WO2006015283A2 - Treatment of benign prostatic hyperplasia - Google Patents

Treatment of benign prostatic hyperplasia Download PDF

Info

Publication number
WO2006015283A2
WO2006015283A2 PCT/US2005/027133 US2005027133W WO2006015283A2 WO 2006015283 A2 WO2006015283 A2 WO 2006015283A2 US 2005027133 W US2005027133 W US 2005027133W WO 2006015283 A2 WO2006015283 A2 WO 2006015283A2
Authority
WO
WIPO (PCT)
Prior art keywords
lonidamine
bph
prostate
day
analog
Prior art date
Application number
PCT/US2005/027133
Other languages
French (fr)
Other versions
WO2006015283A3 (en
Inventor
George Tidmarsh
Original Assignee
Threshold Pharmaceuticals, Inc.
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 Threshold Pharmaceuticals, Inc. filed Critical Threshold Pharmaceuticals, Inc.
Priority to JP2007523870A priority Critical patent/JP2008508316A/en
Publication of WO2006015283A2 publication Critical patent/WO2006015283A2/en
Publication of WO2006015283A3 publication Critical patent/WO2006015283A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the invention relates to treatment and prevention of benign prostatic hyperplasia, and has application in the field of medicine and related fields, including chemistry, medicinal chemistry, and molecular biology.
  • Benign Prostatic Hyperplasia (BPH), a disease in which prostate epithelial cells grow abnormally and block urine flow, afflicts more than 10 million adult males in the United States and many millions more throughout the rest of the world.
  • surgical intervention was the only treatment of the disease, and even today, surgery is the treatment of last resort, almost inevitably relied upon when other treatments are not, or cease to be, effective.
  • Prostate surgery and recovery therefrom is painful, and the surgery itself may not be effective and poses the risk of serious side effects.
  • the invention provides methods and compositions for treating BPH in a human subject by administration of lonidamine or a lonidamine analog to the subject.
  • Pharmaceutical compositions useful for treatment of BPH including sustained release formulations, are also provided.
  • the formulation is orally administered and permits once-a-day dosing of a therapeutically effective dose of the compound.
  • treating refers to taking steps to obtain beneficial or desired results, including clinical results, for the patient.
  • beneficial or desired clinical results include, but are not limited to, alleviation or amelioration of one or more symptoms of BPH, diminishment of extent of disease, delay or slowing of disease progression, amelioration, palliation or stabilization of the disease state, and other beneficial results described below.
  • “reduction” of a symptom or symptoms means decrease of the severity or frequency of the symptom(s), or elimination of the symptom(s).
  • administering or "administration of a drug to a subject (and grammatical equivalents of this phrase) includes both direct administration, including self-administration, and indirect administration, including the act of prescribing a drug.
  • direct administration including self-administration
  • indirect administration including the act of prescribing a drug.
  • a physician who instructs a patient to self-administer a drug and/or provides a patient with a prescription for a drug is administering the drug to the patient.
  • a "manifestation" of BPH refers to a symptom, sign, anatomical state (e.g., prostate size), physiological state (e.g., PSA level), or report (e.g., AUASI score) characteristic of a subject with BPH.
  • a "therapeutically effective amount" of a drug is an amount of a drug that, when administered to a subject with BPH, will have the intended therapeutic effect, e.g., alleviation, amelioration, palliation or elimination of one or more manifestations of BPH in the subject. The full therapeutic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses. Thus, a therapeutically effective amount may be administered in one or more administrations.
  • a prophylactically effective amount of a drug is an amount of a drug that, when administered to a subject, will have the intended prophylactic effect, e.g., preventing or delaying the onset (or reoccurrence) of disease or symptoms, or reducing the likelihood of the onset (or reoccurrence) of disease or symptoms.
  • the full prophylactic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses.
  • a prophylactically effective amount may be administered in one or more administrations.
  • TID and “QD” have their ordinary meanings of "three times a day” and “once daily,” respectively.
  • alkyl refers to a monovalent alkane (hydrocarbon) derived radical containing from 1 to 15 carbon atoms. It may be straight, branched or cyclic and may be unsubstituted or substituted with substituent groups including but not limited to hydroxyl, halide, alkoxyl, and nitrile. Alkoxy groups that can be used include but are not limited to methooxy. Illustrative straight or branched alkyl groups include methyl, ethyl, propyl, isopropyl, butyl and t-butyl.
  • aryl refers to moieties that include one or more monocyclic or fused ring aromatic systems. Such moieties include any moiety that includes one or more monocyclic or bicyclic fused ring aromatic systems, including but not limited to phenyl and naphthyl. Aryl groups may be unsubstituted or substituted with substituent groups as listed for the particular substituted aryl.
  • heteroaryl refers to monocyclic aromatic groups having 5 or 6 ring atoms, or fused ring bicyclic aromatic groups having 8 to 10 atoms, in which the ring atoms are C, O, S, SO, SO 2 , or N and at least one of the ring atoms is a heteroatom, i.e., O, S, SO, SO 2 , or N.
  • Heteroaryl groups may be unsubstituted or substituted with substituent groups as listed for the particular substituted heteroaryl. Examples of monocyclic aromatic heteroaryl groups include but are not limited to pyridyl.
  • bicyclic fused ring heteroaryl groups include but are not limited to indazolyl, pyrrolopyrymidinyl, indolizinyl, pyrazolopyridinyl, triazolopyridinyl, pyrazolopyrimidinyl, triazolopyrimidinyl, pyrrolotriazinyl, pyrazolotriazinyl, triazolotriazinyl, pyrazolotetrazinyl, hexaaza- indenly, and heptaaza-indenyl.
  • the arrangement of the hetero atoms within the ring may be any arrangement allowed by the bonding characteristics of the constituent ring atoms.
  • heterocycloalkyl and “heterocyclyl” refer to a monocyclic or fused ring multicyclic cycloalkyl group at least a portion of which is not aromatic and in which one or more of the carbon atoms in the ring system is replaced by a heteroatom selected from O, S, SO, SO 2 , or N.
  • heterocyclyl groups include but are not limited to piperidinyl, morpholinyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydroimidazo [4,5-c] pyridinyl, imidazolinyl, piperazinyl, pyrrolidine-2-onyl, and piperidin-2-onyl.
  • cycloalkyl refers to a monocyclic or fused ring multicyclic group at least a portion of which is not aromatic and in which the ring atoms are carbon.
  • heterocycloalkenyl refers to a monocyclic or fused ring multicyclic group in which one or more of the carbon ring atoms is replaced by a hetero atom, the ring system is at least partially not aromatic, and the ring system includes at least one carbon-carbon double bond.
  • the present invention provides compositions and methods useful in the treatment of benign prostatic hyperplasia (BPH).
  • BPH benign prostatic hyperplasia
  • the invention relates to the use of lonidamine (LND) for the treatment or prevention of BPH.
  • LND lonidamine
  • the invention relates to the use of lonidamine analogs for the treatment or prevention of BPH.
  • BPH also referred to as benign prostatic hyperplasia
  • properties of lonidamine and its bioactive analogs is provided below.
  • BPH involves overgrowth (hyperplasia) of cells in the prostate, resulting in enlargement of the prostate and leading to lower urinary tract symptoms and disease.
  • the prostate gland contains secretory epithelial cells in a stroma of connective tissue and smooth muscle (see Barry, 2003, for a more detailed description of prostate anatomy), and BPH involves hyperplasia of the epithelial component.
  • the secretory epithelial component in the normal prostate is remarkable in that the level of zinc in this tissue is very high compared to other normal tissues.
  • a consequence of the high zinc levels is that, through a mechanism involving zinc inhibition of the enzyme m-aconitase, the generation of energy via the tricarboxylic acid (TCA) cycle and oxidative phosphorylation is substantially reduced in the secretory epithelium, making this tissue far more dependent than other organs and tissues upon glycolysis as an energy source.
  • TCA tricarboxylic acid
  • the zinc inhibition of m-aconitase a key enzyme in the TCA cycle, results in at least a substantial reduction in, and perhaps a near complete blockade of, the TCA cycle in prostate epithelial cells.
  • Another physiological result of the zinc-based inhibition of m-aconitase is the diversion of citrate from the TCA cycle, enabling the prostate to secret large quantities of citrate, used by the sperm as an energy source, into the seminal fluid. See, generally, Costello, 1999; Costello et al., 2000; Costello and Franklin, 2000.
  • prostate epithelial cells are uniquely dependent on glycolysis (anaerobic metabolism).
  • the present invention relates in part to the discovery of these cells' susceptibility to the drug lonidamine, which allows lonidamine to be administered, as described herein, to treat or prevent BPH in humans.
  • Lonidamine is the generic name for 1-(2,4-dichlorobenzyl)-1 H-indazole-3- carboxylic acid, and has also been referred to in the medical literature as 1-[(2,4- dichlorophenyl)methyl]-1H-indazole-3-carboxylic acid, AF1890, diclondazolic acid (DICA), and DoridaminaTM (ACRAF; Rome, Italy). See Figure 1. Lonidamine was first identified as an antispermatogenic agent and subsequently used in the treatment of breast, cervical, lung and prostate cancers, in a few countries in Europe. See Silvestrini, 1981 ; Gatto et al., 2002.
  • lonidamine's anticancer properties result at least in part from a lonidamine-mediated disruption of the mitochondrial membrane, resulting in reduced activity of mitochondrially-bound hexokinase and interference with ATP production by the glycolytic pathway and oxidative phosphorylation.
  • Floridi et al., 1981 Fanciulli et al., 1996, and references numbered 15-22 therein; and Gatto, 2002. Also see Kaplan, 2000.
  • lonidamine inhibits glycolysis and/or impairs the already diminished mitochondrial function in prostate epithelial cells, starving these cells, relative to the normal cells in the body, of energy.
  • lonidamine inhibits glycolysis and/or impairs the already diminished mitochondrial function in prostate epithelial cells, starving these cells, relative to the normal cells in the body, of energy.
  • a specific mechanism it is believed that, due to this energy deprivation, enough of the hyperplastic, epithelial cells are destroyed or otherwise reduced in size to reduce the size of the prostate and thereby relieve the condition and its clinical consequences.
  • administering provides benefits such as reduction of severity or frequency of one or more symptoms, reduction in prostate size or rate of enlargement, improvement in perceived quality of life, and reversion of other manifestations of BPH toward a more normal state.
  • administration of lonidamine to a human subject in need of prophylaxis for BPH provides benefits such as a reduction in likelihood that BPH or symptoms associated with BPH will appear, reappear or progress in the subject.
  • administration of a lonidamine analog to a human subject is similarly effective for treatment and prophylaxis of BPH.
  • lonidamine or its analogs to a human subject as described herein can be efficacious in the treatment of acute urinary retention.
  • Section 3 describes certain lonidamine analogs useful for treatment and prophylaxis of BPH.
  • Section 4 relates to synthesis and forms of lonidamine and lonidamine analogs.
  • Section 5 describes patient populations for whom administration of lonidamine and lonidamine analogs provides benefit.
  • Section 6 describes methods of administration of lonidamine (e.g., dose, route, schedule and duration of administration).
  • Section 7 describes combination therapies in which lonidamine or an analog is administered in combination with another drug or therapy.
  • Section 8 describes exemplary dosage forms.
  • Section 9 describes monitoring PSA levels in subjects treated with lonidamine or a lonidamine analog.
  • Section 10 provides examples of the use and effects of lonidamine. The description below is organized into sections for convenience only, and disclosure found in any organizational section is applicable to any aspect of the invention.
  • lonidamine in addition to lonidamine, a variety of compounds related to lonidamine are useful for the treatment and prevention of BPH.
  • Useful compounds are generally structurally similar to, are bioisosteres of, or are pharmacophores of lonidamine, as described below, and have biological activity(s) similar to those of lonidamine, as also discussed below.
  • Such compounds can be referred as "bioactive lonidamine analogs,” “lonidamine analogs,” or, in some cases, simply, “analogs.”
  • Ri represents -COOH or a derivative or bioisostere of the -COOH group.
  • Ri is usually selected from an acid group of formula -COOH; an amide of formula -CONR 3 R 4 , where R 3 and R 4 may be independently alkyl or hydrogen, with hydrogen preferred; a hydrazide of formula -CONHNR 6 Rr, where R 6 and R 7 are usually -H or -CH 3 ; a substituted ester of formula -COOR 5 , with R 5 being a residue easily hydrolyzed in the subject after administration and generally a straight chain or branched chain alkyl group substituted with one or more hydroxyl groups, more usually a straight chain or branched chain methyl, ethyl, or propyl group substituted with one or more hydroxyl groups, more usually still an ethyl group substituted with one hydroxyl group or
  • Ri may also be the carboxylate anion of formula -COO " , in which case the lonidamine or lonidamine analog will be associated with a counter ion, Z + , where Z + is a pharmaceutically acceptable cation.
  • R 2 represents a substituted or unsubstituted aryl or heteroaryl group.
  • R 2 is a substituted aryl group; more usually, a substituted phenyl group; more usually still, a phenyl group substituted by one, two, or three substituents independently selected from halo and alkyl substituents, particularly -Cl, -Br, -I, CF 3 and -CH 3 substituents.
  • R 2 is a substituted phenyl group
  • R 2 is usually -Cl, -Br, -I, CF 3 Or -CH 3 , monosubstituted phenyl, substituted at the 2 ,3, or 4 position; dichloro, dibromo, dimethyl, or chloro and methyl disubstituted phenyl, substituted at the 2 and 3 or 2 and 4 positions; or 2, 4, 5 trichlophenyl.
  • R 2 is a substituted phenyl group
  • R 2 is more usually 2,4-dichlorophenyl or 4-chloro-2-methylphenyl.
  • X represents a straight chain or branched chain, saturated or unsaturated hydrocarbon linkage group.
  • Y represents a moiety of formula -CHR 7 -, where R 7 is hydrogen or a straight chain or branched chain alkyl group; more usually R 7 is hydrogen or a straight chain alkyl group; more usually still R 7 is hydrogen, methyl, ethyl, or n-propyl; more usually still Ry is hydrogen or methyl; and most usually Ry is hydrogen (i.e., Y is most usually -CH 2 -).
  • n is zero or, most usually, one.
  • v- ⁇ is a core ring system that may generally be an aryl, heteroaryl, cycloalkyl or heterocyclyl ring system.
  • the Ar core ring system usually includes 2 fused rings.
  • the fused rings may generally be 4-, 5-, 6-, 7-, or 8-membered rings, more usually 5- or 6-membered rings.
  • the core ring system is most usually fused 5- and 6-membered rings.
  • the fused ring atoms may generally be any atom, usually carbon or hetero atoms, more usually carbon and nitrogen group atoms, and more usually still carbon and nitrogen.
  • the number of carbon atoms in the core ring system is usually 7.
  • the core ring system usually contains 2 hetero atoms, where the preferred hetero atom is nitrogen.
  • one or more of the fused rings may be aromatic.
  • the core ring system is fused 5- and 6-membered rings, the core ring system is usually aromatic over both fused rings.
  • the fused 5- and 6-membered ring system is most usually an indazole.
  • lonidamine analogs for use according to the methods of the invention include analogs of the formula
  • R 1 , R 2 , X, Y, and n are generally as above or, in a preferred version,
  • the lonidamine analog is a 1 ,3-substituted- indazole, such as a 1-halobenzyM H-indazole.
  • the lonidamine analog is a 3-substituted 1-benzyl-1H-indazole.
  • the lonidamine analog is a i-substituted-indazole-3-carboxylic acid, such as a 1- halobenzyl-1 H-indazole-3-carboxylic acid.
  • Bioisosteres include bioisosteres and pharmacophores of lonidamine and analogs described herein.
  • Bioisosterism is a well-known tool for predicting the biological activity of compounds, based upon the premise that compounds with similar size, shape, and electron density can have similar biological activity.
  • To form a bioisostere of a given molecule one replaces one or more atoms or groups with known bioisosteric replacements for that atom or group.
  • lonidamine analogs that may be used in the methods of the invention can generally be any pharmacophore of lonidamine and the lonidamine analogs described above. Often a reasonable quantitative prediction of the binding ability of a known molecule can be made based on the spatial arrangement of a small number of atoms or functional groups in the molecule. Such an arrangement is called a pharmacophore, and once the pharmacophore or pharmacophores in a molecule have been identified, this information can be used to identify other molecules containing the same or similar pharmacophores.
  • Such methods are well known to persons of ordinary skill in the art of medicinal chemistry, and as the structural information described herein identifies the pharmacophore of lonidamine and the lonidamine analogs relevant to treatment of BPH, those of skill in the art can identify other LND analogs that comprise the pharmacophore and so are useful in treating BPH.
  • An example of programs available to perform pharmacophore-related searches is the program 3D Pharmacophore search from the Chemical Computing Group (see http://www.chemcomp.com/ fdept/prodinfo.htm).
  • a lonidamine analog of particular interest is tolnidamine (1-(4-chloro-2- methylbenzyl)-1H-indazole-3-carboxylic acid, AF 1923); see Ansari et al., 1998; Corsi et al., 1976.
  • Tolnidamine (TND) differs from lonidamine by the presence of a methyl substituent, rather than a chlorine substituent, in position 2 of the benzyl group.
  • TTD Tolnidamine
  • Other analogs of lonidamine with biological activity have been described in the following publications: U.S. Patent No.
  • Lonidamine analogs suited for use in the invention are those that interfere with cellular energy metabolism of prostate epithelial cells when administered to a human, non-human primate, or other mammal.
  • a compound e.g., lonidamine
  • Active forms can be identified by routine screening of analogs for the activity of the parent compound.
  • a variety of assays and tests can be used to assess pharmacological activity of lonidamine analogs, including in vitro assays, such as those described below and elsewhere herein, in vivo assays of prostate function (including citrate production and ATP production) in humans, non-human primates and other mammals, in vivo assays of prostate size in humans, non-human primates and other mammals, and/or clinical studies.
  • Apoptosis assay in cell lines Lonidamine induces apoptosis in cell lines derived from human prostate cells. The induction of apoptosis is significantly greater in LNCaP cells (ATCC NO. CLR-1740), a prostate-derived cell line that is citrate-producing, than in PC3 cells (ATCC NO. CLR-1435), a prostate-derived cell line that is citrate-oxidizing, consistent with the susceptibility of the citrate-producing prostate cells to metabolic inhibitors such as lonidamine.
  • an analog with similar apoptosis-inducing activity is selected.
  • a lonidamine analog that induces apoptosis (enhances caspase 3 activity) in citrate-producing prostate cells, such as LNCaP cells is administered to treat BPH.
  • a lonidamine analog that induces apoptosis in LNCaP cells to a significantly greater degree than in PC3 cells is administered to treat BPH.
  • the induction of apoptosis by the lonidamine analog is at least about 2-fold greater in LNCaP cells than in PC3 cells (and sometimes at least about 3-fold greater, at least about 4-fold greater, or at least about 10-fold greater) when assayed at the concentration of analog at which the difference in the level of apoptosis in the two cell lines is greatest (provided that the concentration of analog used in the assay is not greater than 1 mM).
  • apoptosis is significantly greater in primary cultures of prostate epithelial cells than in primary cultures of human prostate stromal cells, consistent with the susceptibility of citrate-producing prostate cells to metabolic inhibitors such as lonidamine.
  • a lonidamine analog in which a lonidamine analog is administered for treatment or prevention of BPH or its manifestations, an analog with apoptosis- inducing activity similar to that of lonidamine is selected.
  • a lonidamine analog that induces apoptosis in prostate epithelial cells is administered to treat BPH.
  • a lonidamine analog that induces apoptosis in primary cultures of prostate epithelial cells to a significantly greater degree than in primary cultures of human prostate stromal cells is used. In some embodiments of the invention, the lonidamine analog does not significantly induce apoptosis in stromal cells.
  • induction of apoptosis by the lonidamine analog is at least 2-fold greater in epithelial cells than in stromal cells (and sometimes at least 4- fold greater, sometimes at 10-fold greater, and sometimes at least 20-fold greater) when assayed at the concentration of analog at which the difference in the level of apoptosis in the two cell lines is greatest (provided that the concentration of analog used in the assay is not greater than 1 mM).
