![]() PHARMACEUTICAL COMPOSITIONS UNDERSTANDING SELECTIVE BCL-2 INHIBITORS
专利摘要:
Pharmaceutical compositions comprising selective bcl-2 inhibitors This invention is directed to methods of treating systemic lupus erythematosus, lupus nephritis or sjogren's syndrome with compounds that selectively inhibit the activity of bcl-2 anti-apoptotic proteins. Specifically, the present invention is directed to treatment with compounds that selectively inhibit the activity of bcl-2 proteins, with a lower affinity for inhibiting the activity of other bcl-2 family proteins, including bcl-x1. 公开号:BR112013012854B1 申请号:R112013012854-2 申请日:2011-11-22 公开日:2019-08-06 发明作者:Steven Elmore;Andrew Souers;Li Chun Wang;Tariq Ghayur;Stuart J. Perper 申请人:Abbvie Ireland Unlimited Company; IPC主号:
专利说明:
"PHARMACEUTICAL COMPOSITIONS UNDERSTANDING SELECTIVE BCL-2 INHIBITORS" RELATED ORDER INFORMATION This application claims the benefit of U.S. Application No. 61 / 416,689, filed on November 23, 2010, content of which is incorporated herein for reference. FIELD OF THE INVENTION This invention pertains to methods of treating systemic lupus erythematosus, lupus nephritis or Sjogren's Syndrome with compounds that selectively inhibit the activity of anti-apoptotic Bcl-2 proteins. Specifically, the current invention is directed to treatment with compounds that selectively inhibit the activity of Bcl-2 proteins, with a lower affinity for inhibiting the activity of other proteins of the BCL2 family, including Bcl- x L. BACKGROUND OF THE INVENTION Anti-apoptotic Bcl-2 proteins are associated with a wide range of diseases. There is, therefore, a need in the therapeutic arts for compounds that inhibit the activity of anti-apoptotic Bcl-2 proteins. The Bcl-2 family of proteins is the key regulator of mitochondria-dependent apoptosis in nucleated cells and consists of both anti-apoptotic members (Bcl-x L , Bcl-2, Bcl-w, A1, Mcl-1) and pro- apoptotic (Bak, Bax, Bid, Bim, Bad, Bik, Bmf, Noxa, Puma). Generally, Bcl-2 protein expression is associated with several physiological functions, including inhibition of apoptosis in the body, in some cases resulting in the proliferation of cells affected by Bcl-2 inhibition. As such, inhibition of Bcl-2 protein can reduce cell proliferation, leading to better results related to cancer treatment and prevention. The involvement of Bcl-2 proteins in bladder cancer, brain cancer, breast cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia, follicular lymphoma, cell lymphoid malignancies T or B cell origin, melanoma, myeloid leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, and the like are described in the PCT US 2004/36770 of common ownership, published as WO 2005/049593, and PCT US 2004/37911, published as WO 2005/024636. SUMMARY OF THE INVENTION One embodiment of the current invention belongs to a method for the treatment of systemic lupus erythematosus (SLE) or lupus nephritis by administering a therapeutically effective amount of a compound that selectively inhibits Bcl-2 proteins. Selective Bcl-2 inhibitors generally have the following Formula (I): 2/55 (D. Where A 1 is N or CH; B 1 is OR 1 or NHR 1 ; Γ is CN, NO 2i CF 3 , F or Cl; R 1 is (CH 2 ) n R 2 ; R 2 is cycloalkyl or heterocycle; where the heterocycle and cycloalkyl are optionally substituted by one or more independently selected R 4 , OR 4 , OH, CN, or F; R 3 is heteroaryl; where the heteroaryl is optionally substituted by one or more independently selected NH 2 , Cl, or F; R 4 is alkyl, cycloalkyl, heterocycle, or spiroheterocyclic; where the alkyl is optionally substituted by one or more F; R 5 is deuterium; each R 6 is independently selected from CH 3 , spiroheterocyclic and OH; m is 0, 1, 2, 3, 4, 5, or 6; n is 0 or 1; and p is 0, 1, or 2. The method for treating SLE or lupus nephritis may also comprise administering a pharmaceutically acceptable salt of a selective Bcl-2 inhibitor. Generally, the selective Bcl-2 inhibitor has a binding affinity (K |) of less than about 1 nanomolar for Bcl-2. In another embodiment, a selective Bcl-2 inhibitor has a binding affinity (K |) of less than about 100 picomolar for Bcl-2. A selective Bcl-2 inhibitor may also have a binding affinity (K |) for Bcl-2 that is approximately 3/55 t and 500 times less than the binding affinity for Bcl-x L. In this embodiment, the selective Bcl-2 inhibitor may include N - ({5-chloro-6 - [(4-fluorotetrahydro-2H-pyran-4-yl) methoxy] pyridin- 3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -2 - [(6 - fluoro-1H-indazol-4-yl) oxy] benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1ii] metii} piperazin-1-yl) -N - {[4 - ({[(2S) -4 -cyclopropylmorpholin-2-yl] methyl} amino) -3-nitrophenyl] sulfonyl} - 2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; N - ({5-chloro-6 - [(4-fluorotetrahydro-2H-pyran-4yl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -2- (1 H-indazol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1 -yl) -2 - [(6-fluoro-1 H-indole -5-yl) oxy] -N - ({4 - [(4fluorotetrahydro-2H-pyran-4-yl) methoxy] -3-nitrophenyl} sulfonyl) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(4 - {[(4,4-difluorocyclohexyl ) methyl] amino} -3nitrophenyl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2- (1H-benzimidazol-4-yloxy) -4 (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - ({5-fluoro-6 - [(4fluorotetrahydro-2H-pyran-4-yl) methoxy] pyridin-3-yl} sulfonyl) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - ({3-nitro-4 - [(tetrahydro- 2H-pyran-4ylmethyl) amino] phenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; N - ({3-chloro-4 - [(4fluorotetrahydro-2H-pyran-4-yl) methoxy] phenyl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex- 1en-1-yl] methyl} piperazin-1-yl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2- (1H-benzimidazole- 4-yloxy) -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(4- { [(4-cyanocyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] benzamide; N - ({5-chloro-6 - [(cis-4-hydroxy-4methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4- dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -2- (1H-indazol-4-yloxy) benzamide; N - [(3-chloro-4 - {[4-fluoro-1- (oxetan-3yl) piperidin-4-yl] methoxy} phenyl) sulfonyl] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - ({5-cyano-6 - [(4- fluorotetrahydro-2H-pyran-4yl) methoxy] pyridin-3-yl} sulfonyl) -2- (1H-indol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1 -yl) -N - [(4 - {[(4-fluorotetrahydro-2H -pyran-4yl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; N - ({3-chloro4 - [(4-fluorotetrahydro-2H-pyran-4-yl) methoxy] phenyl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1- en-1-yl] methyl} piperazin-1-yl) -2- (1H-indazol-4-yloxy) benzamide; 4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - ({5-fluoro-6 - [(4- fluorotetrahydro2H-pyran-4-yl) methoxy] pyridin-3-yl} sulfonyl) -2- (1H-indazol-4-yloxy) benzamide; 4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - {[4 - ({[(2R) -4 -cyclopropylmorpholin- 2-yl] methyl} amino) -3-nitrophenyl] sulfonyl} -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(4 - {[(trans-4cyanocyclohexyl) methyl ] amino} -3-nitrophenyl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; Trans-2 - [(6-amino-5-chloropyridin-3-yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-14/55 yl ] methylJiperiperin-1-yl) -N - ({4 - [(4-morpholin-4-ylcyclohexyl) amino] -3-nitrophenyl} sulfonyl) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 -yljmethyljpiperazin-1 -yl) -N - {[4 - ({(3R) -1 - [2fluoro -1- (fluoromethyl) ethyl] pyrrolidin-3-yl} amino) -3-nitrophenyl] sulfonyl} -2- (1 H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide; Trans-N - ({5-chloro-6 - [(4-hydroxycyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[25 (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) -2- (1H-indazol-4yloxy) benzamide; N - ({3-chloro-4 - [(trans-4-hydroxycyclohexyl) methoxy] phenylJsulfonyl) -4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -2- (1H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide; N - ({5-chloro-6 - [(trans-4-hydroxycyclohexyl) methoxy] pyridin-3-ylSulfonyl) -4- (4 - {[2 (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en -1-yl-methyl-piperazin-1-yl) -2 - [(6-fluoro-1 H-indazol-410 yl) oxy] benzamide; 2 - [(6-amino-5-chloropyridin-3-yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex1 -en-1-yl] methyl IJpi perazin- 1 -yl) -N - [(4 - {[trans-4- (morpholin-4-yl) cyclohexyl] aminoJ-3nitrophenyl) sulfonyl] benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - [(4 - {[(cis-4- hydroxy-4-methylcyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] - 2- (1 H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methylJpiperazin-1-yl) -N - ({5-cyano-6 - [(4- fluorotetrahydro-2H-pyran-4-yl) methoxy] pyridin-3yl} sulfonyl) -2- (1H-indazol-4-yloxy) benzamide; N - [(5-chloro-6 - {[4-fluoro-1- (oxetan-3-yl) piperidin-4yl] methoxyJpiridin-3-yl) sulfonyl] -4- (4 - {[2- (4- chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2 - [(6-amino-5-chloropyridin- 3- yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - ({3 -nitro-4- [(tetrahydro-2H-pyran-4-ylmethyl) amino] phenyl} sulfonyl) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - ({4 - [(4-methylpiperazin-1- il) amino] -3nitrophenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; Trans-4- (4 - {[2- (4-chlorophenyl) - 4,4-dimethylcyclohex-1-en-1-yl] methylJpiperazin-1-yl) -N - [(4 - {[(4-methoxycyclohexyl) methyl] aminoJ-3nitrophenyl) sulfonyl] -2- (1H-pyrrole [ 2,3-b] pyridin-5-yloxy) benzamide; Trans-4- (4 - {[2- (4-chlorophenyl) - 4,4-dimethylcyclohex-1 -en-1 -yljmethyljiperiperin-1-yl) -N - ({4 - [(4-morpholin-4-ylcyclohexyl) amino] -3nitrophenyl} sulfonyl) -2- (1H-pyrrole [ 2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1-en-1-yl] methylJpiperazin-1-yl) -N - ({4 - [(4-fluorotetrahydro-2H-piran- 4-yl) methoxy] -3nitrophenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2 - [(6-amino-5-chloropyridin-3yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yljmethyljpiperazin-1-yl) -N - [(4 - {[(3R) -1 30 (2,2-difluoroethyl) pyrrolidin-3-yl] aminoJ-3-nitrophenyl) sulfonyl] benzamide; N - ({5-chloro-6 - [(trans-4hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-ylSulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex1 - en-1-yl] methyl} piperazin-1-yl) -2- (1 H-indazol-4-yloxy) benzamide; N - ({5-chloro-6 - [(cis-1-fluoro-4hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-ylSulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4, 4-dimethylcyclohex1-en-1-yl] methyl} piperazin-1-yl) -2- (1H-indazol-4-yloxy) benzamide; 2 - [(6-amino-5-chloropyridin-335 yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yljmethyljpiperazin-1-yl ) -N - [(4 - {[(4methoxycyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] benzamide; N - ({5-chloro-6 - [(trans-1-fluoro-4hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) - 4,4-dimethylcyclohex5 / 55 -en-1-yl] methyl} piperazin-1-yl) -2- (1 H-indazol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1 -en-1-yl] methyl Piperazin-1-yl) -N - [(4 - {[(trans-4-hydroxy- 4methylcyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2 - [(3-amino-1 H-indazol-4-yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - [(4 - {[(trans-4-methoxycyclohexyl) methyl] amino} -3nitrophenyl) sulfonyl] benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - ({3-nitro-4 - [(2 -oxaspiro [3.5] non-7-ylmethyl) amino] phenyl} sulfonyl) -2- (1Hpirrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - ({5-cyano-6 - [(trans -4-hydroxy-4-methylcyclohexyl) methoxy] pyridin-3yl} sulfonyl) -2- (1H-indazol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -2 - [(6-fluoro-1H-indole-5- il) oxy] -N - {[3-nitro-4 - ({[4- (oxetan-3-yl) morpholin-2yl] methyl} amino) phenyl] sulfonyl} benzamide; N - ({5-chloro-6 - [(trans-4-hydroxy-4methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4- dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -2 - [(6-fluoro-1H-indazol-4-yl) oxy] benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1 -yl) -N - [(5-cyano-6 - {[4- fluoro-1 - (oxetan-3-yl) piperidin- 4-yl] methoxy} pyridin-3-yl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2 - [(6-amino-5-chloropyridin-3-yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin- 1-yl) -N [(4 - {[(4-hydroxycyclohexy)) methyl] amino} -3-nitrophenyl) sulfonyl] benzamide; N - ({5-chloro-6 - [(trans-4hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -2 - [(3-chloro-1H-indazol-4-yl) oxy] - 4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) benzamide; 4- [4 - {[2- (4-chlorophenyl) ~ 4 I 4-dimethylcyclohex-1-en-1-yl] methyl} ( 2 H 8 ) piperazin-1-yl] -N - ({3-nitro -4 - [(tetrahydro-2H-pyran-4ylmethyl) amino] phenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide, · N - ({5-chloro- 6 - [(trans1-fluoro-4-hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1-en- 1-yl] methyl} piperazin-1-yl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4 (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(6 - {[(cis-4- hydroxy-4methylcyclohexyl) methyl] amino} -5-nitropyridin-3-yl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1 -yl) -N ({5-nitro-6 - [(tetrahydro -2H-pyran-4-ylmethyl) amino] pyridin-3-yl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin- 5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 -i IJmeti l} piperazin-1 -i I) -N- ({6 - [(trans-4 -hydroxy-4-methylcyclohexyl) methoxy] -5- (trifluoromethyl) pyridin-3-yl} sulfonyl) -2- (1Hindazol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin1-yl) -N - [(4 - {[(cis-4-ethyl -4-hydroxycyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1H-pyrrolo [2,3b] pyridin-5-yloxy) benzamide; and 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1 yl] methyl} piperazin-1-yl) -2- (1H-indol-5-yloxy) - N - (((3-nitro-4 - [(tetrahydro-2H-pyran-4ylmethyl) amino] phenyl} sulfonyl) benzamide. Another embodiment of the current invention pertains to a method for the treatment of Sjogren's Syndrome by administering a therapeutically effective amount of one with 6/55 since it selectively inhibits Bcl-2 proteins. Selective Bcl-2 inhibitors generally have the following Formula (I): (D. Where A 1 is N or CH; B 1 is OR 1 or NHR 1 ; I 1 is CN, NO 2i CF 3i F or Cl; R 1 is (CH 2 ) n R 2 ; R 2 is cycloalkyl or heterocycle; where the heterocycle and cycloalkyl are optionally substituted by one or more independently selected R 4 , OR 4 , OH, CN, or F; R 3 is heteroaryl; where the heteroaryl is optionally substituted by one or more independently selected NH 2 , Cl, or F; R 4 is alkyl, cycloalkyl, heterocycle, or spiroheterocycle; where the alkyl is optionally substituted by one or more F; R 5 is deuterium; each R 6 is independently selected from CH 3 , spirocyclopropyl and OH; m is 0.1, 2, 3, 4, 5, or 6; n is 0 or 1; and p is 0, 1, or 2. The method for treating Sjogren's Syndrome may also comprise administering a pharmaceutically acceptable salt of a selective Bcl-2 inhibitor. Generally, the selective Bcl-2 inhibitor has a binding affinity (K.) of less than about 1 7/55 nanomolar. In another embodiment, a selective Bcl-2 inhibitor has a binding affinity (Kj) of less than about 100 picomolar. The selective inhibitor of Bcl-2 may also have a binding affinity (Kj) for Bcl-2 that is approximately 500 times less than the binding affinity (K) for Bcl-x L. In this embodiment, the selective inhibitor of Bcl-2 5 may include N - ({5-chloro-6 - [(4-fluorotetrahydro-2H-pyran-4-yl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2 (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl-methyl-piperazin-1-yl) -2 - [(6-fluoro-1 H-indazol-4yl) oxy] benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl-methyl-piperazin-1-yl) -N - {[4 ({[(2S) -4-cyclopropylmorfolin- 2-yl] methyl} amino) -3-nitrophenyl] sulfonyl} -2- (1H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide; N - (((5-chloro-6 - [(4-fluorotetrahydro-2H-pyran-4-yl) methoxy] pyridin-3-yl} sulfonyl) -4- (410 {[2- (4-chlorophenyl) -4 , 4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) -2- (1 H-indazol-4yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4 , 4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) -2 - [(6fluoro-1H-indol-5-yl) oxy] -N - ({4 - [(4-fluorotetrahydro -2H-pyran-4-yl) methoxy] -3nitrophenyl} sulfonyl) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin- 1-yl) -N - [(4 - {[(4,4-difluorocyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1H15 pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2- (1 H-benzimidazol-4-yloxy) -4- (4 - ([2- (4-chlorophenyl) -4,4 dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - (((5-fluoro-6 - [(4-fluorotetrahydro-2H-pyran-4yl) methoxy] pyridin-3-yl} sulfonyl) benzamide; 4- (4 - {[2- (4-chlorophenyl) - 4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - ({3-nitro-4 - [(tetrahydro-2H-pyran-4-ylmethyl) amino] phenyl} sulfonyl) -2- (1Hpirrolo [2,3-b] pyridin-5-yloxy) benzamide; N - ((3-chloro-4 - [(4-fluorotetrahydro-2H-pir an-420 yl) methoxy] phenyl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) 2 - (1 H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2- (1 H-benzimidazol-4-yloxy) -4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 -yl] methyl} piperazin-1-yl) -N - [(4 - {[(4cyanocyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] benzamide; N - (((5-chloro-6 - [(cis-4-hydroxy-4methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4- dimethylcyclohex-1-en-125 yl] methyl} piperazin-1-yl) -2- (1 H-indazol-4-yloxy) benzamide; N - [(3-chloro-4 - ([4-fluoro-1 - (oxetan-3yl) piperidin-4-yl] methoxy} phenyl) sulfonyl] -4- (4 - ([2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1 -yl) -2- (1 H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4 dimethylcyclohex-1-en-1 -yl] methyl} piperazin-1-yl) -N - (((5-cyano-6 - [(4-fluorotetrahydro-2H-pyran-4yl) methoxy] pyridin-3-yl} sulfonyl) -2- (1H- indol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,430 dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(4- ( [(4-fluorotetrahydro-2H-pyran-4yl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; N - ((3 -chloro-4 [(4-fluorotetrahydro-2H-pyran-4-yl) methoxy] phenyl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex1-en-1- yl] methyl} piperazin-1-yl) -2- (1H-indazol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4 dimethylcyclohex-1-en -1-yl] methyl} piperazin-1-yl) -N - (((5-fluoro-6 - [(4-fluorotetrahydro-2H-pyran-435 yl) methoxy] pyridin-3-yl} sulfonyl) -2- (1H-indazol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1 -en-1 -yl] methyl} piperazin-1 -yl) -N - ([4 - ({[(2R) -4 -cyclopropylmorpholin-2yl] methyl} amino) -3-nitrophenyl] sulfonyl} -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - ((2- (4 8/55 chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(4 - {[(trans-4cyanocyclohexyl) methyl] amino} -3-nitrophenyl ) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; Trans-2 - [(6-amino-5-chloropyridin-3-yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1illmethyl} piperazin- 1-yl) -N - ({4 - [(4-morpholin-4-ylcyclohexyl) amino] -3-nitrophenyl} sulfonyl) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1 -yl) -N - {[4 - ({(3R) - 1 - [2 fluoro-1- (fluoromethyl) ethyl] pyrrolidin-3-yl} amino) -3-nitrophenyl] sulfonyl} -2- (1 H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide; Trans-N - ({5-chloro-6 - [(4-hydroxycyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2 (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) -2- (1H-indazol-4yloxy) benzamide; N - ({3-chloro-4 - [(trans-4-hydroxycyclohexyl) methoxy] phenyl} sulfonyl) -4- (4 - {[2- (410 chlorophenyl) -4,4-dimethylcyclohex-1-en-1 -yl] methyl} piperazin-1-yl) -2- (1H-pyrrolo [2,3-b] pyridin-5yloxybenzamide; N - ({5-chloro-6 - [(trans-4-hydroxycyclohexyl) methoxy] pyridin -3-yl} sulfonyl) -4- (4 - {[2 (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 -yl] methyl} piperazin-1 -yl) -2 - [(6 -fluoro-1 H-indazol-4yl) oxy] benzamide; 2 - [(6-amino-5-chloropyridin-3-yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4 , 4-dimethylcyclohex1 -en-1 -yl] methyl} pi perazin-1 -yl) -N - [(4 - {[trans-4- (morpholin-4-yl) cyclohexyl] amino} -315 nitrophenyl) sulfonyl] benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - [(4 - {[(cis- 4-hydroxy-4-methylcyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] - 2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -N - ({5-cyano-6 - [(4- fluorotetrahydro-2H-pyran-4-yl) methoxy] pyridin-3yl} sulfonyl) -2- (1H-indazol-4-yloxy) benzamide; N - [(5-chloro-6 - {[4-fluoro-1- (oxetan-3-yl) piperidin-4-yl] methoxy} pyridin-3-yl) sulfonyl] -4- (4 - {[2 - (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1 yl] methyl} piperazin-1-yl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2 - [(6-amino-5-chloropyridin- 3- yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - ({3 -nitro-4 [(tetrahydro-2H-pyran-4-ylmethyl) amino] phenyl} sulfonyl) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - ({4 - [(4-methylpiperazin-1- il) amino] -3-nitrophenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; Trans-4- (4 - {[2- (4-chlorophenyl) - 4.4- dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(4 - {[(4-methoxycyclohexyl) methyl] amino} -3nitrophenyl) sulfonyl] -2- (1H-pyrrole [2,3-b] pyridin-5-yloxy) benzamide; Trans-4- (4 - {[2- (4-chlorophenyl) - 4.4- dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - (((4 - [(4-morpholin-4-ylcyclohexyl) amino] -3- nitrophenyl} sulfonyl) -2- ( 1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,430 dimethylcyclohex-1-en-1-yl] methyl} piperazin-1- il) -N - ({4 - [(4-fluorotetrahydro-2H-pyran-4-yl) methoxy] -3nitrophenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2 - [(6-amino-5-chloropyridin-3yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 -iljmetyljpi perazin-1 -yl) -N - [(4 - {[(3R) -1 (2,2-difluoroethyl) pyrrolidin-3-yl] amino} -3-nitrophenyl) sulfonyl] benzamide; N - ({5-chloro-6 - [(trans-4hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex35 1 -en-1-yl] methyl} piperazin-1-yl) -2- (1 H-indazol-4-yloxy) benzamide; N - ({5-chloro-6 - [(cis-1-fluoro-4hydroxy-4-methylcyclohexyl) methoxy] pyridin -3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex1-en-1-yl] methyl} piperazin-1-yl) -2- (1H-indazole -4-yloxy) benzaniide; 2 - [(6-amino-5-chloropyridin-39/55 yl) oxy] -4 - (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(4 - {[(4methoxycyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] benzamide; N - ({5-chloro-6 - [(trans-1-fluoro-4hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) - 4,4-dimethylcyclohex- 1- en-1-yl] methyl} piperazin-1-yl) -2- (1H-indazol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 -yl] methyl} piperazin-1 -yl) -N - [(4 - {[(trans-4 -hydroxy-4methylcyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2- [(3-amino-1 H-indazol-4-yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl } piperazin-1-yl) -N - [(4 - {[(trans-4-methoxycyclohexyl) methyl] amino} -3nitrophenyl) sulfonyl] benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -N - ({3-nitro-4 - [(2- oxaspiro [3.5] non-7-ylmethyl) amino] phenyl} sulfonyl) -2- (1Hpirrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -N - ({5-cyano-6 - [(trans- 4-hydroxy-4-methylcyclohexyl) methoxy] pyridin-3yl} suphonyl) -2- (1 H-indazol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -2 - [(6-fluoro-1H-indole-5 -yl) oxy] -N - {[3-nitro-4 - ({[4- (oxetan-3-yl) morpholin-2yl] methyl} amino) phenyl] sulfonyl} benzamide; N - ({5-chloro-6 - [(trans-4-hydroxy-4methylcyclohexyl) methoxy] pyridin-3-i} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4- dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -2 - [(6-fluoro-1H-indazol-4-yl) oxy] benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1 -en-1-yl-methylpiperazi n-1 -yl) -N - [(5-cyano-6 - {[4-fluoro-1 - (oxetan-3-yl) piperidine- 4-yl] methoxy} pyridin-3-yl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2 - [(6-amino-5-chloropyridin-3-yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N [(4 - {[(4-hydroxycyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] benzamide; N - ({5-chloro-6 - [(trans-4hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -2 - [(3-chloro-1H-indazol-4-yl) oxy] - 4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) benzamide; 4- [4 - {[2- (4-chlorophenyl) - 4,4-dimethylcyclohex-1-en-1-yl] methyl} ( 2 H 8 ) piperazin-1-yl] -N - ({3-nitro-4 - [(tetrahydro-2H-pyran-4ylmethyl) amino] phenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; N - ({5-chloro-6 - [(trans1-fluoro-4-hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) - 4,4 dimethylcyclohex-1-en-1-yl-methyl} piperazin-1-yl) -2- (1 H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(6 - {[(cis-4 -hydroxy-4methylcyclohexyl) methyl] amino} -5-nitropyridin-3-yl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N ({5-nitro-6 - [(tetrahydro -2H-pyran-4-ylmethyl) amino] pyridin-3-yl} sulfonyl) -2- (1 H-pyrrolo [2,3-b] pyridin- 5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N- ({6 - [(trans-4- hydroxy-4-methylcyclohexyl) methoxy] -5- (trifluoromethyl) pyridin-3-yl} sulfonyl) -2- (1Hindazol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin1-yl) -N - [(4 - {[(cis-4-ethyl -4-hydroxycyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1H-pyrrolo [2,3b] pyridin-5-yloxy) benzamide; and 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -2- (1H-indol-5-yloxy) -N - ({3-nitro-4 - [(tetrahydro-2H-pyran-410/55 ylmethyl) amino] phenyl} sulfonyl) benzamide. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 illustrates the effect (A) of treatment with a non-selective Bcl-2 inhibitor (Compound 2) on both lymphocyte (left) and platelet (right) counts, as well as the effects (B) of treatment with a selective Bcl-2 inhibitor (Compound 1) in both lymphocyte numbers (left) and platelet count (right) in the peripheral blood of mice (NZBxNZW) F !. (C), (D), (E), (F), (G), (H) and (I) further illustrate the effects of Compounds 1, 17, 5, 7, 8, 9, and 12, respectively, lymphocyte numbers (left) and platelet count (right) in peripheral blood of C57BL / 6 mice. Figure 2 illustrates the effect of treatment with a selective Bcl-2 inhibitor (Compound 3) in mice on lymphocytes and platelets. Specifically, Figure 2 illustrates the effects of treatment in NZBWF1 mice with Compound 3 at doses of 30 mg / kg and 100 mg / kg, compared to treatment with the phosphate vehicle control, and also illustrates the exposure of Compound 3, as measured 24 hours after the last dose. Figure 3 illustrates the reduction in T cells and B cells in mice treated with various doses of a selective Bcl-2 inhibitor (Compound 3), when compared to treatment with a phosal vehicle. Specifically, Figure 3 illustrates the reduction in CD4 + T cells, CD8 + T cells, and CD19 + B cells in mice treated with doses of Compound 3 including 30 mg / kg, 100 mg / kg, and 300 mg / kg. Figure 4 illustrates the effectiveness of treatment with a selective Bcl-2 inhibitor (Compound 1) in a spontaneous murine model (NZBxNZW) Fi of lupus as assessed by the incidence (A) of severe proteinuria (PU> 300 mg / dL) and (B) Kaplan-Meier cumulative survival. Figure 4 further illustrates the effectiveness of treatment with a selective Bcl-2 inhibitor (Compound 17) in a spontaneous murine model (NZBxNZWJF! Lupus as assessed by the incidence (C) of severe proteinuria (PU> 300 mg / dL) and (D) Kaplan-Meier cumulative survival, asterisks represent a statistical significance of P <0.05. Figure 5 illustrates the efficacy of treatment with a selective Bcl-2 inhibitor (Compound 1), as well as treatment with mycophenolate mofetil (MMF), in inhibiting the titer production of anti-ds DNA in a spontaneous SLE model for the Compound 1 to 30 mg / kg and 100 30 mg / kg, and MMF at 100 mg / kg compared to control vehicles. Asterisks represent a statistical significance of P <0.05. Figure 6 illustrates the representative images demonstrating a reduction of infiltrates in the renal tissue of mice with spontaneous lupus nephritis, after dosing with a selective Bcl-2 inhibitor (Compound 1), as evidenced by histological evaluation35. 200x magnification. Figure 7 illustrates changes in histological sites for renal tissue treated with one of three treatment regimens: treatment with the vehicle, treatment with the Compound 11/55 at doses of 30 mg / kg and 100 mg / kg, and MMF treatment at a dose of 100 mg / kg, as they relate to histological signs of glomerulonephritis, tubular changes, and perivascular infiltrates. Specifically, figure 7 illustrates a statistically significant improvement in glomerulonephritis, tubular changes, and perivascular infiltrates in mice receiving treatment with 30 mg / kg and 100 mg / kg of Compound 1. Figure 8 illustrates an immunoglobulin G (IgG), a B cell, and a deposition of T cell in renal tissue for mice with spontaneous lupus nephritis dosed with a phosphate vehicle, Compound 1 in doses of 30 mg / kg and 100 mg / kg, and MMF at doses of 100 mg / kg. Specifically, the mice treated with Compound 1 showed a decreasing deposition of IgG, B cells, and T cells, when compared to the phosal vehicle. Figure 9 illustrates the effectiveness of treatment with a selective Bcl-2 inhibitor (Compound 1), MMF and BAFFR3-lg in an IFNa-induced SLE model as assessed by the (A) incidence of severe proteinuria (PU £ 300 mg / dL ) and (B) KaplanMeier cumulative survival. Asterisks represent a statistical significance of P <0.05. Figure 10 illustrates an anti-DNA DNA production in IFNa-induced IF (NZBxNZW) IF mice treated with a selective Bcl-2 inhibitor (Compound 1), in doses ranging from 3-30 mg / kg, purchased with the control vehicle. , as evidenced by the levels of Immunoglobulin G (IgG). Figure 11 illustrates representative images demonstrating the reduction of periductular infiltrates in the submandibular salivary gland of mice of the spontaneous murine model treated with a selective inhibitor of Bcl-2 (Compound 1), with a magnification of 200x. Figure 12 illustrates the histology points for submandibular salivary gland tissue treated with one of three treatment regimens: treatment with a phosphate vehicle, treatment with a selective Bcl-2 inhibitor (Compound 1) in doses of 10 mg / kg; and treatment with Compound 1 in doses of 30 mg / kg and 100 mg / kg. DETAILED DESCRIPTION The methods of the present invention are directed to the treatment of various disease states by administering to a patient in need of him a therapeutically effective amount of a selective Bcl-2 inhibitor. The expression Bcl-2 may play a role in the development and progression of the disease state associated with a number of autoimmune disorders. The Bcl-2 protein inhibitor may also have a positive impact on the treatment of autoimmune disorders including systemic lupus erythematosus (SLE), lupus nephritis and Sjogren's syndrome. The involvement of Bcl-2 proteins in autoimmune and immune disorders is described in Current Allergi and Astma Reports 2003, 3, 378-384; British Journal of Haematologi 2000, 110 (3), 584-90; Blood 2000, 95 (4), 1283-92; and New England Journal of Medicine 2004, 12/55 351 (14), 1409-1418. The involvement of Bcl-2 proteins in arthritis is disclosed in the United States Provisional Patent Application Serial No. 60 / 988,479 of common ownership. The involvement of Bcl-2 proteins in bone marrow transplant rejection is disclosed in the commonly owned United States Patent Application Serial No. 11 / 941,196. Unless otherwise defined herein, the scientific and technical terms used in connection with the present invention should have the meaning that is commonly understood by those of ordinary skill in the art. The meaning and scope of the terms must be clear, however, in the case of latent ambiguity, the definitions provided here take precedence over any dictionary or intrinsic definition. In this order, the use of "or" means "and / or" unless otherwise stated. In addition, the use of the term "including", as well as other forms, such as "includes" and "included", is not limiting. With reference to the use of the words "understand" or "understand" or "understanding" in this patent application (including the claims), the applicant notes that unless the context otherwise requires, those words are used based on clear understanding that they are not to be interpreted inclusive, rather than exclusively, and that the applicants intend that one of those words be so interpreted in the construction of this patent application, including the claims below. For a variant that occurs more than once in any substituent or in the compound of the invention or in any other formula here, its definition in each occurrence is independent of this definition in each other occurrence. Combinations of substituents are possible only if such combinations result in stable compounds. Stable compounds are compounds that can be isolated to a useful degree of purity from the reaction mixture. The terms "treat", "treating" and "treatment" refer to a method of alleviating or revoking a disease and / or its present symptoms. The terms "prevent", "preventing" and "prevention" refer to a method of preventing the onset of a disease and / or its present symptoms or preventing a subject from acquiring a disease. As used here, "prevent", "preventing" and "prevention" also include delaying the onset of a disease and / or its present symptoms and reducing a subject's risk of acquiring a disease. The term "therapeutically effective amount" refers to that amount of the compound being administered sufficiently to prevent the development or to some extent alleviate one or more of the symptoms of the condition or disorder being treated. The term "modular" refers to the ability of a compound to increase or decrease the function, or activity, of a Bcl-2 protein. The term “composition” as used here is intended to cover a product comprising the specified ingredients in the specified amounts, as well as any 13/55 product that results, directly or indirectly, from a combination of the specified ingredients in the specified quantities. By "pharmaceutically acceptable" is meant that the carrier, the diluent, or an excipient must be compatible with the other ingredients of the formulation and not harmful to the container thereof. The “subject is also included here animals such as mammals, including, but not limited to, primates (example, humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice and the like. In compatible modalities, the subject is a human. The state of the disease to be treated with the methods of the current invention includes those that result from an inflammatory and autoimmune process, specifically systemic lupus erythematosus (SLE), lupus nephritis, Sjogren's Syndrome and combinations thereof. SLE is a chronic systemic autoimmune disease that can affect any part of the body. As with other autoimmune diseases, the inume system attacks tissues and cells in the body, resulting in inflammation and tissue damage. The most common SLE affects the heart, joints, skin, lungs, blood vessels, liver, rihs, and the nervous system. Therefore, the disease state in which SLE has affected the kidneys can be called lupus nephritis. Additionally, Sjogren's Syndrome, also known as “Sicca's Syndrome”, also known as “Mikulicz's Disease” and “Sicca's Syndrome”, is a systemic autoimmune disease in which the immune cells attack and destroy the exocrine glands that produce the tears and saliva. As such, patients suffering from Sjogren's Syndrome typically suffer from a decreased ability to properly produce saliva and tears, resulting in excess dry eyes and mouth. Therefore, in one embodiment, the methods of the current invention are aimed at treating SLE and lupus nephritis. In another embodiment, the methods of the invention are directed to the treatment of Sjogren's Syndrome. As noted, the methods of the present invention comprise treatment with a selective Bcl-2 inhibitor. It is important to note that the term “Ki” is used as an objective measure for binding affinity. A compound that has a higher binding affinity for the target substrate compared to a second substrate, will show a Ki value for the target substrate that is less than the second substrate. This is due to the fact that the compound has a greater affinity for the target substrate, and a lower concentration of the compound is required to bind to and cause an effect on the target substrate, when compared to the second substrate. For example, selective Bcl-2 inhibitors have a greater affinity for Bcl-2 proteins compared to Bcl-xL proteins, meaning that a lower concentration of the selective Bcl-2 inhibitor is required to have an effect on the Bcl- 2 inhibitor. 2, when compared to the concentration required to cause an effect on Bcl-xL proteins. As referenced here, the language stating that a compound has a 14/55 competitive binding affinity (Kj) for Bcl-2 which is less than the binding affinity (Kj) for Bcl-x L should be constructed to mean that the compound has a higher selective affinity for Bcl-2 than for Bcl-x L , as evidenced by a K value, for Bcl-2 which is less than the Kj value for Bcl-x L. o Specifically, the phrase '' selective inhibitor ae tíci-z refers to compounds that have a binding affinity (Kj) of (a) less than about 500 nanomolar, less than about 400 nanomolar, less than about 300 nanomolar, less than about 200 nanomolar, less than about 100 nanomolar, less than about 50 nanomolar, less than about 25 nanomolar, less than about 10 nanomolar, less than about 5 nanomolar, less than about 1 nanomolar, less than about 900 picomolar, less than about 800 picomolar, less than about 700 picomolar, less than about 600 picomolar, less than about 500 picomolar, less than about 400 picomolar, less than about 300 picomolar, less than about 200 picomolar, less than about 100 picomolar, and less than about 50 molar pico15 for Bcl-2; and (b) a competitive binding affinity (K |) for Bcl-2 that is at least about 500, at least about 1000, at least about 2000, at least about 2500, at least about 3000, at least about 3500, and at least about 4000 times less than the binding affinity (K) for Bcl-x L. In some embodiments of the present invention, the selective Bcl-2 inhibitory compounds that can be used in the methods of the present invention are those having a binding affinity (Kj) less than about 100 picomolar for Bcl-2 and an affinity of binding (K,) for Bcl-2 which is approximately 500 times less than the binding affinity for Bcl-x L , a binding affinity (K) less than about 100 picomolar for Bcl-2 and a binding affinity (Kj) for Bcl-2 which is approximately 1000 times less than the binding affinity for Bcl-x L , a binding affinity (K) less than about 100 picomolar for Bcl-2 and a binding affinity ( K f ) for Bcl-2 which is approximately 2000 times less than the binding affinity for Bcl-x L , a binding affinity (K |) less than about 100 picomolar for Bcl-2 and a binding affinity ( K) for Bcl-2 which is approximately 2500 times less than the binding affinity o for Bcl-X | _, a binding affinity (Ki) less than about 100 picomolar for Bcl-2 and a binding affinity (Ki) for Bcl-2 which is approximately 3000 times less than the affinity of binding for Bcl- x L , a binding affinity (Ki) less than about 100 picomolar for Bcl-2 and a binding affinity (Ki) for Bcl-2 which is approximately 3500 times less than the binding affinity for Bcl-2 Bcl- x L , a binding affinity (Ki) less than 35 than about 100 picomolar for Bcl-2 and a binding affinity (Ki) for Bcl-2 which is approximately 4000 times less than the binding affinity for Bcl - x L. The selective inhibitory compounds of Bcl-2 that can be used in the methods of The current invention can generally be considered to be any compound having the following Formula (I): (D, where A 1 is N or CH; B 1 is OR 1 or NHR 1 ; Γ is CN, NO 2 , CF 3i F or Cl; R 1 is (CH 2 ) n R 2 ; R 2 is cycloalkyl or heterocycle; where the heterocycle and cycloalkyl are optionally substituted by one or more independently selected R 4 , OR 4 , OH, CN, or F; R 3 is heteroaryl; where the heteroaryl is optionally substituted by one or more independently selected NH 2 , Cl, or F; R 4 is alkyl, cycloalkyl, heterocycle, or spiroheterocycle; where the alkyl is optionally substituted by one or more F; R 5 is deuterium; each R 6 is independently selected from CH 3 , spirocyclopropyl and OH; m is 0, 1, 2, 3, 4, 5, or 6; n is 0 or 1; and p is 0, 1, or 2. Methods for making selective Bcl-2 inhibitors, such as those covered by Formula (I) and which can be used in the methods of the present invention are described in US Serial No. 12 / 787,682, filed on May 26, 2010, US Serial No. 12 / 793,418, filed on June 3, 2010 which is a continuation in part of US Se 16/55 rial No. 12 / 631,404, deposited on December 4, 2009, and US Serial No. 12 / 793,413, deposited on June 3, 2010 which is a continuation in part of US Serial No. 12 / 631,367, deposited on 4 December 2009, the contents of each of which are incorporated herein. In relation to the various starting compounds seieuvus a and Óci-2 aoranged by the current invention, it must be understood that the variable halves here are represented by the identifiers (capital letters with numerals and / or alphabetic superscripts) and can be specifically incorporated. It is intended to be understood that the valences proper to the halves are maintained for all halves and combinations thereof, that monovalent halves having more than one atom are drawn from left to right and are attached through two ends of the left, and that the divalent halves are also drawn from left to right. It is intended to be understood that a specific modality of a variable half here can be the same or different as another specific modality having the same identifier. The term "alkenyl" as used herein, means a straight or branched hydrocarbon chain containing from 2 to 10 carbons and containing at least one carbonocarbon double bond. The term "C x -Cj alkyl" means a straight or branched hydrocarbon chain containing at least one carbon-carbon double bond containing two carbon atoms. The term "C2-C4 alkenyl" means an alkenyl group containing 2-4 carbon atoms. Representative examples of alkenyl include, but are not limited to, buta-2,3-dienyl, ethenyl, 2-propenyl, 2-methyl-2-propenyl, 3-butenyl, 4-pentenyl, 5-hexenyl, 2-heptenyl, 2-methyl- 1-heptenyl, and 3-decenyl. The term alkenylene means a divalent group derived from a straight or branched hydrocarbon chain of 2 to 4 carbon atoms and contains at least one carbon-carbon double bond. The term “C X -C | alkylene ”means a divalent group derived from a straight or branched hydrocarbon chain containing at least one carbon-carbon double bond and containing xai carbon atoms. Representative examples of alkenylene include, but are not limited to, alkenylene include, -CH = CH- and -CH 2 CH = CH-. The term alkyl as used here, means a straight or branched saturated hydrocarbon chain containing from 1 to 10 carbon atoms. The term “C x -C | alkyl ”means a straight or branched saturated hydrocarbon chain containing xai carbon atoms. For example, "C 2 -C 10 alkyl" means a straight or branched saturated hydrocarbon chain containing from 2 to 10 carbon atoms. Examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, 17/55 isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2,2-dimethylpentyl, 2,3-dimethylpentyl, n-heptyl, n-octyl, nnonyl, and n-decyl. The term "alkylene means a divalent group derived from a straight or branched saturated hydrocarbon chain of 1 to 10 carbon atoms, for example, 1 to 4 carono atoms. A term 'Córr aiquiieno Sigrinica' is a modified group oivai and a straight or branched saturated hydrocarbon chain containing xai carbon atoms. For example, "C 2 -C 6 alkylene" means a straight or branched saturated hydrocarbon chain containing 2 to 6 carbon atoms. Examples of alkylene include, but are not limited to, -CH 2 -, -CH 2 CH 2 -, -CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 -, and -CH 2 CH ( CH 3 ) CH 2 -. The term alkynyl as used herein, means a straight or branched hydrocarbon chain group containing from 2 to 10 carbon atoms and containing at least one carbon-carbon triple bond. The term "C x -Ci alkynyl" means a straight or branched hydrocarbon chain group containing xai carbon atoms. Representative examples of alkynyl include, but are not limited to, acetylenyl, 1-propynyl, 2-propynyl, 3-butynyl, 2-pentynyl, and 1-butynyl. The term alkynylene, as used herein, means a divalent radical derived from a straight or branched hydrocarbon chain group containing from 2 to 10 carbon atoms and containing at least one carbon-carbon triple bond. The term aryl (alone or in combination with another term (s)) means an aromatic carbocicyl containing 6 to 14 atoms in the carbon ring. An aryl can be monocyclic or polycyclic (that is, it can contain more than one ring). In the case of aromatic polycyclic rings, only one ring of the polycyclic system is required to be unsaturated while the remaining ring (s) can be saturated, partially saturated or unsaturated. Examples of aris include phenyl, naphthalenyl, indenyl, indanyl, and tetrahydronaphthyl. The term carbocycle (alone or in combination with another term (s)) means a completely unsaturated (i.e., "aryl"), saturated cyclic (i.e., cycloalkyl) or partially saturated (i.e., cycloalkenyl) hydrocarbon containing 3 to 14 atoms in the carbon ring (“carbon rings” are the atoms linked together to form the ring or rings of a cyclic substituent). A carbocycle can be a single ring (monocyclic) or polycyclic ring structure. A carbocycle can be a single ring structure, which typically contains 3 to 8 rings of atoms, more typically 3 to 6 rings of atoms, and even more typically 5 to 6 rings of atoms. Examples of such single ring carbocycles include cyclopropyl (cyclopropanyl), cyclobutyl (cyclobutanil), cyclopentyl (cyclopentanyl), cyclopentenyl, cyclopentadienyl, cyclohexyl (cyclohexanil), cyclohexenyl, cyclohexadienyl, and phenyl. A carbocyclyl may alternatively be polycyclic (that is, it may contain more than one ring). Examples of polycyclic carbocycles include spirocyclic, fused and bridge carbocycles. On a carbocycle 18/55 polycyclic, an atom is common to different rings. An example of a substituted spirocyclic carbocycle is spirocyclopropyl. In a spirocyclic carbocycle, an atom is common to two different rings. An example of a spirocyclic carbocycle is spiropentanil. In a bridged carbocycle, the rings share at least two common non-adjacent atoms. The examples of bicyclic bridged carbocycles (2.z. I jnepiariii, oícicio | 2.2. Ijhepí-2-enii, and adamantanil. In a fused ring carbocycle system, two or more rings can be fused together, in a that the rings share a common bond. Examples of two or three fused ring carbocycles include naphthalenyl, tetrahydronaphthalenyl (tetralinyl), indenyl, indanyl (dihydroindenyl), anthracenyl, phenanthrenyl, and decalinyl. The term "cyclic half, as used herein, means benzene, phenyl, phenylene, cycloalkane, cycloalkyl, cycloalkylene, cycloalkylene, cycloalkenyl, cycloalkenylene, cycloalkylene, cycloalkyl, cycloalkylene, heteroaryl, heteroaryl, heterocycloalkyl, heterocycloalkyl, heterocycloalkyl, heterocycloalkyl, heterocycloalkyl, heterocycloalkyl, heterocycloalkyl. The term cycloalkylene or cycloalkyl or "cycloalkane" as used here, means a bridged or monocyclic hydrocarbon ring system. Monocyclic cycloalkyl is a carbocyclic ring system containing three to eight carbon atoms, zero heteroatoms and zero double bonds. Examples of monocyclic ring systems include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohepty, and cyclooctyl. The monocyclic ring may contain one or more alkylene bridges, each consisting of one, two or three carbon atoms, each linking two non-adjacent carbon atoms in the ring system. Non-limiting examples of such cycloalkyl bridge ring systems include bicycles [3.1.1] heptane, bicycles [2.2.1] heptane, bicycles [2.2.2] octane, bicycles [3.2.2] nonane, bicycles [3.3. 