Analogs of thalidomide as potential angiogenesis inhibitors

Figg , et al. May 6, 2

Patent Grant 8716315

U.S. patent number 8,716,315 [Application Number 11/880,404] was granted by the patent office on 2014-05-06 for analogs of thalidomide as potential angiogenesis inhibitors. This patent grant is currently assigned to N/A, The United States of America as represented by the Secretary of the Department of Health and Human Services. The grantee listed for this patent is Kurt Eger, William D. Figg, Michael Guetschow, Sunna Hauschildt, Thomas Hecker, Uwe Teubert, Michael Weiss. Invention is credited to Kurt Eger, William D. Figg, Michael Guetschow, Sunna Hauschildt, Thomas Hecker, Uwe Teubert, Michael Weiss.


United States Patent 8,716,315
Figg ,   et al. May 6, 2014

Analogs of thalidomide as potential angiogenesis inhibitors

Abstract

A number of thalidomide metabolites having superior anti-angiogenic properties have now been isolated and identified. In addition, thalidomide analogs that mimic the effects of the isolated and identified active thalidomide metabolites, and variations of such thalidomide analogs, have been developed. Such thalidomide analog compounds show enhanced potency in the inhibition of undesirable angiogenesis without the undesirable effects of administration of thalidomide.


Inventors: Figg; William D. (Fairfax, VA), Eger; Kurt (Leipzig, DE), Teubert; Uwe (Hameln, DE), Weiss; Michael (Bethesda, MD), Guetschow; Michael (Bonn, DE), Hecker; Thomas (Erfurt, DE), Hauschildt; Sunna (Leipzig, DE)
Applicant:
Name City State Country Type

Figg; William D.
Eger; Kurt
Teubert; Uwe
Weiss; Michael
Guetschow; Michael
Hecker; Thomas
Hauschildt; Sunna

Fairfax
Leipzig
Hameln
Bethesda
Bonn
Erfurt
Leipzig

VA
N/A
N/A
MD
N/A
N/A
N/A

US
DE
DE
US
DE
DE
DE
Assignee: The United States of America as represented by the Secretary of the Department of Health and Human Services (Washington, DC)
N/A (N/A)
Family ID: 23037728
Appl. No.: 11/880,404
Filed: July 19, 2007

Prior Publication Data

Document Identifier Publication Date
US 20070293519 A1 Dec 20, 2007

Related U.S. Patent Documents

Application Number Filing Date Patent Number Issue Date
10469359 7320991
PCT/US02/05868 Feb 26, 2002
60271941 Feb 27, 2001

Current U.S. Class: 514/323
Current CPC Class: C07D 401/04 (20130101); A61K 31/513 (20130101); A61K 31/506 (20130101); C07D 403/04 (20130101); A61K 31/454 (20130101); A61P 43/00 (20180101); C07D 209/48 (20130101); A61K 31/4035 (20130101); A61P 35/02 (20180101); A61P 35/00 (20180101); A61P 9/00 (20180101); A61K 31/4035 (20130101); A61K 2300/00 (20130101); A61K 31/454 (20130101); A61K 2300/00 (20130101); A61K 31/506 (20130101); A61K 2300/00 (20130101)
Current International Class: A61K 31/445 (20060101)
Field of Search: ;514/323

References Cited [Referenced By]

U.S. Patent Documents
2830991 April 1958 Keller et al.
3314953 April 1967 Vazakas et al.
3320270 May 1967 Grogan et al.
3560495 February 1971 Frankus et al.
3794641 February 1974 Gorog et al.
4092147 May 1978 Ashkar
4291048 September 1981 Gold et al.
5434170 July 1995 Andrulis, Jr.
5593990 January 1997 D'Amato
5605684 February 1997 Piacquadio
5629327 May 1997 D'Amato
5712291 January 1998 D'Amato
5783605 July 1998 Kuo et al.
5789434 August 1998 Kluender et al.
5840724 November 1998 Fenton et al.
6071948 June 2000 D'Amato
6080742 June 2000 Germann et al.
6096768 August 2000 Ashton et al.
6110941 August 2000 Zimmer et al.
6124322 September 2000 Bjoerkman et al.
6235756 May 2001 D'Amato
6306879 October 2001 Germann et al.
6417197 July 2002 Schneider et al.
6420414 July 2002 D'Amato
6429212 August 2002 Hashimoto
6458810 October 2002 Muller et al.
6469045 October 2002 D'Amato
6500845 December 2002 Boehlke et al.
6518298 February 2003 Green et al.
6555554 April 2003 Muller et al.
6762195 July 2004 Muller et al.
2001/0041716 November 2001 Laing et al.
2002/0022627 February 2002 Dannenberg
2002/0161023 October 2002 D'Amato
2003/0013739 January 2003 Masferrer
2003/0181428 September 2003 Green et al.
2005/0004087 January 2005 D'Amato et al.
Foreign Patent Documents
2104776 Aug 1992 CA
2251060 Mar 1997 CA
2228385 Jan 1998 CA
2248838 Oct 1998 CA
2302886 Mar 2000 CA
33 32 633 Apr 1985 DE
0 688 771 Dec 1995 EP
0 856 513 Aug 1996 EP
0 908 176 Apr 1999 EP
0 688 771 Jul 2001 EP
1 336 602 Aug 2003 EP
962857 Jul 1964 GB
1075420 Jul 1967 GB
1049283 Nov 1996 GB
10-109975 Apr 1998 JP
WO 92/18496 Oct 1992 WO
WO 94/20085 Sep 1994 WO
WO 95/17154 Jun 1995 WO
WO 96/20705 Jul 1996 WO
WO 96/20926 Jul 1996 WO
WO 97/12652 Apr 1997 WO
WO 97/23457 Jul 1997 WO
WO 97/37988 Oct 1997 WO
WO 97/45117 Dec 1997 WO
WO 98/03502 Jan 1998 WO
WO 98/19649 May 1998 WO
WO 98/25895 Jun 1998 WO
WO 99/13873 Mar 1999 WO
WO 99/58096 Nov 1999 WO
WO 99/59960 Nov 1999 WO
WO 01/74362 Oct 2001 WO
WO 02/064083 Aug 2002 WO
WO 02/068414 Sep 2002 WO
WO 03/014315 Feb 2003 WO
WO 03/097052 Nov 2003 WO
WO 2004/085422 Oct 2004 WO
WO 2005/016325 Feb 2005 WO
WO 2005/016326 Feb 2005 WO