  • HIF-1 alpha expression assays Lonidamine reduced HIF-1 alpha expression/accumulation (measured in the nuclear fraction) in cells cultured under conditions of hypoxia by almost 2-fold at 200 micromolar and by more than 5 fold and more than 10-fold at higher lonidamine concentrations.
  • a lonidamine analog reduces HIF-1 alpha expression (prevents HIF-1 alpha accumulation) in LNCaP cells cultured under hypoxic conditions by at least about 2-fold, at least about 5-fold or at least about 10-fold compared to culture in the absence of such lonidamine analog.
  • some lonidamine analogs useful in the present invention have hexokinase inhibitory activity as great or greater than that of lonidamine.
  • Assays for hexokinase activity are known in the art. See Fanciulli et al., 1996, and Floridi et al., 1981.
  • Antispermatogenic activity Likewise, it is believed that the antispermatogenic activity of lonidamine results, at least in part, from energolytic (inhibition of energy production) effects in germ cells.
  • Some lonidamine analogs useful in the present invention have antispermatogenic activity as great, or greater, than that of lonidamine.
  • Assays for antispermatogenic activity are known in the art. See, e.g., Grima et al., 2001; Lohiya et al., 1991.
  • lonidamine analogs can be evaluated in vivo for suitability for use in the methods of the invention.
  • suitable assays include measurements of prostate function and activity.
  • In vivo measurements of prostate function The effect of a compound on prostate function, and, in particular, on respiration, can be assessed by monitoring prostate tissue metabolism following administration of the compound.
  • Some lonidamine analogs useful in the present invention will detectably reduce ATP 1 citrate, and/or lactate production by the prostate in animals (including humans, non- human primates and other mammals).
  • ATP, citrate, and/or lactate levels can be monitored directly and/or indirectly in vivo using techniques of magnetic resonance spectroscopy (MRS) or other methods.
  • MRS magnetic resonance spectroscopy
  • prostate size In vivo measurements of prostate size.
  • the effect of a compound on prostate size can be assessed following administration of the compound using standard methods (for example, ultrasonography or digital rectal examination, for humans, and ultrasonography and/or comparison of organ weight in animals).
  • Assays can be conducted in humans or, more usually, in healthy non-human animals such as in the monkey, dog, rat, or other animal models of BPH (see, Jeyaraj et a/., 2000; Lee et al., 1998; Mariotti et al., 1982), Some lonidamine analogs useful in the present invention will detectably reduce prostate size in such assays and animal models.
  • Clinical trials such as that described for lonidamine in the Example, infra, can be used to assess the therapeutic effects of lonidamine analogs.
  • lonidamine analog of interest in any of the aforementioned assays, models, and trials can be compared with that of lonidamine to provide guidance concerning dosage schedules for the compound, and other information.
  • lonidamine analogs with greater biological activity per mg than lonidamine are of special interest.
  • Some lonidamine analogs of interest have a longer in vivo half-life and/or greater bioavailability than lonidamine.
  • Lonidamine and lonidamine analogs and derivatives can be prepared using well known synthetic methods and this disclosure. Synthesis of lonidamine is described in U.S. Patent No. 3,895,026 and Germany Patent No. 2,310,031. Synthesis of exemplary lonidamine analogs, including tolnidamine (TND), is described in the art (see, e.g., Corsi et al., 1976, "1-Halobenzyl-1H-lndazole-3- Carboxylic Acids.
  • lonidamine analogs useful in the practice of the invention are not limited to those for which specific structures are provided in this disclosure or the cited references, and that the compounds described above are provided for illustration and not to limit the present invention.
  • Lonidamine analogs useful in the methods of the present invention are not limited to those now described herein or elsewhere in the pharmaceutical and patent literature; the ordinarily skilled practitioner guided by the present disclosure can synthesize novel analogs suitable for use according to the present invention using routine methods of medicinal chemistry and this disclosure.
  • Exemplary methods for synthesis of lonidamine and analogs (indazole derivatives) useful in the practice of the present invention are described in U.S. provisional application No. 60/576968 (filed June 20, 2004) and U.S. provisional application No. 60/588,694 (filed July 15, 2004), both incorporated herein by reference.
  • lonidamine or a lonidamine analog is provided in the form of a pharmaceutically acceptable salt.
  • Pharmaceutically acceptable salts include addition salts with acids, as well as the salts with bases.
  • Salts with bases are, for example, alkali metal or alkaline earth metal salts, such as sodium, potassium, calcium or magnesium salts, or ammonium salts, such as those with ammonia or suitable organic amines, e.g. diethylamine, di-(2-hydroxyethyl)-amine or tri-(2-hydroxyethyl)-amine.
  • Suitable acids for the formation of acid addition salts are, for example, mineral acids, such as hydrochloric, hydrobromic, sulphuric or phosphoric acid, or organic acids, such as organic sulphonic acids, for example, benzenesulphonic, 4-toluenesulphonic or methanesulphonic acid, and organic carboxylic acids, such as acetic, lactic, palmitic, stearic, malic, maleic, fumaric, tartaric, ascorbic or citric acid.
  • mineral acids such as hydrochloric, hydrobromic, sulphuric or phosphoric acid
  • organic acids such as organic sulphonic acids, for example, benzenesulphonic, 4-toluenesulphonic or methanesulphonic acid
  • organic carboxylic acids such as acetic, lactic, palmitic, stearic, malic, maleic, fumaric, tartaric, ascorbic or citric acid.
  • ester, amide and prodrug derivatives of lonidamine and analogs is also contemplated in the practice of the present invention (see, e.g., U.S. Pat. No. 6,146,658, for general information regarding preparation of such derivatives from a compound of interest) as is administration of polymorphic forms, enantiomeric forms, tautomeric forms, solvates, hydrates, and the like.
  • Exemplary prodrug forms of lonidamine and lonidamine analogs are described in U.S. provisional application no. 60/257026, entitled “Tertiary amine prodrugs of lonidamine and analogs" (filed July 8, 2004), incorporated herein by reference. 5.
  • lonidamine or a lonidamine analog is administered to a subject in need of treatment for BPH.
  • the subject in need of treatment is a human male who does not have cancer.
  • a subject in need of treatment for BPH is a man diagnosed with BPH. BPH can be diagnosed using art-known methods and criteria. The most common test is the digital rectal examination in which a physician determines whether the prostate is of a normal size and firmness.
  • diagnostic assays include a urine flow rate test, determination of post void residual urine volume (e.g., by palpitation of the abdomen, drainage of residual urine, x-ray urogramography, or ultrasonography), moderate or severe symptom scores on the American Urologic Association Symptom Index (AUASI; Barry et al., 1992) or International Prostate Symptom Score (IPSS; Barry et a/., 2001), and other tests known in the art.
  • AUASI American Urologic Association Symptom Index
  • IPSS International Prostate Symptom Score
  • Desired clinical results of treatment for BPH include, but are not limited to, alleviation or amelioration of one or more symptoms of BPH (see below), a reduction in prostate size (see below), a reduction in AUASI or IPSS scores compared to baseline measurements prior to commencement of therapy (for example, by 3 points or more, such as by 5 points or more), AUASI or IPSS scores less than 8, a reduction in serum PSA by at least about 20% (as discussed below, a transient rise in serum PSA can occur upon initial administration of lonidamine or a lonidamine analog), such as by at least about 40%, a serum PSA less than 4, such as less than 2, improvement in urodynamic parameters, and other desired results that will be recognized by a treating physician as indicative of a reduction in severity of BPH in a subject.
  • An assessment of the response to treatment can be made at any time following the first administration of the drug. For example, an assessment is made about 30 days, about 60 days, or about 90 days after beginning treatment. Alternatively, assessment can be made about 6, 12, 18, 24 or more months after beginning treatment. Alternatively, an assessment can be made less than about 30 days, about 30 days, about 60 days, or about 90 days after a course of treatment ends.
  • lonidamine or a lonidamine analog is administered to a human subject exhibiting a symptom associated with BPH to reduce the frequency or severity of the symptom.
  • a symptom associated with BPH refers to any one or more of the following symptoms: (1) urinary urgency; (2) terminal dribbling of urine; (3) frequent urination; (4) nocturia; (5) a weak/slow stream of urine; (6) a sense of incomplete emptying; (7) intermittency; (8) straining; (9) dysuria; (10) hematuria; (11) acute urinary retention; (12) urinary tract infection; (13) incontinence.
  • lonidamine or a lonidamine analog is administered to treat erectile dysfunction.
  • Administration of lonidamine or a lonidamine analog according to the methods of the invention typically results in a reduction in severity, or elimination, of one or more of these symptoms; usually results in either a reduction in severity of, or elimination of, all of these symptoms; and may result in elimination of all of these symptoms.
  • lonidamine or a lonidamine analog is administered to reduce prostate size in a human subject in need of such reduction.
  • a subject in need of reduction of prostate size is a man having an enlarged prostate gland as determined by (1 ) imaging (e.g., ultrasonography, magnetic resonance imaging) or (2) digital rectal examination or (3) one or more signs or symptoms resulting directly or indirectly from compression of the urethra by the prostate (e.g., including the symptoms of BPH discussed herein).
  • a reduction in serum PSA prostate specific antigen
  • enlarged prostates often exceed 30 grams, 40 grams, or 50 grams in size.
  • the degree of reduction of prostate size will vary from subject to subject due to a number of factors, including the degree of enlargement at the time of onset of therapy, but will typically be a reduction of at least about 10% volume, more often at least about 25%, sometimes at least about 40%, and sometimes an even greater than 40% reduction in prostate size is observed. This reduction can be determined by imaging or other methods. Serum PSA can also in some instances serve as a useful proxy for prostate volume. [0057] In a related aspect, lonidamine or an analog is administered to a subject with a serum PSA level greater than 2 ng/ml. PSA is secreted only by the epithelial cells of the prostate.
  • lonidamine treatment can provide a therapeutic benefit to subjects with PSA levels greater than 2 ng/ml.
  • subjects predicted to benefit significantly from treatment in accordance with the invention can be selected in a population of men with BPH by identifying subjects with a serum PSA value greater than 2 ng/ml.
  • the subject has a PSA level greater than about 4 ng/ml. Because higher PSA levels are also, and perhaps more closely, associated with prostate cancer than with BPH, in one embodiment, the subject selected for therapy with lonidamine or an analog has a PSA level less than about 10 ng/ml.
  • lonidamine or a lonidamine analog is administered to a subject who would benefit from prophylaxis of BPH.
  • a subject who would benefit from prophylaxis of BPH is a man previously treated for BPH by surgery, transurethral microwave thermotherapy, transurethral needle ablation, transurethral electrovaporization, laser therapy, balloon dilatation, prostatic urethral stent, drug therapy, or other therapy and not currently diagnosed with or exhibiting symptoms of BPH.
  • a subject who would benefit from prophylaxis of BPH is a man at increased risk for developing BPH due to age (e.g., men older than 40, older than 50, older than 60, or older than 70 years of age).
  • a subject who would benefit from prophylaxis of BPH is a man who is asymptomatic, or has symptoms sufficiently mild so that no clear diagnosis of BPH can be made, but who has an elevated serum PSA level (e.g., PSA > 2 ng/ml or, in some cases, >4 ng/ml).
  • the subject to whom lonidamine is administered in accordance with the methods of the invention is a man who has previously been treated for BPH, while in other cases the subject is a man who has not previously been treated for BPH.
  • any references herein to administration of lonidamine apply equally to administration of a biologically active lonidamine analog.
  • the subject in need of treatment or prophylaxis for BPH either is not also under treatment for cancer or does not have cancer.
  • the subject in need of treatment or prophylaxis for BPH has not been diagnosed as having cancer.
  • the subject in need of treatment or prophylaxis for BPH does not have cancer.
  • the subject in need of treatment has a cancer other than prostate cancer but does not have prostate cancer.
  • cancer has its ordinary medical meaning and refers to a malignancy (including head, neck, prostate and breast cancers, leukemias and lymphomas), generally characterized by clonality, autonomy, anaplasia, and metastasis (see Mendelsohn, 1991).
  • the invention provides a method of treating BPH in a patient by administering lonidamine to the patient.
  • the invention provides a method for treating BPH comprising (a) administering lonidamine to a patient diagnosed with BPH and (b) determining whether one or more manifestations of BPH are reduced in the patient.
  • the invention provides a method for treating BPH by (a) diagnosing BPH in a patient, (b) administering lonidamine to the patient and (c) determining whether one or more manifestations of BPH are reduced in said patient.
  • the invention provides a method of treating BPH in a patient by administering a lonidamine analog to the patient.
  • the invention provides a method for treating BPH comprising (a) administering a lonidamine analog to a patient diagnosed with BPH and (b) determining whether one or more manifestations of BPH are reduced in the patient. In one embodiment, the invention provides a method for treating BPH by
  • the subject is not diagnosed with or under treatment for cancer; optionally has a PSA less than or equal to 2 ng/ml; or optionally has a PSA greater than 2 ng/ml and less than 10 ng/ml.
  • the invention provides a method entailing (a) advertising the use of lonidamine, or a lonidamine analog, for treatment of BPH, and
  • step (b) selling lonidamine or a lonidamine analog to individuals for use in the treatment of BPH.
  • the advertising makes reference to a trademark that identifies a lonidamine (or lonidamine analog) product and the lonidamine (or analog) sold in step (b) is identified by the same trademark.
  • the individuals to whom lonidamine is sold include corporate persons (corporations) and the like and "selling BPH to individuals for use for treatment of BPH" includes selling to, for example, a medical facility for distribution to patients for treatment of BPH.
  • the invention provides a method of treating acute urinary retention in a human by administering lonidamine or a lonidamine analog to the human in need of such treatment. Because acute urinary retention can be a symptom of BPH, this embodiment of the invention is applicable to any subject who suffers from acute urinary retention but has not been diagnosed as having BPH when lonidamine or a Ionidamine analog is first administered.
  • a variety of routes and dosage schedules are appropriate for administration of lonidamine and lonidamine analogs according to the invention.
  • a preferred mode of delivery of lonidamine and lonidamine analogs to a patient is oral delivery.
  • Preferred dosage forms for oral administration are pills, tablets, capsules, caplets, and the like, especially as formulated for sustained release.
  • Other suitable forms for oral administration include troches, elixirs, suspensions, syrups, wafers, lozenges, and the like.
  • Lonidamine and lonidamine analogs may be formulated in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration.
  • the dosage form is the 150 mg unit dosage form marketed for the treatment of cancer by ACRAF in Italy under the tradename Doridamina.
  • Doridamina contains lonidamine 150 mg; starch 87 mg; microgranular cellulose 55 mg; lactose 40 mg; sodium carboxymethyl starch 15 mg; hydroxypropyl methylcellulose 11 mg; precipitated silica 4.5 mg; magnesium stearate 3.5 mg; titanium dioxide 0.4 mg; polyethylene glycol 400 0.4 mg.
  • Another formulation is 150 mg lonidamine (intra-granular), 87 mg cornstarch (intra-granular), 40 mg lactose (intra-granular), 3 mg colloidal silicon dioxide (intra-granular), 1.5 mg colloidal silicon dioxide (extra-granular), 55 mg microcrystalline cellulose (extra-granular), 15 mg sodium starch glycolate (extra-granular), 3.5 mg magnesium stearate (extra- granular), 9 mg hydroxypropyl methylcellulose (extra-granular), 10.9 mg OPADRY white (coating).
  • the dose, schedule and duration of administration of lonidamine and lonidamine analogs will depend on a variety of factors, including the age, weight and health of the subject, the severity of BPH symptoms, if any, the subject's medical history, co-treatments, therapeutic goal (e.g., therapy or prophalaxis), the mode of administration of the drug, the formulation used, patient response to the drug, and the like.
  • therapeutic goal e.g., therapy or prophalaxis
  • three general categories of dosing for administration of lonidamine and lonidamine analogs can be described: high dosing, low dosing, and intermediate dosing.
  • the standard lonidamine dose used for the treatment of cancer for which lonidamine has been approved in a few countries in Europe is 150 mg po TID for about thirty days or longer.
  • Low dosing Low dosing is contemplated for the treatment and prophalaxis of BPH.
  • Exemplary low doses of lonidamine or a lonidamine analog include, without limitation, doses in the range of 1-300 mg per day (total daily dosage), more often in the range of 5-300 mg/day, or sometimes in the range of 5-70 mg/day.
  • exemplary low dose ranges include 1-25 mg/day, 20-45 mg/day, 40- 65 mg/day, 40-70 mg/day, 50-100 mg/day, 80-100 mg/day, 50-200 mg/day, and 50- 300 mg/day.
  • the low dose is 150 mg administered orally once per day; the Doridamina unit dose form can be used in this embodiment.
  • the low dose is 75 mg administered orally once or twice daily.
  • the Doridamina unit dose form can be used in this embodiment by splitting it into two equal parts.
  • the daily dosages recommended herein may be divided for, for example, two-, three- or four-times per day administration.
  • the drug is formulated for administration once per day.
  • the drug is formulated for administration less frequently than once per day.
  • a modified-release form of the drug is used.
  • Administration of low doses of lonidamine can be daily, every other day, five days on, two days off, and other schedules selected in accordance with the teachings herein by the administering physician.
  • An advantage of low dose schedules of the invention is that this dose may be continued to be administered for weeks to months while limiting or eliminating the unwanted, albeit usually mild, side effects reported for higher doses of lonidamine (principally myalgia and testicular pain).
  • a low dose schedule can be used for therapy or prophalaxis.
  • a low dose form is used for a maintenance dose after a higher initial, priming or loading dose.
  • BPH is treated in accordance with the methods of the invention by administering to a BPH patient a higher dose of lonidamine or a lonidamine analog (usually for a shorter period of time than for low dosing).
  • exemplary high doses include, without limitation, total daily doses greater than 0.5 g, such as doses in the range 0.5 - 5 g/day, 0.5 - 3 g/day, 0.5 - 1 g/day and 1-3 g/day, or higher doses.
  • the daily dosages may be divided, for example, for two-, three- or four-time per day administration.
  • the drug is formulated for administration once-per day, or less frequently than once per day.
  • a modified-release form of the drug is used.
  • a high dose can be administered on a one-time, once-a-week, once every two weeks, or once-a-month basis (e.g., 0.5 - 5 g/administration) or by other schedules selected in accordance with the teachings herein by the administering physician.
  • a high dose schedule can be used for therapy or prophalaxis.
  • a high dose is administered in combination with, or following, a surgical or other non-drug treatment for BPH.
  • BPH is treated in accordance with the methods of the invention by administering lonidamine or a lonidamine analog to a BPH patient at a dose intermediate between a high dose and a low dose.
  • exemplary intermediate doses include, without limitation, doses greater than 300 and less than 500 mg/day, such as doses in the range >300-400 or 400 ⁇ 500 (e.g., 450 mg/day).
  • the daily dosages may be divided, for example, for two-, three- or four-times per day administration.
  • the drug is formulated for administration once-per day or less frequently than once per day.
  • a modified-release form of the drug is used.
  • this intermediate dose can be administered on a one-time, once-a-week, once every two weeks, or once-a-month basis (e.g., 300 - 500 mg/administration) or by other schedules to be determined by the administering physician.
  • the daily dosage is 150 mg of lonidamine or a lonidamine analog taken three times a day.