1] nonane, bicycles [4.2.1] nonane, tricycle [3.3.1.0 3.7 ] nonane (octahydro-2,5 methanopentalene or noradamantane), and tricycle [3.3.1.1 3.7 ] decane (adamantane). The bridged and monocyclic cycloalkyl 25 can be attached to the parent molecular moiety through any replaceable atom contained within the ring system. The term cycloalkenylene, or cycloalkenyl or "cycloalkene" as used here, means a bridged or monocyclic hydrocarbon ring system. The monocyclic cycloalkenyl has four-, five-, six-, seven- or eight carbon atoms and zero hetero atoms. Four-membered ring systems having one, two or three double bonds. Representative examples of monocyclic cycloalkenyl groups include, but are not limited to, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl. The monocyclic cycloalkenyl ring may contain one or two alkylene bridges, each consisting of one, two or three carbon atoms, each linking two non-adjacent carbon atoms in the ring system. Representative examples of bicyclic cycloalkenyl groups include, but are not limited to, 4,5,6,7-tetrahydro-3aH-indene, octahydronaphthalenyl, and 1,6dihydro-pentalene. Bicyclic and monocyclic cycloalkenyl can be attached to one half 19/55 parent molecular through any replaceable atom contained within the ring systems. The term cycloalkine, or cycloalkynyl, or cycloalkynylene, as used herein, means a bridged or monocyclic hydrocarbon ring system. Cycloalkynyl is monocyclic with one or more kioono atoms, zero neceo atoms and one or more triple bonds. The monocyclic cycloalkynyl ring may contain one or two alkylene bridges, each consisting of one, two, or three carbon atoms, each linking two non-adjacent carbon atoms in the ring system. The bridged and monocyclic cycloalkynyl can be attached to a parent molecular moiety through any replaceable atom 10 contained within the ring systems. The term heteroarene, ”or heteroaryl, or heteroarylene, as used herein, means a six-membered or five-membered aromatic ring having at least one carbon atom and one or more than an independently selected nitrogen, oxygen or sulfur atom. The heteroarenes of this invention are connected through any 15 adjacent atoms in the ring, as long as the proper valences are maintained. Specifically, the term heteroaryl (alone or in combination with another term (s)) means an aromatic heterocycle containing 5 to 14 ring atoms. A heteroaryl can be a single ring or 2 or 3 fused rings. Examples of heteroaryl substituents include 6-membered ring substituents such as pyridyl, pyrazyl, pyrimidinyl, pyridazinyl, and 1,3,5-, 1,2,420 or 1,2,3-triazinyl; 5-membered ring substituents such as imidazyl, furanyl, thiophenyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, 1,2,3-, 1,2,4-, 1,2,5-, or 1,3,4- oxadiazolyl and isothiazolyl; 6/5-member fused ring substituents such as benzothiofuranyl, benzisoxazolyl, benzoxazolyl, imidazolyl, indolyl, benzoimidazolyl, pyrrolo [2,3-b] pyridinyl, purinil, and anthranilyl; and 6/6-member fused rings such as benzopyranyl, quinolinyl, isoquinolinyl, cinnoiinyl, quinazolinyl, and benzoxazinyl. The term heterocycle (alone or in combination with another term (s)) means a ring structure (i.e., "heteroaryl) completely unsaturated, partially saturated (i.e.," heterocycloalkenyl ") or saturated (i.e., heterocycloalkyl) containing a total of 3 to 14 ring atoms. At least one of the ring atoms is a hetero atom (i.e., oxygen, nitrogen or sulfur), with the remaining ring atoms being selected independently from a group consisting of carbon, oxygen, nitrogen and sulfur. A heterocycle can be a single ring (monocyclic) or polycyclic ring structure. A heterocycle can be a single ring, which typically contains 3 to 7 ring atoms, more typically 3 to 6 ring atoms, and even more typically 5 to 6 ring atoms. Examples of single ring heterocycles include furanyl, dihydrofuranyl, tetrahydrofuranyl, tetrahydropyranyl, thiophenyl (thiophuranyl), dihydrothiophenyl, tetrahydrothiophenyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, pyridazolyl, pyridazolyl, pyridazolyl, pyridinol, pyridinol, pyridine 20/55 tetrazolyl, oxazolyl, oxazolidinyl, isoxazolidinyl, isoxazolyl, thiazolyl, isothiazolyl, thiazolinyl, isothiazolinyl, thiazolidinyl, isothiazolidinyl, thiodiazolyl, oxadiazolyl (including 1,2,3-oxadiazolyl, 1,2,4 oxadiazolyl (furazanil), or 1,3,4-oxadiazolyl), oxatriazolyl (including 1,2,3,4oxatriazolyl or 1,2,3,5-oxatriazolyl), oxetanil, dioxazolyl (including 1,2,3-dioxazolyl, 1,2,4ó aioxazoni, 1,3,2-ciioxazoiii, or i, 3,4-aioxazoiii), oxauazoiü, oxatioui, oxauoianii, pirami, aihydropyranyl, thiopyranil, tetrahydrothiopyranyl, pyridinyl (azinyl), piperidinyl, diazinil (including pyridinyl) (1,2-diazinyl), pyrimidinyl (1,3-diazinyl), or pyrazinyl (1,4-diazinyl)), piperazinyl, triazinyl (including 1,3,5-triazinyl, 1,2,4-triazinyl, and 1,2,3-triazinyl)), oxazinyl (including 1,2-oxazinyl, 1,3-oxazinyl, or 1,4-oxazinyl)), oxatiazinyl (including 1,2,3-oxatiazinyl, 1,2,4-oxatiazinyl, 1,2,5-oxatiazinyl, or 10 1,2,6-oxatiazinyl)), oxadiazinyl (including 1,2,3-oxadiazinyl, 1,2,4-oxadiazinyl, 1,4,2-oxadiazinyl, or 1,3,5-oxadiazinyl)), morpholinyl, azepinyl, oxepinyl, tiepinyl, and diazepinil. A heterocycle may alternatively be polycyclic (that is, it may contain more than one ring). Examples of polycyclic heterocycles include bridged, fused and spirocyclic heterocycles. In an spirocyclic heterocycle, an atom is common to two different rings. Examples of spirocyclic heterocycles include 2-oxaspiro [3.5] nonanil. Examples of polycyclic heterocycles include bridged, fused and spirocyclic heterocycles. In a spirocyclic heterocycle, an atom is common to two different rings. Examples of a spirocyclic heterocycle include 2-oxaspiro [3,5] nonanil. In a bridged heterocycle, the rings share at least two common non-adjacent atoms. In a fused ring heterocycle 20, two or more rings can be fused together, so that the rings share a common bond. Examples of fused ring heterocycles containing two or more rings include indolizinyl, pyranopyrrolyl, 4H-quinolizinyl, purinyl, naphthyridinyl, pyrrole [2,3bjpiridinyl, pyridopyridinyl (including pyrido [3,4-b] -pyridinyl, pyrido [3,2 -b] -pyridinyl, or pyrido [4,3-b] pyridinyl), and pteridinyl. Other examples of fused ring heterocycles include benzo-fused heterocycles, such as indolyl, indazoyl, isoindolyl (isobenzazolyl, pseudoisoindolyl), indoleninyl (pseudoindolyl), isoindazolyl (benzopyrazolyl), benzoimidazolyl, benzazinyl (quinoline) isoquinolinyl (2-benzazinyl)), phthalazinyl, quinoxalinyl, quinazolinyl, benzodiazinyl (including cinnolinyl (1,2-benzodiazinyl) or quinazolinyl (1,3-benzodiazinyl)), benzopyranyl (including chromanil or isochromanyl), benzoxazinil (including 1.3 , 2-benzoxazinyl, 1,4,230 benzoxazinyl, 2,3,1-benzoxazinyl, or 3,1,4-benzoxazinyl), and benzisoxazinyl (including 1,2benzisoxazinyl or 1,4-benzisoxazinyl). The term heterocycloalkyl (alone or in combination with another term (s)) means a saturated heterocycle. The term heterocycloalkene, or heterocycloalkenyl, or heterocycloalkenylene, 35 as used herein, means a ring of three-, fourth-, five-, six-, seven-, or eight monocyclic or bridged members containing at least one heteroatom independently selected from of a group consisting of O, N, and S and one or more double bonds. The 21/55 bridged and monocyclic heterocycloalkenes are connected to the parent molecular moiety through any replaceable carbon or any replaceable nitrogen atom contained within the rings. The nitrogen and sulfur hetero atoms in the heterocyclic rings can optionally be oxidized and the nitrogen atoms can be optionally quernized. Representative examples of heterocycloalkene groups include, but are not limited to, tetrahydrooxocinyl, 1,4,5,6-tetrahydropyridazinyl, 1,2,3,6-tetrahydropyridinyl, dihydropyranyl, imidazolinyl, isothiazolinyl, oxadiazolinyl, isoxazolinyl, oxoxazolinyl, oxoxazoline , pyrazolinyl, pyrrolinyl, thiadiazolinyl, thiazolinyl, and thiopyranil. The term phenylene, as used here, means a divalent radical formed by the removal of a hydrogen atom from a phenyl. The term spiroalkyl, as used here, means alkylene, both ends of which are attached to the same carbon atom and is exemplified by C 2 -spiroalkyl, C 3 -spiroalkyl, C 4 -spiroalkyl, C 5 -spiroalkyl, C 6 - spiroalkyl, C 7 -spiroalkyl, C 8 -spiroalkyl, Cg-spiroalkyl and the like. The term spiroheteroalkyl, as used here, means spiroalkyl having one or two halves of CH 2 replaced by O, C (O), CNOH, CNOCH 3 , S, S (O), SO 2 or NH independently selected and one or two halves of CH unsubstituted or substituted by N. The term spiroheteroalkenyl, as used here, means spiroalkenyl having a 20 or two halves of CH 2 replaced by O, C (O), CNOH, CNOCH 3 , S, S (O), SO 2 or NH independently selected and one or two halves of CH unsubstituted or substituted by N and also means spiroalkenyl having one or more CH 2 halves unsubstituted or substituted by O, C (O), CNOH, CNOCH 3 , S, S (O), SO 2 or NH independently selected and one or two halves of CH replaced by N. The term spirocycle, as used here, means two substituents on the same carbon atom, which together with the carbon atom to which they are attached, to form a cycloalkane, heterocycloalkane, cycloalkene, or heterocycloalkene ring. The term The term C 2 -C 5 -spiroalkyl, as used herein, means C 2 -spiroalkyl, C 3 -spiroalkyl, C 4 -spiroalkyl, and C 5 -spiroalkyl. The term C 2 -spiroalkyl, ”as used here, means et-1,2-ylene, both ends of which replace the hydrogen atoms of the same half of CH 2 . The term C 3 -spiroalkyl, ”as used here, means prop-1,3-ylene, both ends of which replace the hydrogen atoms of the same half of CH 2 . The term C 4 -spiroalkyl, ”as used here, means but-1,4-ylene, both ends of which replace the hydrogen atoms of the same half of CH 2 . The term C 5 -spiroalkyl, ”as used here, means pent-1,5-ylene, both ends of which replace the hydrogen atoms of the same half of CH 2 . 22/55 The term C 6 -spiroalkyl, ”as used here, means hex-1,6-ylene, both ends of which replace the hydrogen atoms of the same half of CH 2 . The term protecting group NH, as used herein, means trichloroethoxycarbonyl, tribromoethoxycarbonyl, benzyloxycarbonyl, para-nitrobenzylcarbonyl, ortho-bromobenzyloxycarbonyl, chloroacetyl, dichloroacetyl, trichloroacetyl, trifluoroacetii, phenylacetyl, benzyl, tertiary, acetyl, tertiary, -methoxybenzyloxycarbonyl, 3.4- dimethoxybenzyl-oxycarbonyl, 4- (phenylazo) benzyloxycarbonyl, 2-furfuryl-oxycarbonyl, diphenylmethoxycarbonyl, 1,1-dimethylpropoxy-carbonyl, isopropoxycarbonyl, phthaloyl, succinyl, alanyl, leucyl, 1adamantyloxycarbonyl, trilinyl, methyl-benzyl, trilinyl, 8-quinolyl , 2-nitrophenylthio, methanesulfonyl, para-toluenesulfonyl, Ν, Ν-dimethylaminomethylene, benzylidene, 2-hydroxybenzylidene, 2-hydroxy-5-chlorobenzylidene, 2-hydroxy-1-naphthyl-methylene, 3-hydroxy-4-pyridylmethyl-ethylene-2-hydroxy-ethylene-cyclohexyl , 2-ethoxycarbonylcyclopentylidene, 2-acetylcyclohexylidene, 3,3-dimethyl-5-oxycyclohexylidene, diphenylphosphoryl, dibenzylphosphoryl, 5-methyl2-oxo-2H-1,3-dioxol-4-yl-methyl, trimethylsilyl, and triethylsyl triphenylsilyl. The term protecting group C (O) OH, as used herein, means methyl, ethyl, n-propyl, isopropyl, 1,1-dimethylpropyl, n-butyl, tert-butyl, phenyl, naphthyl, benzyl, diphenylmethyl, triphenylmethyl, paranitrobenzyl , para-methoxybenzyl, bis (para-methoxyphenyl) methyl, acetylmethyl, benzoylmethyl, paranitrobenzoylmethyl, para-bromobenzoylmethyl, para-methanesulfonylbenzoylmethyl, 2-tetrahydropyranyl 2-tetrahydrofuranyl, 2,2,2-trichlorethylethyl, ethyl , acetoxymethyl, propionyloxymethyl, pivaloyloxymethyl, phthalimidomethyl, succinimidomethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methoxymethyl, methoxyethoxymethyl, 2- (trimethylsilyl) ethoxymethyl, benzyloxymethyl, 2-methylthiomethyl, 2-methylethyl-methylthiomethyl 3-methyl-3-butenyl, allyl, trimethylsilyl, triethylsilyl, triisopropylsilyl, diethylisopropylsilyl, tert-butyldimethylsilyl, tert-butyldiphenylsilyl, diphenylmethylsilyl, and tert-butylmethoxyphenylsilyl. The term OH or SH protecting group, as used herein, means benzyloxycarbonyl, 4-nitrobenzyloxycarbonyl, 4-bromobenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 3.4- dimethoxybenzyloxycarbonyl, methoxycarbonyl, ethoxycarbonyl, tert-butoxycarbonyl, 1,1-dimethylpropoxycarbonyl, isopropoxycarbonyl, isobutyloxycarbonyl, diphenylmethoxycarbonyl, 2,2,2-trichloroethoxycarbonyl, 2,2,2-tribromoethoxycarbonyl, 2- (trimethylsilyl) ethoxycarbonyl, 2- (phenylsulfonyl) ethoxycarbonyl, 2- (triphenylethylphosphonyl, 2) -furfuryloxycarbonyl, 1adamantyloxycarbonyl, vinyloxycarbonyl, allyloxycarbonyl, S-benzylthiocarbonyl, 4-ethoxy-1naftyloxycarbonyl, 8-quinolyloxycarbonyl, acetyl, formyl, chloroacetyl, dichloroacetyl, trichloroacetyl, trichloroacetyl, trifluoroacetyl, trifluoroacetyl, trifluoroacetyl, trichloroacetyl, trichloroacetyl, trichloroacetyl , 2,2-trichloroethyl, 2-trimethylsilylethyl, 1,1-dimethyl-2-propenyl, 3-methyl-3-butenyl, allyl, benzyl (phenylmethyl), para-methoxybenzyl, 3,4-dimethoxybenzyl, diphenylmethyl, triphenylmethyl, tetrahydrofuryl, tetrahydropyranyl, tetrahydrothiopyranyl, methoxymethyl, methylthiomethyl, benzyloxymethyl, 2-methoxyethoxymethyl, 2,2,2-trichloroethoxymethyl, 2- (trimethylsilyl) ethoxymethyl, 1-ethoxyethyl, methanesulfonyl, para-toluenes 23/55 triethylsilyl, triisopropylsilyl, diethylisopropylsilyl, tert-butyldimethylsilyl, tert-butyldiphenylsilyl, diphenylmethylsilyl, and tert-butylmethoxyphenylsilyl. If a substituent is described as being "substituted", non-hydrogen radiation is in place of a hydrogen radical in a carbon or nitrogen substituent. Thus, for example, a suosutuent ue aiquii südsiííuicío and a auu suosiíiuiiiie in which at least one non-hydrogen radical is in place of a hydrogen radical in the alkyl substituent. To illustrate, monofluoralkyl is alkyl substituted with a fluorine radical, and difluoralkyl is alkyl substituted by two fluorine radicals. It must be recognized that if there is more than one substitution in a substituent, each non-hydrogen radical can be identical or different (unless otherwise stated). If a substituent is described as being "optionally substituted", the substituent can be either (1) unsubstituted or (2) substituted. If a substituent is described as being optionally substituted by up to a particular number of non-hydrogen radicals, that substituent can be either (1) unsubstituted; or (2) replaced by up to 15 that particular number of non-hydrogen radicals or by a maximum number of replaceable positions in the substituent, whichever is less. Thus, for example, if a substituent is described as a heteroaryl optionally substituted with up to 3 non-hydrogen radicals, then any heteroaryl with less than 3 substitutable positions could optionally be substituted by up to just as much as non-hydrogen radicals the 20 heteroaryl has replaceable positions. To illustrate, tetrazolyl (which has only one replaceable position) would be optionally substituted by up to a non-hydrogen radical. To further illustrate, if an amino nitrogen is described as being optionally substituted by up to 2 hydrogen radicals, then a primary amino nitrogen will be optionally substituted by up to 2 non-hydrogen radicals, whereas a second amino nitrogen will optionally be replaced by up to only 1 non-hydrogen radical. This patent application uses the terms "substituent" and "radical" interchangeably. As stated, the selective Bcl-2 inhibitory compounds of the current invention encompass all possible combinations of the substituents for the compound genus that within the scope of the current invention include, but are not limited to, N - ({5-chloro-630 [(4-fluorotetrahydro-2H-pyran-4-yl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4 dimethylcyclohex-1 -en-1 - iIjmetiljpiperazin-1-yl) -2 - [(6-fluoro-1H-indazol-4-yl) oxy] benzamide; 4- (4 {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yljmethyljpiperazi n-1 -yl) -N - {[4 - ({[(2S) -4cyclopropylmorfolin-2 -yl] methyl} amino) -3-nitrophenyl] sulfonyl} -2- (1H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide; N - ({5-chloro-6 - [(4-fluorotetrahydro-2H-pyran-4-yl) methoxy] pyridin-3-yl} sulfonyl) -4- (435 {[2- (4-chlorophenyl) -4 , 4-dimethylcyclohex-1-en-1-yl-methyl-piperazin-1-yl) -2- (1 H-indazol-4yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl-methyl-piperazin-1-yl) -2 - [(6-fluor-1H-indol-5-yl) oxy] -N - ({4 - [(4-fluorotetrahydro-2H-pyran-4-yl) methoxy] -324/55 nitrophenyl} sulfonyl) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - [(4 - ([(4,4- difluorocyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1Hpyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2- (1H-benzimidazol-4-yloxy) -4- (4- ([2- (4-chlorophenyl) -4,4 dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - (((5-fluor-6 - [(4-fluorotetrahydro-2H-pyran -4yl) methoxy] pyridin-3-yl} sulfonyl) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -N - ((3-nitro-4 - [(tetrahydro-2H-pyran-4-ylmethyl) amino] phenyl} sulfonyl) -2- (1Hpyrrolo [2,3-b] pyridin-5-yloxy) benzamide; N - (((3-chloro-4 - [(4-fluorotetrahydro-2H-pyran-4yl) methoxy] phenyl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) 2- (1 H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2- (1 H-benzimidazol-4-yloxy) - 4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(4 - {[(4cyanocyclohexyl) methyl] amino } -3-nitrophenyl) sulfonyl] benzamide; N - ((5-chloro-6 - [(cis-4-hydroxy-4methylcyclohexyl) methoxy] pyridin- 3-yl} sulfonyl) -4- (4 - {[2- (4-chloroOphenyl) -4,4-dimethylcyclohex-1-en-1i IJmeti l} piperazin-1 -yl) -2- (1 H-indazole -4-yloxy) benzamide; N - [(3-chloro-4 - {[4-fluoro-1 - (oxetan-3yl) piperidin-4-yl] methoxy} phenyl) sulfonyl] -4- (4 - ([2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[ 2- (4-chlorophenyl) -4,4dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - ((5-cyano-6 - [(4-fluorotetrahydro-2H-pyran-4il ) methoxy] pyridin-3-yl} sulfonyl) -2- (1H-indol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4 dimethylcyclohex-1-en-1- il] methyl} piperazin-1-yl) -N - [(4 - ([(4-fluorotetrahydro-2H-pyran-4yl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1H-pyrrole [2 , 3-b] pyridin-5-yloxy) benzamide; N - ((3-chloro4 - [(4-fluorotetrahydro-2H-pyran-4-yl) methoxy] phenyl} siylphonyl) -4- (4 - {[2 - (4-chlorophenyl) -4 > 4dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -2- (1H-indazol-4-yloxy) benzamide; 4- (4 - ((2- (4chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - (((5-fluoro-6 - [(4-fluorotetrahydro2H-pyran-4-yl) methoxy ] pyridin-3-yl} sulfonyl) -2- (1H-indazol-4-yloxy) benzamide; 4- (4 - ((2- (4chlorophenyl) -4,4-dimethylcyclohex-1-en-1-i l] methyl} piperazin-1-yl) -N - ([4 - ({[(2R) -4-cyclopropylmorpholin2-yl] methyl} amino) -3-nitrophenyl] sulfonyl} -2- (1H-pyrrole [2 , 3-b] pyridin-5-yloxy) benzamide; 4- (4 - ((2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(4 - ([(trans-4cyanocyclohexyl) methyl ] amino} -3-nitrophenyl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; Trans-2 - [(6-amino-5-chloropyridin-3-yl) oxy] -4- (4 - ([2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -N - ((4 - [(4-morfolin -4-ylcyclohexyl) amino] -3-nitrophenyl} sulfonyl) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-i ImethylJpiperazin-1 -yl) -N - ([4 - ((((3R) -1 - [2fluoro-1- (fluoromethyl) ethyl] pyrrolidin-3-yl} amino) -3-nitrophenyl] sulfonyl} -2- (1 H-pyrrole [2 , 3-b] pyridin-5yloxy) benzamide; Trans-N - (((5-chloro-6 - [(4-hydroxycyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2 (4 -chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) -2- (1 