Other References

Niwayama et al. "Enhanced potency . . . " Bioorganic & Med.Chemi Lett. v.8 p. 1071-1076 (1998). cited by examiner .
Shibata et al. "Pnhenylphthimides . . . " chem. Pharm. Bull. v. 44, p. 156-162 (1996). cited by examiner .
Gutschow et al. "Aza analogues . . . " Bioorg. Med. Chem. vo. 9, p. 1059-1065 (2001). cited by examiner .
MPEP 2132, p. 1-2, electronic pages (2013). cited by examiner .
NOT-OD-05-022 "Polucy on enhancing . . . " p. 1-14 (2005). cited by examiner .
Bartlett et al., "Phase I study to determine the safety, tolerability and immunostimulatory activity of thalidomide analogue CC-5013 in patents with metastatic malignant melanoma and other advanced cancers," British Journal of Cancer 90:955-961, 2004. cited by applicant .
Bauer et al., "Inhibition of Angiogenesis by Thalidomide Requires Metabolic Activation, Which is Species-dependent," Biochemical Pharmacology 55:1827-1834, 1998. cited by applicant .
Bray et al., "Improved Procedures for the Preparation of (+)--(1R, 2S, 4R)-4-Amino-2-Hydroxy-1-Hydroxymethyl Cyclopentane," Tetrahedron Letters 36(25):4483-4486, 1995. cited by applicant .
Cava et al., "Thionation Reactions of Lawesson's Reagents," Tetrahedron 41(22):5061-5087, 1985. cited by applicant .
Ching et al., "Interaction of thalidomide, phthalimide analogues of thalidomide and pentoxifylline with the anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid: concomitant reduction of serum tumour necrosis factor--alpha and enhancement of anti-tumour activity," British Journal of Cancer 78(3):336-343, 1998. cited by applicant .
Corral et al., "Differential Cytokine Modulation and T Cell Activation by Two Distinct Classes of Thalidomide Analogues That Are Potent Inhibitors of TNF-.alpha.," The Journal of Immunology 163:380-386, 1999. cited by applicant .
D'Amato et al., "Thalidomide is an Inhibitor of Angiogenesis," Proc. Nat'l. Acad. Sci. USA 91:4082-4085, 1994. cited by applicant .
Davies et al., "Thalidomide (Thal) and Immunomodulatory Derivatives (IMiDs) Augment Natural Killer (NK) Cell Cytotoxicity in Multiple Myeloma (MM)," American Society of Hematology, 42.sup.nd Annual Meeting, San Francisco, CA, Dec. 1-5, 2000 (Abstract No. 3617). cited by applicant .
Davies et al., "Thalidomide (Thal) and Immunomodulatory Derivatives (IMiDs) Augment Natural Killer (NK) Cell Cytotoxicity in Multiple Myeloma (MM)," VIIIth International Myeloma Workshop, Banff, Canada, May 4-8, 2001 (Abstract No. P222). cited by applicant .
Davies et al., "Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma," Blood 98(1):210-216, 2001. cited by applicant .
Dalgleish et al., "Thalidomide Analogues CC-5013 and CC-4047 Induce T cell Activation and IL-12 Production in Patients with Both Solid Tumours and Relapsed and Refractory Multiple Myeloma," British Journal of Cancer 88(Suppl. 1)S25-S54 (Abstract P14), 2003. cited by applicant .
De et al., "Possible Antineoplastic Agents: III--Synthesis of 6-Alkyl-2-[4'Methoxyphalimido] and 6-Alkyl-3-[3'-4'-Dimethoxyphenyl] Glutarimides," J. Indian Chem. Soc. 53:1122-1125, 1976. cited by applicant .
De et al., "Possible Antineoplastic Agents: Part IV--Synthesis & Antineoplastic Potency of N-Substituted .alpha.-(4,5-Dimethoxyphthalimido)glutarimides & N-Substituted .beta.-)4-Bromophenyl)gludarimides," Indian J. of Chem. 16B:510-512, 1978. cited by applicant .
Deckers et al., "Effect on Angiogenic and Antiangiogenic Compounds on the Outgrowth of Capillary Structures from Fetal Mouse Bone Explants," Laboratory Investigation 81(1):5-15, 2001. cited by applicant .
Dibbs et al., "Thalidomide and Thalidomide Analogs Suppress TNF.alpha. Secretion by Myocytes," Circulation 98(17(Suppl), Abstract No. 1284):I247, 1998. cited by applicant .
Dredge et al., "A costimulatory thalidomide analog enhances the partial anti-tumor immunity of an autologous vaccination in a model of colorectal cancer," American Association for Cancer Research, 93.sup.rd AnnualMeeting, San Francisco, CA, Apr. 6-10, 2002 (Abstract No. 491). cited by applicant .
Dredge et al., "Protective Antitumor Immunity Induced by a Costimulatory Thalidomide Analog in Conjunction with Whole Tumor Cell Vaccination Is Mediated by Increased Th1-Type Immunity," The Journal of Immunology 168:4914-4919, 2002. cited by applicant .
Dredge et al., "Novel thalidomide analogues display anti-angiogenic activity independently of immunomodulatory effects," British Journal of Cancer 87:1166-1172, 2002. cited by applicant .
Dredge et al., "Recent developments in antiangiogenic therapy," Expert Opin. Biol. Ther. 2(8):953-966, 2002. cited by applicant .
Dredge et al., "Immunological Effects of Thalidomide and Its Chemical and Functional Analogs," Critical Reviews in Immunology 22(5&6):425-437, 2002. cited by applicant .
Dredge et al., "Thalidomide analogs as emerging anti-cancer drugs," Anti-Cancer Drugs 14:331-335, 2003. cited by applicant .
Dredge et al., "Angiogenesis inhibitors in cancer therapy," Current Opinion in Investigational Drugs 4(6):667-674, 2003. cited by applicant .
Eisen et al., "Continuous low dose Thalidomide: a phase II study in advanced melanoma, renal cell, ovarian and breast cancer," British Journal of Cancer 82(4):812-817, 2000. cited by applicant .
Fine et al., "Phase II Trial of the Antiangiogenic Agent Thalidomide in Patients with Recurrent High-Grade Gliomas," Journal of Clinical Oncology 18(4):708-715, 2000. cited by applicant .
Folkes et al., "Oxidative activation of indole-3-acetic acids to cytotoxic species--a potential new role for plant auxins in cancer therapy," Biochemical Pharmacology 61(2):129-136, 2001. cited by applicant .
Greig et al., "New Therapeutic Strategies and Drug Candidates for Neurodegenerative Diseases. p53 and TNF-.alpha. Inhibitors, and GLP-1 Receptor Agonists," Ann. N.Y. Acad. Sci. 1035:290-315, 2004. cited by applicant .
Greig et al., "Thalidomide-based TNF-.alpha. Inhibitors for neurodegenerative diseases," Acta Neurobiol Exp 64(1):1-9, 2004. cited by applicant .
Gupta et al., "Adherence of multiple myeloma cells to bone morrow stromal cells upregulates vascular endothelial growth factor secretion: therapeutic applications," Leukemia 15:1950-1961, 2001. cited by applicant .
Gutschow et al., "Aza analogues of thalidomide: Synthesis and evaluation as inhibitors of tumor necrosis factor-alpha production in vitro" Bioorganic & Medical Chemistry 9:1059-1065, 2001. cited by applicant .
Hashimoto, "Novel Biological Response Modifier Derived from Thalidomide," Current Medicinal Chemistry 5(3):163-178, 1998. cited by applicant .
Hashimoto et al., "Structural Development of Biological Response Modifiers Based on Thalidomide," Bioorganic & Medicinal Chemistry 10:461-479, 2002. cited by applicant .
Haslett et al., "Thalidomide and a Thalidomide Analogue Drug Costimulate Virus-Specific CD8.sup.+ T Cells in Vitro," The Journal of Infectious Diseases 187:946-955, 2003. cited by applicant .
Hayashi et al., "Mechanisms Whereby Immunomodulatory Analogs of Thalidomide Augment Autologous NK Cell Anti-Myeloma Immunity," American Society of Hematology, 44.sup.th Annual Meeting, Philadelphia, PA, Dec. 6-10, 2002 (Abstract No. 3219). cited by applicant .
He et al., "Synthesis of Thalidomide Analogs and Their Biological Potential for Treatment of Graft Versus Host Disease (GVHD)," American Chemical Society (Abstract No. 216), 1993. cited by applicant .
Hernandez-Ilizaliturri et al., "Addition of Immunomodulatory Drugs CC5013 or CC4047 to Rituximab Enhances Anti-Tumor Activity in a Severe Combined Immunodeficiency (SCID) Mouse Lymphoma Model," American Society of Hematology, 45.sup.th Annual Meeting, San Diego, CA, Dec. 6-9, 2003 (Abstract No. 235). cited by applicant .
Hess et al., "Synthesis and immunological activity of water-soluble thalidomide prodrugs," Bioorganic and Medicinal Chem. 9(5):1279-1291, 2001. cited by applicant .
Hideshima et al., "Thalidomide (Thal) and Its Analogs Overcome Drug Resistance of Human Multiple Myeloma (MM) Cells to Conventional Therapy," American Society of Hematology, 42.sup.nd Annual Meeting, San Francisco, CA, Dec. 1-5, 2000 (Abstract No. 1313). cited by applicant .
Isner et al., "Therapeutic Angiogenesis," Frontiers in Bioscience 3:e49-69, 1998. cited by applicant .
Jonsson, "Chemical structure and teratogenic properties," Acta Pharm. Suecica 9:521:542, 1972. cited by applicant .
Karbownik et al., "Indole-3-propionic acid, a melatonin-related molecule, protects hepatic microsomal membranes from iron-induced oxidative damage: relevance to cancer reduction," Journal of Cellular Biochemistry 81(3):507-513, 2001. cited by applicant .
Kim et al., "Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo," Nature 362:841-844, 1993. cited by applicant .
Kumar et al., "Antimyeloma activity of two novel N-substituted and tetraflourinated thalidomide analogs," Leukemia 19:1253-1261, 2005. cited by applicant .
Lentzsch et al., "S-3-amino-phthalimido-glutarimide inhibits angiogenesis and growth of B-cell neoplasias in mice," Cancer Research 62:2300-2305, 2002. cited by applicant .
Lentzsch et al., "Immunomodulatory Derivatives of Thalidomide (IMiD CC-4047) Down Regulates CAAT/Enhancer-Binding Protein .beta.(C/EBP.beta.) in Multiple Myeloma (MM)", American Society of Hematology, 45.sup.th Annual Meeting, San Diego, CA, Dec. 6-9, 2003 (Abstract No. 3456). cited by applicant .
Lentzsch et al., "Immunomodulatory Derivatives of Thalidomide (IMiD CC-4047) Determine the Lineage Commitment of Hematopoietic Progenitors by Down Regulation of GATA-1 and Modulation of Cytokine Secretion," American Society of Hematology, 45.sup.th Annual Meeting, San Diego, CA, Dec. 6-9, 2003 (Abstract No. 3073). cited by applicant .
Lepper et al., "Comparative Molecular Field Analysis and Comparative Molecular Similarity Indices Analysis of Thalidomide Analogues as Angiogenesis Inhibitors," J Medicinal Chem. 47(9):2219-2227, 2004. cited by applicant .
Little et al., "Activity of Thalidomide in AIDS-Related Kaposi's Sarcoma," Journal of Clinical Oncology 18(13):2593-2602, 2000. cited by applicant .
Luzzio et al., "Thalidomide metabolites and analogs. Part 2: Cyclic derivatives of 2-N-phthalimido-2S,3S (3-hydroxy) ornithine," Tetrahedron Letters 41:7151-7155, 2000. cited by applicant .
Luzzio et al., "Thalidomide Metabolites and Analogues. 3. Synthesis and Antiangiogenic Activity of the Teratogenic and TNF.alpha.-Modulatory Thalidomide Analogue 2-(2,6-Dioxopiperidine-3-yl)phthalimidine," Journal of Medicinal Chemistry 46(18):3793-3799, 2003. cited by applicant .
Man et al., ".alpha.-Fluoro-Substituted Thalidomide Analogues," Bioorganic & Medicinal Chemistry Letters 13:3415-3417, 2003. cited by applicant .
Marks et al., "Effects of Putative Hydroxylated Thalidomide Metabolites on Blood Vessel Density in the Chorioallantoic Membrane (CAM) Assay and on Tumor and Endothelial Cell Proliferation," Biol. Pharm. Bull. 25(5):597-604, 2002. cited by applicant .
Marriott et al., "CC-3052: A Water-Soluble Analog of Thalidomide and Potent Inhibitor of Activation-Induced TNF-.alpha. Production," The Journal of Immunology 161:4236-4243, 1998. cited by applicant .
Marriott et al., "Immunotherapeutic and antitumour potential of thalidomide analogues," Expert Opin. Biol. Ther. 1(4):1-8, 2001. cited by applicant .
Marriott et al., "Thalidomide and its analogues have distinct and opposing effects on TNF-.alpha. and TNFR2 during co-stimulation of both CD4.sup.+ and CD8.sup.+ T cells," Clin Exp Immunol 130:75-84, 2002. cited by applicant .
Marriott et al., "A Novel Subclass of Thalidomide Analogue with Anti-Solid Tumor Activity in Which Caspase-dependent Apoptosis is Associated with Altered Expression of bcl-2 Family Proteins," Cancer Research 63:593-599, 2003. cited by applicant .
Marriott et al., "Thalidomide Derived Immunomodulatory Drugs (IMiDs) as Potential Therapeutic Agents," Current Drug Targets--Immune, Endocrine & Metabolic Disorders 3:181-186, 2003. cited by applicant .
Meierhofer et al., "New insights into the pharmacological and toxicological effects of thalidomide," Current Opinion in Drug Discovery & Development 6(1):92-99, 2003. cited by applicant .
Mitsiades et al., "Apoptotic signaling induced by immunomodulatory thalidomide analogs in human multiple myeloma cells: therapeutic implications," Blood 99(12):4525-4530, 2002. cited by applicant .
Moutouh de Parseval et al., "Novel immunomodulatory drugs (IMiDs.RTM.): A potential, new therapy for .beta.-hemoglobinopathies," American Society of Hematology, 46.sup.th Annual Meeting, San Diego, CA, Dec. 4-7, 2004 (Abstract No. 3740). cited by applicant .
Miyashi et al., "Inducer-Specific Regulators of tumor Necrosis Factor Alpha Production," Chem. Pharm. Bull. 44(10):1980-1982, 1996. cited by applicant .
Miyashi et al., "Novel Biological Response Modifiers: Phthalimides with Tumor Necrosis Factor-.alpha. Production-Regulating Activity," J. Med. Chem. 40:2858-2865, 1997. cited by applicant .
Muller et al., "Amino-Substituted Thalidomide Analogs: Potent Inhibitors of TNF-.alpha. Production," Bioorganic & Medicinal Chemistry Letters 9:1625-1630, 1999. cited by applicant .
Ng et al., "Antiangiogenic Activity of N-substituted and Tetrafluorinated Thalidomide Analogues," Cancer Research 63(12):3189-3194, 2003. cited by applicant .
Ng et al., "Antitumor Effects of Thalidomide Analogs in Human Prostate Cancer Xenografts Implanted in Immunodeficient Mice," Clinical Cancer Research 10:4192-4197, 2004. cited by applicant .
Ni et al., "Experimental study on activity of human recombinant adenovirus vector expressing human endostatin in vitro," Dier Junyi Daxue Xuebao Bianjibu 23(3):261-263, 2002 (Abstract only). cited by applicant .
Niwayama et al., "Enhanced Potency of Perfluorinated Thalidomide Derivatives for Inhibition of LPS-Induced Tumor Necrosis Factor-.alpha. Production is Associated with a Change of Mechanism of Action," Bioorganic & Medicinal Chemistry Letters 8:1071-1076, 1998. cited by applicant .
Niwayama et al., "Potent Inhibition of Tumor Necrosis Factor-.alpha. Production by Tetrafluorothalidomide and Tetrafluorophthalimides," J. Med. Chem. 39:3044-3045, 1996. cited by applicant .
Neumann et al., "N-(Sulfonyloxy)phthalimides and Analogues Are Potent Inactivators of Serine Proteases," The Journal of Biological Chemistry 269(34):21561-21567, 1994. cited by applicant .
Orzeszko et al., "Tumor necrosis factor-alpha production-regulating activity of phthalimide derivatives in genetically modified murine melanoma cells B78H1," Il Farmaco 58:371-376, 2003. cited by applicant .
Pagani et al., "Effect of a series of substances related to N-alpha-naphthylphthalamic acid on root geotropism of Lens esculanta Moench s.1. seeds," FRPSX, Farmaco Ed. Sci. 25:203-224, 1970. cited by applicant .
Patten et al., "The Early Use of the Serum Free Light Chain Assay in Patients with Relapsed Refractory Myeloma Receiving Treatment with a Thalidomide Analogue (CC-4047)," American Society of Hematology, 45.sup.th Annual Meeting, San Diego, CA, Dec. 6-9, 2003 (Abstract No. 1640). cited by applicant .
Payvandi et al., "The thalidomide analogs IMiDs enhance expression of CD69 stimulatory receptor on natural killer cells," American Association of Cancer Research, 92.sup.nd Annual Meeting, New Orleans, LA, Mar. 24-28, 2001 (Abstract No. 1793). cited by applicant .
Payvandi et al., "Thalidomide analogs IMiDs inhibit expression of cyclooxygenase-2 in multiple myeloma cell line and LPS stimulated PBMCs," American Society of Hematology, 43.sup.rd Annual Meeting, Orlando, FL, Dec. 7-11, 2001 (Abstract No. 2689). cited by applicant .
Payvandi et al., "Thalidomide and IMiDs Inhibit Microvessel Formation from Human Arterial Rings in the Absence of Human Liver Microsomes," American Society of Hematology, 44.sup.th Annual Meeting, Philadelphia, PA, Dec. 6-10, 2002 (Abstract No. 5046). cited by applicant .
Payvandi et al., "CC-5013 inhibits the expression of adhesion molecules ICAM-1 and CD44 and prevents metastasis of B16 F10 mouse melanoma cells in an animal model," American Society of Clinical Oncology, 39.sup.th Annual Meeting, Chicago, IL, May 31-Jun. 3, 2003 (Abstract No. 992). cited by applicant .
Payvandi et al., "Immunomodulatory drugs inhibit expression of cyclooxygenase-2 from TNF-.alpha., IL-1.beta., and LPS-stimulated human PBMC in a partially IL-10-dependent manner," Cellular Immunology 230:81-88, 2004. cited by applicant .
Pratt et al., "Phthalic Acid Derivatives: Constitution and Color, XIV..sup.I Some Derivatives of Tetrabromophthalimide," JACSAT J. Amer. Chem. Soc. 40:1420, 1918. cited by applicant .
Schafer et al., "Enhancement of Cytokine Production and AP-1 Transcriptional Activity in T Cells by Thalidomide-Related Immunomodulatory Drugs," The Journal of Pharmacology and Experimental Therapeutics 305(3):1222-1232, 2003. cited by applicant .
Schey et al., "Phase I Study of an Immunomodulatory Thalidomide Analog, CC-4047, in Relapsed or Refractory Multiple Myeloma," Journal of Clinical Oncology 22(16):1-8, 2004. cited by applicant .
Sedlak et al., "Preparation, 1H and 13C NMR spectra of substituted 2-benzoylaminocarboxamides," Collection of Czechoslovak Chemical Communications, Academic Press 60:150-160, 1995. cited by applicant .
Shah et al., "Synehesis and Enantiomeric Separation of 2-Phthalimidino-glutaric Acid Analogues: Potent Inhibitors of Tumor Metastasis," J. Med. Chem. 42:3014-3017, 1999. cited by applicant .
Shaughnessy et al., "Global Gene Expression Analysis Shows Loss of C-MYC and IL-6 Receptor Gene mRNA After Exposure of Myeloma to Thalidomide and IMiD," The American Society of Hematology, 42.sup.nd Annual Meeting, San Francisco, CA, Dec. 1-5, 2000 (Abstract No. 2485). cited by applicant .
Shimazawa et al., "Antiangiogenic Activity of Tumor Necrosis Factor-.alpha. Production Regulators Derived from Thalidomide," Biol. Pharm. Bull. 22(2):224-226, 1999. cited by applicant .
Shimazawa et al., "Nonpeptide Small-Molecular Inhibitors of Dipeptidyl Peptidase IV:N-Phenylphthalimide Analogs," Bioorganic & Medicinal Chemistry Letters 9:559-562, 1999. cited by applicant .
Shimazawa et al., "Novel Small Molecule Nonpeptide Aminopeptidase N inhibitors with a Cyclic Imide Skeleton," J. Enzyme Inhibition 14:259-275, 1999. cited by applicant .
Shire et al., "TNF-.alpha. inhibitors and rheumatoid arthritis," Exp. Opin. Ther. Patents 8(5):531-544, 1998. cited by applicant .
Singhal et al., "Antitumor Activity of Thalidomide in Refractory Multiple Myeloma," The New England Journal of Medicine 341(21):1565-1571, 1999. cited by applicant .
Singhal et al., "Thalidomide in Cancer. Potential Uses and Limitations," BioDrugs 15(3):163-172, 2001. cited by applicant .
Streetly et al., "Thalidomide analogue CC-4047 is effective in the treatment of patients with relapsed and refractory multiple myeloma (MM) and induces T-cell activation and IL-12 production," International Multiple Myeloma Workshop, IXth International Conference, Salamanca, Spain, May 23-27, 2003 (Abstract No. 367). cited by applicant .
Streetly et al., "An Update of the Use and Outcomes of the New Immunomodulatory Agent CC-4047 (Actimid) in Patients with Relapsed/Refractory Myeloma," American Society of Hematology, 45.sup.th Annual Meeting, San Diego, CA, Dec. 6-9, 2003 (Abstract No. 829). cited by applicant .
Streetly et al., "Changes in Neutrophil Phenotype Following the Administration of CC-4047 (Actimid) to Patients with Multiple Myeloma," American Society of Hematology, 45.sup.th Annual Meeting, San Diego, CA, Dec. 6-9, 2003 (Abstract No. 2543). cited by applicant .
Suzuki, et al., "Use of a new protecting group in an attempted synthesis of cyclopropyldihydroxyphenylalanine," Journal of Organic Chemistry 48(24):4769-4771, 1983. cited by applicant .
Teo et al., "Chiral Inversion of the Second Generation IMiD.TM. CC-4047 (ACTIMID.TM.) in Human Plasma and Phosphate-Buffered Saline," Chirality 15:348-351, 2003. cited by applicant .
Teubert et al., "5'-Substituted Thalidomide Analogues as Modulators of TNF-.alpha.," Arch. Pharm. Pharm. Med. Chem. 331:7-12, 1998. cited by applicant .
"The Merck Index," Merck & Co. 2001. cited by applicant .
Tsenova et al., "Use of IMiD3, a Thalidomide Analog, as an Adjunct to Therapy for Experimental Tuberculous Meningitis," Antimicrobial Agents and Chemotherapy 46(6):1887-1895, 2002. cited by applicant .
Tweedie et al., "TNF-.alpha. Synthesis Inhibitors on the 3-Phthalimidoglutarimide Backbone as Therapeutic Candidates for Neurodegenerative Diseases," 7.sup.th International Conference on Alzheimer's and Parkinson's Disease, pp. 77-86, Sorrento, Italy, Mar. 9-13, 2005. cited by applicant .
Understanding Angiogenesis "About Angiogenesis," Angiogenesis Foundation from the website, 2005. cited by applicant .
Weinz et al., "Investigation of the in vitro biotransformation and simultaneous enantioselective separation of thalidomide and its neutral metabolites by capillary electrophoresis," Journal of Chromatography B 674:287-292, 1995. cited by applicant .
Ye et al., "Novel IMiD Drugs Enhance Expansion and Regulate Differentiation of Human Cord Blood CD34+ Cells with Cytokines," American Society of Hematology, 44.sup.th Annual Meeting, Philadelphia, PA, Dec. 6-10, 2002 (Abstract No. 4099). cited by applicant .
Zeldis et al., "Potential New Therapeutics for Waldenstrom's Macroglobulinemia," Seminars in Oncology 30(2):275-281, 2003. cited by applicant .
Zeldis et al., "Update on the evolution of the IMiD.TM.," International Society for Biological Therapy of Cancer, 18.sup.th Annual Meeting, Bethesda, MD, Oct. 30-Nov. 2, 2003 (Oral Abstract). cited by applicant .
Zhang et al., "CC-5079, a novel microtubule and TNF-a inhibitor with anti-angiogenic and antimetastasis activity," American Association for Cancer Research, National Cancer Institute, and European Organization for Research and Treatment of Cancer, International Conference on Molecular Targets and Cancer Therapeutics, Boston, MA, Nov. 17-21, 2003 (Abstract No. B012). cited by applicant .
Zhang et al., "Preparation of water-soluble thalidomide derivatives," Database CA Online, Chemical Abstracts Service, Database Accession No. 2004:817880, 2004 (Abstract). cited by applicant .
Zhu et al., "Thiothalidomides: Novel Isosteric Analogues of Thalidomide with Enhanced TNF-.alpha. Inhibitory Activity," J. Med. Chem. 46(24):5222-5229, 2003. cited by applicant .
Shibata et al., "Phenylphthalimides with Tumor Necrosis Factor Alpha Production-Enhancing Activity," Chem. Pharm. Bull. 44(1):156-162, Jan. 1996. cited by applicant.

Primary Examiner: Chang; Celia
Attorney, Agent or Firm: Klarquist Sparkman, LLP

Parent Case Text



PRIORITY CLAIM

This is a continuation of U.S. patent application Ser. No. 10/469,359, filed Nov. 14, 2003, which is a .sctn.371 U.S. national stage of PCT/US02/05868, filed Feb. 26, 2002, which was published in English under PCT Article 21(2), and claims the benefit of U.S. Provisional Application No. 60/271,941, filed Feb. 27, 2001, all of which are incorporated by reference in their entirety.
Claims



We claim:

1. A composition that inhibits angiogenesis in a subject when administered in an effective amount, wherein the composition comprises one or more compounds having the following formula or pharmaceutically acceptable salts thereof; ##STR00012## wherein R4 through R7 are, independently, a halogen, or R5 and R6 are hydrogen and R4 and R7 are methyl groups.

2. The composition of claim 1, wherein the compound is: ##STR00013## or a pharmaceutically acceptable salt thereof.

3. The composition of claim 1, wherein the compound is: ##STR00014## or a pharmaceutically acceptable salt thereof.

4. The composition of claim 1, further comprising a pharmaceutically acceptable carrier.

5. The composition of claim 2, further comprising a pharmaceutically acceptable carrier.

6. The composition of claim 3, further comprising a pharmaceutically acceptable carrier.
Description



FIELD OF THE INVENTION

The present invention concerns anti-angiogenesis compositions and methods, and particularly thalidomide analogs that actively inhibit angiogenesis in humans and animals.

BACKGROUND OF THE INVENTION

Angiogenesis is the formation of new blood vessels from pre-existing vessels. Angiogenesis is prominent in solid tumor formation and metastasis. A tumor requires formation of a network of blood vessels to sustain the nutrient and oxygen supply for continued growth. Some tumors in which angiogenesis is important include most solid tumors and benign tumors, such as acoustic neuroma, neurofibroma, trachoma, and pyogenic granulomas. Prevention of angiogenesis could halt the growth of these tumors and the resultant damage due to the presence of the tumor.

It has been shown that there is a direct correlation between tumor microvessel density and the incidence of metastasis. Tumor cells themselves can produce factors that stimulate the proliferation of endothelial cells and new capillary growth. Angiogenesis is important in two stages of tumor metastasis. The first stage where angiogenesis stimulation is important is in the vascularization of the tumor, which allows tumor cells to enter the blood stream and to circulate throughout the body. After the tumor cells have left the primary site, and have settled into the secondary, metastasis site, angiogenesis must occur before the new tumor can grow and expand. Therefore, prevention of angiogenesis could lead to the prevention of metastasis of tumors and possibly contain the neoplastic growth at the primary site. These observations have led to the investigation of anti-angiogenic agents as possible therapeutic options for various cancers.

In the 1950's, thalidomide was marketed as a sedative in Europe but was withdrawn from the market when it was found to be a potent teratogen. Recently, thalidomide has been promoted as a possible inhibitor of angiogenesis. Studies have indicated, however, that thalidomide itself is not sufficiently active to inhibit angiogenesis. Instead, the anti-angiogenic activity or effects previously attributed to thalidomide are the resulting effects of compounds that are only present following metabolic activation of thalidomide (i.e., "active" thalidomide metabolites). D'Amato, R.; Loughman Flynn, E.; Folkman, J., Thalidomide as an Inhibitor of Angiogenesis. Proc. Nat'l. Acad. Sci., 1994, 91, 4082-4085; M.; Bauer, K.; Dixon, S.; Figg, W. Inhibition of Angiogenesis by Thalidomide Requires Metabolic Activation, Which Is Species-dependent. Biochem. Pharmacology, 1998, 55, 1827-1834. Accordingly, it has been speculated that certain metabolites of thalidomide rather than thalidomide itself are responsible for its anti-angiogenic properties. However, the specific thalidomide metabolites responsible for the anti-angiogenic properties have not yet been isolated and identified.

There are hundreds, if not thousands of compounds formed as a result of metabolism of thalidomide and the actively metabolized products of hydrolysis compounds of the thalidomide. Many of the thalidomide metabolites are inactive and/or unstable. There is no way to predict which metabolite(s) will have superior anti-angiogenic properties. As such, "active" thalidomide metabolites (or "active" thalidomide analogs) having superior anti-angiogenic properties are not yet available.

If the anti-angiogenic activity can be attributed to one or a small number of thalidomide metabolites and those metabolites could be isolated and identified, then active thalidomide analogs may be synthesized to provide exceptionally effective compounds inhibiting angiogenic effects. This is especially true when comparing thalidomide to "active" thalidomide analogs. To obtain such active compounds from thalidomide, thalidomide must first be activated via metabolism; only a very small amount of thalidomide would actually be metabolized to one or more "active" metabolites. Further, it may be possible to administer such "active" thalidomide analogs in lower amounts and still achieve the desired anti-angiogenic effects. Moreover, such "active" thalidomide analogs could be safer than thalidomide in avoiding undesirable side effects, e.g., teratogenicity or neurotoxicity, and may be more specific to tumor angiogenesis than thalidomide-thalidomide has a host of undesirable biological activities.

Accordingly, there is a need for isolation and identification of the thalidomide metabolites having superior anti-angiogenic properties. Further, there is a need for the synthesis of purified thalidomide analogs that can mimic the effects of the isolated and identified thalidomide metabolites that display such anti-angiogenic activity. In addition, there is a need for a method for treating undesired angiogenesis using such active thalidomide analogs.

SUMMARY OF THE INVENTION

The present invention provides compounds having superior anti-angiogenic properties. More specifically, a number of thalidomide metabolites having superior anti-angiogenic properties have now been isolated and identified. Accordingly, the present invention provides active thalidomide analogs that mimic the effects of the isolated and identified active thalidomide metabolites, and variations of such thalidomide analogs. Such thalidomide analog compounds of the present invention show enhanced potency in the inhibition of undesirable angiogenesis.

The present method further provides for inhibiting unwanted angiogenesis in a human or animal by administering to the human or animal with the undesired angiogenesis a composition comprising an effective amount of active thalidomide analog of the present invention. Specifically, the invention includes a method of inhibiting angiogenesis by exposing the mass having the undesirable angiogenesis to an angiogenesis inhibiting amount of one or more of the present invention thalidomide analogs (and variations of the same) or pharmaceutically acceptable salts of such compounds, wherein such thalidomide analogs (and variations of the same) have the following general formula (Formula A):

##STR00001## wherein R1 is a hydrogen when R2 is methyl alcohol, a branched or unbranched alkyl alcohol, alkyl acid or amino acid, alkylamine, substituted cycloalkyl, substituted alkylphenyl, or phenylalkyl or R1 is a hydroxyl group, a substituted or unsubstituted cycloalkyl aryl or heteroaryl when R2 is a hydrogen, methyl alcohol, a branched alkylalcohol, alkyl acid, amino acid, alkylamino, substituted cycloalkyl, substituted phenylalkyl or alkylphenyl. Additionally, the phthalimid moiety may be replaced by bicyclo [2,2,1]hepten-icarboxylicimid.

In another embodiment the thalidomide analogs (and variations of the same) have the following general formula (Formula B):

##STR00002## wherein R4 through R7 are fluoride or are another halogen, R4 through R7 may comprise the same or different halogens or R5 and R6 are hydrogen and R4 and R7 are methyl groups. Alternatively, there may be substitutions on the isoindole ring, e.g., R4 through R7 may comprise different groups on the isoindole ring to obtain 4-chloro; 4-nitro; 5,6-dichloro; 4-methyl; 5-methyl; 5,6-dimethyl; and 4,5,6,7-tetrachloro. Further, the isoindole ring may be replaced with succinimides or maleimides. Additionally, other halogens may be substituents at the phenyl ring. For example, rather than 2,4-fluoro, the following groups may be substituents on the phenyl ring: 2,3-difluoro; 2,5-difluoro; 2,6-difluoro; 3,4-difluoro; 3,5-difluoro; 2,3-dichloro; 2,4-dichloro; 2,4-dichloro; 2,6-dichloro; 2,4-dibromo; 2,5-dibromo; 2,6-dibromo; 2-fluoro; 3-fluoro; 4-fluoro; 2-chloro; 3-chloro; 4-chloro; 2-bromo; 3-bromo; 4-bromo; 2,3,4-trifluoro. Additionally, the phthalimid moiety may be replaced by bicyclo [2,2,1]hepten-icarboxylicimid.

In another aspect of the invention, the thalidomide analogs (and variations of the same) have the following general formula (Formula C):

##STR00003## wherein R8 through R11 are hydrogen, R12 is an alkyl residue, R13 is a double-bonded oxygen, sulfur or nitrogen, and R14 is an alkyl, cycloalkyl, substituted phenyl, or a cyclic alkyl, such as cyclo-hexane or wherein R8 through R11 are fluoride or are one or more other halogens, R12 is an alkyl residue, R13 is a double-bonded oxygen, sulfur or nitrogen and R14 is benzene or wherein R8 through R11 are fluoride or are one or more other halogens, and R12 through R14 are hydrogen. In addition, the N--H may be substituted by R15, wherein R15 is an alkylamine, substituted cycloalkyl, substituted alkylphenyl or phenylalkyl, methyl alcohol, branched or unbranched alkyl alcohol, alkyl acid or amino acid.

The invention also includes pharmaceutical compositions that include one or more of the compounds of the present invention, or pharmaceutically acceptable salts thereof, and pharmaceutically acceptable carriers. Further, it is to be understood that the compounds included as part of the present invention shown generally in Formulas A-C above, but include all other compounds that are members of the genus described by such general formulas.

Examples of a couple specific thalidomide analog compounds (having superior anti-angiogenic activity) that are members of the genus of Formula A of the present invention are

##STR00004## i.e., 2-(5-hydroxy-2,6-dioxo-piperidin-3-yl)-1H-isoindole-1,3[2H]-dione, and

##STR00005## i.e., 2-(1-hydroxymethyl-2,6-dioxo-piperidin-3-yl)-1,3-dihydro-2H-isoindole-1,3- -dione.

Examples of some specific thalidomide analog compounds (having superior anti-angiogenic activity) that are members of the genus of Formula B of the present invention are

##STR00006## i.e., 2-(2,4-difluorophenyl)-4,7-dimethyl-1H-isoindole-1,3(2H)-dione, and

##STR00007## i.e., 2-(2,4-difluorophenyl)-4,5,6,7-tetrafluoro-1H-isoindole-1,3(2H)-dione.

Examples of specific thalidomide analog compounds (having superior anti-angiogenic activity) that are members of the genus of Formula C of the present invention are

##STR00008## i.e., 1-cyclohexyl-5-ethyl-phthalimidobarbituric acid, and

##STR00009## i.e., 5-ethyl-1-phenyl-5-(tetrafluorophthalimido)barbituric acid, and

##STR00010## i.e., 5-(tetrafluorophthalimido)pyrimidine-2,4(1H,3H)-dione.

The invention also includes pharmaceutical compositions that include one or more of the above-described compounds.

The foregoing and other objects, features, and advantages of the invention will become more apparent from the following detailed description of particular examples that proceed with reference to the accompanying figures.

BRIEF DESCRIPTION OF THE FIGURES

FIG. 1 is a photomicrograph of a control comprising a rat aorta ring treated with DMSO.

FIG. 2 is a photomicrograph of a rat aorta ring treated with CPS3.

FIG. 3 is a photomicrograph of a rat aorta ring treated with about 100 .mu.M of CPS11.

FIG. 4 is a photomicrograph of another rat aorta ring treated with about 100 .mu.M of CPS11.

FIG. 5 is a photomicrograph of another rat aorta ring treated with about 100 .mu.M of CPS11.

FIG. 6 is a photomicrograph of a rat aorta ring treated with about 100 .mu.M of CPS44.

FIG. 7 is a photomicrograph of a rat aorta ring treated with about 100 .mu.M of CPS45.

FIG. 8 is a photomicrograph of another rat aorta ring treated with about 100 .mu.M of CPS48.

FIG. 9 is a photomicrograph of another rat aorta ring treated with about 100 .mu.M of CPS49.

DETAILED DESCRIPTION OF PARTICULAR EXAMPLES

Definitions

The term "halogen" refers to fluoro, bromo, chloro and iodo substituents.

A "pharmaceutical agent" or "drug" refers to a chemical compound or composition capable of inducing a desired therapeutic or prophylactic effect when properly administered to a subject.

The pharmaceutically acceptable salts of the compounds of this invention include those formed from cations such as sodium, potassium, aluminum, calcium, lithium, magnesium, zinc, and from bases such as ammonia, ethylenediamine, N-methyl-glutamine, lysine, arginine, ornithine, choline, N,N'-dibenzylethylenediamine, chloroprocaine, diethanolamine, procaine, N-benzylphenethylamine, diethylamine, piperazine, tris(hydroxymethyl)aminomethane, and tetramethylammonium hydroxide. These salts may be prepared by standard procedures, for example by reacting the free acid with a suitable organic or inorganic base. Any chemical compound recited in this specification may alternatively be administered as a pharmaceutically acceptable salt thereof.

All chemical compounds include both the (+) and (-) stereoisomers, as well as either the (+) or (-) stereoisomer.

A thalidomide "analog" as used herein is a synthetic chemical compound using the thalidomide structure as a backbone (i.e., side groups have been added or such groups have been deleted from the parent structure). The analog differs in structure from thalidomide and its metabolite compounds such as by a difference in the length of an alkyl chain, a molecular fragment, by one or more functional groups, or a change in ionization. Thalidomide analogs generally are not naturally occurring compounds. That is, thalidomide analogs generally cannot be enzymatically or nonenzymatically formed in the body by administration of thalidomide.

A thalidomide "metabolite" is a thalidomide derivative that is formed by enzymatic action, i.e., metabolism of thalidomide in the body. The metabolite is formed by phase-one reactions (e.g., oxidation, reduction, and hydrolysis) or by phase-two reactions (e.g., conjugations). Thalidomide metabolites require an enzyme reaction to be produced.

"Angiogenesis" refers to the development of blood vessels. Accordingly, "anti-angiogenic activity" refers to the inhibition and/or complete cessation of angiogenesis.

"Tumor" refers to a mass of cells resulting from excessive cellular multiplication.

The term "halogen" refers to fluoro, bromo, chloro and iodo substituents.

The term "alcohol" refers to any member of a class of organic compounds in which a hydrogen atom of a hydrocarbon has been replaced by a hydroxy (--OH) group. Unless otherwise mentioned, such an alcohol contains one to twelve carbon atoms.

The term "acid" refers to a compound capable of transferring a hydrogen atom in solution.

The term "alkyl" refers to a cyclic, branched, or straight chain alkyl group containing only carbon and hydrogen, and unless otherwise mentioned contains one to twelve carbon atoms. This term is further exemplified by groups such as methyl, ethyl, n-propyl, isobutyl, t-butyl, pentyl, pivalyl, heptyl, adamantyl, and cyclopentyl. Alkyl groups can either be unsubstituted or substituted with one or more substituents, e.g., halogen, alkyl, alkoxy, alkylthio, trifluoromethyl, acyloxy, hydroxy, mercapto, carboxy, aryloxy, aryloxy, aryl, arylalkyl, heteroaryl, amino, alkylamino, dialkylamino, morpholino, piperidino, pyrrolidin-1-yl, piperazin-1-yl, or other functionality.

The term "amino acid" refers to any of the organic compounds that contain one or more basic amino groups (--NH.sub.2) and one or more acidic carboxyl groups (--COOH) and that are polymerized to form peptides and proteins.

The term "aryl" refers to a monovalent unsaturated aromatic carbocyclic group having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl), which can optionally be unsubstituted or substituted with, e.g., halogen, alkyl, alkoxy, mercapto (--SH), alkylthio, trifluoromethyl, acyloxy, hydroxy, mercapto, carboxy, aryloxy, aryl, arylalkyl, heteroaryl, amino, alkylamino, dialkylamino, morpholino, piperidino, pyrrolidin-1-yl, piperazin-1-yl, or other functionality.

The term "alkyl residue" refers to a branched or straight chain alkyl group containing only carbon and hydrogen, and unless otherwise mentioned contains one to twelve carbon atoms. The term is further exemplified by groups such as methyl, ethyl, n-propyl, isobutyl, pentyl, pivalyl and heptyl. Alkyl groups can either be substituted or unsubstituted.

Other chemistry terms herein are used according to conventional usage in the art, as exemplified by The McGraw-Hill Dictionary of Chemical Terms (1985), The Condensed Chemical Dictionary (1981), and Dorland's Illustrated Medical Dictionary (1974).

A "mammal" includes both human and non-human mammals. Similarly, the term "subject" includes both human and veterinary subjects.

An "animal" is a living multicellular vertebrate organism, a category that includes, for example, mammals and birds.

"Thalidomide" or N-(2,6-dioxopiperidin-3-yl)phthalimide has the following chemical structure:

##STR00011## Materials and Methods

Where necessary, solvents were dried and purified according to the recommended procedures. Organic solutions were dried over NaSO.sub.4. Evaporation refers to removal of solvent on a Vacuubrand rotary evaporator under reduced pressure of from about 200 to about 15 mbar. Melting points were determined using a Boetius apparatus and are uncorrected. .sup.1H NMR spectra (300 Mhz), .sup.13C NMR spectra (75 MHz), and .sup.19F spectra (188 MHz) were recorded on a Varian Gemini 300 spectrometer with tetramethylsilane as internal standard; the values of chemical shifts (.delta.) are given in ppm and coupling constants (J) in Hz. Mass spectral data were determined by direct insertion at 70 eV with a Varian MAT CH6 spectrometer as well as a HP-MS Engine 5989A. Yields refer to purified products and are not optimized.

Compound Reference Numbers

Compounds are identified throughout this detailed description using alpha-numeric references in bold, which correspond to the identification of the compounds as set forth in the Summary of the Invention, and in the following examples.

EXAMPLE 1

Synthesis of and Analytical Results for 2-(5-hydroxy-2,6-dioxo-piperidin-3-yl)-1H-isoindole-1,3[2H]-dione (CPS3)

This example illustrates the preparation of 2-(5-hydroxy-2,dioxo-piperidin-3(2H)-dione) having a molecular weight of about 274.2. A mixture of about 1.0 g (about 3.2 mmol) of acetic acid 5-(1,3-dioxo-1,3-dihydro-isoindol-2-yl)-2,6-dioxo-piperidin-3-yl-ester (single diastereomer) prepared according to Teubert, U. et al., Arch. Pharm. Pharm. Med. Chem., 1998, 331, 7 (incorporated herein by reference) and about 0.3 g (about 1.6 mmol) of p-toluenesulfonic acid was refluxed in about 30 ml of methanol for about 5 hours. The solution was allowed to cool to about room temperature. After cooling, the precipitated product was filtered and recrystallized from acetone/petroleum ether (having a boiling point of about 60.degree. to about 80.degree. C.). Alternatively, the precipitated, filtered product was recrystallized from acetonitrile. A yield of about 0.52 g (about a 60% yield) of 2-(5-hydroxy-2,6-dioxo-piperidin-3-yl)-1H-isoindole-1,3[2H]-dione having a melting point of about 195.degree. to about 230.degree. C. resulted.

The .sup.1H NMR spectral analysis results were as follows: (DMSO-d6) 2.27-2.53 (m, 2H, 4'-H), 4.53-4.57 (m, 1H, 5'-H), 5.29 (dd, J=13.1, 5.2 Hz, 1H, 3'-H), 5.82 (d, J=6.0 Hz, 1H, OH), 7.90-7.94 (m, 4H, aromatic H), 11.22 (s, 1H, NH). The .sup.13C NMR spectral analysis results were as follows: (DMSO-d6) 31.3 (C-4'), 48.22 (C-3'), 66.33 (C-5'), 123.29, 131.19, 138.88 (C-aromatic), 166.86, 176.17, 169.70, 174.71 (C.dbd.O). Mass spectrometry analysis (EI) results yielded, m/z (relative intensity), 274 (13)[M+].

EXAMPLE 2

Synthesis of and Analytical Results for 2-(1-hydroxymethyl-2,6-dioxo-piperidin-3-yl)-1,3-dihydro-2H-isoindole-1,3- -dione (CPS11)

This example illustrates the preparation and analysis of 2-(1-hydroxymethyl-2,6-dioxo-piperidin-3-yl)-1,3-dihydro-2H-isoindole-1,3- -dione having a molecular weight of about 288.25.

A suspension of about 12.9 g (about 50 mmol) of rac-thalidomide in about 100 mL of an about 35% aqueous formaldehyde solution was refluxed until dissolved. The solution was then allowed to cool to room temperature. After about 24 hours, the precipitate was collected by filtration and washed with about 3% aqueous formaldehyde solution and was then dried with Na.sub.2SO.sub.4. A yield of about 10.1 g (about a 70% yield) of 2-(1-hydroxymethyl-2,6-dioxo-piperidin-3-yl)-1,3-dihydro-2H-isoindole-1,3- -dione having a melting point of about 165.degree. C. resulted.

The .sup.1H NMR spectral analysis results were as follows: (DMSO-d6) 2.16-2.67 (m, 2H, 4'-H), 2.87-3.11 (m, 2H, 5'-H), 5.08 (d, J=7.2 Hz, 2H, NCH.sub.2OH), 5.52 (m, 1H, CHCH.sub.2), 6.17 (t, Jab=7.2 Hz, Jbc=7.2 Hz, 1H, OH), 7.92 (s, 4H, Harom.). Analytical values for the compound C.sub.14H.sub.12N.sub.2O.sub.5 were carbon about 58.27%, hydrogen about 4.09%, and nitrogen about 9.52%.

EXAMPLE 3

Synthesis of and Analytical Results for 2-(2,4-difluorophenyl)-4,7-dimethyl-1H-isoindole-1,3(2H)-dione (CPS42)

This example illustrates the preparation and analysis of 2-(2,4-difluorophenyl)-4,7-dimethyl-1H-isoindole-1,3(2H)-dione having an exact mass of 287.08, a molecular weight of about 287.6 (carbon about 66.90%, hydrogen about 3.86%, fluoride about 13.23%, nitrogen about 4.88%, and oxygen about 11.14%).

A mixture of about 2 g (about 15.5 mmol) of 2,4-difluoroaniline, about 2.46 g (about 14 mmol) of 3,6-dimethylphthalic anhydride, and about 100 mL of glacial acetic acid was refluxed for about 3.5 hours. The 3,6-dimethylphthalic anhydride was prepared according to Newman, M. S.; Lord, B. T., J. Am. Chem. Soc., 1944, 66, 733, which is incorporated herein by reference.

The solvent was evaporated to dryness under reduced pressure of from about 200 to about 15 mbar. The residue was dissolved in about 150 mL of CH.sub.2C.sub.12. The solution was washed three times with about 50 mL of about 0.1 M HCl and twice with about 50 mL of H.sub.2O and was then dried with Na.sub.2SO.sub.4. After removal of the solvent, the residue was recrystallized from ethyl alcohol to yield about 1.27 g (32%) 2-(2,4-difluorophenyl)-4,7-dimethyl-1H-isoindole-1,3(2H)-dione having a melting point of about 212.degree. to about 212.5.degree. C.

The .sup.1H NMR spectral analysis results were as follows: (DMSO-d6) 2.59 (s, 6H), 7.24-7.32 (m, 1H), 7.46-7.56 (m, 1H), 7.54 (s, 2H) 7.56-7.66 (m, 1H). The .sup.13C NMR spectral analysis results were as follows: (DMSO-d6) 16.87 (CH.sub.3), 104.93 (dd, 2J=27.1, 24.2 Hz, C-3'), 112.08 (dd, 2J=22.6, 4J=3.6 Hz, C-5'), 115.93 (dd, 2J=13.2, 4J=3.9 Hz, C-1'), 128.15 (C-4, C-7), 132.07 (dd, 3J=10.1, 2.1 Hz, C-6') 135.37 (C-3a, C-7a), 136.62 (C-5, C-6), 157.81 and 162.08 (d, J=264.1 Hz and d, J=235.6 Hz, C-2' and C-4'), 166.64 (C-1, C-3). Mass spectrometry analysis (EI) results yielded, m/z (relative intensity), 287 (M+, 100), 259 (81c). Analytically calculated values for the compound C.sub.16H.sub.11NO.sub.2F.sub.2 were carbon 66.90%, hydrogen 3.86%, and nitrogen 4.88%. As determined from the NMR and mass spectrometry analysis result values for the compound C.sub.16H.sub.11NO.sub.2F.sub.2 were carbon about 67.20%, hydrogen about 3.77%, and nitrogen about 4.59%.

EXAMPLE 4

Synthesis of and Analytical Results for 1-cyclohexyl-5-ethyl-5-phthalimidobarbituric acid (CPS44)

This example illustrates the preparation and analysis of 1-cyclohexyl-5-ethyl-5-phthalimidobarbituric acid (C.sub.20H.sub.21N.sub.3O.sub.5) having an exact mass of 283.15 and a molecular weight of about 383.4 (carbon about 62.65%, hydrogen about 5.52%, nitrogen about 10.96%, and oxygen about 20.87%). The same compound might alternatively be named, e.g., 1-cyclohexyl-5-(1,3-dioxo-1,3-dihydro-isoindol-2-yl)-5-ethyl-pyrimidine-2- ,4,6(1H,3H,5H)-trione or 1-cyclohexyl-5-(1,3-dioxo-1,3-dihydro-isoindol-2-yl)-5-ethyl-pyrimidine-2- ,4,6-trione.

A mixture of about 500 mg (about 2 mmol) of 5-amino-1-cyclohexyl-5-ethylbarbituric acid available from 5-azido-1-cyclohexyl-5-ethylbarbituric acid as described in Guetschow, M. et al., Synthesis, 1999, 410-414 (incorporated herein by reference) and about 300 mg (about 2 mmol) of phthalic anhydride and about 20 mL of glacial acetic acid was refluxed for about 5 hours. The mixture was cooled to room temperature and the precipitate was filtered off. The precipitate was next washed with H.sub.2O and dried to give about 470 mg of 1-cyclohexyl-5-ethyl-5-phthalimidobarbituric acid (a yield of about 61%). The 1-cyclohexyl-5-ethyl-5-phthalimidobarbituric acid had a melting point of about 248 to about 253.degree. C.

The .sup.1H NMR spectral analysis results were as follows: (DMSO-d.sub.6)* 0.99 (t, 3H, J=7.4 Hz), 1.01-2.21 (m, 10H), 2.68 (q, 2H, J=7.4 Hz), 4.13-4.29 (m, 1H), 7.90 (s, 4H), 12.09 (s, 1H). The .sup.13C NMR spectral analysis results were as follows: (DMSO-d.sub.6)* 9.10, 24.83, 25.62, 25.71, 27.23, 27.92, 29.05, 54.66, 67.83, 123.74, 130.32, 135.56, 149.24, 167.20, 167.79, 168.28. The mass spectral (EI) analysis results were as follows: m/z (relative intensity) 383 (M.sup.+, 5), 302 (M.sup.+, 100), 105 (52). Analytically calculated values for the compound C.sub.20H.sub.21N.sub.3O.sub.5 were carbon 62.65%, hydrogen 5.52%, and nitrogen 10.96%. As determined from the NMR and mass spectrometry analysis results values for the compound C.sub.20H.sub.21N.sub.3O.sub.5 were carbon about 62.30%, hydrogen about 5.85%, nitrogen about 10.89%, and oxygen about 20.87%.

EXAMPLE 5

Synthesis of and Analytical Results for 5-ethyl-1-phenyl-5-(tetrafluorophthalimido)barbituric acid (CPS45)

This example illustrates the preparation and analysis of 5-ethyl-1-phenyl-5-(tetrafluorophthalimido)barbituric acid (C.sub.20H.sub.11N.sub.3O.sub.5F.sub.4) having an exact mass of 377.10 and a molecular weight of about 377.4 (carbon about 63.66%, hydrogen about 4.01%, nitrogen about 11.14%, and oxygen about 21.20%). The same compound might alternatively be named, e.g., 5-(1,3-dioxo-1,3-dihydro-4,5,6,7-tetrafluoro-isoindol-2-yl)-5-ethyl-1-phe- nyl-pyrimidine-2,4,6-(1H,3H,5H)-trione.

A mixture of about 250 mg (about 1 mmol) of 5-amino-5-ethyl-1-phenylbarbituric acid, about 260 mg (about 1.2 mmol) of tetrafluorophthalic anhydride, and about 7 mL of acetic acid was refluxed for about 3 hours. The solvent was evaporated to dryness under reduced pressure of from about 200 to about 15 mbar. The residue was recrystallized from ethyl alcohol to yield about 270 mg 5-ethyl-1-phenyl-5-(tetrafluorophthalimido)barbituric acid (a 60% yield) having a melting point of about 218.degree. to about 220.degree. C.

The .sup.1H NMR spectral analysis results were as follows: (DMSO-d6), 1.09 (t, 3H, J=7.2 Hz), 2.86 (q, 2H, J=7.3 Hz), 7.20-7.32 (m, 2H), 7.45-7.59 (m, 3H). The .sup.13C NMR spectral analysis results were as follows: (DMSO-d6), 9.36, 26.90, 68.10, 112.20, 112.65, 128.05, 128.56, 129.27, 133.91, 141.10, 147.05, 148.88, 162.51, 166.85, 167.18. The mass spectral (EI) analysis results were as follows: m/z (relative intensity), 449 (M.sup.+, 62), 421 (M.sup.+, 20), 230 (39), 176 (70), 119 (100). Analytically calculated values for the compound C.sub.20H.sub.11N.sub.3O.sub.5F.sub.4 were carbon 53.46%, hydrogen 2.47%, and nitrogen 9.35%. As determined from the NMR and mass spectrometry analysis results values for the compound C.sub.20H.sub.11N.sub.3O.sub.5F.sub.4 were carbon about 53.26%, hydrogen about 2.78%, oxygen about 21.20%, and nitrogen about 9.04%.

EXAMPLE 6

Synthesis of and Analytical Results for 5-(tetrafluorophthalimido)pyrimidine-2,4(1H,3H)-dione (CPS48)

This example illustrates the preparation and analysis of 5-(tetrafluorophthalimido)pyrimidine-2,4(1H,3H)-dione (C.sub.12H.sub.3F.sub.4N.sub.3O.sub.4) having an exact mass of 329.01 and a molecular weight of about 329.2 (carbon about 43.79%, hydrogen about 0.92%, fluoride about 23.09%, nitrogen about 12.7%, and oxygen about 19.44%). The same compound might alternatively be named, e.g., 2-(2,4-dioxo-1,2,3,4-tetrahydro-pyrimidin-5-yl)-4,5,6,7-tetrafluoro-1H-is- oindole-1,3(2H)-dione.

A mixture of about 440 mg anhydride (about 2 mmol) of tetrafluorophthalic, about 254 mg (about 2 mmol) of 5-aminouracil, and about 50 mL of glacial acetic acid was refluxed for about 6 hours. The solution was allowed to cool to about room temperature. After cooling, the precipitated product was filtered, washed with water, and dried with Na.sub.2SO.sub.4 to yield about 260 mg (about 40%) of 5-(tetrafluorophthalimido)pyrimidine-2,4(1H,3H)-dione with a melting point of about 3500 to about 355.degree. C.

The .sup.1H NMR spectral analysis results were as follows: (DMSO-d6) 7.89 (d, 1H, J=6.2 Hz), 11.47 (d, 1H, J=6.2 Hz), 11.72 (s, 1H). The .sup.19F NMR spectral analysis results were as follows: (DMSO-d6/CFCl3) -142.0 (m, 2F), -136.5 (m, 2F). The mass spectral (EI) analysis results were as follows: m/z (relative intensity), 329 (M.sup.+, 50), 176 (100). Analytically calculated values for the compound C.sub.12H.sub.3F.sub.4N.sub.3O.sub.4 were carbon about 43.79%, hydrogen about 0.92%, and nitrogen about 12.77%. As determined from the NMR and mass spectrometry analysis result values for the compound C.sub.12H.sub.3F.sub.4N.sub.3O.sub.4 were carbon about 43.60%, hydrogen about 1.16%, nitrogen about 12.50%.

EXAMPLE 7

Synthesis of and Analytical Results for (CPS49) 2-(2,4-difluoro-phenyl)-4,5,6,7-tetrafluoro-1H-isoindole-1,3(2H)-dione

This example illustrates the preparation and analysis of 2-(2,4-difluoro-phenyl)-4,5,6,7-tetrafluoro-1H-isoindole-1,3(2H)-dione (C.sub.14H.sub.3F.sub.6NO.sub.2) having an exact mass of 331.01 and a molecular weight of about 331.2 (carbon about 50.77%, hydrogen about 0.91%, fluoride about 34.42%, nitrogen about 4.23%, and oxygen about 9.66%).

A mixture of about 1 g (about 7.75 mmol) of 2,4-difluoroaniline, about 1.54 g (about 7 mmol) of tetrafluorophthalic anhydride and about 50 mL of glacial acetic acid was refluxed for about 3.5 hours. The solvent was evaporated to dryness under reduced pressure of from about 200 to about 15 mbar. The residue was dissolved in about 75 mL of CH.sub.2C.sub.12. The solution was washed three times with about 25 mL of about 0.1 M HCl and twice with about 25 mL of water. The residue was then dried with Na.sub.2SO.sub.4. After removal of the solvent, the residue was recrystallized from ethyl alcohol to yield about 980 mg (about 42%) of 2-(2,4-difluorophenyl)-4,5,6,7-tetrafluoro-1H-isoindole-1,3(2H)-dione with a melting point of about 145.degree. to about 146.degree. C.

The .sup.1H NMR spectral analysis results were as follows: (DMSO-d6) 7.22-7.38 (m, 1H), 7.50-7.66 (m, 2H). The .sup.19F NMR spectral analysis results were as follows: (DMSO-d6/CFCl3) -142.3 (m, 2F), -136.9 (m, 2F), -113.8 (m, 1F), -105.6 (m, 1F). The mass spectral (EI) analysis results were as follows: m/z (relative intensity), 331 (M.sup.+, 65), 287 (68), 148 (100). Analytically calculated values for the compound C.sub.14H.sub.3NO.sub.2F.sub.6 were carbon about 50.78%, hydrogen about 0.91%, and nitrogen about 4.23%. As determined from the NMR and mass spectrometry analysis results values for the compound C.sub.14H.sub.3NO.sub.2F.sub.6 were carbon about 50.60%, hydrogen about 0.83%, and nitrogen about 3.95%.

EXAMPLE 8

HUVEC MTT Assay for Selected Present Invention Thalidomide Analogs

HUVEC MTT assays were performed for the selected thalidomide analogs of the present invention to determine a rough estimate of the efficacy of such compounds in the inhibition of angiogenesis. For the MTT assay, 1.0 to 2.5.times.10.sup.3 cells per well were plated in 96-well plates in 0.1 ml medium, in triplicate. After 24 hours, the cells were exposed to treatment for 5 days. One plate was analyzed every 24 hours by the addition of 20 .mu.L of 5 mg/ml MTT solution (available from Sigma of St. Louis, Mo.) in PBS, to each well for 4 hours. The MTT solution was aspirated and 170 .mu.L DMSO was added to each well to dissolve the formazan crystals. The absorbance at 540 nm was measured using a Biokinetics plate reader (available from Bio-Tek Instruments of Winooski, Vt.). Triplicate wells were assayed for each condition. The assay protocol described herein stems from Kruger et al., a protein kinase C inhibitor, inhibits endothelial cell proliferation and angiogenic hypoxic response, Invasion and Metastasis, 18(4): 209-218 (incorporated herein by reference).

The following results for the selected compounds were determined measured utilizing growth curves comparing control wells to treated wells using the MTT assays.

2-(5-hydroxy-2,6-dioxo-piperidin-3-yl)-1H-isoindole-1,3[2H]-dione (CPS 3)--No cytostatic activity was noted at either 100 .mu.M or at 10 .mu.M.

2-(1-hydroxymethyl-2,6-dioxo-piperidin-3-yl)-1,3-dihydro-2H-isoindole-1,3- -dione (CPS11)--Potent inhibition (>90%) at 100 .mu.M was found at about 72 hours. An inhibition of about 60% was found at 10 .mu.M was noted at about 72 hours.

2-(2,4-difluorophenyl)-4,7-dimethyl-1H-isoindole-1,3(2H)-dione (CPS42)--An inhibition of about 44% was found at 100 .mu.M but no such activity was found at a level of about 10 .mu.M.

1-cyclohexyl-5-ethyl-5-phthalimidobarbituric acid (CPS44)--An inhibition of about 50% was found at 100 .mu.M but no such activity was found at a level of about 10 .mu.M.

5-ethyl-1-phenyl-5-(tetrafluorophthalimido)barbituric acid (CPS45)--Potent inhibition (>90%) at 100 .mu.M and at 10 .mu.M was noted.

5-(tetrafluorophthalimido)pyrimidine-2,4(1H,3H)-dione (CPS48) Potent inhibition (about 85%) at 100 .mu.M and at 10 .mu.M was noted.

2-(2,4-difluoro-phenyl)-4,5,6,7-tetrafluoro-1H-isoindole-1,3(2H)-dione (CPS49)--A potent inhibition of about 90% was found at 100 .mu.M but no such activity was found at a level of about 10 .mu.M.

EXAMPLE 9

Anti-angiogenic Activity Analysis Results for Selected Present Invention Thalidomide Analogs Measured Utilizing Rat Aortic Rings

The efficacy of selected thalidomide analogs of the present invention was studied by five-day treatment of rat aortic rings (utilizing Sprague-Dawley rats available from Charles River Labs) with varied doses of the analogs. A DMSO control was utilized (FIG. 1). The results, determined using image analysis comparing control rings versus the treated rings (using NIM Image software), of the studies are as follows:

2-(5-hydroxy-2,6-dioxo-piperidin-3-yl)-1H-isoindole-1,3[2H]-dione (CPS3)--A daily dosage of 100 .mu.M showed about a 50% angiogenesis inhibition activity (FIG. 2).

2-(1-hydroxymethyl-2,6-dioxo-piperidin-3-yl)-1,3-dihydro-2H-isoindole-1,3- -dione (CPS11)--A daily dosage of 100 .mu.M showed a potent 90% angiogenesis inhibition activity (FIGS. 3-5).

1-cyclohexyl-5-ethyl-5-phthalimidobarbituric acid (CPS44)--No inhibition of angiogenesis was found at 100 .mu.M (FIG. 6).

5-ethyl-1-phenyl-5-(tetrafluorophthalimido)barbituric acid (CPS45) --A daily dosage of 100 .mu.M showed a potent 90% angiogenesis inhibition activity (FIG. 7).

5-(tetrafluorophthalimido)pyrimidine-2,4(1H,3H)-dione (CPS48)--A daily dosage of 100 .mu.M showed a potent 90% angiogenesis inhibition activity (FIG. 8).

2-(2,4-difluoro-phenyl)-4,5,6,7-tetrafluoro-1H-isoindole-1,3(2H)-dione (CPS49)--A daily dosage of 100 .mu.M showed extensive angiogenesis inhibition activity (FIG. 9).

EXAMPLE 10

Anti-angiogenic Activity Analysis Results for 2-(1-hydroxymethyl-2,6-dioxo-piperidin-3-yl)-1,3-dihydro-2H-isoindole-1,3- -dione (CPS11) Measured Utilizing Human Saphenous Vein

The efficacy of 2-(1-hydroxymethyl-2,6-dioxo-piperidin-3-yl)-1,3-dihydro-2H-isoindole-1,3- -dione (CPS11) was studied by 14 day treatment of human saphenous veins (obtained through an IRB-approved protocol, Surgery Brand NCI) with 100 .mu.M doses of the analog. A CAI, carboxyamido-triazole, 12 .mu.g/ml control was utilized. The results of such studies using image analysis as discussed above, indicate that daily dosages of 100 .mu.M of 2-(1-hydroxymethyl-2,6-dioxo-piperidin-3-yl)-1,3-dihydro-2H-isoindole-1,3- -dione (CPS11) showed a potent 90% angiogenesis inhibition activity level.

EXAMPLE 11

Toxicology Screen Analysis Results for Selected Present Invention Thalidomide Analogs

Toxicology screen studies have been performed for the thalidomide analogs of the present invention. The results of such toxicology screening studies for selected thalidomide analogs are as follows:

2-(1-hydroxymethyl-2,6-dioxo-piperidin-3-yl)-1,3-dihydro-2H-isoindole-1,3- -dione (CPS11)--Treatment was safe at dosage levels of 10 and 100 mg/kg, i.p., single dose. Some amount of sedation was noted.

2-(2,4-difluorophenyl)-4,7-dimethyl-1H-isoindole-1,3(2H)-dione (CPS42)--Treatment was safe at a dosage level 200 mg/kg, i.p., single dose. Slight sedation was noted within 15 minutes of injection.

5-ethyl-1-phenyl-5-(tetrafluorophthalimido)barbituric acid (CPS45) --A dose of 200 mg/kg, i.p., single dose, was a lethal dose. Animals treated with such dosage died within 2.5 hours of treatment.

2-(2,4-difluoro-phenyl)-4,5,6,7-tetrafluoro-1H-isoindole-1,3(2H)-dione (CPS49)--A dose of 200 mg/kg, i.p., single dose, was a lethal dose. Animals treated with such dosage died within 18 hours of treatment.

EXAMPLE 12

Methods of Treatment

The present invention includes a treatment for undesirable angiogenesis and angiogenesis dependent or associated diseases, in a subject such as an animal, for example a rat or human. The method includes administering one or more of the compounds of the present invention, or a combination of one or more of the compounds and one or more other pharmaceutical agents, to the subject in a pharmaceutically compatible carrier. The administration is made in an amount effective to inhibit the development or progression of angiogenesis and diseases associated with the same. Although the treatment can be used prophylactically in any patient in a demographic group at significant risk for such diseases, subjects can also be selected using more specific criteria, such as a definitive diagnosis of the condition.

The vehicle in which the drug is delivered can include pharmaceutically acceptable compositions of the drugs, using methods well known to those with skill in the art. Any of the common carriers, such as sterile saline or glucose solution, can be utilized with the drugs provided by the invention. Routes of administration include but are not limited to oral and parenteral routes, such as intravenous (iv), intraperitoneal (ip), rectal, topical, ophthalmic, nasal, and transdermal.

The drugs may be administered in a suitable manner now known or later developed, e.g., orally or intravenously, in any conventional medium. For example, intravenous injection may be by an aqueous saline medium. The medium may also contain conventional pharmaceutical adjunct materials such as, for example, pharmaceutically acceptable salts to adjust the osmotic pressure, lipid carriers such as cyclodextrins, proteins such as serum albumin, hydrophilic agents such as methyl cellulose, detergents, buffers, preservatives and the like. A more complete explanation of parenteral pharmaceutical carriers can be found in Remington: The Science and Practice of Pharmacy (19.sup.th Edition, 1995) in chapter 95.

Embodiments of other pharmaceutical compositions can be prepared with conventional pharmaceutically acceptable carriers, adjuvants and counterions as would be known to those of skill in the art. The compositions are preferably in the form of a unit dose in solid, semi-solid and liquid dosage forms such as tablets, pills, powders, liquid solutions or suspensions.

The compounds of the present invention are ideally administered as soon as possible after detected unwanted angiogenesis. For example, once unwanted angiogenesis has been confirmed or the presence of a tumor has been identified, a therapeutically effective amount of the drug is administered. The dose can be given orally or by frequent bolus administration.

Therapeutically effective doses of the compounds of the present invention can be determined by one of skill in the art, with a goal of achieving a desired level of anti-angiogenesis as illustrated in the foregoing examples. The relative toxicities of the compounds make it possible to administer in various dosage ranges. An example of such a dosage range is from about 0.5 to about 50 mg/kg body weight orally in single or divided doses. Another example of a dosage range is from about 0.5 to about 50 mg/kg body weight orally in single or divided doses. For oral administration, the compositions are, for example, provided in the form of a tablet containing from about 25 to about 500 mg of the active ingredient, particularly 100 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject being treated.

The specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors, including the activity of the specific compound, the extent of existing angiogenic activity, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, and severity of the condition of the host undergoing therapy.

The pharmaceutical compositions can be used in the treatment of a variety of diseases mediated by angiogenesis. Examples of such diseases include all types of cancer, ocular neovascular disease, solid tumor formation and metastasis in solid tumors such as rhabdomyosarcomas, retinoblastoma, Ewing sarcoma, neuroblastoma, osteosarcoma, colon, prostate, head and neck, breast, bladder, liver, pancreatic, lung, CNS, and blood-born tumors such as leukemia, also diseases such as hemangioma, ulcerative colitis, Crohn's disease, diabetic retinopathy, macular degeneration, sickle cell anemia, sarcoid, syphilis, pseudoxanthoma elasticum, Paget's disease, vein occlusion, artery occlusion, carotid obstructive disease, chronic uveitis/vitritis, mycobacterial infections, Lyme's disease, systemic lupus erythematosis, retinopathy of prematurity, Eale's 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.

EXAMPLE 13

Combination Therapy

The present invention also includes combinations of the thalidomide analogs of the present invention and/or combination of the same with various other angiogenesis inhibitor compounds. For example, the compounds of this invention may be administered in combination with effective doses of other anti-angiogenic agents. The term "administration" refers to both concurrent and sequential administration of the active agents. Examples of anti-angiogenic agents that can be used in combination with the thalidomide analogs of the present invention are TNP-470, carbonic anhydrase inhibitors, endostatin, angiostatin, 2-methoxyestradiol, IMiD (Immune-modulating inhibitor drug) CC5013, matrix metalloproteinase inhibitors, and COL-3. In addition, the thalidomide analogs of this invention may be used in combination with other forms of cancer therapy, e.g., chemotherapy, radiation therapy, hormonal therapy).

In view of the many possible embodiments to which the principles of our invention may be applied, it should be recognized that the illustrated embodiment is only a preferred example of the invention and should not be taken as a limitation on the scope of the invention. Rather, the scope of the invention is defined by the following claims. We therefore claim as our invention all that comes within the scope and spirit of these claims.

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed