IE20070466A1 - Selective estrogen receptor modulators - Google Patents
Selective estrogen receptor modulators Download PDFInfo
- Publication number
- IE20070466A1 IE20070466A1 IE20070466A IE20070466A IE20070466A1 IE 20070466 A1 IE20070466 A1 IE 20070466A1 IE 20070466 A IE20070466 A IE 20070466A IE 20070466 A IE20070466 A IE 20070466A IE 20070466 A1 IE20070466 A1 IE 20070466A1
- Authority
- IE
- Ireland
- Prior art keywords
- compound
- nhr
- alkyl
- och
- tcx
- Prior art date
Links
Landscapes
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
One aspect of the present invention relates to compounds and derivatives thereof generated from a virtual screening process, and their use as estrogen receptors and as such may be useful for treatment or prevention of a variety of conditions related to estrogen functioning including: bone loss, bone fractures, osteoporosis, cartilage degeneration, endometriosis, uterine fibroid disease, hot flashes, increased levels of LDL cholesterol, cardiovascular disease, impairment of cognitive functioning, cerebral degenerative disorders, restenosis, gynecomastia, vascular smooth muscle cell proliferation, obesity, incontinence, and cancer, in particular of the breast, uterus and prostate.
Description
Selective Estrogen Receptor Modulators
BACKGROUND OF THE INVENTION ! ι , . ί ..
i
Structure-based virtual screening (VS) is typically performed by docking a f 3S molecule into a receptor active site and determining the optimal orientation by conformational, translational and rotational movement. Bajorath, J., Integration of virtual and high-throughput screening. Nat Rev Drug Discov 2002, 1, (11), 882-94; Kitchen, D. B.; Decornez, H.; Furr, J. R.; Bajorath, J., Docking and scoring in virtual screening for drug discovery: methods and applications. Nat Rev Drug Discov 2004, 3, (11), 935-49; and Schneider, G.; Bohm, H. J., Virtual screening and fast automated docking methods. Drug Discov Today 2002, 7, (1), 64-70. Subsequent scoring of these complexes is undertaken to assess the correct binding modes of the complexes, allowing ranking by affinity. Lyne, P.
D., Structure-based virtual screening: an overview. Drug Discov Today 2002, 7, (20), 104755. This ranking allows prioritization and selection of compounds for biological testing.
Several studies have examined the ability of docking and scoring combinations to retrieve a set of known actives from databases of decoys. Kellenberger, E.; Rodrigo, J.; Muller, P.; Rognan, D., Comparative evaluation of eight docking tools for docking and virtual screening accuracy. Proteins 2004, 57, (2), 225-42; Perola, E.; Walters, W. P.; Charifson, P. S., A detailed comparison of current docking and scoring methods on systems of pharmaceutical relevance. Proteins 2004, 56, (2), 235-49; Schulz-Gasch, T.; Stahl, M., Binding site characteristics in structure-based virtual screening: evaluation of current docking tools. J Mol Model (Online) 2003,9, (1), 47-57; Warren, G. L., Molecular docking and high-throughput screening for novel inhibitors of protein tyrosine phosphatase-IB. J Med Chem 2005; and Bissantz, C.; Folkers, G.; Rognan, D., Protein-based virtual screening of chemical databases. 1. Evaluation of different docking/scoring combinations. J Med Chem 2000,43, (25), 4759-67. Evaluation of their efficacy has been determined through analyses of Enrichment (E) rates or their ability to ‘correctly’ reproduce binding modes observed in crystal structures as measured by RMSD. Perola, E.; Walters, W. P.; Charifson, P. S., A detailed comparison of current docking and scoring methods on systems of pharmaceutical relevance. Proteins 2004, 56, (2), 235-49; and Krovat, Ε. M.; Langer, T., Impact of scoring functions on enrichment in docking-based virtual screening: an application study on renin inhibitors. J Chem Inf Comput Sci 2004,44, (3), 1123-9. The
minimum requirement of a docking tool would be to reproduce known binding modes for a set of complexes as measured by RMSD, however, this metric loses information about intermolecular interactions focussing only on ligand co-ordinates. Cole et al importantly point out that RMSD calculations can be flawed because docked solutions can exhibit a low RMSD, but they can also have substituents oriented incorrectly with respect to residues of the active site. Cole, J. C.; Murray, C. W.; Nissink, J. W.; Taylor, R. D.; Taylor, R., Comparing protein-ligand docking programs is difficult. Proteins 2005, 60, (3), 325-32. Marcou et al recommend omitting the use of RMSD as a measure of quantifying docking success. Marcou, G.; Rognan, D., Optimizing Fragment and Scaffold Docking by Use of Molecular Interaction Fingerprints. J. Chem. Inf. Model. 2006. It is imperative that good enrichment rates are achieved based on accurate identification of true binding modes, to ensure false positives resulting from conformational artifacts are minimized. Recent studies have focused on the use of interaction fingerprints as a post-docking strategy to reduce false positives. Marcou, G.; Rognan, D., Optimizing Fragment and Scaffold Docking by Use of Molecular Interaction Fingerprints. J. Chem. Inf. Model. 2006; Deng, Z.; Chuaqui, C.; Singh, J., Structural interaction fingerprint (SIFt): a novel method for analyzing threedimensional protein-ligand binding interactions. J Med Chem 2004,47, (2), 337-44; and Deng, Z.; Chuaqui, C.; Singh, J., Knowledge-based design of target-focused libraries using protein-ligand interaction constraints. J Med Chem 2006,49, (2), 490-500. Rognan’s group show scoring by similarity of these fingerprints outperforms conventional scoring functions. The importance of post-docking filters has also been highlighted with the introduction of standalone programs such as VISCANA, Silver, and PostDock. Amari, S.; Aizawa, M.; Zhang, J.; Fukuzawa, K.; Mochizuki, Y.; Iwasawa, Y.; Nakata, K.; Chuman, H.; Nakano, T., VISCANA: visualized cluster analysis of protein-ligand interaction based on the ab initio fragment molecular orbital method for virtual ligand screening. J Chem Inf Model 2006, 46, (1), 221-30; Silver:
www.ccdc.cam.ac.uk/products/life_sciences/gold/index.php#silver; and Springer, C.; Adalsteinsson, H.; Young, Μ. M.; Kegelmeyer, P. W.; Roe, D. C., PostDOCK: a structural, empirical approach to scoring protein ligand complexes. J Med Chem 2005, 48, (22), 682131.
However, to date, no truly generic virtual screening (VS) platform exists to produce high hit retrieval rates across numerous targets, indeed a tendency towards delivery of generic screening tools has negatively impacted on the perceived robustness of virtual
TCX-003.88
screening as a discovery tool. Selecting the most appropriate docking tool that will produce the best result for a given target is not straightforward. Herein a three step scoring and filtering procedure is described, furnishing target specific virtual screening (TS-VS), which serves to minimize false positives resulting from conformational artifacts of the docking process. This approach may be widely applied through analysis of key protein-ligand contacts for a variety of targets (e.g. Estrogen Receptor alpha (ERa).
ESTROGEN RECEPTORS
The estrogen receptor is responsible, among other functions, as a ligand-inducible nuclear transcription factor, for the mediation of the physiological effects of estrogen steroid hormones. Katzenellenbogen, B.S. Estrogen receptors: bioactivities and interactions with cell signalling pathways. Biol. Reprod. 1996, 54,287-293. Through binding to the ligand-binding domain of the receptor, hormone ligands initiate a cascade of molecular and biochemical events which ultimately can express themselves in the growth of certain tissues through the activation or inactivation of particular genes. Beato, M.; Sanchez-Pacheco, A. Interaction of steroid hormone receptors with the transcription initiation complex. Endocr. Rev. 1996,17, 587-609. Non-steroidal antiestrogens, by definition, antagonise the activity of estrogenic species. One such compound is tamoxifen ((Z)-l-[4-(2-dimethylamino-ethoxy)-phenyl]-l,2-diphenyl-l-butene), which has been used extensively in the treatment of hormone-sensitive breast cancers, and has become the firstline endocrine therapy for all stages of breast cancer in pre- and post-menopausal women. Lerner, L.J.; Jordan, V.C. Development of antiestrogens and their use in breast cancer : Eighth Cain Memorial Award Lecture. Cancer Res. 1990,50,4177-4189. Now classified as a selective estrogen receptor modulator (SERM) by virtue of its estrogen-like effects in certain tissues, the antiestrogenic properties of this compound are related to its ability to compete for estrogen binding sites in target tissues such as the breast. In very general terms, the estrogen receptors (ER) function as follows upon receipt/binding of a suitable ligand to the ligand binding domain (LBD). Agonist (estrogen) binding to the LBD causes receptor dissociation from its location in heat-shock protein, dimerisation and eventual transcription. The physical binding of a ligand initiates a conformational change in the receptor. If the ligand is an agonist, the receptor folds in such a way that helix 12 (Hl 2) of the protein closes tightly over the top of the ligand binding domain, and this folding forms a hydrophobic cavity or cleft on the surface of the protein. This cleft acts as a nuclear receptor coactivator binding site on the surface and facilitates nuclear transcription by the
TCX-003.88
receptor. Feng W, Ribeiro RCJ, Wagner RL, Nguyen H, Apriletti JW, Fletterick RJ, Baxter JD, Kushner PJ, West BL: Hormone dependant coactivator binding to a hydrophobic cleft on nuclear receptors. Science (1998) 280:1747-1749. Contrarily, on binding an antagonist, Hl2 can no longer fold over the LBD and the coactivator binding site is not formed, thus preventing the receptor from fulfilling its role in transcription. The ER should not be thought of as the controller of gene transcription but rather as a choreographer in the transcription ballet. The action of a compound as a selective estrogen receptor modulator may be rationalised as follows. Different modulators will interact with the ligand binding domain of an estrogen receptor in differing ways so as to subtly affect not only the orientation of the region identified as Hl 2 within the ligand binding domain of the receptor, but the overall bound conformation of the entire receptor. Such conformational changes and differences in receptor-ligand complexes inherently affect the coactivator binding sites of the receptor. This affection of coactivator binding sites influences the manner and degree to which the receptor will function in its transcription role - the potential for different levels of estrogenic or antiestrogenic activity may be rationalised - the basis of SERM action.
It was thought until quite recently that these physiological effects arose through the influence of a single receptor. The discovery of a second receptor subtype, resulted in the classification of two isoforms - the ERa and ERp. Kuiper GGJM, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA: Cloning of a novel estrogen receptor expressed in rat prostrate and ovary. Proc Natl Acad Sci USA (1996) 93:5925-5930. Even more recent is the discovery of perhaps another variant of isoform ERP - ERP2 - with significant variation in its ligand-binding domain. It is thought that this ‘new’ isoform may function as a negative regulator of estrogen action. Maruyama K, Endoh H, Sasaki-Iwaoka H, Kanou H, Shimaya E, Hashimoto S, Kato S, Kawashima H: A novel isoform of the rat estrogen receptor beta with 18 amino acid insertion in the ligand binding domain as a putative dominant regulator of estrogen action. Biochem Biophys Res Comm (1998) 246:142-147. The ERa dominates in reproductive tissues such as the uterus and breast, whereas ER$ has a diverse tissue distribution, being expressed in the central nervous system, the gastrointestinal tract, the kidneys and the lungs - it is the β form which predominates in the ovaries however. Both ERa and ER$ are found in breast tissue, with the alpha isoform apparently playing the more important role. Gustafsson J-A: Estrogen receptor β - a new dimension in estrogen mechanism of action. J Endocinol (1999) 163: 379-383.
TCX-003.88 |£0 70 4
Compounds which modulate the ER, as either agonists or antagonists, or in a tissue selective manner are recognised for their pharmaceutical utility in the treatment of a wide variety of estrogen-related conditions, including conditions related to the central nervous system, skeletal system, reproductive system, cardiovascular system, skin, hair follicles, immune system, bladder and prostrate as well as estrogen receptor- and non-estrogen receptor-expressing tumors. See, for example, U.S. Patent 7,138,426 (DiNinno); hereby incorporated by reference. In addition to such estrogen related conditions, some estrogen receptor modulators have been shown to inhibit the proliferation of certain cell-lines not only through estrogen antagonism, but also, through the sustained induction of programmed cell death, apoptosis. Budtz PE: Role of proliferation and apoptosis in net growth rates of human breast cancer cells (MCF-7) treated with oestradiol and/or tamoxifen. Cell Proliferat (1999) 32:289-302; and Johnston SRD, Boeddinghaus IM, Riddler S, Haynes BP, Hardcastle IR, Rowlands M, Grimshaw R, Jarman M, Dowsett M: Idoxifene antagonises estradiol-dependent MCF-7 breast cancer xenograft growth through sustained induction of apoptosis. Cancer Res (1999) 59: 3646-3651. Apoptotic cell death can be induced by a variety of drugs with diverse chemical structures and different mechanisms of action. Among the list of apoptosis-inducing agents are a wide range of anti-cancer drugs. Given the importance of the estrogen receptor and the potential application of modulators in so many disease processes the design of therapeutics which modulate this target continues to generate considerable interest both industrial and academic. Lloyd, D.G.; Meegan, M.J. Recent advances in estrogen receptor antagonists. IDrugs 2000, 3, (6) 632 -642 and references cited therein. It has been suggested that building flexibility into the rigid backbone of antiestrogens could enhance their activity and binding affinity for the estrogen receptor. Bradbury, S.P.; Mekenyan, O.G.; Ankley, G.T. The role of ligand flexibility in predicting biological activity : Structure-activity relationships for aryl hydrocarbons, estrogen and androgen receptor binding affinity. Environ. Toxicol. Chem.1998,17,15-25.
HEAT-SHOCK PROTEINS
Heat shock or stress dramatically increases cellular production of several classes of highly conserved chaperone proteins, commonly known as heat-shock proteins (HSPs). These chaperones, including the members of the HSP60, HSP70, and HSP90 families, are ATP-dependent molecules that facilitate/ensure proper client protein (e.g. protein that requires interaction with the chaperones for its activity and stability) folding, prevent nonspecific aggregations, and maintain active protein conformations.
TCX-003.88 ** 0 7 0
The HSP90 family, comprised of HSP90a and β, Grp94 and TRAP-1, represents one of the most abundant cellular proteins, accounting for 1-2% of total protein in a mammalian cell under normal conditions. HSP90 is unique among cellular chaperones in that it is not required for general co-translational protein folding but is instead dedicated to a unique set of cellular proteins, many of which are key signaling molecules critically involved in cell growth, differentiation, and apoptosis. So far over 100 proteins have been documented to associate with HSP90 and this list of client proteins is expanding rapidly.
Crystallographic studies have revealed the existence of an unconventional low affinity ATP binding cleft at their N-terminal domain that is well conserved among the four HSP90 family members. ATP binding and hydrolysis play an essential role in the regulation of chaperone functions. The occupancy of the ATP binding site by the ansamycin antibiotics geldanamycin (GM) and herbimycin A (HA), as well as the structurally unrelated fugal metabolite radicicol, inhibits the intrinsic ATPase activity of HSP90 and blocks the ATP/ADP-regulated association-dissociation cycles between HSP90 and client proteins. Consequently, ATP-competitive HSP90 inhibitors induce destabilization and eventual ubiquitin-dependent degradation of client proteins.
HSP90 has generated tremendous interest as a novel anti-cancer target following the realization that many of its clients are bona fide oncoproteins that are frequently overexpressed, mutated, or constitutively active in tumor cells. These include well known and established cancer drug targets such as receptor tyrosine kinases (HER-2/neu, epidermal growth factor receptor EGFR, Met and insulin-like growth factor-1 receptor IGF1R), metastable serine/threonine kinases (Akt and Raf-1), mutated signaling proteins (Flt3, v-Src), chimeric oncoproteins (Bcr-Abl, NPM-ALK), cell-cycle regulators (CDK4 and CDK6), transcription factors (estrogen and androgen receptors ER and AR, hypoxiainducible factor HIF-Ια) and apoptosis regulators (Survivin and Apaf-1). It is notable that HSP90 client proteins functionally contribute to all of the six “hallmarks of cancer”, which include (with examples of relevant HSP90 client proteins in parenthesis) 1) self-sufficiency in growth signals (ErbB2, Raf-1), 2) insensitivity to growth suppression signals (Plk,
Mytl), 3) evasion of apoptosis (Akt, RIP), 4) acquisition of limitless replicative potential (hTERT), 5) sustained angiogenesis,(HIF-1.alpha., FAK) and 6) invasion and metastasis (Met). The association with HSP90 ensures that these otherwise unstable oncoproteins function properly in multiple signaling pathways that are essential in maintaining the unregulated growth and the malignant phenotypes of tumors.
TCX-003.88 ΙΕΰ 7Q 4
Inactivation of HSP90 by an ATP-competitive inhibitor will induce simultaneous depletion of multiple oncoproteins and cause concurrent inhibition of various oncogenic signaling pathways. Therefore, by disrupting the function of a single molecular entity HSP90, an HSP90 inhibitor may uniquely provide a combinatorial attack on multistep oncogenesis and block all of the six hallmarks of cancer. Depending on cellular contexts, HSP90 inhibitors effectively cause growth arrest, differentiation, or apoptosis of tumor cells both in vitro and in vivo. In addition, HSP90 itself is overexpressed (about 2-20 fold) in multiple tumor types as a result of oncogenic transformation (e.g. accumulation of mutated proteins) and cellular stress (e.g. low pH and lack of nutrients). Overexpression of Hsp90 has been shown to correlate with poor prognosis in breast cancer.
Cancer cells are highly adaptive to hostile microenvironments and are capable of acquiring drug resistance, in part due to their inherent genetic instability and plasticity. Moreover, most forms of cancer are polygenic and harbor multiple signaling aberrations. HSP90 may be a key component of the very machinery that allows certain cancer cells to escape apoptotic death and evoke alternative or overlapping signaling to efficiently develop resistance to a specific drug treatment. Consequently, inhibition of HSP90 by concurrently disrupting a wide range of oncogenic pathways may prove to be a very effective approach to combat a variety of hard-to-treat tumors. The cancers include, for example, breast cancer, ovarian, prostate, chronic myelogenous leukemia (CML), melanoma, gastrointestinal stromal tumors (GISTs), master cell leukemia, testicular tumor, acute myelogenous leukemia, gastric tumor, lung, head and neck, glioblastoma, colon, thyroid, stomach, liver, multiple myeloma, renal, and lymphoma.
In addition to cancers, HSP90 inhibitors may also have the potential to treat nononcological indications where diseased cells show increased expression and usage of HSP90. These include, but are not limited to viral diseases mediated by hepatitis B virus (HBV), hepatitis C virus (HCV) and herpes simplex virus type 1 (HSV-1) as well as autoimmune diseases including those mediated by persistent lymphocyte activation. In all these cases, elevated HSP90 activity either facilitates virus assembly and replication or is required for aberrant signaling transduction in inappropriately activated lymphocyte. Furthermore, HSP90 inhibitors are also known to induce upregulation of other heat shock proteins (e.g. HSP70), which may offer neuroprotection and cardioprotection against ischemic injury as well as damages caused by protein-aggregation. Therefore, HSP90
TCX-003.88
ΙΕΟ 7 Q 4 inhibitors offer therapeutic potential in treatment of central nervous system (CNS) disorders and cardiovascular diseases.
ANGIOGENESIS
Angiogenesis is the generation of new blood vessels into a tissue or organ. Under normal physiological conditions, humans and animals undergo angiogenesis only in very specific, restricted situations. For example, angiogenesis is normally observed in wound healing, fetal and embryonal development, and formation of the corpus luteum, endometrium and placenta.
Angiogenesis is controlled through a highly regulated system of angiogenic stimulators and inhibitors. The control of angiogenesis has been found to be altered in certain disease states and, in many cases, pathological damage associated with the diseases is related to uncontrolled angiogenesis. Both controlled and uncontrolled angiogenesis are thought to proceed in a similar manner. Endothelial cells and pericytes, surrounded by a basement membrane, form capillary blood vessels. Angiogenesis begins with the erosion of the basement membrane by enzymes released by endothelial cells and leukocytes. Endothelial cells, lining the lumen of blood vessels, then protrude through the basement membrane. Angiogenic stimulants induce the endothelial cells to migrate through the eroded basement membrane. The migrating cells form a sprout off the parent blood vessel where the endothelial cells undergo mitosis and proliferate. The endothelial sprouts merge with each other to form capillary loops, creating a new blood vessel.
Persistent, unregulated angiogenesis occurs in many disease states, tumor metastases, and abnormal growth by endothelial cells. The diverse pathological disease states in which unregulated angiogenesis is present have been grouped together as angiogenic-dependent or angiogenic-associated diseases.
One example of a disease dependent on angiogenesis is ocular neovascular disease. This disease is characterized by invasion of new blood vessels into the structures of the eye, such as the retina or cornea. It is the most common cause of blindness and is involved in approximately twenty eye diseases. In age-related macular degeneration, the associated visual problems are caused by an ingrowth of choroidal capillaries through defects in Bruch's membrane with proliferation of fibrovascular tissue beneath the retinal pigment epithelium. Angiogenic damage is also associated with diabetic retinopathy, retinopathy of prematurity, corneal graft rejection, neovascular glaucoma, and retrolental fibroplasia.
TCX-003.88 «0 70 4
Other diseases associated with corneal neovascularization include, but are not limited to, epidemic keratoconjunctivitis, Vitamin A deficiency, contact lens overwear, atopic keratitis, superior limbic keratitis, and pterygium keratitis sicca. Other diseases associated with undesirable angiogenesis include Sjogren's syndrome, acne rosacea, phylectenulosis, syphilis, Mycobacteria infections, lipid degeneration, chemical bums, bacterial ulcers, fungal ulcers, Herpes simplex infection, Herpes zoster infections, protozoan infections, Kaposi's sarcoma, Mooren's ulcer, Terrien's marginal degeneration, marginal keratolysis, rheumatoid arthritis, systemic lupus, polyarteritis, trauma, Wegener's sarcoidosis, scleritis, Stevens-Johnson's disease, pemphigoid, and radial keratotomy.
Diseases associated with neovascularization include, but are not limited to, retinal/choroidal neovascularization, diabetic retinopathy, macular degeneration, sickle cell anemia, sarcoidosis, syphilis, pseudoxanthoma elasticum, Paget's disease, vein occlusion, artery occlusion, carotid obstructive disease, chronic uveitis/vitritis, Mycobacteria infections, lyme's disease, systemic lupus erythematosis, retinopathy of prematurity, Eales' disease, Behcet's disease, infections causing retinitis or choroiditis, presumed ocular histoplasmosis, Best's disease, myopia, optic pits, Stargardt's disease, pars planitis, chronic retinal detachment, hyperviscosity syndromes, toxoplasmosis, trauma and post-laser complications. Other eye-related diseases include, but are not limited to, diseases associated with rubeosis (neovascularization of the angle) and diseases caused by the abnormal proliferation of fibrovascular or fibrous tissue, including all forms of prolific vitreoretinopathy.
Another angiogenesis associated disease is rheumatoid arthritis. The blood vessels in the synovial lining of the joints undergo angiogenesis. In addition to forming new vascular networks, the endothelial cells release factors and reactive oxygen species that lead to pannus growth and cartilage destruction. Angiogenesis may also play a role in osteoarthritis. The activation of the chondrocytes by angiogenic-related factors contributes to the destruction of the joint. At a later stage, the angiogenic factors promote new bone growth. Therapeutic intervention that prevents the cartilage destruction could halt the progress of the disease and provide relief for persons suffering with arthritis.
Chronic inflammation may also involve pathological angiogenesis. Such diseases as ulcerative colitis and Crohn's disease show histological changes with the ingrowth of new blood vessels into inflamed tissues. Bartonelosis, a bacterial infection found in South
TCX-003.88 /£0 70 4
America, can result in a chronic stage that is characterized by proliferation of vascular endothelial cells. Another pathological role associated with angiogenesis is found in atherosclerosis. The plaques formed within the lumen of blood vessels have been shown to have angiogenic stimulatory activity.
Angiogenesis has been associated with a number of different types of cancer, including solid tumors and blood-bome tumors. Solid tumors with which angiogenesis has been associated include, but are not limited to, rhabdomyosarcomas, retinoblastoma, Ewing's sarcoma, neuroblastoma, and osteosarcoma. Angiogenesis is also associated with blood-bome tumors, such as leukemias, any of various acute or chronic neoplastic diseases of the bone marrow in which unrestrained proliferation of white blood cells occurs, usually accompanied by anemia, impaired blood clotting, and enlargement of the lymph nodes, liver and spleen. It is believed that angiogenesis plays a role in the abnormalities in the bone marrow that give rise to leukemia tumors and multiple myeloma diseases.
One of the most frequent angiogenic diseases of childhood is the hemangioma. A hemangioma is a tumor composed of newly formed blood vessels. In most cases the tumors are benign and regress without intervention. In more severe cases, the tumors progress to large cavernous and infiltrative forms and create clinical complications. Systemic forms of hemangiomas, hemangiomatoses, have a high mortality rate. Therapy-resistant hemangiomas exist that cannot be treated with therapeutics currently in use.
Angiogenesis is also responsible for damage found in heredity diseases such as Osler-Weber-Rendu disease, or heredity hemorrhagic telangiectasia. This is an inherited disease characterized by multiple small angiomas, tumors of blood or lymph vessels. The angiomas are found in the skin and mucous membranes, often accompanied by epitaxis (nose bleeds) or gastrointestinal bleeding and sometimes with pulmonary or hepatitic arteriovenous fistula.
Angiogenesis is also involved in normal physiological processes, such as reproduction and wound healing. Angiogenesis is an important step in ovulation and also in implantation of the blastula after fertilization. Prevention of angiogenesis could be used to induce amenorrhea, to block ovulation, or to prevent implantation by the blastula.
In wound healing, excessive repair or fibroplasia can be a detrimental side effect of surgical procedures and may be caused or exacerbated by angiogenesis. Adhesions are a frequent complication of surgery and lead to problems such as small bowel obstruction.
TCX-003.88
ΙΕο
Accordingly there is a general need in the art for effective enzyme modulators, and more specifically for potent compounds which possess a degree of molecular flexibility and can demonstrate positive induction of apoptosis in key cell lines, including pharmaceutical compositions comprising such compounds as well as methods relating to their use.
SUMMARY
Herein is disclosed the use of a target specific virtual screening (TS-VS) method to generate what are believed to be a novel anti-estrogenic chemical scaffold. In particular, the TS-VS method used herein serves to minimize false positives resulting from conformational artifacts of the docking process and is optimized to converge on novel chemotypes of estrogen receptor alpha (ERa). The application of the TS-VS method resulted in the identification of both previously known and novel putative ER scaffolds; application of distance constraints within TS-VS allowed facile identification of three novel active ligands with low ERa binding affinities. Importantly, these compounds all exhibited ERa over ERp selectivity, with the most selective being 17-fold. Remarkably, these compounds also displayed low micomolar antiproliferative activity in the human MCF-7 breast cancer cell line. Based on these results, it is believed that compounds of the invention may find utility in the treatment of a wide variety of estrogen-related conditions, including conditions related to the CNS, skeletal system, reproductive system, cardiovascular system, skin, hair follicles, immune system, bladder and prostrate, as well as estrogen receptor-and non-receptor expressing tumors.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 depicts active ligands extracted from literature. NC represents the Normalised Complementarity value. The references for each structure are: (1) Harper, M. J.; Walpole, A. L., A new derivative of triphenylethylene: effect on implantation and mode of action in rats. J Reprod Fertil 1967, 13, (1), 101-19; (2) Jones, C. D.; Jevnikar, M. G.; Pike, A. J.; Peters, Μ. K.; Black, L. J.; Thompson, A. R.; Falcone, J. F.; Clemens, J. A., Antiestrogens. 2. Structure-activity studies in a series of 3-aroyl-2-arylbenzo[b]thiophene derivatives leading to [6-hydroxy-2-(4-hydroxyphenyl)benzo[b]thien-3-yl] [4-(2-(1piperidinyl)ethoxy]-phenyl]methanone hydrochloride (LY156758), a remarkably effective estrogen antagonist with only minimal intrinsic estrogenicity. J Med Chem 1984, 27, (8), i
TCX-003.88 £θ70466
1057-66; (3) Lloyd, D. G.; Hughes, R. B.; Zisterer, D. M.; Williams, D. C.; Fattorusso, C.; Catalanotti, B.; Campiani, G.; Meegan, M. J., Benzoxepin-derived estrogen receptor modulators: a novel molecular scaffold for the estrogen receptor. J Med Chem 2004, 47, (23), 5612-5; (4) Yang, X.; Reinhold, A. R.; Rosati, R. L.; Liu, Κ. K., Enzyme-catalyzed asymmetric deacylation for the preparation of lasofoxifene (CP-336156), a selective estrogen receptor modulator. Org Lett 2000, 2, (25), 4025-7; (5) Greenberger, L. M.; Annable, T.; Collins, Κ. I.; Komm, B. S.; Lyttle, C. R.; Miller, C. P.; Satyaswaroop, P. G.; Zhang, Y.; Frost, P., A new antiestrogen, 2-(4-hydroxy-phenyl)-3-methyl-l-[4-(2-piperidinl-yl-ethoxy)-benzyl]-lH-in dol-5-ol hydrochloride (ERA-923), inhibits the growth of tamoxifen-sensitive and -resistant tumors and is devoid of uterotropic effects in mice and rats. Clin Cancer Res 2001, 7, (10), 3166-77; (6) Gottardis, Μ. M.; Jiang, S. Y.; Jeng, M. H.; Jordan, V. C., Inhibition of tamoxifen-stimulated growth of an MCF-7 tumor variant in athymic mice by novel steroidal antiestrogens. Cancer Res 1989,49, (15), 4090-3; (7) Stauffer, S. R.; Coletta, C. J.; Tedesco, R.; Nishiguchi, G.; Carlson, K.; Sun, J.;
Katzenellenbogen, B. S.; Katzenellenbogen, J. A., Pyrazole ligands: structureaffinity/activity relationships and estrogen receptor-alpha-selective agonists. J Med Chem 2000,43, (26), 4934-47; (8) Renaud, J.; Bischoff, S. F.; Buhl, T.; Floersheim, P.; Fournier, B.; Halleux, C.; Kallen, J.; Keller, H.; Schlaeppi, J. M.; Stark, W., Estrogen receptor modulators: identification and structure-activity relationships of potent ERalpha-selective tetrahydroisoquinoline ligands. J Med Chem 2003,46, (14), 2945-57; (9) Ke, Η. Z.; Simmons, H. A.; Pirie, C. M.; Crawford, D. T.; Thompson, D. D., Droloxifene, a new estrogen antagonist/agonist, prevents bone loss in ovariectomized rats. Endocrinology 1995, 136, (6), 2435-41; (10) Gauthier, S.; Caron, B.; Cloutier, J,; Dory, Y. L.; Favre, A.; Larouche, D.; Mailhot, J.; Ouellet, C.; Schwerdtfeger, A.; Leblanc, G.; Martel, C.; Simard,
J.; Merand, Y.; Belanger, A.; Labrie, C.; Labrie, F., (S)-(+)-4-[7-(2,2-dimethyl-1oxopropoxy)-4-methyl-2-[4-[2-(l-piperidinyl)- ethoxy]phenyl]-2H-l-benzopyran-3-yl]phenyl 2,2-dimethylpropanoate (EM-800): a highly potent, specific, and orally active nonsteroidal antiestrogen. J Med Chem 1997,40, (14), 2117-22; (11) Lubczyk, V.; Bachmann, H.; Gust, R., Antiestrogenically active l,l,2-tris(4-hydroxyphenyl)alkenes without basic side chain: synthesis and biological activity. J Med Chem 2003,46, (8), 148491; and (12) Meegan, M. J.; Hughes, R. B.; Lloyd, D. G.; Williams, D. C.; Zisterer, D. M., Flexible estrogen receptor modulators: design, synthesis, and antagonistic effects in human MCF-7 breast cancer cells. J Med Chem 2001,44, (7), 1072-84.
TCX-003.88 /04
Figure 2 depicts additional active ligands extracted from literature. NC represents the Normalised Complementarity value. The references for each structure are: (13) Kim,
S.; Wu, J. Y.; Birzin, Ε. T.; Frisch, K.; Chan, W.; Pai, L. Y.; Yang, Y. T.; Mosley, R. T.; Fitzgerald, P. M.; Sharma, N.; Dahllund, J.; Thorsell, A. G.; DiNinno, F.; Rohrer, S. P.; Schaeffer, J. M.; Hammond, M. L., Estrogen receptor ligands. II. Discovery of benzoxathiins as potent, selective estrogen receptor alpha modulators. J Med Chem 2004, 47, (9), 2171-5; (14) Brady, H.; Doubleday, M.; Gayo-Fung, L. M.; Hickman, M.; Khammungkhune, S.; Kois, A.; Lipps, S.; Pierce, S.; Richard, N.; Shevlin, G.; Sutherland, Μ. K.; Anderson, D. W.; Bhagwat, S. S.; Stein, B., Differential response of estrogen receptors alpha and beta to SP500263, a novel potent selective estrogen receptor modulator. Mol Pharmacol 2002,61, (3), 562-8; (15) Weatherman, R. V.; Carroll, D. C.; Scanlan, T.
S., Activity of a tamoxifen-raloxifene hybrid ligand for estrogen receptors at an AP-1 site. Bioorg Med Chem Lett 2001, 11, (24), 3129-31; (16) Grese, T. A.; Pennington, L. D.; Sluka, J. P.; Adrian, M. D.; Cole, H. W.; Fuson, T. R.; Magee, D. E.; Phillips, D. L.; Rowley, E. R.; Shetler, P. K.; Short, L. L.; Venugopalan, M.; Yang, N. N.; Sato, M.; Glasebrook, A. L.; Bryant, H. U., Synthesis and pharmacology of conformationally restricted raloxifene analogues: highly potent selective estrogen receptor modulators. J Med Chem 1998,41, (8), 1272-83; (17) Wallace, Ο. B.; Lauwers, K. S.; Jones, S. A.; Dodge, J. A., Tetrahydroquinoline-based selective estrogen receptor modulators (SERMs). Bioorg Med Chem Lett 2003,13, (11), 1907-10; (18) Blizzard, T. A.; Morgan, J. D., 2nd; Mosley, R. T.; Birzin, Ε. T.; Frisch, K.; Rohrer, S. P.; Hammond, M. L., 2-Phenylspiroindenes: a novel class of selective estrogen receptor modulators (SERMs). Bioorg Med Chem Lett 2003, 13, (3), 479-83; and (19) Sharma, A. P.; Saeed, A.; Durani, S.; Kapil, R. S., Structure-activity relationship of antiestrogens. Phenolic analogues of 2,3-diaryl-2H-lbenzopyrans. J Med Chem 1990, 33, (12), 3222-9.
Figure 3 depicts Tables 1,2, and 3. Table 1 depicts the results ofthe enrichment of inhibitors for ERa using 15 different scoring functions. Table 2 depicts the results ofthe comparison of E rates for ChemGauss2 before (wo) and after addition (w) of distance constraints. Table 3 depicts the results of the comparison of FP rates for ChemGauss2 before and after addition of distance constraints.
Figure 4 depicts [A] several known selected scaffolds identified by vHTS protocol outlined; and [B] a quinoline structure developed by American Home Products.
TCX-003.88 tin
Figure 5 depicts compounds 1-7 identified by vHTS and chosen for biochemical testing
Figure 6 depicts an X-ray of 4-hydroxytamoxifen in active site of ERa (3ERT) with docked structure of compound 1 overlaid.
Figure 7 depicts an X-ray of 4-hydroxytamoxifen in active site of ERa (3ERT) with docked structure of compound 2 overlaid.
Figure 8 depicts an X-ray of 4-hydroxytamoxifen in active site of ERa (3ERT) with docked structure of compound 3 overlaid.
Figure 9 depicts an X-ray of 4-hydroxytamoxifen in active site of ERa (3ERT) with docked structure of compound 4 overlaid.
Figure 10 depicts an X-ray of 4-hydroxytamoxifen in active site of ERa (3ERT) with docked structure of compound 5 overlaid.
Figure 11 depicts an X-ray of 4-hydroxytamoxifen in active site of ERa (3ERT) with docked structure of compound 6 overlaid.
Figure 12 depicts an X-ray of 4-hydroxytamoxifen in active site of ERa (3ERT) with docked structure of compound 7 overlaid.
Figure 13 depicts a table showing a comparison of LPC output of compound 4 docked versus 4-hydoxytamoxifen (3ERT).
Figure 14 depicts a scheme showing one synthetic approach to compounds of formula I.
DETAILED DESCRIPTION
One aspect of the present invention relates to 2-amino-4-phenyl-4H-chromene compounds and derivatives thereof, their synthesis, and their use as estrogen receptor modulators. In certain embodiments, the compounds of the instant invention are ligands for estrogen receptors and as such may be useful for treatment or prevention of a variety of conditions related to estrogen functioning including: bone loss, bone fractures, osteoporosis, cartilage degeneration, endometriosis, uterine fibroid disease, hot flashes, increased levels of LDL cholesterol, cardiovascular disease, impairment of cognitive functioning, cerebral degenerative disorders, restenosis, gynecomastia, vascular smooth
TCX-003.88 ?0 4 muscle cell proliferation, obesity, incontinence, and cancer, in particular of the breast, uterus and prostate.
The present invention relates to compounds that are capable of treating and/or preventing a variety of conditions related to estrogen functioning. One aspect of the present invention is illustrated by a compound of formula I, II, III, or IV, as well as isomers, prodrugs and pharmaceutically acceptable salts thereof:
III IV wherein R1 to R9, as well as m and n, are as defined elsewhere in the application.
It is appreciated that judicious selection of, and variation in, any oxygen or nitrogen protecting groups of the aforementioned compounds may provide enhanced potential metabolic profiles for these compounds.
From studies of the binding mode of ligands within the LBD of estrogen receptors, see below, certain compounds containing hydrogen bond donors and receptors, e.g., hydroxy-containing compounds, were found to interact strongly with glutamine, arginine and histidine amino acid residues responsible for ligand anchoring within the ER active site. Such compounds were found to exhibit high antiproliferative potencies and increased ER binding affinity.
In one embodiment the invention provides a compound wherein the compound is antiosteoporotic. In another embodiment the invention provides a compound wherein the compound inhibits the proliferation of and/or induces apoptosis in human breast cancer
TCX-003.88 cells. Preferably the invention provides a compound wherein the compound is a modulator of the estrogen receptor(s).
The invention further provides a pharmaceutical composition comprising a compound of the invention. Preferably the pharmaceutical composition is in combination with a pharmaceutically acceptable carrier or diluent. Most preferably in combination with a pharmaceutically active compound. In certain embodiments, the pharmaceutically active compound is an anti-cancer drug, e.g., cisplatin.
The pharmaceutical composition of the invention may be administered in the form of an emulsion, liposome, patch, powder and/or complex tablet, capsule, syrup, dosemetered inhaler. In certain embodiments, the pharmaceutical composition is in a form for oral, intravenous, intramuscular, intraperitoneal, intradermal, intravesicular and/or rectal administration. In one embodiment, the present invention relates to a pharmaceutical composition comprising a compound of the invention for use in the preparation of a medicament for the prophylaxis and/or treatment of estrogen related conditions and/or conditions where the induction of apoptosis is desirable. In certain embodiments, the condition is any one or more of obesity, hormone dependent breast cancer, osteoporosis, estrogen deficiency, arthritis, cardiovascular disease, ovarian cancer, artherosclerosis, colon tumor, endometriosis, Alzheimer’s disease, non-insulin dependent (type II) diabetes, infertility, prostrate tumor, melanoma, acne, hypercholesterolemia, CNS disease, contraception, conditions related to hair follicles, macular degeneration, urinary incontinence, estrogen receptor-expressing and estrogen receptor-expressing tumors, leukaemia.
In one embodiment the invention provides use of a compound of the invention in inhibiting the proliferation of and/or induction of apoptosis in breast cancer cells.
The invention also provides a compound of the invention for the preparation of a medicament for use in the prophylaxis and/or treatment of an estrogen related disease.
Most preferably in the prophylaxis and/or treatment of breast cancer.
The invention further provides a method for the treatment and/or prophylaxis of an estrogen related disease comprising administering an effective amount of a compound of the invention or a composition of the invention.
TCX-003.88
DEFINITIONS
For convenience, certain terms employed in the specification, examples, and appended claims are collected here.
The indefinite articles “a” and “an,” as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean “at least one.”
The phrase “and/or,” as used herein in the specification and in the claims, should be understood to mean “either or both” of the elements so conjoined, i.e., elements that are conjunctively present in some cases and disjunctively present in other cases. Multiple elements listed with “and/or” should be construed in the same fashion, i.e., “one or more” of the elements so conjoined. Other elements may optionally be present other than the elements specifically identified by the “and/or” clause, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
As used herein in the specification and in the claims, “or” should be understood to have the same meaning as “and/or” as defined above. For example, when separating items in a list, “or” or “and/or” shall be interpreted as being inclusive, i.e., the inclusion of at least one, but also including more than one, of a number or list of elements, and, optionally, additional unlisted items. Only terms clearly indicated to the contrary, such as “only one of’ or “exactly one of,” or, when used in the claims, “consisting of,” will refer to the inclusion of exactly one element of a number or list of elements. In general, the term “or” as used herein shall only be interpreted as indicating exclusive alternatives (i.e., “one or the other but not both”) when preceded by terms of exclusivity, such as “either,” “one of,” “only one of,” or “exactly one of.” “Consisting essentially of,” when used in the claims, shall have its ordinary meaning as used in the field of patent law.
As used herein in the specification and in the claims, the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the
TCX-003.88 /ίθ°<«6 list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified. Thus, as a nonlimiting example, “at least one of A and B” (or, equivalently, “at least one of A or B,” or, equivalently “at least one of A and/or B”) can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.
It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.
In the claims, as well as in the specification above, all transitional phrases such as “comprising,” “including,” “carrying,” “having,” “containing,” “involving,” “holding,” “composed of,” and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases “consisting of’ and “consisting essentially of’ shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03.
As used herein, the term “composition” is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
The term “therapeutically effective amount” as used herein means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician.
The terms “treating” or “treatment” of a disease as used herein includes: preventing the disease, i.e., causing the clinical symptoms of the disease not to develop in a mammal that may be exposed to or predisposed to the disease but does not yet experience or display
TCX-003.88 «0 70 4 symptoms of the disease; inhibiting the disease, i.e., arresting or reducing the development of the disease or its clinical symptoms; or relieving the disease, i.e., causing regression of the disease or its clinical symptoms.
The term “bone resorption,” as used herein; refers to the process by which osteoclasts degrade bone.
Estrogen includes, but is not limited to naturally occurring estrogens, estradiol (E2), estrone (Ei), and estriol (E3), synthetic conjugated estrogens, oral contraceptives and sulfated estrogens. See, Gruber C J, Tschugguel W, Schneeberger C, Huber J C., Production and actions of estrogens N Engl J Med 2002 January 31 ;346(5):340 52.
Estrogen receptor modulators refers to compounds which interfere or inhibit the binding of estrogen to the receptor, regardless of mechanism. Examples of estrogen receptor modulators include, but are not limited to, estrogen, progestogen, estradiol, droloxifene, raloxifene, lasofoxifene, TSE-424, tamoxifen, idoxifene, LY353381,
LY117081, toremifene, fulvestrant, 4-(7-(2,2-dimethyl-l-oxopropoxy-4-methyl-2-[4[2-(lpiperidinyl)ethoxy]phe- nyl]-2H-1 -benzopyran-3-yl]-phenyl-2,2-dimethylpropanoate, 4,4'dihydroxybenzophenone-2,4-dinitrophenyl-hydrazone, and SH646.
As used herein, Hsp90 includes each member of the family of heat shock proteins having a mass of about 90-kiloDaltons. For example, in humans the highly conserved Hsp90 family includes cytosolic Hsp90a and Hsp90a isoforms, as well as GRP94, which is found in the endoplasmic reticulum, and HSP75/TRAP1, which is found in the mitochondrial matrix.
The term administration and variants thereof (e.g., administering a compound) in reference to a compound of the invention means introducing the compound or a prodrug of the compound into the system of the animal in need of treatment. When a compound of the invention or prodrug thereof is provided in combination with one or more other active agents (e.g., a bisphosphonate, etc.), administration and its variants are each understood to include concurrent and sequential introduction of the compound or prodrug thereof and other agents. The present invention includes within its scope prodrugs of the compounds of this invention. In general, such prodrugs will be functional derivatives of the compounds of this invention which are readily convertible in vivo into the required compound. Thus, in the methods of treatment of the present invention, the term administering shall encompass the treatment of the various conditions described with the compound specifically disclosed
TCX-003.88 ISO 7q , or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Design of Prodrugs, ed. H. Bundgaard, Elsevier, 1985, which is incorporated by reference herein in its entirety. Metabolites of these compounds include active species produced upon introduction of compounds of this invention into the biological milieu.
The terms “co-administration” and “co-administering” refer to both concurrent administration (administration of two or more therapeutic agents at the same time) and time varied administration (administration of one or more therapeutic agents at a time different from that of the administration of an additional therapeutic agent or agents), as long as the therapeutic agents are present in the patient to some extent at the same time.
The term “heteroatom” is art-recognized and refers to an atom of any element other than carbon or hydrogen. Illustrative heteroatoms include boron, nitrogen, oxygen, phosphorus, sulfur and selenium.
The term “alkyl” is art-recognized, and includes saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups. In certain embodiments, a straight chain or branched chain alkyl has about 80 or fewer carbon atoms in its backbone (e.g., Ci-Cgo for straight chain, C3-C80 for branched chain), and alternatively, about 30 or fewer. Likewise, cycloalkyls have from about 3 to about 10 carbon atoms in their ring structure, and alternatively about 5, 6 or 7 carbons in the ring structure. As used herein, “fluoroalkyl” denotes an alkyl where one or more hydrogens have been replaced with fluorine; “perfluoroalkyl” denotes an alkyl where all the hydrogens have been replaced with fluorine.
Unless the number of carbons is otherwise specified, “lower alkyl” refers to an alkyl group, as defined above, but having from one to about ten carbons, alternatively from one to about six carbon atoms in its backbone structure. Likewise, “lower alkenyl” and “lower alkynyl” have similar chain lengths.
The terms “alkenyl” and “alkynyl” are art-recognized and refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
TCX-003.88 llo
The term “aralkyl” and “heteroaralkyl” is art-recognized and refers to an alkyl group substituted with an aryl group or heteroaromatic group, respectively.
The term “aryl” is art-recognized and refers to 5-, 6- and 7-membered single-ring aromatic groups, for example, benzene, naphthalene, anthracene, and the like. The aromatic ring may be substituted at one or more ring positions with such substituents as described herein, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, trifluoromethyl, cyano, or the like. The term “aryl” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are “fused rings”) wherein at least one of the rings is aromatic, e.g., the other cyclic rings may be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls.
The terms ortho, meta and para are art-recognized and refer to 1,2-, 1,3- and 1,4disubstituted benzenes, respectively. For example, the names 1,2-dimethylbenzene and ortho-dimethylbenzene are synonymous.
The terms “heterocyclyl”, “heteroaryl”, or “heterocyclic group” are art-recognized and refer to 3- to about 10-membered ring structures, alternatively 3- to about 7-membered rings, whose ring structures include one to four heteroatoms. Heterocycles may also be polycycles. Heterocyclyl groups include, for example, thiophene, thianthrene, furan, pyran, isobenzofuran, chromene, xanthene, phenoxanthene, pyrrole, imidazole, pyrazole, isothiazole, isoxazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, pyrimidine, phenanthroline, phenazine, phenarsazine, phenothiazine, furazan, phenoxazine, pyrrolidine, oxolane, thiolane, oxazole, piperidine, piperazine, morpholine, lactones, lactams such as azetidinones and pyrrolidinones, sultams, sultones, and the like. The heterocyclic ring may be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, trifluoromethyl, cyano, or the like.
TCX-003.88 ^°7°466
It should be understood that heterocycles may be attached to the rest of the compound by any atom of the heterocycle which results in the creation of a stable structure.
The terms “polycyclyl” or “polycyclic group” are art-recognized and refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are “fused rings”. Rings that are joined through non-adjacent atoms are termed “bridged” rings. Each of the rings of the polycycle may be substituted with, for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, trifluoromethyl, cyano, or the like. Examples of polycyclyls include, but are not limited to, cyclopentane, cyclopentene, indane, indene, cyclohexane, cyclohexene, cyclohexadiene, benzene, tetrahydronaphthalene, decahydronaphthalene, naphthalene, pyridine, piperidine, pyridazine, pyrimidine, pyrazine, 1,2,3-triazine, 1,2,4-triazine, 1,3,5-triazine, 1,2,3,4tetrazine, 1,2,4,5-tetrazine, 1,2,3,4-tetrahydroquinoline, quinoline, 1,2,3,4tetrahydroisoquinoline, isoquinoline, cinnoline, phthalazine, quinazoline, quinoxaline, 1,5naphthyridine, 1,6-naphthyridine, 1,7-naphthyridine, 1,8-naphthyridine, 2,6-naphthyridine, 2,7-naphthyridine, pteridine, acridine, phenazine, 1,10-phenatroline, dibenzopyrans, 1benzopyrans, phenothiazine, phenoxazine, thianthrene, dibenzo-p-dioxin, phenoxathiin, phenoxthionine, morpholine, thiomorpholine, tetrahydropyan, pyran, benzopyran, 1,4dioxane, 1,3-dioxane, dihyropyridine, dihydropyran, 1-pyrindine, quinuclidine, triazolopyridine, β-carboline, indolizine, quinolizidine, tetrahydronaphtheridine, diazaphenanthrenes, thiopyran, tetrahydrothiopyran, benzodioxane, furan, benzofuran, tetrahydrofuran, pyrrole, indole, thiophene, benzothiopene, carbazole, pyrrolidine, pyrazole, isoxazole, isothiazole, imidazole, oxazole, thiazole, 1,2,3-triazole, 1,2,4-triazole, 1,2,3oxadiazole, 1,2,4-oxadiazole, 1,3,4 oxadiazole, 1,2,5-oxadiazole, 1,2,3-thiadiazole, 1,2,4thiadiazole, 1,3,4-thiadiazole, 1,2,5 thiadiazole, tetrazole, benzothiazole, benzoxazole, benzotriazole, benzimidazole, benzopyrazole, benzisothiazole, benzisoxazole and purine.
The term “carbocyclyl” or “carbocycle” is art-recognized and refers to an aromatic or non-aromatic ring in which each atom of the ring is carbon.
TCX-003.88
ΙΕο
The definition of each expression, e.g., R, alkyl, m, n, and the like, when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure.
The abbreviations Me, Et, Ph, Tf, Nf, Ts, and Ms represent methyl, ethyl, phenyl, trifluoromethanesulfonyl, nonafluorobutanesulfonyl, p-toluenesulfonyl and methanesulfonyl, respectively. A more comprehensive list of the abbreviations utilized by organic chemists of ordinary skill in the art appears in the first issue of each volume of the Journal of Organic Chemistry; this list is typically presented in a table entitled Standard List of Abbreviations.
Certain compounds contained in compositions of the present invention may exist in particular geometric or stereoisomeric forms. In addition, polymers of the present invention may also be optically active. The present invention contemplates all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention. Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.
If, for instance, a particular enantiomer of compound of the present invention is desired, it may be prepared by asymmetric synthesis, or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomers. Alternatively, where the molecule contains a basic functional group, such as amino, or an acidic functional group, such as carboxyl, diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomers.
It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, or other reaction.
TCX-003.88
The term “substituted” is also contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, those described herein above. The permissible substituents may be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. This invention is not intended to be limited in any manner by the permissible substituents of organic compounds.
As used herein and unless otherwise indicated, the term “prodrug” means a derivative of a compound that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide a compound of this invention. Prodrugs may become active upon such reaction under biological conditions, or they may have activity in their unreacted forms. Examples of prodrugs contemplated in this invention include, but are not limited to, analogs or derivatives of compounds of formulas (I) through (IV) that comprise biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues. Other examples of prodrugs include derivatives of compounds of formulas (I) through (IV) that comprise -NO, -NO2, -ONO, or -ONO2 moieties. Prodrugs can typically be prepared using well-known methods, such as those described by 1 Burger's Medicinal Chemistry and Drug Discovery (1995) 172-178, 949-982 (Manfred E. Wolff ed., 5th ed).
As used herein and unless otherwise indicated, the terms “biohydrolyzable amide”, “biohydrolyzable ester”, “biohydrolyzable carbamate”, “biohydrolyzable carbonate”, “biohydrolyzable ureide” and “biohydrolyzable phosphate analogue” mean an amide, ester, carbamate, carbonate, ureide, or phosphate analogue, respectively, that either: 1) does not destroy the biological activity of the compound and confers upon that compound advantageous properties in vivo, such as improved water solubility, improved circulating half-life in the blood (e.g., because of reduced metabolism of the prodrug), improved uptake, improved duration of action, or improved onset of action; or 2) is itself biologically inactive but is converted in vivo to a biologically active compound. Examples of biohydrolyzable amides include, but are not limited to, lower alkyl amides, α-amino acid
TCX-003.88 amides, alkoxyacyl amides, and alkylaminoalkylcarbonyl amides. Examples of biohydrolyzable esters include, but are not limited to, lower alkyl esters, alkoxyacyloxy esters, alkyl acylamino alkyl esters, and choline esters. Examples of biohydrolyzable carbamates include, but are not limited to, lower alkylamines, substituted ethylenediamines, aminoacids, hydroxyalkylamines, heterocyclic and heteroaromatic amines, and polyether amines.
For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, “Handbook of Chemistry and Physics”, 67th Ed., 1986-87, inside cover.
While several embodiments of the present invention are described and illustrated herein, those of ordinary skill in the art will readily envision a variety of other means and/or structures for performing the functions and/or obtaining the results and/or one or more of the advantages described herein, and each of such variations and/or modifications is deemed to be within the scope of the present invention. More generally, those skilled in the art will readily appreciate that all parameters, dimensions, materials, and configurations described herein are meant to be exemplary and that the actual parameters, dimensions, materials, and/or configurations will depend upon the specific application or applications for which the teachings of the present invention is/are used. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. It is, therefore, to be understood that the foregoing embodiments are presented by way of example only and that, within the scope of the appended claims and equivalents thereto, the invention may be practiced otherwise than as specifically described and claimed. The present invention is directed to each individual feature, system, article, material, kit, and/or method described herein. In addition, any combination of two or more such features, systems, articles, materials, kits, and/or methods, if such features, systems, articles, materials, kits, and/or methods are not mutually inconsistent, is included within the scope of the present invention.
VIRTUAL SCREENING METHODS
Rather than comparing the performance of post-docking filters with scoring functions, the approach disclosed herein implemented both methods concurrently, and thereby ensured that only true binding modes were ranked. The resultant platform, TS-VS, consists of a rigid-body docking algorithm (LIGIN), a ‘rough’ scoring function TCX-003.88
Normalised Complementarity (NC), a post-docking filter - Ligand Protein Contacts (LPC), and final ranking with an empirical scoring function. TS-VS was conceived as a method to target specific biological systems of study, rather than deliver a generic tool and its validation was based on the retrieval of active modulators of the human Estrogen Receptor (ER).
As a stand-alone docking tool LIGIN has been previously tested in CASP2 experiments involving binding pocket identification, modeling the quinone binding site in the DI protein of photosystem-2 reaction centre, and the inhibitory/stimulatory binding sites for tentoxin within chloroplast FOFl-ATPase. Sobolev, V.; Moallem, Τ. M.; Wade, R. C.; Vriend, G.; Edelman, M., CASP2 molecular docking predictions with the LIGIN software. Proteins 1997, Suppl 1, 210-4; Sobolev, V.; Edelman, M., Modeling the quinoneB binding site of the photosystem-II reaction center using notions of complementarity and contact-surface between atoms. Proteins 1995,21, (3), 214-25; and Sobolev, V.; Niztayev, A.; Pick, U.; Avni, A.; Edelman, M., A proteomic approach to resolving the binding sites for tentoxin in plastid CFl-ATPase. Proceedings of the 12th International Congress on Photosynthesis. However, in the context of virtual screening, LIGIN has not yet been evaluated. The LIGIN methodology is fully described elsewhere, the main features are described in Example 1 of the Exemplification. Sobolev, V.; Wade, R. C.; Vriend, G.; Edelman, M., Molecular docking using surface complementarity. Proteins 1996,25, (1), 120-9.
LIGIN docks ligands based on maximization of surface complementarity of atoms of the ligand with those of the receptor. Atoms are assigned a chemical type (8 classes defined) according to their chemical properties and they participate in non-bonding interactions with residues of the active site. Interactions are quantified by ‘legitimate’ (complementary) and ‘illegitimate’ (uncomplimentary) contact assignments designated by the two atoms involved. The basic presumption is that two atoms will be in contact if they share a common surface area with a distance between them smaller than Ra + Rb + 2Rw, where Ra and Rb are van der Waals radii of the atoms and Rw is that of the solvent molecule. A final evaluation of the fit of a molecule in the active site is given by calculation of a complementarity function (CF):
CF = Sl-Si-£ (Eqn. 1)
TCX-003.88
where SI and Si are the sum of all ‘legitimate’ (complimentary) and ‘illegitimate’ (uncomplimentary) contact surface areas respectively between ligand and residues of receptor. E is a repulsion term similar to that used in energy force fields. A ‘wall’ term is also incorporated, similar to the repulsive term used in the Lennard-Jones potential to account for intermolecular clashes. As the CF value would be ultimately dependent on the size of the ligand, it is normalised by dividing by the solvent accessible surface of the uncomplexed ligand, producing the Normalised Complementarity value.
Post docking, the Normalised Complementarity (NC) function is calculated using Ligand Protein Contacts (LPC) software as it differs from the function in LIGIN by inclusion of a wall term that accounts for interatomic clashes. Sobolev, V,; Sorokine, A.; Prilusky, J.; Abola, Ε. E.; Edelman, M., Automated analysis of interatomic contacts in proteins. Bioinformatics 1999,15, (4), 327-32. A threshold value is set allowing removal of docked ligands overly exposed to solvent because of ‘poor’ positioning in the active site. This step is highly beneficial when applied to docking against targets such as nuclear receptors, which inherently possess well-buried cavities. The large quantity of co-crystal data for the nuclear receptor superfamily, our previous work on this target and knowledge about its modulation through key interactions with residues Glu353, Arg394, His524, Asp351 (antagonist) prompted us to use it in this study. Knox, A. J. S., Meegan M.J., Lloyd D.G, Estrogen Receptors: Molecular interactions, virtual screening and future prospects. Current Topics in Medicinal Chemistry 2006,6, (3), 211-237; Lloyd, D. G.; Hughes, R. B.; Zisterer, D. M.; Williams, D. C.; Fattorusso, C.; Catalanotti, B.; Campiani, G.; Meegan, M. J., Benzoxepin-derived estrogen receptor modulators: a novel molecular scaffold for the estrogen receptor. J Med Chem 2004,47, (23), 5612-5; and Meegan, M. J.; Hughes, R. B.; Lloyd, D. G.; Williams, D. C.; Zisterer, D. M., Flexible estrogen receptor modulators: design, synthesis, and antagonistic effects in human MCF-7 breast cancer cells. J Med Chem 2001,44, (7), 1072-84. The desire was to deliver a target specific virtual screening utility which would yield high enrichment, low false positives and be clearly validated through experiment.
Post-docking, LPC also generates information about interatomic distances between ligands and residues of the active site of the ER, with specific details about H-bonding interactions. Only those ligands exhibiting true binding modes based on two types of distance constraints, nearest-atom and H-bonding are retained. Using nearest-atom distance constraints it is possible suggest novel chemistry by permitting compounds whose scaffolds
TCX-003.88
Ιίο may be adequately oriented for ER binding but may not be revealed in a focused virtual screen due to the presence of inappropriate substituents. Alternatively, applying specific Hbonding distance constraints only those molecules bearing substituents that could interact through H-bonding to specific residues (eg. Thr347, Glu353, Leu387, Arg394) are retained. Importantly, Asp351 was not included in the filter list of essential H-bonding residues so as to permit the identification of modulating scaffolds - not necessarily antagonists - whose activity could be optimized towards agonism or antagonism by enumeration of a virtual library and follow-up synthesis.
Following application of either nearest-atom or specific H-bonding constraints, a final scoring component was executed on the remaining complexes to allow prioritization. In delivery of a target-specific virtual screening utility, selection of an appropriate scoring function component is paramount. Arbitrary or uninformed use of scoring functions can and does lead to negatively impacted hit rates and compound rankings, with increased false positive rates and multiple false negative annotations. To select the scoring function most applicable to docking in ERa, 15 popular scoring functions, i.e., X-Score, Fresno, six scoring functions implemented in Sybyl6.91 (D-Score, PMF-Score, G-Score, ChemScore, F-Score, and DrugScore), six implemented in FRED2.il (ChemScore, ChemGauss, ChemGauss2, PLP, ScreenScore, ShapeGauss) and one from Surflex (Hammerhead), were evaluated. This evaluation was performed without the implementation of distance constraints in the process so as to prevent any undue bias. The optimal individual scoring function was then selected and included as the last component in the protocol so that only reasonably docked structures were actually scored. Integration of these ‘in silico’ methods with wet-lab experiment has allowed us to optimize our suite of algorithms and discover lead compounds of the ERa, delivering a validated target specific VS platform.
A common problem in VS is that some compounds are ranked well by scoring functions post-docking although their respective pose is barely in the binding site leading to false enrichment. Leach, A. R.; Shoichet, Β. K.; Peishoff, C. E., Prediction of ProteinLigand Interactions. Docking and Scoring: Successes and Gaps. J Med Chem 2006,49, (20), 5851-5855. Secondly, and importantly, Warren et al have recently observed that from an assessment of 35 scoring functions, none were able to reliably identify the best-docked pose against a set of different targets. Warren, G. L.; Andrews, C. W.; Capelli, A. M.; Clarke, B.; Lalonde, J.; Lambert, Μ. H.; Lindvall, M.; Nevins, N.; Semus, S. F.; Senger, S.; Tedesco, G.; Wall, I. D.; Woolven, J. M.; Peishoff, C. E.; Head, M. S., A Critical
TCX-003.88
Assessment of Docking Programs and Scoring Functions. J Med Chem 2006,49, (20), 5912-5931. Obviously, this leads to difficulty in choosing the optimal docking algorithm to select in a VS campaign against a particular target. To overcome this pitfall we have introduced the TS-VS platform to ensure only realistic binders and not conformational artifacts are prioritized.
The initial scoring function discerns the ‘buriedness’ of a pose within the binding site termed Normalised Complementarity (NC) as calculated by LPC software. Sobolev,
V.; Sorokine, A.; Prilusky, J.; Abola, Ε. E.; Edelman, M., Automated analysis of interatomic contacts in proteins. Bioinformatics 1999, 15, (4), 327-32. A docked molecule producing a score of about one is one that is 100% contained in the binding site and thus the solvent accessible surface is zero. This first filter is sensitive enough to ensure that molecules are actually docked in the cavity of the receptor and not overly exposed to solvent. Figures 1 and 2 show nineteen active ligands extracted from literature with demonstrated modulation of ERa. These ligands were used to inform the process to set a threshold value (0.8) that must be overcome to allow a molecule to move to the next stage in the process.
Applying this methodology to the validation set of 1000 compounds seeded with 19 actives, only 860 passed (all seeded actives included) and docked sufficiently well within the hydrophobic cavity of the ER.
It was necessary to select the optimal scoring function at this stage before application of the distance constraint filters to prevent introduction of bias into the final scoring process. The intention, however was to use the optimal scoring function as the final component in the full VS procedure after application of the distance constraint filters to score only true binders.
The ability of 15 scoring functions to discriminate between actives and inactives from the remaining 860 docked poses was examined. Table 1 in Figure 3 depicts the Enrichment calculated for 0.6% (top 5), 1.2% (top 10), 1.8% (top 15) and 2.4% (top 20) of the ranked hitlist for each scoring function. It is immediately clear that ~ 50% of the scoring functions provide no enrichment at all. The increasing order of merit of each is D_Score = PMFScore = FScore = G_Score = Drugscore = FRED_Chemscore = HammerHead < Fresno < Chemscore < Pip < Xscore < Screenscore < Shapegauss < Chemgauss < Chemgauss2. Chemgauss2 is a smooth Gaussian function composed of shape-based
TCX-003.88
interactions between all heavy atoms, hydrogen bonding interactions, and aromatic interactions. From Table 1, Chemgauss2 is undoubtedly the best performing scoring function.
Having identified ChemGauss2 as the optimal scoring function for the ER for use in the final step in the protocol, it was necessary to take the 860 molecules that passed the NC threshold of 0.8 and re-filter applying distance constraints between the nearest interacting atom of a ligand and atom of the residues Thr347, Glu353, Leu387, Arg394. The residues, Met343, Leu349, Leu384, and His524 were added to the list for constraining, as they appear to provide additional important interactions in the binding process for antiestrogens. Manas, E. S.; Unwalla, R. J.; Xu, Ζ. B.; Malamas, M. S.; Miller, C. P.; Harris, H. A.; Hsiao, C.; Akopian, T.; Hum, W. T.; Malakian, K.; Wolfrom, S.; Bapat, A.; Bhat, R. A.; Stahl, M. L.; Somers, W. S.; Alvarez, J. C., Structure-based design of estrogen receptor-beta selective ligands. J Am Chem Soc 2004,126, (46), 15106-19. A large reduction the number of docked complexes needing to be re-scored resulted from this process, with all actives and only 52 inactives passing this stage. It is important to note that this reduction in the docked molecule listing makes prioritization significantly easier for any scoring function and assists in ranking - particularly if sub-optimal scoring functions are the only tools available to the cheminformatician. Corroborating this, Chuaqui et al described a ID profile-based approach, Structural Interaction Fingerprint (p-SIFT) to filter out poorly docked poses and found that once incorrect poses that contribute to false positive scores were removed, differences in the performance of individual scoring functions were factored out. Chuaqui, C.; Deng, Z.; Singh, J., Interaction profiles of protein kinase-inhibitor complexes and their application to virtual screening. J Med Chem 2005,48, (1), 121-33.
As illustrated in Table 2 in Figure 3, the optimized protocol significantly improves Enrichment over the dataset. Prior to incorporation of distance constraints the E rates observed using Chemgauss2 were 36.21 in the first 0.6% and 31.68 for the first 1.2% respectively. On addition of the target-specific constraints to ‘focus’ the docking, a maximum enrichment of 45.26 was observed for both 0.6% and 1.2% levels of the dataset. What is important to note at this stage is that no specific H-bonding constraints have been applied, but only interatomic distance constraints. Halgren et al also point out that the common definition of Enrichment does not account for the actual rank of each active in a scored hitlist, and for this reason we also calculate False Positive (FP) rates for our program in the validation process as another indicator of success. Again it can be observed that the
TCX-003.88
FP rates are significantly lower after introduction of distance constraints, as evidenced in Table 3 in Figure 3. Halgren, T. A.; Murphy, R. B.; Friesner, R. A.; Beard, H. S.; Frye, L. L.; Pollard, W. T.; Banks, J. L., Glide: a new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screening. J Med Chem 2004,47, (7), 1750-9.
Finally, to test more rigorously the procedure and its application the ER, 10,000 compounds (known inactive but with similar molecular properties to known antiestrogens) were screened, seeded with a single known antiestrogen used previously by us to show the importance of pre-processing a database prior to docking. Knox, A. J.; Meegan, M. J.; Carta, G.; Lloyd, D. G., Considerations in compound database preparation -- hidden impact on virtual screening results. J Chem Inf Model 2005,45, (6), 1908-19. From the ranked database of 10,000 compounds our procedure managed to select the single antiestrogen in 14th place. Typically, on completion of a virtual screen, a certain percentage of the top ranking compounds is biologically evaluated. In this test case, and in most screens in our laboratory, the top 0.5% is ordered for further biological testing - translating to the top 50 compounds selected in this case. Therefore, the single antiestrogen would have been retrieved from the set successfully, with significant enrichment and savings over random or traditional HTS.
The procedure detailed above at this stage is sufficient for discrimination between actives and inactives in a training set. It is clear that if a molecule from a compound collection docks in the correct manner with appropriate interatomic distances from the required residues to pass the LPC filter it may not be immediately active, because the necessary H-bonding substituents would not be present. Generally, a VS platform with pharmacophoric preferences incorporated (eg. Flexx-Pharm, Gemdock) will retrieve molecules that fulfill a number or features and as a result novel chemotypes are retrieved, but many potential ones are also missed because of the specificity of such protocols.
Hindle, S. A.; Rarey, M.; Buning, C.; Lengaue, T., Flexible docking under pharmacophore type constraints. J Comput Aided Mol Des 2002,16, (2), 129-49; and Yang, J. M.; Chen, C. C., GEMDOCK: a generic evolutionary method for molecular docking. Proteins 2004,55, (2), 288-304. Advantageously, at this stage of the TS-VS procedure, all scaffolds that adopt the correct shape within the active site are retained. While these ligands may not currently possess the appropriate H-bonding substituents to be immediate binders, they could be tailored to do so using classical chemical modification. Thus TS-VS is serving to suggest novel scaffolds for follow up studies. Final scoring of the selected scaffolds with
TCX-003.88
ChemGauss2 allows prioritization according to those compounds with the best interactions. To highlight this process, we again carried out a virtual screen of the SPECS database (Release: Aug2005, 202054 compounds in total) employing this method. A selection of some the known scaffolds obtained are illustrated in Figure 4a.
Scaffolds B and F represent known scaffolds present in the raloxifene moiety and ZK-119010 moiety respectively. Scaffold A represents a triarylimidazole-type scaffold. Stauffer et al 46 have detailed the differences in binding affinity when the cores (diazoles, imidazoles, pyrazoles) are replaced by one another and with the rings containing the same substituents in the same positions. Although the imidazole core still permitted binding to the ER it was less efficacious than the pyrazole core. Fink et al have also previously demonstrated the high affinity binding of 1,3,5-triaryl-alkyl-pyrazoles to ERa. Fink, Β. E.; Mortensen, D. S.; Stauffer, S. R.; Aron, Z. D.; Katzenellenbogen, J. A., Novel structural templates for estrogen-receptor ligands and prospects for combinatorial synthesis of estrogens. Chem Biol 1999, 6, (4), 205-19. Scaffolds C, D and E all contain a quinoline core that has been previously shown to be effective when incorporated in an antiestrogenic moiety as illustrated in Figure 4b.
Next, specific H-bonding interactions were incorporated in this section to allow immediate identification of potential ER binders from the same screen of the SPECS database carried out to suggest novel scaffolds. The set were filtered using LPC with Hbond constraints set (Arg394, Glu353, Thr347, Leu387). Following visual inspection (compounds selected based on number of interactions with key residues) of 13 ranked compounds remaining, and as a proof of concept, a set of 7 compounds from the original database of greater than 200,000 (Figure 5) were selected and purchased, and evaluated for their ability to bind to the ER by fluorescence binding assay using 10 μΜ as an activity cutoff. Of these 7 tested ligands, 3 demonstrated ERa binding affinities above the 10 μΜ cutoff - with measured binding affinities of 1.1 μΜ (compound 2), 53 nM (compound 4) and 56 nM (compound 5) for human ERa. Figures 6-12 illustrate predicted binding modes of the 7 compounds in ERa.
From Figures 9 and 10 it is clear that compounds 4 and 5 adopt an orientation very close to that of 4-hydroxytamoxifen. The presence of free hydroxy substituents allows interaction with Glu353 and Arg394 and also His524 as observed from the LPC output of the docked complex of compound 4. These compounds were shown to possess the best
TCX-003.88
binding affinity, validating and corroborating the computational analysis. Compound 4 possesses fluorine on the ortho position which appears to reduce the binding affinity because of steric interactions with Asp351, compared with compound 5 which possesses a methyl group on the para position of the side-chain ring. The affinity of compound 2 for ERa is probably due to the presence of a meta methoxy group and para hydroxy group near Glu353 and Arg394 which permits a strong H-bond interaction to occur.
To determine whether our TS-VS methodology was indeed specific enough to select hits that would preferentially bind ERa over the ERP isoform, we examined binding of these compounds to ERP by the same experimental method. Compounds 2,4 and 5 exhibited binding IC50 values of 6.2 μΜ, 780 nM and 915 nM respectively, demonstrating 4.4, 13.7 and 17-fold selectivity for ERa over ERp.
Finally, to evaluate the ability of these compounds to inhibit proliferation of human MCF-7 breast cancer cells, an MTT functional assay was also carried out. The compounds exhibited 15μΜ (compound 2), 11.4 μΜ (compound 4) and 7 μΜ (compound 5) inhibitory activity comparing well with the clinical standard, Tamoxifen (4.6 μΜ). All compounds assayed for their antiproliferative effects were concurrently tested to assess the extent of their cytotoxity using the LDH assay outlined in the experimental section. All three compounds possessed cytotoxic effects comparable to Tamoxifen indicating their actions to be also cytostatic rather than cytotoxic.
As discussed elsewhere, the key to turning an estrogenic substance into an antiestrogen is by inclusion of a basic side-chain such as that of a dimethylaminoethyl chain of tamoxifen. Knox, A. J. S., Meegan M.J., Lloyd D.G, Estrogen Receptors: Molecular interactions, virtual screening and future prospects. Current Topics in Medicinal Chemistry 2006,6, (3), 211-237. It is interesting to note that none of the compounds possessed the predicted ability to interact directly (i.e., via H-bonding) with what is usually considered to be the key antiestrogenic residue, Asp351, but yet they all exhibited inhibitory activity close to that of Tamoxifen.
Compounds 4 and 5 have been previously synthesized by Elagamey et al, and compound 2 synthesized by Chemobrovin et al, however no indication of their pharmacological activity has been reported with respect to the ER to date. Elagamey, A. G. A.; El-Taweel, F. M. A. A., Nitriles in heterocyclic synthesis: Synthesis of condensed pyrans. Indian J. Chem. Sect. B 1990,29, (9), 885-886; and Chemobrovin, Ν. I. K., Yu. V.;
TCX-003.88
ΙΕο /0 4
Bobrovskaya, Ο. V.; Syropyatov, B. Ya., Synthesis and biological activity of 1-acetyl-2,3diaryl-l,2,3,4-tetrahydroquinazoline-4-ones. Khim. Farm. Zh. 1991,25, (5), 37-39, Gungor et al have independently described the synthesis and biological activity of a different series of arylquinazolinone and 3-arylquinazolinethione derivatives that possess low nM ER binding, and preferential binding affinity to ERp. Gungor, T.; Chen, Y.; Golla, R.; Ma, Z.; Corte, J. R.; Northrop, J. P.; Bin, B.; Dickson, J. K.; Stouch, T.; Zhou, R.; Johnson, S. E.; Seethala, R.; Feyen, J. H., Synthesis and characterization of 3-arylquinazolinone and 3arylquinazolinethione derivatives as selective estrogen receptor beta modulators. J Med Chem 2006, 49, (8), 2440-55. The 4H-chromene-3-carboxylate scaffold of compounds 4 and 5 has been incorporated in ethyl 2-amino-6-bromo-4-(l-cyano-2-ethoxy-2-oxoethyl)4H-chromene-3-carboxylate and found to induce apoptosis in tumor cells by binding to bcl2 protein. Manero, F.; Gautier, F.; Gallenne, T.; Cauquil, N.; Gree, D.; Cartron, P. F.; Geneste, O,; Gree, R.; Vallette, F. M.; Juin, P., The small organic compound HA14-1 prevents Bcl-2 interaction with Bax to sensitize malignant glioma cells to induction of cell death. Cancer Res 2006,66, (5), 2757-64. Herein is the first report of associated ER activity for these compounds and additional virtual and chemical library enumerations incorporating focused structural modifications to the core scaffolds are also reported herein (e.g., compounds of formulae I, II, III, and IV).
As indicated previously, a common occurrence and pitfall in many VS campaigns is the retrieval of false positives among true positives. It has been shown that incorporation of two components from our TS-VS protocol, namely normalized complementarity (NC) scoring and distance constraints, can significantly reduce false positives and subsequent ranking by a universal scoring function is far more effective after their inclusion. The choice of docking algorithm to use in VS is highly dependent on the target of interest and suggests that assessing the binding modes of a set of known actives using distance constraints may be more effective than calculation of RMSD as RMSD only accounts for ligand co-ordinates and negates any information about potential interactions. Constraining the distances in different ways, i.e., Nearest-atom or H-bonding, has generated both new scaffolds and also retrieved actual validated hits of both ERa and ER[3. The full TS-VS procedure positively identified 1 micromolar (compound 2 = 1.4uM) and two novel nanomolar (compound 4 = 56nM, compound 5 = 53nM) ligands of ERa by virtual screening of 202054 compounds, of which only 7 were selected for biological testing. The compounds also exhibit low micromolar inhibition of MCF-7 proliferation and were also
TCX-003.88 l£o shown to be selective in targeting ERa over ER[f (e.g., compound 5 = 17-fold selective). The procedure is fully automated and access to a mid-sized 130 Intel Xeon 3.06GHz processor cluster 68 allows one to carry out VS via these methods in a short time. This procedure may be extended to carry out virtual screening to identify compounds selective for ERp.
As mentioned above, in one embodiment of the invention the TS-VS approach has been optimized to converge on novel chemotypes of Estrogen Receptor alpha (ERa). Therefore, in certain aspects of the invention are directed to estrogen receptor modulators (encompassing antagonists and/or agonists), pharmaceutical compositions comprising such modulators and their use in methods for treating estrogen related conditions and conditions wherein the induction of apoptosis is desirable. Such conditions are discussed in detail below, and generally include (but are not limited to) obesity, hormone dependent breast cancer, osteoporosis, estrogen deficiency, arthritis, cardiovascular disease, ovarian cancer, artherosclerosis, colon tumor, endometriosis, Alzheimers disease, non-insulin dependent (type II) diabetes, infertility, prostrate tumor, melanoma, acne, hypercholesterolemia, CNS disease, contraception, conditions related to hair follicles, macular degeneration, urinary incontinence, estrogen receptor-expressing and non-estrogen receptor-expressing tumors, leukaemia.
Throughout the specification the term estrogen agonist refers to a compound that binds to an estrogen receptor (ER) and mimics the action of estrogen in one or more tissues. An antagonist binds to ER and blocks the action of estrogen in one or more tissues.
COMPOUNDS AND COMPOSITIONS
The compounds of the invention discussed below are grouped under four generic types (formulae I, II, III, and IV) for ease of description. The invention includes stereoisomers, geometric isomers, prodrugs and pharmaceutically acceptable salts of the compounds.
The compounds of this invention may be made by one skilled in organic synthesis by known techniques, as well as by the synthetic routes disclosed hereafter. For example, a general reaction scheme for the formation of representative compounds of the invention is shown in Figure 14. See also, for example, Fujimoto, A. “A New Selective Preparation of 4H-Chromenes by Reaction of alkyl cyanoacetate with 3,5-Dibromosalicylaldehyde in the Presence of Ammonium Acetate.” Synthesis 871-872, 1977; Roudier, J., & Foucaud, A. “A
TCX-003.88
Convenient Systhesis of 4H-Chromosomes.” Synthesis 159-160,1984; Shestopalov A.M. et al. “One-step synthesis of substituted 2-amino-4H-chromenes and 2-amino-4Hbenzo[f]chromenes. Molecular and crystal structure of 2-amino-3-cyano-6-hydroxy-4phenyl-4H-benzo[f]chromene,” Russian Chemical Bulletin, 51(12) December 2002, pp. 2238-2243(6); Athanasellis, G. et al. “A simple synthesis of functionalized 2-amino-3cyano-4-chromones by application of the N-hydroxybenzotriazole methodology”
ARKIVOC 2006 (x) 28-34; Sharanin, Yu.A. and Klokol, G.V., Synthesis of 2-Amino-4Hchromenes, J. Org. Chem. USSR 19:1582-1583 (1983); U.S Patent 6,906,203 (Drewe); and U.S. Patent 6,660,871 (Huang); all of which are hereby incorporated by reference.
Pharmaceutical chemists will recognize that physiologically active compounds containing one or more accessible hydroxyl moieties are frequently administered in the form of pharmaceutically acceptable esters or as prodrugs, which is well documented in prior literature. Balant, L.P., Doelker, E. Metabolic considerations in prodrug design. In: Wolff, M.E. (Ed.) Burger’s Medicinal Chemistry and Drug Discovery. 1994, 5th edition, Vol. 1: Principles & Practice. Wiley: New York - pp. 949-977 and references cited therein. Prodrugs are covalently bound carriers that release a parent compound in vivo (believed to be mediated through metabolism), and would include for example compounds of the invention wherein accessible hydroxy groups were bonded to any group that, when administered to a patient, cleaves or is metabolised to form the hydroxy group. It is known in the pharmaceutical field to adjust the rate or duration of action of a compound by appropriate choices of such covalently bound groups. To this end, prodrugs are also included within the context of the invention.
One aspect of the invention relates to a compound of formula I:
wherein, independently for each occurrence, m is 0,1, 2, 3, or 4;
6
TCX-003.88 too n is 0, 1, 2, or 3;
R is alkyl, polycyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl;
R1 is -H, -R, or -C(=O)R; or the two R1 taken together with the nitrogen to which they are bound represent -N-C(R)OR;
R2 is -C=N, -C(=O)OH, -C(=O)OR, -CH2OH, or -CH2OR;
R3 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, -CH2F, -CHF2, -CF3, -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NHz, -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2, -N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, -I, or (CH2)b-A;
A is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, -CH2F, -CHF2, -CF3, -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2, -N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, or -I; b is 0,1,2, 3,4, or 5;
R4 is -OCH2ON(R7)2, -OCH2CH2N(R7)2, -CH2CH2ON(R7)2, -CH2CH2CH2N(R7)2, -N(R)CH2ON(R7)2, -N(H)CH2ON(R7)2, -N(H)CH2CH2N(R7)2, or -N(R)CH2CH2N(R7)2;
R5is -OR1, or-N(R*)2;
R6is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, -CH2F, -CHF2, -CF3, -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2j -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2, -N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, -I, or (CH2)b-A; and
TCX-003.88
* τ
R is -H, -R, or -C(=O)R; or the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical.
In certain embodiments, the invention relates to the aforementioned compound, wherein m is 1.
In certain embodiments, the invention relates to the aforementioned compound, wherein m is 0.
In certain embodiments, the invention relates to the aforementioned compound, wherein n is 1.
In certain embodiments, the invention relates to the aforementioned compound, wherein n is 0.
In certain embodiments, the invention relates to the aforementioned compound, wherein R is alkyl or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R is -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH3, -CH2CH(CH3)CH3, or -CH2C6H5.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -H, alkyl, or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -H, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH2CH3, -CH2CH(CH3)CH2CH3, or-CH2C6H5.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -C(=O)R,
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -C(=O)R; and R is alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R1 taken together with the nitrogen to which they are bound represent -N=C(R)OR.
TCX-003.88
ΙΕο 70 4
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; and R is alkyl or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R2 is -C=N, -C(=O)OH, -C(=O)OR, -CH2OH, or -CH2OR; and R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R2 is -C^N, -C(=O)OH, -C(=O)OCH3, -C(=O)OCH2CH3, or -CH2OH.
In certain embodiments, the invention relates to the aforementioned compound, wherein R3 is -H, -R, -OR, -Cl, -F, -Br, -NO2, -NH2, -NHR, or -NR2; and R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R3 is -H, -OCH3, -Cl, -F, -Br, -NO2, -NH2, or -CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 1.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -OCH2ON(R7)2, -OCH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -OCH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -OCH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -CH2CH2ON(R7)2, or -CH2CH2CH2N(R7)2,
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -CH2CH2CH2N(R7)2,
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -N(R)CH2ON(R7)2, -N(H)CH2ON(R7)2, -N(R)CH2CH2N(R7)2, or -N(H)CH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -N(R)CH2CH2N(R7)2, or -N(H)CH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R5 is -OR1.
TCX-003.88 '£oo<66
In certain embodiments, the invention relates to the aforementioned compound, wherein R5 is -OR1; and R1 is -H, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH2CH3, -CH2CH(CH3)CH2CH3, -CH2C6H5, or -C(=O)CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R5 is -OH, -OCH3, -OCH2CH3, or -C(=O)CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R5 is -N(R*)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R5 is -N(R*)2; and R1 is -H, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH3, -CH2CH(CH3)CH3, -CH2C6H5, or -C(=O)CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R5 is -NH2, -NH(CH3), -N(CH3)2, -NH(CH2C6H5), -N(CH3)(CH2C6H5), -N(H)C(=O)CH3, or -N(CH3)C(=O)CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R6 is -H, -R, -OR, -Cl, -F, -Br, -NO2, -NH2, -NHR, or -NR2; and R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R6 is -H, -OCH3, -Cl, -F, -Br, -NO2, -NH2, or -CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R7 is -H, -R, or -C(=O)R; and R is alkyl or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R7 is -H, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH3, -CH2CH(CH3)CH3, -CH2C6H5, or-C(=O)CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical.
In certain embodiments, the invention relates to the aforementioned compound, •7 wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; and the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine,
TCX-003.88
pyrrolidine, piperidine, piperazine, morpholine, or diazadecaline (1,2,3,4,5,6,7,8octahydroquinoxaline).
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, piperidine, piperazine, morpholine, or diazadecaline (1,2,3,4,5,6,7,8-octahydroquinoxaline); and said optionally substituted monocyclic or bicyclic heterocycle radical is substituted with one or more substituents selected from the group consisting of -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, -ch2f, -chf2, CF3, -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2, -N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, -1, and -(CH2)b-A.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; and the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, or piperidine.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, piperidine, or piperazine; and said optionally substituted monocyclic or bicyclic heterocycle radical is substituted with one or more substituents selected from the group consisting of -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, CH2F, -CHF2, -CF3, -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2, -N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R,
-S(=O)2R, -SO3H, -Cl, -Br, -F, -I, and -(CH2)„-A.
TCX-003.88
One aspect of the invention relates to a compound of formula II:
wherein, independently for each occurrence, m is 0, 1, 2, 3, or 4;
n is 0,1, 2, or 3;
R is alkyl, polycyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl;
R1 is -H, -R, or -C(=O)R; or the two R1 taken together with the nitrogen to which they are bound represent -N=C(R)OR;
R2 is -C=N, -C(=O)OH, -C(=O)OR, -CH2OH, or -CH2OR;
R3 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, -CH2F, -CHF2, -CF3, -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2,
-N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2,
-N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, -I, or (CH2)b-A;
A is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, -CH2F, -CHF2, -CF3, -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR,
-OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R,
-N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2, -N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, or -I;
b is 0,1, 2, 3,4, or 5;
TCX-003.88
R4 is -OCH2ON(R7)2, -OCH2CH2N(R7)2, -CH2CH2ON(R7)2, -CH2CH2CH2N(R7)2,
-N(R)CH2ON(R7)2, -N(H)CH2ON(R7)2, -N(H)CH2CH2N(R7)2, or
-N(R)CH2CH2N(R7)2;
Rsis -OR1, or-NCR1^;
R6 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2,
-C(=O)R, -CH2F, -CHF2, -CF3, -ChN, -OH, -OR, -OC(=O)R, -OC(=O)OR,
-OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R,
-N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2,
-N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2,
-N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, -1, or (CH2)b-A; and
R7 is -H, -R, or -C(=O)R; or the two R7 taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical.
in certain embodiments, the invention relates to the aforementioned compound, wherein m is 1.
In certain embodiments, the invention relates to the aforementioned compound, wherein m is 0.
In certain embodiments, the invention relates to the aforementioned compound, wherein n is 1.
In certain embodiments, the invention relates to the aforementioned compound, wherein n is 0.
In certain embodiments, the invention relates to the aforementioned compound, wherein R is alkyl or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R is -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH3, -CH2CH(CH3)CH3, or -CH2C6H5.
TCX-003.88 lEo
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -H, alkyl, or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -H, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH3, -CH2CH(CH3)CH3, or -CH2C6H5.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -C(=O)R.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -C(=O)R; and R is alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R* taken together with the nitrogen to which they are bound represent -N=C(R)OR.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; and R is alkyl or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R2 is -C^N, -C(=O)OH, -C(=O)OR, -CH2OH, or -CH2OR; and R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R2 is -C=N, -C(=O)OH, -C(=O)OCH3, -C(=O)OCH2CH3, or -CH2OH.
In certain embodiments, the invention relates to the aforementioned compound, wherein R3 is -H, -R, -OR, -Cl, -F, -Br, -NO2, -NH2, -NHR, or -NR2; and R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R3 is -H, -OCH3, -Cl, -F, -Br, -NO2, -NH2, or -CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 1.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -OCH2ON(R7)2, -OCH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -OCH2CH2N(R7)2.
TCX-003.88
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -OCH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -CH2CH2ON(R7)2, or -CH2CH2CH2N(R7)2,
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -CH2CH2CH2N(R7)2,
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -N(R)CH2ON(R7)2, -N(H)CH2ON(R7)2, -N(R)CH2CH2N(R7)2, or -N(H)CH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -N(R)CH2CH2N(R7)2, or -N(H)CH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R5 is -OR1.
In certain embodiments, the invention relates to the aforementioned compound, wherein R5 is -OR1; and R1 is -H, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH3, -CH2CH(CH3)CH3, -CH2C6H5, or -C(=O)CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R5 is -OH, -OCH3, -OCH2CH3, or -C(=O)CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R5 is -NCR1^.
In certain embodiments, the invention relates to the aforementioned compound, wherein R5 is -NCR1^; and R1 is -H, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH3, -CH2CH(CH3)CH3, -CH2C6H5, or -C(=O)CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R5 is -NH2, -NH(CH3), -N(CH3)2, -NH(CH2C6H5), -N(CH3)(CH2C6H5), -N(H)C(=O)CH3, or -N(CH3)C(=O)CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R6 is -H, -R, -OR, -Cl, -F, -Br, -NO2, -NH2, -NHR, or -NR2; and R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R6 is -H, -OCH3, -Cl, -F, -Br, -NO2, -NH2, or -CH3.
TCX-003.88
IE 0 7 /) /
In certain embodiments, the invention relates to the aforementioned compound, wherein R7 is -H, -R, or -C(=O)R; and R is alkyl or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R7 is -H, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH3, -CH2CH(CH3)CH3, -CH2C6H5, or -C(=O)CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R7 taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; and the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, piperidine, piperazine, morpholine, or diazadecaline (1,2,3,4,5,6,7,8octahydroquinoxaline).
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, piperidine, piperazine, morpholine, or diazadecaline (1,2,3,4,5,6,7,8-octahydroquinoxaline); and said optionally substituted monocyclic or bicyclic heterocycle radical is substituted with one or more substituents selected from the group consisting of -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, -ch2f, -chf2, CF3, -ON, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2j -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2, -N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, -I, and -(CH2)„-A.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R7 taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; and the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, or piperidine.
TCX-003.88
In certain embodiments, the invention relates to the aforementioned compound, •γ wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, piperidine, or piperazine; and said optionally substituted monocyclic or bicyclic heterocycle radical is substituted with one or more substituents selected from the group consisting of -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, CH2F, -CHF2, -CF3, -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2, -N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, -I, and -(CH2)b-A.
One aspect of the invention relates to a compound of formula III:
wherein, independently for each occurrence, m is 0, 1,2,3, or 4;
n is 0,1, 2, or 3;
R is alkyl, polycyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl;
R1 is -H, -R, or -C(=O)R; or the two R1 taken together with the nitrogen to which they are bound represent -N=C(R)OR;
R2 is -C=N, -C(=O)OH, -C(=O)OR, -CH2OH, or -CH2OR;
R3 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, -CH2F, -CHF2, -CF3, -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2,
TCX-003.88
-N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, -I, or (CH2)b-A;
A is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, -CH2F, -CHF2, -CF3, -C^N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2, -N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, or -I; b is 0, 1,2, 3,4, or 5;
R6 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, -CH2F, -CHF2, -CF3, -C^N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2, -N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, -I, or (CH2)b-A;
R8 is -(CH2)PSR10, -(CH2)pS(=O)R10, -(CH2)PS(=O)2R10, -(CH2)pC(=O)N(R‘)R10, -(CH/pNCR'jCH^HjCHjSR10, -(CH2)PN(R1)CH2CH2CH2S(=O)R10, or -(CfFjpNiR'jCILCHzCHzS^OhR10;
p is 5, 6, 7, 8, 9, or 10;
R9 is -H, -OH, or -OR; and
R10 is alkyl, fluoroalkyl or perfluoroalkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein m is 1.
In certain embodiments, the invention relates to the aforementioned compound, wherein m is 0.
In certain embodiments, the invention relates to the aforementioned compound, wherein n is 1.
In certain embodiments, the invention relates to the aforementioned compound, wherein n is 0.
/10
TCX-003.88
ΙΕο
In certain embodiments, the invention relates to the aforementioned compound, wherein R is alkyl or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R is -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH3, -CH2CH(CH3)CH3, or -CH2C6H5.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -H, alkyl, or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -H, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH3, -CH2CH(CH3)CH3, or -CH2C6H5.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -C(=O)R.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -C(=O)R; and R is alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R1 taken together with the nitrogen to which they are bound represent -N=C(R)OR.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; and R is alkyl or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R2 is -C=N, -C(=O)OH, -C(=O)OR, -CH2OH, or -CH2OR; and R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R2 is -C=N, -C(=O)OH, -C(=O)OCH3, -C(=O)OCH2CH3, or -CH2OH.
In certain embodiments, the invention relates to the aforementioned compound, wherein R3 is -H, -R, -OR, -Cl, -F, -Br, -NO2, -NH2, -NHR, or -NR2; and R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R3 is -H, -OCH3, -Cl, -F, -Br, -NO2, -NH2, or -CH3.
4Π
TCX-003.88
ΙΕο 70 4 65
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -OCH2ON(R7)2, -OCH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -OCH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -OCH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -CH2CH2ON(R7)2, or -CH2CH2CH2N(R7)2,
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -CH2CH2CH2N(R7)2,
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -N(R)CH2ON(R7)2, -N(H)CH2ON(R7)2, -N(R)CH2CH2N(R7)2, or -N(H)CH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -N(R)CH2CH2N(R7)2, or -N(H)CH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R6 is -H, -R, -OR, -Cl, -F, -Br, -NO2, -NH2, -NHR, or -NR2; and R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R6 is -H, -OCH3, -Cl, -F, -Br, -NO2, -NH2, or -CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)PSR10, -(CH2)PS(=O)R10, or -(CH2)PS(=O)2R10.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)PSRI(), -(CH2)PS(=O)R10, or -(CH2)PS(=O)2R10; and R10 is fluoroalkyl.
in certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)PSCH2CH2CH2CF2CF3, -(CH2)PS(=O)CH2CH2CH2CF2CF3, -(CH2)PS(=O)2CH2CH2CH2CF2CF3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)pC(=O)N(R')R1().
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CHzjpC^OjNiR’jR10; and R10 is alkyl.
cn
TCX-003.88 toO 70 4
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)pC(=O)N(CH3)CH2CH2CH2CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)pN(R')CH2CH2CH2SR1(), -(CH2)pN(R')CH2CH2CH2S(=O)R1(), or -(CH2)pN(R1)CH2CH2CH2S(=O)2R10.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)pN(R')CH2CH2CH2SR10, -(CH2)pN(Rl)CH2CH2CH2S(=O)R10, or -(CH2)pN(R')CH2CH2CH2S(=O)2R10; and R10is alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)pN(CH3)CH2CH2CH2SCH2CH2CH2CH2CH3, -(CH2)pN(CH3)CH2CH2CH2S(=O)CH2CH2CH2CH2CH3, or -(CH2)PN(CH3)CH2CH2CH2S(-O)2CH2CH2CH2CH2CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 5.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 6.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 7.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 8.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 9.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 10.
In certain embodiments, the invention relates to the aforementioned compound, wherein R9 is -H.
In certain embodiments, the invention relates to the aforementioned compound, wherein R9 is -OH.
In certain embodiments, the invention relates to the aforementioned compound, wherein R9 is -OR; R is alkyl.
TCX-003.88 «070466
In certain embodiments, the invention relates to the aforementioned compound, wherein R9 is -OCH3, -OCH2CH3, -OCH2CH2CH3, -OCH(CH3)CH3, -OCH2CH2CH2CH3, -OCH(CH3)CH2CH3, -OCH2CH(CH3)CH3, -OCH2C6H5, or -OC(=O)CII3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R10 is alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R10 is fluoroalkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R10 is perfluoroalkyl.
One aspect of the invention relates to a compound of formula IV:
wherein, independently for each occurrence, m is 0,1,2, 3, or 4;
n is 0, 1,2, or 3;
p is 5, 6, 7, 8, 9, or 10
R is alkyl, polycyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl;
R1 is -H, -R, or -C(=O)R; or the two R1 taken together with the nitrogen to which they are bound represent -N=C(R)OR;
R2 is -C=N, -C(=O)OH, -C(=O)OR, -CH2OH, or -CH2OR;
R3 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, -CH2F, -CHF2, -CF3, -C^N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2j -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2,
TCX-003.88 «0 70 4
-N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, -I, or (CH2)b-A;
A is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2, -C(=O)NHR, -C(=O)NR2, -C(=O)R, -CH2F, -CHF2, -CF3, -C^N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2, -N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, or -I; b is 0,1,2, 3,4, or 5;
R6is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH2j -C(=O)NHR, -C(=O)NR2, -C(=O)R, -CH2F, -CHF2, -CF3, -C^N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH2, -OC(=O)NHR, -OC(=O)NR2, -NH2, -NHR, -NR2, -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH2, -N(R)C(=O)NH2, -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR2, -N(R)C(=O)NR2, -NO2, -SH, -SR, -S(=O)R, -S(=O)2R, -SO3H, -Cl, -Br, -F, -I, or (CH2)b-A;
R8 is -(CH2)PSR10, -(CH2)pS(=O)R10, -(CH2)pS(=O)2R10, -(ClfyjpCfyOjNCR'jR10, -(CH2)pN(R1)CH2CH2CH2SR10, -(CH2)pN(R1)CH2CH2CH2S(=O)R10, or -(CH2)pN(Rl)CH2CH2CH2S(=O)2R!0;
R9 is -H, -OH, or -OR; and
R10 is alkyl, fluoroalkyl or perfluoroalkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein m is 1.
In certain embodiments, the invention relates to the aforementioned compound, wherein m is 0.
In certain embodiments, the invention relates to the aforementioned compound, wherein n is 1.
In certain embodiments, the invention relates to the aforementioned compound, wherein n is 0.
rTCX-003.88
IE 0 7 0 4 g g
In certain embodiments, the invention relates to the aforementioned compound, wherein R is alkyl or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R is -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH3, -CH2CH(CH3)CH3, or -CH2C6H5.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -H, alkyl, or aralkyl.
in certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -H, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)CH3, -CH2CH2CH2CH3, -CH(CH3)CH2CH3, -CH2CH(CH3)CH3, or -CH2C6H5.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -C(=O)R.
In certain embodiments, the invention relates to the aforementioned compound, wherein R1 is -C^OjR; and R is alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R1 taken together with the nitrogen to which they are bound represent -N=C(R)OR.
In certain embodiments, the invention relates to the aforementioned compound, wherein the two R1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; and R is alkyl or aralkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R2 is -C=N, -C(=O)OH, -C(=O)OR, -CH2OH, or -CH2OR; and R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R2 is -C=N, -C(=O)OH, -C(=O)OCH3, -C(O)0CH2CH3, or -CH2OH.
In certain embodiments, the invention relates to the aforementioned compound, wherein R3 is -H, -R, -OR, -Cl, -F, -Br, -NO2, -NH2, -NHR, or -NR2; and R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R3 is -H, -OCH3, -Cl, -F, -Br, -NO2, -NH2, or -CH3.
TCX-003.88 /£0 70 4 6β
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -OCH2ON(R7)2, -OCH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -OCH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -OCH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -CH2CH2ON(R7)2, or -CH2CH2CH2N(R7)2,
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -CH2CH2CH2N(R7)2,
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -N(R)CH2ON(R7)2, -N(H)CH2ON(R7)2, -N(R)CH2CH2N(R7)2, or -N(H)CH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R4 is -N(R)CH2CH2N(R7)2, or -N(H)CH2CH2N(R7)2.
In certain embodiments, the invention relates to the aforementioned compound, wherein R6 is -H, -R, -OR, -Cl, -F, -Br, -NO2, -NH2, -NHR, or -NR2; and R is lower alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R6 is -H, -OCH3, -Cl, -F, -Br, -NO2, -NH2, or -CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)PSR10, -(CH2)PS(=O)R10, or -(CH2)PS(=O)2R10.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)PSR10, -(CH2)PS(=O)R10, or -(CH2)PS(=O)2R10; and R10is fluoroalkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)PSCH2CH2CH2CF2CF3, -(CH2)PS(=O)CH2CH2CH2CF2CF3, -(CH2)PS(=O)2CH2CH2CH2CF2CF3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)PC(=O)N(R1)R10.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)pC(=O)N(R’)R10; and R10 is alkyl.
TCX-003.88
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)pC(=O)N(CH3)CH2CH2CH2CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CHUpN^CHzCI^CIfySR10, -(CHOpN^CHzCHzCHzS^OjR10, or -(CIOpNCR'iCIhCifyC^S^OhR10.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)PN(R1)CH2CH2CH2SR10, -(CH2)pN(R')CH2CH2CH2S(-O)R10, or “(CIi2)pN(Rl)CH2CH2CH2S(=O)2R10; and R10 is alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R8 is -(CH2)pN(CH3)CH2CH2CH2SCH2CH2CH2CH2CH3, -(CII2)PN(CH3)CH2CH2CH2S(-O)CH2CH2CH2CH2CH3, or -(CH2)pN(CH3)CH2CH2CH2S(=O)2CH2CH2CH2CH2CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 5.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 6.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 7.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 8.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 9.
In certain embodiments, the invention relates to the aforementioned compound, wherein p is 10.
In certain embodiments, the invention relates to the aforementioned compound, wherein R9 is -H.
In certain embodiments, the invention relates to the aforementioned compound, wherein R9 is -OH.
In certain embodiments, the invention relates to the aforementioned compound, wherein R9 is -OR; R is alkyl.
TCX-003.88 *0 70 4 6 6
In certain embodiments, the invention relates to the aforementioned compound, wherein R9 is -OCH3, -OCH2CH3, -OCH2CH2CH3, -OCH(CH3)CH3, -OCH2CH2CH2CH3, -OCH(CH3)CH2CH3, -OCH2CH(CH3)CH3, -OCH2C6H5, or -OC(=O)CH3.
In certain embodiments, the invention relates to the aforementioned compound, wherein R10 is alkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R10 is fluoroalkyl.
In certain embodiments, the invention relates to the aforementioned compound, wherein R10 is perfluoroalkyl.
In certain embodiments, the invention relates to a pharmaceutical composition, comprising any one of the aforementioned compounds; and a pharmaceutically acceptable carrier.
In certain embodiments, the invention relates to a pharmaceutical composition, comprising any one of the aforementioned compounds; and a pharmaceutically active compound.
In certain embodiments, the invention relates to the aforementioned pharmaceutical composition, wherein the pharmaceutically active compound is an anti-cancer drug.
In certain embodiments, the invention relates to the aforementioned pharmaceutical composition, wherein the anti-cancer drug is cisplatin.
PHARMACEUTICAL COMPOSITIONS AND THERAPEUTIC ADMINISTRATION
A utility of these compounds in any of the cited disease states or conditions would involve the administration of an effective amount of a compound of this invention, preferably in the form of a pharmaceutical composition to an animal in need thereof.
The compounds of the invention may be administered to animals (including humans) orally or parenterally in the conventional form of preparation such as capsules, microcapsules, tablets, granules, powder, troches, pills, suppositories, injections, suspensions and syrups. Suitable formulations may be prepared by methods commonly employed using conventional organic and inorganic additives such as an excipient, a binder, a disintegrator, a lubricant, a flavouring agent, a preservative, a stabilizer, a suspending
TCX-003.88
IE ® 7 0 4 β | agent, a dispersing agent, a diluent and base wax. The amount of active ingredient in the medical composition may be at a level that will exercise the desired therapeutic effect.
In certain aspects, the present invention provides a pharmaceutical composition comprising an effective amount of a compound of the invention and a pharmaceutically acceptable carrier or vehicle. The pharmaceutical compositions are suitable for veterinary or human administration.
The pharmaceutical compositions of the present invention can be in any form that allows for the composition to be administered to a subject, said subject preferably being an animal, including, but not limited to a human, mammal, or non-human animal, such as a cow, horse, sheep, pig, fowl, cat, dog, mouse, rat, rabbit, guinea pig, etc. In a preferred embodiment, the subject is a mammal, and most preferably a human.
The compositions of the invention can be in the form of a solid, liquid or gas (aerosol). Typical routes of administration may include, without limitation, oral, topical, parenteral, sublingual, rectal, vaginal, ocular, and intranasal. Parenteral administration includes subcutaneous injections, intravenous, intramuscular, intraperitoneal, intrapleural, intrasternal injection or infusion techniques. The compositions may be administered parenterally. The compositions may be administered intravenously. Pharmaceutical compositions of the invention can be formulated so as to allow a compound of the invention to be bioavailable upon administration of the composition to a subject. Compositions can take the form of one or more dosage units, where, for example, a tablet can be a single dosage unit, and a container of a compound of the invention in aerosol form can hold a plurality of dosage units.
Materials used in preparing the pharmaceutical compositions can be non-toxic in the amounts used. It will be evident to those of ordinary skill in the art that the optimal dosage of the active ingredient(s) in the pharmaceutical composition will depend on a variety of factors. Relevant factors include, without limitation, the type of subject (e.g., human), the overall health of the subject, the type of cancer the subject is in need of treatment of, the use of the composition as part of a multi-drug regimen, the particular form of the Compound of the Invention, the manner of administration, and the composition employed.
The pharmaceutically acceptable carrier or vehicle may be particulate, so that the compositions are, for example, in tablet or powder form. The carriers) can be liquid, with the compositions being, for example, an oral syrup or injectable liquid. In addition, the e· π
TCX-003.88 /£θ7Ο carrier(s) can be gaseous, so as to provide an aerosol composition useful in, e.g., inhalatory administration.
The composition may be intended for oral administration, and if so, the composition is preferably in solid or liquid form, where semi-solid, semi-liquid, suspension and gel forms are included within the forms considered herein as either solid or liquid.
As a solid composition for oral administration, the composition can be formulated into a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like form. Such a solid composition typically contains one or more inert diluents. In addition, one or more of the following can be present: binders such as ethyl cellulose, carboxymethylcellulose, microcrystalline cellulose, or gelatin; excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, com starch and the like; lubricants such as magnesium stearate or Sterotex; glidants such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin, a flavoring agent such as peppermint, methyl salicylate or orange flavoring, and a coloring agent.
When the pharmaceutical composition is in the form of a capsule, e.g., a gelatin capsule, it can contain, in addition to materials of the above type, a liquid carrier such as polyethylene glycol, cyclodextrin or a fatty oil.
The pharmaceutical composition can be in the form of a liquid, e.g., an elixir, syrup, solution, emulsion or suspension. The liquid can be useful for oral administration or for delivery by injection. When intended for oral administration, a composition can comprise one or more of a sweetening agent, preservatives, dye/colorant and flavor enhancer. In a composition for administration by injection, one or more of a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and isotonic agent can also be included.
The liquid compositions of the invention, whether they are solutions, suspensions or other like form, can also include one or more of the following: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer’s solution, isotonic sodium chloride, fixed oils such as synthetic mono or digylcerides which can serve as the solvent or suspending medium, polyethylene glycols, glycerin, cyclodextrin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents
TCX-003.88 *070466 for the adjustment of tonicity such as sodium chloride or dextrose. A parenteral composition can be enclosed in ampoule, a disposable syringe or a multiple-dose vial made of glass, plastic or other material. Physiological saline is a preferred adjuvant. An injectable composition is preferably sterile.
The amount of the compound of the invention that is effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques. In addition, in vitro or in vivo assays can optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the compositions will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient’s circumstances. When a compound according to this invention is administered into a human subject, the daily dosage will normally be determined by the prescribing physician with the dosage generally varying according to the age, weight, and response of the individual patient, as well as the severity of the patient's symptoms.
In certain applications, a suitable amount of compound is administered to a mammal undergoing treatment. Oral dosages of the present invention, when used for the indicated effects, will range between about 0.01 mg per kg of body weight per day (mg/kg/day) to about 100 mg/kg/day, preferably 0.01 to 10 mg/kg/day, and most preferably 0.1 to 5.0 mg/kg/day. For oral administration, the compositions are preferably provided in the form of tablets containing 0.01,0.05,0.1, 0.5,1.0,2.5, 5.0,10.0,15.0, 25.0, 50.0,100 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably, from about 1 mg to about 100 mg of active ingredient. Intravenously, the most preferred doses will range from about 0.1 to about 10 mg/kg/minute during a constant rate infusion. Advantageously, compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three or four times daily. Furthermore, preferred compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittant throughout the dosage regimen.
ΖΆ
TCX-003.88
The pharmaceutical compositions comprise an effective amount of a compound of the invention such that a suitable dosage will be obtained. Typically, this amount is at least 0.01% of a compound of the invention by weight of the composition. When intended for oral administration, this amount can be varied to be between 0.1% and 80% by weight of the composition. Preferred oral compositions can comprise from between 4% and 50% of the compound of the invention by weight of the composition. Preferred compositions of the present invention are prepared so that a parenteral dosage unit contains from between 0.01% and 2% by weight of the compound of the invention.
Generally, the dosage of a compound of the invention administered to a subject is typically between 0.1 mg/kg and 100 mg/kg of the subject’s body weight. In one embodiment, the dosage administered to a subject is between 0.5 mg/kg and 50 mg/kg of the subject’s body weight, more preferably between 1 mg/kg and 25 mg/kg of the subject’s body weight.
In a specific embodiment, when the compounds of the invention are used in combination with radiotherapy, a compound of the invention can be administered in amounts that result in concentrations in the fluid of a target tissue that are less than about twice the IC50 concentration for the particular compound, more preferably about equal to the IC50 concentration. The IC50 concentration is defined as the concentration of the compound of the invention that kills 50% of cells following treatment with the compound of the invention.
In another embodiment, the Compounds of the Invention may be administered at amounts lower than the IC50 concentration, such as about 50% of the IC50 concentration, about 40% of the IC50 concentration, about 30% of the IC50 concentration, about 20% of the IC50 concentration, about 10% or about 5% of the IC50 concentration, at the target tissue.
In still another embodiment, the compounds of the invention may be administered locally so that the concentration at the target tissue is in the effective range and the concentration in non-target tissue is minimized.
In another embodiment, the dosage of the compound of the invention results in a concentration at a target tissue that does not promote apoptosis of cells in culture yet is effective in increasing cell death in neoplastic cells exposed to radiation or recognized chemotherapeutic chemical agents. Concentrations that produce these effects can be f- 1
TCX-003.88 *0 70 4 determined for a compound of the invention by one of skill in the art using markers of apoptosis, including, but not limited to, the apoptotic index and caspase activities.
The compounds of the invention can be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.). Administration can be systemic or local. Various delivery systems are known, e.g., microparticles, microcapsules, capsules, etc., and may be useful for administering a compound of the invention. In certain embodiments, more than one compound of the invention is administered to a subject. Methods of administration may include, but are not limited to, oral administration and parenteral administration; parenteral administration including, but not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous; intranasal, epidural, sublingual, intranasal, intracerebral, intraventricular, intrathecal, intravaginal, transdermal, rectally, by inhalation, or topically to the ears, nose, eyes, or skin. The preferred mode of administration is left to the discretion of the practitioner, and will depend in-part upon the site of the medical condition (such as the site of cancer, a cancerous tumor or a precancerous condition).
In one embodiment, the compounds of the invention are administered orally. In another embodiment, the compounds of the invention are administered parenterally. In still another embodiment, the compounds of the invention are administered intravenously.
In specific embodiments, it can be desirable to administer one or more compounds of the invention locally to the area in need of treatment. This can be achieved, for example, and not by way of limitation, by local infusion during surgery; topical application, e.g., in conjunction with a wound dressing after surgery; by injection; by means of a catheter; by means of a suppository; or by means of an implant, the implant being of a porous, nonporous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. In one embodiment, administration can be by direct injection at the site (or former site) of a cancer, tumor, or precancerous tissue. In certain embodiments, it can be desirable to introduce one or more Compounds of the Invention into the central nervous system by any suitable route, including intraventricular and intrathecal injection. Intraventricular injection can be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir.
TCX-003.88
ΙΕο 70 4
Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent, or via perfusion in a fluorocarbon or synthetic pulmonary surfactant. In certain embodiments, the compounds of the invention can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
In one embodiment, the compounds of the invention can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.).
In yet another embodiment, the compounds of the invention can be delivered in a controlled release system. In one embodiment, a pump can be used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)). In another embodiment, polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J. Macromol. Sci. Rev. Macromol. Chem. 23:61 (1983); see also Levy et al., Science 228:190 (1985); During et al., Ann. Neurol. 25:351 (1989); Howard et al., J. Neurosurg. 71:105 (1989)). In yet another embodiment, a controlled-release system can be placed in proximity of the target of the compounds of the invention, e.g., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)). Other controlled-release systems discussed in the review by Langer (Science 249:1527-1533 (1990)) can be used.
The term “carrier” refers to a diluent, adjuvant or excipient, with which a compound of the invention is administered. Such pharmaceutical carriers can be liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. The carriers can be saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea, and the like. In addition, auxiliary, stabilizing, thickening, lubricating and coloring agents can be used. In one embodiment, when administered to a subject, the compounds of the invention and pharmaceutically acceptable carriers are sterile. Water is a preferred carrier when the compound of the
TCX-003.88 «070466 invention is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical carriers also include excipients such as starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The present compositions, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
The present compositions can take the form of solutions, suspensions, emulsion, tablets, pills, pellets, capsules, capsules containing liquids, powders, sustained-release formulations, suppositories, emulsions, aerosols, sprays, suspensions, or any other form suitable for use. In one embodiment, the pharmaceutically acceptable carrier is a capsule (see e.g., U.S. Patent No. 5,698,155; hereby incorporated by reference). Other examples of suitable pharmaceutical carriers are described in “Remington’s Pharmaceutical Sciences” by E.W. Martin, hereby incorporated by reference.
Sustained or directed release compositions that may be formulated include, but are not limited to liposomes or other formulations wherein the active component is protected with differentially degradable coatings, e.g., by microencapsulation, multiple coatings, etc. It is also possible to freeze-dry the new compositions and use the lyophilizates obtained, for example, for the preparation of products for injection.
In one embodiment, the compounds of the invention are formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to animals, particularly human beings. Typically, the carriers or vehicles for intravenous administration are sterile isotonic aqueous buffer solutions. Where necessary, the compositions can also include a solubilizing agent. Compositions for intravenous administration can optionally comprise a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where a compound of the invention is to be administered by infusion, it can be dispensed, for example, with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the compound of the invention is
TCX-003.88 «070466 administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
Compositions for oral delivery can be in the form of tablets, lozenges, aqueous or oily suspensions, granules, powders, emulsions, capsules, syrups, or elixirs, for example. Orally administered compositions can contain one or more optionally agents, for example, sweetening agents such as fructose, aspartame or saccharin; flavoring agents such as peppermint, oil of Wintergreen, or cherry; coloring agents; and preserving agents, to provide a pharmaceutically palatable preparation. Moreover, where in tablet or pill form, the compositions can be coated to delay disintegration and absorption in the gastrointestinal tract thereby providing a sustained action over an extended period of time. Selectively permeable membranes surrounding an osmotically active driving complex are also suitable for orally administered compositions of the invention. In these later platforms, fluid from the environment surrounding the capsule is imbibed by the driving complex, which swells to displace the agent or agent composition through an aperture. These delivery platforms can provide an essentially zero order delivery profde as opposed to the spiked profiles of immediate release formulations. A time-delay material such as glycerol monostearate or glycerol stearate can also be used. Oral compositions can include standard carriers such as mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Such carriers are preferably of pharmaceutical grade.
The pharmaceutical compositions of the invention can be intended for topical administration, in which case the carrier can be in the form of a solution, emulsion, ointment or gel base. The base, for example, can comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, beeswax, mineral oil, diluents such as water and alcohol, and emulsifiers and stabilizers. Thickening agents can be present in a composition for topical administration. If intended for transdermal administration, the composition can be in the form of a transdermal patch or an iontophoresis device. Topical formulations can comprise a concentration of a compound of the invention of from between 0.01% and 10% w/v (weight per unit volume of composition).
The compositions can include various materials that modify the physical form of a solid or liquid dosage unit. For example, the composition can include materials that form a coating shell around the active ingredients. The materials that form the coating shell are
TCX-003.88 IE 0 J Q 4 e typically inert, and can be selected from, for example, sugar, shellac, and other enteric coating agents. Alternatively, the active ingredients can be encased in a gelatin capsule.
The compositions can consist of gaseous dosage units, e.g., it can be in the form of an aerosol. The term aerosol is used to denote a variety of systems ranging from those of colloidal nature to systems consisting of pressurized packages. Delivery can be by a liquefied or compressed gas or by a suitable pump system that dispenses the active ingredients. Aerosols of the compositions can be delivered in single phase, bi-phasic, or triphasic systems in order to deliver the composition. Delivery of the aerosol includes the necessary container, activators, valves, subcontainers, Spacers and the like, which together can form a kit. Preferred aerosols can be determined by one skilled in the art, without undue experimentation.
The pharmaceutical compositions can be prepared using methodology well known in the pharmaceutical art. For example, a composition intended to be administered by injection can be prepared by combining a compound of the invention with water so as to form a solution. A surfactant can be added to facilitate the formation of a homogeneous solution or suspension. Surfactants are complexes that can non-covalently interact with a compound of the invention so as to facilitate dissolution or homogeneous suspension of the compound of the invention in the aqueous delivery system.
Compounds of the present invention may also be delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropyl-methacrylamide-phenol, polyhydroxy-ethylaspartamide-phenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues. Furthermore, the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polyactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and crosslinked or amphipathic block copolymers of hydrogels.
In one embodiment, the pharmaceutical compositions of the present invention may comprise one or more known therapeutically active agents. Examples of specific agents are discussed in more detail below. In one embodiment, the pharmaceutical compositions of
TCX-003.88 the present invention may comprise one or more additional anticancer agents. In another embodiment, the pharmaceutical compositions of the present invention can be administered prior to, at the same time as, or after an known therapeutic agent, or on the same day, or within 1 hour, 2 hours, 12 hours, 24 hours, 48 hours or 72 hours of each other,
METHODS
In certain embodiments, the compounds of the present invention are selective modulators of estrogen receptors and are therefore useful to treat or prevent a variety of diseases and conditions related to estrogen receptor functioning in mammals, preferably humans. Specifically, the compounds of the present invention exhibit a potent, selective affinity for ERa. They also may act as antagonists on breast and uterine tissue and as agonists on bone and lipids.
“A variety of diseases and conditions related to estrogen receptor functioning” includes, but is not limited to, bone loss, bone fractures, osteoporosis, glucocorticoid induced osteoporosis, Paget's disease, abnormally increased bone turnover, periodontal disease, tooth loss, rheumatoid arthritis, osteoarthritis, periprosthetic osteolysis, osteogenesis imperfecta, metastatic bone disease, hypercalcemia of malignancy, and multiple myeloma, cartilage degeneration, endometriosis, uterine fibroid disease, cancer of the breast, uterus or prostate, hot flashes, cardiovascular disease, impairment of cognitive function, cerebral degenerative disorders, restenosis, gynecomastia, vascular smooth muscle cell proliferation, obesity and incontinence. In treating such conditions with the instantly claimed compounds, the required therapeutic amount will vary according to the specific disease and is readily ascertainable by those skilled in the art. Both treatment and prevention are contemplated by the scope of the invention.
In certain embodiments, the compounds of the present invention are useful in treating estrogen related diseases which are not directly modulated through the estrogen receptor. In certain embodiments, the compounds of the invention act by modulating estrogen binding to other enzymes. In certain embodiments, the compounds of the invention effect the action of protein kinase C (PKC), calmodulim (CaM)-dependent enzymes, acyl coenzyme A or cholesterol acyl transferase (CAT).
In certain embodiments, the present invention also relates to methods for eliciting an estrogen receptor modulating effect in a mammal in need thereof by administering the compounds and pharmaceutical compositions of the present invention.
TCX-003.88 ιεο 70 4 66
In certain embodiments, the present invention also relates to methods for eliciting an estrogen receptor agonizing effect in a mammal in need thereof by administering the compounds and pharmaceutical compositions of the present invention. The estrogen receptor agonizing effect can be either an ERa agonizing effect, an ERp agonizing effect or a mixed ERa and ERp agonizing effect.
In certain embodiments, the present invention also relates to methods for treating or preventing disorders related to estrogen functioning, bone loss, bone fractures, osteoporosis, glucocorticoid induced osteoporosis, Paget's disease, abnormally increased bone turnover, periodontal disease, tooth loss, rheumatoid arthritis, osteoarthritis, periprosthetic osteolysis, osteogenesis imperfecta, metastatic bone disease, hypercalcemia of malignancy, and multiple myeloma, cartilage degeneration, endometriosis, uterine fibroid disease, cancer of the breast, uterus or prostate, hot flashes, cardiovascular disease, impairment of cognitive function, cerebral degenerative disorders, restenosis, gynecomastia, vascular smooth muscle cell proliferation, obesity and incontinence in a mammal in need thereof by administering the compounds and pharmaceutical compositions of the present invention. Exemplifying the invention is a method of treating or preventing osteoporosis. Exemplifying the invention is a method of treating or preventing bone loss. Exemplifying the invention is a method of treating or preventing metastatic bone disease. Exemplifying the invention is a method of treating or preventing cancer. Exemplifying the invention is a method of treating or preventing cardiovascular disease.
An embodiment of the invention is a method for treating or preventing cancer, especially of the breast, uterus or prostate, in a mammal in need thereof by administering the compounds and pharmaceutical compositions of the present invention. The utility of SERMs for the treatment of breast, uterine or prostate cancer is known in the literature, see T. J. Powles, Breast cancer prevention, Oncologist 2002; 7(1):60 4; Park, W. C. and Jordan, V. C., “Selective estrogen receptor modulators (SERMS) and their roles in breast cancer prevention.” Trends Mol Med. 2002 February;8(2):82 8; Wolff, A. C. et al., “Use of SERMs for the adjuvant therapy of early-stage breast cancer,” Ann Ν Y Acad Sci. 2001 December;949:80 8; Steiner, M. S. et al., “Selective estrogen receptor modulators for the chemoprevention of prostate cancer,” Urology 2001 April; 57(4 Suppl 1):68 72.
Another embodiment of the invention is a method of treating or preventing metastatic bone disease in a mammal in need thereof by administering to the mammal a
TCX-003.88 £θ 70 4 g g therapeutically effective amount of any of the compounds or pharmaceutical compositions described above. The utility of SERMs in the treatment of metastatic bone disease is known in the literature, see, Campisi, C. et al., “Complete resoultion of breast cancer bone metastasis through the use of beta-interferon and tamoxifen,” Eur J Gynaecol Oncol
1993; 14(6):479 83.
Another embodiment of the invention is a method of treating or preventing gynecomastia in a mammal in need thereof by administering to the mammal a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above. The utility of SERMs in the treatment of gynecomastia is known in the literature, see, Ribeiro, G. and Swindell R., “Adjuvant tamoxifen for male breast cancer.”
Br J Cancer 1992; 65:252 254; Donegan, W., “Cancer of the Male Breast,” JGSM Vol. 3, Issue 4,2000.
Another embodiment of the invention is a method of treating or preventing postmenopausal osteoporosis, glucocorticoid osteoporosis, hypercalcemia of malignancy, bone loss and bone fractures in a mammal in need thereof by administering to the mammal a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above. The utility of SERMs to treat or prevent osteoporosis, hypercalcemia of malignancy, bone loss or bone fractures is known in the literature, see Jordan, V. C. et al., Selective estrogen receptor modulation and reduction in risk of breast cancer, osteoporosis and coronary heart disease, Natl Cancer Inst 2001 October; 93(19):1449 57; Bjarnason, N H et al., “Six and twelve month changes in bone turnover are realted to reduction in vertebral fracture risk during 3 years of raloxifene treatment in postemenopausal osteoporosis,” Osteoporosis Int 2001; 12(11):922 3; Fentiman I. S., “Tamoxifen protects against steroid-induced bone loss,” Eur J Cancer 28:684 685 (1992); Rodan, G. A. et al., “Therapeutic Approaches to Bone Diseases,” Science Vol 289,1 Sep. 2000.
Another embodiment of the invention is a method of treating of preventing periodontal disease or tooth loss in a mammal in need thereof by administering to the mammal a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above. The use of SERMs to treat periodontal disease or tooth loss in a mammal is known in the literature, see Rodan, G. A. et al., “Terapeutic Approaches to Bone Diseases,” Science Vol 289, 1 Sep. 2000 pp. 1508 14.
TCX-003.88
Another embodiment of the invention is a method of treating of preventing Paget's disease in a mammal in need thereof by administering to the mammal a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above. The use of SERMs to treat Paget’s disease in a mammal is known in the literature, see Rodan, G. A. et al., “Therapeutic Approaches to Bone Diseases,” Science Vol 289, 1 Sep. 2000 pp. 1508 14.
Another embodiment of the invention is a method of treating or preventing uterine fibroid disease in a mammal in need thereof by administering to the mammal a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above. The use of SERMs to treat uterine fibroids, or uterine leiomyomas, is known in the literature, see Palomba, S., et al, “Effects of raloxifene treatment on uterine leiomyomas in postmenopausal women,” Fertil Steril. 2001 July;76(l):38 43.
Another embodiment of the invention is a method of treating or preventing obesity in a mammal in need thereof by administering to the mammal a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above. The use of SERMs to treat obesity is known in the literature, see Picard, F. et al., “Effects of the estrogen antagonist EM-652. HCI on energy balance and lipid metabolism in ovariectomized rats,” Int J Obes Relat Metab Disord. 2000 July;24(7):830 40.
Another embodiment of the invention is a method of treating or preventing cartilage degeneration, rheumatoid arthritis or osteoarthritis in a mammal in need thereof by administering to the mammal a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above. The use of SERMs to treat cartilage degeneration, rheumatoid arthritis or osteoarhritis is known in the literature, see Badger, A. M. et al., “Idoxifene, a novel selective estrogen receptor modulator, is effective in a rat model of adjuvant-induced arthritis.” J Pharmacol Exp Ther. 1999 December;291(3):1380 6.
Another embodiment of the invention is a method of treating or preventing endometriosis in a mammal in need thereof by administering to the mammal a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above. The use of SERMs to treat endometriosis is known in the art, see Steven R. Goldstein, “The Effect of SERMs on the Endometrium,” Annals of the New York Academy of Sciences 949:237 242 (2001).
TCX-003.88 «07046g
Another embodiment of the invention is a method of treating or preventing urinary incontinence in a mammal in need thereof by administering to the mammal a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above. The use of SERMs to treat urinary incontinence is known in the art, see, Goldstein, S. R., “Raloxifene effect on frequency of surgery for pelvic floor relaxation,” Obstet Gynecol. 2001 July;98(l):91 6 and Matsubara, S., et al., Estrogen Levels Influence Beta-3-adrenoreceptor-mediated Relaxation of the Female Rat Detrusor Muscle, Urology 59: 621 625,2002.
Another embodiment of the invention is a method of treating or preventing cardiovascular disease, restenosis, lowering levels of LDL cholesterol and inhibiting vascular smooth muscle cell proliferation in a mammal in need thereof by administering to the mammal a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above. The utility of SERMs in treating or preventing cardiovascular disease, restenosis, lowering levels of LDL cholesterol and inhibiting vascular smooth muscle cell proliferation is known in the art, see Nuttall, Μ E et al., “Idoxifene: a novel selective estrogen receptor modulator prevents bone loss and lowers cholesterol levels in ovariectomized rats and decreases uterine weight in intact rats,” Endocrinology 1998 December;139(12):522 434; Jordan, V. C. et al., “Selective estrogen receptor modulation and reduction in risk of breast cancer, osteoporosis and coronary heart disease,” Natl Cancer Inst 2001 October; 93(19):1449 57; Guzzo J A., “Selective estrogen receptor modulators-a new age of estrogens in cardiovascular disease?,” Clin Cardiol 2000 January;23(l):15 7; Simoncini T, Genazzani A R., “Direct vascular effects of estrogens and selective estrogen receptor modulators,” Curr Opin Obstet Gynecol 2000 June;12(3):181 7.
Another embodiment of the invention is a method of treating or preventing the impairment of cognitive functioning or cerebral degenerative disorders in a mammal in need thereof by administering to the mammal a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above. The utility of SERMs to prevent the impairment of cognitive functioning is known in the art, see Yaffe, Κ., K. Krueger, S. Sarkar, et al. 2001, “Cognitive function in postmenopausal women treated with raloxifene,” N. Eng. J. Med. 344: 1207 1213.
The present invention provides compounds effective in treating diseases characterized by abnormal mitosis and/or abnormal angiogenesis. See, US Pat. App. Pub.
TCX-003.88
IE o 7 θ θ ι
No. 2007/0010505, hereby incorporated by reference. Preferred compositions may also exhibit a change (increase or decrease) in estrogen receptor binding, improved absorption, transport (e.g., through blood-brain barrier and cellular membranes), biological stability, or decreased toxicity. The invention also provides compounds useful in the method, as described by the general formulae of the claims.
A mammalian disease characterized by undesirable cell mitosis, includes but is not limited to excessive or abnormal stimulation of endothelial cells (e.g., atherosclerosis), solid tumors and tumor metastasis, benign tumors, for example, hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas, vascular malfunctions, abnormal wound healing, inflammatory and immune disorders, Bechet's disease, gout or gouty arthritis, abnormal angiogenesis accompanying: rheumatoid arthritis, skin diseases, such as psoriasis, diabetic retinopathy and other ocular angiogenic diseases such as retinopathy of prematurity (retrolental fibroplasic), macular degeneration, corneal graft rejection, neovascular glaucoma and Osler Weber syndrome (Osler-Weber-Rendu disease). Other undesired angiogenesis involves normal processes including ovulation and implantation of a blastula. Accordingly, the compositions described above can be used to block ovulation and implantation of a blastula or to block menstruation (induce amenorrhea).
In certain embodiments, diseases associated with neovascularization can be treated according to the present invention. Such diseases include, but are not limited to, diabetic retinopathy, retinopathy of prematurity, corneal graft rejection, neovascular glaucoma and retrolental fibroplasias, epidemic keratoconjunctivitis, Vitamin A deficiency, contact lens overwear, atopic keratitis, superior limbic keratitis, pterygium keratitis sicca, Sjogren's, acne rosacea, phylectenulosis, syphilis, Mycobacteria infections, lipid degeneration, chemical burns, bacterial ulcers, fungal ulcers, Herpes simplex infections, Herpes zoster infections, protozoan infections, Kaposi's sarcoma, Mooren's ulcer, Terrien's marginal degeneration, marginal keratolysis, trauma, rheumatoid arthritis, systemic lupus, polyarteritis, Wegener's sarcoidosis, Scleritis, Steven-Johnson disease, pemphigoid, radial keratotomy, and corneal graph rejection.
Other diseases associated with neovascularization can be treated according to the present invention. Such diseases include, but are not limited to, diabetic retinopathy, macular degeneration, sickle cell anemia, sarcoid, syphilis, pseudoxanthoma elasticum,
TCX-003.88
IE
Paget's disease, vein occlusion, artery occlusion, carotid obstructive disease, chronic uveitis/vitritis, mycobacterial infections, Lyme's disease, systemic lupus erythematosis, retinopathy of prematurity, Eales' disease, Bechet's disease, infections causing a retinitis or choroiditis, presumed ocular histoplasmosis, Best's disease, myopia, optic pits, Stargart's disease, pars planitis, chronic retinal detachment, hyperviscosity syndromes, toxoplasmosis, trauma and post-laser complications. Other diseases include, but are not limited to, diseases associated with rubeosis (neovasculariation of the angle) and diseases caused by the abnormal proliferation of fibrovascular or fibrous tissue including all forms of proliferative vitreoretinopathy, whether or not associated with diabetes.
The present invention may also be used to treat cancerous diseases. Cancerous diseases include, but are not limited to, rhabdomyosarcoma, retinoblastoma, Ewing sarcoma, neuroblastoma, osteosarcoma, acoustic neuromas, neurofibromas, hemangiomas, breast cancer, prostrate cancer, renal cell cancer, brain tumors, ovarian cancer, colon cancer, bladder cancer, cutaneous melanoma, liver cancer, and lung cancer.
Another disease that can be treated according to the present invention is rheumatoid arthritis. It is believed that the blood vessels in the synovial lining of the joints undergo angiogenesis. In addition to forming new vascular networks, the endothelial cells release factors and reactive oxygen species that lead to pannus growth and cartilage destruction. The factors involved in angiogenesis may actively contribute to, and help maintain, the chronically inflamed state of rheumatoid arthritis.
Other diseases that can be treated according to the present invention are hemangiomas, Osler-Weber-Rendu disease, or hereditary hemorrhagic telangiectasia, solid or blood borne tumors and acquired immune deficiency syndrome.
In addition, the invention can be used to treat a variety of post-menopausal symptoms, osteoporosis, cardiovascular disease, Alzheimer's disease, to reduce the incidence of strokes, and as an alternative to prior estrogen replacement therapies. The compounds of the present invention can work by estrogenic and non-estrogenic biochemical pathways.
Also contemplated by the present invention are implants or other devices comprised of the compounds described herein or prodrugs thereof where the drug or prodrug is formulated in a bio-degradable or non-biodegradable polymer for sustained release. Nonbiodegradable polymers release the drug in a controlled fashion through physical or
mechanical processes without the polymer itself being degraded. Biodegradable polymers are designed to gradually be hydrolyzed or solubilized by natural processes in the body, allowing gradual release of the admixed drug or prodrug. Both bio-degradable and nonbiodegradable polymers and the process by which drugs are incorporated into the polymers for controlled release are well known to those skilled in the art. Examples of such polymers can be found in many references such as Brem et al, J. Neurosurg 74: pp. 441-446 (1991). These implants or devices can be implanted in the vicinity where delivery is desired, for example, at the site of a tumor or a stenosis.
Because anything not formed in the body as a natural component may elicit extreme and unexpected responses, such as blood vessel closure due to thrombus formation or spasm, and because damage to blood vessels by the act of insertion of a vascular stent may be extreme and unduly injurious to the blood vessel surface, it is prudent to protect against such events. Restenosis is a re-narrowing or blockage of an artery at the same site where treatment, such as an angioplasty or stent procedure, has already taken place. If restenosis occurs within a stent that has been placed in an artery, it is technically called “in-stent restenosis,” the end result being a nanowing in the artery caused by a build-up of substances that may eventually block the flow of blood. The compounds that are part of the present invention are especially useful to coat vascular stents to prevent restenosis, The coating should preferably be a biodegradable or non-biodegradable polymer that allows for a slow release of a compound of the present invention thereby preventing the restenosis event.
In one embodiment, provided are compounds, compositions, and methods for treating a condition by modulating HSP90 activity. See, US Pat. App. Pub. Nos. 2007/0087998 and 2007/0123546, both of which are hereby incorporated by reference. In some aspects, the condition is a cellular proliferative, viral, autoimmune, cardiovascular, or central nervous system disease.
In one embodiment, provided are compounds, compositions, and methods for treating cancers such as, for example, lung and bronchus; prostate; testicular tumor; breast; pancreas; colon and rectum; thyroid; stomach; liver and intrahepatic bile duct; kidney and renal; pelvis; urinary bladder; uterine corpus; uterine cervix; ovary; multiple myeloma; esophagus; acute myelogenous leukemia; chronic myelogenous leukemia; lymphocytic leukemia; lymphoma; myeloid leukemia; master cell leukemia, brain; oral cavity and
TCX-003.88
pharynx; larynx; head; neck; glioblastoma; small intestine; gastrointestinal stromal tumors (GISTs); gastric tumor; non-hodgkin lymphoma; melanoma; and villous colon adenoma.
In one embodiment, provided are compounds, compositions, and methods tor treating a viral disease. Such diseases include, for example, viral diseases mediated by hepatitis B virus (HBV), hepatitis C virus (HCV), or herpes simplex virus type 1 (HSV-1).
In one embodiment, provided are compounds, compositions, and methods for treating an autoimmune disease. In some aspects, the autoimmune disease is mediated by persistent lymphocyte activation.
In one embodiment, provided are compounds, compositions, and methods for treating a cardiovascular or central nervous system disease.
As discussed elsewhere herein, the methods of the invention include administration of an effective amount of a compound of the invention, or a salt thereof as the active ingredient. Pharmaceutically acceptable salts are typically salts of non-toxic type commonly used, such as salts with organic acids, inorganic acids and amino acids. These salts may be prepared by the methods known to chemists of ordinary skill.
In certain embodiments, the present invention relates to a method of eliciting an estrogen receptor modulating effect in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions.
In certain embodiments, the present invention relates to the aforementioned method, wherein the estrogen receptor modulation effect is an estrogen receptor agonizing effect.
In certain embodiments, the present invention relates to the aforementioned method, wherein the estrogen receptor agonizing effect is an ERa receptor agonizing effect.
In certain embodiments, the present invention relates to the aforementioned method, wherein the estrogen receptor modulation effect is an estrogen receptor antagonizing effect.
In certain embodiments, the present invention relates to the aforementioned method, wherein the estrogen receptor antagonizing effect is an ERa receptor antagonizing effect.
In certain embodiments, the present invention relates to a method of treating a disease in a mammal in need thereof by administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or aforementioned
TCX-003.88
pharmaceutical compositions, wherein said disease is selected from the group consisting of bone loss, bone fractures, cartilage degeneration, uterine fibroid disease, hot flashes, increased levels of LDL cholesterol, impairment of cognitive functioning, cerebral degenerative disorders, restenosis, gynecomastia, vascular smooth muscle cell proliferation, obesity, incontinence, obesity, hormone dependent breast cancer, osteoporosis, estrogen deficiency, arthritis, cardiovascular disease, ovarian cancer, artherosclerosis, colon tumor, endometriosis, Alzheimer’s disease, non-insulin dependent (type II) diabetes, infertility, prostrate tumor, melanoma, acne, hypercholesterolemia, CNS disease, contraception, conditions related to hair follicles, macular degeneration, urinary incontinence, estrogen receptor-expressing and estrogen receptor-expressing tumors, and leukaemia.
In certain embodiments, the present invention relates to the aforementioned method, wherein the disease is hormone dependent breast cancer.
In certain embodiments, the present invention relates to the aforementioned method, wherein the disease is estrogen dependent breast cancer.
In certain embodiments, the present invention relates to the aforementioned method, wherein the disease is tamoxifene-resistant breast cancer.
In certain embodiments, the present invention relates to a method of modulating the activity of Hsp90 in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions.
In certain embodiments, the present invention relates to a method of modulating tublin polymerization/depolymerization in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions.
In certain embodiments, the present invention relates to a method of modulating the activity of protein kinase C in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions.
In certain embodiments, the present invention relates to a method of modulating the activity of calmodulin-dependent enzymes in a mammal in need thereof, comprising
TCX-003.88 «θ7046δ administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions.
In certain embodiments, the present invention relates to a method of modulating the activity of acyl coenzyme A in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions.
In certain embodiments, the present invention relates to a method of modulating the activity of cholesterol acyl transferase in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions.
COMBINATION WITH OTHER KNOWN THERAPEUTICALLY ACTIVE AGENTS
The instant compounds are also useful in combination with known agents useful for treating or preventing bone loss, bone fractures, osteoporosis, glucocorticoid induced osteoporosis, Paget's disease, abnormally increased bone turnover, periodontal disease, tooth loss, rheumatoid arthritis, osteoarthritis, periprosthetic osteolysis, osteogenesis imperfecta, metastatic bone disease, hypercalcemia of malignancy, and multiple myeloma, cartilage degeneration, endometriosis, uterine fibroid disease, cancer of the breast, uterus or prostate, hot flashes, cardiovascular disease, impairment of cognitive function, cerebral degenerative disorders, restenosis, gynecomastia, vascular smooth muscle cell proliferation, obesity and incontinence. Combinations of the presently disclosed compounds with other agents useful in treating or preventing osteoporosis or other bone disorders are within the scope of the invention. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the disease involved. Such agents include the following: an organic bisphosphonate; a cathepsin K inhibitor; an estrogen or an estrogen receptor modulator; an androgen receptor modulator; an inhibitor of osteoclast proton ATPase; an inhibitor of HMG-CoA reductase; an inhibitor of cholesxterol ester transfer protein; an integrin receptor antagonist; an osteoblast anabolic agent, such as PmH; calcitonin; Vitamin D or a synthetic Vitamin D analogue; an aromatase inhibitor; selective serotonin reuptake inhibitors (SSRIs); an anticancer agent; and the pharmaceutically acceptable salts and mixtures thereof.
One combination is a compound of the present invention and an organic bisphosphonate. Another combination is a compound of the present invention and a
-77TCX-003.88 it cathepsin K inhibitor. Another combination is a compound of the present invention and an estrogen. Another combination is a compound of the present invention and an androgen receptor modulator. Another combination is a compound of the present invention and an osteoblast anabolic agent.
The compounds of the present invention can be used in combination with other agents useful for treating estrogen-mediated conditions. The individual components of such combinations can be administered separately at different times during the course of therapy or concurrently in divided or single combination forms. The instant invention is therefore to be understood as embracing all such regimes of simultaneous or alternating treatment and the term “administering” is to be interpreted accordingly. It will be understood that the scope of combinations of the compounds of this invention with other agents useful for treating cathepsin-mediated conditions includes in principle any combination with any pharmaceutical composition useful for treating disorders related to estrogen functioning.
The scope of the invention therefore encompasses the use of the instantly claimed compounds in combination with a second agent selected from: an organic bisphosphonate; a cathepsin K inhibitor; an estrogen; an estrogen receptor modulator; an androgen receptor modulator; an inhibitor of osteoclast proton ATPase; an inhibitor of HMG-CoA reductase; an integrin receptor antagonist; an osteoblast anabolic agent; calcitonin; Vitamin D; a synthetic Vitamin D analogue; a selective serotonin reuptake inhibitor; an aromatase inhibitor; and the pharmaceutically acceptable salts and mixtures thereof.
In addition, the scope of the invention further encompasses the use of the instantly claimed compounds in combination with a second agent selected from the group consisting of gemcitabine, capecitabine, methotrexate, taxol, taxotere, mercaptopurine, thioguanine, hydroxyurea, cytarabine, cyclophosphamide, ifosfamide, nitrosoureas, cisplatin, carboplatin, mitomycin, dacarbazine, procarbizine, etoposide, teniposide, campathecins, bleomycin, doxorubicin, idarubicin, daunorubicin, dactinomycin, plicamycin, mitoxantrone, L-asparaginase, doxorubicin, epirubicin, 5-fluorouracil (5-FU), taxanes such as docetaxel and paclitaxel, leucovorin, levamisole, irinotecan, estramustine, etoposide, nitrogen mustards, BCNU, nitrosoureas such as carmustine and lomustine, vinca alkaloids such as vinblastine, vincristine and vinorelbine, platinum complexes such as cisplatin, carboplatin and oxaliplatin, imatinib mesylate, hexamethylmelamine, topotecan, tyrosine kinase inhibitors, tyrphostins herbimycin A, genistein, erbstatin, and lavendustin A.
-78TCX-003.88
In addition, the scope of the invention further encompasses the use of the instantly claimed compounds in combination with a second agent selected from the group consisting of alkylating agents, nitrogen mustards, cyclophosphamide, ifosfamide, trofosfamide, chlorambucil, nitrosoureas, carmustine (BCNU), lomustine (CCNU), alkylsulphonates, busulfan, treosulfan, triazenes, dacarbazine, platinum complexes, cisplatin, carboplatin, oxaliplatin, plant alkaloids, vinca alkaloids, vincristine, vinblastine, vindesine, vinorelbine), taxoids, paclitaxel, docetaxel, DNA topoisomerase inhibitors, epipodophyllins, etoposide, teniposide, topotecan, 9-aminocamptothecin, camptothecin, crisnatol, mitomycins, mitomycin C, anti-metabolites, anti-folates, DHFR inhibitors, methotrexate, trimetrexate, IMP dehydrogenase inhibitors, mycophenolic acid, tiazofurin, ribavirin, EICAR, ribonuclotide reductase inhibitors, hydroxyurea, deferoxamine, pyrimidine analogs, uracil analogs, 5-fluorouraciI, floxuridine, doxifluridine, ratitrexed, cytosine analogs, cytarabine (ara C), cytosine arabinoside, fludarabine, gemcitabine, capecitabine, purine analogs, mercaptopurine, thioguanine, DNA antimetabolites, 3-HP, 2’-deoxy-5-fluorouridine, 5-HP, alpha-TGDR, aphidicolin glycinate, ara-C, 5-aza-2’-deoxycytidine, beta-TGDR, cyclocytidine, guanazole (inosine glycodialdehyde), macebecin II, pyrazoloimidazole, hormonal therapies, receptor antagonists, anti-estrogen, tamoxifen, raloxifene, megestrol, LHRH agonists, goserelin, leuprolide acetate, anti-androgens, flutamide, bicalutamide, retinoids/deltoids, cis-retinoic acid, vitamin A derivative, all-trans retinoic acid (ATRAIV), vitamin D3 analogs, EB 1089, CB 1093, KH 1060, photodynamic therapies, vertoporfin, BPD-MA, phthalocyanine, photosensitizer Pc4, demethoxy-hypocrellin A (2BA-2-DMHA), cytokines, interferon-a, interferon-β, interferon-γ, tumor necrosis factor, angiogenesis inhibitors, angiostatin (plasminogen fragment), antiangiogenic antithrombin III, angiozyme, ABT-627, Bay 12-9566, benefin, bevacizumab, BMS-275291, cartilagederived inhibitor (CDI), CAI, CD59 complement fragment, CEP-7055, Col 3, combretastatin A-4, endostatin (collagen XVIII fragment), fibronectin fragment, Gro-beta, halofuginone, heparinases, heparin hexasaccharide fragment, HMV833, human chorionic gonadotropin (hCG), IM-862, interferon inducible protein (IP-10), interleukin-12, kringle 5 (plasminogen fragment), marimastat, metalloproteinase inhibitors (TIMPs), 2methoxyestradiol, MMI270 (CGS 27023A), MoAb IMC-1C11, neovastat, NM-3, panzem, PI-88, placental ribonuclease inhibitor, plasminogen activator inhibitor, platelet factor-4 (PF4), prinomastat, prolactin 16kD fragment, proliferin-related protein (PRP), PTK 787/ZK 222594, retinoids, solimastat, squalamine, SS 3304, SU 5416, SU 6668, SU 11248,
-79TCX-003.88
If &
tetrahydrocortisol-S, tetrathiomolybdate, thalidomide, thrombospondin-1 (TSP-1), TNP470, transforming growth factor-beta (TGF-β), vasculostatin, vasostatin (calreticulin fragment), ZD 6126, ZD 6474, famesyl transferase inhibitors (FTI), bisphosphonates, antimitotic agents, allocolchicine, halichondrin B, colchicine, colchicine derivative, dolstatin 10, maytansine, rhizoxin, thiocolchicine, trityl cysteine, isoprenylation inhibitors, dopaminergic neurotoxins, l-methyl-4-phenylpyridinium ion, cell cycle inhibitors, staurosporine, actinomycins, actinomycin D, dactinomycin, bleomycins, bleomycin A2, bleomycin B2, peplomycin, anthracycline, daunorubicin, doxorubicin (adriamycin), idarubicin, epirubicin, pirarubicin, zorubicin, mitoxantrone, MDR inhibitors, verapamil,
Ca ATPase inhibitors, and thapsigargin.
In yet other embodiments the scope of the invention further encompasses the use of the instantly claimed compounds in combination with a second agent selected from the group consisting of acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; cedefmgol; chlorambucil; cirolemycin; cisplatin; cladribine; crisnatol mesylate; cyclophosphamide; cytarabine; dacarbazine; dactinomycin; daunorubicin hydrochloride; decitabine; dexormaplatin; dezaguanine; dezaguanine mesylate; diaziquone; docetaxel; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflomithine hydrochloride; elsamitrucin; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; erbulozole; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; fadrozole hydrochloride; fazarabine; fenretinide; floxuridine; fludarabine phosphate; fluorouracil; flurocitabine; fosquidone; fostriecin sodium; gemcitabine hydrochloride; hydroxyurea; idarubicin hydrochloride; ifosfamide; ilmofosine; interleukin II (including recombinant interleukin II, or rIL2), interferon alfa-2a; interferon alfa-2b; interferon alfa-nl; interferon alfa-n3; interferon beta-la; interferon gamma-Ib; iproplatin; irinotecan hydrochloride; lanreotide acetate; letrozole; leuprolide acetate; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate;
-80TCX-003.88
ΙΕο 7 0 4 6 6 melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; metoprine; meturedepa; mitindomide; mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazole; nogalamycin; ormaplatin; oxisuran; paclitaxel; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; riboprine; rogletimide; safingol; safmgol hydrochloride; semustine; simtrazene; sparfosate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin; streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan sodium; tegafur; teloxantrone hydrochloride; temoporfin; teniposide; teroxirone; testolactone; thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; toremifene citrate; trestolone acetate; triciribine phosphate; trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard; uredepa; vapreotide; verteporfin; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorozole; zeniplatin; zinostatin; zorubicin hydrochloride.
Further anti-cancer drugs that can be used in the present invention include, but are not limited to: 20-epi- 1,25-dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morphogenetic protein 1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara CDP DL PTBA; arginine deaminase; asulacrine; atamestane; atrimustine; axinastatin 1; axinastatin 2; axinastatin 3; azasetron; azatoxin; azatyrosine; baccatin III derivatives; balanol; batimastat; BCR/ABL antagonists; benzochlorins; benzoylstaurosporine; beta lactam derivatives; beta alethine; betaclamycin B; betulinic acid; bFGF inhibitor; bicalutamide; bisantrene;
bisaziridinylspermine; bisnafide; bistratene A; bizelesin; breflate; bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C; camptothecin derivatives; canarypox IL2; carboxamide amino triazole; carboxyamidotriazole; CaRest M3; CARN 700; cartilage derived inhibitor; carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B;
-81 TCX-003.88
ΙΕο 7 0 4 6 6 cetrorelix; chlorlns; chloroquinoxaline sulfonamide; cicaprost; cis porphyrin; cladribine; clomifene analogues; clotrimazole; collismycin A; collismycin B; combretastatin A4; combretastatin analogue; conagenin; crambescidin 816; crisnatol; cryptophycin 8; cryptophycin A derivatives; curacin A; cyclopentanthraquinones; cycloplatam; cypemycin; cytarabine ocfosfate; cytolytic factor; cytostatin; dacliximab; decitabine; dehydrodidemnin B; deslorelin; dexamethasone; dexifosfamide; dexrazoxane; dexverapamil; diaziquone; didemnin B; didox; diethylnorspermine; dihydro 5 azacytidine; dihydrotaxol, 9 ; dioxamycin; diphenyl spiromustine; docetaxel; docosanol; dolasetron; doxifluridine; droloxifene; dronabinol; duocarmycin SA; ebselen; ecomustine; edelfosine; edrecolomab; eilornithine; elemene; emitefur; epirubicin; epristeride; estramustine analogue; estrogen agonists; estrogen antagonists; etanidazole; etoposide phosphate; exemestane; fadrozole; fazarabine; fenretinide; filgrastim; finasteride; flavopiridol; flezelastine; fluasterone; fludarabine; fluorodaunorunicin hydrochloride; forfenimex; formestane; fostriecin; fotemustine; gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase inhibitors; gemcitabine; glutathione inhibitors; hepsulfam; heregulin; hexamethylene bisacetamide; hypericin; ibandronic acid; idarubicin; idoxifene; idramantone; ilmofosine; ilomastat; imidazoacridones; imiquimod; immunostimulant peptides; insulin like growth factor 1 receptor inhibitor; interferon agonists; interferons; interleukins; iobenguane; iododoxorubicin; ipomeanol, 4 ; iroplact; irsogladine; isobengazole; isohomohalicondrin B; itasetron; jasplakinolide; kahalalide F; lamellarin N triacetate; lanreotide; leinamycin; lenograstim; lentinan sulfate; leptolstatin; letrozole; leukemia inhibiting factor; leukocyte alpha interferon; leuprolide+estrogen+progesterone; leuprorelin; levamisole; liarozole; linear polyamine analogue; lipophilic disaccharide peptide; lipophilic platinum complexes; lissoclinamide 7; lobaplatin; lombricine; lometrexol; lonidamine; losoxantrone; lovastatin; loxoribine; lurtotecan; lutetium texaphyrin; lysofylline; lytic peptides; maitansine; mannostatin A; marimastat; masoprocol; maspin; matrilysin inhibitors; matrix metalloproteinase inhibitors; menogaril; merbarone; meterelin; methioninase; metoclopramide; MIF inhibitor; mifepristone; miltefosine; mirimostim; mismatched double stranded RNA; mitoguazone; mitolactol; mitomycin analogues; mitonafide; mitotoxin fibroblast growth factor saporin; mitoxantrone; mofarotene; molgramostim; monoclonal antibody, human chorionic gonadotrophin; monophosphoryl lipid A+myobacterium cell wall sk; mopidamol; multiple drug resistance gene inhibitor; multiple tumor suppressor 1 based therapy; mustard anti-cancer agent; mycaperoxide B; mycobacterial cell wall extract;
-82TCX-003.88
myriaporone; N acetyldinaline; N substituted benzamides; nafarelin; nagrestip; naloxone+pentazocine; napavin; naphterpin; nartograstim; nedaplatin; nemorubicin; neridronic acid; neutral endopeptidase; nilutamide; nisamycin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; 06 benzylguanine; octreotide; okicenone; oligonucleotides; onapristone; ondansetron; ondansetron; oracin; oral cytokine inducer; ormaplatin; osaterone; oxaliplatin; oxaunomycin; paclitaxel; paclitaxel analogues; paclitaxel derivatives; palauamine; palmitoylrhizoxin; pamidronic acid; panaxytriol; panomifene; parabactin; pazelliptine; pegaspargase; peldesine; pentosan polysulfate sodium; pentostatin; pentrozole; perflubron; perfosfamide; perillyl alcohol; phenazinomycin; phenylacetate; phosphatase inhibitors; picibanil; pilocarpine hydrochloride; pirarubicin; piritrexim; placetin A; placetin B; plasminogen activator inhibitor; platinum complex; platinum complexes; platinum triamine complex; porfimer sodium; porfiromycin; prednisone; propyl bis acridone; prostaglandin J2; proteasome inhibitors; protein A based immune modulator; protein kinase C inhibitor; protein kinase C inhibitors, microalgal; protein tyrosine phosphatase inhibitors; purine nucleoside phosphorylase inhibitors; purpurins; pyrazoloacridine; pyridoxylated hemoglobin polyoxyethylene conjugate; raf antagonists; raltitrexed; ramosetron; ras famesyl protein transferase inhibitors; ras inhibitors; ras GAP inhibitor; retelliptine demethylated; rhenium Re 186 etidronate; rhizoxin; ribozymes; RII retinamide; rogletimide; rohitukine; romurtide; roquinimex; rubiginone BI; ruboxyl; safmgol; saintopin; SarCNU; sarcophytol A; sargramostim; Sdi 1 mimetics; semustine; senescence derived inhibitor 1; sense oligonucleotides; signal transduction inhibitors; signal transduction modulators; single chain antigen binding protein; sizofiran; sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; somatomedin binding protein; sonermin; sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin 1; squalamine; stem cell inhibitor; stem cell division inhibitors; stipiamide; stromelysin inhibitors; sulfinosine; superactive vasoactive intestinal peptide antagonist; suradista; suramin; swainsonine; synthetic glycosaminoglycans; tallimustine; tamoxifen methiodide; tauromustine; tazarotene; tecogalan sodium; tegafur; tellurapyrylium; telomerase inhibitors; temoporfin; temozolomide; teniposide; tetrachlorodecaoxide; tetrazomine; thaliblastine; thiocoraline; thrombopoietin; thrombopoietin mimetic; thymalfasin; thymopoietin receptor agonist; thymotrinan; thyroid stimulating hormone; tin ethyl etiopurpurin; tirapazamine; titanocene bichloride; topsentin; toremifene; totipotent stem cell factor; translation inhibitors; tretinoin; triacetyluridine; triciribine; trimetrexate; triptorelin; tropisetron;
-83TCX-003.88
IE turosteride; tyrosine kinase inhibitors; tyrphostins; UBC inhibitors; ubenimex; urogenital sinus derived growth inhibitory factor; urokinase receptor antagonists; vapreotide; variolin B; vector system, erythrocyte gene therapy; velaresol; veramine; verdins; verteporfm; vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeniplatin; zilascorb; and zinostatin stimalamer,
It is a further aspect of the invention the compounds of the invention can be administered in conjunction with chemical agents that are understood to mimic the effects of radiotherapy and/or that function by direct contact with DNA. Preferred agents for use in combination with the Compounds of the Invention for treating cancer include, but are not limited to cis-diamminedichloro platinum (II) (cisplatin), doxorubicin, 5-fluorouracil, taxol, and topoisomerase inhibitors such as etoposide, teniposide, irinotecan and topotecan.
KITS
The invention encompasses kits that can simplify the administration of the compounds of the invention or composition of the invention to a subject.
A typical kit of the invention comprises unit dosages of the compounds of the invention. In one embodiment, the unit dosage form is in a container, which can be sterile, containing an effective amount of one of the compounds of the invention and a pharmaceutically acceptable carrier or vehicle. In another embodiment, the unit dosage form is in a container containing an effective amount of one of the compounds of the invention as a lyophilate. In this instance, the kit can further comprise another container which contains a solution useful for the reconstitution of the lyophilate. The kit can also comprise a label or printed instructions for use of the compounds of the invention. In one embodiment, the kit comprises multiple containers: (a) a first container containing an unit dosage form of compound of the invention, and (b) one or more additional containers each containing a unit dosage form of one or more additional agents or pharmaceutically acceptable salts thereof.
In a further embodiment, the kit comprises a unit dosage form of a pharmaceutical composition of the invention.
Kits of the invention can further comprise one or more devices that are useful for administering the unit dosage forms of the compounds of the invention or a pharmaceutical composition of the invention. Examples of such devices include, but are not limited to, a syringe, a drip bag, a patch or an enema, which optionally contain the unit dosage forms.
-84TCX-003.88 assays IEO 7 0 4 6 6
The utility of the compounds of the instant invention can be readily determined by methods well known to one of ordinary skill in the art. These methods may include, but are not limited to, the assays described in detail below, as well as the assays described in the
Exemplification. The assays described in the Exemplification were assays used as part of the Virtual Screening process described herein.
ESTROGEN RECEPTOR BINDING ASSAY. The estrogen receptor ligand binding assays are designed as scintillation proximity assays employing the use of tritiated estradiol and recombinant expressed estrogen receptors. The full length recombinant human ERalpha and ER-beta proteins are produced in a bacculoviral expression system. ER-alpha or ER-beta extracts are diluted 1:400 in phosphate buffered saline containing 6 mM alphamonothiolglycerol. 200 pL aliquots of the diluted receptor preparation are added to each well of a 96-well Flashplate. Plates are covered with Saran Wrap and incubated at 4 °C. overnight.
The following morning, a 20 pL aliquot of phosphate buffered saline containing 10% bovine serum albumin is added to each well of the 96 well plate and allowed to incubate at 4 °C for 2 hours. Then the plates are washed with 200 pL of buffer containing 20 mM Tris (pH 7.2), 1 mM EDTA, 10% Glycerol, 50 mM KCl, 5 and 6 mM alphamonothiolglycerol. To set up the assay in these receptor coated plates, add 178 pL of the same buffer to each well of the 96 well plate. Then add 20 pL of a 10 nM solution of 3Hestradiol to each well of the plate.
Test compounds are evaluated over a range of concentrations from 0.01 nM to 1000 nM. The test compound stock solutions should be made in 100% DMSO at lOO.times. the final concentration desired for testing in the assay. The amount of DMSO in the test wells of the 96 well plate should not exceed 1%. The final addition to the assay plate is a 2 pL aliquot of the test compound which has been made up in 100% DMSO. Seal the plates and allow them to equilibrate at room temperature for 3 hours. Count the plates in a scintillation counter equipped for counting 96 well plates.
OVARIECTOMIZED RAT ASSAY. In the ovariectomized (OVX) Rat Assay, estrogen-deficiency is used to induce cancellous osteopenia (e.g., low bone mineral density [BMD; mg/cm2]), associated with accelerated bone resorption and formation. Both the BMD and bone resorption/formation outcomes are used to model the changes in bone that
-85TCX-003.88 |Γ occur as women pass through menopause. The OVX Rat Assay is the principal in vivo assay used by all major academic and industrial laboratories studying the efficacy of new chemical entities in preventing estrogen-deficiency bone loss.
Sprague-Dawley female rats aged 6 8 months are OVXd and, within 24 hours, started on treatment for 42 days with vehicle or multiple doses of test compound. Untreated sham-OVX and alendronate-treated (0.003 mg/kg s.c., q.d.) or 17-beta-estradiol-treated (0.004 mg/kg s.c., q.d.) groups are included as positive controls. Test compounds may be administered orally, subcutaneously, or by infusion through subcutaneously-implanted minipump. Before necropsy, in vivo dual labeling with calcein (8 mg/kg by subcutaneous injection), a bone seeking fluorochrome, is completed. At necropsy, blood, femurs, a vertebral body segment, and the uterus, are obtained.
The routine endpoints for the OVX Rat Assay include assessments of bone mass, bone resorption, and bone formation. For bone mass, the endpoint is BMD of the distal femoral metaphysis, a region that contains about 20% cancellous bone. The vertebral segment, a region with about 25% cancellous bone may also be used for BMD determination. The BMD measurement is made by dual energy x-ray absorptiometry (DXA, Hologic 4500A; Waltham, Mass.). For bone resorption, the endpoint is urinary deoxypyridinoline crosslinks, a bone collagen breakdown product (uDPD; expressed as nM DPD/nM creatinine). This measurement is made with a commercially available kit (Pyrilinks; Metra Biosystems, Mountain View, Calif.). For bone formation, the endpoints are mineralizing surface and mineral apposition rate, histomorphometric measures of osteoblast number and activity. This measurement is done on 5 . pm sections of the nondecalcified proximal tibial metaphysis, using a semi-automated system (Bioquant; R&M Biometrics; Nashville, Tenn.). Similar endpoints and measuring techniques for each endpoint are commonly used in postmenopausal women.
RA T CHOLESTEROL LOWERING ASSA Y. Sprague-Dawley rats (5 per group) weighing about 250 g were subcutaneously dosed with compounds of the present invention dissolved in propylene glycol for 4 days. A group of 5 rats was dosed with vehicle only. On the fifth day, rats were euthanized with carbon dioxide and their blood samples were obtained. Plasma levels of cholesterol were assayed from these samples with commercially available cholesterol determination kits from Sigma.
-86TCX-003.88
IS 0 7 ο 4 ο θ
MCF-7 ESTROGEN DEPENDENT PROLIFERATION ASSAY. MCF-7 cells (ATCC #HTB-22) are human mammary gland adenocarcinoma cells that require estrogen for growth. The growth media (GM) for the MCF-7 cells is Minimum Essential Media (without phenol red) supplemented with fetal bovine serum (FBS) to 10%. The FBS serves as the sole source of estrogen and this GM supports the full growth of the cells and is used for the routine growth of the cell cultures. When MCF-7 cells are placed in a media in which 10% Charcoal-Dextran treated fetal bovine serum (CD-FBS) is substituted for FBS, the cells will cease to divide but will remain viable. The CD-FBS does not contain detectable levels of estrogen and the media containing this sera is referred to as Estrogen Depleted Media (EDM). The addition of estradiol to EDM stimulates the growth of the MCF-7 cells in a dose dependent manner with an EC50 of 2 pM.
Growing MCF-7 cells are washed several times with EDM and the cultures then maintained in EDM for a minimum of 6 days in order to deplete the cells of endogenous estrogen. On day 0 (at the startof the assay), these estrogen depleted cells are plated into 96well cell culture plates at a density of 1000 cells/well in EDM in a volume of 180 pL/well. On day 1 test compounds are diluted in a 10-fold dilution series in EDM and 20 pL of these dilutions added to the 180 pL of media in the appropriate well of the cell plate resulting in a further 1:10 dilution of the test compounds. On days 4 and 7 of the assay, the culture supernatant is aspirated and replaced with fresh EDM and test compound dilutions as above. The assay is terminated at day 8-10 when the appropriate controls reach 80 90% confluency. At this point, the culture supernatants are aspirated, the cells washed 2 times with PBS, the wash solution aspirated and the protein content of each well determined.
Each drug dilution is evaluated on a minimum of 5 wells and the range of dilution of the test compounds in the assay is 0.001 nM to 1000 nM. The assay in the above format is employed to determine the estradiol agonist potential of a test compound.
In order to evaluate the antagonist activity of a test compound, the MCF-7 cells are maintained in EDM for a minimum of 6 days. Then on day 0 (at the start of the assay), these estrogen depleted cells are plated into 96-well cell culture plates at a density of 1000 cells/well in EDM in a volume of 180 pL/well. On day 1 the test compounds in fresh media containing 3 pM estradiol are applied to the cells. On days 4 and 7 of the assay, the culture supernatant is aspirated and replaced with fresh EDM containing 3 pM estradiol and the test compound. The assay is terminated at day 8-10 when the appropriate controls reach 80 90% confluency and the protein content of each well is determined as above.
-87TCX-003.88
ΙΕΟ 70 4 66
RAT ENDOMETRIOSIS MODEL. Animals: Species: Rattus norvegicus; Strain: Sprague-Dawley CD; Supplier: Charles River Laboratories, Raleigh, N.C.; Sex: Female; Weight: 200 240 gram. Rats are single-housed in polycarbonate cages and are provided Teklad Global Diet 2016 (Madison, Wis.) and bottled reverse osmosis purified H2O ad libitum. They are maintained on a 12/12 light/dark cycle.
Rats are anesthetized with Telazo.TM. (20 mg/kg, ip) and oxymorphone (0.2 mg/kg sc) and positioned dorsoventrally on a sterile drape. Body temperature is maintained using a underlying circulating water blanket. The surgical sites are shaved with clippers and cleaned using three cycles of betadine/isopropyl alcohol or Duraprep.RTM. (3M). The incisional area is covered with a sterile drape.
Using aseptic technique, a 5 cm midline lower abdominal incision is made through the skin, subcutaneous and muscle layers. A bilateral ovariectomy is performed. The left uterine blood vessels are ligated and a 7 mm segment of the left uterine horn is excised. The uterus is closed with 4-0 gut suture. The myometrium is aseptically separated from the endometrium and trimmed to 5.times.5 mm. The trimmed section of the endometrium is transplanted to the ventral peritoneal wall with the epithelial lining of the segment opposed to the peritoneal wall. The explanted endometrial tissue is sutured at its four corners to the body wall using sterile 6-0 silk. The abdominal muscular layer is closed using sterile 4-0 chromic gut. The skin incision is closed using sterile stainless surgical clips. A sterile 90day sustained release estrogen pellet (Innovative Research of America, 0.72 ng/pellet; circulating estrogen equivalent of 200 250 pg/mL) is implanted subcutaneously in the dorsal lateral scapular area. A sterile implantable programmable temperature transponder (RPTM) (BMDS, Seaford, Del.) is injected subcutaneously in the dorsoscapular region. The rats are observed until fully ambulatory, and allowed to recover from surgery undisturbed for 3 weeks.
Three weeks after transplantation of the endometrial tissue, the animals undergo a repeat laparotomy using aseptic surgical site preparation and technique. The explant is evaluated for graft acceptance, and the area is measured with calipers and recorded. The animals with rejected grafts are removed from the study. Animals are sorted to create similar average explant volume per group.
TCX-003.88 «070405
Drug or vehicle (control) treatment is initiated one day after the second laparotomy and continued for 14 days. Body temperature is recorded every other day at 10:00 am using the BMDS scanner.
At the end of the 14 day treatment period, the animals are euthanized by CO2 overdose. Blood is collected by cardiocentesis for circulating estrogen levels. The abdomen is opened, the explant is examined, measured, excised, and wet weight is recorded. The right uterine horn is excised, and wet and dry weights are recorded.
ANTI-PROLIFERA TIVE ACTIVITY IN SITU. Anti-proliferative activity can be evaluated in situ by testing the ability of an improved estradiol derivative to inhibit the proliferation of new blood vessel cells (angiogenesis). A suitable assay is the chick embryo chorioallantoic membrane (CAM) assay described by Crum et al. Science 230:1375 (1985). See also, U.S. Pat. No. 5,001,116, hereby incorporated by reference, which describes the CAM assay. Briefly, fertilized chick embryos are removed from their shell on day 3 or 4, and a methylcellulose disc containing the drug is implanted on the chorioallantoic membrane. The embryos are examined 48 hours later and, if a clear avascular zone appears around the methylcellulose disc, the diameter of that zone is measured.
ANTI-PROLIFERATIVE ACTIVITY IN VITRO. Assays relevant to these mechanisms of action and inhibition of cell proliferation are well-known in the art. For example, anti-mitotic activity mediated by effects on tubulin polymerization activity can be evaluated by testing the ability of an estradiol derivative to inhibit tubulin polymerization and microtubule assembly in vitro. Microtubule assembly can be followed in a Gilford recording spectrophotometer (model 250 or 2400S) equipped with electronic temperature controllers. A reaction mixture typically contains 1.0 M monosodium glutamate (pH 6.6), 1.0 mg/ml (10 μΜ) tubulin, 1.0 mM MgCl2,4% (v/v) dimethylsulfoxide and 20-75 .mu.M of a composition to be tested. The reaction mixtures are incubated for 15 min. at 37.degree. C. and then chilled on ice. After addition of 10 pL 2.5 mM GTP, the reaction mixture is transferred to a cuvette at 0° C., and a baseline established. At time zero, the temperature controller of the spectrophotometer is set at 37° C. Microtubule assembly is evaluated by increased turbity at 350 nm. Alternatively, inhibition of microtubule assembly can be followed by transmission electron microscopy as described in Example 2 of U.S. Pat. Nos. 5,504,074, 5,661,143, and 5,892,069, the disclosures of which are incorporated herein by reference.
- 80 TCX-003.88
Other such assays include counting of cells in tissue culture plates or assessment of cell number through metabolic assays or incorporation into DNA of labeled (radiochemically, for example .sup.3H-thymidine, or fluorescently labeled) or immunoreactive (BrdU) nucleotides. In addition, antiangiogenic activity may be evaluated through endothelial cell migration, endothelial cell tubule formation, or vessel outgrowth in ex-vivo models such as rat aortic rings.
INHIBITOR BINDING POTENCY: TRF BINDING ASSA Y. The binding potency of HSP90 inhibitors can be measured by a TRF binding assay. TRF competition binding assays may be performed to determine the binding potency (IC50 values) of HSP90 inhibitors. Purified His-tagged N-terminal ATP binding domain (amino acid residues 9236) of HSP90a (HSP90a GenelD: 3320; mRNA Sequence NM-005348) was incubated for two hours at room temperature in binding buffer (50 mM HEPES, 6 mM MgCl2, 20 mM KC1 and 0.1% BSA) with biotinylated radicicol and progressively higher concentrations of the competing compounds. A fraction of the mixture was transferred to capture plates (coated with streptavidin) and incubated for one hour at room temperature. After washing with DELFIA wash buffer, europium-labeled anti-his antibody was added and incubated for two hours at room temperature, followed by washing with DELFIA buffer. DELFIA enhancement solution was then added. After gentle shaking for 10 minutes, the plates were read in VICTOR for europium counts.
Note: IC50 values can also be determined using published methods in the following references: 1. Carreras, C. W., A. Schirmer, et al. (2003). Filter binding assay for the geldanamycin-heat shock protein 90 interaction. Anal Biochem 317(1): 40-6; 2. Kim, J., S. Felts, et al. (2004). Development of a fluorescence polarization assay for the molecular chaperone HSP90. J Biomol Screen 9(5): 375-81; and 3. Zhou, V., S. Han, et al. (2004).
A time-resolved fluorescence resonance energy transfer-based HTS assay and a surface plasmon resonance-based binding assay for heat shock protein 90 inhibitors. Anal Biochem 331(2): 349-57.
EXEMPLIFICATION
The invention now being generally described, it will be more readily understood by reference to the following examples which are included merely for purposes of illustration
- 90TCX-003.88 ί£θ7θ4 of certain aspects and embodiments of the present invention, and are not intended to limit the invention.
EXAMPLE 1 - COMPUTATIONAL EXEMPLIFICATION [A] ACTIVE AND DECOY SETS. A total of forty known antiestrogens were selected from literature with activities ranging from nanomolar to low micromolar potency and converted to SMILES format using ACD/ChemSketch 8.17. The set was passed through FILTER to remove those antiestrogens that were not considered to be ‘drug-like’ leaving only 19 remaining. FILTER, distributed by Openeye Scientific Software. Our laboratory and others have highlighted the importance of incorporating a set of actives in a decoy set that reflect the properties of the rest of the decoy set when validating a VS protocol. Knox, A. J. S., Meegan M.J., Lloyd D.G, Estrogen Receptors: Molecular interactions, virtual screening and future prospects. Current Topics in Medicinal Chemistry 2006, 6, (3), 211-237; and Verdonk, M. L.; Berdini, V.; Hartshorn, M. J.; Mooij, W. T.; Murray, C. W.; Taylor, R. D.; Watson, P., Virtual screening using protein-ligand docking: avoiding artificial enrichment. J Chem Inf Comput Sci 2004,44, (3), 793-806. We sought to optimize the protocol towards discovery of inhibitors of ERa that would also possess more ‘drug-like’ properties and our choices of filter parameters reflected this. A subset of the Derwent World Drug Index (WDI) was then extracted and passed through FILTER using the same filtering properties, such as molecular weight < 200 or > 550, number of hydrogen bond donors 0 < x < 6 and acceptors 0 < x < 10, calculated logP < 7. The set remaining totaled 10343 compounds. From this, five hundred molecules with stereochemical information denoted and 481 without were randomly selected. This was done to best reflect the portion of marketed drugs that contain chiral centers, so representing a ‘real world’ virtual dataset. The two sets were merged with the 19 actives to produce a set of 1000 compounds with similar characteristics.
A larger database comprising 9,999 compounds was formed using the WDI and CHEMBANK. A single potent antiestrogen was added to the set to make up the 10,000. Meegan, M. J.; Hughes, R. B.; Lloyd, D. G.; Williams, D. C.; Zisterer, D. M., Flexible estrogen receptor modulators: design, synthesis, and antagonistic effects in human MCF-7 breast cancer cells. J Med Chem 2001,44, (7), 1072-84. This set has been previously described in our study of database pre-processing, Knox, A. J.; Meegan, M. J.; Carta, G.; Lloyd, D. G., Considerations in compound database preparation-hidden impact on virtual
-91 TCX-003.88 «070466 screening results. J Chem Inf Model 2005,45, (6), 1908-19. This set was employed to validate the vHTS protocol more thoroughly.
[B] CONFORMER GENERATION AND STORAGE. Cheminformatic preprocessing of databases of molecules has been assessed by our group in relation to ERa in a previous study. Knox, A. J.; Meegan, M. J.; Carta, G.; Lloyd, D. G., Considerations in compound database preparation-hidden impact on virtual screening results. J Chem Inf Model 2005,45, (6), 1908-19. We have demonstrated the impact it has in the context of virtual screening and prioritization of compounds for biological evaluation using the rigidexhaustive docking algorithm, FRED 2.01. FRED (version 2.0.1) was developed and is distributed by Openeye Scientific Software; URL:http://www.eyesopen.com. Multiple protonated, tautomeric, stereochemical and conformational states were enumerated and their associated effects on Enrichment (E) rates and False Positive (FP) rates were examined using datasets of 1000 and 10,000 compounds respectively. Unexpectedly, the initial SMILES representation of a compound prior to pre-processing had a significant impact on the Enrichment obtained. Weininger, D., SMILES: A Chemical Language and Information System J. Chem. Inf. Comput 1988,28, 31-36. It is concluded that only generation of 10 conformers of each compound using OMEGA 1.81 are needed to produce excellent enrichment when docking in the ERa. OMEGA 1.8.1 is distributed by Openeye Scientific Software Noteworthy, this is ER specific and might not translate to other targets. Interestingly, addition of multiple protonation, tautomeric and stereochemical states does not provide additional benefit. As a result, we have chosen the same method of conformer sampling using OMEGA 1.81 in the current protocol.
To begin with, OMEGA 1.81 was utilized to convert all databases from sdf format to a multi-conformer mol2 database, with subsequent conversion to multi-PDB file using OpenBabel 1.100.2. OpenBabel, http://openbabel.sourceforge.net/. A set of C subroutines automates these processes and also splitting of the multi-conformer file in to separate conformers. An open source database, MYSQL, is used as the core information storage system for the conformers for docking. MYSQL, URL:http://www.mysql.com. Fields containing conformer id, INPUT files, PROTEIN id, SMILES id, and job status ensure an elegant structure to the database in maintained.
[C] PROTEIN PREPARATION. The crystal structure 3ERT was downloaded from the Protein Data Bank and crystallographic waters were removed. The subsequent structure
-92TCX-003.88 *0 was imported to Macromodel 6.5 and re-written in PDB format to ensure bonds were represented correctly in this format. Macromodel v6.5, Schrodinger Inc.: Portland, OR 97201, http.//www.schrodinger.com/Products/macromodel. html. LIGIN does not take hydrogen atoms into account in the docking process and so no addition or minimization of them was needed, [D] DOCKING PROTOCOL. A description of the LIGIN docking program employed at this stage is provided. LIGIN is executed when three main fdes are present, INPUT, PROT and LIG. The LIG file consists of each conformation of a ligand in the database in standard PDB format. The PROT file is generated from the crystal structure (3ERT), and contains the coordinates of the protein atoms and other atoms in the target and but does not include information about the ligand chosen. Shiau, A. K.; Barstad, D.; Loria, P. M.; Cheng, L.; Kushner, P. J.; Agard, D. A.; Greene, G. L., The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen. Cell 1998, 95, (7), 927-37. The input file is then generated from a set of arbitrary rules that classify and assign a number to particular atom types numbered 1-8: (1) Hydrohilic: - N and 0 atoms that can donate and accept H-bonds (e.g., oxygen of hydroxyl group of Ser or Tyr); (2) Acceptor: N or 0 atoms that can only accept H-bond; (3) Donor:
N atom that can only donate H-bond; (4) Hydrophobic: Cl, Br, I and all C atoms that are not in aromatic rings and do not have a covalent bond to a hydrophilic atom; (5) Aromatic: C atoms in aromatic rings, (6) Neutral: C atoms that have a covalent bond to at least one atom of class 1, or two or more atoms from class 2 or 3; N atom if it has covalent bonds with 3 carbon atoms; S and F atoms in all cases, (7) Neutral-donor: C atom that has a covalent bond with only one atom of class 3; and (8) Neutral-acceptor: C atom that has covalent bond with only one atom of class 2.
In order to reduce the sampling time, the co-ordinates of the LIG files are translated to those of the co-crystallised ligand (4-Hydroxytamoxifen), to ensure docking begins in the binding site. LIGIN begins by generating a number of ligand positions in 6-dimensions in the binding site of the receptor. Each of the ligand positions has their respective binding modes assessed according to the complementary function as given in equation (1). After generating random positions of conformers within the binding site, the program then optimizes these positions with simplex optimixation methods and using NC as the scoring function. The docked positions obtained have their respective hydrogen bond lengths optimized to allow for refinement of the final structure. After searching for the global
......-...........- -.....-93- .........................
TCX-003.88 ΐΕ0 70 4 β6 maximum of the complementarity function, the programme creates less than or equal to 20 files (CR1, CR2, CR3...) containing the coordinates of the ligand in PDB format that correspond to the ‘global’ (CR1) and ‘local’ maxima (CR2, CR3...). Merging of the PROT file and each CR file is carried out to produce the final docked complexes. Each step in the process, namely, extraction of ligand information from an SQL database, generation of each INPUT file for the associated LIG files, translating the co-ordinates of the LIG files to the endogenous ligand, execution of LIGIN, and merging of the output CR files with PROT file are all carried out by a series of C routines that construct the fully automated suite.
[E] TS-VS VALIDATION: NC, DISTANCE CONSTRAINTS & SCORING. In a prescreening phase, the Normalized Complementarity (NC) according to LPC was calculated for each of the docked complexes for a set of 19 active ERa inhibitors known to potently modulate ERa. The lowest NC value was set as the threshold value for follow-up docking studies. Sobolev, V.; Sorokine, A.; Prilusky, J.; Abola, Ε. E.; Edelman, M., Automated analysis of interatomic contacts in proteins. Bioinformatics 1999, 15, (4), 327-32.
Next, a post-docking filter was introduced, consisting of distance thresholds set between atoms of a ligand and certain residues known to be important in the ER binding process. From a calculation of the interatomic contacts using LPC on the crystal structure 3ERT, the core putative H-bonds were deemed to be Glu353, Arg394, Leu387, Thr347, and Asp351. The 19 actives used earlier to assess the lowest NC value were re-docked and distance thresholds were set for each of these residues according to the range of distances observed for all 19 actives (eg. 2.4 <= $glu353 && $glu353 <= 4).
A decoy set of 1000 compounds seeded with the same 19 actives was subsequently docked according to the above procedure with the NC threshold set, but without any distance constraints set. The remaining docked complexes were scored using the following scoring functions: F-Score, D-Score, PMF-Score, G-Score, Chemscore, and Drugscore as implemented in Sybyl6.91, Chemscore, Chemgauss, Chemgauss2, Shapegauss, PLP, and Screenscore as implemented in FRED 2.11, Hammerhead as implemented in Surflex, and two standalone scoring functions, Xscore and Fresno. The optimal scoring function was selected through analysis of enrichment data and now became the final component of the procedure.
Finally, the two training sets (1000 & 10,000 molecules) described in the previous section were docked, filtered and scored accordingly.
- - 94 TCX-003.88 110 70 4 6 [F] VIRTUAL SCREENING - PROOF OF CONCEPT A virtual screen of the SPECS database screening collection (Release: Aug2005,202054 compounds in total) was carried out using our fully optimized protocol to initially suggest new scaffolds of inhibitors of ERa. Ten conformers of each molecule were generated using OMEGA 1.81 and docked and scored according to the protocol detailed in the TS-VS validation section. A visual inspection of the compounds that passed was undertaken and a number of scaffolds selected.
Taking the same docked complexes, the set was re-filtered using LPC with additional constraints imposed to guarantee H-bonding of ligand atoms occurred with residues Thr347, Glu353, Leu387 and Arg394. Asp351 was not selected as a H-bonding constraint to allow both agonist and antagonist cores to be discovered, as agonists can readily be converted to antagonists through addition of an antiestrogenic side-chain. The remaining compounds were finally scored with the last scoring component of the procedure,
EXAMPLE 2 - BIOCHEMICAL TESTING [A] RECEPTOR BINDING ASSAY. Competitive binding affinity experiments were carried out using purified baculovirus-expressed human ER-alpha (HrERa) and applying Fluoromone (ES2), a fluoresceinlabeled estrogen ligand. Both were contained in the Estrogen Receptor (ER) Competitor Assay Kits, Green obtained from Invitrogen Corporation. HrERa was stored at -80°C, and not subjected to any vortexing.
Upon binding of a fluorescent molecule to the HrERa and formation of the receptorES2 complex, it tumbles slowly resulting in a high polarization value. Polarization, measured in mP units, is directly related to the molecular volume of the tumbling molecule.
Thus, if a competing compound displaces ES2, it causes the molecule to tumble rapidly in solution and results in a low polarization value being obtained. This change in mP value allows determination of the binding affinity of the compound. The concentration of the competitor that results in a half-maximum shift in polarization is equivalent to the IC50 value, and a curve can be plotted using:
Y = mP|Oo% + (mP0% - mP100%)/l + 10((LogIC5°'x) *Hi,,slope) (Eqn. 2) where; Y = mP, X = Log performing, mP100% = 100% inhibition, and mP0% = 0% inhibition.
-95TCX-003.88 IE 0 ? 0 4 6 6
Method 1. HrERa was serially diluted from 400nM to 0.391 nM in screening buffer (40 mM Tris-HCl, pH 7.5; 50 mM KC1; 5% glycerol; 10% dimethylformamide; 0.02% sodium azide; 50 pg/ml bovine gamma globulin) to a final volume of 100 μΐ in borosilicate test tubes. ES2 was added to each tube at a concentration of InM and the tubes were mixed by shaking lightly. After incubation for 1 hour at room temperature, the FP assays were carried out using a Beacon 2000 fluorescence polarization instrument (PanVera Corporation) with 360 nm excitation filter and 530 nm emission filter. Fluorescence anisotropy was measured for each solution and the amount of ER that gives 80% of the maximal shift in mP was selected as the concentration to use for competitive binding studies.
Competing compounds were prepared at a standard concentration of 10 mM in DMSO. HrERa and ES2 were combined on ice (4°C) in a glass vial to produce the receptor-fluoromone complex. The vial was gently inverted 2-3 times, again ensuring no vortexing of the mixture. In duplicate, the compounds were serially diluted in ethanol to ensure the final concentration of DMSO and ethanol was below 1% in solution. 1 pL of each solution was diluted in 49 pL of buffer and added to 50 pL of the receptor-fluoromone complex in borosilicate test tubes. Following 45 min incubation, the samples underwent FP measurement. E2 was used as a negative control and 50 pL of the receptor-fluoromone complex in 50 pL buffer as the positive control.
Method 2. The recombinant ER and the fluorescent estrogen ligand were removed from the -80°C freezer and thawed on ice for one-hour prior to use. The assay was performed using a protocol described by the manufacturer. The fluorescent estrogen (2 nM) was added to the ER (30 nM for ERa and 20 nM for ERp), screening buffer (100 nM potassium phosphate (pH 7.4), 100 pg/ml BGG, 0.02 M NaN3) was added to make up a final volume that was dependent on the number of tubes used.
Test compound, 1 pL, in varying concentrations, was added to 49 pi screening buffer in borosilicate tubes (6 mm diameter). 50 pi of the fluorescent estrogen/ER complex was added to make up the total volume to 100 pL. A vehicle control contained 1 % ethanol (v/v). A negative control contained 50 pL of screening buffer and 50 pL of fluorescent estrogen/ER complex. This control was used to determine the polarization value when no competitor was present (theoretical maximum polarization). 1 pL of 1 mM estradiol (final concentration 10 pM) was used as complete displacement (minimum polarization value).
-96TCX-003.88
The tubes were incubated in the dark at room temperature for 2 hours and were mixed by shaking on a plate shaker. The polarization instrument contained 485 nM excitation and 530 nM emission interference filters.
[B] DISPLACEMENT BINDING ASSAY. Binding affinity (Ki value) for the estrogen receptor can be measured by the ability of a compound to displace tritium-labelled estradiol from the receptor site. A general procedure for determining the binding affinity of a compound to the estrogen receptor is provided below.
Estrogen receptor-rich cytosol is obtained from the uteri of humanely sacrificed Sprague-Dawley immature rats (100-150 g mass). Briefly, the uteri are homogenized in 0.01 M sodium phosphate buffer, pH 7.3, containing 0.15 M NaCI, 0.1% gelatin and 0.01% sodium azide. The homogenate is then centrifuged at 100,000 x g, 4 °C. The cytosol thus isolated is pretreated with dextran coated charcoal (DCC on ice) and re-isolated using centrifugation, before freezing at -20 °C for later use. Fishman, J.H. Stabilization of estradiol-receptor complexes by elimination of cytosolic factors. Biophys. Res. Commun. 1983, 110, 713-718. The protein concentration of cytosol samples is determined using a standard Bradford protein assay and an appropriate protein concentration range (150 pg protein in a total volume of 0.14 mL) for assay prepared. The required amounts of tritiumlabeled (hot) and non-labeled (cold) estradiol are calculated using standard saturation curve techniques. A fresh buffer solution is prepared (Tris[tris(hydroxymethyl)aminomethane]buffer - 10 mM, pH 7.4, containing 1.5 mM EDTA and 3 mM sodium azide). Displacement testing of the compounds is facilitated through the incubation of a buffered solution of a known concentration of the test compound with hot estradiol solution (specific activity 157 Ci/mmol - final cone. 5 nM / tube), followed by the addition of estrogen receptor-rich cytosol (150 pg protein). Total and non-specific binding control assays are determined in the absence and presence of 14 pL of a 0.2 mM cold estradiol solution respectively, properly corrected for the presence of ethanol in the test (displacement) samples. Samples are vortexed to ensure homogeneity and refrigerated at 4°C for 16-20 h. After this time the samples were retreated with DCC on ice and centrifuged for ten min at 3500 x g. A 170 pL sample is pipetted from each vial and diluted with 10 mL scintillation fluid (Ecoscint). A scintillation control containing 28 pL of 5 nM hot estradiol in 10 mL scintillation fluid is also prepared to facilitate theoretical activity calculations. The samples thus prepared are counted for radioactivity by liquid scintillation counting. Binding values are obtained as counts per minute (cpm) and were converted to ’ ..............'............................ ........-97-........................‘..................-.— ·
TCX-003.88 «070466 disintegrations per minute (dpm) and computationally analyzed using sigmoidal curve fitting programs EBDA and LIGAND to fit the displacement curves and to calculate binding affinity values (Ki) for the test compounds. Munson, P.J.; Rodbard, D. LIGAND: a versatile computerised approach for the characterisation of ligand binding systems. Anal
Biochem. 1980,107,220-239.
EXAMPLE 3 - ANTIPROLIFERATIVE STUDIES
The following tests were carried out to determine the activity of the compounds of the invention. The compounds have potent anti-proliferative effects with desirable cytotoxicity profiles in human breast tumour MCF-7 cell lines in vitro. Such compounds therefore have potential in estrogen related diseases and as antiosteoporotic compounds.
[A] MIT ASSAY. MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] is a yellow tetrazolium salt which is taken up only by metabolically active cells and subsequently cleaved by mitochondrial dehydrogenases to yield a purple crystalline formazan dye. On solubilisation this purple colour may be read spectrophotometrically. The absorbance measured at this wavelength is directly proportional to the amount of viable cells present. The MTT assay was carried out as previously described. Mosmann, T., Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 1983, 65, (1-2), 55-63. The ER(+) breast cancer MCF-7 cell line was maintained in 75-cm2 culture flasks (Greiner) containing (Dulbecco) Eagles minimum essential medium in a 5% CO2 atmosphere with 10% fetal calf serum. The medium was also supplemented with 1% nonessential amino acids.
Method 1. Cells were trypsinized and seeded at a density of 1.5 χ 104 in a 96-well plate and incubated at 37 °C, 5% CO2 atmosphere for 24 h. All compounds were prepared at a standard concentration of 10 mM in DMSO and serially diluted to produce a range of concentrations spanning 1 ηΜ to 100 μΜ. 2 μΕ of each compound solution were added to the cells and reincubated for an additional 72 hours. Control wells contained 2 pL of vehicle (DMSO) in all cases. At the end of the incubation period, culture medium was removed and all cells were washed with 100 pL PBS. 50 pL of MTT solution was added to each well and the plates were incubated in darkness for about 2 hours at 37 °C. The converted dye was solubilised with 200 pL DMSO and pipetted up and down several times to ensure the dye dissolves completely. Absorbance of the converted dye was measured at 570 nm with control cells set to 100% cell viability.
. — —' ...................-98- #0 7(,4^
Method 2. MCF-7 cells were pelleted, counted and complete medium was added to give a density of 2.5 x 104 cells/mL. The cells were placed in 96-well plates at 200 pL per well. The outside wells were not used. Two compounds could be tested per plate over seven concentrations and with a control, vehicle and blank for each compound. The cells were left at 37°C under a humidified atmosphere of 95 % O2, 5 % CO2 for 24 hours. A stock solution of the compounds was made up in ethanol and stored at -20°C until further use. The cells were treated with a 2 pL of vehicle (1 % (v/v) ethanol) or a range of concentrations of the synthesised compounds 2 pL (final concentration 1 nM - 50 pM) and again placed in the incubator for 72 hours. Following the incubation period, 20 pL of lysis solution was added to the ‘blank’ wells and left for 1 hour to ensure 100 % cytotoxicity, 50 pL was then removed from each well and transferred into a new 96-well plate for use in the LDH assay.
The MTT plate was inverted to remove all medium from the wells. The wells were washed with phosphate buffered saline (PBS) solution to remove remaining medium and inverted again to remove all PBS solution. The plates were dried and 50 pi of MTT solution (1 mg/ml) was added to each well. The cells were incubated in the dark for 2-3 hours at 37°C. Purple crystals were formed and dissolved using 200 pi of dimethyl sulfoxife (DMSO). The absorbance was read at a wavelength of 595 nm in a Dynatech MR5000 plate reader and cell viability expressed as a percent of control. IC50 values were calculated for each compound.
[B] CYTOTOXICITYSTUDIES/LDHASSAY. Lactate dehydrogenase (LDH) is a cytosolic enzyme released upon cell lysis (death). Through the use of a commercial LDH assay kit, released LDH converts a substrate tetrazolium salt into a soluble red formazan product. The absorbance of this dye can be measured directly at 490 nm, the colour formed is proportional to the number of lysed cells, and as such the extent of cytotoxic activity for the compound added may be assessed. The lactate dehydrogenase (LDH) assay was used to measure cellular toxicity effects of the various doses of each compound and was examined using a colorimetric determination kit (Promega). CytoTox 96® Non-Radioactive Assay, URL: http://www.promega.com/; Allen, M. J.; Rushton, N. Promega Notes 1994,45, 7-10; and Nachlas, Μ. M.; Margulies, S. I.; Goldberg, J. D.; Seligman, A. M. Analytical Biochemistry 1960,1,317-326.
- 99 TCX-003.88 ic ν / IM β g
Method 1. The assay was carried out concurrently with the MTT assay (Method 1) following dosing of the compounds and incubation for 72 hrs as above. Prior to removal of the culture medium in the MTT assay, 50 pL aliquots of medium were removed to a fresh 96-well plate. 50 pL of LDH solution was added to each well and the plate was left in darkness for about 20 to 30 minutes at room temperature. 50 pL stop solution was then added to each well and the absorbance read at 490 nm on a micro-plate reader. Control of 100% lysis was measured by addition of 20 pL lysing solution 45 minutes prior to harvesting.
Method 2. MCF-7 cells were seeded in 96-well plates, incubated for 24 hours and then treated with compounds. After 72 hours 20 pL of lysis solution (10X) was added to the ‘blank’ wells, they were then left for 1 hour to ensure 100 % death, 50 pL was removed from each well and transferred into a new 96-well plate for use in the LDH assay. 50 pL of substrate mix from the LDH assay kit was added and the plate was placed in the dark at room temperature for 30 minutes. After this period, 50 pL of stop solution was added to each well before reading the absorbance at a wavelength of 490 nm using a Dynatech MR5000 plate reader. Percentage death was calculated at 10 pM.
EXAMPLE 4 - ASSAY FOR DETERMINING APOPTOTIC INDUCTION
A general procedure for Fluorescence Activated Cell Sorting is provided below.
Following treatment of MCF-7 cells with apoptotic compounds, the cells are washed three times with PBS before being trypsinised and centrifuged at 300 g for 5 min. They are then resuspended in 200 pL PBS, made up to 2 mL with ice-cold ethanol (70% v/v) and left to sit on ice for at least 1 h to fix them. Approximately 1 h prior to use they are centrifuged at 300 g for 3 min and the supernatant carefully pipeted off. The pellet is resuspended in 800 pL PBS, RNAase (100 pL; 1 mg/mL) and 100 pL of the fluorescent dye propidium iodide (PI; 400 pg/mL) which binds DNA is added. The tubes are vortexed and are incubated at 37°C for 30 min. Flow cytometry is performed with a FACS calibur flow cytometer from Becton Dickinson. FACS data is analysed using the programme Cell Quest.
In addition, the ability of the compounds to stimulate uterine cell growth may be assessed by an alkaline phosphatase assay in human endometrial Ishikawa and in SaOs-2 osteoblast cells respectively, as described previously. Littlefield, B.A., Gurpide, E., Markiewicz, L., McKinley, B. and Hochberg, R.B. A simple and sensitive microtiter plate : ' ....... ...............—......-.........-.................-........ !00 - ............. -.....
TCX-003.88 «Ο 70 4 66 estrogen bioassay based on stimulation of alkaline phosphatase in Ishikawa cells: estrogenic action ofadrenal steroids. Endocrinol. 1990 127,2757-2762. It will be appreciated that the compounds may have useful pharmacological properties other than those described above.
EXAMPLE 5 - SYNTHESIS OF COMPOUNDS OF FORMULA I
The compounds of formula I can be synthesized in a variety of ways. One approach to the synthesis is a two step method, as shown in Figure 14.
[A] STEP ONE. The appropriate phenol (1 equiv., 10 mmol) is dissolved in 100 ml of dry acetone and placed in a round-bottom flask. Anhydrous potassium carbonate (0.16 mol, 22 g) [16 equiv] is added. They are stirred gently for 10 minutes under a N2 atmosphere and the corresponding basic side chain is then added (40 mmol, 5.78 g) [4 equiv]. The reaction is refluxed until complete on thin layer chromatography. On completion the solution is filtered and the solvent is removed under reduced pressure to afford product (a protected aldehyde) used without further purification.
For example, 4-(2-pyrrolidin-l-yl-ethoxy)benzaldehyde (8) was prepared from 415 hydroxybenzaldehyde using the general procedure outlined above and was obtained as an orange gel (yield 65 %). IR: NaCl υ: 1691.70 cm'1 (CHO), (absence of broad peak at 3369.22 cm'1 indicates loss of OH group) 'H NMR (400 MHz, CDClj): δ 1.72 (m, 4H, C(CH2)2-C), δ 2.55 (m, 4H, CH2-N-CH2), δ 2.86-2.89 (t, 2H, J=6.02 Hz (av.), CH2-N), δ 4.12-4.15 (t, 2H, J=6.02 Hz, CH2-N), δ 6.96-6.98 (d, 2H, J=9.04 Hz, Ar-H (3’,5’), δ 7.7620 7.78 (d, 2H, >9.04 Hz, Ar-H (2’,6’)), δ 9.82 (s, 1H, CHO).
[B] STEP TWO. The protected aldehyde (1 equiv., 10 mmol) is placed in round bottom flask. Ethyl cyanoacetate (10 mmol) and resorcinol (20 mmol) are added. Ethanol (30 mL) is added and reaction is refluxed. 0.5 mL of piperidine is then added dropwise and the reaction is refluxed for 2 hours. Solvent is removed under reduced pressure.
For example, 2-amino-7-hydroxy-4-[4-(2-pyrrolidin-l-yl-ethoxy)-phenyl]-4Hchromeme-3-carboxylic acid ethyl ester (9) was prepared from 4-(2-pyrrolidin-l-ylethoxy)-benzaldehyde (8; 10 mmol, 1.97 g) using the general procedure outlined above and was obtained as a brown gel. Column chromatography was carried out using DCM:methanol as the solvent system (10 %) and the compound was obtained as yellow crystals (yield 10 %). IR: KBr υ: 3416.27 cm'1,3304.14 cm'1 (NH2), 1674.57 cm'1 (C=O). JH NMR (400 MHz, CDC13): δ 1.18-1.22 (t, 3H, C-CH3), δ 1.78 (s, 4H, C-(CH2)2-C), δ 2.13 (s, 4H, CH2-N-CH2), δ 3.61-3.63 (t, 2H, J=4.28 Hz (av.), CH2-N), δ 4.07-4.08 (q, 2H,
TCX-003.88 >7.04 Hz, O-CH2), δ 4.28-4.30 (t, 2H, >4.52 Hz, CH2-N), δ 4.85 (s, 1H, CH, H4), δ 6.566.59 (q, 1H, >2.24 Hz (av.), Ar-H (H6)), δ 6.60-6.61 (d, 1H, >2 Hz, Ar-H (H8)), δ 6.906.92 (d, 2H, >8.56 Hz, Ar-H (3’,5’)), δ 6.94-6.96 (d, 1H, Ar-H (H5)), δ 7.17-7.19 (d, 2H, >8.52 Hz, Ar-H (2’,6’)) 13C NMR (100 MHz, CDCI3): δ 13.08 (CH3), δ 22.14 ((CH2)2-), δ
38.40 (CH, C4), δ 53.32 (N-CH2), δ 53.84 (CH2-N-CH2), δ 58.53 (O-CH2), δ 62.56 (C-CH20), δ 77.09 (C-C, C3), δ 101.73 (Ar-CH (C8), δ 111.44 (Ar-CH (C6)), δ 113.62 (Ar-C-H (3’,5’), δ 116.96 (Ar-C-C (C4-C-C5)), δ 127.76 (Ar-CH (2’,6’)), δ 129.22 (Ar-CH (C5)), δ 131.38 (Ar-C-C (1’)), δ 148.92 (Ar-C-C-O), δ 155.48 (Ar-C-0 (4’)), δ 156.12 (Ar-C-OH, C7), δ 169.20 (Ar-C-NH2 (C2)). EIMS (HR): C24H28N2O5, Calculated M+ + 1: 425.2076,
Observed M++ 1: 425.2081, Error: + 1.1 ppm
INCORPORATION BY REFERENCE
All of the U.S. patents and U.S. published patent applications cited herein are hereby incorporated by reference.
EQUIVALENTS
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
Claims (10)
1. We claim: wherein, independently for each occurrence, m is 0,1, 2, 3, or 4; n is 0, 1,2, or 3; R is alkyl, polycyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl; R 1 is -H, -R, or -C(=O)R; or the two R 1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; R 2 is -C^N, -C(=O)OH, -C(=O)OR, -CH 2 OH, or -CH 2 OR; R 3 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -1, or (CH 2 )b-A; A is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, or -I; b is 0, 1, 2, 3, 4, or 5; TCX-003.88 R 4 is -OCH2ON(R 7 )2, -OCH 2 CH 2 N(R 7 )2, -CH2CH2ON(R 7 )2, -CH 2 CH 2 CH 2 N(R 7 ) 2 , -N(R)CH 2 ON(R 7 )2, -N(H)CH2ON(R 7 )2, -N(H)CH 2 CH 2 N(R 7 ) 2 , or -N(R)CH 2 CH 2 N(R 7 ) 2 ; R 5 is -OR 1 , or -NCR'h; R 6 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -I, or (CH 2 ) b -A; and R 7 is -H, -R, or -C(=O)R, or the two R 7 taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical.
2. The compound of claim 1, wherein m is 1.
3. The compound of claim 1, wherein m is 0.
4. The compound of any of claims 1 to 3, wherein n is 1. 5. -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -1, or (CH 2 ) b -A; and R 7 is -H, -R, or -C(=O)R ; or the two R 7 taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic 5 172. The compound of any of claims 132 to 167, wherein p is 9. 173. The compound of any of claims 132 to 167, wherein p is 10. 174. The compound of any of claims 132 to 173, wherein R 9 is -H. 175. The compound of any of claims 132 to 173, wherein R 9 is -OH. 176. The compound of any of claims 132 to 173, wherein R 9 is -OR; R is alkyl. 10 177. The compound of claim 176, wherein R 9 is -OCH 3 , -OCH2CH3, -OCH2CH2CH3, -OCH(CH 3 )CH 3 , -OCH2CH2CH2CH3, -OCH(CH3)CH 2 CH 3 , -OCH 2 CH(CH 3 )CH 3 , -OCH 2 C 6 H 5 , or -OC(=O)CH 3 . 178. The compound of any of claims 132 to 177, wherein R 10 is alkyl. 179. The compound of any of claims 132 to 177, wherein R 10 is fluoroalkyl. 15 180. The compound of any of claims 132 to 177, wherein R 10 is perfluoroalkyl. 181. A pharmaceutical composition comprising a compound according to any one of claims 1-180 and a pharmaceutically acceptable carrier. 182. A pharmaceutical composition comprising a compound as claimed in any one of claims 1-180 in combination with a pharmaceutically active compound. 20 183. The pharmaceutical composition of claim 182, wherein the pharmaceutically active compound is an anti-cancer drug. 184. The pharmaceutical composition of claim 183, wherein the anti-cancer drug is cisplatin. 185. A method of eliciting an estrogen receptor modulating effect in a mammal in need 25 thereof, comprising administering to the mammal a therapeutically effective amount of a compound according to any one of claims 1-180, or a pharmaceutical composition according to any one of claims 181-184. TCX-003.88 ,Ε 0 7 0 4 6 g 186. The method according to claim 185, wherein the estrogen receptor modulation effect is an estrogen receptor agonizing effect. 187. The method according to claim 185, wherein the estrogen receptor agonizing effect is an ERa receptor agonizing effect. 188. The method according to claim 185, wherein the estrogen receptor modulation effect is an estrogen receptor antagonizing effect. 189. The method according to claim 185, wherein the estrogen receptor antagonizing effect is an ERa receptor antagonizing effect. 190. A method of treating a disease in a mammal in need thereof by administering to the mammal a therapeutically effective amount of a compound or pharmaceutical composition according to any one of claims 1-184, wherein said disease is selected from the group consisting of bone loss, bone fractures, cartilage degeneration, uterine fibroid disease, hot flashes, increased levels of LDL cholesterol, impairment of cognitive functioning, cerebral degenerative disorders, restenosis, gynecomastia, vascular smooth muscle cell proliferation, obesity, incontinence, obesity, hormone dependent breast cancer, osteoporosis, estrogen deficiency, arthritis, cardiovascular disease, ovarian cancer, artherosclerosis, colon tumor, endometriosis, Alzheimer’s disease, non-insulin dependent (type II) diabetes, infertility, prostrate tumor, melanoma, acne, hypercholesterolemia, CNS disease, contraception, conditions related to hair follicles, macular degeneration, urinary incontinence, estrogen receptor-expressing and estrogen receptor-expressing tumors, and leukaemia. 191. The method of claim 190, wherein the disease is hormone dependent breast cancer. 192. The method of claim 191, wherein the disease is estrogen dependent breast cancer. 193. The method of claim 192, wherein the disease is tamoxifene-resistant breast cancer. 194. A method of modulating the activity of Hsp90 in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions. 195. A method of modulating tublin polymerization/depolymerization in a mammal in need thereof, comprising administering to the mammal a therapeutically effective TCX-003.88 ΙΕο 70 4 66 amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions. 196. A method of modulating the activity of protein kinase C in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions. 197. A method of modulating the activity of calmodulin-dependent enzymes in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions. 198. A method of modulating the activity of acyl coenzyme A in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions. 199. A method of modulating the activity of cholesterol acyl transferase in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of any one of the aforementioned compounds or the aforementioned pharmaceutical compositions. 200. A compound of formula V r5 V wherein, independently for each occurrence, m is 0, 1, 2, 3, or 4; n is 0,1, 2, or 3; R is alkyl, polycyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl; TCX-003.88 «0 70 4 R 1 is -H, -R, or -C(»)R; or the two R 1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; R 2 is -C=N, -C(=O)OH, -C(=O)OR, -CH 2 OH, or -CH 2 OR; R 3 is -R, -C(=0)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3s -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -I, or (CH 2 ) b -A; A is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -ON, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, or -I; b is 0,1,2, 3,4, or 5; R 4 is -OCH2ON(R 7 )2, -OCH 2 CH 2 N(R 7 )2, -CH2CH2ON(R 7 )2, -CH 2 CH 2 CH 2 N(R 7 ) 2 , -N(R)CH 2 ON(R 7 )2, -N(H)CH2ON(R 7 )2, -N(H)CH 2 CH 2 N(R 7 ) 2 , or -N(R)CH 2 CH 2 N(R 7 )2,-OR’, or -NiR'h, -H, -OH, or -OR or -(CH2)PSR 10 , -(CH2) P S(=O)R 10 , -(CH2)pS(=O)2R 10 , -(CH2) p C(-O)N(R')R 10 , -(CH2)pN(R')CH2CH2CH2SR 1() , -(CH2) p N(R 1 )CH2CH2CH2S(=O)R 10 , or -(CH2) p N(R')CH 2 CH 2 CH 2 S(=O) 2 R 10 ’ where p is 5,6, 7, 8,9, or 10 and R 10 is alkyl, fluoroalkyl or perfluoroalkyl; R 5 is -OR 1 , or -N(R’)2, -OCH2ON(R 7 )2, -OCH 2 CH 2 N(R 7 )2, -CH2CH2ON(R 7 )2, -CH 2 CH 2 CH 2 N(R 7 ) 2 , -N(R)CH 2 ON(R 7 )2, -N(H)CH2ON(R 7 )2, -N(H)CH 2 CH 2 N(R 7 ) 2 , or -N(R)CH 2 CH 2 N(R 7 ) 2 , -H, -OH, or -OR; or -(CH 2 ) P SR 10 , -(CH 2 ) p S(=O)R 10 , -(CH2)pS(=O)2R 10 , -(CH2) p C(=O)N(R')R 10 , -(CH 2 ) p N(R’)CH 2 CH 2 CH 2 SR 10 , -(CIOpNiR’lCOCHzCHjStoOiR 10 , or -(CH2)pN(R 1 )CH2CH 2 CH 2 S(^O) 2 R 10 where R 10 is alkyl, fluoroalkyl or perfluoroalkyl and p is 5, 6, 7, 8, 9, or 10; TCX-003.88 IE 0 7 0 4 g g R 6 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2j -CF 3 , -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , 5 -N(H)CH 2 CH 2 N(R 7 ) 2 . 157. The compound of claims 132 to 156, wherein R 6 is -H, -R, -OR, -Cl, -F, -Br, -NO 2 , -NH 2 , -NHR, or -NR 2 ; and R is lower alkyl. 158. The compound of claim 157, wherein R 6 is -H, -OCH 3 , -Cl, -F, -Br, -NO 2 , -NH 2 , or -CH 3 . 10 159. The compound of any of claims 132 to 158, wherein R 8 is -(CH 2 ) P SR 10 , -(CH 2 ) p SfyO)R 10 , or -(CH 2 ) P S(=O) 2 R I(> . 160. The compound of claim 159, wherein R 8 is -(CH2)pSR 10 , -(CH2) P S(=O)R 1() , or -(CH 2 ) p S(=O) 2 R 10 ; and R 10 is fluoroalkyl. 161. The compound of claim 160, wherein R 8 is -(CH 2 ) p SCH 2 CH 2 CH 2 CF 2 CF 3 , 15 -(CH 2 ) p S(=O)CH 2 CH 2 CH 2 CF 2 CF 3 , -(CH 2 ) p S(=O) 2 CH 2 CH 2 CH 2 CF 2 CF 3 162. The compound of any of claims 132 to 158, wherein R 8 is -(CH 2 ) P C(=O)N(R I )R 10 . 163. The compound of claim 162, wherein R 8 is -(CH 2 ) P C(=O)N(R 1 )R 10 ; and R 10 is alkyl. 164. The compound of any of claims 132 to 158, wherein R 8 is -(CH 2 ) P C(=O)N(CH 3 . )CH 2 CH 2 CH 2 CH 3 . 20 165. The compound of any of claims 132 to 158, wherein R 8 is -(CH2)pN(R')CH2CH2CH2SR ,() , -(CH2)pN(R 1 )CH2CH2CH2S(=O)R IQ , or -(CH2) p N(R’)CH 2 CH 2 CH 2 S(=O) 2 R 10 . 166. The compound of claim 165, wherein R 8 is -(CH2)pN(R 1 )CH2CH 2 CH 2 SR ll) , -(CH2)pN(R 1 )CH2CH 2 CH 2 Sfy ())R 10 , or -(CH2)PN(R 1 )CH2CH 2 CH 2 S(=O)2R 10 ; and 25 R 10 is alkyl. 167. The compound of claim 166, wherein R is -(CH 2 ) P N(CH 3 )CH 2 CH 2 CH 2 SCH 2 CH 2 CH 2 CH 2 CH 3 , -(CH 2 ) p N(CH 3 )CH 2 CH 2 CH 2 S(=O)CH 2 CH 2 CH 2 CH 2 CH 3 ,or -(CH 2 ) P N(CH 3 )CH 2 CH 2 CH 2 S(=O) 2 CH 2 CH 2 CH 2 CH 2 CH 3 . TCX-003.88 IE 0 7 O 4 168. The compound of any of claims 132 to 167, wherein p is 5. 169. The compound of any of claims 132 to 167, wherein p is 6. 170. The compound of any of claims 132 to 167, wherein p is 7. 171. The compound of any of claims 132 to 167, wherein p is 8. 5 125. 126. 127. 128. Ο 129. 130. 131. 132. The compound of any of claims 83 to 118, wherein p is 7. The compound of any of claims 83 to 118, wherein p is 8. The compound of any of claims 83 to 118, wherein p is 9. The compound of any of claims 83 to 118, wherein p is 10. The compound of any of claims 83 to 124, wherein R 9 is -H. The compound of any of claims 83 to 124, wherein R 9 is -OH. The compound of any of claims 83 to 124, wherein R 9 is -OR; R is alkyl. The compound of claim 127, wherein R 9 is -OCH 3 , -OCH 2 CH 3 , -OCH 2 CH 2 CH 3 , -OCH(CH 3 )CH 3 , -OCH 2 CH 2 CH 2 CH 3 , -OCH(CH 3 )CH 2 CH 3 , -OCH 2 CH(CH 3 )CH 3 , -OCH 2 C 6 H 5 , or -OC(=O)CH 3 . The compound of any of claim 83 to 128, wherein R 10 is alkyl. The compound of any of claim 83 to 128, wherein R 10 is fluoroalkyl. The compound of any of claim 83 to 128, wherein R 10 is perfluoroalkyl. A compound of formula IV: wherein, independently for each occurrence, m is 0, 1,2, 3, or 4; n is 0, 1,2, or 3; p is 5, 6, 7, 8, 9, or 10 R is alkyl, polycyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl; R 1 is -H, -R, or -C(=O)R; or the two R 1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; R 2 is -C=N, -C(=O)OH, -C(=O)OR, -CH 2 OH, or -CH 2 OR; TCX-003.88 133. 134. 135. 136. /Εθ 70 4 66 R 3 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2j -OC(=O)NHR, -OC(=O)NR 2 , -NH 2j -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -1, or (CH 2 ) b -A; A is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, or -I; b is 0, 1, 2, 3, 4, or 5; R 6 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -C^N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -I, or (CH 2 ) b -A; R 8 is -(CH2)pSR 10 , -(CH2) p S(=O)R 10 , -(CH2)pS(=O)2R 10 , -(CH2)pC(=O)N(R')R 1() , -(CIOpNfR'iCHzClhCHzSR 10 , -(CH2)pN(R')CH2CH2CH2S(=O)R 1(> , or -(CH2) p N(R')CH 2 CH 2 CH 2 S(=O) 2 R 11) ; R 9 is -H, -OH, or -OR; and R 10 is alkyl, fluoroalkyl or perfluoroalkyl. The compound of claim 132, wherein m is 1. The compound of claim 132, wherein m is 0. The compound of any of claims 132 to 134, wherein n is 1. The compound of any of claims 132 to 134, wherein n is 0. TCX-003.88 IEO 70 4 66 137. The compound of any of claims 132 to 136, wherein R is alkyl or aralkyl. 138. The compound of claim 137, wherein R is lower alkyl. 139. The compound of claim 138, wherein R is -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , or -CH 2 C 6 H 5 . 140. The compound of any of claims 132 to 139, wherein R 1 is -H, alkyl, or aralkyl. 141. The compound of claim 140, wherein R 1 is -H, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , or -CH 2 C 6 H 5 . 142. The compound of any of claims 132 to 139, wherein R 1 is -C(=O)R. 143. The compound of claim 142, wherein R 1 is -C(=O)R; and R is alkyl. 144. The compound of any of claims 132 to 139, wherein the two R 1 taken together with the nitrogen to which they are bound represent -N^C/RjOR. 145. The compound of claim 144, wherein the two R 1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; and R is alkyl or aralkyl. 146. The compound of any of claims 132 to 145, wherein R 2 is -C=N, -C(=O)OH, -C(=O)OR, -CH 2 OH, or -CH 2 OR; and R is lower alkyl. 147. The compound of claim 146, wherein R 2 is -C=N, -C(=O)OH, -C(=O)OCH 3 , -C(=O)OCH 2 CH 3 , or -CH 2 OH. 148. The compound of any of claims 132 to 147, wherein R 3 is -H, -R, -OR, -Cl, -F, -Br, -NO 2 , -NH 2j -NHR, or -NR 2 ; and R is lower alkyl. 149. The compound of claim 148, wherein R 3 is -H, -OCH 3 , -Cl, -F, -Br, -NO 2 , -NH 2 , or -CH 3 . 150. The compound of any of claims 132 to 149, wherein R 4 is -OCH2ON(R 7 )2, -OCH 2 CH 2 N(R 7 ) 2 . 151. The compound of claim 132, wherein R 4 is -OCH2ON(R 7 )2 152. The compound of claim 132, wherein R 4 is -OCH2CH2N(R 7 )2. 153. The compound of any of claims 132 to 149, wherein R 4 is -CH2CH2ON(R 7 )2, or -CH 2 CH 2 CH 2 N(R 7 ) 2 , TCX-003.88 ΙΕΟ 70 4 66 154. The compound of claim 153, wherein R 4 is -CH2CH2CH2N(R 7 )2, 155. The compound of any of claims 132 to 149, wherein R 4 is -N(R)CH 2 ON(R 7 )2, -N(H)CH2ON(R 7 )2, -N(R)CH 2 CH 2 N(R 7 ) 2 , or -N(H)CH 2 CH 2 N(R 7 ) 2 . 156. The compound of claim 155, wherein R 4 is -N(R)CH2CH2N(R 7 )2, or
5. The compound of any of claims 1 to 3, wherein n is 0,
6. The compound of any of claims 1 to 5, wherein R is alkyl or aralkyl.
7. The compound of claim 6, wherein R is lower alkyl.
8. The compound of claim 7, wherein R is -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , or -CH 2 C 6 H 5 .
9. The compound of any of claims 1 to 8, wherein R 1 is -H, alkyl, or aralkyl. 10. The compound of claim 9, wherein R 1 is -H, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 2 CH 3 , -CH 2 CH(CH 3 )CH 2 CH 3 , or -CH 2 C 6 H 5 . 11. The compound of any of claims 1 to 8, wherein R 1 is -C(=O)R. 12. The compound of claim 11, wherein R 1 is -C(=O)R; and R is alkyl. ΙΕο 70 4 6 6 TCX-003.88 13. The compound of any of claims 1 to 8, wherein the two R 1 taken together with the nitrogen to which they are bound represent -N=C(R)OR. 14. The compound of claim 13, wherein the two R 1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; and R is alkyl or aralkyl. 15. The compound of any preceding claim, wherein R is -C=N, -C(=O)OH, -C(=O)()R, -CH2OH, or -CH 2 OR; and R is lower alkyl. 16. The compound of claim 15, wherein R 2 is -C^N, -C(=O)OH, -C(=O)OCH 3 , -C(=O)OCH 2 CH 3 , or -CH 2 OH. 17. The compound of any preceding claim, wherein R is -H, -R, -OR, -Cl, -F, -Br, -NO 2 , -NH 2 , -NHR, or -NR 2 ; and R is lower alkyl. 18. The compound of claim 17, wherein R 3 is -H, -OCH 3 , -Cl, -F, -Br, -NO 2 , -NH 2 , or -CH 3 . 19. The compound of any preceding claim, wherein p is 1. 20. The compound of any preceding claim, wherein R 4 is -OCH2ON(R 7 )2, -OCH 2 CH 2 N(R 7 ) 2 . 21. The compound of claim 20, wherein R 4 is -OCH2ON(R 7 )2,. 22. The compound of claim 20, wherein R 4 is -OCH2CH2N(R 7 )2. 23. The compound of any of claims 1 to 19, wherein R 4 is -CH2CH2ON(R 7 )2, or -CH 2 CH 2 CH 2 N(R 7 ) 2 , 24. The compound of claim 23, wherein R 4 is -CH2CH2CH2N(R 7 )2, 25. The compound of any of claims 1 to 19, wherein R 4 is -N(R)CH2ON(R 7 )2, -N(H)CH 2 ON(R 7 )2, -N(R)CH2CH2N(R 7 )2, or -N(H)CH 2 CH 2 N(R 7 ) 2 . 26. The compound of claim 25, wherein R 4 is -N(R)CH2CH2N(R 7 )2, or -N(H)CH 2 CH 2 N(R 7 ) 2 . 27. The compound of any preceding claim, wherein R 5 is -OR 1 . 28. The compound of claim 27, wherein R 5 is -OR 1 ; and R 1 is -H, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 2 CH 3 , -CH 2 CH(CH 3 )CH 2 CH 3 , -CH 2 C 6 H 5 , or -C(=O)CH 3 . 29. The compound of claim 28, wherein R 5 is -OH, -OCH3, -OCH 2 CH 3 , or -C(=O)CH 3 . TCX-003.88 30. The compound of any of claims 1 to 26, wherein R 5 is -NCR 1 ^. 31. The compound of claim 30, wherein R 5 is -NiR 1 ^; and R 1 is -H, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , -CH 2 C 6 H 5 , or -C(=O)CH 3 . 32. The compound of claim 31, wherein R 5 is -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -NH(CH 2 C 6 H 5 ), -N(CH 3 )(CH 2 C 6 H 5 ), -N(H)C(=O)CH 3 , or -N(CH 3 )C(=O)CH 3 . 33. The compound of any preceding claim, wherein R 6 is -H, -R, -OR, -Cl, -F, -Br, -NO 2 , -NH 2s -NHR, or -NR 2 ; and R is lower alkyl. 34. The compound of claim 33, wherein R 6 is -H, -OCH 3 , -Cl, -F, -Br, -NO 2 , -NH 2 , or -CH 3 . 35. The compound of any preceding claim, wherein R is -H, -R, or -C(=O)R; and R is alkyl or aralkyl. 36. The compound of claim 35, wherein R 7 is -H, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , -CH 2 C 6 H 5 , or -C(=O)CH 3 . γ 37. The compound of any of claims 1 to 34, wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical. 38. The compound of claim 37, wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; and the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, piperidine, piperazine, morpholine, or diazadecaline (1,2,3,4,5,6,7,8-octahydroquinoxaline). 39. The compound of claim 37 or claim 38, wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, piperidine, piperazine, morpholine, or diazadecaline (1,2,3,4,5,6,7,8-octahydroquinoxaline); and said optionally substituted monocyclic or bicyclic heterocycle radical is substituted with one or more substituents selected from the group consisting of -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, - TCX-003.88 0 4 βθ CHF 2 , -CF 3 , -C^N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -I, and -(CH 2 ) b -A. 40. The compound of any of claims 1 to 34, wherein the two R 7 taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; and the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, or piperidine. 41. The compound of claim 40, wherein the two R 7 taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, piperidine, or piperazine; and said optionally substituted monocyclic or bicyclic heterocycle radical is substituted with one or more substituents selected from the group consisting of-R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -ch 2 f, CHF 2 , -CF 3 , -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -0C(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -I, and -(CH 2 ) b -A. 42. A compound of formula II: wherein, independently for each occurrence, m is 0, 1,2, 3, or 4; n is 0, 1,2, or 3; TCX-003.88 «0 70 4 R is alkyl, polycyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl; R 1 is -H, -R, or -C(=O)R; or the two R 1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; R 2 is -C^N, -C(=O)OH, -C(=O)OR, -CH 2 OH, or -CH 2 OR; R 3 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -I, or (CH 2 ) b -A; A is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, or -I; b is 0, 1,2, 3,4, or 5; R 4 is -OCH2ON(R 7 )2, -OCH 2 CH 2 N(R 7 )2, -CH2CH2ON(R 7 )2, -CH 2 CH 2 CH 2 N(R 7 ) 2 , -N(R)CH 2 ON(R 7 )2, -N(H)CH2ON(R 7 )2, -N(H)CH 2 CH 2 N(R 7 ) 2 , or -N(R)CH 2 CH 2 N(R 7 ) 2 ; R 5 is -OR 1 , or-NiR 1 ^; R 6 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -I, or (CH 2 )b-A; and TCX-003.88 /f 0 7 Ο 4 g g R 7 is -H, -R, or -C(=O)R ; or the two R 7 taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical. 43. The compound of claim 42, wherein m is 1. 5 44. The compound of claim 42, wherein m is 0. 45. The compound of any of claims 42 to 44, wherein n is 1. 46. The compound of any of claims 42 to 44, wherein n is 0. 47. The compound of any of claims 42 to 46, wherein R is alkyl or aralkyl. 48. The compound of claim 47, wherein R is lower alkyl. 10 49. The compound of claim 48, wherein R is-CH 3 ,-CH2CH3,-CH2CH2CH3, -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , or -CH 2 C 6 H 5 . 50. The compound of any of claims 42 to 49, wherein R 1 is -H, alkyl, or aralkyl. 51. The compound of claim 50, wherein R 1 is -H, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , 15 -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , or -CH 2 C 6 H 5 . 52. The compound of any of claims 42 to 51, wherein R 1 is -C(=O)R. 53. The compound of claim 52, wherein R 1 is -C(=O)R; and R is alkyl. 54. The compound of any of claims 42 to 53, wherein the two R 1 taken together with 20 the nitrogen to which they are bound represent -N=C(R)OR. 55. The compound of claim 54, wherein the two R 1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; and R is alkyl or aralkyl. 56. The compound of any of claims 42 to 55, wherein R 2 is -C=N, -C(=O)OH, -C(=O)OR, -CH 2 OH, or -CH 2 OR; and R is lower alkyl. 25 57. The compound of claim 56, wherein R 2 is -ON, -C(=O)OH, -C(=O)OCH 3 , -C(=O)OCH 2 CH 3 , or -CH 2 OH. 58. The compound of any of claims 42 to 57, wherein R 3 is -H, -R, -OR, -Cl, -F, -Br, -NO 2 , -NH 2 , -NHR, or -NR 2 ; and R is lower alkyl. TCX-003.88 fcu 70 4 66 59. The compound of claim 58, wherein R 3 is -H, -OCH 3 , -Cl, -F, -Br, -NO 2 , -NH 2 , or -CH 3 . 60. The compound of any of claims 42 to 59, wherein p is 1. 61. The compound of any of claims 42 to 60, wherein R 4 is -OCH2ON(R 7 )2, -OCH 2 CH 2 N(R 7 ) 2 . 62. The compound of claim 61, wherein R 4 is -OCH2CH2N(R 7 )2. 63. The compound of claim 42, wherein R 4 is -OCH2CH2N(R 7 )2. (claim 62 & 63 are identical) 64. The compound of any of claims 42 to 60, wherein R 4 is -CH2CH2ON(R 7 )2, or -CH 2 CH 2 CH 2 N(R 7 ) 2 , 65. The compound of claim 64, wherein R 4 is -CH2CH2CH2N(R 7 )2, 66. The compound of any of claims 42 to 60, wherein R 4 is -N(R)CH 2 ON(R 7 )2, -N(H)CH2ON(R 7 )2, -N(R)CH 2 CH 2 N(R 7 ) 2 , or -N(H)CH 2 CH 2 N(R 7 ) 2 . 67. The compound of claim 66, wherein R 4 is -N(R)CH2CH2N(R 7 )2, or -N(H)CH 2 CH 2 N(R 7 ) 2 . 68. The compound of any of claims 42 to 67, wherein R 5 is -OR 1 . 69. The compound of claim 68, wherein R 5 is -OR 1 ; and R 1 is -H, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3j -CH 2 C 6 H 5 , or-C(=O)CH 3 . 70. The compound of claim 69, wherein R 5 is -OH, -OCH 3 , -OCH 2 CH 3 , or -C(=O)CH 3 . 71. The compound of any of claims 42 to 67, wherein R 5 is -N(R ! ) 2 . 72. The compound of claim 71, wherein R 5 is -NiR'h; and R 1 is -H, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , -CH 2 C 6 H 5 , or -C(=O)CH 3 . 73. The compound of claim 72, wherein R 5 is -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -NH(CH 2 C 6 H 5 ), -N(CH 3 )(CH 2 C 6 H 5 ), -N(H)C(=O)CH 3 , or -N(CH 3 )C(=O)CH 3 . 74. The compound of any of claims 42 to 73, wherein R 6 is -H, -R, -OR, -Cl, -F, -Br, -NO 2 , -NH 2 , -NHR, or -NR 2 ; and R is lower alkyl. TCX-003.88 IE 0 7 0 4 § β 75. The compound of claim 74, wherein R 6 is -H, -OCH 3 , -Cl, -F, -Br, -NO 2 , -NH 2 , or -CH 3 . 76. The compound of any of claims 42 to 75, wherein R 7 is -H, -R, or -C(=O)R; and R is alkyl or aralkyl. 77. The compound of claim 76, wherein R 7 is -H, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , -CH 2 C 6 H 5 , or -C(=O)CH 3 . 78. The compound of any of claims 42 to 67, wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical. 79. The compound of claim 78, wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; and the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, piperidine, piperazine, morpholine, or diazadecaline (1,2,3,4,5,6,7,8-octahydroquinoxaline). 80. The compound of claim 79, wherein the two R 7 taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, piperidine, piperazine, morpholine, or diazadecaline (1,2,3,4,5,6,7,8-octahydroquinoxaline); and said optionally substituted monocyclic or bicyclic heterocycle radical is substituted with one or more substituents selected from the group consisting of -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , ON, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(-O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -I, and -(CH 2 ) b -A. *7 81. The compound of any of claims 42 to 67, wherein the two R taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; and the optionally substituted TCX-003.88 *°70466 monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, or piperidine. 82. The compound of claim 81, wherein the two R 7 taken together with the nitrogen to which they are attached represent an optionally substituted monocyclic or bicyclic heterocyclic radical; the optionally substituted monocyclic or bicyclic heterocycle radical is the radical of azetidine, pyrrolidine, piperidine, or piperazine; and said optionally substituted monocyclic or bicyclic heterocycle radical is substituted with one or more substituents selected from the group consisting of -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -ch 2 f, CHF 2 , -CF 3 , -ON, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -I, and -(CH 2 ) b -A. 83. A compound of formula III: wherein, independently for each occurrence, m is 0, 1, 2, 3, or 4; n is 0, 1,2, or 3; R is alkyl, polycyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl; R 1 is -H, -R, or -C(=O)R; or the two R 1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; R 2 is -C=N, -C(=O)OH, -C(=O)OR, -CH 2 OH, or -CH 2 OR; R 3 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -ON, -OH, -OR, -OC(=O)R, -OC(=O)OR, TCX-003.88 »070466 -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -I, or (CH 2 ) b -A; A is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2j -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, or -I; b is 0, 1,2, 3,4, or 5; R 6 is -R, -C(=O)H, -C(=O)OH, -C(=O)OR, -C(=O)NH 2 , -C(=O)NHR, -C(=O)NR 2 , -C(=O)R, -CH 2 F, -CHF 2 , -CF 3 , -C=N, -OH, -OR, -OC(=O)R, -OC(=O)OR, -OC(=O)NH 2 , -OC(=O)NHR, -OC(=O)NR 2 , -NH 2 , -NHR, -NR 2 , -N(H)C(=O)R, -N(R)C(=O)R, -N(H)C(=O)OR, -N(R)C(=O)OR, -N(H)C(=O)NH 2 , -N(R)C(=O)NH 2 , -N(H)C(=O)NHR, -N(R)C(=O)NHR, -N(H)C(=O)NR 2 , -N(R)C(=O)NR 2 , -NO 2 , -SH, -SR, -S(=O)R, -S(=O) 2 R, -SO 3 H, -Cl, -Br, -F, -I, or (CH 2 ) b -A; R 8 is -(CH2)PSR 10 , -(CH2) P S(=O)R 10 , -(CH2)pS(=O)2R 10 , -(CH2) p C(=O)N(R 1 )R 10 , -(CH2)pN(R’)CH2CH2CH2SR 1() , -(CH2) p N(R')CH 2 CH 2 CH 2 S(=O)R' 0 , or -(CH2)pN(R 1 )CH2CH 2 CH 2 SfoO) 2 R i0 ; p is 5,6, 7, 8, 9, or 10; R 9 is -H, -OH, or -OR; and R 10 is alkyl, fluoroalkyl or perfluoroalkyl. 84. The compound of claim 83, wherein m is 1. 85. The compound of claim 83, wherein m is 0. 86. The compound of any of claims 83 to 85, wherein n is 1. 87. The compound of any of claims 83 to 85, wherein n is 0. 88. The compound of any of claims 83 to 85, wherein R is alkyl or aralkyl. TCX-003.88 «070466 89. The compound of claim 88, wherein R is lower alkyl. 90. The compound of claim 89, wherein R is -CH 3 , -CH2CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , or -CH 2 C 6 H 5 . 5 91. The compound of any of claims 83 to 90, wherein R 1 is -H, alkyl, or aralkyl. 92. The compound of claim 91, wherein R 1 is -H, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , or -CH 2 C 6 H 5 . 93. The compound of any of claims 83 to 90, wherein R 1 is -C(=O)R. 10 94. The compound of claim 93, wherein R 1 is -C(=O)R; and R is alkyl. 95. The compound of any of claims 83 to 90, wherein the two R 1 taken together with the nitrogen to which they are bound represent -N=C(R)OR. 96. The compound of claim 95, wherein the two R 1 taken together with the nitrogen to which they are bound represent -N=C(R)OR; and R is alkyl or aralkyl. 15 97. The compound of any of claims 83 to 96, wherein R 2 is -C=N, -C(=O)OH, -C(=O)OR, -CH 2 OH, or -CH 2 OR; and R is lower alkyl. 98. The compound of claim 97, wherein R 2 is -C=N, -C(=O)OH, -C(=O)OCH 3 , -C(=O)OCH 2 CH 3 , or -CH 2 OH. 99. The compound of any of claims 83 to 98, wherein R 3 is -H, -R, -OR, -Cl, -F, -Br, 20 -NO 2 , -NH 2 , -NHR, or -NR 2 ; and R is lower alkyl. 100. The compound of claim 99, wherein R 3 is -H, -OCH 3 , -Cl, -F, -Br, -NO 2 , -NH 2 , or -CH 3 . 101. The compound of any of claims 83 to 100, wherein R 4 is -OCH2ON(R 7 )2, -OCH 2 CH 2 N(R 7 ) 2 . 25 102. The compound of claim 83, wherein R 4 is -OCH2ON(R 7 )2. 103. The compound of claim 83, wherein R 4 is -OCH2CH2N(R 7 )2. 104. The compound of any of claims 83 to 100, wherein R 4 is -CH2CH2ON(R 7 )2, or -CH 2 CH 2 CH 2 N(R 7 ) 2 , 105. The compound of claim 104, wherein R 4 is -CH2CH2CH2N(R 7 )2, TCX-003.88 ΙΕΟ 7 0 4 6 6 106. The compound of any of claims 83 to 100, wherein R 4 is -N(R)CH 2 ON(R 7 )2, -N(H)CH2ON(R 7 )2, -N(R)CH 2 CH 2 N(R 7 ) 2 , or -N(H)CH 2 CH 2 N(R 7 ) 2 . 107. The compound of claim 106, wherein R 4 is -N(R)CH2CH2N(R 7 )2, or -N(H)CH 2 CH 2 N(R 7 ) 2 . 5 108. The compound of any of claims 83 to 107, wherein R 6 is -H, -R, -OR, -Cl, -F, -Br, -NO 2 , -NH 2j -NHR, or -NR 2 ; and R is lower alkyl. 109. The compound of claim 108, wherein R 6 is -H, -OCH 3 , -Cl, -F, -Br, -NO 2 , -NH 2 , or -ch 3 . 110. The compound of any of claims 83 to 109, wherein R 8 is -(CH 2 ) P SR 10 , 10 -(CH 2 ) p S(=O)R 10 , or -(CH 2 ) p S(=O) 2 R 10 . 111. The compound of claim 110, wherein R 8 is -(CH2)PSR 10 , -(CH2) p S(=O)R 10 , or -(CH 2 ) P S(=O) 2 R 10 ; and R 10 is fluoroalkyl. 112. The compound of claim 111, wherein R 8 is -(CH 2 ) P SCH 2 CH 2 CH 2 CF 2 CF 3 , -(CH 2 ) P S(=O)CH 2 CH 2 CH 2 CF 2 CF 3 ,-(CH 2 ) P S(=O) 2 CH 2 CH 2 CH 2 CF 2 CF 3 15 113. The compound of any of claims 83 to 109, wherein R 8 is -(CH 2 ) P C(=O)N(R I )R 10 . 114. The compound of claim 113, wherein R 8 is -(CH 2 ) p C(=O)N(R’)R 10 ; and R 10 is alkyl. 115. The compound of claim 114, wherein R 8 is -(CH 2 ) P C(=O)N(CH 3 )CH 2 CH 2 CH 2 CH 3 . 116. The compound of any of claims 83 to 109, wherein R 8 is -(CH2)PN(R 1 )CH2CH 2 CH 2 SR 10 , -(CH2)pN(R 1 )CH2CH 2 CH 2 S(-O)R 10 , or -(CH2)PN(R ! )CH20 2CH 2 CH 2 S(=O) 2 R ,() . 117. The compound of claim 116, wherein R 8 is -(CH2)PN(R I )CH2CH 2 CH 2 SR 10 , -(CH2)pN(R , )CH2CH 2 CH 2 S(=O)R’°, or -(CH 2 ) p N(R i )CH 2 CH 2 CH 2 S(=O) 2 R 10 ; and R 10 is alkyl. o 118. The compound of claim 117, wherein R is 25 -(CH 2 ) P N(CH 3 )CH 2 CH 2 CH 2 SCH 2 CH 2 CH 2 CH 2 CH 3 , -(CH 2 ) p N(CH 3 )CH 2 CH 2 CH 2 SfyO)CH 2 CH 2 CH 2 CH 2 CH 3 , or -(CH 2 ) p N(CH 3 )CH 2 CH 2 CH 2 S(=O) 2 CH 2 CH 2 CH 2 CH 2 CH 3 . 119. The compound of any of claims 83 to 118, wherein p is 5. 120. The compound of any of claims 83 to 118, wherein p is 6. TCX-003.88 ΙΕΟ 7 0 4 6 6 121. 122. 123. 124.
10. Heterocyclic radical.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
IE20070466A IE20070466A1 (en) | 2007-06-28 | 2007-06-28 | Selective estrogen receptor modulators |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
IE20070466A IE20070466A1 (en) | 2007-06-28 | 2007-06-28 | Selective estrogen receptor modulators |
Publications (1)
Publication Number | Publication Date |
---|---|
IE20070466A1 true IE20070466A1 (en) | 2009-03-18 |
Family
ID=40513677
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
IE20070466A IE20070466A1 (en) | 2007-06-28 | 2007-06-28 | Selective estrogen receptor modulators |
Country Status (1)
Country | Link |
---|---|
IE (1) | IE20070466A1 (en) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20210015793A1 (en) * | 2018-03-19 | 2021-01-21 | Eth Zurich | Compounds for treating cns- and neurodegenerative diseases |
WO2023172608A3 (en) * | 2022-03-08 | 2023-12-07 | University Of Pittsburgh-Of The Commonwealth System Of Higher Education | Enhancing estrogen receptor alpha in osteoarthritis |
-
2007
- 2007-06-28 IE IE20070466A patent/IE20070466A1/en not_active Application Discontinuation
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20210015793A1 (en) * | 2018-03-19 | 2021-01-21 | Eth Zurich | Compounds for treating cns- and neurodegenerative diseases |
WO2023172608A3 (en) * | 2022-03-08 | 2023-12-07 | University Of Pittsburgh-Of The Commonwealth System Of Higher Education | Enhancing estrogen receptor alpha in osteoarthritis |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
EP3184519B1 (en) | Acid-addition salt of trk-inhibiting compound | |
CN102405043B (en) | Preceding neurogenic compounds | |
Zhang et al. | Identification of N-propylnoraporphin-11-yl 5-(1, 2-dithiolan-3-yl) pentanoate as a new anti-Parkinson's agent possessing a dopamine D2 and serotonin 5-HT1A dual-agonist profile | |
CN102046167A (en) | Solid forms comprising (+)-2-[1-(3-ethoxy-4-methoxyphenyl)-2-methylsulfonylethyl]-4-acetylaminoisoindoline-1,3-dione, compositions thereof, and uses thereof | |
da Silva et al. | Thiazolidin-4-ones from 4-(methylthio) benzaldehyde and 4-(methylsulfonyl) benzaldehyde: synthesis, antiglioma activity and cytotoxicity | |
JP2021193149A (en) | Crystalline spirocyclic compound inhibitor of tryptophan hydroxylase 1 (tph1) for treating diseases or disorders associated with peripheral serotonin | |
CN107007587A (en) | For inducing chondrogenetic Compounds and methods for | |
JP6977238B2 (en) | Compounds for the treatment of hypoproliferative disorders | |
JP5913651B2 (en) | Compounds and methods for the treatment of pain and other diseases | |
CA3098219C (en) | A 10h-phenothiazine ferroptosis inhibitor as well as the preparative method and the use thereof | |
EA035989B1 (en) | Crystalline forms and processes for the preparation of cannabinoid receptor modulators | |
AU2011360973A1 (en) | Compounds and methods for the treatment of pain and other disorders | |
CA3119313A1 (en) | Pharmaceutical composition comprising histone deacetylase 6 inhibitors | |
JPWO2005079845A1 (en) | Migraine prophylaxis | |
IE20070466A1 (en) | Selective estrogen receptor modulators | |
CN105884645B (en) | A kind of rhein compound and application thereof | |
US20130338116A1 (en) | Compounds and methods for the treatment of pain and other diseases | |
CN102702070A (en) | Solid form containing (+)-2-[1-(3-oxethyl-4-methoxyphenyl)-2-methylsulfonylethyl]-4-acetylaminoisoindoline-1,3-dione, composition and application thereof | |
US20110293745A1 (en) | Aurora kinase inhibitors | |
TW201609106A (en) | BTK inhibitors for the treatment of CNS malignancies | |
CN117003737A (en) | Substituted imidazolyl phenyl ketone compound, derivative and pharmaceutical composition thereof | |
CN112689528B (en) | Pyrimidinyl-heteroaryloxy-naphthyl compounds and methods of use | |
JP2006515275A (en) | Benzopyranone compounds, compositions thereof, and methods of treating or preventing cancer | |
Byrtus et al. | Synthesis and 5-HT1A, 5-HT2A receptor activity of new β-tetralonohydantoins | |
Kovacikova et al. | Substituted pyridoindoles as biological antioxidants: drug design, chemical synthesis, and biological activity |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
FC9A | Application refused sect. 31(1) |