H-indazol-4yloxy) benzamide; N - ((3-chloro-4- [ (trans-4-hydroxycyclohexyl) methoxy] phenyl} sulfonyl) -4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) - 2- (1H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide; N - ((5-chloro-6 - [(trans-4-hydro xicyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2 (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 -i IJmetylJpiperazin-1-yl) -2- [ (6-fluoro-1 H-indazol-425/55 yl) oxy] benzamide; 2 - [(6-amino-5-chloropyridin-3-yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex1 -en-1-yl] methyl} piperazin- 1 -yl) -N - [(4 - {[trans-4- (morpholin-4-yl) cyclohexyl] amino} -3nitrophenyl) sulfonyl] benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - [(4 - {[(cis-4- hydroxy-4-methylcyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] - 2- (1 H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - ({5-cyano-6 - [(4 -fluorotetrahydro-2H-pyran-4-yl) methoxy] pyridin-3ylsulfonyl) -2- (1 H-indazol-4-yloxy) benzamide; N - [(5-chloro-6 - {[4-fluoro-1 - (oxetan-3-yl) piperidin-4yl] methoxy} pyridin-3-yl) sulfonyl] -4- (4 - {[2- ( 4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2 - [(6-amino-5-chloropyridin- 3- yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - ({3 -nitro-4 [(tetrahydro-2H-pyran-4-ylmethyl) amino] phenyl} sulfonyl) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1 -en-1 -yl] methyl} piperazin-1 -yl) -N - ({4 - [(4-methylpiperazin-1 - il) amino] -3nitrophenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; Trans-4- (4 - {[2- (4-chlorophenyl) - 4.4- dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(4 - {[(4-methoxycyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1 H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; T rans-4- (4 - {[2- (4-chlorophenyl) - 4.4- dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - ({4 - [(4-morpholin-4-ylcyclohexyl) amino] -3nitrophenyl} sulfonyl) -2- (1H- pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - ({4 - [(4-fluorotetrahydro-2H- pyran-4-yl) methoxy] -3nitrophenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2 - [(6-amino-5-chloropyridin-3yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-ylmethyl} piperazin-1 -yl) -N - [(4 - {[(3R) -1 (2,2-difluoroethyl) pyrrolidin-3-yl] amino} -3-nitrophenyl) sulfonyl] benzamide; N - ({5-chloro-6 - [(trans-4hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4- dimethylcyclohex- -en-1-yl] methyl} piperazin-1-yl) -2- (1 H-indazol-4-yloxy) benzamide; N - ({5-chloro-6 - [(cis-1-fluor-4hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) - 4,4-dimethylcyclohex- 1- en-1-yl] methyl} piperazin-1-yl) -2- (1H-indazol-4-yloxy) benzamide; 2 - [(6-amino-5-chloropyridin-3-yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl) piperazin-1-yl) -N - [(4 - {[(4methoxycyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] benzamide; N - ({5-chloro-6 - [(trans-1-fluoro-4hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) - 4,4-dimethylcyclohex1 -en-1-yl-methylpiperazi n-1-yl) -2- (1 H-indazol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1 -en-1 -yljmethyljiperiperin-1 -yl) -N - [(4 - {[(trans-4-hydroxy-4methylcyclohexyl) methyl ] amino} -3-nitrophenyl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2- [(3-amino-1 H-indazol-4-yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl } piperazin-1-yl) -N - [(4 - {[(trans-4-methoxycyclohexyl) methyl] amino} -3nitrophenyl) sulfonyl] benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - ({3-nitro-4 - [(2 -oxaspiro [3,5] non-7-ylmethyl) amino] phenyl} sulfonyl) -2- (1Hpirrolo [2,3-b] pyridin-5-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - ({5-cyano-6 - [(trans -4-hydroxy-4-methylcyclohexyl) methoxy] pyridin-326/55 yl} sulfonyl) -2- (1H-indazol-4-yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -2 - [(6-fluor-1H-indoi-5- il) oxy] -N - {[3-nitro-4 - ({[4- (oxetan-3-yl) morpholin-2yl] methyl} amino) phenyl] sulfonyl} benzamide; N - ({5-chloro-6 - [(trans-4-hydroxy-4methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4- dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -2 - [(6-tluoro-1H-inaazoi-4-ii) oxijoenzarniaa; 4- (4 - ((2- (4-cioroyen) -4,4 dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) -N - [(5-cyano-6 - {[4- fluoro-1 - (oxetan-3-yl) piperidin- 4-yl] methoxy} pyridin-3-yl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 2 - [(6-amino-5-chloropyridin-3-yl) oxy] -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl) piperazin- 1-yl) -N [(4 - {[(4_hydroxycyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] benzamide; N - ({5-chloro-6 - [(trans-4hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -2 - [(3-chloro-1H-indazol-4-yl) oxy] - 4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) benzamide; 4- [4 - {[2- (4-chlorophenyl) - 4,4-dimethylcyclohex-1-en-1-yl] methyl} ( 2 H 8 ) piperazin-1-yl] -N - ({3-nitro-4 - [(tetrahydro-2H-pyran-4ylmethyl) amino] phenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; N - ({5-chloro-6 - [(trans1-fluoro-4-hydroxy-4-methylcyclohexyl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) - 4,4 dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -2- (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide; 4 (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(6 - {[(cis-4- hydroxy-4methylcyclohexyl) methyl] amino} -5-nitropyridin-3-yl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1 -yl) -N - ({5nitro-6 - [(tetrahydro- 2H-pyran-4-ylmethyl) amino] pyridin-3-yl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) -N - ({6 [(trans-4-hydroxy -4-methylcyclohexyl) methoxy] -5- (trifluoromethyl) pyridin-3-yl} sulfonyl) -2- (1H-indazol-4yloxy) benzamide; 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -N - [(4 {[(cis-4- ethyl-4-hydroxycyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide; and 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) -2 (1H-indol-5-yloxy) - N - ({3-nitro-4 - [(tetrahydro-2H-pyran-4-ylmethyl) amino] phenyl} sulfonyl) benzamide. Each of the compounds and methods for making these compounds are described in US Serial No. 12 / 787,682, filed on May 26, 2010, US Serial No. 12 / 793,418, filed on June 3, 2010 which is a continuation in part of US Serial No. 12 / 631,404 filed on December 4, 2009 and US Serial No. 12 / 793,413 filed on June 3, 2010 which is a continuation in part of US Serial No. 12 / 631,367, filed on December 4, 2009 2009, the contents of which are incorporated here for reference. The Bcl-2 inhibitor compounds used in the methods of the present invention can also include a pharmaceutically acceptable salt form of a compound having Formula (I). The phrase pharmaceutically acceptable salt (s), as used herein, means those salts of the selective Bcl-2 inhibitors of the invention that are safe and effective for administration to a patient and that do not adversely affect the therapeutic qualities of the compound. Pharmaceutically acceptable salts include salts of basic or acidic groups. 27/55 cos present in the compounds of the invention. Pharmaceutically acceptable acid addition salts include, but are not limited to, hydrochloride, hydrobromide, iodate, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, pantothenate, bitartarate, salts ascorbate, succinate, maleate, gentisinate, fumarate, glucone, io, giucaronaio, sucrate, yormaio, òenzoaio, giuiarnate, irietanosuuOnaio, eianosuiionato, benzenosulfonate, p-toluenesulfonate and pamoate (ie, 1, 2-r-methylene bis -3naftoate)). Certain compounds of the invention can form pharmaceutically acceptable salts with various amino acids. Compatible base salts include, but are not limited to, aluminum, calcium, lithium, magnesium, potassium, sodium, zinc, and diethanolamine salts. For a review of pharmaceutically acceptable salts see Berge et al., 66 J. Farm. Know.,. 1-19 (1977), incorporated herein for reference in its entirety. The compounds used in the methods of the present invention can also comprise geometric isomers. The compounds of these inventions may contain carbon-carbon double bonds or carbon-nitrogen double bonds in the E or Z configuration, where the term "E" represents higher order substituents on the opposite sides of the carbon-carbon or carbon double bond -nitrogen and the term "Z" represents higher-order substituents on the same side of the carbon-carbon or carbon-nitrogen double bond as determined by the Cahn-Ingold-Prelog Priority Rules. The compounds of this invention can also exist as a mixture of the Έ "and" Z "isomers. Substituents around 20 of a cycloalkyl or heterocycloalkyl are designated to be of the cis or trans configuration. In addition, the invention contemplates the various isomers and mixtures thereof resulting from the arrangement of the substituents around an adamantane ring system. Two substituents around a single ring within an adamantane ring system are designated as being of Z or E relative configuration. For examples, see C. D. Jones, 25 M. Kaselj, R. N. Salvatore, W. J. le Noble J. Org. Chem. 1998, 63, 2758-2760 and E. L. Eliel, and S.H. Wilen. (1994) Stereochemistry of Organic Compounds. New lork, NI: John Wiley & Sons, Inc. The selective inhibitory compounds of Bcl-2 can also contain asymmetrically substituted carbon atoms in the R or S configuration, in which the terms R and S 30 are defined by the IUPAC 1974 Recommendations for Section E, Fundamental Stereochemistry, Pure Appl. Chem. (1976) 45.13-10. Compounds having asymmetrically substituted carbon atoms with equal amounts of R and S configurations are rancemic on these carbon atoms. Atoms with an excess of configuration over the others are attributed to the present configuration in the largest amount, such an excess of about 85% 35 90%, an excess of about 95% -99%, or an excess greater than about 99 %. Therefore, this invention includes rancemic mixtures, absolute and relative stereoisomers, and mixtures of absolute and relative stereoisomers. 28/55 The selective inhibitory compounds of Bcl-2 used in the methods of the present invention containing halves of NH, C (O) OH, OH or SH may have attached halves forming pro-drugs there. The halves forming prodrugs are removed by metabolic processes and release the compounds having the hydroxyl, amino or carboxylic acid released in vivo. The Pro5 drugs are useful for adjusting such iarmacokineic properties or their sensitivity and / or hydrophobicity, absorption in the gastrointestinal tract, bioavailability, tissue penetration, and release rate. The compounds used in the various modalities can also exist in the classified isotope form or enriched by isotope containing one or two atoms having an atomic mass or mass number different from the atomic mass or mass number most abundantly found in nature. Isotopes can be radioactive or non-radioactive isotopes. Isotopes of atoms such as hydrogen, carbon, phosphorus, sulfur, fluorine, chlorine, and iodine include, but are not limited to, 2 H, 3 H, 13 C, 14 C, 15 N, 18 0, 32 P, 35 S, 18 F, 36 CI, and 125 l. Compounds containing other isotopes of these and / or other atoms are within the scope of this invention. In another embodiment, compounds classified as isotopes contain deuterium ( 2 H), tritium ( 3 H) or 14 C isotopes. The isotope classified compounds of this invention can be prepared by general methods well known to a person skilled in the art. Such isotope classified compounds can be conveniently prepared by performing the procedures disclosed in the Examples disclosed here and in the Schemes by replacing a readily available isotope classified reagent with an unclassified reagent. In some cases, compounds can be treated with reagents classified as isotopes to exchange a normal atom with its isotope, for example, hydrogen for deuterium can be exchanged for a deuteric acid such as D2SO4 / D2O. In addition to the above, relevant and intermediate procedures are disclosed, for example, in Lizondo, J et al., Drugs Fut, 21 (11), 1116 (1996); Brickner, SJ et al., J Med Chem, 39 (3), 673 (1996); Mallesham, B et al., Org Lett, 5 (7), 963 (2003); PCT publications WO1997010223, W02005099353, WO1995007271, W02006008754; US Patent Nos. 7538189; 7534814; 7531685; 7528131; 7521421; 7514068; 7511013; and US Patent Application Publication Nos. 20090137457; 20090131485; 20090131363; 20090118238; 20090111840; 20090105338; 20090105307; 20090105147; 20090093422; 20090088416; and 20090082471, the methods are incorporated herein for reference. Compounds classified as isotopes of the invention can be used as standards to determine the effectiveness of Bcl-2 inhibitors in binding assays. The 35 stations containing isotopes have been used in pharmaceutical research to investigate the metabolic fate of compounds by assessing the mechanism of action and metabolic pathway of the parent compound classified as non-isotope (Blake et al. J. Farm. Sci. 64, 3, 29/55 367-391 (1975)). Such metabolic studies are important in the design of safety, effective therapeutic drugs, whether because of the active compound in vivo administered to the patient or because of the metabolites produced from a parent compound prove to be toxic or carcinogenic (Foster et al., Advances in Drug Research Vol. 14, pp. 2-36, ò Acaüeuuc press, Loúoon, laoó; Kaío et <ái., j. ί-αΟαιι & ϋ i <aciiOi <aiiíícàOi., 932 (1995); Kushneretal., Can. J. Fisiol. Pharmacol., 77, 79-88 (1999). In addition, no drugs containing non-radioactive isotopes, such as deuterated drugs called "heavy drugs", can be used to treat diseases and conditions related to Bcl-2 activity. Increasing the amount of an isotope present 10 in a compound above its natural abundance is called enrichment. Examples of the enrichment amount include about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 16, 21, 25, 29, 33, 37, 42, 46, 50, 54, 58, 63, 67, 71, 75, 79, 84, 88, 92, 96, at about 100 mol%. The replacement of up to about 15% of the normal atom with a heavy isotope has been carried out and maintained for a period of days to weeks in mammals, including 15 rodents and dogs, with minimal observance of side effects (Czajka DM and Finkel AJ, Ann. Nl Acad. Sci. 1960 84: 770; Tomson JF, Ann. New lork Acad. Sci. 1960 84: 736; Czakja DM et al., Am. J. Fisiol. 1961 201: 357). An acute substitution of as high as 15% -23% in human fluids with deuterium was found not to be the cause of toxicity (Blagojevic N et al. In Dosimetri & Treatment Planning for Neutron Capture 20 Terapi, Zamenhof R, Solares G and Harling Eds. 1994. Advanced Medical Publishing, Madison Wis. Pp.125-134; Diabetes Metab. 23: 251 (1997)). The classification of a drug's stable isotopes can alter its physicochemical properties such as pKa and liposolubility. These effects and changes can affect the pharmacodynamic response of the drug molecule if the replacement of the isotope affects the region involved in a binding receptor interaction. While some of the physical properties of a molecule classified as stable isotope are different from those not classified, the biological and chemical properties are the same, an important exception: due to the increased mass of the heavy isotope, any bond involving the heavy isotope and another atom will be more stronger than the same bond between the light isotope and that atom. Therefore, the incorporation of an isotope into a metabolism or enzyme transformation site will potentially delay such reactions by altering the pharmacokinetic profile or the relative effectiveness for the non-isotopic compound. Current methods can be incorporated into a pro-drug form of the selective inhibitor compound of Bcl-2. Prodrugs are derivatives of an active drug designed to improve some identified unwanted physical or biological properties. Physical properties are usually solubility (aqueous solubility or not enough lipid or too much) or related stability, while problematic biological properties 30/55 include poor bioavailability or very fast metabolism which itself can be related to physiochemical property. Prodrugs are usually prepared by: a) ester formation, hemi esters, carbonate esters, nitrate esters, amides, hydroxanic acids, carbamates, imines, Mannich bases, phosphates, phosphate esters, and □ drug enammas active, b) functionalization of the drug with functional groups of azo, glycoside, peptide, and ether, c) use of forms of aminal, hemi-aminal, polymers, salts, complexes, phosphoramides, acetals, hemiacetals and ketals. For example, see Andrejus Korolkovas's, Essentials of Medicinal Chemistri ', John Wilei-lnterscience Publications, John Wilei and Sons, New lork (1988), pp. 97-118, which is incorporated here in its entirety for reference. In addition, the methods of the current invention may involve administering compounds having Formula (i) by, for example, at least one mode selected from parenteral, subcutaneous, intramuscular, intravenous, intra-articular, intrabronchial, intraabdominal, intracapsular , intracartilaginous, intracavitary, intracelial, intracerebellar, intra15 cerebroventricular, intracólico, intracervical, intragastric, intrahepatic, intramoocardial, intraosteal, intrapelvic, intrapericardíaco, intraperitoneal, intrapleural, intraprostatic, intrapulmonaria, intraretal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, and transdermal. As previously stated, the "therapeutically effective amount" of the current invention refers to that amount of the compound being administered sufficiently to prevent the development of or to some extent alleviate one or more of the symptoms of the condition or disorder being treated. The therapeutically effective amounts of the compounds having Formula (I) depend on the treatment container, the disorder being treated and the severity of the treatment, the composition containing the compound, the time of administration, the route of administration, the duration of treatment, the potency of the compound, its release rate and whether or not it is co-administered. Generally, the methods of the present invention involve administering a dose of the selective Bcl-2 inhibitor ranging from about 0.001 mg / kg to about 1000 mg / kg. In one embodiment, the methods involve administering a dose of the selective Bcl-2 inhibitor ranging from about 0.01 mg / kg to about 500 mg / kg. In yet another embodiment, the methods involve administering a dose of the selective Bcl-2 inhibitor ranging from about 0.1 mg / kg to about 300 mg / kg. The methods of the current invention may have illustrated the improved efficacy in treating established diseases such as SLE, lupus nephritis, and Sjogren's Syndrome compared to methods currently known in the art due to the fact that the 35 compounds disclosed here may selectively inhibit the protein Bcl-2. The Bcl-2 protein family is a group of proteins that have a regulatory effect on various functions of homeostasis and development, such as apoptosis (programmed cell death). The Bcl family 31/55 includes other proteins including Bcl-x L and Bcl-w. However, inhibition of the Bcl-x L protein has been shown to have an adverse impact on platelet counts, in some cases resulting in thrombocytopenia. The selective inhibitory compounds of Bcl-2 having Formula (I) have shown a higher binding affinity (as evidenced by the low Kj values) for Bcl-2 compared to the other proteins in the lamina bci-2, such as bcix L and Bcl-w. As such, the methods of the current invention provide the advantages of inhibiting Bcl-2 protein, a decreasing risk of side effects associated with inhibition of BcI-Xl and Bcl-w. The binding affinity for the various proteins is measured as a K it value which represents the amount of the compound required to inhibit a physiological process or compound (such as the protein) by 50%. The selective Bcl-2 compounds used in the methods of the present invention generally have a binding affinity (Kj) of less than about 1 micromolar, less than about 500 nanomolar, less than about 400 nanomolar, less than about 300 nanomolar, less than about 200 nanomolar, less than about 100 nanomolar, less than about 50 nanomolar, less than about 25 nanomolar, less than about 10 nanomolar, less than about 5 nanomolar, less than about 1 nanomolar, less than about 900 picomolar, less than about 800 picomolar, less than about 700 picomolar, less than about 600 picomolar, less than about 500 picomolar, less than about 400 picomolar, less than about 300 picomolar, less than about 200 picomolar, and less than about 100 picomolar for Bcl-2. The selective Bcl-2 inhibitors used in the methods of the present invention selectively bind to and elicit a response in Bcl-2 proteins at much lower concentrations than those required to bind to and elicit a response in Bcl-x L. As such, when the selective Bcl-2 inhibitor is administered to the patient, the inhibitor is more inclined to inhibit Bcl-2, rather than Bcl-x L. The selective inhibitors used in the methods of the present invention tend to have a competitive binding affinity (K>) for Bcl-2 that is at least about 500, at least about 1000, at least about 2000, at least about 2500 at least about 3000, at least about 3500, and at least about 4000 times less than the binding affinity for Bcl-x L. As such, even at low concentrations (ie, picomolar concentrations), the selective Bcl-2 inhibitor will bind to and inhibit the Bcl-2 protein. In addition, the methods of the invention include administering compounds having Formula (I) with or without an excipient. Excipients include, for example, encapsulated materials or additives such as absorption accelerators, antioxidants, binders, buffers, coating agents, coloring agents, diluents, disintegrating agents, emulsifiers, extenders, fillers, flavoring agents, humectants, lubricants, perfumes, condoms, propellants, release agents, sterilizing agents, sweeteners 32/55, solubilizers, wetting agents and mixtures thereof. Excipients for preparing compositions comprising a compound of Formula (I) to be administered orally in a solid dosage form include, for example, agar, alaaminic acid, aluminum hydroxide, benzyl alcohol, benzyl benzoate, ó 1.3 - gucoi buuíii, camomeros, oieo beaver, cellulose, oio ceiosis, cocoa butter, corn starch, corn oil, cottonseed oil, cross povidone, diglycerides, ethanol, ethyl cellulose, ethyl laureate, ethyl oleate, fatty acid esters, gelatin, germ oil, glucose, glycerol, peanut oil, hydroxypropylmethyl cellulose, isopropanol, isotonic saline, lactose, magnesium hydroxide, magnesium stearate, malt, mann10 tol, monoglycerides, olive oil, olive oil peanuts, potassium phosphate salts, potato starch, povidone, propylene glycol, Ringer's solution, safflower