  • 150 mg of lonidamine or a lonidamine analog is administered one time per day for 28 days.
  • lonidamine or a lonidamine analog is administered at a dose of 5 mg, 25 mg, 50 mg, 75 mg or 100 mg per day for 4 to 26 weeks or longer.
  • An intermediate dose schedule may be used for therapy or prophylaxis.
  • an intermediate dose is administered in combination with, or following, a surgical or other non-drug treatment for BPH.
  • these dosing schedules are for illustration and not limitation, and that a dosing schedule may change during a course of therapy based on, for example, a patient's response to the therapy or the use of a lonidamine analog that has an activity/dose profile significantly different from that of lonidamine.
  • lonidamine or the lonidamine analog can be administered once or many times over periods as long as several months or years.
  • lonidamine or an analog is administered to a symptomatic (e.g., experiencing difficulty in urination) BPH patient only until the symptoms abate or disappear, and then treatment is stopped unless and until symptoms reappear. When symptoms reappear, administration of lonidamine or an analog is resumed.
  • treatment continues after symptoms disappear or are reduced to an acceptable target level, at least for a period of time, such as a week, two weeks, a month or several months.
  • the drug is administered to an asymptomatic subject to prevent the development or reoccurrence of symptoms (i.e., prophylactically administered).
  • Lonidamine and lonidamine analogs can be administered to a BPH patient in combination with other agents or procedures intended to treat BPH, ameliorate symptoms of BPH, potentiate the effects of the lonidamine or lonidamine analog, or provide other therapeutic benefit.
  • Administration of an agent "in combination with” includes parallel administration (administration of both agents to the patient over a period of time, such as administration of lonidamine and tamsulosin on alternate days for one month), co-administration (in which the agents are administered at approximately the same time, e.g., within about a few minutes to a few hours of one another), and co-formulation (in which the agents are combined or compounded into a single dosage form suitable for oral, parenteral, or other administration).
  • Exemplary agents for administration in combination with lonidamine or lonidamine analogs include, but are not limited to, zinc, alpha-blockers, 5-alpha- reductase inhibitors, treatments for urinary incontinence, and plant extracts.
  • agents for administration in combination with lonidamine or lonidamine analogs include other metabolic inhibitors, including but not limited to other hexokinase inhibitors and other inhibitors of glycolysis, including but not limited to 2-deoxy-D- glucose and an inhibitor, direct or indirect, of HIF-1alpha.
  • Zinc As discussed above, high concentrations of zinc in the secretory epithelial cells of the prostate inhibit m-aconitase, increasing the dependence of that tissue on glycolysis for energy production.
  • zinc e.g., zinc chloride, zinc gluconate, zinc sulfate, zinc acetate, zinc aspartate, zinc citrate, zinc glycerate, zinc oxide, zinc picolinate, and the like
  • a drug composition of the invention to maximize the efficacy of the treatment.
  • zinc e.g., zinc chloride, zinc gluconate, zinc sulfate, zinc acetate, zinc aspartate, zinc citrate, zinc glycerate, zinc oxide, zinc picolinate, and the like
  • 15-300 mg/day zinc can be administered for this purpose; typically 30-50 mg/day is administered.
  • Alpha-Adrenerqic-Blockers alleviate some symptoms of BPH, without curing the underlying disease. These agents work by relaxing the muscles at the neck of the bladder and in the prostate, reducing the pressure on the urethra.
  • Exemplary alpha-blockers include doxazosin (Cardura), terazosin (Hytrin), tamsulosin (Flomax), alfuzosin (Xatral), and prazosin (Hypovase).
  • an alpha blocker is administered in combination with lonidamine or a lonidamine analog to treat BPH.
  • the alpha- blocker is administered at a lower dosage (amount) or less frequently (e.g., alternate days rather than daily) than the "standard" dosage (the dosage that would be indicated for the subject in the absence of lonidamine administration) in combination with lonidamine or a lonidamine analog.
  • 5-Alpha-Reductase Inhibitors inhibit the conversion of testosterone to dihydrotestosterone 2 (DHT), an androgen that contributes to prostate enlargement.
  • An exemplary 5-alpha-reductase inhibitor is finasteride (Proscar).
  • Another exemplary 5-alpha-reductase inhibitor is Avodart® (dutasteride).
  • a 5-alpha-reductase inhibitor is administered in combination with lonidamine or an analog to treat BPH.
  • the 5-alpha-reductase inhibitor is administered at a lower dosage (amount) or less frequently (e.g., alternate days rather than daily) than the "standard" dosage (the dosage that would be indicated for the subject in the absence of lonidamine administration) in combination with lonidamine (or a lonidamine analog).
  • a lower dosage or less frequently (e.g., alternate days rather than daily) than the "standard" dosage (the dosage that would be indicated for the subject in the absence of lonidamine administration) in combination with lonidamine (or a lonidamine analog).
  • Treatments for urinary incontinence include alpha-adrenergic agonists such as pseuodoephedrine hydrochloride (Sudafed), and phenylpropanolamine; anticholinergics such as dicyclomine hydrochloride (Bentyl), and hyoscyamine sulfate (Levsin SL, Levsin, Levsinex, Cystospaz-M, Levbid); and antispasmodic drugs including darifenacin (Enablex), solifenacin succinate (VESIcare), oxybutynin (Ditropin), tolterodine L- tartrate (Detrol and Detrol LA), and trospium (Sanctura).
  • alpha-adrenergic agonists such as pseuodoephedrine hydrochloride (Sudafed), and phenylpropanolamine
  • anticholinergics such as
  • Glycolytic and Mitochondrial Function Inhibitors are known in the art, and include those described in PCT patent publications WO 01/82926 published 8 November 2001 ; U.S. Patent Nos. 6,670,330; 6,218,435; 5,824,665; 5,652,273; and 5,643,883; U.S. patent application publication nos.
  • Such inhibitors can be administered in combination with lonidamine or lonidamine analogs for therapeutic benefit in the treatment of BPH.
  • Pygeum Africanum or an extract thereof can be administered in combination with lonidamine or lonidamine analogs for therapeutic benefit in the treatment of BPH.
  • lonidamine or a lonidamine analog may be administered in combination with, or prior to, procedures for treatment of BPH including surgery (transurethral resection of the prostate; transurethral incision of the prostate; or open prostatectomy), laser therapy, transurethral microwave thermotherapy, balloon dilatation, placement of a prostatic urethral stent, transurethral needle ablation, transurethral electrovaporization of the prostate, or other non-drug therapies.
  • Unit Dosage Forms The compounds used in the methods of the present invention are formulated in compositions suitable for therapeutic administration.
  • the methods of the invention are practiced with lonidamine in the unit dosage form marketed as Doridamina (by ACRAF) in Italy.
  • New dosage forms of lonidamine are also provided by the invention.
  • the present invention provides a unit dosage pharmaceutical formulation of lonidamine that is suitable for oral administration (including tablets, capsules, caplets, and pills) and contains, in various embodiments, an amount of lonidamine in a range bounded by a lower limit of (in mg) 1 , 5, 10, and 50 and an upper limit of 10, 20, 40, 50, 70 and 100 (where the higher limit is in mg and greater than the lower limit) and is especially convenient for certain low dose schedules.
  • the unit dosage form contains an amount of drug in a range bounded by a lower limit of (in mg) 200, 300, 500 or 1000 and an upper limit of 500, 1000, 3000 or 5000 (where the higher limit is in mg and greater than the lower limit) and is especially convenient for certain high dose schedules.
  • the formulation contains between 100 and 200 mg of compound (e.g., 150 mg), between 200 and 5000 mg, between 200 and 1000 mg, or between 500 and 1000 mg of the compound.
  • Lonidamine analogs can be similarly formulated. Dosage forms of particular interest contain 5, 25, 50, 100 or 150 mg lonidamine.
  • solid unit dosage forms of the invention generally include a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier refers to a solid or liquid filler, diluent, or encapsulating substance, including for example excipients, fillers, binders, and other components commonly used in pharmaceutical preparations, including, but not limited to, those described below.
  • Methods for formulation of drugs generally are well known in the art, and the descriptions herein are illustrative and not limiting. See, e.g., Ansel et al., 1999; Marshall, 1979.
  • Hydrophilic binders suitable for use in the formulations of the invention include copolyvidone (cross-linked polyvinylpyrrolidone), polyvinylpyrrolidone, polyethylene glycol, sucrose, dextrose, corn syrup, polysaccharides (including acacia, guar, and alginates), gelatin, and cellulose derivatives (including HPMC, HPC, and sodium carboxymethylcellulose).
  • Water-soluble diluents suitable for use in the formulations of the invention include sugars (lactose, sucrose, and dextrose), polysaccharides (dextrates and maltodextrin), polyols (mannitol, xylitol, and sorbitol), and cyclodextrins.
  • Non-water-soluble diluents suitable for use in the formulations of the invention include calcium phosphate, calcium sulfate, starches, modified starches, and microcrystalline cellulose.
  • Surfactants suitable for use in the formulations of the invention include ionic and non-ionic surfactants or wetting agents such as ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglycerides, sorbitan fatty acid esters, poloxamers, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene derivfatives, nonoglycerides or ethoxylated derivatives thereof, sodium lauryl sulfate, lecithins, alcohols, and phospholipids.
  • ionic and non-ionic surfactants or wetting agents such as ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglycerides, sorbitan fatty acid esters, poloxamers, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene derivfatives, nonoglycerides or ethoxylated derivatives thereof, sodium
  • Disintegrants suitable for use in the formulations of the invention include starches, clays, celluloses, alginates, gums, cross-linked polymers (PVP, sodium carboxymethyl-cellulose), sodium starch glycolate, low-substituted hydroxypropyl cellulose, and soy polysaccharides.
  • PVP cross-linked polymers
  • Preferred disintegrants include a modified cellulose gum such as cross-linked sodium carboxymethylcellulose.
  • Lubricants and glidants suitable for use in the formulations of the invention include talc, magnesium stearate, calcium stearate, stearic acid, colloidal silicon dioxide, magnesium carbonate, magnesium oxide, calcium silicate, microcrystalline cellulose, starches, mineral oil, waxes, glyceryl behenate, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, sodium lauryl sulfate, sodium stearyl fumarate, and hydrogenated vegetable oils.
  • Preferred lubricants include magnesium stearate and talc and combinations thereof.
  • the preferred range of total mass for the tablet or capsule may be from about 40 mg to 2 g, from about 100 mg to 1000 mg, and from about 300 mg to 750 mg.
  • the present invention provides unit dosage forms that are sustained release formulations of lonidamine or a lonidamine analog to allow once a day (or less) oral dosing, a frequency sometimes preferred by patients over multiple day dosing.
  • sustained release formulations including tablets, capsules, caplets and pills
  • Such sustained release formulations usually contain between 1 mg and 3 g of the active compound, with various alternative embodiments including those described above for conventional oral unit doses, such as an amount of drug in a range bounded by a lower limit of (in mg) 1 , 5, 10, and 50 and an upper limit of 10, 20, 40, 50, 70 and 100 (where the higher limit is in mg and greater than the lower limit) and are especially convenient for certain low dose or intermediate dose schedules.
  • the unit dosage form contains an amount of drug in a range bounded by a lower limit of (in mg) 200, 300, 500, 750 or 1000 and an upper limit of 500, 1000, 2000, 3000 or 5000 (where the higher limit is in mg and greater than the lower limit).
  • lonidamine or a lonidamine analog in the sustained release formulations is released over a period of time greater than 6 hours, e.g., greater than 12 hours, after administration.
  • the sustained release formulation allows once a day dosing to achieve a pharmacodynamic profile therapeutically equivalent to dosing 150 mg of lonidamine three times a day.
  • sustained-release formulations for other drugs that can be modified in accordance with the teachings herein to be useful in the present invention are well known in the art, and are, for example, described in U.S. Pat. Nos. 5,968,551 ; 5,266,331 ; 4,970,075; 5,549,912; 5,478,577; 5,472,712; 5,356,467; 5,286,493; 6,294,195; 6,143,353; 6,143,322; 6,129,933; 6,103,261 ; 6,077,533; 5,958,459; and 5,672,360.
  • sustained-release formulations are also discussed in the scientific literature, e.g., in ORAL SUSTAINED RELEASE FORMULATIONS: DESIGN AND EVALUATION, edited by A. Yacobi and E. Halperin-Walega, Pergamon Press, 1988, which describes a variety of types of sustained-release dosage forms and drug release mechanisms, for example single unit (e.g., matrix tablets, coated tablets, capsules), multiple unit (e.g., granules, beads, micro-capsules), inert, insoluble matrix, hydrophilic gel matrix (e.g., bioadhesive, erodible, non-erodible), and ion- exchange resin sustained-release dosage forms.
  • single unit e.g., matrix tablets, coated tablets, capsules
  • multiple unit e.g., granules, beads, micro-capsules
  • inert e.g., insoluble matrix, hydrophilic gel matrix (e.g., bioadhesive, erod
  • the present invention provides a method of treating BPH, by administering once daily to a patient in need of such treatment a sustained release tablet dosage form comprising a daily therapeutic dose of lonidamine from about 1 mg to 2 g in a hydrophilic matrix.
  • the matrix can be, for example and without limitation, selected from the group consisting of hydroxypropylmethyl cellulose (by weight percent of about 20-40%), lactose (5-15%), microcrystalline cellulose (4-6%), and silicon dioxide (1-5%) having an average particle size ranging from 1-10 microns, often ranging from 2-5 microns, and most often ranging from about 2-3 microns.
  • Illustrative preferred sustained release formulations of the invention include formulations A and B in Table 1.
  • HPMC Metal K15M, CR 8 30 .
  • Methyl cellulose (Methocel, K100L, CR) 18 0 Anydrous lactose 12.2 8.2
  • the sustained release formulations of the invention may be in the form of a compressed tablet containing an intimate mixture of lonidamine and a partially neutralized pH-dependent binder that controls the rate of drug dissolution in aqueous media across the range of pH in the stomach (typically ⁇ 2) and intestine (typically -5.5).
  • phthalic acid derivatives such as the phthalic acid derivatives of vinyl polymers and copyolymers, hydroxyalkylcellulose, alkylcelluloses, cellulose acetates, hydroxyalkylcellulose acetates, cellulose ethers, alkylcellulose acetates, and esters thereof, and polymers and copoloymers of lower alkyl acrylic acids and lower alkyl acrylates, and the partial esters thereof.
  • pH-dependent binder materials are methacrylic acid copolymers. Such a copolymer is commercially available from Rohm Pharma as EudragitTM L- 100-55 as a powder or L30D-55 as a 30% dispersion in water.
  • Other pH-dependent binder materials which may be used alone or in combination include hydroxypropyl cellulose phthalate, hydroxypropyl methylcellulose phthalate, cellulose acetate phthalate, polyvinylacetate phthalate, polyvinylpyrrolidone phthalate, and the like.
  • One or more pH-dependent binders are present in the sustained release oral dosage forms of the invention in an amount ranging from about 1 to 20 weight percent, or from 5 to 12 weight percent, or about 10%.
  • the pH-independent binders or viscosity enhancing agents contained in the sustained release formulations of the invention include substances such as hydroxypropyl methylcellulose, hydroxypropyl cellulose, methylcellulose, polyvinylpyrrolidone, neutral poly(meth)acrylate esters, and the like.
  • the pH- independent binders are in an amount ranging from 1 to 10 weight percent or from 1 to 3 weight percent, or about 2%.
  • the sustained release formulations of the invention also contain in some embodiments one or more pharmaceutical excipients intimately mixed with the lonidamine or lonidamine analog and the pH-dependent binder, such as pH- independent binders or film-forming agent, starch, gelatin, sugars, carboxymethylcellulose, and the like, as well as other useful pharmaceutical diluents such as lactose, mannitol, dry starch, micro-crystalline cellulose, and the like, and surface active agents such as polyoxyethylene sorbitan esters, sorbitan esters, and the like; and coloring agents and flavoring agents.
  • Lubricants such as talc and magnesium stearate and tableting aids can also be used.
  • the sustained release formulations of the invention include any of the commercially available polymers suitable for use in such formulations, including but not limited to cellulose, ethyl cellulose, methyl cellulose, carboxymethyl cellulose, hydroxypropylmethyl cellulose, hydroxypropylmethyl cellulose phthalate, hydroxypropyl cellulose, microcrystalline cellulose, sodium carboxymethyl cellulose, cellulose acetate phthalate, polyvinyl acetate phthalate, polyvinylpyrrolidone, polyethylene oxide, polyethylene glycol, zein, alginate, hypromellose phthalate, methacrylic acid copolymer, Crospovidone, silica aerogel, pregelatinized starch, corn starch, croscarmellose sodium, sodium starch glycolate, candelilla wax, paraffin wax, carnauba wax, montan glycol wax, white wax, Eudragit (polymethacrylic acid esters), Aquacoat (ethyl cellulose, cellulose acetate phthal
  • microencapsulation and coated granule systems in which particles of drug (or particles of drug and polymer) as small as 1 micron are coated in a polymeric membrane, including embodiments in which particles coated with polymers with different release characteristics are delivered together in a capsule;
  • solvent-activated systems including (i) osmotically controlled devices (e.g. OROS) in which an osmotic agent and the drug are encapsulated in a semi-permeable membrane, water is pulled into device due to osmotic gradient, and increased pressure drives drug out of device through a laser drilled hole; (ii) a hydrogel swelling system in which drug is dispersed in a polymer and/or a polymer is coated onto a particle of drug, and the polymer swells on contact with water (swelling is in some embodiments pH or enyzmatically controlled), allowing diffusion of drug out of the device; (iii) a microporous membrane system in which drug is encapsulated in a membrane that has a component that dissolves on contact with water (in some embodiments, dissolution is pH or enyzmatically controlled), leaving pores in the membrane through which the drug diffuses; and (iv) a wax matrix system in which the drug and an additional soluble component are disper
  • polymeric degradation systems including (i) bulk degradation, in which drug is dispersed in polymeric matrix, and degradation occurs throughout the polymeric structure in a random fashion, allowing drug release; and (ii) surface erosion, in which drug is dispersed in polymeric matrix and delivered as the surface of the polymer erodes.
  • the invention provides a method for treating BPH by administering a unit dose oral pharmaceutical composition that is a sustained- release formulation containing an effective amount of lonidamine, such as described above, once per day.
  • This example describes results of a Phase Il Open-Label Study to evaluate the efficacy and safety of oral lonidamine (LND) treatment in subjects with symptomatic benign prostatic hyperplasia (BPH) by measuring the change from baseline of: total prostate volume by transrectal ultrasound (TRUS), maximum flow rate (Q ma ⁇ ) on uroflowmetry, international prostatic symptoms score (I-PSS) and prostate-specific antigen (PSA).
  • TRUS transrectal ultrasound
  • Q ma ⁇ maximum flow rate
  • I-PSS international prostatic symptoms score
  • PSA prostate-specific antigen
  • Pretreatment evaluation included recording of medications taken 2 weeks prior to study entry; serum chemistry panel including sodium, potassium, chloride, carbon dioxide, blood urea nitrogen (BUN), creatinine, AST and ALT; hormonal profile (FSH, LH, PRL, testosterone, free testosterone); TRUS measurement of prostate volume; PSA; uroflowmetry; and I-PSS.
  • serum chemistry panel including sodium, potassium, chloride, carbon dioxide, blood urea nitrogen (BUN), creatinine, AST and ALT
  • hormonal profile FSH, LH, PRL, testosterone, free testosterone
  • the primary efficacy endpoints were the change from baseline to Day-28 in prostate volume, Q max , I-PSS and PSA. Efficacy was analyzed by paired t-tests of the percent change in prostate volume and PSA and absolute change in Q max and I- PSS from baseline. Non-parametric tests were also performed to examine the robustness of the results. All tests were two-sided and no adjustments were made for multiple tests. Missing data for Day-14 and Day-28 were replaced using the last observation carried forward procedure.
  • TBV total prostate volume
  • Table 10 shows the change in prostate volume compared to baseline in subjects treated with lonidamine.
  • Urine flow was measured at 0, 14, 28 and 56 days for all individuals treated with lonidamine .
  • Table 11 shows AUA and PSA scores for 8 individuals treated with lonidamine measured at baseline and 28 days.
  • WO 01/82926 Manipulation of Oxidative Phosphorylation for Hypersensitizing Tumor Cells to Glycolytic Inhibitors.

