335A2 (en) Pharmaceutical compositions useful in the prevention and treatment of beta-amyloid protein-induced disease
AU2589900A (en) Synthesis of dihydrohonokiol compositions
JPH10505348A (en) Methods for identifying and treating resistant tumors
AU4085799A (en) Novel diphenylethylene compounds
DE3832362A1 (en) NEW CYCLOPEPTOLIDES, PROCESS FOR THEIR PREPARATION AND THEIR USE
JP4714304B2 (en) Use of 5-methyl-1,3-benzenediol or its derivatives in the preparation of pharmaceuticals and functional foods for the treatment or prevention of depression
US5543428A (en) Method for treating resistant tumors
US4639466A (en) 3&#39;,6-diisopentenyl-2&#39;,4&#39;-dihydroxy-5,7-dimethoxy-isoflavan and related compounds useful as anxiolytic agents and avermectin antidotes
WO2012103813A1 (en) Danshensu and chuanxiongqin derivatives, process for preparation, and use thereof
WO2006089881A1 (en) Use of phenanthrene derivatives as anti-inflammatory agents, synthesis method and intermediate products
KR20210022504A (en) Novel sesquiterpene derivatives and use thereof
US9428431B2 (en) Compositions comprising CB receptor agonists, uses thereof and methods for their preparation
KR100542323B1 (en) Preparation method of compounds with apoptosis-inducing activity on cells from the Machilus thunbergii
CN115025075B (en) New use of FK3 medicine
JP2003073292A (en) New cholesterol synthesis inhibitor
KR100487112B1 (en) A new sphingolipid, (4E, 6E, 2S, 3R)-2-N-Eicosanoyl-4,6-tetradeca sphingenine of Beauveria bassiana 101A having enhancement of the neurite outgrowth
Widyasti Chemical constituents of fan-si abutilon indicum stems