oil, sesame oil, sodium carboxymethyl cellulose, sodium phosphate salts, s sodium lauryl ulfate, sodium sorbitol, soybean oil, stearic acids, steraryl fumarate, sucrose, surfactants, talc, tragacanth, tetrahydrofurfuryl alcohol, triglycerides, water, and mixtures thereof. Excipients for preparing compositions comprising a compound of this invention having Formula (I) to be administered ophthalmically or orally in liquid dosage forms include, for example, 1,3-butylene glycol, castor oil, corn oil, oil cottonseed seed, ethanol, sorbitan fatty acid esters, germ oil, peanut oil, glycerol, isopropanol, olive oil, polyethylene glycols, propylene glycol, sesame oil, water and mixtures thereof. Excipients for the preparation of compositions comprising a compound of this invention having Formula (I) to be administered osmotically include, for example, chlorofluorohydrocarbons, ethanol, water and mixtures thereof. Excipients for the preparation of compositions comprising a compound of the invention having Formula (I) to be administered parenterally include, for example, 1,3-butanediol, castor oil, corn oil, cottonseed oil, dextrose, oil germ oil, peanut oil, liposomes, oleic acid, olive oil, peanut oil, Ringer's solution, safflower oil, sesame oil, soybean oil, USP or isotonic sodium chloride solution, water and mixtures thereof. Excipients for preparing compositions comprising a compound of this invention having Formula (I) to be administered rectally or vaginally include, for example, cocoa butter, polyethylene glycol, wax and mixtures thereof. The methods of the current invention encompass methods for administering a selective Bcl-2 inhibitor alone or in combination with other therapeutic products. Several proteins have been implicated in general to autoimmune and inflammatory responses. Therefore, 35 it may be possible to combine selective Bcl-2 inhibitors with compounds capable of altering the function of other proteins generally implicated in autoimmune and inflammatory responses. Examples of proteins associated with the autoimmune and inflammatory response 33/55 include C5, CCL1 (I-309), CCL11 (eotaxin), CCL13 (mcp-4), CCL15 (MIP-1d), CCL16 (HCC-4), CCL17 (TARC), CCL18 (PARC), CCL19 , CCL2 (mcp-1), CCL20 (MIP-3a), CCL21 (MIP-2), CCL23 (MPIF-1), CCL24 (MPIF-2 / eotaxin-2), CCL25 (TECK), CCL26, CCL3 (MIP -1a), CCL4 (MIP-1b), CCL5 (RANTES), CCL7 (mcp-3), CCL8 (mcp-2), CXCL1, CXCL10 5 (iF-iÜ), CXCLIÍ (i-íAú / ír- ») , UÀoLiZ pwr i), VÀU.IJ, ^ aULí4, UAUL2, GXvLó, CXCL5 (ENA-78 / LIX), CXCL6 (GCP-2), CXCL9, IL13, IL8, CCL13 (mcp-4), CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CX3CR1, IL8RA , XCR1 (CCXCR1), IFNA2, IL10, IL13, IL17C, IL1A, IL1B, IL1F10, IL1F5, IL1F6, IL1F7, IL1F8, IL1F9, IL22, IL5, IL8, IL9, LTA, LTB, MIF, SCIE1 (endocytokine activating monocyte activating monocyte ), SPP1, TNF, 10 TNFSF5, IFNA2, IL10RA, IL10RB, IL13, IL13RA1, IL5RA, IL9, IL9R, ABCF1, BCL6, C3, C4A, CEBPB, CRP, ICEBERG, IL1R1, IL1RN, IL8RB, LTB4R, TOLLIP, FADD, IRAK1, IRAK2, MID88, NCK2, TNFAIP3, TRADD, TRAF1, TRAF2, TRAF3, TRAF4, TRAF5, TRAF6, ACV1 ACVR2B, ACVRL1, CD28, CD3E, CD3G, CD3Z, CD69, CD80, CD86, CNR1, CTLA4, CISLTR1, FCER1A, FCER2, FCGR3A, GPR44, HAVCR2, OPRD1, 15 P2RX7, TLR2, TLR3, TLR4, TLR7 , TLR8, TLR9, TLR10, BLR1, CCL1, CCL2, CCL3, CCL4, CCL5, CCL7, CCL8, CCL11, CCL13, CCL15, CCL16, CCL17, CCL18, CCL19, CCL20, CCL21, CCL22, CCL23, CCL24, CCL25, CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR5, CCR6, CCR5 CCR8, CCR9, CX3CL1, CX3CR1, CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL10, CXCL11, CXCL12, CXCL13, CXCR4, GPR2, SCIE1, SDF2, XCL1, XCL2, XPR1, BMPR1, 20 , C19orf10 (IL27w), CER1, CSF1, CSF2, CSF3, DKFZp451J0118, FGF2, GFI1, IFNA1, IFNB1, IFNG, IGF1, IL1A, IL1B, IL1R1, IL1R2, IL2, IL2RA, IL2RB, IL2RG, IL3, IL4, IL4R, IL5, IL5RA, IL6, IL6R, IL6, IL7 IL8RA, IL8RB, IL9, IL9R, IL10, IL10RA, IL10RB, IL11, IL11RA, IL12A, IL12B, IL12RB1, IL12RB2, IL13, IL13RA1, IL13RA2, IL15, IL15RA, IL16, IL17, IL17R, IL18, IL18R1, IL19, IL18R1, IL19 KITLG, LEP, 25 LTA, LTB, LTB4R, LTB4R2, LTBR, MIF, NPPB, PDGFB, TBX21, TDGF1, TGFA, TGFB1, TGFB1I1, TGFB2, TGFB3, TGFBI, TGFBR1, TGFBR2, TGFBR3, T1L, TNF, TNFRSF1A, TNFRSF1B, TNFRSF7, TNFRSF8, TNFRSF9, TNFRSF11A, TNFRSF21, TNFF, TNF, TNF, TNF, TNF, FGF, PLGF, DLL4, and NPR-1. It is to be understood that the invention can be used alone or in combination with an additional agent, for example, a therapeutic agent, the therapeutic agent being selected by an expert for the intended purpose. For example, the additional agent may be a therapeutic agent, recognized in the art as being useful for treating the disease or condition being treated in the present invention. The additional agent can also be an agent that imparts a beneficial attribute to the therapeutic composition, for example, an agent that affects the viscosity of the composition. It should also be understood that the combinations that are to be included within this invention are those combinations comprising a treatment with the inhibitors 34/55 selective Bcl-2 described here and one or more additional therapeutic agents. Combinations to treat autoimmune and inflammatory diseases are non-steroidal anti-inflammatory drugs also referred to as NSAIDS which include ibuprofen-type drugs. Other combinations are corticosteroids including prednisolone; side effects oem uotineciucs <jü ubo uc eaíeiuiúda püUcdTi sct cu úíó íúcóhíu eliminated by tapering the required steroid dose when treating patients in combination with this invention. Non-limiting examples of therapeutic agents for lupus with this invention can be combined including the following: cytokine suppressive anti-inflammatory drug (CSAIDs); antibodies to or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL -8, IL-9, IL-10, IL-12, IL-13, IL-15, IL-16, IL-17A, IL-17F, IL-18, IL-21, IL-22, IL-23 , IL-25, IL33, interferons (e.g., alpha, beta, gamma, etc.), Tweak, BAFF / BLiS, April, chemokines. The invention can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD16, CD19, CD20, CD22, CD25, CD28, CD30, CD32, CD40, 15 CD45, CD47, CD52, CD54, CD64, CD69, CD72, CD79, CD80 (B7,1), CD86 (B7,2), CD90, CD100, CD200, CTLA, ICOS-1, B7RP, BR3, TACI, BCMA, or other linkers including CD154 (gp39 or CD40L). The invention can also be combined with agents, such as mycophenolate mofetil (MMF), cytoxane, Bortezomib, methotrexate, 6-MP, azathioprine sulfasalazine, mesala20 zine, chloroquine / hydroxychloroquine olsalazine, penicillamine, aurothiomalate (intramuscular), intramuscular and oral, thyrathiomalate (intramuscular) colchicine, corticosteroids (oral, inhaled and local injection), selective glucocorticoid receptor modulators (SGRMs), beta-2 adrenoreceptor agonists (salbutamol, terbutaline, salmeterol), xanthines (theophylline, aminophylline), chromoglycate, nedocromil, katrotene, and katrotene, katrotene, and katroten, oxytropium, cyclosporine, FK506, rapamycin, mycophenolate mofetil, nominated leflu25, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adensosin agonists, antithrombotic agents, complement inhibitors, signaling agents, adrenergic agents by pro-inflammatory cytokines such as TNF-q or IL-1 (for example, IRAK, NIK, IKK, p38 or MAP kinase inhibitors), enzyme inhibitors converting IL-1 β, btk inhibitors, 30 sik inhibitors, inhibitors of the PKC family, enzyme inhibitors converting TNF- (TACE), inhibitors of T cell signaling such as inhibitors of kinase, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, enzyme converting angiotensin inhibitors, soluble cytokine receptors and derivatives thereof (for example, soluble p55 or p75 TNF receptors and p75TNFRIgG derivatives (Enbrel ™ and p55TNFRI and p55TNFRI )), sIL-IRI, slL-1 R11, slL-6R), anti-inflammatory cytokines (for example, IL-4, IL-6, IL-10, IL-11, IL12, IL-13, IL-17, IL -18, IL-33 and TGFP), celecoxib, folic acid, hydroxychloroquine sulfate, rofecoxib, etanercept, infliximab, naproxen, valdecoxib, sulfasalazine, methylpred 35/55 nisolonea meloxicam, methylprednisolone acetate, sodium aurothiomalate, aspirin, triamcinolone acetonide, propoxyphene napsylate / apap, folate, nabumetone, diclofenac, piroxicam, etodolac, sodium diclofenac, oxydapathone, oxydaphazine, oxycypro sodium / misoprostol, fentanyl, anakinra, human recombinant, tramadol HCI, salsalate, sulindac, cyanocobalamin / fa / pyridoxine, acetaminophen, sodium alendronate, prednisolone, morphine sulfate, lidocaine hydrochloride, indomethacin, glucosamine sulfate / chondroitin , sulfadiazine, oxycodone HCI / acetaminophen, olopatadine HCI, misoprostol, sodium naproxen, omeprazole, cyclophosphamide, rituximab, IL-1 TRAP, MRA, CTLA4-IG, IL-18 BP, anti-IL-18, Anti-IL15, BIRB -796, SCIO-469, VX-702, AMG-548, VX-740, Roflumilast, IC-485, CDC-801, and Mesopram. The combinations include methotrexate or leflunomide, cyclosporine and S1P agonists. Examples of therapeutic agents for SLE (Lupus) and lupus nephritis, in which the invention can be combined include the following: NSAIDs, for example, diclofenac, naproxen, ibuprofen, piroxicam, indomethacin; COX2 inhibitors, for example, Celecoxib, rofecoxib, valdecoxib; anti-malarials, for example, hydroxychloroquine; Steroids, for example, prednisone, prednisolone, budesonide, dexamethasone; Cytotoxic, for example, azathioprine, cyclophosphamide, mycophenolate mofetil, methotrexate; PDE4 inhibitors or purine synthase inhibitors, for example, Cellcept. Binding proteins incorporated in the methods of the invention, can also be combined with agents such as sulfasalazine, 5-aminosalicylic acid, olsalazine, Imuran and agents that interfere with the synthesis, production or proinflammatory action of cytokines such as IL-1, for example, IL-Ιβ type caspase inhibitors converting enzyme and IL-1 ra inhibitors. The invention can also be used with inhibitors signaling T cells, for example, tyrosine kinase inhibitors; or molecules that target T cell activation molecules, for example, CTLA-4IgG or antibodies of the anti-B7 family, antibodies of the anti-PD-1 family. The invention can be combined with IL-11 or anti-cytokine antibodies, for example, phonotolizumab (anti-IFNg antibody), anti-interferon alpha, or anti-receptor receptor antibodies, for example, anti-IL-6 receptor antibody and antibodies for de cell surface molecules. The invention can also be used with HMGB1, HDGF inhibitors. The invention can also be used with Toll-like receptor inhibitors 1, 2, 3, 4, 7, and 9. The invention can also be used with BDCA-1, 2 and 3 dendritic cell marker inhibitors. it can also be used with agents that promote regulatory T cell function. The invention can also be used with LJP 394 (abetimus), agents that inhibit complement, for example, anti-C5, anti-C5a, destroy or inactivate B cells, for example, Rituximab (anti-CD20 antibody), limfostat- B (anti-BliS antibody), anti-CD22, TNF antagonists, for example, anti-TNF antibodies, Adalimumab (PCT Publication No. WO 97/29131; HUMIRA), CA2 (REMICADE), CDP 571, TNFR-lg constructs, (p75TNFRIgG (ENBREL ) and 36/55 p55TNFRIgG (LENERCEPT)) and inhibitors of other members of the bcl-2 family such as Bclx L , Mcl-1, A-1 etc. Examples of therapeutic agents used to treat Sjogren's Syndrome, which can be combined with selective Bcl-2 inhibitors include, but are not limited to, artificial tears, cyclosporine, cevimeline, pilocarpine, NSAIDs, corticosteroids, immunosuppressants, anti- disease-modifying rheumatic diseases (DMARDs) such as methotrexate, and hydroxychloroquine. It may also be possible to combine the selective Bcl-2 inhibitor with a binding protein to further improve the compound to the desired site of action. In one embodiment the binding protein used in the methods of the invention has a constant rate (Ko n ) for one or more targets selected from a group consisting of: at least about 10 2 M ' 1 s'1; at least about 10 3 M ' 1 s'1; at least about 10 4 M ' 1 s'1; at least about 10 5 M ' 1 s'1; and at least about 10 6 M ' 1 s' 1 , as measured by a surface plasmon resonance. In one embodiment, the binding protein of the invention has a constant rate (Kon) for one or more targets between 10 2 M ' 1 s' 1 and 10 3 M ' 1 s'1; between 10 3 M ' 1 s' 1 and 10 4 M ' 1 s'1; between 10 4 M ' 1 s' 1 and 10 5 M _1 s · 1 ; or between 10 5 M ' 1 s' 1 and 10® M ' 1 s' 1 , as measured by surface plasmon resonance. In another embodiment, the binding protein has a constant discount rate (Koff) for one or more targets selected from a group consisting of: at most about 10 ' 3 s'1; at most about 10V; at most about 10 ' 5 s'1; and at most about 10®s' 1 , as measured by a surface plasmon resonance. In one embodiment, the binding protein of the invention has a constant discount rate (Koff) for one or more targets from 10 ' 3 s' 1 to 10V; from 10 ' 4 s' 1 to 10 ' 5 s'1; or from 10 ' 5 s' 1 to 10¾ 1 . as measured by a surface plasmon resonance. In another embodiment, the binding protein has a constant dissociation (K D ) for one or more targets selected from a group consisting of: at most about 10 ' 7 M; maximum of about 10® M; at most about 10 ' 9 M; at most about 10 ' 1 ° M; at most about 10 '11 M; at most about 10'12 M; and at most 10 "13 M. In one embodiment, the binding protein of the invention has a dissociation constant (KD) to its targets of 10" 7 M to 10 "8 M; from 10® M to 10 ' 9 M; from 10 ' 9 M to 10' 1 ° M; from 10 ' 1 ° to 10' 11 M; from 10 '11 M to 10' 12 M; or from 10 '12 to Μ 10' 13 M. In another aspect, the binding protein is a conjugate comprising a binding protein and an agent selected from the group consisting of an immunoadhesion molecule, an imaging agent, a therapeutic agent, and a cytotoxic agent. Examples of imaging agents include a radiolabel, an enzyme, a fluorescent classification, a luminescent classification, a bioluminescent classification, a magnetic classification, and biotin. Examples of radiolabels include 3H, 14C, 35S, 37/55 90Ι, 99Tc, 1111η, 1251, 1311, 177Lu, 166Ho, and 153Sm. In yet another embodiment, the cytotoxic or therapeutic agent is selected from a group consisting of an antimetabolite, an alkylating agent, an antibiotic, a growth factor, a cytokine, an anti-angiogenic agent, an anti-mitotic agent, an anthracycline, toxin, and an apoptotic agent. In another aspect, the binding protein is a crystallized binding protein, for example, a pharmaceutically controlled release crystal free from the carrier. In yet another embodiment, the crystallized binding protein has a longer half-life in vivo than the soluble homolog of a binding protein. In yet another embodiment, the crystallized binding protein retains biological activity. In another embodiment, the binding protein described here is glycosylated. For example, glycosylation is a human glycosylation pattern. Another aspect of the invention pertains to a nucleic acid encoding any of a binding protein disclosed herein. Another embodiment provides a vector comprising the nucleic acid disclosed here where the vector is selected from a group consisting of pcDNA; pTT (Durocher et al., Nucleic Acids Research 2002, Vol 30, No.2); pTT3 (pTT with additional multiple cloning sites; pEFBOS (Mizushima, S. and Nagata, S., (1990) Nucleic Acids Research Vol 18, No. 17); pBV; pJV; pcDNA3.1 TOPO, pEF6 TOPO and pBJ. In one embodiment, the vector is a vector disclosed in US Patent Application Serial No. 61 / 021,282. In another aspect, a host cell is transformed with the vector disclosed here. In one embodiment, the host cell is a prokaryotic cell. In another embodiment, the host cell is an E.Coli. In a related embodiment the host cell is a eukaryotic cell. In another embodiment, the eukaryotic cell is selected from a group consisting of a protist cell, an animal cell, an avian cell, a plant cell and a fungal cell. In yet another embodiment, the host cell is a mammalian cell including, but not limited to, CHO, COS; NS0, SP2, PER.C6 or a fungal cell such as Saccharomices cerevisiae; or an insect cell such as Sf9. Another aspect of the invention provides a method of producing a binding protein disclosed herein comprising culturing any of the host cells also disclosed here in a culture medium under conditions sufficient to produce a binding protein. In one embodiment, 50% -75% of the binding protein produced by this method is a specific double tetravalent binding protein. In a particular embodiment, 75% -90% of the binding protein produced by this method is a specific double tetravalent binding protein. In a particular embodiment, 90% -95% of the produced binding protein is a specific double tetravalent binding protein. 38/55 One embodiment provides a composition for releasing a binding protein where the composition comprises a formulation which in part comprises a crystallized binding protein, as disclosed herein, and an ingredient, and at least a polymeric carrier. For example, the polymeric carrier is a polymer selected from one or more of the selected group consisting of: poii (acrylic acid ;, pon (cyanoacrylates), poly (amino acids), poly (anhydrides), poly (depsipeptide), poly (esters), poly (lactic acid), poly (co-glycolic lactic acid) or PLGA, poly (b-hydroxybutriate), poly (caprolactone), poly (dioxanone); poly (ethylene glycol), poly ((hydroxypropyl) methacrylamide , poly [(organo) phosphazene], poly (ortho esters), poly (vinyl alcohol), poly (vinyl pyrrolidone), maleic anhydride-vinyl ether copolymers, pluronic polyols, albumin, alginate, cellulose and cellulose derivatives, collagen, fibrin, gelatin, hyaluronic acid, oligosaccharides, glycaminoglycans, sulfated polysaccharides, mixtures and copolymers thereof, for example, the ingredient is selected from a group consisting of albumin, sucrose, trehalose, lactitol, gelatin, hydroxypropyl-β- cyclodextrin, me toxipolethylene glycol and polyethylene glycol. Another embodiment provides a method for treating a mammal comprising the step of administering to a mammal an effective amount of the composition disclosed herein. The invention also provides a pharmaceutical composition comprising a binding protein, as disclosed herein, and a pharmaceutically acceptable carrier. In another embodiment, the pharmaceutical composition comprises at least one additional therapeutic agent for treating the disease. For example, the additional agent is selected from a group consisting of: a therapeutic agent, an imaging agent, a cytotoxic agent, an angiogenesis inhibitor (including, but not limited to, an anti-VEGF antibody or a VEGF-trap ), a kinase inhibitor (including, but not limited to, a KDR inhibitor and a TIE-2), a costimulation molecule blocker (including, but not limited to, an anti-B7,1, anti-B7, 2, CTLA4-lg, anti-CD20), an adhesion molecule blocker (including, but not limited to, an anti-LFA-1 antibody, an anti-E / L selectin antibody, a small molecule inhibitor), an antibody anti-cytokine or functional fragments thereof (including, but not limited to, an anti-IL-18, an anti-TNF, and an anti-IL-6 / cytokine receptor antibody), methotrexate, cyclosporine, rapamycin, FK506, a re30 detectable carrier or classifier, TNF antagonist, antirheumatic, muscle relaxant, narcotic, non-inflammatory drug steroid (NSAID), analgesic, anesthetic, sedative, local anesthetic, neuromuscular blocker, antimicrobial, antipsoriatic, corticosteroid, an anabolic steroid, erythropoietin, immunization, immunoglobulin, immunosuppressive, hormone de growth, a hormone replacement drug, a radiopharmaceutical, an antidepressant, an antipsychotic, a stimulant, an asthma medication, a beta agonist, an inhaled steroid, an epinephrine or analog, a cytokine and an antagonist cytokine. 39/55 In another aspect the invention provides a method for treating a patient suffering from a disease comprising the step of administering any of the binding proteins disclosed herein, concurrently, or after administration of a second agent, as discussed here. In a particular modality, the second agent is selected from a group that is very cohesive, uuuesoríiüei, íchW úü uieuciíiicíiíü epiuefiíiai, üüíiÍooebtóiüides, cyclosporine, sulfasalazine, aminosalicine, methazine, azalea, azalea, antioxidants, thromboxane inhibitors, IL-1 receptor antagonists, anti-IL-Ιβ mAbs, anti-IL-6 or IL-6 mAbs receptor, growth factors, elastase inhibitors, imidazole pyridinyl10 compounds, antibodies or TNF agonists , LT, IL-1, IL-2, IL-6, IL-7, IL-8, IL-12, IL-13, IL-15, IL-16, IL-18, IL-23, EMAP-II , GM-CSF, FGF, and PDGF, antibodies to CD2, CD3, CD4, CD8, CD-19, CD25, CD28, CD30, CD40, CD45, CD69, CD90 or their ligands, methotrexate, cyclosporine, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, ibuprofen, corticosteroids, prednisolone, phosphodiesterase inhibitors, adenosine agonists, agents 15 antithrombotics, complement inhibitors, adrenergic agents, IRAK, NIK, IKK, p38, MAP kinase inhibitors, IL-1 β converting enzyme inhibitors, TNFa converting enzyme inhibitors, T cell signaling inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, enzyme inhibitors converting angiotensin, soluble cytokine receptors, soluble TNF p55 receptor, soluble TNF p75 receptor, 20 slL-1 RI, slL-1 Rll, slL-6R, anti-inflammatory cytokines, IL-4, IL -10, IL-11, IL-13 and TGFβ. One aspect of the invention provides at least one anti-idiotype antibody to at least one binding protein of the present invention. The anti-idiotype antibody includes any protein or peptide containing a molecule that comprises at least a part of an immunoglobulin molecule such as, but not limited to, at least one complementary determining region (CDR) of a light or heavy chain or a part ligand binding region, a heavy chain or light chain region, a heavy chain or light chain variable region, a heavy chain or light chain constant region, a structure region, or any part thereof, which may be incorporated into a binding protein of the present invention. A binding protein of the invention can be used alone or in combination to treat such diseases. It is to be understood that a binding protein can be used alone or in combination with an additional agent, for example, a therapeutic agent, the additional sense agent selected by one skilled in the art for its intended purpose. For example, the additional agent may be an agent recognized in the art as being useful for the treatment of the disease or condition being treated by an antibody of the present invention. The additional agent can also be an agent that imparts a beneficial attribute to the therapeutic composition, for example, an agent that affects the viscosity of the composition. 