Abstract

A method for treatment or prophylaxis of benign prostatic hyperplasia by administration of lonidamine or a lonidamine analog is provided. Also provided are unit dosage forms of lonidamine and lonidamine analogs useful for such treatment and prophylaxis.

Description

Patent Application
TREATMENT OF BENIGN PROSTATIC HYPERPLASIA
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims benefit under 35 U.S.C. 119(e) of U.S. provisional application No. 60/592,833 (filed July 29, 2004) and provisional application No. 60/661 ,067 (filed March 11 , 2005) the entire contents of which are incorporated by reference herein.
FIELD OF THE INVENTION
[0002] The invention relates to treatment and prevention of benign prostatic hyperplasia, and has application in the field of medicine and related fields, including chemistry, medicinal chemistry, and molecular biology.
BACKGROUND OF THE INVENTION
[0003] Benign Prostatic Hyperplasia (BPH), a disease in which prostate epithelial cells grow abnormally and block urine flow, afflicts more than 10 million adult males in the United States and many millions more throughout the rest of the world. Until relatively recently, surgical intervention was the only treatment of the disease, and even today, surgery is the treatment of last resort, almost inevitably relied upon when other treatments are not, or cease to be, effective. Prostate surgery and recovery therefrom is painful, and the surgery itself may not be effective and poses the risk of serious side effects. For a recent review, see Barry, 2001 (full citations are provided below).
[0004] Only two classes of drugs are currently available to treat the symptoms of BPH. One class includes compounds that inhibit production of the active form of testosterone (dihyrdotestosterone or DHT). Use of drugs in this class can cause a loss of libido and loss of muscle mass and tone in males and is associated with an increased occurrence of high grade prostate cancer. In addition, this therapy is limited by the very long delay (months) between first administering the drug and significant reduction in prostate size in the patient. The second class of currently used drugs for BPH, alpha adrenergic blockers, acts by relaxing the smooth muscles, allowing urine to pass through the urethra more freely. While this class of drugs reduces symptoms more rapidly than the first, it does not reduce the size of the prostate or prevent it from growing larger, which can lead to eventual surgical intervention.
[0005] Thus, there is a significant, unmet need for drugs that can treat the underlying disease condition of BPH without serious side effects. The present invention meets that need.
SUMMARY OF THE INVENTION
[0006] The invention provides methods and compositions for treating BPH in a human subject by administration of lonidamine or a lonidamine analog to the subject. Pharmaceutical compositions useful for treatment of BPH, including sustained release formulations, are also provided. In one embodiment, the formulation is orally administered and permits once-a-day dosing of a therapeutically effective dose of the compound.
DETAILED DESCRIPTION OF THE INVENTION 1. Definitions
[0007] The following definitions are provided to assist the reader. Unless otherwise defined, all terms of art, notations and other scientific or medical terms or terminology used herein are intended to have the meanings commonly understood by those of skill in the chemical and medical arts. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not be construed to represent a substantial difference over the definition of the term as generally understood in the art.
[0008] As used herein, "treating" a condition or patient refers to taking steps to obtain beneficial or desired results, including clinical results, for the patient. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, alleviation or amelioration of one or more symptoms of BPH, diminishment of extent of disease, delay or slowing of disease progression, amelioration, palliation or stabilization of the disease state, and other beneficial results described below. [0009] As used herein, "reduction" of a symptom or symptoms (and grammatical equivalents of this phrase) means decrease of the severity or frequency of the symptom(s), or elimination of the symptom(s).
[0010] As used herein, "administering" or "administration of a drug to a subject (and grammatical equivalents of this phrase) includes both direct administration, including self-administration, and indirect administration, including the act of prescribing a drug. For example, as used herein, a physician who instructs a patient to self-administer a drug and/or provides a patient with a prescription for a drug is administering the drug to the patient.
[0011] As used herein, a "manifestation" of BPH refers to a symptom, sign, anatomical state (e.g., prostate size), physiological state (e.g., PSA level), or report (e.g., AUASI score) characteristic of a subject with BPH. [0012] As used herein, a "therapeutically effective amount" of a drug is an amount of a drug that, when administered to a subject with BPH, will have the intended therapeutic effect, e.g., alleviation, amelioration, palliation or elimination of one or more manifestations of BPH in the subject. The full therapeutic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses. Thus, a therapeutically effective amount may be administered in one or more administrations.
[0013] As used herein, a "prophylactically effective amount" of a drug is an amount of a drug that, when administered to a subject, will have the intended prophylactic effect, e.g., preventing or delaying the onset (or reoccurrence) of disease or symptoms, or reducing the likelihood of the onset (or reoccurrence) of disease or symptoms. The full prophylactic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses. Thus, a prophylactically effective amount may be administered in one or more administrations.
[0014] As used herein, "TID" and "QD" have their ordinary meanings of "three times a day" and "once daily," respectively.
[0015] As used herein, "alkyl" refers to a monovalent alkane (hydrocarbon) derived radical containing from 1 to 15 carbon atoms. It may be straight, branched or cyclic and may be unsubstituted or substituted with substituent groups including but not limited to hydroxyl, halide, alkoxyl, and nitrile. Alkoxy groups that can be used include but are not limited to methooxy. Illustrative straight or branched alkyl groups include methyl, ethyl, propyl, isopropyl, butyl and t-butyl. [0016] As used herein, "aryl" refers to moieties that include one or more monocyclic or fused ring aromatic systems. Such moieties include any moiety that includes one or more monocyclic or bicyclic fused ring aromatic systems, including but not limited to phenyl and naphthyl. Aryl groups may be unsubstituted or substituted with substituent groups as listed for the particular substituted aryl. [0017] As used herein, "heteroaryl" refers to monocyclic aromatic groups having 5 or 6 ring atoms, or fused ring bicyclic aromatic groups having 8 to 10 atoms, in which the ring atoms are C, O, S, SO, SO2, or N and at least one of the ring atoms is a heteroatom, i.e., O, S, SO, SO2, or N. Heteroaryl groups may be unsubstituted or substituted with substituent groups as listed for the particular substituted heteroaryl. Examples of monocyclic aromatic heteroaryl groups include but are not limited to pyridyl. Examples of bicyclic fused ring heteroaryl groups include but are not limited to indazolyl, pyrrolopyrymidinyl, indolizinyl, pyrazolopyridinyl, triazolopyridinyl, pyrazolopyrimidinyl, triazolopyrimidinyl, pyrrolotriazinyl, pyrazolotriazinyl, triazolotriazinyl, pyrazolotetrazinyl, hexaaza- indenly, and heptaaza-indenyl. Unless indicated otherwise, the arrangement of the hetero atoms within the ring may be any arrangement allowed by the bonding characteristics of the constituent ring atoms.
[0018] As used herein, the terms "heterocycloalkyl" and "heterocyclyl" refer to a monocyclic or fused ring multicyclic cycloalkyl group at least a portion of which is not aromatic and in which one or more of the carbon atoms in the ring system is replaced by a heteroatom selected from O, S, SO, SO2, or N. Examples of heterocyclyl groups include but are not limited to piperidinyl, morpholinyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydroimidazo [4,5-c] pyridinyl, imidazolinyl, piperazinyl, pyrrolidine-2-onyl, and piperidin-2-onyl.
[0019] As used herein, "cycloalkyl" refers to a monocyclic or fused ring multicyclic group at least a portion of which is not aromatic and in which the ring atoms are carbon.
[0020] As used herein "heterocycloalkenyl" refers to a monocyclic or fused ring multicyclic group in which one or more of the carbon ring atoms is replaced by a hetero atom, the ring system is at least partially not aromatic, and the ring system includes at least one carbon-carbon double bond.
2. Benign Prostatic Hyperplasia and the Effects of Lonidamine and Lonidamine Analogs
[0021] The present invention provides compositions and methods useful in the treatment of benign prostatic hyperplasia (BPH). In particular, the invention relates to the use of lonidamine (LND) for the treatment or prevention of BPH. Additionally, the invention relates to the use of lonidamine analogs for the treatment or prevention of BPH. To aid in understanding the invention, a brief discussion of BPH (also referred to as benign prostatic hyperplasia) and the properties of lonidamine and its bioactive analogs is provided below.
[0022] BPH involves overgrowth (hyperplasia) of cells in the prostate, resulting in enlargement of the prostate and leading to lower urinary tract symptoms and disease. The prostate gland contains secretory epithelial cells in a stroma of connective tissue and smooth muscle (see Barry, 2003, for a more detailed description of prostate anatomy), and BPH involves hyperplasia of the epithelial component. The secretory epithelial component in the normal prostate is remarkable in that the level of zinc in this tissue is very high compared to other normal tissues. A consequence of the high zinc levels is that, through a mechanism involving zinc inhibition of the enzyme m-aconitase, the generation of energy via the tricarboxylic acid (TCA) cycle and oxidative phosphorylation is substantially reduced in the secretory epithelium, making this tissue far more dependent than other organs and tissues upon glycolysis as an energy source. The zinc inhibition of m-aconitase, a key enzyme in the TCA cycle, results in at least a substantial reduction in, and perhaps a near complete blockade of, the TCA cycle in prostate epithelial cells. Another physiological result of the zinc-based inhibition of m-aconitase is the diversion of citrate from the TCA cycle, enabling the prostate to secret large quantities of citrate, used by the sperm as an energy source, into the seminal fluid. See, generally, Costello, 1999; Costello et al., 2000; Costello and Franklin, 2000. [0023] As other normal cells in the body do not accumulate zinc to a level inhibitory to the metabolism of citrate, prostate epithelial cells are uniquely dependent on glycolysis (anaerobic metabolism). The present invention relates in part to the discovery of these cells' susceptibility to the drug lonidamine, which allows lonidamine to be administered, as described herein, to treat or prevent BPH in humans. Lonidamine is the generic name for 1-(2,4-dichlorobenzyl)-1 H-indazole-3- carboxylic acid, and has also been referred to in the medical literature as 1-[(2,4- dichlorophenyl)methyl]-1H-indazole-3-carboxylic acid, AF1890, diclondazolic acid (DICA), and Doridamina™ (ACRAF; Rome, Italy). See Figure 1. Lonidamine was first identified as an antispermatogenic agent and subsequently used in the treatment of breast, cervical, lung and prostate cancers, in a few countries in Europe. See Silvestrini, 1981 ; Gatto et al., 2002. The mechanisms of action of lonidamine in spermatogenesis and cancer may not be completely understood. However, it has been suggested that lonidamine's anticancer properties result at least in part from a lonidamine-mediated disruption of the mitochondrial membrane, resulting in reduced activity of mitochondrially-bound hexokinase and interference with ATP production by the glycolytic pathway and oxidative phosphorylation. See, Floridi et al., 1981 , Fanciulli et al., 1996, and references numbered 15-22 therein; and Gatto, 2002. Also see Kaplan, 2000. Without intending to be bound by a specific mechanism for the effects of lonidamine in benign prostatic hyperplasia, it is believed that lonidamine inhibits glycolysis and/or impairs the already diminished mitochondrial function in prostate epithelial cells, starving these cells, relative to the normal cells in the body, of energy. Without intending to be bound by a specific mechanism, it is believed that, due to this energy deprivation, enough of the hyperplastic, epithelial cells are destroyed or otherwise reduced in size to reduce the size of the prostate and thereby relieve the condition and its clinical consequences. [0024] Accordingly, administration of lonidamine to a human subject diagnosed with, or exhibiting symptoms of, BPH provides benefits such as reduction of severity or frequency of one or more symptoms, reduction in prostate size or rate of enlargement, improvement in perceived quality of life, and reversion of other manifestations of BPH toward a more normal state. Further, administration of lonidamine to a human subject in need of prophylaxis for BPH provides benefits such as a reduction in likelihood that BPH or symptoms associated with BPH will appear, reappear or progress in the subject. Still further, administration of a lonidamine analog to a human subject is similarly effective for treatment and prophylaxis of BPH. In another embodiment, administration of lonidamine or its analogs to a human subject as described herein can be efficacious in the treatment of acute urinary retention. These and other aspects of the invention are discussed in greater detail below. Section 3, below, describes certain lonidamine analogs useful for treatment and prophylaxis of BPH. Section 4 relates to synthesis and forms of lonidamine and lonidamine analogs. Section 5 describes patient populations for whom administration of lonidamine and lonidamine analogs provides benefit. Section 6 describes methods of administration of lonidamine (e.g., dose, route, schedule and duration of administration). Section 7 describes combination therapies in which lonidamine or an analog is administered in combination with another drug or therapy. Section 8 describes exemplary dosage forms. Section 9 describes monitoring PSA levels in subjects treated with lonidamine or a lonidamine analog. Section 10 provides examples of the use and effects of lonidamine. The description below is organized into sections for convenience only, and disclosure found in any organizational section is applicable to any aspect of the invention.
3. Lonidamine Analogs
[0025] As noted above, in addition to lonidamine, a variety of compounds related to lonidamine are useful for the treatment and prevention of BPH. Useful compounds are generally structurally similar to, are bioisosteres of, or are pharmacophores of lonidamine, as described below, and have biological activity(s) similar to those of lonidamine, as also discussed below. Such compounds can be referred as "bioactive lonidamine analogs," "lonidamine analogs," or, in some cases, simply, "analogs."
[0026] Structural characteristics of lonidamine analogs. Based, in part, on the structure of lonidamine and related compounds known to have pharmaceutical activities similar to that of lonidamine, certain lonidamine analogs, including novel analogs provided by the present invention, suitable for use in treatment or prophalaxis of BPH are described by the formula,
Figure imgf000008_0001
( Ar j where R1, R2, X, Y, n and w1 are defined below: [0027] Ri represents -COOH or a derivative or bioisostere of the -COOH group. Ri is usually selected from an acid group of formula -COOH; an amide of formula -CONR3R4, where R3 and R4 may be independently alkyl or hydrogen, with hydrogen preferred; a hydrazide of formula -CONHNR6Rr, where R6 and R7 are usually -H or -CH3; a substituted ester of formula -COOR5, with R5 being a residue easily hydrolyzed in the subject after administration and generally a straight chain or branched chain alkyl group substituted with one or more hydroxyl groups, more usually a straight chain or branched chain methyl, ethyl, or propyl group substituted with one or more hydroxyl groups, more usually still an ethyl group substituted with one hydroxyl group or a straight chain or branched chain propyl group substituted with two hydroxyl groups, and most usually -CH2CH2OH, -CH2CH(OH)CH2OH, or - CH2(CH2OH)2. Ri may also be the carboxylate anion of formula -COO", in which case the lonidamine or lonidamine analog will be associated with a counter ion, Z+, where Z+ is a pharmaceutically acceptable cation.
[0028] R2 represents a substituted or unsubstituted aryl or heteroaryl group.
Usually, R2 is a substituted aryl group; more usually, a substituted phenyl group; more usually still, a phenyl group substituted by one, two, or three substituents independently selected from halo and alkyl substituents, particularly -Cl, -Br, -I, CF3 and -CH3 substituents. When R2 is a substituted phenyl group, R2 is usually -Cl, -Br, -I, CF3 Or -CH3, monosubstituted phenyl, substituted at the 2 ,3, or 4 position; dichloro, dibromo, dimethyl, or chloro and methyl disubstituted phenyl, substituted at the 2 and 3 or 2 and 4 positions; or 2, 4, 5 trichlophenyl. When R2 is a substituted phenyl group, R2 is more usually 2,4-dichlorophenyl or 4-chloro-2-methylphenyl. [0029] X represents a straight chain or branched chain, saturated or unsaturated hydrocarbon linkage group. When X is a saturated hydrocarbon linkage group, X is usually a straight chain linkage group and usually X has the formula -(CH2)p-, with p equal to 1 , 2, or 3. When X is a saturated hydrocarbon linkage group, X is most usually a methylene group, -(CH2)-. When X is an unsaturated hydrocarbon linkage group, X is usually a straight chain linkage group, most usually - (CH=CH)-.
[0030] Y represents a moiety of formula -CHR7-, where R7 is hydrogen or a straight chain or branched chain alkyl group; more usually R7 is hydrogen or a straight chain alkyl group; more usually still R7 is hydrogen, methyl, ethyl, or n-propyl; more usually still Ry is hydrogen or methyl; and most usually Ry is hydrogen (i.e., Y is most usually -CH2-).
[0031] n is zero or, most usually, one.
[0032] v-^ is a core ring system that may generally be an aryl, heteroaryl, cycloalkyl or heterocyclyl ring system. The Ar core ring system usually includes 2 fused rings. The fused rings may generally be 4-, 5-, 6-, 7-, or 8-membered rings, more usually 5- or 6-membered rings. The core ring system is most usually fused 5- and 6-membered rings. The fused ring atoms may generally be any atom, usually carbon or hetero atoms, more usually carbon and nitrogen group atoms, and more usually still carbon and nitrogen. The number of carbon atoms in the core ring system is usually 7. The core ring system usually contains 2 hetero atoms, where the preferred hetero atom is nitrogen. Generally, one or more of the fused rings may be aromatic. When the core ring system is fused 5- and 6-membered rings, the core ring system is usually aromatic over both fused rings. The fused 5- and 6-membered ring system is most usually an indazole.
[0033] More particularly, lonidamine analogs for use according to the methods of the invention, and certain of the novel analogs provided by the invention, include analogs of the formula
Figure imgf000010_0001
where R1, R2, X, Y, and n are generally as above or, in a preferred version,
R2 is -Cl, -Br, -I, or -CH3, monosubstituted phenyl, substituted at the 2, 3, or 4 position; dichloro, dibromo, dimethyl, or chloro and methyl disubstituted phenyl, substituted at the 2 and 3 or 2 and 4 positions; or 2, 4, 5 trichlophenyl; Y is -(CH2)-; and n is zero, and R1 is -COOH, -CONH2, -CONHNH2, -CONHN(CH3)2) - CH2CH2OH, -CH2CH(OH)CH2OH, or CH2(CH2OH)2; or n is one, R1 is -COOH, and X is -CH=CH-.
[0034] In one embodiment, the lonidamine analog is a 1 ,3-substituted- indazole, such as a 1-halobenzyM H-indazole. In another embodiment, the lonidamine analog is a 3-substituted 1-benzyl-1H-indazole. In another embodiment, the lonidamine analog is a i-substituted-indazole-3-carboxylic acid, such as a 1- halobenzyl-1 H-indazole-3-carboxylic acid.
[0035] Bioisosteres. In addition, lonidamine analogs that may be used in the treatment methods of the invention include bioisosteres and pharmacophores of lonidamine and analogs described herein. Bioisosterism is a well-known tool for predicting the biological activity of compounds, based upon the premise that compounds with similar size, shape, and electron density can have similar biological activity. To form a bioisostere of a given molecule, one replaces one or more atoms or groups with known bioisosteric replacements for that atom or group. Known bioisosteric replacements include, for example, the interchangeability of -F1 -OH, - NH2, -Cl, and -CH3; the interchangeability of -Br and -/-C3H7; the interchangeability of -I and -f-C4H9; the interchangeability of -O-, -S-, -NH-, -CH2, and -Se-; the interchangeability of -N=, -CH=, and -P= (in cyclic or noncyclic moieties); the interchangeability of phenyl and pyridyl groups; the interchangeability of -C=C- and - S- (for example, benzene and thiophene); the interchangeability of an aromatic nitrogen (R-ι-N(R3)-R2) for an unsaturated carbon (R1-C(=R3)-R2); and the interchangeability of -CO-, -SO-, and -SO2- . These examples are not limiting on the range of bioisosteric equivalents and one of skill in the art will be able to identify other bioisosteric replacements known in the art. See, e.g., Patani and LaVoie, 1996; and Burger, 1991.
[0036] Pharmacophores. In addition to the lonidamine analogs described herein, lonidamine analogs that may be used in the methods of the invention can generally be any pharmacophore of lonidamine and the lonidamine analogs described above. Often a reasonable quantitative prediction of the binding ability of a known molecule can be made based on the spatial arrangement of a small number of atoms or functional groups in the molecule. Such an arrangement is called a pharmacophore, and once the pharmacophore or pharmacophores in a molecule have been identified, this information can be used to identify other molecules containing the same or similar pharmacophores. Such methods are well known to persons of ordinary skill in the art of medicinal chemistry, and as the structural information described herein identifies the pharmacophore of lonidamine and the lonidamine analogs relevant to treatment of BPH, those of skill in the art can identify other LND analogs that comprise the pharmacophore and so are useful in treating BPH. An example of programs available to perform pharmacophore-related searches is the program 3D Pharmacophore search from the Chemical Computing Group (see http://www.chemcomp.com/ fdept/prodinfo.htm). [0037] A lonidamine analog of particular interest is tolnidamine (1-(4-chloro-2- methylbenzyl)-1H-indazole-3-carboxylic acid, AF 1923); see Ansari et al., 1998; Corsi et al., 1976. Tolnidamine (TND) differs from lonidamine by the presence of a methyl substituent, rather than a chlorine substituent, in position 2 of the benzyl group. Other analogs of lonidamine with biological activity have been described in the following publications: U.S. Patent No. 3,895,026 entitled "Substituted 1-Benzyl- 1 H-lndazole-3-Carboxylic Acids and Derivatives Thereof;" Corsi et al., 1976, "1- Halobenzyl-1 H-lndazole-3-Carboxylic Acids. A New Class of Antispermatogenic Agents," Journal of Medicinal Chemistry 19:778-83; Silvestrini, 1981 , "Basic and Applied Research in the Study of Indazole Carboxylic Acids," Chemotherapy 27:9- 20; Lobl et al., 1981 , "Effects of Lonidamine (AF 1890) and its analogues on follicle- stimulating hormone, luteinizing hormone, testosterone and rat androgen binding protein concentrations in the rat and rhesus monkey," Chemotherapy 27:61-76; U.S. Patent 6,001 ,865 entitled "3-Substituted 1-Benzyl-1 H-lndazole Derivatives As Antifertility Agents"; and Cheng et al., 2001 , "Two new male contraceptives exert their effects by depleting germ cells prematurely from the testis," Biol. Reprod. 65:449-61 , which describe AF-2364 and AF-2785 and other compounds. The structures of lonidamine (I1 R = Cl), tolnidamine (I, R= CH3), AF-2364 (II) and AF- 2785 (III) are shown below.