40/55 It should also be understood that the combinations that are to be included within this invention are those combinations useful for their intended purpose. The agents set out below are illustrative for the purpose and are not intended to be limited. The combinations, which are part of this invention, can be the antibodies of the present invention ò and at least one ugenie óeieò <^ neuo α ράιίιϊ ss noiá cimuiaó. Comination also may include more than one additional agent, for example, two or three additional agents if the combination is such that the formed composition can perform its intended functions. Combinations to treat autoimmune and inflammatory diseases are non-steroidal anti-inflammatory drugs also referred to as NSAIDS which include ibuprofen-type drugs. Other combinations are corticosteroids including prednisolone; the well-known side effects of steroid use can be reduced or even eliminated by tapering the required dose when treating patients in combination with the DVD Igs of this invention. Non-limiting examples of therapeutic agents for rheumatoid arthritis with which an antibody, or an antibody portion of the invention can be combined include the following: anti-inflammatory cytokine drugs (CSAIDs); antibodies to or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL- 8, IL-15, IL-16, IL-18, IL-21, IL-23, interferons, EMAP-11, GM-CSF, FGF, and PDGF. The binding proteins incorporated in the methods 20 of the invention, or antigen binding parts thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD80 (B7.1), CD86 (B7.2), CD90, CTLA or other ligands including CD154 (gp39 or CD40L). Combinations of therapeutic agents can interfere at different points in the subsequent and autoimmune inflammatory cascade; examples include chimeric TNF antagonists, human or humanized TNF antibodies, Adalimumab, (WO 97/29131), CA2 (Remicade ™), CDP 571, and soluble p55 or p75 TNF receptors, derived therefrom, (p75TNFR1gG (Enbrel ™) or p55TNFR1gG (Lenercept), and also TNFα converting enzyme inhibitors (TACE); IL-1 inhibitors similarly (conver30 enzyme inhibitors having Interleukin-1, IL-1 RA etc.) can be effective for the same reason. The other combinations include Interleukin 11. Yet another combination includes the autoimmune response protagonists who can act in parallel to, depending on or in conjunction with IL-12 function; especially they are 1L-18 antagonists including IL-18 antibodies or soluble IL18 receptors, or IL-18 binding proteins. It has been shown that IL-12 and IL-18 have over 35 positions, but distinct functions and a combination of antagonists for both may be the most effective, yet another combination is non-depleting inhibitors. anti-CD4 action, yet other combinations include CD80 (B7,1) or CD86 co-stimulatory pathway antagonists 41/55 (B7,2) including antibodies, soluble receptors or antagonistic ligands. The binding protein incorporated in the methods of the invention can also be combined with agents, such as methotrexate, 6-MP, azathioprine sulfasalazine, mesalazine, olsalazine chloroquine / hydroxychloroquine, pencilamine, aurothiomalate (intramuscular and oral), azathioprine, coccinic, corticosteroids ( oral, inhaled and local injection), beta-2 adrenoreceptor agonists (salbutamol, terbutaline, salmeteral), xanthines (theophylline, aminophylline), chromoglycate, nedocromil, ketotifen, ipratropium and oxitrope, cyclosporine, FK506, rapamycin, rapamine, micamide, micamide; , for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adenosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents that interfere with pro-inflammatory cytokine signaling such as TNF-g or IL-1 ( for example, IRAK, NIK, IKK, p38 or MAP kinase inhibitors), enzyme inhibitors converting IL-Ιβ, TNFα converting enzyme inhibitors (TACE), T cell signaling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6 mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (for example , soluble p55 or p75 TNF receptors and the p75TNFRIgG derivatives (Enbrel ™ and p55TNFRIgG (Lenercept)), sIL-IRI, slL-1RII, slL-6R), anti-inflammatory cytokines (for example, IL-4, 1L-10, IL- 11, IL-13 and TGFa), celecoxib, folic acid, hydroxychloroquine sulfate, rofecoxib, etanercept, infliximab, naproxen, valdecoxib, sulfasalazine, methylprednisolone, meloxicam, methylprednisolone acetate, sodium acetate, triaxyl acetate, aspirin, sodium acetate, triaxyl acetate, aspirin, sodium acetate; / apap, folate, nabumetone, diclofenac, piroxicam, etodolac, sodium diclofenac, oxaprozine, oxycodone HCI, hydrocodone bitartrate / apap, sodium diclofenac / misoprostol, fentanyl, anakinra, r human combination, tramadol HCI, salsalate, sulindac, cyanocobalamin / fa / pyridoxine, acetaminophen, sodium alendronate, prednisolone, morphine sulfate, lidocaine hydrochloride, indomethacin, glucosamine sulf / chondroitin, amitriptyline HCI, sulfadiazine, oxycodone, oxycodone HCI, misoprostol, sodium naproxen, omeprazole, cyclophosphamide, rituximab IL-1 TRAP, MRA, CTLA4-IG, IL-18 BP, anti-IL-18, Anti-IL15, BIRB-796, SCIO-469, VX-702 , AMG-548, VX-740, Roflumilast, IC-485, CDC-801, and Mesopram. The combinations include methotrexate or leflunomide and in cases of severe or moderate rheumatoid arthritis, cyclosporine. Examples of therapeutic agents for SLE (Lupus) and lupus nephritis, in which the binding proteins incorporated in the methods of the invention can be combined include the following: NSAIDs, for example, diclofenac, naproxen, ibuprofen, piroxicam, indomethacin; COX2 inhibitors, for example, Celecoxib, rofecoxib, valdecoxib; antimalarials, for example, hydroxychloroquine; Steroids, for example, prednisone, prednisolone, budesonide, dexamethasone; Cytotoxic, eg, azathioprine, cyclophosphamide, mico 42/55 mofetil phenolate, methotrexate; PDE4 inhibitors or purine synthase inhibitors, for example, Cellcept. Binding proteins incorporated in the methods of the invention, can also be combined with agents such as sulfasalazine, 5-aminosalicylic acid, olsalazine, Imuran and agents that interfere with the synthesis, production or proinflammatory action of cytokines such as IL-1, for example, IL-1 β caspase inhibitors converting enzyme and IL-lra inhibitors. The binding proteins incorporated in the methods of the invention can also be used with T cell signaling inhibitors, for example, tyrosine kinase inhibitors; or molecules that target T cell activation molecules, for example, CTLA-4-lgG or antibodies of the anti-B7 family, antibodies of the anti-PD-1 family. The binding proteins incorporated in the methods of the invention can be combined with IL-11 or anti-cytokine antibodies, for example, phonotolizumab (anti-IFNg antibody), or anti-receptor receptor antibodies, for example, anti-IL-6 receptor antibody and antibodies to the B cell surface molecules. The antibodies of the invention or antigen binding part thereof can also be used with LJP 394 (abetimus), agents that deplete or inactivate B cells, for example, Rituximab (anti -CD20), limfostat-B (anti-BliS antibody), TNF antagonists, for example, anti-TNF antibodies, Adalimumab (PCT Publication No. WO 97/29131; HUMIRA), CA2 (REMICADE), CDP 571, TNFR-lg, (p75TNFRIgG (ENBREL) and p55TNFRIgG (LENERCEPT)) and bcl-2 inhibitors, because overexpression of bcl-2 in transgenic mice has been shown to cause phenotype-type lupus (see Marquina, Regina et al ., Journal of Immunologi (2004), 172 (11 ), 7177-7185), therefore, the inhibition is expected to have therapeutic effects. The pharmaceutical compositions of the invention can include a therapeutically effective amount or a prophylactically effective amount of a binding protein of the invention. A therapeutically effective amount refers to an effective amount, in dosages and for periods of time necessary, to achieve a desired therapeutic result. A therapeutically effective amount of a binding protein can be determined by a person skilled in the art and can vary according to factors such as the disease stage, age, sex and weight of the individual, and the ability of the binding protein to elicit a response desired in the individual. A therapeutically effective amount is also one in which any toxic or harmful effects of the antibody, or part of the antibody, are outweighed by the therapeutically beneficial effects. A prophylactically effective amount refers to an effective amount, in dosages and for periods of time necessary, to achieve a desired prophylactic result. Typically, since a prophylactic dose is used in subjects before or at an earlier stage of the disease, the prophylactically effective amount will be less than the therapeutically effective amount. Dosage regimens can be adjusted to provide a response of 43/55 be optimal (for example, a therapeutic or prophylactic response). For example, a single bolus can be administered, several divided doses can be administered over a period, or the dose can be proportionally reduced or increased by the requirements of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in unit dosage form to facilitate administration and uniformity of dosage. The dosage unit form as used herein refers to different physically compatible units as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined amount of the active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the unit dosage forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular prophylactic or therapeutic effect to be achieved, and (b) the limitations inherent in the art of the composition such as an active compound for the treatment of sensitivity in individuals. An exemplary non-limiting range for a therapeutically or prophylactically effective amount of a binding protein of the invention is 0.1-20 mg / kg, for example, 1-10 mg / kg. It should also be noted that dosage values may vary with the type and severity of the condition to be relieved. It is still to be understood that for any particular subject, the specific dosage regimens must be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the established dosage ranges here they are exemplary only and are not intended to limit the scope or practice of the claimed composition. It will be readily apparent to those skilled in the art that other compatible modifications and adaptations of the methods of the invention described here are obvious and can be made using compatible equivalents without departing from the scope of the invention or the modalities disclosed herein. Having now described the present invention in detail, it is more clearly understood with reference to the following examples, which are included for purposes of illustration only and are not intended to be limiting of the invention. Example 1: In Vitro Competitive Binding Affinity Assays for Bcl-2 Selective Inhibitors In order to test the selective binding affinity for Bcl-2 receptors, in vitro testing of certain selective Bcl-2 inhibitors was performed and compared to a non-selective Bcl-2 inhibitor. Specifically, two compounds: 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1en-1-yl] methyl} piperazin-1-yl) -N - ({3-nitro-4 - [(tetrahydro-2H-pyran-4-ylmethyl) amino] phenyl} sulfonyl) -2 (1H-pyrrolo [2,3-b] pyridin-5-yloxy) benzamide (hereinafter “Compound 1,” a selective inhibitor of Bcl-2); and N- (4- (4 - ((2- (4-chlorophenyl) -5,5-dimethyl-1-cyclohex-1-en-1-yl) methyl) piperazin-1-yl) benzoyl) 4- ( ((1 R) -3- (morpholin-4-yl) -1 - ((phenylsulfanyl) methyl) propyl) amino) -344/55 ((trifluoromethyl) sulfonyl) benzenesulfonamide (hereinafter “Compound 2,” a non-selective inhibitor of Bcl-2) were introduced into murine cells (FL5.12) engineered to depend on both Bcl-2 (FL5.12-Bcl-2) and Bcl-x L (FL5.12-Bcl-x L ) for survival. These compounds as well as the additional compounds listed in Table 1A have also been introduced into human tumor cell lines that have previously been shown to be predominantly dependent on both Bcl-2 (RS4; 11) and Bcl-xL (H146) for survival, and the effect of the compounds measured. A comparison of binding affinity for each of the compounds was performed to determine the target affinity, as measured by the Resolved Resonant Time Fluorescence Energy Transfer (TR FRET). The test was also performed to determine the effective concentration required to inhibit at least 50% of the target protein, as measured by the EC50 value, for all compounds (Ref. WO2010 / 138588A2). The results of the in vitro test for the compounds in Table 1A are provided in Table 1B below: Table 1A: Compound Number Listing and the name of the associated compound Compound Number Name 1 4- (4 - ([2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - ({3-nitro-4 - [(tetrahydro -2H-pyran-4ylmethyl) amino] phenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide 2 N- (4- (4 - ((2- (4-chlorophenyl) -5,5-dimethyl-1-cyclohex-1-en-1-yl) methyl) piperazin-1-yl) benzoyl) -4 - (( (1 R) -3- (morpholin-4-yl) -1 (((phenylsulfanyl) methyl) propyl) amino) -3 ((trifluoromethyljsulfonyl) benzenesulfonamide 3 3 is 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -2- (1H-indol-5-yloxy) - N - ({3-nitro-4 [(tetrahydro-2H-pyran-4ylmethyl) amino] phenyl} sulfonyl) benzamide. 4 Trans-4- (4 - ([2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - ({4 - [(4-morpholin -4ylcyclohexyl) amino] -3-nitrophenyl} sulfonyl) -2- (1H-pyrrolo [2,3b] pyridin-5-yloxy) benzamide 5 Trans-4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - [(4 - {[(4methoxycyclohexyl) methyl] amino} -3-nitfophenyl) sulfonyl] -2- (1Hpyrrolo [2,3-b] pyridin-5-yloxy) benzamide 6 Trans-N - ({5-chloro-6 - [(4-hydroxycyclohexyl) methoxy] pyridin-3yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1- en1 -yl] methyl} piperazin-1 -yl) -2- (1 H-indazol-4-yloxy) benzamide 7 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - {[4 - ({[(2S) - 4-cyclopropylmorpholin2-yl] methyl} amino) -3-nitrophenyl] sulfonyl} -2- (1H-pyrrolo [2,3b] pyridin-5-yloxy) benzamide 8 4- (4 - {[2- (4-chlorophenyl) -4 l 4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - [(4 - {[(cis-4- hydroxy-4metiicyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1Hpyrrolo [2,3-b] pyridin-5-yloxy) benzamide 45/55 9 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - [(4 - {[(trans-4- hydroxy-4methylcyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1Hpyrrole [2,3-b] pyridin-5-yloxy) benzamide 10 4- (4 - {[2- (4-dorophenyl) -4,4-dimethylcyclohex-1-en-1yl] methyl} piperazin-1-yl) -N - ({4 - [(4-methylpiperazin- 1-yl) amino] 3-nitrophenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin-5yloxy) benzamide 11 N - [(3-chloro-4 - {[4-fluoro-1- (oxetan-3-if) piperidin-4yl] methoxy} phenyl) sulfonyl] -4- (4 - {[2- (4-chlorophenyl) -4,4 dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) -2- (1Hpyrrolo [2,3-b] pyridin-5-yloxy) benzamide 12 N - ({5-chloro-6 - [(4-fluorotetrahydro-2H-pyran-4yl) methoxy] pyridin-3-yl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4, 4dimethylcyclohex-1-en-1-yl] methyl} piperazin-1-yl) -2- (1Hindazol-4-yloxy) benzamide 13 N - [(5-chloro-6 - {[4-fluoro-1- (oxetan-3-yl) piperidin-4yl] methoxy} pyridin-3-yl) sulfonyl] -4- (4 - {[2- ( 4-chlorophenyl) -4,4dimethylcyclohex-1 -en-1 -i)] methyl} piperazin-1-yl) -2- (1Hpyrrolo [2,3-b] pyridin-5-yloxy) benzamide 14 N - ({5-chloro-6 - [(trans-4-hydroxycyclohexyl) methoxy] pyridin-3yl} sulfonyl) -4- (4 - {[2- (4-chlorophenH) -4,4-dimethylcyclohex-1- en1-yl] methyl} piperazin-1-yl) -2 - [(6-fluoro-1 H-indazol-4yl) oxy] benzamide 15 N - ({3-chloro-4 - [(4-fluorotetrahydro-2H-pyran-4yl) methoxy] phenyl} sulfonyl) -4- (4 - {[2- (4-chlorophenyl) -4,4dimethylcyclohex-1- en-1-yl] methyl} piperazin-1-yl) -2- (1Hpirrolo [2,3-b] pyridin-5-yloxy) benzamide 16 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 yl] methyl} piperazin-1-yl) -N - ({4 - [(4-fluorotetrahydro-2H -pyran-4yl) methoxy] -3-nitrophenyl} sulfonyl) -2- (1H-pyrrolo [2,3-b] pyridin5-yloxy) benzamide 17 Trans-2 - [(6-amino-5-chloropyridin-3-yl) oxy] -4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1 yl) -N - ({4 - [(4-morpholin-4-ylcyclohexyl) amino] -3nitrophenyl} sulfonyl) benzamide Table 1B. Binding affinities for Bcl-2 family proteins and cell efficacy in Bcl-2 or Bcl-x L dependent cell lines for representative compounds. Target AFTR FRET, [nM] Cell Effectiveness, ECso. [nM] FL5.12, 3% FBS 1Q% HS Human Tumor Cell Lines Compound Bcl-2 BcI-Xl Bcí-w Mcl-1 Bcl-2 BcI-xl RS4; 11 (Bcl-2) H146 (BcI-xl) 2 0.04 0.05 7 > 224 20 13 110 75 1 <0.01 48 21 > 440 4 261 12 3,600 3 0.119 151111 12 1060 41 > 5000 4 <0.01 7.2 1.2 70 32 > 5000 5 <0.01 126 1.2 67 8 > 5000 6 <0.01 27> 224 23 3657 7 <0.01 9.4 2.7 926 46/55 8 <0.01 16 405 > 440 4.0 3343 9 <0.01 16 227 > 440 2.5 3757 10 0.02 21> 440 12 97 o » 21 2904 11 <0.01 12 12 2994 12 <0.01 23 2.3 59 17 2747 13 <0.01 9 167 > 440 7 3158 14 <0.01 20 22 3543 15 <0.01 61 35 > 5000 16 <0.01 15 0.7 31 2.1 3931 17 <0.01 12 357 > 440 4.0 65 17 3236 As illustrated in Table 1E 1, Compound 1 has an affinity picomolar to Bcl- 2, but> 4,000-times less affinity for Bcl-x L in competitive binding assays. The significantly higher affinity for Bcl-2 when compared to Bcl-x L suggests a selective binding affinity. In addition, Compound 1 potentially killed FL5.12-Bcl-2 cells (EC50 = 4 nM), but exhibited very weak activity against FL5.12BcI-Xl cells (EC50 = 261 nM), still indicating functional selectivity for Bcl-2. In addition, Compound 1 potentially killed RS4; 11 cells (EC50 = 12 nM), but exhibited very weak activity against H146 cells (EC50 = 3600 nM), still indicating functional selectivity for Bcl-2. As also shown in Table 1B, the additional compounds show a selective binding affinity for Bcl-2 over Bcl-xL and other proteins of the Bcl-2 family. The additional compounds also inhibit Bcl-2-dependent cell lines than Bcl-xL-dependent cell lines. Cellular inhibition and death by Compound 1 exhibit the death marks of apoptotic cells, including the rapid release of cytochrome c, caspase-3 and -7 activation, and the externalization of the phosphatidylserine (PS) membrane. Compound 1 killing cells is dependent on caspase and can be abolished by the caspase inhibitor pan z-VAD-fmk. Cell death is completely inhibited when Bax and Bak, the essential downstream effectors, are genetically ablated. These data indicate that Compound 1 potentially and selectively interrupts Bcl-2 protein-protein interactions and induces a cell death based on mechanism in Bcl-2 dependent cells for survival. Example 2: Pharmacodynamic Response with Compound 1 Selective Bcl-2 Inhibitor It is known within the art that the inhibition of certain members of the Bcl-2 family of 47/55 proteins can induce thrombocytopenia by limiting the dose. Thrombocytopenia limiting the dose that severely limited the therapeutic use of some non-selective Bcl-2 inhibitors for autoimmune indications is considered to be due to Bcl-x L (See Mason, KD, et al., Programmed to nudge cell deat delimits platelet life span, Cell, 2007. 128 (6): p. 117386). Therefore, the effect of sparing Bcl-2 / Bcl-x L Compound 1, on immune cells of peripheral blood and platelets was evaluated in mice (NZBxNZW) FI. The mice were treated four days with Compound 1 (1-100 mg / kg, orally every day) and the number of cells was measured with a Cell Din hematology analyzer. As shown in figure 1B, Compound 1 resulted in a dose-dependent decrease in lymphocytes while maintaining a normal platelet count compared to the control. As shown in figure 1A, Compound 2 also resulted in a decrease in lymphocytes, but caused a significant decrease in platelet counts. These data are selectively consistent with the in vitro profile and highlight the essential role of Bcl-2 in lymphocytes and BcI-Xl in platelet survival respectively. The data also establish lymphopenia as a convenient mechanism marker for Compound 1. The effects of the selective Bcl-2 / Bcl-x L compounds spared in peripheral blood immune cells were also evaluated in the C57BL / 6 mice. The mice were treated four days with individual compounds (100 mg / kg, orally every day) and the number of cells was measured with a Cell Din hematological analyzer. As shown in Table 2, treatment with all compounds resulted in a decrease in lymphocytes after a single oral dose of 100 mg / kg and after 4 oral doses of 100 mg / kg. Table 2. Number of lymphocytes and degree of reduction in C57BL / 6 mice treated with 1 and 4 does of a selective Bcl-2 inhibitor (100 mg / kg) - | Compound Lymphocytes (x 10 e ) % Reduction vs. Vehicle lymphocytes (x 10 a ) % Reduction vs. Vehicle 1 1.38 83 1.32 83 3 2.3 63 1.34 79 4 2.62 57 1.72 73 5 0.98 84 1.02 84 6 1.91 69 0.98 85 7 0.89 85 0.95 85 8 0.92 85 1.02 84 9 0.75 88 0.88 86 10 2.25 65 2.48 71 11 1.76 73 1.71 80 48/55 12 1.44 78 2.21 74 13 2.32 64 2.61 70 14 3.02 54 2.33 73 15 1.85 72 1.65 81 16 1.77 73 1.99 77 17 1.84 77 1.30 83 Example 3: Pharmacodynamic response with Compound 3 Selective Bcl-2 inhibitor An experiment was carried out to evaluate the effect of an additional selective inhibitor compound of Bcl-2, 4- (4 - {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) -2 (1H-indol-5-yloxy) -N - ({3-nitro-4 - [(tetrahydro-2H-pyran-4-ylmethyl) amino] phenyl} sulfonyl) benzamide, hereinafter “ Compound 3, ”in immune cells and platelets, as assessed in mice (NZBxNZW) FI. Mice were treated for four days with Compound 3 (doses of 30 mg / kg and 100 mg / kg, administered by intraperitoneal injection every day) and the number of cells was measured with a Cell Din hematological analyzer. Compound exposure was calculated 24 hours after the last dose. The results of this experiment are illustrated in figures 2 and 3. As shown in figure 2, Compound 3 resulted in a dose-dependent decrease in the lymphocytes while maintaining normal platelet count compared to the phosphate vehicle control. Specifically, the doses of 30 mg / kg and 100 mg / kg of Compound 3 resulted in a reduction of lymphocytes by 47% and 66%, respectively, without significantly affecting platelet counts. In addition, Figure 3 illustrates a statistically significant decrease in CD4 + T cells, CD8 + T cells, and CD19 + B cells, for doses of Compound 3 comprising 100 mg / kg and 300 mg / kg. As such, these pharmacodynamic studies illustrate the ability of Compound 3, a selective Bcl-2 inhibitor to effectively reduce lymphocytes, without the side effects associated with non-selective Bcl-2 inhibitors. Example 4: Treatment with Bcl-2 Selective Inhibitors in the Spontaneous Murine Model of Lupus To demonstrate that Compound 1 and Compound 17 are viable therapies for human SLE or lupus nephritis, the experiments were conducted in a spontaneous murine (NZBxNZW) Fi model of lupus. (See Liu, K. and C. Mohan, What do mouse models teach us about human SLE Clin Immunol, 2006. 119 (2): p. 123-30). This model has been well characterized in relation to the duck-physiological changes analogous to those of the human SLE. They exhibit a tendency towards female in the prevalence of the disease and high serum titers of anti-dsDNA IgG antibodies, which are major brands of human SLE, with monitoring of renal IgG deposition. Renal histopathological changes include severe glomeronephritis, peripheral and mesangial proliferative changes, membrane thickening 49/55 capillary, tubular atrophy and infiltration of lymphocytes and monocytes / macrophages as in most human SLE patients. These changes result in disruption of kidney function as evidenced by severe proteinuria (PU) greater than 300mg / dL as measured by albumin rods, followed by mortality as measured by survival. Two of the clinical parameters for lupus nephritis, MMF and cyclophosphamide, have been shown to decrease the autoantibody titer, improve renal pathology, delay the onset of severe proteinuria and prolong survival in these animals. (See Gelfand, MC and AD Steinberg, Terapeutic studies in NZB-W mice. II. The relative efficacy of azathioprine, cyclophosphamide and methylprednisolone. Artritis Rheum, 1972. 15 (3): p. 247-52; and Ramos, MA, et al., Modulation of autoantibodi production bi micophenolate mofetil: effects on te development of SLE in (NZB x NZW) F1 mice. Nefrol Dial Transplant, 2003.18 (5): p. 878-83) Females (NZB x NZW) F1 were purchased from Te Jackson Laboratory (Bar Harbor, Maine, USA) and kept in a conventional animal housing facility throughout the experiment. Anti-ds DNA was measured in the 25 week old (NZBxNZW) FI mice and the animals were distributed into various treatment groups (N = 1418 / group) at 26 weeks of age and administered daily with oral doses of Compound 1 or Compound 17 ranging from 1 to 100 mg / kg, or mycophenolate mofetil (MMF) in a dose of 100 mg / kg. Proteinuria (PU) and survival were monitored weekly, followed by measurements twice a week of lymphocytes and platelet counts, and the production of anti-DNA DNA. PK parameters were also measured throughout the study. The impact on IgG deposition and renal pathology was attributed at the conclusion of the study. Severe PU was defined by two consecutive weekly measurements of PU 300 mg / dl using Albustix (VWR) test strips. When the mice became dying, they were sacrificed according to the protocols of the Institutional Animal Use and Care Committee. PU and survival data were presented as KaplanMeier survival curves using Grafpad Prism software and group differences were considered significant at p <0.05. Histological scores were analyzed using ANOVA analysis. The anti-ds DNA were analyzed using a one-way ANOVA analysis and the Tukei post test. As illustrated in figure 4A, treatment with Compound 1 resulted in delayed onset of severe PU in a dose-dependent manner, reaching significance at 10, 30 and 100 mg / kg. In addition, treatment with Compound 1 to 3 , 10, 30, and 100 mg / kg significantly prolonged survival as illustrated in Figure 4B and Table 3A. These data also correlated with a sustained and dose-dependent reduction of lymphocytes in peripheral blood, with 30 and 100 mg / kg of Compound 1 dosage both resulting in 70% lymphopenia. The efficacy at both disease endpoints at the dose of 30 mg / kg of Compound 1 was comparable to MMF treatment at 100 mg / kg. The exposure on this model in 50/55 dosage of 30 mg / kg was 40 pgh / ml. Similar efficacy results were also obtained for Compound 17 as shown in Figure 4 C, D and Table 3B. Table 3A. Compound 1 PU and Survival Effectiveness at Week 47 Treatment Groups % of mice with PU <300 mg / dL % of Survival Compound 1 Control Vehicle 7 19 1 mg / kg 51 60 3mg / kg 50 70 * 10mg / kg 66 * 83 * 30mg / kg 94 * 100 * 100mg / kg 100 * 100 * MMF Control Vehicle 13 13 100mg / kg 94 * 100 * * p <0.05 Table 3B. Compound 17 PU and the Effectiveness of Survival at Week 39 Treatment Groups % of mice with PU <300 mg / dL % of Survival 17 Control Vehicle 71 93 1 mg / kg 58 67 3mg / kg 67 92 10mg / kg 71 93 30mg / kg 86 100 100mg / kg 100 * 100 * p <0.05 Example 5: Effectiveness of Bcl-2 Selective Inhibitors on the anti-ds DNA titer in the Spontaneous Mouse Model Compound 1 is believed to trigger an apoptosis of the lymphocytes that are responsible for the production of antibodies, which play a role in the progression of systemic lupus erythematosus, as well as Sjogren's syndrome. As such, it was hypothesized that treatment with Compound 1 would decrease the anti-DNA titer titer. Specifically, the levels of anti-dsDNA antibodies were measured by ELISA and the arbitrary unit-of-activity concentrations assigned per ml relative to the standard plasma tank derived from the NZB / W 9-10 month old proteinuric mice. The ELISA assay was performed by coating the plates with poly-L-lysine followed by calf-thymus DNA. The diluted mouse plasma was incubated and developed using conjugated anti-IgG HRP antibodies and the main DO from duplicate sources was compared to the standard titrated curve of the pooled titrated anti-dsDNA plasma. The standard undiluted plasma tank was arbitrarily assigned a value of 1000 anti-dsDNA Units / ml. Linear regression analysis was then used to calculate the relative units of a given sample multiplied by the given dilution factor. The results of this experiment are shown in Figure 5. In control animals, IgG levels of anti-DNA DNA increased from an average of <100 unit / mL at baseline (25 weeks) to an average of ~ 600 unit / mL in week 32, with another increase to -1800 unit / mL at week 40. the apparent increase in anti-ds DNA titer occurred with a concomitant increase in disease severity and incidence as measured by PU and 51/55 survival. There was no appreciable reduction in the anti-ds DNA titer in the groups treated with 1, 3 and 10 mg / kg of Compound 1. However, at week 40, treatment with Compound 1 at 30 and 100 mg / kg significantly inhibited the titer anti-ds DNA compared to the control vehicle, comparable to the effect observed with MMF. Example 6: Infiltration of Renal Tissue with Selective Bcl-2 Inhibitors An experiment was carried out to determine the extent to which selective Bcl-2 inhibitors infiltrated kidney tissue. Specifically, a histological assignment of the penetration of selective Bcl-2 inhibitors into the renal tissue of the spontaneous murine model for lupus was performed. The spontaneous murine model of lupus, as described in Example 3, was used for the histological assignment described here. The kidneys were divided and then fixed in 10% neutral buffered formalin or cryopreserved (closed freeze). For H&E staining, 5-pm sections of tissues embedded in paraffin were semiquantitatively scored (0-4) by a pathologist for glomerulonephritis and tubular changes (dilation and cylinders). For IgG immunohistochemistry, 5-pm cryosections were fixed with acetone, washed and blocked with 10% normal goat serum. The sections were then incubated with FITO conjugated goat anti-mouse IgG (Cappel / ICN Pharmaceuticals) or HRP-goat IgG negative control (Jackson ImmunoResearch Laboratories) and coverslips using 4 ', 6-diamidino-2-phenylindole (Vector Laboratories) . Sections were assessed for severity of IgG deposition using a semi-quantitative scoring system (from 0-4). To identify B and T cells, immunohistochemistry for CD45R (B cells) and CD3 (T cells) was completed in the paraffin sections. Figures 6 and 7 illustrate the results of the histological assignment, and figure 8 illustrates the effectiveness of treatment with selective Bcl-2 inhibitors in the deposition of IgG, B cells, and T cells in the kidneys. As illustrated in figure 6, mice with spontaneous lupus nephritis dosed with a phosphate vehicle typically had extensive renal infiltrations, as evidenced by tubular cylinders, dilated tubules, glomeruloclerosis and infiltrated cells. The renal tissue infiltrated in mice with spontaneous lupus nephritis dosed with 30 or 100 mg / kg of Compound 1 was small, different and less frequent. In addition, figure 7 includes a bar graph illustrating the difference in histological scores for untreated renal tissue, tissue treated with Compound 1 at doses of 30 mg / kg and 100 mg / kg, and treated tissue with MMF at a dose of 100 mg / kg. As noted in Figure 7, tissue treated with Compound 1 at doses of 30 mg / kg and 100 mg / kg showed a statistically significant improvement (decrease in severity) in histological scores, while belonging to glomerulonephritis, tubular changes and perivascular infiltrations. In addition, figure 8 illustrates a decrease in IgG deposition in renal tissue treated with Compound 1 at a dose of 30 mg / kg, as well as decreasing the number of B cells and 52/55 T cells in renal tissue treated with Compound 1 at a dose of 100 mg / kg. Therefore, the selective inhibitor of Bcl-2, Compound 1, illustrated in the deposition of IgG, in the infiltration and expansion of B cells, and T cells in renal tissue, and also resulted in a statistically significant improvement in histological scores, when compared to treatment with a phosphate vehicle. Example 7: Treatment with a Selective Bcl-2 Inhibitor in an Interferon-a Accelerated Lupus Model Due to the fact that studies on the spontaneous (NZBxNZW) FI model require 6-8 months to complete due to the slow development of the manifestations of the disease, additional tests for selective Bcl-2 inhibitors have been carried out on alternative models. To provide a faster measurement, an accelerated lupus model IFNa was established and used to assign the therapeutic potential of compound 1. An increased IFNa serum level and an increased “IFNa responsive gene signature” concomitance was reported in a subset of patients with SLE (See Kwok, SK, et al., Disfunctional interferon-alfa production by peripheral plasmacitoid dendritic cells upon Toll-like receptor-9 stimulation in patients with sistemic lupus eritematosus. Artritis Res Ter, 2008. 10 (2): p. R29; and Rong, Z., et al., Effect of Interferon-alfa in sistemic lupus eritematosus (SLE) serum on te differentiation and maturation of dendritic cells derived from CD34 + hematopoietic precursor cells. Journal of Nanjing Medicai Universiti, 2009. 23 (6): pp. 380-385) A drug-induced SLE type disease has also been reported in patients with HCV given IFNa therapy. (See Wilson, LE, et al., Autoimmune disease complicating antiviral terapy for hepatitis C virus infection. Semin Artritis Rheum, 2002. 32 (3): p. 163-73) These observations highlight an important role for IFNa signaling in pathogenesis SLE. To recapitulate the IFNa effect in rodents, inventors and others in the art (See Bardwell, PD, et al., Te Bcl-2 family antagonist ABT-737 significantly inhibits multiple animal models of autoimmunity. J Immunol, 2009. 182 (12) : p. 7482-9; and Matian, A., et al., IFN-alfa induces early letal lupus in preautoimmune (New Zealand Black x New Zealand White) F1 but not in BALB / c mice. J Immunol, 2005. 174 (5): p. 2499-506) established an IFNa-induced lupus (NZBxNZW) Fi model mediated by adenovirus, which characterizes a severe and rapid disease with several characteristics similar to mice with spontaneous lupus (NZBxNZW) Fi, including death due to the severity of glomerulonephritis. However, there are also differences between these two: (1) a supraphysiological production of IFNq in the blood is required for severe lupus nephritis (see Matian, A., et al., IFN-alfa induces earli letal lupus in preautoimmune (New Zealand Black x New Zealand White) F1 but not in BALB / c mice, J Immunol, 2005. 174 (5): p. 2499-506); (2) a sustained,> 50% peripheral blood lymphopenia is observed within 2 weeks of IFNa treatment; (3) the increase in disease is not associated with a robust increase in humoral autoimmunity such as the titer 53/55 anti-ds DNA as seen in spontaneous (NZBxNZW) Fi mice. The data collected in the experiment were consistent with recent findings in mice with B6.Sle123 treated IFN-α adenovirus (See Fairhurst, AM, et al., Sistemic IFN-alfa drives kidnei nefritis in B6.Sle123 mice. Eur J Immunol., 2008. 38 (7): p. 1948-60) and supported the idea that the majority of the effects of IFNa treatment on the pathogenesis of the disease in this model is to lead to organ disease in the end, possibly through the activation of various types immune cells and pro-inflammatory cytokine production. Compound 1, MMF and BAFFR3-lg (a substitute for Belimumab) were evaluated in the IFNa-induced mouse model (NZBxNZW) Fi. BAFFR3-lg specifically blocks the binding of BAFF / BLiS to its cognate receptor BAFFR3, resulting in a systemic reduction in the number of B cells in the lymphoid organs (See Kaiagaki, N., et al., BAFF / BLiS receptor 3 binds te B cell survival factor BAFF ligand trough a discrete surface loop and promotes processing of NF-kappaB2 Immuniti, 2002. 17 (4): p. 515-24). Treatment was started in a late prophylactic mode (7 days after IFNa adenovirus). Specifically, the F1 mice (NZB x NZW) (Te Jackson Laboratori), aged 13-15 weeks old, were injected with a single intravenous dose of IFN-α adenovirus (Abbott) at a concentration of 5 x 10 9 viral particles / mice. The treatment groups consisted of the administration of Compound 1 in doses ranging from 1-100mg / kg / day, given orally; mycophenolate mofetil (MMF) in a dose of 100 mg / kg / day, given orally; and BAFFR3-lg (BAFF / BLiS blocker) at a dose of 15 mg / kg, 3x / week, by intraperitoneal injection. All treatment groups were given treatment 7 days after injection of the adenovirus. Following adenovirus injection, mice were monitored weekly for proteinuria (PU) using Albustix test strips (VWR). Severe PU was defined by consecutive weekly PU doctors è 300 mg / dl. When the mice became dying, they were sacrificed according to the protocols of the Institutional Animal Use and Care Committee. The results of these experiments are illustrated in figure 9, and in Table 4 below. Consistent with the discovery in spontaneous lupus mice, treatment with both 30 and 100 mg / kg of Compound 1 significantly delayed the onset of severe PU and prolonged survival. Efficacy was compared to, if not better than, MMF and BAFFR3-lg, and correlated with sustained lymphopenia. The target effectiveness exposure was approximately 40 pg h / ml. Table 4 PU and Survival Effectiveness in Compound 1, MMF and BAFF-R3-lg Treated Animals Treatment Groups % of mice with PU <300 mg / dL % of Survival Compound 1 Control Vehicle 0 16 30 mg / kg 77 * 82 * MMF Control Vehicle 0 42 54/55 100 mg / kg 40 * 72 * BAFF-R3-lg PBS 10 60 15 mg / kg 70 * 95 * *: ρ <0.05 Example 8: Effect of Selective Bcl-2 Inhibitors on the title enti-ds DNA in the Interferon-a Accelerated Lupus Model An additional experiment was carried out to determine the effects of treatment with the selective inhibitor of Bcl-2 on the anti-DS DNA titer in the accelerated lupus model of Interferon-a. Anti-dsDNA antibody levels were measured by ELISA and arbitrary activity unit concentrations assigned per ml relative to a standard plasma tank derived from the 9-10 month old NZB / W proteinuric mouse. The ELISA assay was performed by coating the plates with poly-L-lysine followed by calf-timus DNA. The diluted mouse plasma was incubated and developed using conjugated anti-IgG HRP antibodies and the main DO of the duplicated cells was compared to a standard titrated curve of the pooled high-titrated anti-dsDNA plasma. The standard undiluted plasma tank was arbitrarily assigned a value of 1000 anti-dsDNA Units / ml. Linear regression analysis was then used to calculate the relative units of a given sample multiplied by a given dilution factor. The results of this experiment are illustrated in figure 10. In contrast to the findings from the spontaneous F1 mouse (NZBxNZW) model, shown in Figure 5, Compound 1 did not significantly inhibit the anti-dsDNA titer in the interferon-induced model, as shown in Figure 10. There were two turns, but the non-statistically significant induction of anti-DNA titer between day 42 and 55, which was not dose dependent. It should be noted that at 30 mg / kg, the Compound 1 treatment of these animals maintained a lymphopenia of> 70% in the treatment through the blood and protected the animals from lupus nephritis. Example 9: Infiltration of Bcl-2 Selective Inhibitors into Salivary Glands It is recognized within the art that Sjogren's Syndrome is a chronic disease state that affects the body's moisture-producing glands, including the salivary glands of the mouth. As such, it has been theorized that the penetration of selective Bcl-2 inhibitors into the salivary glands of patients with Sjogren's Syndrome could provide an effective treatment to decrease lymphocytes associated with Sjogren's Syndrome, without the dose-limiting thrombocytopenia associated with non-selective Bcl-2 inhibitor. A histological assessment of the penetration of selective Bcl-2 inhibitors into the salivary glands of the spontaneous murine model for lupus was performed to test this theory. The spontaneous murine model of lupus, as described in Example 3, was used for a histological assessment described above. Specifically, the sublingual and submandibular salivary glands were fixed in 10% neutral buffered formalin and embedded paraffin. Five pm sections 55/55 were stained with H&E and semiquantitatively scored (0-4) by an experienced pathologist for infiltration of inflammatory cells. The scoring categories of the salivary gland infiltrate: (1) 3 or slightly smaller periductal foci, (2) 3 or more medium-sized foci, (3) foci of severe extension, and (4) coalescent to diffuse infiltrates. The nisioiogical evaluation of the sections u or the suornanai tissue uses the illusion in Figure i i. As illustrated in figure 11, the mouse with spontaneous lupus nephritis dosed by a phosphate vehicle was typically extended to coalescent periductular infiltrates. The infiltrates in the salivary glands in mice with spontaneous lupus nephritis dosed with 30 or 100 mg / kg of Compound 1 were smaller, different and less frequent. In addition, figure 12 includes a bar graph illustrating the difference in histological scores for submandibular tissue that was not treated, tissue treated with Compound 1 at a dose of 10 mg / kg, and tissue treated with Compound 1 in doses of 30 mg / kg and 100 mg / kg. As noted in figure 11, tissue treated with Compound 1 at doses of 30 mg / kg and 100 mg / kg, the histological score shown showed a statistically significant improvement in histological points, as evidenced by the fact that the histological score to 30 mg / kg and 100 mg / kg of treatment was less severe. Therefore, Sjogren's Syndrome is an inflammatory disease that affects the body's moisture-producing glands, including the salivary gland. A histological evaluation of the salivary glands in spontaneous merino models was performed to determine whether treatment with the selective inhibitor Bcl-2 could decrease the inflammatory processes in the salivary gland, and finally provide a treatment option for Sjogren's Syndrome. Histological evaluation showed that doses of 30 mg / kg and 100 mg / kg of Compound 1 resulted in decreased inflammation in the salivary glands, as evidenced by the improvement in the histological score of 3-4 in mice that were not treated for a score of 1-2 in mice that have been treated. Thus, it was determined that selective Bcl-2 inhibitors can provide an effective treatment for patients with Sjogren's Syndrome.
权利要求:
Claims (11) [1] 1. Pharmaceutical composition CHARACTERIZED by the fact that it comprises a selective inhibitor compound of Bcl-2 for the treatment of systemic lupus erythematosus, lupus nephritis or Sjogren's syndrome in a patient, in which the deleterious inhibitor compound of Bcl2 is 4- (4- {[2- (4-chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-i] methyl Piperazi n-1 -i I) -N - ({3-nitro-4tetrahydro-2H-pyran-4 -ylmethyl) amino] phenyl} sulfonyl) -2- (1 H-pyrrolo [2,3-b] pyridine-5-yloxy) benzamide or a pharmaceutically acceptable salt thereof or 4- (4 - {[2- (4 -chlorophenyl) -4,4dimethylcyclohex-1 -en-1 -i I] methyl Piperazi n-1 -yl) -N- [4 - {[(trans-4-hydroxy-4methylcyclohexyl) methyl] amino} -3-nitrophenyl ) sulfonyl] -2- (1 H-pyrrolo [2,3-b] pyridine-5-yloxy) benzamide or a pharmaceutically acceptable salt thereof. [2] 2. Pharmaceutical composition according to claim 1, CHARACTERIZED by the fact that the composition comprises a dose of the selective inhibitor compound of Bcl2 ranging from 0.001 mg / kg to 1000 mg / kg. [3] 3. Pharmaceutical composition according to claim 1, CHARACTERIZED by the fact that the composition comprises a dose of the selective inhibitor compound of Bcl2 ranging from 0.01 mg / kg to 500 mg / kg. [4] 4. Pharmaceutical composition according to claim 1, CHARACTERIZED by the fact that the composition comprises a dose of the selective inhibitor compound of Bcl2 ranging from 0.1 mg / kg to 300 mg / kg. [5] 5. Pharmaceutical composition according to claim 1, CHARACTERIZED by the fact that it traditionally comprises a binding protein. [6] Pharmaceutical composition according to any one of claims 1 to 5, CHARACTERIZED by the fact that the selective inhibitor compound of Bcl-2 is 4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1 -en-1-yl] methyl} piperazin-1-yl) -N - ({3-nitro-4-tetrahydro-2H-pyran-4ylmethyl) amino] phenyl} sulfonyl) -2- (1 H-pyrrole [2 , 3-b] pyridine-5-yloxy) benzamide or a pharmaceutically acceptable salt thereof. [7] 7. Use of the pharmaceutical composition, as defined in claim 6, CHARACTERIZED by the fact that it is in the preparation of a medication to treat systemic lupus erythematosus or lupus nephritis. [8] 8. Use, according to claim 7, CHARACTERIZED by the fact of systemic lupus erythematosus. [9] Pharmaceutical composition according to any one of claims 1 to 5, CHARACTERIZED by the fact that the selective inhibitor compound of Bcl-2 is 4- (4 - {[2- (4chlorophenyl) -4,4-dimethylcyclohex-1 -en-1 -i I] methylpiperazi n-1 -yl) -N- [4 - {[(trans-4-hydroxy-4methylcyclohexyl) methyl] amino} -3-nitrophenyl) sulfonyl] -2- (1 H -pyrrole [2,3-b] pyridine-5-yloxy) benzamide or a pharmaceutically acceptable salt thereof. [10] 10. Use of the pharmaceutical composition, as defined in claim 9, 2/2 CHARACTERIZED by the fact that it is in the preparation of a medication to treat systemic lupus erythematosus or lupus nephritis. [11] 11. Use, according to claim 10, CHARACTERIZED by the fact of systemic lupus erythematosus.
类似技术:
公开号 | 公开日 | 专利标题 US20200222413A1|2020-07-16|Methods of treatment using selective bcl-2 inhibitors BR112020020246A2|2021-01-12|AXL KINASE INHIBITORS AND USE OF THE SAME JP2019534304A|2019-11-28|Compounds for the treatment of inflammatory diseases and pharmaceutical compositions thereof BR112019027967A2|2020-07-14|isolated crystalline form b, pharmaceutical composition, method for treating a disorder, method of a compound or composition, and, process for producing crystalline form b. WO2021194991A1|2021-09-30|Use of losmapimod for treatment of covid-19 US20190328729A1|2019-10-31|Uses of Bcl-2 Antagonists for Treating Cancer and Diagnostics Related Thereto BR112021007643A2|2021-07-27|heterocyclic kinase inhibitors and their uses JP2021500413A|2021-01-07|IL-8 inhibitor for use in the treatment of some sarcomas WO2020087565A1|2020-05-07|Indazole kinase inhibitor and use thereof
同族专利:
公开号 | 公开日 US20150150870A1|2015-06-04| NZ610746A|2015-06-26| US20120129853A1|2012-05-24| BR112013012854A8|2018-01-16| CA2820618C|2020-04-21| GT201300136A|2014-08-05| ES2603129T3|2017-02-23| TW201630885A|2016-09-01| AU2011332000B2|2016-07-07| EP2642999B1|2016-09-28| CY1118442T1|2017-06-28| EP3028702B1|2018-09-12| BR112013012854A2|2016-08-23| WO2012071374A1|2012-05-31| PT2642999T|2017-01-05| CR20130268A|2013-10-17| JP2013543896A|2013-12-09| AU2011332000A1|2013-06-06| MY165884A|2018-05-18| HUE031267T2|2017-06-28| AR083946A1|2013-04-10| US20170232000A9|2017-08-17| RU2013128544A|2014-12-27| RU2593231C2|2016-08-10| US20200222413A1|2020-07-16| KR20180030257A|2018-03-21| IL226490D0|2013-07-31| IL251845A|2019-06-30| HRP20161762T1|2017-02-24| KR101841084B1|2018-03-23| SG10201509631SA|2015-12-30| UA119150C2|2019-05-10| NZ708508A|2016-06-24| US9872861B2|2018-01-23| CN103402521B|2016-01-20| JP6215368B2|2017-10-18| IL226490A|2017-05-29| US20160235760A1|2016-08-18| EP2642999A1|2013-10-02| AU2016204396A1|2016-07-14| US20180369250A1|2018-12-27| JP2016117744A|2016-06-30| TW201302705A|2013-01-16| TWI620736B|2018-04-11| DOP2017000056A|2017-03-31| MX2013005849A|2013-07-05| KR20190109590A|2019-09-25| SI2642999T1|2017-01-31| AU2016204396B2|2017-10-26| CN103402521A|2013-11-20| EP3028702A1|2016-06-08| ES2701603T3|2019-02-25| ECSP13012710A|2013-08-30| US9345702B2|2016-05-24| SG190399A1|2013-06-28| IL251845D0|2017-06-29| CN105664164A|2016-06-15| UA113157C2|2016-12-26| UY33746A|2012-06-29| PL2642999T3|2017-04-28| CN105664164B|2020-05-05| LT2642999T|2017-01-25| JP5876067B2|2016-03-02| TWI535701B|2016-06-01| PH12016500939A1|2017-06-14| ZA201303589B|2014-07-30| PH12016500939B1|2017-06-14| MX346201B|2017-03-09| DK2642999T3|2017-01-09| KR20140004659A|2014-01-13| CA2820618A1|2012-05-31| CL2013001458A1|2013-10-04| RS55545B1|2017-05-31|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 US2128208A|1937-02-27|1938-08-23|Du Pont|Alkoxy derivatives of methacrylic acid esters and method of producing them| MY115155A|1993-09-09|2003-04-30|Upjohn Co|Substituted oxazine and thiazine oxazolidinone antimicrobials.| FR2736550B1|1995-07-14|1998-07-24|Sandoz Sa|PHARMACEUTICAL COMPOSITION IN THE FORM OF A SOLID DISPERSION COMPRISING A MACROLIDE AND A VEHICLE| JPH11512429A|1995-09-15|1999-10-26|ファルマシア・アンド・アップジョン・カンパニー|Aminoaryloxazolidinone N-oxide| PL188192B1|1996-02-09|2004-12-31|Abbott Lab Bermuda Ltd|Human antibodies capable to bond human tnf alpha| US20030220234A1|1998-11-02|2003-11-27|Selvaraj Naicker|Deuterated cyclosporine analogs and their use as immunodulating agents| AT416772T|1998-07-06|2008-12-15|Squibb Bristol Myers Co|BIPHENYLSULFONAMIDE AS A DOUBLE-ACTIVE RECEPTOR ANTAGONIST OF ANGIOTENSIN AND ENDOTHELIN| US6492389B1|1998-07-21|2002-12-10|Thomas Jefferson University|Small molecule inhibitors of BCL-2 proteins| DE19913692A1|1999-03-25|2000-09-28|Basf Ag|Mechanically stable pharmaceutical dosage forms containing liquid or semi-solid surface-active substances| DE19929361A1|1999-06-25|2001-01-04|Basf Ag|Mechanically stable pharmaceutical dosage forms containing liquid or semi-solid surface-active substances| US20030236236A1|1999-06-30|2003-12-25|Feng-Jing Chen|Pharmaceutical compositions and dosage forms for administration of hydrophobic drugs| GB9918037D0|1999-07-30|1999-09-29|Biochemie Gmbh|Organic compounds| MXPA02006474A|1999-12-28|2002-11-29|Eisai Co Ltd|Heterocyclic compounds having sulfonamide groups.| US20020055631A1|2000-09-20|2002-05-09|Augeri David J.|N-acylsulfonamide apoptosis promoters| UY26942A1|2000-09-20|2002-04-26|Abbott Lab|PROMOTING N-ACILSULFONAMIDS OF APOPTOSIS| US6720338B2|2000-09-20|2004-04-13|Abbott Laboratories|N-acylsulfonamide apoptosis promoters| US6995162B2|2001-01-12|2006-02-07|Amgen Inc.|Substituted alkylamine derivatives and methods of use| US20090247613A1|2002-02-20|2009-10-01|Sirna Therapeutics, Inc.|RNA INTERFERENCE MEDIATED INHIBITION OF B-CELL CLL/LYMPHOMA-2 GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID | CZ20033296A3|2001-06-06|2004-04-14|Eli Lilly And Company|Benzoyl sulfonamides and sulfonyl benzamidines functioning as antineoplastic compounds| GB0123400D0|2001-09-28|2001-11-21|Novartis Ag|Organic compounds| RU2318518C2|2002-02-26|2008-03-10|Астразенека Аб|Pharmaceutical composition possessing anti-proliferative effect , method for its preparing and methods for its using| ES2342660T3|2002-02-26|2010-07-12|Astrazeneca Ab|NEW CRYSTAL FORMS OF THE ANTICHANCERIGENO COMPOUND ZD1839.| FR2836914B1|2002-03-11|2008-03-14|Aventis Pharma Sa|SUBSTITUTED INDAZOLES, COMPOSITIONS CONTAINING SAME, METHOD OF MANUFACTURE AND USE| TWI262077B|2002-04-29|2006-09-21|Merck & Co Inc|Tetrahydropyranyl cyclopentyl tetrahydropyridopyridine modulators of chemokine receptor activity| JP4336678B2|2003-09-04|2009-09-30|株式会社日立超エル・エス・アイ・システムズ|Semiconductor device| US8614318B2|2003-11-13|2013-12-24|Abbvie Inc.|Apoptosis promoters| US7767684B2|2003-11-13|2010-08-03|Abbott Laboratories|Apoptosis promoters| WO2005049593A2|2003-11-13|2005-06-02|Abbott Laboratories|N-acylsulfonamide apoptosis promoters| US7642260B2|2003-11-13|2010-01-05|Abbott Laboratories, Inc.|Apoptosis promoters| US7973161B2|2003-11-13|2011-07-05|Abbott Laboratories|Apoptosis promoters| US7790190B2|2004-03-20|2010-09-07|Yasoo Health, Inc.|Aqueous emulsions of lipophile solubilized with vitamin E TPGS and linoleic acid| WO2005099353A2|2004-04-19|2005-10-27|Symed Labs Limited|A novel process for the preparation of linezolid and related compounds| EP1768967B1|2004-07-20|2009-04-22|Symed Labs Limited|Novel intermediates for linezolid and related compounds| ES2306216T3|2004-08-27|2008-11-01|Bayer Pharmaceuticals Corporation|PHARMACEUTICAL COMPOSITIONS IN THE FORM OF SOLID DISPERSIONS FOR CANCER TREATMENT.| WO2006039164A2|2004-09-29|2006-04-13|Amr Technology, Inc.|Novel cyclosporin analogues and their pharmaceutical uses| EP2757099B1|2005-05-12|2017-08-23|AbbVie Bahamas Limited|Apoptosis promoters| US8624027B2|2005-05-12|2014-01-07|Abbvie Inc.|Combination therapy for treating cancer and diagnostic assays for use therein| WO2006124863A2|2005-05-16|2006-11-23|Irm Llc|Pyrrolopyridine derivatives as protein kinase inhibitors| TW200716636A|2005-05-31|2007-05-01|Speedel Experimenta Ag|Heterocyclic spiro-compounds| RU2565071C2|2005-06-22|2015-10-20|Плекссикон, Инк|PYRROLO[2,3-b]PYRIDINE DERIVATIVES AS PROTEIN KINASE INHIBITORS| US7514068B2|2005-09-14|2009-04-07|Concert Pharmaceuticals Inc.|Biphenyl-pyrazolecarboxamide compounds| CA2631760A1|2005-12-02|2007-06-07|Robert A. Brodsky|Use of high-dose oxazaphosphorine drugs for treating immune disorders| EP1880715A1|2006-07-19|2008-01-23|Abbott GmbH & Co. KG|Pharmaceutically acceptable solubilizing composition and pharmaceutical dosage form containing same| WO2008030836A2|2006-09-05|2008-03-13|Abbott Laboratories|Bcl inhibitors treating platelet excess| US8796267B2|2006-10-23|2014-08-05|Concert Pharmaceuticals, Inc.|Oxazolidinone derivatives and methods of use| WO2008064116A2|2006-11-16|2008-05-29|Abbott Laboratories|Method of preventing or treating organ, hematopoietic stem cell or bone marrow transplant rejection| AU2008214774A1|2007-02-15|2008-08-21|F. Hoffmann-La Roche Ag|2-aminooxazolines as TAAR1 ligands| WO2008124878A1|2007-04-13|2008-10-23|Cryptopharma Pty Ltd|Non-steroidal compounds| KR20100012031A|2007-04-19|2010-02-04|콘서트 파마슈티컬즈, 인크.|Deuterated morpholinyl compounds| US7531685B2|2007-06-01|2009-05-12|Protia, Llc|Deuterium-enriched oxybutynin| WO2009035598A1|2007-09-10|2009-03-19|Concert Pharmaceuticals, Inc.|Deuterated pirfenidone| US20090118238A1|2007-09-17|2009-05-07|Protia, Llc|Deuterium-enriched alendronate| US20090088416A1|2007-09-26|2009-04-02|Protia, Llc|Deuterium-enriched lapaquistat| US20090082471A1|2007-09-26|2009-03-26|Protia, Llc|Deuterium-enriched fingolimod| CN101842362A|2007-09-27|2010-09-22|沃尔特及伊莱萨霍尔医学研究院|Benzothiazole compounds| WO2009045564A1|2007-10-01|2009-04-09|Arkema Inc.|Blends of biodegradable polymers and acrylic copolymers| CA2701172A1|2007-10-01|2009-04-09|The Johns Hopkins University|Methods of treating neurological autoimmune disorders with cyclophosphamide| EP2209774A1|2007-10-02|2010-07-28|Concert Pharmaceuticals Inc.|Pyrimidinedione derivatives| US20090105338A1|2007-10-18|2009-04-23|Protia, Llc|Deuterium-enriched gabexate mesylate| WO2009051782A1|2007-10-18|2009-04-23|Concert Pharmaceuticals Inc.|Deuterated etravirine| JP2011502963A|2007-10-19|2011-01-27|バーンハムインスティテュートフォーメディカルリサーチ|Naphthalene inhibitors that are anti-apoptotic proteins| CA2703591C|2007-10-26|2013-05-07|Concert Pharmaceuticals, Inc.|Deuterated darunavir| RU2472509C2|2007-11-16|2013-01-20|Эбботт Лэборетриз|Method of treating arthritis| CA2708223A1|2007-12-06|2009-06-11|Andrew Krivoshik|Oral compositions of abt-263 for treating cancer| CN101481363B|2008-01-10|2011-05-04|江苏豪森药业股份有限公司|Method for preparing repaglinide| US8187836B2|2008-01-15|2012-05-29|Abbott Laboratories|Mammalian expression vectors and uses thereof| US20090258914A1|2008-04-15|2009-10-15|Burnham Institute For Medical Research|Inhibitor of Anti-Apoptotic Proteins| WO2010041051A1|2008-10-07|2010-04-15|Astrazeneca Uk Limited|Pharmaceutical formulation 514| US8557983B2|2008-12-04|2013-10-15|Abbvie Inc.|Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases| ES2622344T3|2008-12-04|2017-07-06|Abbvie Inc.|Apoptosis inducing agents for the treatment of cancer and immune and autoimmune diseases| US20100160322A1|2008-12-04|2010-06-24|Abbott Laboratories|Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases| CN102307872B|2008-12-05|2016-06-15|Abbvie公司|Selectively bring out the sulfamide derivative of Apoptosis medicament as the BCL-2-that is used for the treatment of cancer and immunological diseases| US8563735B2|2008-12-05|2013-10-22|Abbvie Inc.|Bcl-2-selective apoptosis-inducing agents for the treatment of cancer and immune diseases| US8586754B2|2008-12-05|2013-11-19|Abbvie Inc.|BCL-2-selective apoptosis-inducing agents for the treatment of cancer and immune diseases| WO2010077740A2|2008-12-09|2010-07-08|Cytokine Pharmasciences, Inc.|Novel antiviral compounds, compositions, and methods of use| US20120094963A1|2008-12-23|2012-04-19|of Queen Elizabeth near Dublin|Targeting prodrugs and compositions for the treatment of gastrointestinal diseases| JP5613177B2|2009-01-19|2014-10-22|アッヴィ・インコーポレイテッド|Apoptosis inducer for the treatment of cancer and immune and autoimmune diseases| NZ593593A|2009-01-19|2013-11-29|Abbvie Inc|Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases| US20100297194A1|2009-04-30|2010-11-25|Nathaniel Catron|Formulation for oral administration of apoptosis promoter| US8546399B2|2009-05-26|2013-10-01|Abbvie Inc.|Apoptosis inducing agents for the treatment of cancer and immune and autoimmune diseases| KR101920202B1|2009-05-26|2018-11-21|애브비 아일랜드 언리미티드 컴퍼니|Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases| TWI540132B|2009-06-08|2016-07-01|亞培公司|Pharmaceutical dosage form for oral administration of a bcl-2 family inhibitor| TWI520960B|2010-05-26|2016-02-11|艾伯維有限公司|Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases| EP2613769B1|2010-10-29|2017-09-27|AbbVie Ireland Unlimited Company|Melt-extruded solid dispersions containing an apoptosis-inducing agent| KR20180059560A|2010-10-29|2018-06-04|애브비 인코포레이티드|Solid dispersions containing an apoptosis-inducing agent| UA113157C2|2010-11-23|2016-12-26|METHOD OF TREATMENT WITH THE USE OF SELECTIVE BCL-2 INHIBITORS| EP2643322B1|2010-11-23|2017-08-30|Abbvie Inc.|Salts and crystalline forms of an apoptosis-inducing agent|US8546399B2|2009-05-26|2013-10-01|Abbvie Inc.|Apoptosis inducing agents for the treatment of cancer and immune and autoimmune diseases| US9034875B2|2009-05-26|2015-05-19|Abbvie Inc.|Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases| EA031737B1|2010-06-03|2019-02-28|Фармасайкликс, Инк.|USE OF INHIBITORS OF BRUTON'S TYROSINE KINASEFOR TREATING LEUKEMIA AND LYMPHOMA| KR20180059560A|2010-10-29|2018-06-04|애브비 인코포레이티드|Solid dispersions containing an apoptosis-inducing agent| EP2643322B1|2010-11-23|2017-08-30|Abbvie Inc.|Salts and crystalline forms of an apoptosis-inducing agent| UA113157C2|2010-11-23|2016-12-26|METHOD OF TREATMENT WITH THE USE OF SELECTIVE BCL-2 INHIBITORS| BR112014014529A2|2011-12-13|2019-09-24|Buck Inst For Res On Aging|methods to improve medical therapies| US10954567B2|2012-07-24|2021-03-23|Pharmacyclics Llc|Mutations associated with resistance to inhibitors of Bruton's Tyrosine Kinase | CN111437386A|2012-09-07|2020-07-24|吉宁特有限公司|Combination therapy of type II anti-CD 20 antibodies with selective Bcl-2 inhibitors| WO2014043223A1|2012-09-11|2014-03-20|Hospital For Special Surgery|Irhom2 inhibition for the treatment of complement mediated disorders| PT2970263T|2013-03-13|2018-10-03|Abbvie Inc|Processes for the preparation of an apoptosis-inducing agent| US20140275082A1|2013-03-14|2014-09-18|Abbvie Inc.|Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases| US10328058B2|2014-01-28|2019-06-25|Mayo Foundation For Medical Education And Research|Treating atherosclerosis by removing senescent foam cell macrophages from atherosclerotic plaques| JP6688224B2|2014-01-28|2020-04-28|バック インスティテュート フォー リサーチ オン エイジング|Treatment of osteoarthritis in joints by administration of means that inhibit MDM2| JP2017509336A|2014-03-20|2017-04-06|ファーマサイクリックス エルエルシー|Mutations associated with phospholipase C gamma 2 and resistance| WO2015171591A1|2014-05-05|2015-11-12|Board Of Trustees Of The University Of Arkansas|COMPOSITIONS AND METHODS FOR INHIBITING ANTIAPOPTOTIC Bcl-2 PROTEINS AS ANTI-AGING AGENTS| WO2016024230A1|2014-08-11|2016-02-18|Acerta Pharma B.V.|Therapeutic combinations of a btk inhibitor, a pi3k inhibitor, a jak-2 inhibitor, and/or a bcl-2 inhibitor| US10195213B2|2015-03-13|2019-02-05|Unity Biotechnology, Inc.|Chemical entities that kill senescent cells for use in treating age-related disease| MA43170A|2015-11-03|2018-09-12|Univ Texas|COMBINATION OF A BCL-2 INHIBITOR AND A MEK INHIBITOR FOR THE TREATMENT OF CANCER| CN109152933A|2016-04-21|2019-01-04|生物风险投资有限责任公司|Induce the compound of degradation and application thereof of anti-apoptotic BCL-2 family protein| DK3494115T3|2016-08-05|2021-01-18|Univ Michigan Regents|N-BENZAMIDE DERIVATIVES AS BCL-2 INHIBITORS| CN108658983B|2017-06-26|2019-12-20|深圳市塔吉瑞生物医药有限公司|N-benzenesulfonyl benzamide compound for inhibiting Bcl-2 protein and composition and application thereof| US10285998B1|2018-04-04|2019-05-14|The Menopause Method, Inc.|Composition and method to aid in hormone replacement therapy| CN112739349A|2018-09-18|2021-04-30|圣诺康生命科学公司|Combination therapy for the treatment of leukemia| US20220002290A1|2018-10-29|2022-01-06|Chia Tai Tianqing Pharmaceutical Group Co., Ltd.|Trifluoromethyl-substituted sulfonamide as bcl-2-selective inhibitor| CN113825754A|2019-05-24|2021-12-21|正大天晴药业集团股份有限公司|Selective BCL-2 inhibitors of disubstituted sulfonamides including methyl and trifluoromethyl| CN112707900A|2019-10-24|2021-04-27|上海科技大学|Protein degradation agent and application thereof in disease treatment| WO2021217051A1|2020-04-24|2021-10-28|Genentech, Inc.|Methods of using anti-cd79b immunoconjugates|
法律状态:
2016-11-29| B25A| Requested transfer of rights approved|Owner name: ABBVIE BAHAMAS LTD. (US) | 2017-01-03| B25A| Requested transfer of rights approved|Owner name: ABBVIE IRELAND UNLIMITED COMPANY (BM) | 2018-01-16| B07D| Technical examination (opinion) related to article 229 of industrial property law| 2018-04-03| B06F| Objections, documents and/or translations needed after an examination request according art. 34 industrial property law| 2018-07-31| B07E| Notice of approval relating to section 229 industrial property law| 2019-01-15| B06T| Formal requirements before examination| 2019-06-11| B09A| Decision: intention to grant| 2019-08-06| B16A| Patent or certificate of addition of invention granted|Free format text: PRAZO DE VALIDADE: 20 (VINTE) ANOS CONTADOS A PARTIR DE 22/11/2011, OBSERVADAS AS CONDICOES LEGAIS. (CO) 20 (VINTE) ANOS CONTADOS A PARTIR DE 22/11/2011, OBSERVADAS AS CONDICOES LEGAIS |
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 US41668910P| true| 2010-11-23|2010-11-23| US61/416,689|2010-11-23| PCT/US2011/061769|WO2012071374A1|2010-11-23|2011-11-22|Methods of treatment using selective bcl-2 inhibitors| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|