Figure imgf000013_0001
Il III
[0038] Functional characteristics of lonidamine analogs. Lonidamine analogs suited for use in the invention are those that interfere with cellular energy metabolism of prostate epithelial cells when administered to a human, non-human primate, or other mammal. As is usual in the pharmaceutical arts, not every structural analog of a compound (e.g., lonidamine) is pharmacologically active. Active forms can be identified by routine screening of analogs for the activity of the parent compound. A variety of assays and tests can be used to assess pharmacological activity of lonidamine analogs, including in vitro assays, such as those described below and elsewhere herein, in vivo assays of prostate function (including citrate production and ATP production) in humans, non-human primates and other mammals, in vivo assays of prostate size in humans, non-human primates and other mammals, and/or clinical studies.
[0039] Apoptosis assay in cell lines. Lonidamine induces apoptosis in cell lines derived from human prostate cells. The induction of apoptosis is significantly greater in LNCaP cells (ATCC NO. CLR-1740), a prostate-derived cell line that is citrate-producing, than in PC3 cells (ATCC NO. CLR-1435), a prostate-derived cell line that is citrate-oxidizing, consistent with the susceptibility of the citrate-producing prostate cells to metabolic inhibitors such as lonidamine. In some embodiments of the invention in which a lonidamine analog is used for treatment or prevention of BPH or its manifestations, an analog with similar apoptosis-inducing activity is selected. Thus, in some embodiments of the invention, a lonidamine analog that induces apoptosis (enhances caspase 3 activity) in citrate-producing prostate cells, such as LNCaP cells, is administered to treat BPH. In some embodiments of the invention, a lonidamine analog that induces apoptosis in LNCaP cells to a significantly greater degree than in PC3 cells is administered to treat BPH. In some embodiments of the invention, the induction of apoptosis by the lonidamine analog is at least about 2-fold greater in LNCaP cells than in PC3 cells (and sometimes at least about 3-fold greater, at least about 4-fold greater, or at least about 10-fold greater) when assayed at the concentration of analog at which the difference in the level of apoptosis in the two cell lines is greatest (provided that the concentration of analog used in the assay is not greater than 1 mM). [0040] Apoptosis assay in primary cell cultures. Lonidamine induces apoptosis in primary cultures of human prostate epithelial cells. The induction of apoptosis is significantly greater in primary cultures of prostate epithelial cells than in primary cultures of human prostate stromal cells, consistent with the susceptibility of citrate-producing prostate cells to metabolic inhibitors such as lonidamine. In some embodiments of the invention in which a lonidamine analog is administered for treatment or prevention of BPH or its manifestations, an analog with apoptosis- inducing activity similar to that of lonidamine is selected. Thus, in some embodiments of the invention, a lonidamine analog that induces apoptosis in prostate epithelial cells is administered to treat BPH. In some embodiments of the invention, a lonidamine analog that induces apoptosis in primary cultures of prostate epithelial cells to a significantly greater degree than in primary cultures of human prostate stromal cells is used. In some embodiments of the invention, the lonidamine analog does not significantly induce apoptosis in stromal cells. In some embodiments of the invention, induction of apoptosis by the lonidamine analog is at least 2-fold greater in epithelial cells than in stromal cells (and sometimes at least 4- fold greater, sometimes at 10-fold greater, and sometimes at least 20-fold greater) when assayed at the concentration of analog at which the difference in the level of apoptosis in the two cell lines is greatest (provided that the concentration of analog used in the assay is not greater than 1 mM).
[0041] HIF-1 alpha expression assays. Lonidamine reduced HIF-1 alpha expression/accumulation (measured in the nuclear fraction) in cells cultured under conditions of hypoxia by almost 2-fold at 200 micromolar and by more than 5 fold and more than 10-fold at higher lonidamine concentrations. Thus, in some embodiments of the invention, a lonidamine analog reduces HIF-1 alpha expression (prevents HIF-1 alpha accumulation) in LNCaP cells cultured under hypoxic conditions by at least about 2-fold, at least about 5-fold or at least about 10-fold compared to culture in the absence of such lonidamine analog. [0042] The effect of lonidamine on HIF-1 alpha expression in prostate cells appears more pronounced in LNCaP cells than in PC3 cells cultured under hypoxic conditions (oxygen level <0.1%). Some lonidamine analogs useful for treatment of BPH according to the present invention have a similar effect. [0043] Hexokinase activity. As discussed above, and without intending to be bound to any specific mechanism, the effects of lonidamine on the prostate may be mediated, at least in part, by its effects on mitochondria and mitochondrial hexokinase activity in secretory epithelial cells. Accordingly, some lonidamine analogs useful in the present invention have hexokinase inhibitory activity as great or greater than that of lonidamine. Assays for hexokinase activity are known in the art. See Fanciulli et al., 1996, and Floridi et al., 1981. [0044] Antispermatogenic activity. Likewise, it is believed that the antispermatogenic activity of lonidamine results, at least in part, from energolytic (inhibition of energy production) effects in germ cells. Some lonidamine analogs useful in the present invention have antispermatogenic activity as great, or greater, than that of lonidamine. Assays for antispermatogenic activity are known in the art. See, e.g., Grima et al., 2001; Lohiya et al., 1991.
[0045] In addition to in vitro assays, lonidamine analogs can be evaluated in vivo for suitability for use in the methods of the invention. For example and without limitation, suitable assays include measurements of prostate function and activity. [0046] In vivo measurements of prostate function. The effect of a compound on prostate function, and, in particular, on respiration, can be assessed by monitoring prostate tissue metabolism following administration of the compound. Some lonidamine analogs useful in the present invention will detectably reduce ATP1 citrate, and/or lactate production by the prostate in animals (including humans, non- human primates and other mammals). ATP, citrate, and/or lactate levels can be monitored directly and/or indirectly in vivo using techniques of magnetic resonance spectroscopy (MRS) or other methods. See, for example, Narayan and Kurhanewicz, 1992; Kurhanewicz et a/., 1991 ; Thomas et a/., 1990, for MRS assays that can be applied for this purpose.
[0047] In vivo measurements of prostate size. The effect of a compound on prostate size can be assessed following administration of the compound using standard methods (for example, ultrasonography or digital rectal examination, for humans, and ultrasonography and/or comparison of organ weight in animals). Assays can be conducted in humans or, more usually, in healthy non-human animals such as in the monkey, dog, rat, or other animal models of BPH (see, Jeyaraj et a/., 2000; Lee et al., 1998; Mariotti et al., 1982), Some lonidamine analogs useful in the present invention will detectably reduce prostate size in such assays and animal models.
[0048] Clinical trials. Clinical trials, such as that described for lonidamine in the Example, infra, can be used to assess the therapeutic effects of lonidamine analogs.
[0049] The activity of a lonidamine analog of interest in any of the aforementioned assays, models, and trials can be compared with that of lonidamine to provide guidance concerning dosage schedules for the compound, and other information. Generally, lonidamine analogs with greater biological activity per mg than lonidamine are of special interest. Some lonidamine analogs of interest have a longer in vivo half-life and/or greater bioavailability than lonidamine.
4. Synthesis and Forms of Lonidamine and Lonidamine Analogs [0050] Lonidamine and lonidamine analogs and derivatives can be prepared using well known synthetic methods and this disclosure. Synthesis of lonidamine is described in U.S. Patent No. 3,895,026 and Germany Patent No. 2,310,031. Synthesis of exemplary lonidamine analogs, including tolnidamine (TND), is described in the art (see, e.g., Corsi et al., 1976, "1-Halobenzyl-1H-lndazole-3- Carboxylic Acids. A New Class of Antispermatogenic Agents", Journal of Medicinal Chemistry 19:778-83; Cheng etal., 2001 , "Two new male contraceptives exert their effects by depleting germ cells prematurely from the testis" Biol Reprod. 65:449-61 ; Silvestrini, 1981 , "Basic and Applied Research in the Study of Indazole Carboxylic Acids" Chemotherapy 27:9-20; Lobl et al., 1981 , "Effects of Lonidamine (AF 1890) and its analogues on follicle-stimulating hormone, luteinizing hormone, testosterone and rat androgen binding protein concentrations in the rat and rhesus monkey" Chemotherapy 27 :61 -76; and U.S. Patent Nos. 3,895,026 and 6,001 ,865). ). It will be appreciated, of course, that lonidamine analogs useful in the practice of the invention are not limited to those for which specific structures are provided in this disclosure or the cited references, and that the compounds described above are provided for illustration and not to limit the present invention. Lonidamine analogs useful in the methods of the present invention are not limited to those now described herein or elsewhere in the pharmaceutical and patent literature; the ordinarily skilled practitioner guided by the present disclosure can synthesize novel analogs suitable for use according to the present invention using routine methods of medicinal chemistry and this disclosure. Exemplary methods for synthesis of lonidamine and analogs (indazole derivatives) useful in the practice of the present invention are described in U.S. provisional application No. 60/576968 (filed June 20, 2004) and U.S. provisional application No. 60/588,694 (filed July 15, 2004), both incorporated herein by reference.
[0051] In certain embodiments, lonidamine or a lonidamine analog is provided in the form of a pharmaceutically acceptable salt. Pharmaceutically acceptable salts include addition salts with acids, as well as the salts with bases. Salts with bases are, for example, alkali metal or alkaline earth metal salts, such as sodium, potassium, calcium or magnesium salts, or ammonium salts, such as those with ammonia or suitable organic amines, e.g. diethylamine, di-(2-hydroxyethyl)-amine or tri-(2-hydroxyethyl)-amine. Suitable acids for the formation of acid addition salts are, for example, mineral acids, such as hydrochloric, hydrobromic, sulphuric or phosphoric acid, or organic acids, such as organic sulphonic acids, for example, benzenesulphonic, 4-toluenesulphonic or methanesulphonic acid, and organic carboxylic acids, such as acetic, lactic, palmitic, stearic, malic, maleic, fumaric, tartaric, ascorbic or citric acid.
[0052] Administration of ester, amide and prodrug derivatives of lonidamine and analogs is also contemplated in the practice of the present invention (see, e.g., U.S. Pat. No. 6,146,658, for general information regarding preparation of such derivatives from a compound of interest) as is administration of polymorphic forms, enantiomeric forms, tautomeric forms, solvates, hydrates, and the like. Exemplary prodrug forms of lonidamine and lonidamine analogs are described in U.S. provisional application no. 60/257026, entitled "Tertiary amine prodrugs of lonidamine and analogs" (filed July 8, 2004), incorporated herein by reference. 5. Patients For Whom Administration of Lonidamine Provides Benefit [0053] The present invention provides that administration of lonidamine to men afflicted with, or susceptible to, BPH can be therapeutically effective. Accordingly, in one aspect of the invention, lonidamine or a lonidamine analog is administered to a subject in need of treatment for BPH. In one embodiment, the subject in need of treatment is a human male who does not have cancer. As used herein, "a subject in need of treatment for BPH" is a man diagnosed with BPH. BPH can be diagnosed using art-known methods and criteria. The most common test is the digital rectal examination in which a physician determines whether the prostate is of a normal size and firmness. Other diagnostic assays include a urine flow rate test, determination of post void residual urine volume (e.g., by palpitation of the abdomen, drainage of residual urine, x-ray urogramography, or ultrasonography), moderate or severe symptom scores on the American Urologic Association Symptom Index (AUASI; Barry et al., 1992) or International Prostate Symptom Score (IPSS; Barry et a/., 2001), and other tests known in the art.
[0054] Desired clinical results of treatment for BPH include, but are not limited to, alleviation or amelioration of one or more symptoms of BPH (see below), a reduction in prostate size (see below), a reduction in AUASI or IPSS scores compared to baseline measurements prior to commencement of therapy (for example, by 3 points or more, such as by 5 points or more), AUASI or IPSS scores less than 8, a reduction in serum PSA by at least about 20% (as discussed below, a transient rise in serum PSA can occur upon initial administration of lonidamine or a lonidamine analog), such as by at least about 40%, a serum PSA less than 4, such as less than 2, improvement in urodynamic parameters, and other desired results that will be recognized by a treating physician as indicative of a reduction in severity of BPH in a subject. An assessment of the response to treatment can be made at any time following the first administration of the drug. For example, an assessment is made about 30 days, about 60 days, or about 90 days after beginning treatment. Alternatively, assessment can be made about 6, 12, 18, 24 or more months after beginning treatment. Alternatively, an assessment can be made less than about 30 days, about 30 days, about 60 days, or about 90 days after a course of treatment ends.
[0055] In a related aspect, lonidamine or a lonidamine analog is administered to a human subject exhibiting a symptom associated with BPH to reduce the frequency or severity of the symptom. As used herein, "a symptom associated with BPH" refers to any one or more of the following symptoms: (1) urinary urgency; (2) terminal dribbling of urine; (3) frequent urination; (4) nocturia; (5) a weak/slow stream of urine; (6) a sense of incomplete emptying; (7) intermittency; (8) straining; (9) dysuria; (10) hematuria; (11) acute urinary retention; (12) urinary tract infection; (13) incontinence. In addition, there is an association between BPH and erectile dysfunction. Thus, in one embodiment of the invention, lonidamine or a lonidamine analog is administered to treat erectile dysfunction. Administration of lonidamine or a lonidamine analog according to the methods of the invention typically results in a reduction in severity, or elimination, of one or more of these symptoms; usually results in either a reduction in severity of, or elimination of, all of these symptoms; and may result in elimination of all of these symptoms. [0056] In another related aspect, lonidamine or a lonidamine analog is administered to reduce prostate size in a human subject in need of such reduction. As used herein, "a subject in need of reduction of prostate size" is a man having an enlarged prostate gland as determined by (1 ) imaging (e.g., ultrasonography, magnetic resonance imaging) or (2) digital rectal examination or (3) one or more signs or symptoms resulting directly or indirectly from compression of the urethra by the prostate (e.g., including the symptoms of BPH discussed herein). A reduction in serum PSA (prostate specific antigen) is also a useful proxy for reduction of prostate volume. Although varying among individuals, enlarged prostates often exceed 30 grams, 40 grams, or 50 grams in size. The degree of reduction of prostate size will vary from subject to subject due to a number of factors, including the degree of enlargement at the time of onset of therapy, but will typically be a reduction of at least about 10% volume, more often at least about 25%, sometimes at least about 40%, and sometimes an even greater than 40% reduction in prostate size is observed. This reduction can be determined by imaging or other methods. Serum PSA can also in some instances serve as a useful proxy for prostate volume. [0057] In a related aspect, lonidamine or an analog is administered to a subject with a serum PSA level greater than 2 ng/ml. PSA is secreted only by the epithelial cells of the prostate. For men with BPH, higher PSA levels suggest a relatively higher ratio of epithelial cell proliferation to stromal cell proliferation than in men with lower PSA levels. The present invention provides a number of diagnostic methods suitable for use in determining patients who should respond favorably to treatment with lonidamine or an analog. Thus, lonidamine treatment can provide a therapeutic benefit to subjects with PSA levels greater than 2 ng/ml. Accordingly, subjects predicted to benefit significantly from treatment in accordance with the invention can be selected in a population of men with BPH by identifying subjects with a serum PSA value greater than 2 ng/ml. In one embodiment of the invention, the subject has a PSA level greater than about 4 ng/ml. Because higher PSA levels are also, and perhaps more closely, associated with prostate cancer than with BPH, in one embodiment, the subject selected for therapy with lonidamine or an analog has a PSA level less than about 10 ng/ml.
[0058] In one aspect of the invention, lonidamine or a lonidamine analog is administered to a subject who would benefit from prophylaxis of BPH. In one example, "a subject who would benefit from prophylaxis of BPH" is a man previously treated for BPH by surgery, transurethral microwave thermotherapy, transurethral needle ablation, transurethral electrovaporization, laser therapy, balloon dilatation, prostatic urethral stent, drug therapy, or other therapy and not currently diagnosed with or exhibiting symptoms of BPH. In another example, a subject who would benefit from prophylaxis of BPH is a man at increased risk for developing BPH due to age (e.g., men older than 40, older than 50, older than 60, or older than 70 years of age). In another example, a subject who would benefit from prophylaxis of BPH is a man who is asymptomatic, or has symptoms sufficiently mild so that no clear diagnosis of BPH can be made, but who has an elevated serum PSA level (e.g., PSA > 2 ng/ml or, in some cases, >4 ng/ml).
[0059] Thus, in some cases, the subject to whom lonidamine is administered in accordance with the methods of the invention is a man who has previously been treated for BPH, while in other cases the subject is a man who has not previously been treated for BPH. Similarly, it will be clear that any references herein to administration of lonidamine apply equally to administration of a biologically active lonidamine analog.
[0060] In one embodiment of the invention, the subject in need of treatment or prophylaxis for BPH either is not also under treatment for cancer or does not have cancer. In a related embodiment, the subject in need of treatment or prophylaxis for BPH has not been diagnosed as having cancer. In one embodiment, the subject in need of treatment or prophylaxis for BPH does not have cancer. In one embodiment, the subject in need of treatment has a cancer other than prostate cancer but does not have prostate cancer. As used herein, "cancer" has its ordinary medical meaning and refers to a malignancy (including head, neck, prostate and breast cancers, leukemias and lymphomas), generally characterized by clonality, autonomy, anaplasia, and metastasis (see Mendelsohn, 1991).
[0061] In one embodiment, the invention provides a method of treating BPH in a patient by administering lonidamine to the patient. In a related embodiment, the invention provides a method for treating BPH comprising (a) administering lonidamine to a patient diagnosed with BPH and (b) determining whether one or more manifestations of BPH are reduced in the patient. In one embodiment, the invention provides a method for treating BPH by (a) diagnosing BPH in a patient, (b) administering lonidamine to the patient and (c) determining whether one or more manifestations of BPH are reduced in said patient. In one embodiment, the invention provides a method of treating BPH in a patient by administering a lonidamine analog to the patient. In a related embodiment, the invention provides a method for treating BPH comprising (a) administering a lonidamine analog to a patient diagnosed with BPH and (b) determining whether one or more manifestations of BPH are reduced in the patient. In one embodiment, the invention provides a method for treating BPH by
(a) diagnosing BPH in a patient, (b) administering a lonidamine analog to the patient and (c) determining whether one or more manifestations of BPH are reduced in said patient. In the foregoing embodiments, optionally the subject is not diagnosed with or under treatment for cancer; optionally has a PSA less than or equal to 2 ng/ml; or optionally has a PSA greater than 2 ng/ml and less than 10 ng/ml.
[0062] In another aspect, the invention provides a method entailing (a) advertising the use of lonidamine, or a lonidamine analog, for treatment of BPH, and
(b) selling lonidamine or a lonidamine analog to individuals for use in the treatment of BPH. In one embodiment, the advertising makes reference to a trademark that identifies a lonidamine (or lonidamine analog) product and the lonidamine (or analog) sold in step (b) is identified by the same trademark. It will be appreciated that the individuals to whom lonidamine is sold include corporate persons (corporations) and the like and "selling BPH to individuals for use for treatment of BPH" includes selling to, for example, a medical facility for distribution to patients for treatment of BPH. [0063] In another embodiment, the invention provides a method of treating acute urinary retention in a human by administering lonidamine or a lonidamine analog to the human in need of such treatment. Because acute urinary retention can be a symptom of BPH, this embodiment of the invention is applicable to any subject who suffers from acute urinary retention but has not been diagnosed as having BPH when lonidamine or a Ionidamine analog is first administered.
6. Dose, Route, Schedule and Duration of Administration [0064] A variety of routes and dosage schedules are appropriate for administration of lonidamine and lonidamine analogs according to the invention. [0065] A preferred mode of delivery of lonidamine and lonidamine analogs to a patient is oral delivery. Preferred dosage forms for oral administration are pills, tablets, capsules, caplets, and the like, especially as formulated for sustained release. Other suitable forms for oral administration include troches, elixirs, suspensions, syrups, wafers, lozenges, and the like. Other modes of administration are also contemplated, including parenteral, inhalation spray, transdermal, rectal, intraprostetic injection (e.g., of lonidamine-containing microparticles) and other routes. Lonidamine and lonidamine analogs may be formulated in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration. In one embodiment, the dosage form is the 150 mg unit dosage form marketed for the treatment of cancer by ACRAF in Italy under the tradename Doridamina. Doridamina contains lonidamine 150 mg; starch 87 mg; microgranular cellulose 55 mg; lactose 40 mg; sodium carboxymethyl starch 15 mg; hydroxypropyl methylcellulose 11 mg; precipitated silica 4.5 mg; magnesium stearate 3.5 mg; titanium dioxide 0.4 mg; polyethylene glycol 400 0.4 mg. Another formulation is 150 mg lonidamine (intra-granular), 87 mg cornstarch (intra-granular), 40 mg lactose (intra-granular), 3 mg colloidal silicon dioxide (intra-granular), 1.5 mg colloidal silicon dioxide (extra-granular), 55 mg microcrystalline cellulose (extra-granular), 15 mg sodium starch glycolate (extra-granular), 3.5 mg magnesium stearate (extra- granular), 9 mg hydroxypropyl methylcellulose (extra-granular), 10.9 mg OPADRY white (coating).
[0066] The dose, schedule and duration of administration of lonidamine and lonidamine analogs will depend on a variety of factors, including the age, weight and health of the subject, the severity of BPH symptoms, if any, the subject's medical history, co-treatments, therapeutic goal (e.g., therapy or prophalaxis), the mode of administration of the drug, the formulation used, patient response to the drug, and the like. For illustration rather than limitation, three general categories of dosing for administration of lonidamine and lonidamine analogs can be described: high dosing, low dosing, and intermediate dosing. For reference, the standard lonidamine dose used for the treatment of cancer for which lonidamine has been approved in a few countries in Europe is 150 mg po TID for about thirty days or longer. [0067] Low dosing. Low dosing is contemplated for the treatment and prophalaxis of BPH. Exemplary low doses of lonidamine or a lonidamine analog include, without limitation, doses in the range of 1-300 mg per day (total daily dosage), more often in the range of 5-300 mg/day, or sometimes in the range of 5-70 mg/day. Other exemplary low dose ranges include 1-25 mg/day, 20-45 mg/day, 40- 65 mg/day, 40-70 mg/day, 50-100 mg/day, 80-100 mg/day, 50-200 mg/day, and 50- 300 mg/day. In one embodiment, the low dose is 150 mg administered orally once per day; the Doridamina unit dose form can be used in this embodiment. In another embodiment, the low dose is 75 mg administered orally once or twice daily. The Doridamina unit dose form can be used in this embodiment by splitting it into two equal parts.
[0068] As noted, the daily dosages recommended herein may be divided for, for example, two-, three- or four-times per day administration. In one embodiment, the drug is formulated for administration once per day. In one embodiment, the drug is formulated for administration less frequently than once per day. In another embodiment, a modified-release form of the drug is used. [0069] Administration of low doses of lonidamine can be daily, every other day, five days on, two days off, and other schedules selected in accordance with the teachings herein by the administering physician.
[0070] An advantage of low dose schedules of the invention is that this dose may be continued to be administered for weeks to months while limiting or eliminating the unwanted, albeit usually mild, side effects reported for higher doses of lonidamine (principally myalgia and testicular pain).
[0071] A low dose schedule can be used for therapy or prophalaxis. In one embodiment, a low dose form is used for a maintenance dose after a higher initial, priming or loading dose.
[0072] High dosing. In another embodiment, BPH is treated in accordance with the methods of the invention by administering to a BPH patient a higher dose of lonidamine or a lonidamine analog (usually for a shorter period of time than for low dosing). Exemplary high doses include, without limitation, total daily doses greater than 0.5 g, such as doses in the range 0.5 - 5 g/day, 0.5 - 3 g/day, 0.5 - 1 g/day and 1-3 g/day, or higher doses. The daily dosages may be divided, for example, for two-, three- or four-time per day administration. In one embodiment, the drug is formulated for administration once-per day, or less frequently than once per day. In one embodiment, a modified-release form of the drug is used. Alternatively, a high dose can be administered on a one-time, once-a-week, once every two weeks, or once-a-month basis (e.g., 0.5 - 5 g/administration) or by other schedules selected in accordance with the teachings herein by the administering physician. [0073] A high dose schedule can be used for therapy or prophalaxis. In one embodiment, a high dose is administered in combination with, or following, a surgical or other non-drug treatment for BPH.
[0074] Intermediate dosing. In another embodiment, BPH is treated in accordance with the methods of the invention by administering lonidamine or a lonidamine analog to a BPH patient at a dose intermediate between a high dose and a low dose. Exemplary intermediate doses include, without limitation, doses greater than 300 and less than 500 mg/day, such as doses in the range >300-400 or 400<500 (e.g., 450 mg/day). The daily dosages may be divided, for example, for two-, three- or four-times per day administration. In an embodiment, the drug is formulated for administration once-per day or less frequently than once per day. In one embodiment, a modified-release form of the drug is used. Alternatively, this intermediate dose can be administered on a one-time, once-a-week, once every two weeks, or once-a-month basis (e.g., 300 - 500 mg/administration) or by other schedules to be determined by the administering physician. In one embodiment, the daily dosage is 150 mg of lonidamine or a lonidamine analog taken three times a day. In one embodiment, 150 mg of lonidamine or a lonidamine analog is administered one time per day for 28 days. In other embodiments, lonidamine or a lonidamine analog is administered at a dose of 5 mg, 25 mg, 50 mg, 75 mg or 100 mg per day for 4 to 26 weeks or longer.
[0075] An intermediate dose schedule may be used for therapy or prophylaxis.
In one embodiment, an intermediate dose is administered in combination with, or following, a surgical or other non-drug treatment for BPH. [0076] It will be appreciated that these dosing schedules are for illustration and not limitation, and that a dosing schedule may change during a course of therapy based on, for example, a patient's response to the therapy or the use of a lonidamine analog that has an activity/dose profile significantly different from that of lonidamine.
[0077] Duration. In therapeutic and prophylactic applications, lonidamine or the lonidamine analog can be administered once or many times over periods as long as several months or years. In one embodiment of the invention, lonidamine or an analog is administered to a symptomatic (e.g., experiencing difficulty in urination) BPH patient only until the symptoms abate or disappear, and then treatment is stopped unless and until symptoms reappear. When symptoms reappear, administration of lonidamine or an analog is resumed. In another embodiment, treatment continues after symptoms disappear or are reduced to an acceptable target level, at least for a period of time, such as a week, two weeks, a month or several months. In another embodiment, the drug is administered to an asymptomatic subject to prevent the development or reoccurrence of symptoms (i.e., prophylactically administered).
7. Treatment Combinations
[0078] Lonidamine and lonidamine analogs can be administered to a BPH patient in combination with other agents or procedures intended to treat BPH, ameliorate symptoms of BPH, potentiate the effects of the lonidamine or lonidamine analog, or provide other therapeutic benefit. Administration of an agent "in combination with" includes parallel administration (administration of both agents to the patient over a period of time, such as administration of lonidamine and tamsulosin on alternate days for one month), co-administration (in which the agents are administered at approximately the same time, e.g., within about a few minutes to a few hours of one another), and co-formulation (in which the agents are combined or compounded into a single dosage form suitable for oral, parenteral, or other administration). Exemplary agents for administration in combination with lonidamine or lonidamine analogs include, but are not limited to, zinc, alpha-blockers, 5-alpha- reductase inhibitors, treatments for urinary incontinence, and plant extracts. Other agents for administration in combination with lonidamine or lonidamine analogs include other metabolic inhibitors, including but not limited to other hexokinase inhibitors and other inhibitors of glycolysis, including but not limited to 2-deoxy-D- glucose and an inhibitor, direct or indirect, of HIF-1alpha. [0079] Zinc. As discussed above, high concentrations of zinc in the secretory epithelial cells of the prostate inhibit m-aconitase, increasing the dependence of that tissue on glycolysis for energy production. In accordance with the methods of the present invention, it may in some patients be beneficial to co-administer zinc (e.g., zinc chloride, zinc gluconate, zinc sulfate, zinc acetate, zinc aspartate, zinc citrate, zinc glycerate, zinc oxide, zinc picolinate, and the like) with a drug composition of the invention, to maximize the efficacy of the treatment. For example and not limitation, 15-300 mg/day zinc can be administered for this purpose; typically 30-50 mg/day is administered.
[0080] Alpha-Adrenerqic-Blockers. Alpha-blockers alleviate some symptoms of BPH, without curing the underlying disease. These agents work by relaxing the muscles at the neck of the bladder and in the prostate, reducing the pressure on the urethra. Exemplary alpha-blockers include doxazosin (Cardura), terazosin (Hytrin), tamsulosin (Flomax), alfuzosin (Xatral), and prazosin (Hypovase). In one embodiment of the invention, an alpha blocker is administered in combination with lonidamine or a lonidamine analog to treat BPH. In another embodiment, the alpha- blocker is administered at a lower dosage (amount) or less frequently (e.g., alternate days rather than daily) than the "standard" dosage (the dosage that would be indicated for the subject in the absence of lonidamine administration) in combination with lonidamine or a lonidamine analog.
< [0081] 5-Alpha-Reductase Inhibitors. 5-alpha-reductase inhibitors inhibit the conversion of testosterone to dihydrotestosterone 2 (DHT), an androgen that contributes to prostate enlargement. An exemplary 5-alpha-reductase inhibitor is finasteride (Proscar). Another exemplary 5-alpha-reductase inhibitor is Avodart® (dutasteride). In one embodiment of the invention, a 5-alpha-reductase inhibitor is administered in combination with lonidamine or an analog to treat BPH. In another embodiment, the 5-alpha-reductase inhibitor is administered at a lower dosage (amount) or less frequently (e.g., alternate days rather than daily) than the "standard" dosage (the dosage that would be indicated for the subject in the absence of lonidamine administration) in combination with lonidamine (or a lonidamine analog). [0082] Treatments for Urinary Incontinence. Treatments for urinary incontinence include alpha-adrenergic agonists such as pseuodoephedrine hydrochloride (Sudafed), and phenylpropanolamine; anticholinergics such as dicyclomine hydrochloride (Bentyl), and hyoscyamine sulfate (Levsin SL, Levsin, Levsinex, Cystospaz-M, Levbid); and antispasmodic drugs including darifenacin (Enablex), solifenacin succinate (VESIcare), oxybutynin (Ditropin), tolterodine L- tartrate (Detrol and Detrol LA), and trospium (Sanctura).
[0083] Glycolytic and Mitochondrial Function Inhibitors. Glycolytic inhibitors, such as 2-deoxy-D-glucose and compounds that inhibit glucose transport, mitochondrial function inhibitors, mitochondrial poisons, and hexokinase inhibitors such as 3-bromopyruvate and its analogs can also be used in combination with lonidamine or a lonidamine analog to treat BPH. Such inhibitors are known in the art, and include those described in PCT patent publications WO 01/82926 published 8 November 2001 ; U.S. Patent Nos. 6,670,330; 6,218,435; 5,824,665; 5,652,273; and 5,643,883; U.S. patent application publication nos. 20030072814; 20020077300; 20020035071 , and U.S. patent application Serial No. 10/754,239. Such inhibitors can be administered in combination with lonidamine or lonidamine analogs for therapeutic benefit in the treatment of BPH.
[0084] Plants. Saw Palmetto (Serenoa repens) or an extract thereof, or
Pygeum Africanum or an extract thereof can be administered in combination with lonidamine or lonidamine analogs for therapeutic benefit in the treatment of BPH. [0085] Procedures. In addition, lonidamine or a lonidamine analog may be administered in combination with, or prior to, procedures for treatment of BPH including surgery (transurethral resection of the prostate; transurethral incision of the prostate; or open prostatectomy), laser therapy, transurethral microwave thermotherapy, balloon dilatation, placement of a prostatic urethral stent, transurethral needle ablation, transurethral electrovaporization of the prostate, or other non-drug therapies.
8. Dosage Forms
[0086] Unit Dosage Forms. The compounds used in the methods of the present invention are formulated in compositions suitable for therapeutic administration. In one embodiment, the methods of the invention are practiced with lonidamine in the unit dosage form marketed as Doridamina (by ACRAF) in Italy. New dosage forms of lonidamine are also provided by the invention. For example, the present invention provides a unit dosage pharmaceutical formulation of lonidamine that is suitable for oral administration (including tablets, capsules, caplets, and pills) and contains, in various embodiments, an amount of lonidamine in a range bounded by a lower limit of (in mg) 1 , 5, 10, and 50 and an upper limit of 10, 20, 40, 50, 70 and 100 (where the higher limit is in mg and greater than the lower limit) and is especially convenient for certain low dose schedules. In another embodiment, the unit dosage form contains an amount of drug in a range bounded by a lower limit of (in mg) 200, 300, 500 or 1000 and an upper limit of 500, 1000, 3000 or 5000 (where the higher limit is in mg and greater than the lower limit) and is especially convenient for certain high dose schedules. In yet other embodiments, the formulation contains between 100 and 200 mg of compound (e.g., 150 mg), between 200 and 5000 mg, between 200 and 1000 mg, or between 500 and 1000 mg of the compound. Lonidamine analogs can be similarly formulated. Dosage forms of particular interest contain 5, 25, 50, 100 or 150 mg lonidamine. [0087] In addition to lonidamine and/or lonidamine analogs, solid unit dosage forms of the invention generally include a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" refers to a solid or liquid filler, diluent, or encapsulating substance, including for example excipients, fillers, binders, and other components commonly used in pharmaceutical preparations, including, but not limited to, those described below. Methods for formulation of drugs generally are well known in the art, and the descriptions herein are illustrative and not limiting. See, e.g., Ansel et al., 1999; Marshall, 1979.
[0088] Hydrophilic binders suitable for use in the formulations of the invention include copolyvidone (cross-linked polyvinylpyrrolidone), polyvinylpyrrolidone, polyethylene glycol, sucrose, dextrose, corn syrup, polysaccharides (including acacia, guar, and alginates), gelatin, and cellulose derivatives (including HPMC, HPC, and sodium carboxymethylcellulose).
[0089] Water-soluble diluents suitable for use in the formulations of the invention include sugars (lactose, sucrose, and dextrose), polysaccharides (dextrates and maltodextrin), polyols (mannitol, xylitol, and sorbitol), and cyclodextrins. Non-water-soluble diluents suitable for use in the formulations of the invention include calcium phosphate, calcium sulfate, starches, modified starches, and microcrystalline cellulose.
[0090] Surfactants suitable for use in the formulations of the invention include ionic and non-ionic surfactants or wetting agents such as ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglycerides, sorbitan fatty acid esters, poloxamers, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene derivfatives, nonoglycerides or ethoxylated derivatives thereof, sodium lauryl sulfate, lecithins, alcohols, and phospholipids.
[0091] Disintegrants suitable for use in the formulations of the invention include starches, clays, celluloses, alginates, gums, cross-linked polymers (PVP, sodium carboxymethyl-cellulose), sodium starch glycolate, low-substituted hydroxypropyl cellulose, and soy polysaccharides. Preferred disintegrants include a modified cellulose gum such as cross-linked sodium carboxymethylcellulose. [0092] Lubricants and glidants suitable for use in the formulations of the invention include talc, magnesium stearate, calcium stearate, stearic acid, colloidal silicon dioxide, magnesium carbonate, magnesium oxide, calcium silicate, microcrystalline cellulose, starches, mineral oil, waxes, glyceryl behenate, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, sodium lauryl sulfate, sodium stearyl fumarate, and hydrogenated vegetable oils. Preferred lubricants include magnesium stearate and talc and combinations thereof. [0093] The preferred range of total mass for the tablet or capsule may be from about 40 mg to 2 g, from about 100 mg to 1000 mg, and from about 300 mg to 750 mg.
[0100] Sustained Release Forms. In addition, the present invention provides unit dosage forms that are sustained release formulations of lonidamine or a lonidamine analog to allow once a day (or less) oral dosing, a frequency sometimes preferred by patients over multiple day dosing. Such sustained release formulations (including tablets, capsules, caplets and pills) of the invention usually contain between 1 mg and 3 g of the active compound, with various alternative embodiments including those described above for conventional oral unit doses, such as an amount of drug in a range bounded by a lower limit of (in mg) 1 , 5, 10, and 50 and an upper limit of 10, 20, 40, 50, 70 and 100 (where the higher limit is in mg and greater than the lower limit) and are especially convenient for certain low dose or intermediate dose schedules. In another embodiment, the unit dosage form contains an amount of drug in a range bounded by a lower limit of (in mg) 200, 300, 500, 750 or 1000 and an upper limit of 500, 1000, 2000, 3000 or 5000 (where the higher limit is in mg and greater than the lower limit).
[0101] In one embodiment, lonidamine or a lonidamine analog in the sustained release formulations (also called "modified" or "controlled" release forms) is released over a period of time greater than 6 hours, e.g., greater than 12 hours, after administration. In one embodiment, the sustained release formulation allows once a day dosing to achieve a pharmacodynamic profile therapeutically equivalent to dosing 150 mg of lonidamine three times a day.
[0102] Examples of sustained-release formulations for other drugs that can be modified in accordance with the teachings herein to be useful in the present invention are well known in the art, and are, for example, described in U.S. Pat. Nos. 5,968,551 ; 5,266,331 ; 4,970,075; 5,549,912; 5,478,577; 5,472,712; 5,356,467; 5,286,493; 6,294,195; 6,143,353; 6,143,322; 6,129,933; 6,103,261 ; 6,077,533; 5,958,459; and 5,672,360. Sustained-release formulations are also discussed in the scientific literature, e.g., in ORAL SUSTAINED RELEASE FORMULATIONS: DESIGN AND EVALUATION, edited by A. Yacobi and E. Halperin-Walega, Pergamon Press, 1988, which describes a variety of types of sustained-release dosage forms and drug release mechanisms, for example single unit (e.g., matrix tablets, coated tablets, capsules), multiple unit (e.g., granules, beads, micro-capsules), inert, insoluble matrix, hydrophilic gel matrix (e.g., bioadhesive, erodible, non-erodible), and ion- exchange resin sustained-release dosage forms.
[0103] In one embodiment, the present invention provides a method of treating BPH, by administering once daily to a patient in need of such treatment a sustained release tablet dosage form comprising a daily therapeutic dose of lonidamine from about 1 mg to 2 g in a hydrophilic matrix. The matrix can be, for example and without limitation, selected from the group consisting of hydroxypropylmethyl cellulose (by weight percent of about 20-40%), lactose (5-15%), microcrystalline cellulose (4-6%), and silicon dioxide (1-5%) having an average particle size ranging from 1-10 microns, often ranging from 2-5 microns, and most often ranging from about 2-3 microns.
[0104] Illustrative preferred sustained release formulations of the invention include formulations A and B in Table 1.
Table 1
Formulation A B
(weight percentage) Lonidamine (milled) 53.8 53.8
HPMC (Methocel K15M, CR) 8 30 .
Methyl cellulose (Methocel, K100L, CR) 18 0 Anydrous lactose 12.2 8.2
Microcrystalline cellulose (Avicel PH101) 5 5 Silicon dioxide (1-10 micron; Syloid 244) 3 3 Total Tablet Weight (in grams) 1 1 [0105] The sustained release formulations of the invention may be in the form of a compressed tablet containing an intimate mixture of lonidamine and a partially neutralized pH-dependent binder that controls the rate of drug dissolution in aqueous media across the range of pH in the stomach (typically ~ 2) and intestine (typically -5.5).
[0106] Many materials known in the pharmaceutical art as "enteric" binders and coating agents have the desired pH dissolution properties suitable for use in the sustained formulations of the invention. These include phthalic acid derivatives such as the phthalic acid derivatives of vinyl polymers and copyolymers, hydroxyalkylcellulose, alkylcelluloses, cellulose acetates, hydroxyalkylcellulose acetates, cellulose ethers, alkylcellulose acetates, and esters thereof, and polymers and copoloymers of lower alkyl acrylic acids and lower alkyl acrylates, and the partial esters thereof.
[0107] Preferred pH-dependent binder materials are methacrylic acid copolymers. Such a copolymer is commercially available from Rohm Pharma as Eudragit™ L- 100-55 as a powder or L30D-55 as a 30% dispersion in water. Other pH-dependent binder materials which may be used alone or in combination include hydroxypropyl cellulose phthalate, hydroxypropyl methylcellulose phthalate, cellulose acetate phthalate, polyvinylacetate phthalate, polyvinylpyrrolidone phthalate, and the like. One or more pH-dependent binders are present in the sustained release oral dosage forms of the invention in an amount ranging from about 1 to 20 weight percent, or from 5 to 12 weight percent, or about 10%.
[0108] The pH-independent binders or viscosity enhancing agents contained in the sustained release formulations of the invention include substances such as hydroxypropyl methylcellulose, hydroxypropyl cellulose, methylcellulose, polyvinylpyrrolidone, neutral poly(meth)acrylate esters, and the like. The pH- independent binders are in an amount ranging from 1 to 10 weight percent or from 1 to 3 weight percent, or about 2%.
[0109] The sustained release formulations of the invention also contain in some embodiments one or more pharmaceutical excipients intimately mixed with the lonidamine or lonidamine analog and the pH-dependent binder, such as pH- independent binders or film-forming agent, starch, gelatin, sugars, carboxymethylcellulose, and the like, as well as other useful pharmaceutical diluents such as lactose, mannitol, dry starch, micro-crystalline cellulose, and the like, and surface active agents such as polyoxyethylene sorbitan esters, sorbitan esters, and the like; and coloring agents and flavoring agents. Lubricants such as talc and magnesium stearate and tableting aids can also be used. [0110] The sustained release formulations of the invention include any of the commercially available polymers suitable for use in such formulations, including but not limited to cellulose, ethyl cellulose, methyl cellulose, carboxymethyl cellulose, hydroxypropylmethyl cellulose, hydroxypropylmethyl cellulose phthalate, hydroxypropyl cellulose, microcrystalline cellulose, sodium carboxymethyl cellulose, cellulose acetate phthalate, polyvinyl acetate phthalate, polyvinylpyrrolidone, polyethylene oxide, polyethylene glycol, zein, alginate, hypromellose phthalate, methacrylic acid copolymer, Crospovidone, silica aerogel, pregelatinized starch, corn starch, croscarmellose sodium, sodium starch glycolate, candelilla wax, paraffin wax, carnauba wax, montan glycol wax, white wax, Eudragit (polymethacrylic acid esters), Aquacoat (ethyl cellulose, cellulose acetate phthalate), Carbopol (acrylic acid polyalkeny polyether copolymer), and Macrogol (polyethylene glycol). [0111] The sustained release formulations of the invention include formulations that are diffusion controlled, such as those that employ:
(a) a reservoir system in which the drug is encapsulated in a polymeric membrane, and water diffuses through the membrane to dissolve the drug, which then diffuses out of device;
(b) a monolithic (matrix) system in which the drug is suspended in a polymeric matrix and diffuses out through long pathways;
(c) microencapsulation and coated granule systems in which particles of drug (or particles of drug and polymer) as small as 1 micron are coated in a polymeric membrane, including embodiments in which particles coated with polymers with different release characteristics are delivered together in a capsule;
(d) solvent-activated systems, including (i) osmotically controlled devices (e.g. OROS) in which an osmotic agent and the drug are encapsulated in a semi-permeable membrane, water is pulled into device due to osmotic gradient, and increased pressure drives drug out of device through a laser drilled hole; (ii) a hydrogel swelling system in which drug is dispersed in a polymer and/or a polymer is coated onto a particle of drug, and the polymer swells on contact with water (swelling is in some embodiments pH or enyzmatically controlled), allowing diffusion of drug out of the device; (iii) a microporous membrane system in which drug is encapsulated in a membrane that has a component that dissolves on contact with water (in some embodiments, dissolution is pH or enyzmatically controlled), leaving pores in the membrane through which the drug diffuses; and (iv) a wax matrix system in which the drug and an additional soluble component are dispersed in wax, such that, when water dissolves the component, diffusion of drug from the system is allowed; and
(e) polymeric degradation systems, including (i) bulk degradation, in which drug is dispersed in polymeric matrix, and degradation occurs throughout the polymeric structure in a random fashion, allowing drug release; and (ii) surface erosion, in which drug is dispersed in polymeric matrix and delivered as the surface of the polymer erodes.
[0112] In one aspect, the invention provides a method for treating BPH by administering a unit dose oral pharmaceutical composition that is a sustained- release formulation containing an effective amount of lonidamine, such as described above, once per day.
9. Example
Lonidamine Treatment Significantly Reduces Prostate Volume and Improves Urine Flow in Symptomatic Benign Prostatic Hyperplasia
[0113] This example describes results of a Phase Il Open-Label Study to evaluate the efficacy and safety of oral lonidamine (LND) treatment in subjects with symptomatic benign prostatic hyperplasia (BPH) by measuring the change from baseline of: total prostate volume by transrectal ultrasound (TRUS), maximum flow rate (Qmaχ) on uroflowmetry, international prostatic symptoms score (I-PSS) and prostate-specific antigen (PSA).
[0114] Thirty subjects with symptomatic BPH received oral LND (150 mg/day) once daily for 28 days. Subjects were assessed at baseline, at active-therapy assessment visits (Day-14, Day-28), four weeks post-therapy (Day-56), eight weeks post therapy (Day-84), twelve weeks post therapy (Day-112) and six-months post- therapy (Day-200) for prostate volume by TRUS, Qmax, I-PSS, PSA, serum chemistry and adverse events. RESULTS
[0115] Subjects experienced an average decrease of 11.2% (p<0.001 ) in prostate volume on Day-28, which was maintained at Day-56 (average decrease of 12.0%; p<0.001 ). Qmax improved by a mean of 3.2 mL/sec at Day-28 (p=0.002). I- PSS scores improved from 19.5 prior to treatment to 12.2 at Day-28 (p<0.001). PSA decreased on average by 17.8% from baseline (p=0.001 ) to Day-28. LND was generally well-tolerated, with no reported moderate or severe adverse events.
SUBJECTS
[0116] Between March and August of 2004, 30 subjects were enrolled and received LND therapy. Male subjects between 50 and 80 years of age were eligible for inclusion if they had experienced lower urinary tract symptoms (LUTS) for at least 3 months, had a prostate volume >30 cc as measured by TRUS, a Qmax < 15 mL/sec as measured by uroflowmetry, an I-PSS of >13, PSA > 1.0 ng/mL, and were able to comply with the prescribed treatment protocol and evaluations. Exclusion criteria included prior therapy for BPH other than alpha-blockers; prior surgery of the prostate (except biopsies); uncontrolled diabetes mellitus (fasting blood glucose > 200 mg/dL); and current or past evidence of malignant disease of the prostate; active cardiac, renal, or hepatic disease as evidenced by creatinine ^ .8 mg/dL, ALT or AST ≥2.5x the upper limit of normal, history of myocardial infarction, congestive heart failure, or unstable cardiac arrhythmias within 6 months prior to study entry. Alpha-blocker therapy was not allowed during or for 14 days prior to the study. All subjects gave informed consent according to institutional guidelines. Subject characteristics are summarized in Table 2.
Table 2 Baseline Characteristics of Study Subjects (n=30)
Figure imgf000035_0001
ASSESSMENT
[0117] Pretreatment evaluation included recording of medications taken 2 weeks prior to study entry; serum chemistry panel including sodium, potassium, chloride, carbon dioxide, blood urea nitrogen (BUN), creatinine, AST and ALT; hormonal profile (FSH, LH, PRL, testosterone, free testosterone); TRUS measurement of prostate volume; PSA; uroflowmetry; and I-PSS. On Day-14 and Day-28 subjects were evaluated for prostate volume, PSA, uroflowmetry, adverse event and concomitant medication assessments. On Day-28, subjects also underwent a physical examination including digital rectal exam, temperature and vital signs; evaluation of serum chemistry; hormonal profile; and I-PSS. On Day-56 subjects were also evaluated for prostate volume and uroflowmetry. On Day-84 subjects were evaluated for uroflowmetry. On Day-112 patients were evaluated for uroflowmetry. On Day-200 subjects were evaluated for prostate volume, PSA, and uroflowmetry.
[0118] 30 patients were evaluated at baseline, 29 patients at day 28 (one drop out due to haematuria), 28 patients at day 84 (one drop-out due to noncompliance with protocol), and 26 patients at day 200 (voluntary withdrawal, lost to follow-up).
STATISTICAL ANALYSIS
[0119] The primary efficacy endpoints were the change from baseline to Day-28 in prostate volume, Qmax, I-PSS and PSA. Efficacy was analyzed by paired t-tests of the percent change in prostate volume and PSA and absolute change in Qmax and I- PSS from baseline. Non-parametric tests were also performed to examine the robustness of the results. All tests were two-sided and no adjustments were made for multiple tests. Missing data for Day-14 and Day-28 were replaced using the last observation carried forward procedure.
RESULTS
[0120] The once-daily regimen of oral LND was well tolerated with no significant therapy-related side effects. No occurrences of myalgia, testicular pain, sexual dysfunction or cardiovascular events were reported. One subject experienced hematuria apparently not related to the drug treatment but, for safety reasons, the LND treatment was stopped. One subject was lost to follow-up at 1 month. One patient had a clinically significant elevated ALT and AST values on Day 28. An abdominal ultrasound was negative. The ALT and AST returned to within normal limits. No other clinically significant serum chemistry data were reported. Three subjects had increases in FSH to above normal limits at Day-28 and two subjects had increases in LH. Testosterone levels did not significantly change, while free testosterone levels decreased, although none dropped below the lower limit of normal at Day-28.
Prostate volume
[0121] Mean total prostate volume decreased significantly during the course of treatment. At the end of the treatment (Day-28) 16 subjects (53%) had >_IO% and 7 subjects (23%) had ^>0% reduction in total prostate volume, respectively. Prostate volume continued to decrease even after subjects had been off LND therapy for four weeks. See Tables 3, 4, 5, 6 and 7.
PSA
[0122] At Day-14, PSA levels decreased modestly on average by 1.5% (95% Cl: -14.1 % to 11.2%), but by Day-28 PSA levels had dropped on average 17.8% (95% Cl: -27.2% to -8.4%; p=0.001 ). See Tables 3 and 4.
Uroflowmetry
[0123] Subjects experienced a significant increase in Qmax by Day-14 that continued through Day-28 and Day-56. At the end of the treatment 12 subjects (40%) had more than 3 mL/sec and 7 subjects (24%) more than 5 mL/sec improvement over baseline Qmax- The residual urine (post-void volume) decreased significantly from an average volume of 82.1 mL at baseline to 44.0 ml_ at Day-14, 31.6 mL at Day-28, and 34.8 mL at Day-56. See Tables 3 and 4.
I-PSS
[0124] Subjects' I-PSS symptom scores decreased from an average of 19.5 at baseline to 12.2 on Day-28 (p <0.001). The decreases were consistent across subjects. See Tables 3, 4, and 8.
Comment
[0125] The subjects selected for this study were targeted based on serum PSA > 1.0 ng/mL to ensure inclusion of subjects with a higher degree of epithelial or glandular hyperplasia versus more stromal hyperplasia. Dose levels were selected based on the extensive clinical literature published on LND. The typical dose for cancer subjects of 150 mg p.o. TID was reduced to a once-daily regimen, in order to determine whether the side effects observed in cancer subjects could be mitigated while maintaining therapeutic activity for BPH, and to help foster subject compliance. Interestingly, serum PSA levels decreased moderately at Day-14 but then dropped sharply on average at Day-28, possibly related to prostate-cell apoptosis through the disruption of the citric acid cycle. The magnitude and rapidity of this response for a compound acting on prostate volume is unique in the medical treatment of BPH. Table 3
Summary of Day 14, 28, 56, 84, 112 and 200 Efficacy Endpoints
I-PSS (score in units)
Mean SD
Absolute Absolute
Change Change
Day N from Baseline from Baseline 95% Cl P-value
28 27a -7.3 3.5 (-8.6 , -5.9) <0.001 (t-test); <0.001 (WSR)
Flow Rate (mL/sec)
Mean SD
Absolute Absolute
Change Change
Day N from Baseline from Baseline 95% Cl P-value
14 28b 3.1 5.1 (1.2 , 5.0) 0.002 (t-test); 0.002 (WSR)
28 29b 3.2 5.2 (1.3 , 5.2) 0.002 (t-test); 0.002 (WSR)
56 27 3.3 5.0 (1.3 , 5.3) 0.002 (t-test); 0.001 (WSR)
84 28 3.8 5.3 (1.7 , 5.8) 0.001 (t-test); <0.001 (WSR)
112 28 4.1 5.5 (2.0 , 6.3) <0.001 (t-test); <0.001 (WSR)
200 25 4.2 5.1 (2.1 , 6.3) <0.001 (t-test); <0.001 (WSR)
Prostate Volume
Mean SD
Percent Percent
Change Change
Day N from Baseline from Baseline 95% Cl P-value
14 30 -6.5% 10.9% (-10.6% , -2.4%) 0.003 (t-test); 0.002 (WSR)
28 29b -11.2% 15.2% (-16.8% , -5.5%) <0.001 (t-test); <0.001 (WSR)
56 28 -12.0% 13.7% (-17.3% , -6.7%) <0.001 (t-test); <0.001 (WSR)
200 28 -4.3% 18.4% (-11.7% , 3.1 %) 0.241 (t-test); 0.179 (WSR)
PSA
Mean SD
Percent Percent
Change Change
Day N from Baseline from Baseline 95% Cl P-value
14 28b -1.5% 33.9% (-14.1% , 11.2%) 0.814 (t-test); 0.558 (WSR)
28 29b -17.8% 25.2% (-27.2% , -8.4%) 0.001 (t-test); <0.001 (WSR)
200 26 -14.8% 27.8% (-26.0% , -3.6%) 0.012 (t-test); 0.017 (WSR)
Post Micturitional Residue (ml_)
Mean SD
Absolute Absolute
Change Change
Day N from Baseline from Baseline 95% Cl P-value
14 28b -38.1 61.0 (-60.9 , -15.3) 0.002 (t-test); 0.001 (WSR)
28 29b -50.5 64.9 (-74.7 , -26.3) <0.001 (t-test); <0.001 (WSR)
56 28 -44.9 61.5 (-68.8 , -21.1) 0.001 (t-test); <0.001 (WSR)
84 28 -40.3 64.5 (-65.3 , -15.3) 0.003 (t-test); 0.001 (WSR)
112 28 -34.2 54.4 (-55.3 , -13.1) 0.003 (t-test); 0.002 (WSR)
200 25 -41.9 63.2 (-68.0 , -15.8) 0.003 (t-test); 0.004 (WSR) a Last observation carried forward for 1 patient without Day 28 data and 2 patients with partially incomplete
Day 28 data (analyses include 30 patients). b Last observation carried forward for patients with missing data (analyses include 30 patients). WSR = Wilcoxon signed rank test
Table 4 Summary of Day 14, 28, 56, 84, 112 and 200 Efficacy Endpoints
I-PSS (score in units)
Mean SD Mean Absolute Change Mean Percent Change
Day N (units) (units) from Baseline (units) from Baseline
0 30 19.5 2.9 NA NA
28 27a 12.2 4.4 -7.3 -37.6%
Flow Rate (mL/sec)
Mean SD Mean Absolute Change Mean Percent Change
Day N (mL/sec) (mL/sec) from Baseline (mL/sec) from Baseline
0 30 9.4 2.7 NA NA
14 28b 12.5 5.5 3.1 38.2%
28 29b 12.6 6.0 3.2 35.3%
56 27 12.7 5.4 3.3 38.3%
84 28 13.2 5.7 3.8 43.8%
112 28 13.5 5.9 4.1 47.2%
200 25 13.7 5.5 4.2 48.3%
Prostate Volume
Mean SD Mean Absolute Change Mean Percent Change
Day N (CC) (CC) from Baseline (cc) from Baseline
0 30 55.4 25.4 NA NA
14 30 52.0 25.0 -3.4 -6.5%
28 29b 49.6 24.5 -5.9 -11.2%
56 28 48.2 23.5 -6.2 -12.0%
200 26 52.9 24.4 -2.3 -4.3%
PSA
Mean SD Mean Absolute Change Mean Percent Change
Day N (ng/rπL) (ng/mL) from Baseline (ng/mL) from Baseline
0 30 3.6 1.9 NA NA
14 28b 3.6 2.2 0.0 -1.5%
28 29b 2.8 1.5 -0.7 -17.8%
200 26 3.1 1.8 -0.5 -14.8%
Post Micturitional Residue (mL)
Mean SD Mean Absolute Change Mean Percent Change
Day N (mL) (mL) from Baseline (mL) from Baseline
0 30 82.1 64.1 NA NA
14 28b 44.0 62.5 -38.1 -45.7%
28 29b 31.6 45.3 -50.5 -46.6%
56 28 34.8 41.1 -44.9 -49.6% 84 28 39.5 52.6 -40.3 -42.8 %
112 28 45.6 43.8 -34.2 -33.3%
200 25 39.0 40.3 -41.9 -38.0% a Last observation carried forward for 1 patient without Day 28 data and 2 patients with partially incomplete
Day 28 data (analyses include 30 patients).
Last observation carried forward for patients with missing data (analyses include 30 patients). Baseline equals Day 0 value _
Table 5
Patients with reduction in total prostate volume (TPV) of at least 10% or of at least
20% compared to baseline
Figure imgf000040_0001
Table 6 Percent of patients showing improvement in total prostate volume (TPV)
Figure imgf000040_0002
Table 7 Changes in transition zone volume of prostate
Figure imgf000040_0003
Table 8 Improvement in I-PSS 28 days After Study Initiation
Figure imgf000041_0001
Table 9 Hormone Profile Summary
Figure imgf000041_0002
Interim Results
[0126] Interim results are shown for 15 subjects measured at baseline and 14 days. Of the 15 subjects, 8 subjects were also measured at 28 days. Of these 8, 7 subjects were also measured at 56 days. Where shown, statistical significance was determined using the Wilcox signed rank test. Because the study was ongoing, the final results of the completed study (above) may differ somewhat from those presented below. However, the overall trend from the interim results, which show a decrease in prostate size, an increase in urine flow and a reduction in AUASI, is expected to be maintained.
Table 10 shows the change in prostate volume compared to baseline in subjects treated with lonidamine.
[0127] Urine flow was measured at 0, 14, 28 and 56 days for all individuals treated with lonidamine . The % increase in urine flow from baseline was 40.1% at 14 days (p = 0.057; N=13), 52.2 % at 28 days (p = 0.016; N=8) and 37.0% at 56 days (p= 0.204; N=7).
[0128] Table 11 shows AUA and PSA scores for 8 individuals treated with lonidamine measured at baseline and 28 days.
TABLE 10
Figure imgf000042_0001
TABLE 11
Figure imgf000042_0002
[0129] These interim clinical trial results demonstrate that administration of lonidamine to individuals with BPH can results in striking changes in prostate size, urine flow, PSA levels and AUASI scores consistent with a significant therapeutic effect on BPH.
10. References Cited
Ansari et al., 1998, "Long-term sequelae of tolnidamine on male reproduction and general body metabolism in rabbits" Contraception 57:271-79.
Ansel et al., 1999, Pharmaceutical Dosage Forms and Drug Delivery Systems 7th ed. Lippincott Williams & Wilkins, Philadelphia: pp. 1-562.
Barry et a/., 1992, "Symptom index for benign prostatic hyperplasia" J Urol. 148:1549-57.
Barry et a/., 2001 , "Measuring the symptoms and health impact of benign prostatic hyperplasia and its treatments" BENIGN PROSTATIC HYPERPLASIA (5TH INTERNATIONAL CONSULTATION ON BPH). Health Publication, Ltd.
Barry et a/., 2003, "Benign Prostatic Hyperplasia in SCIENTIFIC AMERICAN MEDICINE, Dale and Federman Eds., WebMD Inc.
Burger, 1991 , "Isosterism and Bioisosterism in Drug Design" A. Prog. Drug Res. 37:287-371.
Cheng et a/., 2001 , "Two new male contraceptives exert their effects by depleting germ cells prematurely from the testis" Biol Reprod. 65:449-61.
Corsi et al., 1976, "I-Halobenzyl-I H-Indazole-S-Carboxylic Acids. A New Class of Antispermatogenic Agents", Journal of Medicinal Chemistry 19:778-83.
Costello & Franklin, 2000, "The intermediary metabolism of the prostate: a key to understanding the pathogenesis and progression of prostate malignancy" Oncology 59:269-82
Costello et a/., 1999, "Citrate in the diagnosis of prostate cancer" Prostate 38:237-45.
Costello et a/., 2000, "Zinc causes a shift toward citrate at equilbrium of the m- aconitase reaction of prostate mitochondria" J. Inorganic Biochemistry 78:161-65. Fanciulli et al., 1996, "Effect of the antitumor drug lonidamine on glucose metabolism of adriamycin-sensitive and -resistant human breast cancer cells" Oncology Research 3:111-120
Floridi et al., 1981 , "Effect of lonidamine on the energy metabolism of Ehrlich ascites tumor cells" Cancer Res. 41 :4661-6.
Franklin et al; 1995, "Regulation of citrate metabolism by androgen in the LNCaP human prostate carcinoma cell line." Endocrine 3:603-607
Gatto et al., 2002, "Recent studies on lonidamine, the lead compound of the antispermatogenic indazol-carboxylic acids" Contraception 65:277-78.
Grima et al., 2001 ; 2001 , "Reversible inhibition of spermatogenesis in rats using a new male contraceptive, 1-(2,4-dichlorobenzyl)-indazole-3-carbohydrazide" Biol Reprod. 64:1500-8.
Heywood et al., "Toxicological studies on 1-substituted-indazole-3-carboxylic acids." 1981 , Chemotherapy 27:91-97.
Jeyaraj et al., 2000, "Effects of long-term administration of androgens and estrogen on rhesus monkey prostate: possible induction of benign prostatic hyperplasia" J Androl. 21 :833-41.
Kaplan, 2000 "Correspondence re: M. Fanciulli et al., Energy metabolism of human LoVo colon carcinoma cells: correlation to drug resistance and influence of lonidamine." Clin Cancer Res. 6:4166-7.
Kurhanewicz et al., 1991 , "31P Spectroscopy of the human prostate gland in vivo using a transrectal probe" Magnetic Resonance in Medicine 22:404-13.
Kurhanewicz et al., 2000, Radiol CHn North Am 38:115-38.
Lee et al., 1998, "Chronology and urodynamic characterization of micturition in neurohormonally induced experimental prostate growth in the rat" Neurourol Urodyn.17:55-69.
Lobl et al., 1981 , "Effects of Lonidamine (AF 1890) and its analogues on follicle-stimulating hormone, luteinizing hormone, testosterone and rat androgen binding protein concentrations in the rat and rhesus monkey" Chemotherapy 27:61 - 76.
Lohiya et al., 1991 , "Antispermatogenic effects of tolnidamine in langur (Presbytis entellus)" Contraception 43:485-96.
Mariotti et al., 1982, "Collagen and cellular proliferation in spontaneous canine benign prostatic hypertrophy" J Urol. 127:795-7. Marshall, 1979."SoNd Oral Dosage Forms," MODERN PHARMACEUTICS, Vol. 7, (Banker and Rhodes, editors), pp. 359-427.
Mendelsohn, 1991 , "Principles of Neoplasia" in HARRISON'S PRINCIPLES OF INTERNAL MEDICINE, Wilson ed., McGraw-Hill, New York, p. 1576.
Narayan and Kurhanewicz, 1992, "Magnetic resonance spectroscopy in prostate disease: diagnostic possibilities and future developments." Prostate Suppl. 4:43-50.
Patani and LaVoie, 1996, "Bioisosterism: A Rational Approach in Drug Design" Chem. Rev. 96:3147-76.
Silvestrini et al., 1984, Prog. Med. Chem. 21, G. P. Ellis and G.B. West, Eds. (Elsevier Science Publishers, Amsterdam), p.111-35.
Silvestrini, 1981 , "Basic and Applied Research n the Study of Indazole Carboxylic Acids" Chemotherapy 27:9-20.
Thomas et al., 1990, "1H MR spectroscopy of normal and malignant human prostates in vivo." Journal of Magnetic Resonance 87:610-19.
Yacobi and Halperin-Walega, 1988, in Oral Sustained Release Formulations: Design and Evaluation, Pergamon Press
US 2003/0072814 "Topical Pharmaceutical Composition for the Treatment of Warts".
US 2002/0077300 "Screening Method for Cancer Therapeutics and Stable Antitumor Drug".
US 2002/0035071 "Mimicking the Metabolic Effects of Caloric Restriction by Administration of Glucose Antimetabolites".
US 3,895,026 "Substituted 1-benzyl-1 H-indazole-3-carboxylic Acids and derivatives thereof.
US 4,970,075 "Controlled Release Bases for Pharmaceuticals".
US 5,266,331 "Controlled Release Oxycodone Compositions".
US 5,286,493 "Stabilized Controlled Release Formulations Having Acrylic Polymer Coating".
US 5,386,467 "Controlled Release Coatings Derived From Aqueous Dispersions of Zein".
US 5,472,712 "Controlled Release Formulations Coated With Aqueous Dispersions of Ethylcellulose". US 5,478,577 "Methods of Treating Pain by Administering 24 Hour Oral Opioid Formulations Exhibiting Rapid Rate of Initial Rise of Plasma Drug Level".
US 5,549,912 "Controlled Release Oxycodone Compositions".
US 5,643,883 "Glucose-6-Phosphate Uptake Inhibitors and Novel Uses Thereof. US 5,652,273 "Reduction of Hair Growth".
US 5,672,360 "Method of Treating Pain by Administering 24 Hour Oral Opioid Formulations".
US 5,824,665 "Reduction of Hair Growth".
US 5,958,459 Opioid Formulations Having Extended Controlled Released".
US 5,968,551 Orally Administrable Opioid Formulations Having Extended Duration of Effect".
US 6,001 ,865 "3-substituted 1-benzyl-1H-indazole derivatives as antifertility agents" .
US 6,077,533 "Powder-Layered Oral Dosage Forms".
US 6,103,261 "Opioid Formulations Having Extended Controlled Release".
US 6,129,933 "Stabilized Controlled Release Substrate Having a Coating Derived From an Aqueous Dispersion of Hydrophobic Polymer".
US 6,143,322 "Method of Treating Humans With Opioid Formulations Having Extended Controlled Release".
US 6,143,353 "Controlled Release Formulations Coated With Aqueous Dispersions of Acrylic Polymers".
US 6,146,658 "Prodrugs, their preparation and use as pharmaceuticals".
US 6,218,435 "Reduction of Hair Growth".
US 6,294,195 Orally Administrable Opioid Formulations Having Extended Duration of Effect".
US 6,670,330 "Cancer Chemotherapy With 2-Deoxy-D-Glucose".
WO 01/82926 "Manipulation of Oxidative Phosphorylation for Hypersensitizing Tumor Cells to Glycolytic Inhibitors".
[0130] Although the present invention has been described in detail with reference to specific embodiments, those of skill in the art will recognize that modifications and improvements are within the scope and spirit of the invention, as set forth in the claims which follow. All publications and patent documents (patents, published patent applications, and unpublished patent applications) cited herein are incorporated herein by reference as if each such publication or document was specifically and individually indicated to be incorporated herein by reference. Citation of publications and patent documents is not intended as an admission that any such document is pertinent prior art, nor does it constitute any admission as to the contents or date of the same. The invention having now been described by way of written description and example, those of skill in the art will recognize that the invention can be practiced in a variety of embodiments and that the foregoing description and examples are for purposes of illustration and not limitation of the following claims.

Claims

1. A method for treating benign prostatic hyperplasia (BPH) comprising administering a therapeutically effective amount of lonidamine or a lonidamine analog to a human subject in need of such treatment.
2. A method of reducing prostate size in a human subject, comprising administering a therapeutically effective amount of lonidamine or a lonidamine analog to the subject.
3. A method for prophylaxis of BPH comprising administering a prophylactically effective amount lonidamine or a lonidamine analog to a human subject.
4. A method for treating BPH comprising (a) diagnosing BPH in a patient, (b) administering lonidamine or a lonidamine analog to the patient and (c) determining whether one or more manifestations of BPH are reduced in said patient.
5. A method for treating BPH comprising (a) administering lonidamine or a lonidamine analog to a patient diagnosed with BPH and (b) determining whether one or more manifestations of BPH are reduced in said patient.
PCT/US2005/027133 2004-07-29 2005-07-29 Treatment of benign prostatic hyperplasia WO2006015283A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2007523870A JP2008508316A (en) 2004-07-29 2005-07-29 Treatment of benign prostatic hyperplasia

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US59283304P 2004-07-29 2004-07-29
US60/592,833 2004-07-29
US66106705P 2005-03-11 2005-03-11
US60/661,067 2005-03-11

Publications (2)

Publication Number Publication Date
WO2006015283A2 true WO2006015283A2 (en) 2006-02-09
WO2006015283A3 WO2006015283A3 (en) 2006-06-22

Family

ID=35787884

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/027133 WO2006015283A2 (en) 2004-07-29 2005-07-29 Treatment of benign prostatic hyperplasia

Country Status (2)

Country Link
JP (1) JP2008508316A (en)
WO (1) WO2006015283A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9989861B2 (en) 2004-04-14 2018-06-05 Asml Netherlands B.V. Lithographic apparatus and device manufacturing method
EP3564214A1 (en) 2018-05-04 2019-11-06 Universita' Degli Studi G. D Annunzio Chieti - Pescara Indazole derivatives as modulators of the cannabinoid system
US11084789B2 (en) 2016-01-14 2021-08-10 Beth Israel Deaconess Medical Center, Inc. Mast-cell modulators and uses thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5458597A (en) * 1993-11-08 1995-10-17 Zomed International Device for treating cancer and non-malignant tumors and methods

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5458597A (en) * 1993-11-08 1995-10-17 Zomed International Device for treating cancer and non-malignant tumors and methods

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9989861B2 (en) 2004-04-14 2018-06-05 Asml Netherlands B.V. Lithographic apparatus and device manufacturing method
US11084789B2 (en) 2016-01-14 2021-08-10 Beth Israel Deaconess Medical Center, Inc. Mast-cell modulators and uses thereof
EP3564214A1 (en) 2018-05-04 2019-11-06 Universita' Degli Studi G. D Annunzio Chieti - Pescara Indazole derivatives as modulators of the cannabinoid system

Also Published As

Publication number Publication date
JP2008508316A (en) 2008-03-21
WO2006015283A3 (en) 2006-06-22

Similar Documents

Publication Publication Date Title
US20050272795A1 (en) Prevention and treatment of benign prostatic hyperplasia
Dunn et al. Tamsulosin: a review of its pharmacology and therapeutic efficacy in the management of lower urinary tract symptoms
BG61798B1 (en) Method and pharmaceutical composition for the treatment of prostatic hyperplasia based on 5alpha -reductase inhibitor
US20070197666A1 (en) Pharmaceutical combination for the treatment of benign prostatic hyperplasia or for the long-term prevention of acute urinary retention
EA013399B1 (en) Treating benign prostate hyperplasia with a selective androgen receptor modulator (sarm)
US11963949B2 (en) Method for treating obesity, diabetes, cardiovascular and kidney diseases by regulating GPR30/GPER activity
WO2006015283A2 (en) Treatment of benign prostatic hyperplasia
TW201400121A (en) Method for treating gynecological diseases
ZA200505396B (en) Treatment of benign prostatic hyperplasia
US20240041899A1 (en) Solid preparation
US7425565B2 (en) Use of benzothiopenes to treat and prevent prostate cancer
JP2006508992A (en) Pharmaceutical composition comprising an LTB4 antagonist and a COX-2 inhibitor or a combined COX1 / 2 inhibitor
KR20220016807A (en) Spray Dried Formulation of Pyridazinone TRPC5 Inhibitor
JP2004529182A (en) Pharmaceutical composition
ZA200407057B (en) Pharmaceutical combination for the treatment of benign prostatic hyperplasia or for the long-term prevention of acute urinary retention

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2007523870

Country of ref document: JP

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase