USOO8309.603B2

(12) United States Patent (10) Patent No.: US 8,309.603 B2 Dalton et al. (45) Date of Patent: Nov. 13, 2012

(54) SARMS AND METHOD OF USE THEREOF 4,191,775 A 3, 1980 Glen 4,239,776 A 12/1980 Glen et al. (75) Inventors: N SR n, t TN (S), 4,282,2184,386,080 A 8,5/1983 1981 GlenCrossley et al. et al. uane D. Miller, Germantown, 4411,890 A 10/1983 Momany et al. (US); Jeff Kearbey, Lakeland, TN (US) 4,465,507 A 8/1984 Konno et al. 4,636,505 A 1/1987 Tucker (73) Assignee: University of Tennessee Research 4,880,839 A 1 1/1989 Tucker Foundation, Knoxville, TN (US) 5,162,504 A 11/1992 Horoszewicz s s 5,179,080 A 1/1993 Rothkopfet al. (*) Notice: Subject to any disclaimer, the term of this ises: A 32. Swissan et al. patent is extended or adjusted under 35 5,656,651 A 8, 1997 Sovak et al. U.S.C. 154(b) by 212 days. 6,019,957. A 2/2000 Miller et al. 6,043,265 A 3/2000 Murugesan et al. 6,071,957 A 6/2000 Miller et al. (21) Appl. No.: 12/730,094 6, 160,011 A 12/2000 Miller et al. 6,482,861 B2 11/2002 Miller et al. (22) Filed: Mar. 23, 2010 6,492,554 B2 12/2002 Dalton et al. 6,548,529 B1 4/2003 Roblet al. (65) Prior Publication Data 6,569,896 B2 5/2003 Dalton et al. 6,777.427 B2 8/2004 Miyakawa et al. US 2010/0249228A1 Sep. 30, 2010 6,838,484 B2 1/2005 Steiner et al. 6,960,474 B2 11/2005 Salvati et al. Related U.S. Application Data 7,026,500 B2 4/2006 Dalton et al. 2001/0012839 A1 8, 2001 Miller et al. (63) Continuation-in-part of application No. 1 1/785,250, 2002/0173445 A1 11/2002 Salvati et al. filed on Apr. 16, 2007, now Pat. No. 7,772,433, which 2004/0214790 A1 10/2004 Borgens is a continuation-in-part of application No. FOREIGN PATENT DOCUMENTS 10/861,905, filed on Jun. 7, 2004, now Pat. No. EP O 040932 12/1981 7,803,970, application No. 12/730,094, which is a EP O 100 172 2, 1984 continuation-in-part of application No. 1 1/723,957, EP OOO 2892 2, 1985 filed on Mar. 22, 2007, now abandoned, and a EP O 253 SO3 1, 1988 continuation-in-part of application No. 1 1/510,844, EP 1221439 T 2002 filed on Aug. 28, 2006, application No. 12/730,094, 1360001 3. 1970 which is a continuation-in-part of application No. E. 5:32 gSo 1 1/892,595, filed on Aug. 24, 2007, now Pat. No. WO WO 89/07110 8, 1989 8,080,682, application No. 12/730,094, which is a WO WO 89/07111 8, 1989 continuation-in-part of application No. 1 1/634,380, WO WO93/04081 3, 1993 filed on Dec. 6, 2006, which is a continuation-in-part (Continued) of application No. 1 1/505.363, filed on Aug. 17, 2006, and a continuation-in-part of application No. OTHER PUBLICATIONS 1 1/505.499, filed on Aug. 17, 2006, now Pat. No. 7,645,898, and a continuation-in-part of application West, Anthony R., Solid State Chemistry and its Applications, Wiley, No. 1 1/826, 195, filed on Jul. 12, 2007. New York, 1988, pp. 358 & 365.* U.S. Appl. No. 09/644,970, filed Aug. 2, 2000, Dalton et al. (60) Provisional application No. 60/712,390, filed on Aug. U.S. Appl. No. 09/935,044, filed Aug. 23, 2001, Dalton et al. 31, 2005, provisional application No. 60/839,665, U.S. Appl. No. 09.935.045, filed Aug. 23, 2001, Dalton et al. filed on Aug. 24, 2006, provisional application No. U.S. Appl. No. 10/298,229, filed Nov. 28, 2002, Miller et al.

60/907,749, filed on Apr. 16, 2007, provisional ES.S. Appl.E. N.No. 857,, US, Tlleds UCL. A.3,.. : , RationLallOn et al. proV1S1onalEpists Notesasso's",applicauon No. ' ' ' '29A. ' U.S. Appl. No. 10/270,233,10/270.732, filed Oct. 15, 2002, Dalton et al. 12, 2006. U.S. Appl. No. 10/310,150, filed Dec. 5, 2002, Steiner et al. (51) Int. Cl. (Continued) A 6LX3L/275 (2006.01) (52) U.S. Cl...... 514/524; 514/522:514/616; 514/617; Primary Examiner — Shailendra Kumar 514f619 (74) Attorney, Agent, or Firm — Pearl Cohen Zedek Latzer, (58) Field of Classification Search ...... 514/522, LLP. Mark S. Cohen 514/524, 616, 617,619 See application file for complete search history. (57) ABSTRACT (56) References Cited This invention is directed to a feed composition and method of affecting the carcass composition by increasing the lean U.S. PATENT DOCUMENTS mass, reducing the fat mass, and/or reducing the percent fat 3,239,345 A 3/1966 Hodge mass comprising SARM compounds. 3,875,229 A 4, 1975 Gold 4,036.979 A 7, 1977 ASato 4,139,638 A 2f1979 Neri et al. 56 Claims, 24 Drawing Sheets US 8,309.603 B2 Page 2

FOREIGN PATENT DOCUMENTS Scription Superfamilies: novel targets for Small-molecule drug dis WO WO95/1977O 7, 1995 covery. J. Med. Chem., 38: 4855, 1995. WO WO9805962 2, 1998 Dalton JT. Mukherjee A. Zhu Z, Kirkovsky L, and Miller DD. Dis WO WO 98.53826 12/1998 covery of Androgens. Biochem. Biophys. Res. Com WO WO98,55153 12/1998 mun.,244(1): 1-4, 1998. WO WO/O1/27086 4/2001 Edwards JP, West SJ, Pooley CLF, Marschke KB, Farmer LJ, and WO WOO127622 4/2001 Jones TK. New nonsteroidal androgen receptor modulators based on WO WOO128990 4/2001 4-(trifluoromethyl)-2-(1H)-Pyrololidino.3.2-gquinolone. Bioorg. WO WOO1 34.563 5, 2001 Med. Chem. Lett., 8: 745, 1998. WO WOO168603 9, 2001 Eliason et al., “High Throughput Fluorescence Polarization-Based WO WOO2 OO617 1, 2002 Screening Assays for the Identification of Novel Nuclear Receptor WO WO O2, 16310 2, 2002 Ligands.” Abstracts of Papers, 223rd ACS National Meeting, WO WO?O2.22585 3, 2002 Orlando, FL, United States, (2002), Apr. 7, 2002. WO WOf O3,O77919 9, 2003 Berger et al., “Concepts and limitations in the application of OTHER PUBLICATIONS radiolabeled antiandrogens, estrogens, or androgens as isotropic Howard Tucker and Glynne J. Chesterson, J. Med Chem. 1988, 31. scanning agents for the prostate'. Invest. Urol, (1975), 1391, 10-16. pp. 885-887, “Resolution of the Nonsteroidal Antiandrogen-4'- Wahner, et al (1984) “Assesment of Bone Mineral Part 1' J Nucl. Cyano-3-(4-fluorophenyl)sulfonyl-2-hydroxy-2-methyl-3'- Medicine, 1134-1141. (trifluoromethyl)-propionanilide and the Determination of the Abso Wahner, et al (1985) “Bone Mineral Density of the Radius' J. Nucl lute Configuration of the Active Enantiomer'. Medicine 26 13-39. Faulkner KG, etal (1991) “Noninvasive measurements of bone mass, D. McKillop, et al., “Enantioselective metabolism and structure, and strength: current methods and experimental tech pharmacokinetics of Casodex in the male rat', Xenobiotica, 1995, niques.” Am J Rosentgenology 157: 1229-1237. vol. 25, No. 6, 623-634. Hanada, K., et al (2003) “Bone anabolic effects of S-40503, a novel Leonid Kirkovsky, et al., “I’I-Radionated Bicalutamide Analogs nonsteroidal selective androgen receptor modulator (SARM), in rat as Potential Imaging Agents for Prostate Cancer', Poster Presenta models of osteoporosis.” Biol. Pharm. Bull. 26:1563-1569. tion MEDI 155, 214th ACS National Meeting, Las Vegas, NV. Sep. Kalu, DN. (1991) “The ovariectomized rat model of postmenopausal 7-11, 1997. Department of Pharmaceutical Sciences, University of bone loss. Bone Miner 15, 175-91. Tennessee, Memphis, TN38163. Langer (1990) “New methods of drug delivery.” Science 249:1527 David T. Baird and Anna F. Glasier, “Hormonal Contraception— 1533. Drug Therapy'. The New England Journal of Medicine, May 27, Negro-Vilar, A. (1999) “Selective androgen receptor modulators 1993, pp. 1543-1549. (SARMs): a novel approach to androgen therapy for the new illen F.C. W. Wu, “Male Contraception: Current Status and Future Pros nium. J. Clin. Endocrin Metabol. 84:3459-3462. pects”. Clinical Endocrinology, (1988), 29, pp. 443-465. Koski GK, Kariko K, Xu S, Weissman D, Cohen PA, Czerniecki B.J. World Health Organisation Task Force on Methods for the Regulation Cutting edge: innate immune system discriminates between RNA of Male Fertility, “Contraceptive efficacy of -induced containing bacterial versus eukaryotic structural features that prime azoospermia in normal men'. The Lancet, vol. 336, Oct. 20, 1990, pp. for high-level IL-12 secretion by dendritic cells. J Immunol. Apr. 1, 955-959and 1517-1518. 2004; 172(7):3989-93. Heil F. Hemmi H. Hochrein H. Ampenberger F. Kirschning C, Akira C. G. Francisco, et al., “Long-acting contraceptive agents: S. Lipford G. Wagner H. Bauer S. Species-specific recognition of testosterone esters of unsaturated acids', , Jan. 1990, vol. 55, single-stranded RNA via toll-like receptor 7 and 8. Science. Mar. 5, Butterworths. 2004:303(5663): 1526-9. John M. Hoberman and Charles E. Yesalis, “The History of Synthetic Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. Innate Testosterone”, Scientific American, Feb. 1995, pp. 76-81. antiviral responses by means of TLR7-mediated recognition of Leonid Kirkovsky, et al., “Approaches to Irreversible non-steroidal single-stranded RNA. Science. Mar. 5, 2004:303(5663): 1529-31. chiral antiandrogens'. Department of Pharmaceutical Sciences, Uni Campfield et al., 1995, “Recombinant mouse OB protein: evidence versity of Tennessee, 47th Southeast/51st Southwest Joint Regional for a peripheral signal linking adiposity and central neural networks' Meeting of the American Chemical Society, Memphis, TN, Nov. Science 269:546-549. 29-Dec. 1, 1995. Considine et al., 1995, “Evidence against either a premature stop David J. Handelsman, “Bridging the gender gap in contraception: codon or the absence of obese gene mRNA in human obesity.” J. Clin. another hurdle cleared” The Medical Journal of Australia, vol. 154, Invest. 95:2986-2988. Feb. 18, 1996, pp. 230-233. Grundy, 1990, Disease-a-Month 36:645-696. Edwards.JP. Higuchi RI, Winn DT. Pooley CLF, Caferro TR, Hamann Halaas et al., 1995. “Weight-reducing effects of the plasma protein LG, Zhi L. Marschke KB, Goldman ME, and Jones TK. Nonsteroidal encoded by the obese gene.” Science 269:543-546. androgen receptor agonists based on 4-(trifluoromethyl)-2H Hamilton et al., 1995, << Increased obese mRNA expression in pyrano.3.2-gquinolin-2-one. Bioorg. Med. Chem. Lett., 9; 1003, omental fat cells from massively obese humans. Nature Med. 1:953. 1999. Lonnguist et al., 1995, Nature Med. 1:950. Zhi L. Tegley CM, Marschke KB, and Jones TK. Switching androgen Matsumoto, 1994. "Hormonal therapy of male hypogonadism” receptor antagonists to agonists by modifying C-ring Substituents on Endocrinol. Met. Clin. N. Am. 23:857-75. piperidino.3.2-gquinolone. Bioorg. Med. Chem. Lett., 9; 1009, Pelleymounter et al., 1995, “Effects of the obese gene product on 1999. body weight regulation in ob?ob mice.” Science 269:540-543. Higuchi RI, Edwards JP Caferro TR. Ringgenberg JD, Kong JW. Singh et al., 2003, 'Androgens stimulate myogenic differentiation Hamann LG, Arienti KL, Marschke KB, Davis RL, Farmer LJ, and and inhibit adipogenesis in C3H10T1/2 pluripotent cells through an Jones TK. 4-Alkyl- and 3,4-diaklyl-1,2,3,4-tetrahydro-8- androgen receptor-mediated pathway.” Endocrinology, pyridono5,6-glguinolines: potent, nonsteroidal androgen receptor 144(11):5081-8. agonists. Bioorg. Med. Chem. Lett., 9:1335, 1999. Sefton, 1987. “Implantable pumps.” CRC Crit. Ref. Biomed. Eng. Hamann LG. Mani NS, Davis RL. Wang XN, Marschke KB, and 14:2O1. Jones TK. Discovery of a potent, orally active nonsteroidal androgen Edwards, J. P. et al., Bio. Med. Chem. Let., 9, 1003-1008 (1999). receptor agonist: 4-ethyl-1,2,3,4-tetrahydro-6-(trifluoromethyl)-8- Hamann, L. G. et al., J. Med. Chem, 42, 210-212 (1999). pyridono.5,6-g-quinoline (LG 121071). J. Med. Chem., 42: 210, Heitzman, Environ Qual Saf Suppl. 1976; (5):89-98. 1999. Treat et al. In Liposomes in the Therapy of Infectious Disease and Rosen J. Day A. Jones TK, Jones ET, Nadzan AM, and Stein RB. Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353 Intracellular receptors and signal transducers and activators of tran 365 (1989). US 8,309.603 B2 Page 3

Sefton, CRC Grit. Ref. Biomed. Eng. 14:201 (1987). to polyethylene glycol in man' Cancer Treat Rep. 65(11-12): 1077 Buchwald et al., Surgery 88:507 (1980). $1 Nov.-Dec.1981. Yr Saudek et al., N. Engl. J. Med. 321:574 (1989). Katre et al. “Chemical modification of recombinant interleukin 2 by polyethylene glycol increases its potency in the murine Meth A Goodson, in Medical Applications of Controlled Release, Supra, vol. sarcoma model” Proc Natl Acad Sci U S A. 84(6): 1487-91. Mar. 2, pp. 116-138 (1984). 1987. Abuchowski et al. “Immunosuppressive properties and circulating life of Achromobacter glutaminase-asparaginase covalently attached * cited by examiner

U.S. Patent Nov. 13, 2012 Sheet 2 of 24 US 8,309.603 B2

Synthetic Scheme for (R)- Compound: H H H N O Y 2N NaOH/acetone wo-se O 9. -- C N XCOgh O-5C/RT132N NaOHacetone hrs - o No NBSRT DM O2-3 V-Br H3C

N1 NeoH 24% HB SOCTHF/O-5CO o, Reflux HO CH3 2^O'',-Br HO HO %,CH3 Br H3C

NC - B -- resoloe Br NC HHO CH

NC C NC

FC Cl J.--e.e re HO O F -2-propanol 82998 - FC Ol N 2. O C H

Figure 1B U.S. Patent Nov. 13, 2012 Sheet 3 of 24 US 8,309.603 B2

Synthetic Scheme for (S)- (oxirane intermediate) Compound:

?yco-h H O C d ACO2Hw 2N NaOH/acetone N - NBS/Dy N O -- N H - O-50C/RT13 hrs e- o, RT o'--xO HC

H O N O 249 HB SOCI/THF/O-5C cryos, O Reflux HO %, Br HC OH ox- HC OH 3. HC

NC NH2 O Ela NRT O -- C NC HCas: OH C,H aK2CO3 Cetone

O K.co NC O FC O N HO Cat-F 2-propanol FC N o () a HCO HC OH F

Figure 1C U.S. Patent Nov. 13, 2012 Sheet 4 of 24 US 8,309.603 B2

Synthetic Scheme for (R)-II (oxirane intermediate) Compound: CyCO2H lo a "2NNaOHlaceaCeOe N. NBS/DMF N 0-5 OCIRT13 hrs O RT O

H (ko w-r-e-24% HBr O ------>SOC/THF/O-50C ex O Reflux Hoxne, HCK. OH O ,-Br HC OH HC

NC FC NH2 Ela.N/RT Ol KCO -- CI & Br FC N-NC Br - 23 e NC HC OH H S aCetOne HC3 OH

NC NC -- -K.co. so- O O C FC N S HO F 2-propanol FC N S Ho3 H3CS OH F

Figure 1D U.S. Patent Nov. 13, 2012 Sheet 5 of 24 US 8,309.603 B2

Synthetic Scheme for (S)- involving B-ring addition prior to A-ring addition:

C -- t y $fCOH 2N NaOH/acetone ( N y CO2HNBS/DMF SkoN O N - Y-e- is V oh, H 0-5 OC/RT13 hrs o? RT O B 3C

H (X O CyN

O N \O Br H Hors, 2-propanolpropNaOH- OHC Alou O - H3C C C F F

o'- F SOC/THF/O-5C...... -ie- a- CC F H NC DO ElginEta NIR R HC OH HC OH

C FCDClius N'-ro C. F HC oH

Figure 1E U.S. Patent Nov. 13, 2012 Sheet 6 of 24 US 8,309.603 B2

Synthetic Scheme of (R)-II Involving B-ring Addition Prior to A-ring Addition:

CO2H N ar 2N NaOH facetone N NBS/DMF - - - - -se- -- e O NH 0-5 OC/RT13 hrs O RT o?Yao°,-Br HC

H

N H O ()'.ON O ConC. HC O -- HO - NaOH D - O Y °,-O m-e- O --Br 2-propanol HC HC C F C F

-T-se-SOCTHF/O-5°C C -- C El:NR,3 HO &k F CI Y'sOS F NC H3C OH HC OH

FC"Our sco C.F Had OH

Figure 1F U.S. Patent Nov. 13, 2012 Sheet 7 of 24 US 8,309.603 B2

Synthetic Scheme of (S)-II Using Chiral Intermediate and Involving B-ring Addition prior to A-ring Addition: t y CO2H H O C CO2H N ( x - 2N NaOH/acetOneTT T. e- N NBS/DME O O H N H 0-5 OC/RT13 hrs o, RT ox Br H3C

H H CBr3 'yo 24% HB O BrC-CHO oo O v O reflux HO HOSS CH3Br CO Cd H2SO2WPV4 2. S B O H3C H3C Br

H. CB H, CB, O CB'sO Concod HCl O O -- 2-propanolNaOH e- c. s - C -e- OHC F H3C Br C F

F F NC HQ2 in-OOH - a SOCITHF/O-5oC-e- C,2 in-oOH C + OHC OHC F3C NH2

NC O C

Enri- F3C ClusN 71, O C. F HC OH Figure 1G U.S. Patent Nov. 13, 2012 Sheet 8 of 24 US 8,309.603 B2

Synthetic Scheme for (R)-II Using Chiral Intermediate and Involving B-ring Addition Prior to A-ring Addition: C y CO2H H CO2H N 2N NaOH facetOne N NBS/DME O H X" O-50C/RT13Noteine-O hrs O spyRT 'l-Br HC

H H CBra (Po –4724% HBP- o^1 - PiaBC-CHO e XO O reflux CH conco H2SO4 HC Hy-CB?. H H,yers CB F O O - NaOHN7 -e- O O -(- Conco HC 2. -- 2-propanol (N-O C OHC F 3 Br C

F F NC HO OH SoCl/THF/O-5 Oc 9 OHC o-K3-c. le'" OHCy Quo C - F3C NH2

NC C

int- F3CDC. N --OSS O F HC OH

Figure 1H U.S. Patent Nov. 13, 2012 Sheet 9 of 24 US 8,309.603 B2

Synthetic Scheme for Racemic Compound Involving Oxazolidinedione Intermediate:

NC NC HO OH thiophosgene Ol -- 2 (Su-o C FC NH2 CHCl, NaHCO3 FC NCS O H3C F

NC -m-m-sersilver trifluoroacetate Ol O -1. NaOH -m-e- EtaN, acetonitrile fic Fo -Ko 2. Acid ^o F

NC C FC JClf-ONXro F Hac OH

Figure I U.S. Patent Nov. 13, 2012 Sheet 10 of 24 US 8,309.603 B2

Racemic Synthetic Scheme for Compound :

SOCl2 FC"O NH2 HO > Cl CH CH3

C NC NC JOI

FC Clf-N one. FCDC N - seri 3 methylethylketone H CH3 HHC, O

FC x- F Had bh

Figure 1. U.S. Patent Nov. 13, 2012 Sheet 11 of 24 US 8,309.603 B2

Large-scale Synthetic Scheme for (S)- : CO2H H O ( x'w 4NNaoh8. N NBS/DMF N o -- N -m-e- H BS/Dy O H <100C, 2 hrs O RT O Br MtBE

H O d O (X O 24%Reflux HBr HO 1. 2. Br- S5 OC - ar.HC OH O Br HC OH 3 HC

FC NH2 O EtN/RT NC Ol NC DO -- cras,HC OH > 3 N1S/H 12 Br

NC C. K2CO NC O FC V, Br --HO O F acetOne F3C H.N ?oH o-O- H3C OH HC F

Figure 1K U.S. Patent Nov. 13, 2012 Sheet 12 of 24 US 8,309.603 B2

Large-Scale Synthetic Scheme for (S)- :

CO2H H

( )? ANNaoh } N1' NBS/Dy N OJ. H NH H <10 OC, 2 hrs O H RT O MtBE H3C

H (X-O 24% HBr O SOCl2 e a', vO Reflux HO 2, Br O HC OH O Br HC OH 5 OC 3 H3C

O NC FC NH2 EtN/RT Ol K2CO3 -- C 1, Br F C N w Br - so NC HC OH 3 Hi?Had O acetone

NC NC re Kocoa, O FC N 2 -propano FC A. C 3 H C O HO F 3 H OH F 3 HC

Figure 1L. U.S. Patent Nov. 13, 2012 Sheet 13 of 24 US 8,309.603 B2

Compound of Formula S-II in castrated Rats

160.0%

140.0%

120.0% E0 mg/day E 100.0% B 0.01 mg/day 3 0.03 mg/day 80.0% 0.1 mg/day E 0.3 mg/day 6 60.0% a 0.75 mg/day SQ 1 mg/day 40.0%

20.0%

O.0% prostate Seminal vesicle levator ani

FIGURE 2 U.S. Patent Nov. 13, 2012 Sheet 14 of 24 US 8,309.603 B2

4 O ?

1 OO –UNo. 8

|0.1}{10OJ0% 6 –oH

4

No. ?aepojenseo !< No.pi-o NLi-o

FIGURE 3 U.S. Patent Nov. 13, 2012 Sheet 15 of 24 US 8,309.603 B2

12 O

100

-Hae,pºlunsao

FIGURE 4

U.S. Patent Nov. 13, 2012 Sheet 16 of 24 US 8,309.603 B2

2.9 3.4 B 28 g 3.3 S af d 9. 2.6 ; 3.2 25 g 3. ar t s 2.4 : 3. 23 s al 2.2 23

R R R sy s Sy cos es e es & so s es & & s & es

CO 34 D 35 3.3 2 0.90 32 3. 0.85 3. is Y gs 8O 2, 3.0 s 2.9 9 0.75 2.8 0.70 is 27 g, SS 2.6

SO 2.

FIGURE 5

U.S. Patent Nov. 13, 2012 Sheet 18 of 24 US 8,309.603 B2

18O

160

140

120

OO

80

60

40

20

Prostate Seminal Vesicles Levator Ani

Figure 7 U.S. Patent Nov. 13, 2012 Sheet 19 of 24 US 8,309.603 B2

200 18O

160 40

120 100

80

60

40

& S.S.S.S.S.S $. S-S-S-S-S-S OSSSSSSSSSSSSS $OSSS23S83.SSSSSS ŠOSSSSSS8, $ SSS&S Prostate Seminal Vesicles levator Ani Figure 8 U.S. Patent Nov. 13, 2012 Sheet 20 of 24 US 8,309.603 B2

80 Levator Ani 160 Emax = 147 it 10 ED = 0.03 it 0.01 140

20

100 POState Seminal Wesicles Emax a 147 - O 80 Emax = 188 it 135 ED = 0.21 it 0.04 ED = 0.2t 0.04 60

40

20

Figure 9

U.S. Patent Nov. 13, 2012 Sheet 22 of 24 US 8,309.603 B2

120.0%

100.0%

9 30 mg/day wn e & 0.01 mg/day XXXXX s & 0.03 mg/day E

SS 0.75 mg/day 0.1 mg/day

prostate Seminal Vesicle evator ani

Figure 11

U.S. Patent Nov. 13, 2012 Sheet 24 of 24 US 8,309.603 B2

160 Levator Ani 140 Emax - 131 2 ED = 0.02+ 0.01 120

S. 1 OO O CD 8O wn Prostate E 60 8 Emax F 83 it 25 O ED = 0.09 + 0.07 SS 40 s Seminal Wesicles Emax - 85 it 11 2O 2. ED = 0.17 it 0.05

Compound S-XXV (mg/day)

FIGURE 13 US 8,309,603 B2 1. 2 SARMS AND METHOD OF USE THEREOF produced each year. There is demand among producers for agents that increase productivity and performance Such as by CROSS REFERENCE TO RELATED increasing the proportion of meat per carcass, decreasing the APPLICATIONS amount of fat, decreasing the percent fat mass, is increasing 5 This application is a Continuation In Part application of the feed efficiency, increasing average daily gain (ADG), U.S. patent application Ser. No. 1 1/785,250, filed Apr. 16, decreasing feed-to-gain ratio (F:G), and modulation of meat 2007, now U.S. Pat. No. 7,772,433, which is a Continuation quality. In-Part application of U.S. patent application Ser. No. 10/861, The androgen receptor (“AR”) is a ligand-activated tran 905, filed Jun. 7, 2004 now U.S. Pat. No. 7,803,970; and this 10 Scriptional regulatory protein that mediates induction of male application is a Continuation-In-Part application of U.S. sexual development and function through its activity with patent application Ser. No. 1 1/723,957, filed on Mar. 22, 2007 endogenous androgens. The androgenic hormones are Ste now abandoned; and this application is a Continuation-In roids which are produced in the body by the testes and the Part application of U.S. patent application Ser. No. 1 1/510, cortex of the adrenal gland or can be synthesized in the 844, filed on Aug. 28, 2006, which claims the benefit of U.S. 15 laboratory. Androgenic steroids play an important role in Provisional Patent Application No. 60/712,390, filed Aug. 31, many physiologic processes, including the development of 2005; and this application is a Continuation in Part applica muscle and bone mass. The endogenous steroidal androgens tion of U.S. patent application Ser. No. 1 1/892.595, filed 24 include testosterone and dihydrotestosterone (“DHT). Other Aug. 2007 now U.S. Pat. No. 8,080,682, which claims the steroidal androgens include esters of testosterone, Such as the benefit of U.S. Provisional Application Ser. No. 60/839,665, cypionate, propionate, phenylpropionate, cyclopentylpropi filed Aug. 24, 2006 and U.S. Provisional Application Ser. No. 60/907,749, filed Apr. 16, 2007; and this application is a onate, isocarporate, enanthate, and decanoate esters, and Continuation in Part application of U.S. patent application other synthetic androgens such as 7-Methyl-Nortestosterone (“MENT) and its acetate ester (Sundaram et al., “7 Alpha Ser. No. 1 1/826, 195, filed Jul. 12, 2007 which claims the Methyl-Nortestosterone (MENT). benefit of U.S. Provisional Application Ser. No. 60/907,748, 25 filed 16 Apr. 2007, 60/839,665, filed Aug. 24, 2006, and Selective Androgen Receptor Modulators (SARMs) 60/830,158, filed Jul. 12, 2006; and this application is a Con include nonsteroidal compounds which retain the anabolic tinuation. In Part application of U.S. patent application Ser. activity of endogenous androgens with reduced androgenic No. 1 1/634,380, filed Dec. 6, 2006, which is a Continuation in activity (i.e., lesser effects in the prostate). SARMs benefi Part application of U.S. patent application Ser. No. 1 1/505, 30 cially promote muscle growth in humans and can treat, Sup 363, filed Aug. 17, 2006, and Ser. No. 1 1/505.499, filed Aug. press, prevent, or inhibit muscle wasting. 17, 2006, now U.S. Pat. No. 7,645,898, which claims priority The ability of a SARM to beneficially promote muscle to U.S. patent application Ser. No. 1 1/353,225, filed Feb. 14, growth in humans can be used as an alternative means to 2006, now U.S. Pat. No. 7,518,013 and Ser. No. 1 1/125,159, promote lean muscle deposition in animals, feedlot animals, filed May 10, 2005, now U.S. Pat. No. 7,205,437, and Ser. No. 35 beef cattle or finishing livestock. 11/062,752, filed Feb. 23, 2005 and Ser. No. 10/863,524, filed Currently, Paylean R (Ractopamine hydrochloride) is the Jun. 9, 2004, now Abandoned, all of which are hereby incor only commercially available feed additive to increase growth porated by reference in their entirety. rates and lean efficiency in finishing Swine. Ractopamine GOVERNMENT INTEREST STATEMENT 40 mediates its effect by stimulating B-adrenergic receptor. B-adrenergic agonists are a class of chemical compounds This invention was made in whole or in part with govern that stimulates B-receptors in the autonomic nervous system. ment support under grant number R01 DK598006, awarded This stimulation of B-receptors has the effect of promoting by the National Institute of Health; under grant number 45 growth in animals when the B-agonists are fed to the animals. R29CA068096 awarded by the National Cancer Institute, Although B-agonists are effective in promoting the produc National Institute of Health; under grant number tion of lean mass, there are three major drawbacks which limit R15HD35329, awarded by the National Institute of Child the market for Paylean R. The most significant drawback to Health and Human Development, National Institute of Health using B-agonists is the increased Susceptibility to stress in and under grant number R01 DK598.00, awarded by the 50 treated animals resulting in increased injuries. Secondly, the National Institute of Health. The government may have cer European Union has banned B-agonists from use in food tain rights in the invention. producing animals since 1996. Finally, treatment with B-ago nists results in rapid desensitization of the B-adrenergic FIELD OF THE INVENTION receptors, thereby limiting the amount of time an animal can 55 This invention is directed to a feed composition and meth benefit from this treatment (typically <4 wks). ods of affecting the carcass composition of an animal by the Accordingly, there is a need for methods in which new administration of SARM compounds. According to this compounds may be is used to improve carcass characteristics invention, SARMs improve meat production in feedlot ani and increase lean mass gain in animals. mals by increasing the lean mass, reducing the fat mass and/or 60 percent fat mass, improving feed efficiency and modulation of meat quality. SUMMARY OF THE INVENTION BACKGROUND OF THE INVENTION 65 In one embodiment, the present invention provides, a feed The production of Swine for food use is an important indus composition for an animal comprising a compound of for try in the United States with more than 100 million pigs mula IIIA: US 8,309,603 B2 4 1F is a synthetic scheme for the preparation of an (R) enan

(IIIA) tiomer of a compound of formula II (R-II) involving B-ring addition prior to A-ring addition. FIG. 1G is a synthetic scheme for the preparation of an (S) enantiomer of a com pound of formula II (S-II) using 2-tribromomethyl-1,3di oxolan-4-one intermediate and involving B-ring addition prior to A-ring addition. FIG. 1H is a synthetic scheme for the preparation of an (R) enantiomer of a compound of formula II (R-II) using 2-tribromomethyl-1,3dioxolan-4-one interme wherein 10 diate and involving B-ring addition prior to A-ring addition. Z is NO, CN, C1, F, Br, I, H, COR, COOH, or CONHR: FIG. 1I is a synthetic scheme for preparation of a racemic Y is CF, alkoxy, alkyl, hydroxyalkyl, alkylaldehyde, formyl, mixture of a compound of formula II, involving oxazo H, F, Br, C1, I, CN, or Sn(R): lidinedione intermediate and Bring addition prior to A ring. R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, CHF 15 FIG. 1J is a synthetic scheme for preparation of a racemic CF, CFCF, aryl, phenyl, halogen, alkenyl or OH: mixture of a compound of formula II, involving an oxirane R is H, F, Cl, Br, I, CH, CF, OH, CN, NO, NHCOCH, intermediate and A ring addition prior to Bring. FIG. 1K is a NHCOCF, NHCOR, alkyl, arylalkyl, OR, NH, NHR, N(R) synthetic scheme for preparation of a large scale of an (S) SR; enantiomer of a compound of formula II (S-II). FIG. 1L is a R is H, F, Cl, Br, I, CN, NO, COR, COOH, CONHR, CF, synthetic scheme for preparation of a large scale of an (S) Sn(R): enantiomer of a compound of formula II (S-II), including an Q is H, alkyl, halogen, CF, CN, C(R) Sn(R), N(R), oxirane intermediate. NHCOCH, NHCOCF, NHCOR, NHCONHR, NHCOOR, FIG. 2: Anabolic and androgenic pharmacology of com OCONHR, CONHR, NHCSCH, NHCSCF, NHCSR, pound of formula S-II in castrated rats (ORX). NHSOCH, NHSOR, OH, OR, COR, OCOR, OSOR, 25 FIG. 3: Levator ani weight effects in castrated rats for a SOR, SR; panel of compounds. n is an integer of 1-4; and FIG. 4: Prostate weight effects in castrated rats for a panel m is an integer of 1-3: of compounds. or its isomer, pharmaceutically acceptable salt, crystal, N-OX FIG. 5: Effect of compound of formula S-II on the growth ide, hydrate or any combination thereof. 30 performance and carcass composition of finishing pigs. FIG. In one embodiment, the present invention provides a 5A shows the increase of average daily gain (ADG) over the method of affecting the carcass composition of an animal course of the study. FIG. 5B shows the decrease feed to gain comprising administering a compound of formula IIIA. ratio. FIG. 5C shows the increased fat free lean gain per day. In one embodiment, the present invention provides a FIG. 5D shows an increase in ADG for days 21-28. method of increasing lean mass of an animal comprising 35 FIG. 6: Depicts a synthetic scheme for the preparation of an administering a compound of formula IIIA or its isomer, (S) enantiomer of a compound of formula XXIII. pharmaceutically acceptable salt, pharmaceutical product, FIG. 7: Pharmacology of a compound of formula S-XXIII crystal, N-oxide, hydrate or any combination thereof. in intact rats. Asterisks represent statistically significant dif In one embodiment, the present invention provides a ferences between the weight of the organ in the indicated method of reducing fat is mass of an animal comprising 40 group and that observed in intact animals treated with vehicle administering a compound of formula IIIA or its isomer, (P<0.05). pharmaceutically acceptable salt, pharmaceutical product, FIG. 8: Organ weights from castrated, compound of for crystal, N-oxide, hydrate or any combination thereof. mula S-XXIII-treated rats presented as a percentage of intact In another embodiment, the methods of this invention are control. * P-value:0.05 versus intact controls. directed to affecting the carcass composition of an animal; 45 FIG. 9: Organ weight maintenance dose-response curves increasing lean mass of an animal and/or reducing fat mass/ for a compound of formula S-XXIII and seminal vesicles percent fat mass of an animal wherein said animal is a feedlot (closed squares) were obtained by nonlinear regression animal, a beef cattle or a finishing livestock. analysis using the sigmoid Emax model in WinNonlinP). FIG.10: Depicts a synthetic scheme for the preparation of BRIEF DESCRIPTION OF THE DRAWINGS 50 an (S) enantiomer of a compound of formula XXIV. FIG. 11: Anabolic and androgenic activity of a compound The present invention will be understood and appreciated of formula S-XXIV in ORX rats. more fully from the following detailed description taken in FIG. 12: Depicts a synthetic scheme for the preparation of conjunction with the appended drawings in which: an (S) enantiomer of a compound of formula XXV. FIG. 1: Synthetic schemes for the preparation of compound 55 FIG. 13: Anabolic and androgenic activity of a compound of formula II. of formula S-XXV in ORX rats. FIG. 1A is a synthetic scheme for the preparation of an (S) enantiomer of a compound of formula II (S-II). FIG. 1B is a DETAILED DESCRIPTION OF THE PRESENT synthetic scheme for the preparation of an (R) enantiomer of INVENTION a compound of formula II (R-II). FIG. 1C is a synthetic 60 scheme for the preparation of an (S) enantiomer of a com In the following detailed description, numerous specific pound of formula II (S-II) including an oxirane intermediate. details are set forth in order to provide a thorough understand FIG. 1D is a synthetic scheme for the preparation of an (R) ing of the invention. However, it will be understood by those enantiomer of a compound of formula II (R-II) including an skilled in the art that the present invention may be practiced oxirane intermediate. FIG. 1E is a synthetic scheme for the 65 without these specific details. In other instances, well-known preparation of an (S) enantiomer of a compound of formula II methods, procedures, and components have not been (S-II) involving B-ring addition prior to A-ring addition. FIG. described in detailso as not to obscure the present invention. US 8,309,603 B2 5 6 This invention provides, in one embodiment, feed compo R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, CHF sition for animals comprising acylanilides characterized by CF, CFCF, aryl, phenyl, F, Cl, Br, I, alkenyl or OH: the structure of formulas I-XXV. In one embodiment, the or its isomer, pharmaceutically acceptable salt, pharmaceuti compound is a SARM. In one embodiment, the compound cal product, N-oxide, hydrate or any combination thereof. and/or feed composition is useful in affecting the carcass In one embodiment, the present invention provides, a com composition, increasing the lean mass, reducing the fat mass pound of formula S-II: of an animal or reducing percent fat mass, increasing feed efficiency, increasing average daily gain (ADG), decreasing feed to gain ratio (F:G) of an animal, including a feedlot S-II 10 animal, a beef cattle or a finishing livestock. In another NC C embodiment, the compound and/or feed composition is use ful in increasing muscle growth of an animal, modulation of meat quality, or enhancing productive life of animals includ FC NH ing feedlot animals, beef cattle and finishing livestock. 15 In one embodiment, the compounds of this invention pro Hd OH vide compounds, compositions and methods of treating a variety of conditions or diseases, including, inter alia, oral or its isomer, pharmaceutically acceptable salt, pharma testosterone replacement therapy, male contraception, main ceutical product, N-oxide, hydrate or any combination taining sexual desire in women, osteoporosis, treating pros thereof. tate cancer and/or imaging prostate cancer. In some embodi In one embodiment, the present invention provides, a com ments, the compounds of this invention are nonsteroidal pound represented by the structure of formula III: ligands for the AR and exhibit androgenic and/or anabolic activity. In some embodiments, the compounds are partial III agonists or partial antagonists in a tissue selective manner. In 25 Some embodiments, the compounds are full agonists or full antagonists in a tissue selective manner, which in some embodiments, allows for tissue-selective androgenic and/or anabolic effects. These agents may be active alone or in combination with progestins or estrogens, or other agents, as 30 herein described. In other embodiments, the agents are ago nists, antagonists, partial agonists or partial antagonists. In some embodiments, this invention provides compounds, wherein which are useful in androgen replacement therapy (ART), X is a bond, O, CH, NH, Se, PR, NO or NR; useful ina) improving body composition; b) increasing bone 35 G is O or S: mineral density (BMD); c) increasing bone mass; d) increas T is OH, OR, NHCOCH, or NHCOR; ing bone strength; e) improving bone function: f) decreasing R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, fracture risk; g) increasing muscle strength; h) increasing CHF, CF, CFCF, aryl, phenyl, halogen, alkenyl or muscle function: i) improving exercise tolerance; j) enhanc OH: ing libido, k) improving sexual performance; and/or 1) 40 improving mood and/or m) improving cognition. R is CH, CHF, CHF, CF, CHCH, or CFCF: In some embodiments, this invention provides synthetic R is H, F, Cl, Br, I, CH, CF, OH, CN, NO, NHCOCH, processes of preparation of the SARM compounds of this NHCOCF, NHCOR, alkyl, arylalkyl, OR, NH, NHR, invention. In some embodiments, the invention provides N(R), SR; compositions comprising the selective androgen receptor 45 R is H, F, Cl, Br, I, CN, NO, COR, COOH,CONHR, CF, modulator compounds or use of the same for binding an AR, Sn(R), or modulating spermatogenesis, bone formation and/or resorp R together with the benzene ring to which it is attached tion, treating muscle wasting or diseases associated with forms a fused ring system represented by the struc muscle wasting, treating prostate cancer, and/or providing ture: hormonal therapy for androgen-dependent conditions. 50 In one embodiment, the present invention provides, a com pound of formula (I):

o O o (I) 55 2 / X/ Y Y

60 Z is NO, CN, C1, F, Br, I, H, COR, COOH, or CONHR: Y is CF, alkoxy, alkyl, hydroxyalkyl, alkylaldehyde, formyl, H, F, Br, Cl, I, CN, or Sn(R): wherein Q, is alkyl, F, Cl, Br, I, CF, CN, C(R), Sn(R), Q is H, alkyl, halogen, CF, CN, C(R), Sn(R), N(R), N(R), NHCOCH, NHCOCF, NHCOR, NHCONHR, NHCOCH, NHCOCF, NHCOR, NHCONHR, NHCOOR, OCONHR, CONHR, NHCSCH, NHCSCF, 65 NHCOOR, OCONHR, CONHR, NHCSCH, NHC NHCSR, NHSOCH, NHSOR, OR, COR, OCOR, OSOR, SCF, NHCSR, NHSOCH, NHSOR, OH, OR, COR, SOR, SR; and OCOR, OSOR, SOR, SR: or Q together with the ben US 8,309,603 B2 7 8 Zene ring to which it is attached is a fused ring system present invention provides a compound of formula III or IIIA represented by structure A, B or C: wherein Z is NO. In another embodiment, the present inven tion provides a compound of formula III or IIIA, whereinY is CH. In another embodiment, the present invention provides A a compound of formula III or IIIA, whereinY is H. In another 21 NH O embodiment, the present invention provides a compound of formula III or IIIA wherein Y is CF. In another embodiment, Ás the present invention provides a compound of formula III or 10 IIIA, wherein Y is C1. In another embodiment, the present 21 NH O B invention provides a compound of formula III or IIIA wherein R is H and none ofY, Z, Q or R are H. In another embodi ment, the present invention provides a compound of formula Ás 21 III or IIIA wherein R is CN. In another embodiment, the C 15 present invention provides a compound of formula III or IIIA 21 NH wherein R is C1. In another embodiment, the present inven tion provides a compound of formula III or IIIA wherein R is F. In another embodiment, the present invention provides a S-N/ compound of formula III or IIIA wherein Q is CN. In another embodiment, the present invention provides a compound of n is an integer of 1-4; and formula III or IIIA wherein Q is F. In another embodiment, m is an integer of 1-3: the present invention provides a compound of formula III or or an isomer, pharmaceutically acceptable salt, pharma IIIA wherein Q is C1. In another embodiment, if R of formula ceutical product, N-oxide, hydrate thereof or any com III or IIIA is H, then none of ZorY or R or Q are H. bination thereof. 25 In one embodiment, the present invention provides, a feed In one embodiment, the present invention provides a com composition for an animal comprising a compound of for pound characterized by the structure of formula IV: mula IIIA:

30 (IIIA) m(R3) (CI)n IV ex S/S Nc SA 2 35

wherein R, m and n are as described for the structure of wherein formula III. Z is NO, CN, C1, F, Br, I, H, COR, COOH, or CONHR: 40 Y is CF, alkoxy, alkyl, hydroxyalkyl, alkylaldehyde, formyl, In one embodiment, this invention provides a compound of H, F, Br, C1, I, CN, or Sn(R): formula S-XXIII: R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, CHF CF, CFCF, aryl, phenyl, halogen, alkenyl or OH: R is H, F, Cl, Br, I, CH, CF, OH, CN, NO, NHCOCH, 45 S-XXIII NHCOCF, NHCOR, alkyl, arylalkyl, OR, NH, NHR, N(R), SR; NC CN. R is H, F, Cl, Br, I, CN, NO, COR, COOH, CONHR, CF, O Sn(R): Q is H, alkyl, halogen, CF, CN, C(R) Sn(R), N(R), 50 FC NH O F NHCOCH, NHCOCF, NHCOR, NHCONHR, NHCOOR, Hd oH OCONHR, CONHR, NHCSCH, NHCSCF, NHCSR, NHSOCH, NHSOR, OH, OR, COR, OCOR, OSOR, SOR, SR; In one embodiment, this invention provides a compound of n is an integer of 1-4; and 55 formula XXIV: m is an integer of 1-3: or its isomer, pharmaceutically acceptable salt, crystal, N-OX ide, hydrate or any combination thereof to said Subject. S-XXIV In one embodiment, the present invention provides a com NC CN. pound of formula III wherein X is O. In another embodiment, 60 the present invention provides a compound of formula III wherein T is OH. In another embodiment, the present inven tion provides a compound of formula III wherein R is CH. C In another embodiment, the present invention provides a compound of formula III or IIIA wherein Z is CN. In another 65 embodiment, the present invention provides a compound of formula III or IIIA wherein Zis F. In another embodiment, the US 8,309,603 B2 10 In one embodiment, this invention provides a compound of SARMs used in the methods of the present invention may formula XXV: existin, and be isolated in, optically-active or racemic forms. Some compounds may also exhibit polymorphism. It is to be understood that the present invention encompasses any race S-XXV 5 mic, optically-active, polymorphic, or stereoisomeric form, NC CN. or mixtures thereof, which form possesses properties useful

in the treatment of androgen-related conditions described herein. In one embodiment, the SARMs are the pure (R)-

FC NH isomers. In another embodiment, the SARMs are the pure 10 (S)-isomers. In another embodiment, the SARMs are a mix Hd oH ture of the (R) and the (S) isomers. In another embodiment, the SARMs are a racemic mixture comprising an equal amount of the (R) and the (S) isomers. It is well known in the In one embodiment, this invention provides an analog of art how to prepare optically-active forms (for example, by the compound of formulas I-XXV. In another embodiment, 15 resolution of the racemic form by recrystallization tech this invention provides a derivative of the compound of for niques, by synthesis from optically-active starting materials, mulas I-XXV. In another embodiment, this invention pro by chiral synthesis, or by chromatographic separation using a vides a prodrug of the compound of formulas I-XXV. In chiral stationary phase). another embodiment, this invention provides a metabolite of In one embodiment, this invention encompasses the use of the compound of formulas I-XXV. In another embodiment, various optical isomers of the SARM compound. It will be this invention provides a pharmaceutically acceptable salt of appreciated by those skilled in the art that the SARMs of the the compound of formulas I-XXV. In another embodiment, present invention contain at least one chiral center. Accord this invention provides a pharmaceutical product of the com ingly, the SARMs used in the methods of the present inven pound of formulas I-XXV. In another embodiment, this tion may exist in, and be isolated in, optically-active or race invention provides a hydrate of the compound of formulas 25 mic forms. Some compounds may also exhibit I-XXV. In another embodiment, this invention provides an polymorphism. It is to be understood that the present inven N-oxide of the compound of formulas I-XXV. In another tion encompasses any racemic, optically-active, polymor embodiment, this invention provides a polymorph of the com phic, or stereoisomeric form, or mixtures thereof, which form pound of formulas I-XXV. In another embodiment, this possesses properties useful in the treatment of androgen invention provides a crystal of the compound of formulas 30 related conditions described herein. In one embodiment, the I-XXV. In another embodiment, this invention provides an SARMs are the pure (R)-isomers. In another embodiment, the impurity of the compound of formulas I-XXV. In another SARMs are the pure (S)-isomers. In another embodiment, the embodiment, this invention provides a combination of any of SARMs are a mixture of the (R) and the (S) isomers. In an analog, derivative, metabolite, isomer, prodrug, pharma another embodiment, the SARMs area racemic mixture com ceutically acceptable salt, pharmaceutical product, poly 35 prising an equal amount of the (R) and the (S) isomers. It is morph, crystal, impurity, hydrate, N-oxide of the compound well known in the art how to prepare optically-active forms of formulas I-XXV. (for example, by resolution of the racemic form by recrystal As contemplated herein, the present invention relates to the lization techniques, by synthesis from optically-active start use of a SARM compound and/or its analog, derivative, iso ing materials, by chiral synthesis, or by chromatographic mer, metabolite, pharmaceutically acceptable salt, pharma 40 separation using a chiral stationary phase). ceutical product, hydrate, N-oxide, polymorph, crystal, The invention includes “pharmaceutically acceptable impurity or combinations thereof. In one embodiment, the salts' of the compounds of this invention, which may be invention relates to the use of an analog of the SARM com produced, by reaction of a compound of this invention with an pound. In another embodiment, the invention relates to the acid or base. use of a derivative of the SARM compound. In another 45 Suitable pharmaceutically-acceptable salts of amines of embodiment, the invention relates to the use of an isomer of formulas I-XXV may be prepared from an inorganic acid or the SARM compound. In another embodiment, the invention from an organic acid. In one embodiment, examples of inor relates to the use of a metabolite of the SARM compound. In ganic salts of amines are bisulfates, borates, bromides, chlo another embodiment, the invention relates to the use of a rides, hemisulfates, hydrobromates, hydrochlorates, 2-hy pharmaceutically acceptable salt of the SARM compound. In 50 droxyethylsulfonates (hydroxyethanesulfonates), iodates, another embodiment, the invention relates to the use of a iodides, isothionates, nitrate, persulfates, phosphate, Sulfates, pharmaceutical product of the SARM compound. In another Sulfamates, Sulfanilates, Sulfonic acids (alkylsulfonates, aryl embodiment, the invention relates to the use of a hydrate of Sulfonates, halogen Substituted alkylsulfonates, halogen Sub the SARM compound. In another embodiment, the invention stituted arylsulfonates), Sulfonates and thiocyanates. relates to the use of an N-oxide of the SARM compound. In 55 In one embodiment, examples of organic salts of amines another embodiment, the invention relates to the use of a may be selected from aliphatic, cycloaliphatic, aromatic, polymorph of the SARM compound. In another embodiment, araliphatic, heterocyclic, carboxylic and Sulfonic classes of the invention relates to the use of a crystal of the SARM organic acids, examples of which are acetates, arginines, compound. In another embodiment, the invention relates to aspartates, ascorbates, adipates, anthranilate, algenate, the use of an impurity of the SARM compound. 60 alkane carboxylates, Substituted alkane carboxylates, algi As defined herein, the term "isomer' includes, but is not nates, benzenesulfonates, benzoates, bisulfates, butyrates, limited to, optical isomers and analogs, structural isomers and bicarbonates, bitartrates, carboxilate, citrates, camphorates, analogs, conformational isomers and analogs, and the like. In camphorsulfonates, cyclohexylsulfamates, cyclopentanepro one embodiment, the term "isomer' is meant to encompass pionates, calcium edetates, camsylates, carbonates, clavulan optical isomers of the SARM compound. It will be appreci 65 ates, cinnamates, dicarboxylates, digluconates, dodecylsul ated by those skilled in the art that the SARMs of the present fonates, dihydrochlorides, decanoates, enanthuates, invention contain at least one chiral center. Accordingly, the ethanesulfonates, edetates, edisylates, estolates, esylates, US 8,309,603 B2 11 12 fumarates, formates, fluorides, galacturonate gluconates, position Suitable for pharmaceutical use (pharmaceutical glutamates, glycolates, glucorate, glucoheptanoates, glycero composition), for example, as described herein. phosphates, gluceptates, glycolylarsanilates, glutarates, An “alkyl group refers, in one embodiment, to a saturated glutamate, heptanoates, hexanoates, hydroxymaleates, aliphatic hydrocarbon, including straight-chain, branched hydroxycarboxlic acids, hexylresorcinates, hydroxyben chain and cyclic alkyl groups. In one embodiment, the alkyl Zoates, hydroxynaphthoate, hydrofluorate, lactates, lacto group has 1-12 carbons. In another embodiment, the alkyl bionates, laurates, malates, maleates, methylenebis(beta-OX group has 1-7 carbons. In another embodiment, the alkyl ynaphthoate), malonates, mandelates, mesylates, methane group has 1-6 carbons. In another embodiment, the alkyl Sulfonates, methylbromides, methylnitrates, methylsul group has 1-4 carbons. The alkyl group may be unsubstituted or to Substituted by one or more groups selected from halo fonates, monopotassium maleates, mucates, monocarboxy 10 gen, hydroxy, alkoxy carbonyl, amido, alkylamido, dialkyla lates, nitrates, naphthalenesulfonates, 2-naphthalene mido, nitro, amino, alkylamino, dialkylamino, carboxyl, thio Sulfonates, nicotinates, napsylates, N-methylglucamines, and thioalkyl. In one embodiment, the alkyl group is CHs. oxalates, octanoates, oleates, pamoates, phenylacetates, An “alkenyl group refers, in another embodiment, to an picrates, phenylbenzoates, pivalates, propionates, phthalates, unsaturated hydrocarbon, including straight chain, branched phenylacetate, pectinates, phenylpropionates, palmitates, 15 chain and cyclic groups having one or is more double bond. pantothenates, polygalacturates, pyruvates, quinates, salicy The alkenyl group may have one double bond, two double lates, succinates, Stearates, Sulfanilate, Subacetates, tart bonds, three double bonds etc. Examples of alkenyl groups arates, theophyllineacetates, p-toluenesulfonates (tosylates), are ethenyl, propenyl, butenyl, cyclohexenyl etc. In one trifluoroacetates, terephthalates, tannates, teoclates, trihalo embodiment, the alkylene group has 1-12 carbons. In another acetates, triethiodide, tricarboxylates, undecanoates and Val embodiment, the alkylene group has 1-7 carbons. In another erates. embodiment, the alkylene group has 1-6 carbons. In another In one embodiment, examples of inorganic salts of car embodiment, the alkylene group has 1-4 carbons. The alkenyl boxylic acids or phenols may be selected from ammonium, group may be unsubstituted or Substituted by one or more alkali metals to include lithium, Sodium, potassium, cesium; groups selected from halogen, hydroxy, alkoxy carbonyl, alkaline earth metals to include calcium, magnesium, alu 25 amido, alkylamido, dialkylamido, nitro, amino, alkylamino, minium; Zinc, barium, cholines, quaternary ammoniums. dialkylamino, carboxyl, thio and thioalkyl. In another embodiment, examples of organic salts of car A "haloalkyl group refers to an alkyl group as defined boxylic acids or phenols may be selected from arginine, above, which is Substituted by one or more halogen atoms, in one embodiment by F, in another embodiment by Cl, in organic amines to include aliphatic organic amines, alicyclic another embodiment by Br, in another embodiment by I. organic amines, aromatic organic amines, benzathines, t-bu 30 An “aryl group refers to an aromatic group having at least tylamines, benethamines (N-benzylphenethylamine), dicy one carbocyclic aromatic group or heterocyclic aromatic clohexylamines, dimethylamines, diethanolamines, ethano group, which may be unsubstituted or substituted by one or lamines, ethylenediamines, hydrabamines, imidazoles, more groups selected from halogen, haloalkyl, hydroxy, lysines, methylamines, meglamines, N-methyl-D-glucam alkoxy carbonyl, amido, alkylamido, dialkylamido, nitro, ines, N,N'-dibenzylethylenediamines, , 35 amino, alkylamino, dialkylamino, carboxy or thio or thio organic amines, ornithines, pyridines, picolies, piperazines, alkyl. Nonlimiting examples of aryl rings are phenyl, naph procain, tris(hydroxymethyl)methylamines, triethylamines, thyl, pyranyl, pyrrolyl, pyrazinyl, pyrimidinyl, pyrazolyl, triethanolamines, trimethylamines, tromethamines and pyridinyl, furanyl, thiophenyl, thiazolyl, imidazolyl, isox leaS. azolyl, and the like. In one embodiment, the aryl group is a 4-8 In one embodiment, the salts may be formed by conven 40 membered ring. In another embodiment, the aryl group is a tional means, such as by reacting the free base or free acid 4-12 membered ring(s). In another embodiment, the aryl form of the product with one or more equivalents of the group is a 6 membered ring. In another embodiment, the aryl appropriate acid or base in a solvent or medium in which the group is a 5 membered ring. In another embodiment, the aryl salt is insoluble or in a solvent such as water, which is group is 2-4 fused ring system. removed in vacuo or by freeze drying or by exchanging the 45 A “hydroxyl group refers to an OH group. It is understood ions of a existing salt for another ion or Suitable ion-exchange by a person skilled in the art that when T is OR, R is not OH. resin. In one embodiment, the term “halogen refers to in one In one embodiment, the invention also includes N-oxides embodiment to F, in another embodiment to Cl, in another of the amino substituents of the compounds described herein. embodiment to Br, in another embodiment to I. Also, esters of the phenolic compounds can be made with 50 An “arylalkyl group refers, in another embodiment, to an aliphatic and aromatic carboxylic acids, for example, acetic alkyl bound to an aryl, wherein alkyl and aryl are as defined acid and benzoic acid esters. above. An example of an arylalkyl group is a benzyl group. This invention provides derivatives of the SARM com In another embodiment, the present invention provides pounds. In one embodiment, “derivatives” includes but is not process for preparing a selective androgen receptor modula limited to ether derivatives, acid derivatives, amide deriva 55 tor (SARM) compound represented by the structure of for tives, ester derivatives and the like. In another embodiment, mula III: this invention further includes hydrates of the SARM com pounds. III In one embodiment, “hydrate' includes but is not limited to hemihydrate, monohydrate, dihydrate, trihydrate and the like. 60 (R3) This invention provides, in other embodiments, metabo lites of the SARM compounds. In one embodiment, “metabo lite” means any Substance produced from another Substance by metabolism or a metabolic process. This invention provides, in other embodiments, pharma 65 ceutical products of the SARM compounds. The term “phar maceutical product” refers, in other embodiments, to a com US 8,309,603 B2 13 14 wherein X is a bond, O, CH, NH, Se, PR, NO or NR; 10 G is O or S: T is OH, OR, NHCOCH, or NHCOR; 5 R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, 2. CHF, CF, CFCF, aryl, phenyl, halogen, alkenyl or T OH: R is CH, CHF, CHF, CF, CHCH, or CFCF: 10 wherein Z.Y. G. R. T. R. and mare as defined above and R is H, F, Cl, Br, I, CH, CF, OH, CN, NO, NHCOCH, L is a leaving group, with a compound of formula II: NHCOCF, NHCOR, alkyl, arylalkyl, OR, NH, NHR,

11 R is H, F, Cl, Br, I, CN, NO, COR, COOH,CONHR, CF, 15 (R2) Sn(R), or R together with the benzene ring to which it is attached forms a fused ring system represented by the struc ture: wherein Q, X, R and n are as defined above. In one embodiment, the coupling step is carried out in the presence of a base. In another embodiment, the leaving group L is Br. In another embodiment, the compound of formula 10 is 25 prepared by: O V M a) preparing a compound of formula 13 by ring opening of a Z X / z1K / cyclic compound of formula 12: Y Y 30 Z is NO, CN, C1, F, Br, I, H, COR, COOH, or CONHR: Y is CF, alkoxy, alkyl, hydroxyalkyl, alkylaldehyde, formyl, H, F, Br, Cl, I, CN, or Sn(R): Q is H, alkyl, halogen, CF, CN, C(R) Sn(R), N(R), 35 HO NHCOCH NHCOCF, NHCOR, NHCONHR, NHCOOR, OCONHR, CONHR, NHCSCH, NHC SCF, NHCSR, NHSOCH, NHSOR, OH, OR, COR, OCOR, OSOR, SOR, SR: or Q together with the ben Zene ring to which it is attached is a fused ring system 40 wherein L, R, G and T are as defined above, and T is O or represented by structure A, B or C: NH; and b) reacting an amine of formula 14:

NH O A 45 14 (R3) 21 NH2 S.Ás 2, B 50 tau 21 NH O wherein Z. Y. R. and m are as defined above, with the Ás 2 compound of formula 13, in the presence of a coupling 55 reagent, to produce the compound of formula 10. C 21 NH 10

60

n is an integer of 1-4; and 2 m is an integer of 1-3: T the process comprising the step of coupling a compound of 65 formula (10): It is understood to a person skilled in the art that when T is O or NH, T in compound 13 is O or NH. Thus, when T in US 8,309,603 B2 15 16 compound 13 is OR, the reaction will involve a further step of another embodiment, the present invention provides a process converting the OH to OR by a reaction with, for example, an for preparing a compound of formula III wherein Q is C1. alkyl halide R X. When T in compound 13 is NHCOR, In another embodiment, the present invention provides a NHCOCH, the reaction will involve a further step of con process for preparing a selective androgen modulator com verting the NH, to NHCOR or NHCOCH, by a reaction with, 5 pound represented by the structure of formula II, as depicted for example, the corresponding acyl chloride CICOR or in FIG. 1 and Example 1: CICOCH. In one embodiment, step (a) is carried out in the presence of HBr. In one embodiment, whereby compound 13 of step (a) is 10 NC reacted with a coupling agent prior to step (b). In one embodiment, the coupling step is carried out in the presence of a base. In another embodiment, the leaving group FC L is Br. 15 In another embodiment, the process further comprises the C step of converting the selective androgen receptor modulator K2CO3 (SARM) compound to its analog, isomer, metabolite, deriva He2-propanol tive, pharmaceutically acceptable salt, pharmaceutical prod HO F uct, polymorph, crystal, impurity, N-oxide, hydrate or any NC C combination thereof. In another embodiment, this invention provides a large scale process for the preparation of compound of formula III, FC F e wherein the process comprises the same steps as described Ye herein above, wherein compound of formula 12 is prepared 25 HC OH according to the following scheme, in the presence of 4N S-II NaOH: In another embodiment, the present invention provides a C 30 process for preparing an (S) enantiomer of SARM compound cott NNaOH represented by the structure of formula S-II: O -- Cx 2" -10 c. his N H MtBE H S-II

O NC C

N H NBSDMF O -e- RT O FC NH e e 40 CH, OH

said process comprising the steps of FIGS. 1K and 1L provide one embodiment of a large scale a) coupling an amine of formula 17: process for the preparation of a large scale synthesis of com 45 pounds of formulas S-II. In one embodiment, the present invention provides a pro 17 cess for preparing a compound of formula III wherein is X is NH2 O. In another embodiment, the present invention provides a 50 process for preparing a compound of formula III wherein T is OH. In another embodiment, the present invention provides a NC process for preparing a compound of formula III wherein is CF R is CH. In another embodiment, the present invention provides a process for preparing a compound of formula III 55 wherein Z is CN, and/or Cland/or F. In another embodiment, with the carboxylic acid of formula R-18 the present invention provides a process for preparing a com pound of formula III, wherein Z is CN. In another embodi ment, the present invention provides a process for preparing a R-18 compound of formula III whereinY is CF and/or CH, and/or 60 H and/or Cl. In another embodiment, the present invention provides a process for preparing a compound of formula III HO Br wherein R is H, and/or CN, and/or C1 and/or F. In another Hd oH embodiment, the present invention provides a process for preparing a compound of formula III wherein Q is CN. In 65 another embodiment, the present invention provides a process in the presence of a coupling reagent, to produce an amide for preparing a compound of formula III wherein Q is F. In of formula R-19 US 8,309,603 B2 17 18 with the carboxylic acid of formula 5-18 R-19 NC S-18

FC C, OH HO Br CH, and 10 b) reacting the amide of formula R-19 with a compound of in the presence of a coupling reagent, to produce an amide formula 20: of formula 5-19

15

2O S-19 NC C

HO F FC to produce a compound of formula S-II. 25 In one embodiment, whereby compound R-18 of step (a) is and reacted with a coupling agent prior to addition of compound b) reacting the amide of formula 5-19 with a compound of of formula 17. formula 20 FIG. 1A and Example 1 provide one embodiment of a process for the preparation of a compound of formula S-II. 30 In another embodiment, the conditions of step (b) of the process outlined hereinabove may comprise potassium car bonate, Sodium carbonate, or cesium carbonate, or another base appropriate for this reaction, using 2-propanol, THF or methylethylketone as a solvent, optionally with a transition 35 catalyst, BTBAC (benzyltributylammonium chloride) or other Suitable agent. HO F In another embodiment, the present invention provides a process for preparing an (R) enantiomer of SARM compound 40 to produce a compound of R-II. represented by the structure of formula R-II: In one embodiment, whereby compound 5-18 of step (a) is reacted with a coupling agent prior to addition of compound of formula 17.

45 FIG. 1B depicts one embodiment of such a process for the R-II preparation of compound of formula R-II. NC Cl; In another embodiment, the conditions of step (b) of the O process outlined hereinabove may comprise potassium car bonate, Sodium carbonate, or cesium carbonate, or another base appropriate for this reaction, using 2-propanol, THF or FC NH O F methylethylketone as a solvent, optionally with a transition H6 aH, catalyst, BTBAC (benzyltributylammonium chloride) or other Suitable agent. In another embodiment, the present invention provides a said process comprising the steps of 55 process for preparing an (S) enantiomer of a SARM com a) coupling an amine of formula 17: pound represented by the structure of formula S-II

17 S-II NH2 60 NC C

NC FC

CF 65 US 8,309,603 B2 19 20 said process comprising the steps of a) coupling an amine of formula 17: R-II NC C O 17 5 NH2 FC NH a- O F H6 H, NC 10 CF said process comprising the steps of a) coupling an amine of formula 17: with the carboxylic acid of formula R-18 17 15 NH2 R-18 O

NC HO Br 2. 2O CF OH

in the presence of a coupling reagent, to produce an amide with the carboxylic acid of formula S-18 of formula R-19 25 S-18

R-19 NC HO Br 30 H6 ah, FC in the presence of a coupling reagent, to produce an amide of formula S-19 b) reacting the amide of formula R-19, with a base to forman 35 oxirane S-21 S-19 NC

S-21

NC 40 FC NH Br; H6 aH, FC b) reacting the amide of formula S-19, with a base to forman 45 oxirane R-21 and c) reacting the oxirane of formula S-21 with a compound of R-21 formula 20: NC 50

2O FC Ol syO C

55 HO F and c) reacting the oxirane of formula R-21 with a compound of to produce a compound of S-II. formula 20: In one embodiment, whereby compound R-18 of step (a) is 60 reacted with a coupling agent prior addition of compound of formula 17. FIG. 1C depicts an embodiment of such a process for the preparation of compound of formula S-II. In another embodiment, the present invention provides a 65 HO F process for preparing an (R) enantiomer of SARM compound represented by the structure of formula R-II: to produce a compound of R-II. US 8,309,603 B2 21 22 In one embodiment, whereby compound 5-18 of step (a) is reacted with a coupling agent prior to addition of compound S-24 of formula 17. Cl; FIG. 1D preparation of compound of formula R-II. In another embodiment, the present invention provides a 5 process for preparing an (S) enantiomer of a SARM com pound represented by the structure of formula S-II.

10 and coupling the carboxylic acid of compound of formula S-24 S-II with the amine of formula 17 NC C O 15 17 NH2 FC NH O F

CH, OH NC 2O said process comprising the steps of CF a) reacting a ring of formula S-22 to produce the compound of formula S-II. 25 FIG. 1E depicts an embodiment of such a process for the preparation of compound of formula S-II. In another embodiment, the present invention provides a S-22 process for preparing an (R) enantiomer of a SARM com H O pound represented by the structure of formula R-II: N wO O & Br

HC 35 R-II NC C with a compound of 20

FC OuNH s O F 40 2. HO CH,

2O said process comprising the steps of 45 a) reacting a ring of formula R-22 HO .F R-22 to produce a compound of formula R-23: H 50 O N R-23 O H O O -Br N 55 H3C O O w with a compound of 20

60 2O C Cl;

HO F 65 a) ring opening of compound of formula R-23 to produce a compound of formula S-24 to produce a compound of formula S-23; US 8,309,603 B2 23 24 said process comprising the steps of S-23 a) reacting the carboxylic acid of formula R-18

5 R-18

HO Br 10 HC OH

with tribromoacetaldehyde to produce a compound of for Cl mula R-25: F 15 b) ring opening of compound of formula S-23 to produce a compound of formula R-24 R-25 H CBr

O X O

Cl; R-24 25 C5’27, H3C Br

F reacting the dioxalane derivative R-25 with a compound of formula 20 30

and 2O coupling the carboxylic acid of compound of formula R-24 C with the amine of formula 17 35 HO F

to produce a compound of formula R-26; 17 40 NH2

NC R-26 H CBr CF 45 O O to produce the compound of formula R-II. y \ FIG. 1F depicts an embodiment of such a process for the so O CH, O Cl; preparation of compound of formula R-II. In another embodiment, the present invention provides a process for preparing an (S) enantiomer of a SARM com pound represented by the structure of formula S-II 55 b) ring opening of compound of formula R-26 to produce a compound of formula S-24 S-II

NC C S-24 60 Cl;

65 US 8,309,603 B2 25 26 and S-26 coupling the carboxylic acid of compound of formula S-24 H CBr with the amine of formula 17: 5 -X, 17 NH2 O Cl;

NC 10 F CF ring opening of compound of formula S-26 to produce a to produce the compound of formula S-II. 15 compound of formula R-24 FIG. 1G depicts an embodiment of such a process for the preparation of compound of formula S-II. R-24

In another embodiment, the present invention provides a Cl; process for preparing an (R) enantiomer of a SARM com pound represented by the structure of formula R-II 2O F

R-II H6, CH, NC C 25 O and coupling the carboxylic acid of compound of formula R-24 FC NH O F with the amine of formula 17: H6 aH, 30 17 said process comprising the steps of: NH2 a) reacting the carboxylic acid of formula S-18 35 NC S-18 CF

HO Br to produce the compound of formula R-II. 40 FIG. 1H depicts an embodiment of such a process for the CH, preparation of compound of formula R-II. In another embodiment, the present invention provides a with tribromoacetaldehyde to produce a compound of for process for preparing a racemic mixture of a SARM com mula S-25: pound represented by the structure of formula II 45

II S-25 NC C H CBr O 50 O O FC NH O F CH, OH Br 55 said process comprising the steps of reacting the dioxalane derivative S-25 with a compound of a) reacting a compound of formula 24 formula 20

24 60 C O OC HO F -X. F 65 CH, OH to produce a compound of formula S-26; US 8,309,603 B2 27 28 with a compound of formula 27 17 NC NH2 27

FC

NC to produce the amide of formula 29, CF 10 29 wherein P is selected from isocyanate (NCO) or isothiocyan NC ate (NCS) to produce a compound of formula 28a or 28b. respectively 15 FC Ol s O 28a. CH3 NC O c) oxidizing an amide of formula 29, to produce the oxirane of formula 21: CH3 FC O Cl; or 21 O O NC F 25 28b Ol O NC FC st O; O CH CH3 30 FC O C and d) reacting the oxirane of formula 21 with a compound of S O formula 20: F 35 b) ring opening of the oxazolidinedione or 2-thioXooxazolid 4-one ring of formula 28a or 28b in a presence of a base to produce a compound of formula II. FIG. 1I depicts an embodiment of such a process for the 40 HO F preparation of racemic compound of formula II. In another embodiment, the present invention provides a to produce the compound. process for preparing a racemic mixture of a SARM com In another embodiment, the oxidizing an amide of formula pound represented by the structure of to formula II: 29 of step (c) to comprises oZone. In another embodiment, the 45 oxidizing agent is a peroxyacid, for example, peracetic acid, II (CHCOOOH). In another embodiment, the oxidizing agent NC C meta-chloroperbenzoic acid (m-CPBA). In another embodi ment, the oxidizing agent is magnesium monoperoxypthalic 50 acid (MMPP). In another embodiment, the oxidizing agent is Ol O O hydrogen peroxide together with catalytic amounts (1.0-0.1 FC six F mol %) of manganese(2) salts. CH, OH FIG. 1J depicts an embodiment of a process for the prepa ration of racemic compound of formula II. said process comprising the steps of 55 In one embodiment, this invention provides a process for a) chlorinating methacrylic acid: preparing pure enantiomers of SARMs compounds of this invention, comprising the steps of a) preparing a racemic SARM compound of this invention; and b) separating pure O O SARM compound of this invention from its racemic mixture. 60 In one embodiment, separation of the optically-active (R) isomer or (S) enantiomer, from the racemic SARM com HO1. C -- pounds of this invention comprises crystallization tech CH3 CH3 niques. In another embodiment, the crystallization tech niques include differential crystallization of enantiomers. In 65 another embodiment, the crystallization techniques include b) coupling an 3-cyano 4-trifluoromethyl aniline of formula differential crystallization of diastereomeric salts (tartaric 17 with methacryloyl chloride: salts or quinine salts). In another embodiment, the crystalli US 8,309,603 B2 29 30 zation techniques include differential crystallization of chiral example p-toluoylsulfonate (tosylate), benzenesulphonate auxiliary derivatives (mentholesters, etc). In another embodi which may be unsubstituted or substituted by methyl, chlo ment, separation of the optically-active (R) isomer or (S) rine, bromine, nitro and the like: NONO or sulfate, sulfite, enantiomer, from the racemic SARM compounds of this phosphate, phosphite, carboxylate, imino ester, N or car invention comprises reacting the racemate mixture with bamate. another chiral group, forming of a diastereomeric mixture According to this aspect of the invention and in one followed by separation of the diastereomers and removing the embodiment, the reaction is carried out in a suitable inert additional chiral group to obtain pure enantiomers. In another Solvent or diluent such as, for example, tetrahydrofuran, embodiment, separation of the optically-active (R) isomer or diethyl ether, acetone, methyl ethyl ketone, 2-propanol, aro (S) enantiomer, from the racemic SARM compounds of this 10 matic amines such as pyridine; aliphatic and aromatic hydro invention comprises chiral synthesis. In another embodiment, carbons such as benzene, toluene, and Xylene; dimethylsul separation of the optically-active (R) isomer or (S) enanti foxide (DMSO), dimethylformamide (DMF), and omer, from the racemic SARM compounds of this invention dimethylacetamide (DMAC). In one embodiment, the reac comprises biological resolution. In another embodiment, tion may be carried out in a suitable inert solvent or diluent as separation of the optically-active (R) isomer or (S) enanti 15 described hereinabove, suitably in the presence of a base such omer, from the racemic SARM compounds of this invention as triethylamine, and at a temperature in the range, as comprises enzymatic resolution. In another embodiment, described above. In one embodiment, the reaction may be separation of the optically-active (R) isomer or (S) enanti carried out at an appropriate temperature, as will be known to omer, from the racemic SARM compounds of this invention one skilled in the art, for example, in the range, of -20 to 120° comprises chromatographic separation using a chiral station C., or for example at or near ambient temperature. ary phase. In another embodiment, separation of the opti The coupling reagent defined hereinabove is a reagent cally-active (R) isomer or (S) enantiomer, from the racemic capable of turning the carboxylic acid/thiocarboxylic acid of SARM compounds of this invention comprises affinity chro formula 24 or 18 into a reactive derivative thereof, thus matography. In another embodiment, separation of the opti enabling coupling with the respective amine to form an cally-active (R) isomer is or (S) enantiomer, from the racemic 25 amide/thioamide bond. A suitable reactive derivative of a SARM compounds of this invention comprises capillary elec carboxylic acid/thiocarboxylic acid is, for example, an acyl trophoresis. In another embodiment, separation of the opti halide/thioacyl halide, for example an acyl/thioacyl chloride cally-active (R) isomer or (S) enantiomer, from the racemic formed by the reaction of the acid/thioacid and an inorganic SARM compounds of this invention comprises forming an acid chloride, for example thionyl chloride; a mixed anhy ester group of the hydroxyl group of the chiral carbon with an 30 dride, for example an anhydride formed by the reaction of the optically-active acid, for example (-)-camphanic acid, sepa acid and a chloroformate Such as isobutyl chloroformate; an rating the diastereomers esters, thus obtained, by fractional active ester/thioester, for example an ester formed by the crystallization or preferably, by flash-chromatography, and reaction of the acid and a phenol Such as pentafluorophenol, then hydrolyzing each separate ester to the alcohol. an ester Such as pentafluorophenyl trifluoroacetate oran alco In another embodiment, the purity, and selectivity of an 35 hol Such as methanol, ethanol, isopropanol, butanol or N-hy enantiomer obtained by the process of this invention, or by droxybenzotriazole; an acyl/thioacyl azide, for example an chiral separation of a racemic mixture of this invention can be azide formed by the reaction of the acid/thioacid and azide determined by HPLC analysis. Such as diphenylphosphoryl azide; an acyl cyanide/thioacyl In another embodiment, the process further comprises the cyanide, for example a cyanide formed by the reaction of an step of converting the SARM compound to its analog, isomer, 40 acid and a cyanide Such as diethylphosphorylcyanide; or the metabolite, derivative, pharmaceutically acceptable salt, product of the reaction of the acid/thioacid and a carbodiim pharmaceutical product, N-oxide, hydrate or any combina ide such as dicyclohexylcarbodiimide. tion thereof. It is to be understood that the process may comprise any According to this aspect of the invention, and in one embodiment described herein, as will be appropriate to pro embodiment, the reagent used for reacting the amide deriva 45 duce a SARM of a corresponding formula, as will be appre tive, for example compound of formula 19 and the phenol ciated by one skilled in the art. derivative such as for example 20 are carried out in the pres In one embodiment, the process for preparing a SARM of ence of a base. Any suitable base that will deprotonate the this invention may involvering opening in the presence of less hydrogen of the —XH moiety (for example, a phenol moiety acidic conditions, which in another embodiment, diminish when X is O) and allow the coupling may be used. Nonlim 50 the likelihood of obtaining SARM compound mixtures, and iting examples of bases are carbonates Such as alkali carbon provide higher yield and purity of a SARM of interest. In one ates, for example Sodium carbonate (Na2CO), potassium embodiment, the ring opening of a process as described carbonate (KCOs) and cesium carbonate (CSCOs); bicar herein, to produce a carboxylic acid of formula 13, is carried bonates Such as alkali metal bicarbonates, for example out in the presence of HBr, which, in one embodiment, is at a sodium bicarbonate (NaHCO), potassium bicarbonate 55 concentration of up to 30%, or in another embodiment, of up (KHCO), alkali metal hydrides such as sodium hydride to 40%, or in another embodiment, is of up to 25%, or in (NaH), potassium hydride (KH) and lithium hydride (LiH), another embodiment, of up to 23%, or in another embodi and the like. ment, of up to between 20-25%. In one embodiment, the The leaving group L. according to this aspect, and in one SARMs of this invention may be produced via large-scale embodiment, may comprise any removable group customar 60 synthesis, providing highly pure products in high yields. ily considered for chemical reactions, as will be known to the In one embodiment, the reaction may be carried out in a person skilled in the art. Suitable leaving groups are halogens, suitable inert solvent or diluent as described hereinabove, for example F, Cl, Brand I: alkylsulfonate esters (—OSOR) Suitably in the presence of a base such as triethylamine, and at wherein R is an alkyl group, for example methanesulfonate a temperature in the range, as described above. (mesylate), trifluoromethanesulfonate, ethanesulfonate, 2.2, 65 In some embodiments the compounds for use in the meth 2-trifluoroethanesulfonate, perfluoro butanesulfonate: aryl ods of this invention are nonsteroidal ligands for the androgen sulfonate esters ( OSOAr) wherein Aris an aryl group, for receptor and may demonstrate tissue-selective androgenic US 8,309,603 B2 31 32 and/or anabolic activity. These novel agents are useful in embodiment, is an androgen receptor, or in another embodi affecting the carcass composition, increasing the lean mass ment, is an , or in another embodiment, is a and/or reducing the fat mass of an animal, reducing percent progesterone receptor, or in another embodiment, is a gluco fat mass, increasing feed efficiency, increasing average daily corticoid receptor. In some embodiments, the multitude of gain (ADG), decreasing feed to gain ratio (F:G), increasing effects may occur simultaneously, as a function of binding to muscle growth, modulation of meat quality, and/or enhancing multiple receptors in the Subject. In some embodiments, the productive life of animals, including feedlot animals, beef tissue selective effects of the compounds as described herein cattle and finishing livestock. These novel agents are useful in provide for simulataneous action on different target organs. males for the treatment of a variety of hormone-related con Compositions and Methods of Use ditions such as sexual dysfunction, decreased sexual libido, 10 In some embodiments, this invention provides methods of erectile dysfunction, hypogonadism, sarcopenia, osteopenia, use which is comprise administering a composition compris osteoporosis, alterations in cognition and mood, depression, ing the described compounds. In one embodiment, a compo anemia, hair loss, obesity, benign prostate hyperplasia and/or sition is a pharmaceutical composition. In one embodiment, a prostate cancer. Further, the compounds are useful as adjunct composition is a feed composition. In one embodiment, feed to androgen-deprivation therapy (ADT) for treating prostate 15 composition may be a pharmaceutical composition. cancer. Further, the compounds are useful for oral testoster As used herein, pharmaceutical composition” means a one replacement therapy, and treating prostate cancer. In “therapeutically effective amount of the active ingredient, other embodiments, the compounds are useful for the treat i.e. the compound of formula III, together with a pharmaceu ment of a variety of hormone-related conditions in females tically acceptable carrier or diluent. A “feed composition including, sexual dysfunction, decreased sexual libido, means an “effective amount. A “therapeutically effective hypogonadism, sarcopenia, osteopenia, osteoporosis, alter amount” and/or an “effective amount’ as used herein, refers ations in cognition and mood, depression, anemia, hair loss, to that amount which provides atherapeutic effect or effect on obesity, endometriosis, infertility, breast cancer, uterine can the carcass of the animal for a given condition and adminis cer and ovarian cancer. In other embodiments, the SARMs are tration regimen. useful for treating, Suppressing, inhibiting or reducing the 25 In one embodiment, the present invention encompasses incidence of diabetes type II, diabetes type I, glucose intol incorporating the compounds into animal feed. In one erance, hyperinsulinemia, insulin resistance, dyslipidemia, embodiment, the present invention encompasses incorporat hypercholesterolemia, high blood pressure, obesity, fatty ing the compounds into a feed composition. In some embodi liver conditions, diabetic nephropathy, diabetic neuropathy, ments, the compounds/compositions of this invention may be diabetic retinopathy, cardiovascular disease, atherosclerosis, 30 administered to any animal as herein described, for example cerebrovascular conditions and stroke. to finishing livestock. Such administration, in Some embodi In some embodiments, the compounds as described herein ments, is accomplished via, inter alia, supplementation in are useful in preventing and treating muscle wasting disor feeds, feed compositions, formulation into feeds, controlled ders, bone related disorders, and diabetes is related disorders. release implants, topical sprays or creams/ointments, disso In some embodiments, the compounds as described herein 35 lution in drinking water, rumen-stable formulations to are useful, either alone or as a combination with beta-agonists include coatings and derivatives, repeated injection, and other as feed composition, pharmaceutical compositions or as means as will be known to the skilled artisan. In one embodi methods for affecting the carcass composition, increasing the ment, the present invention encompasses incorporating the lean mass, reducing the fat mass and/or reducing the percent compounds into other typical pharmaceutical administration fat mass, increasing feed efficiency, increasing average daily 40 routes and pharmaceutical compositions as described herein. gain (ADG), decreasing feed to gain ratio (F:G) of an animal. As used herein, the term "administering refers to bringing In some embodiment, the compounds as described herein are a Subject in contact with a compound of the present invention. useful, either alone or as a combination with beta-agonists as As used herein, administration can be accomplished in vitro, feed composition or as methods for increasing the muscle i.e. in a test tube, or in Vivo, i.e. in cells or tissues of living growth of an animal, decreasing time to market (or time to 45 organisms, for example humans and/or animals. In one slaughter), increasing carcass weight (or slaughter weight) of embodiment, the present invention encompasses administer a feedlot or finishing animal, modulation of meat quality, ing the compounds of the present invention to a subject. enhancing productive life of and/or improving herd health of In one embodiment, the present invention encompasses animals, including feedlot animals, beef cattle and finishing administering the compounds of the present invention via livestock. 50 implants. In one embodiment, administering the compounds In some embodiments, the compounds as described herein of the present invention is via controlled release implants. In are useful, either alone or as a composition, in males and another embodiment of the present invention, administering females for the treatment of a variety of hormone-related the compounds of the present invention is via topical admin conditions, such as hypogonadism, sarcopenia, erectile dys istration. In one embodiment, topical administration is via a function, lack of libido, osteoporosis and fertility. In some 55 topical spray. In one embodiment, topical administration is embodiments, the compounds as described herein are useful via a cream. In one embodiment, topical administration is via in stimulating or promoting or restoring function to various an ointment. In one embodiment, compounds and/or compo processes, which in turn result in the treatment of the condi sitions of this invention are administered via an implant to a tions as herein described, including, inter aila, promoting pig. In one embodiment, compounds and/or compositions of erythropoiesis, osteogenesis, muscle growth, glucose uptake, 60 this invention are administered via topical administration to a insulin secretion, and/or preventing lipidogenesis, clotting, pig. insulin resistance, atherosclerosis, osteoclast activity, and In one embodiment, this invention is directed to a feed others. composition for an animal comprising a compound of this In one embodiment, the methods of this invention make use invention. In one embodiment, this invention is directed to a of the described compound contacting or binding a receptor, 65 feed composition for an animal comprising a compound of and thereby mediating the described effects. In some embodi formula IIIA or its isomer, pharmaceutically acceptable salt, ments, the receptor is a nuclear receptor, which in one crystal, N-oxide, hydrate or any combination thereof. In one US 8,309,603 B2 33 34 embodiment, this invention is directed to a feed composition Zilpaterol hydrochloride (sold under the tradename of Zilmax for an animal comprising a compound of formula I or its available from Invervet of Millsboro, Del.). Other active iso isomer, pharmaceutically acceptable salt, crystal, N-oxide, mers of other drugs with beta-adrenergicagonistic properties, hydrate or any combination thereof. In one embodiment this include for example hexoprenaline, isoprenaline, riniterol, invention is directed to a feed composition for an animal 5 isoetharine, metaproterenol, reproterenol, cimaterol, pro comprising a compound of formula II or its isomer, pharma caterol, carbuterol, tulobuterol, pibuterol, mabuterol, ceutically acceptable salt, crystal, N-oxide, hydrate or any bitolterol, clenbuterol, and bambuterol. Also included may be combination thereof. In one embodiment this invention is tautomers of beta-agonists that are under development, Such directed to a feed composition for an animal comprising a as broXaterol, etanterol, imoxiterol, namiterol, picumeterol, compound of formula XXIII or its isomer, pharmaceutically 10 acceptable salt, crystal, N-oxide, hydrate or any combination RP58802, RU 42173 and ZK 90055. Those skilled in the art thereof. In one embodiment this invention is directed to a feed will also realize that there are many pharmaceutically accept composition for an animal comprising a compound of for able salt forms of these drugs, such as for example Sulfate, mula XXIV or its isomer, pharmaceutically acceptable salt, fumarate, hydrobromide, dihydrochloride, methanesulpho crystal, N-oxide, hydrate or any combination thereof. In one 15 nate, hydroxynaphthoate, hydrochloride or where appropri embodiment this invention is directed to a feed composition ate, one or other of the hydrate forms thereof. for an animal comprising a compound of formula XXV or its In one embodiment, the feed composition of this invention isomer, pharmaceutically acceptable salt, crystal, N-oxide, is prepared as a dry powder or a granulate and added to the hydrate or any combination thereof. animal feed, Such as by mixing. Also, other forms of the The feed composition containing the compounds of this additive may also be appropriate. The additive can be pre invention can be administered as additives to the animal feed. mixed into the feed according to any of the methods known to In one embodiment, the animal feed including the feed com those skilled in the art, or may be mixed or blended into the position of this invention is provided to the animal once a day. feed at the time of feeding. In another embodiment twice a day. In another embodiment In one embodiment, this invention is directed to a pharma once to five times a day. 25 ceutical composition for an animal comprising a compound In another embodiment, the feed composition comprises of this invention. In one embodiment, this invention is between 0.010-50 ppm of a compound of this invention. In directed to a pharmaceutical composition for an animal com another embodiment, the feed composition comprises 0.01-1 prising a compound of formula IIIA or its isomer, pharma ppm of a compound of this invention. In another embodiment, ceutically acceptable salt, crystal, N-oxide, hydrate or any the feed composition comprises 0.10 ppm of a compound of 30 combination thereof. In one embodiment, this invention is this invention. In another embodiment, the feed composition directed to a pharmaceutical composition for an animal com comprises 1 ppm of a compound of this invention. In another prising a compound of formula I or its isomer, pharmaceuti embodiment, the feed composition comprises 3 ppm of a cally acceptable salt, crystal, N-oxide, hydrate or any combi compound of this invention. In another embodiment, the feed nation thereof. In one embodiment this invention is directed composition comprises 10 ppm of a compound of this inven 35 to a pharmaceutical composition for an animal comprising a tion. In another embodiment, the feed composition comprises compound of formula II or its isomer, pharmaceutically 30 ppm of a compound of this invention. acceptable salt, crystal, N-oxide, hydrate or any combination In one embodiment, the animal is fed with the feed com thereof. In one embodiment this invention is directed to a position of this invention after it has reached 60 pounds. In pharmaceutical composition for an animal comprising a com one embodiment, the animal is fed with the feed composition 40 pound of formula XXIII or its isomer, pharmaceutically of this invention after it has reached 50 pounds. In one acceptable salt, crystal, N-oxide, hydrate or any combination embodiment, the animal is fed with the feed composition of thereof. In one embodiment this invention is directed to a this invention before it has reached 50 pounds. pharmaceutical composition for an animal comprising a com In one embodiment, the animal is fed with the feed com pound of formula XXIV or its isomer, pharmaceutically position of this invention for about ten weeks prior to slaugh 45 acceptable salt, crystal, N-oxide, hydrate or any combination ter. In one embodiment, the animal is fed with the feed com thereof. In one embodiment this invention is directed to a position of this invention for about twenty weeks prior to pharmaceutical composition for an animal comprising a com slaughter. In one embodiment, the animal is fed with the feed pound of formula XXV or its isomer, pharmaceutically composition of this invention for about a year prior to slaugh acceptable salt, crystal, N-oxide, hydrate or any combination ter. 50 thereof. In another embodiment, the feed composition of this inven The pharmaceutical composition containing the com tion comprises a combination of a compound of this invention pounds of this invention can be administered as additives to and a beta-agonist. In another embodiment, the feed compo the animal feed. In one embodiment, the animal feed includ sition comprises a compound of formula II and a beta-agonist. ing the pharmaceutical composition of this invention is pro In another embodiment, the feed composition comprises a 55 vided to the animal once a day. In another embodiment twice compound of formula XXIII and a beta-agonist. In another a day. In another embodiment once to five times a day. embodiment, the feed composition comprises a compound of In another embodiment, the pharmaceutical composition formula XXIV and a beta-agonist. In another embodiment, comprises between 0.010-50 ppm of a compound of this the feed composition comprises a compound of formula XXV invention. In another embodiment, the pharmaceutical com and a beta-agonist. 60 position comprises 0.01-1 ppm of a compound of this inven In one embodiment, the animal is raised with a beta-agonist tion. In another embodiment, the pharmaceutical composi enhanced diet during a first time period of time and later fed tion comprises 0.10 ppm of a compound of this invention. In in a diet with Substantially no beta-agonist, but including a another embodiment, the pharmaceutical composition com compound of this invention during a second period of time. prises 1 ppm of a compound of this invention. In another In one embodiment a beta-agonist includes ractopamine 65 embodiment, the pharmaceutical composition comprises 3 hydrochloride (sold under the tradenames Optaflexx or Pay ppm of a compound of this invention. In another embodiment, lean, e.g. and available from Elanco of Greenfield, Ind.) and the pharmaceutical composition comprises 10 ppm of a com US 8,309,603 B2 35 36 pound of this invention. In another embodiment, the pharma vae), primates (gorilla, monkey), rodents (rat, mouse, Squir ceutical composition comprises 30 ppm of a compound of rel, beaver), cetaceans (whale, dolphin), pinnipeds (walrus, this invention. seal), miscellaneous (bear, raccoon, elephant) or others as In one embodiment, the pharmaceutical composition of will be appreciated by one skilled in the art. this invention is in an amount from about 0.0005% to about In some embodiments, the term “finishing livestock” refers 0.1% of the weight of the animal. In another embodiment, the to, interalia, any animal that is normally fattened for the last pharmaceutical composition of this invention is in an amount few months before processing. In one embodiment, finishing from about 0.005% to about 0.01% of the weight of the livestock is a beef cattle. In one embodiment, finishing live animal. In another embodiment, the pharmaceutical compo stock is a pig. In one embodiment, finishing livestock is a sition of this invention is in an amount from about 0.01% to 10 poultry. In one embodiment, finishing livestock is a farmed about 0.05%. fish. In one embodiment, the methods of this invention are used In one embodiment, the compounds, compositions or in a subject, which is a human. In another embodiment, the methods of use thereofmay find application in increasing the Subject is a mammal. In another embodiment, the Subject is an yield of all retail products derived from such feedlot animals. animal. In another embodiment the subject is an invertebrate. 15 For instance, each of the above food animals have different In another embodiment the subject is a vertebrate. In another types of tissues and preparations thereof Such as for Swine: embodiment, the animal is a feedlot animal. In another ham, bacon, sausage, pork bellies, pork chop, ribs, brain, embodiment, the animal is a beef cattle. In another embodi chitterling, tripe, tenderloin, etc. ment, the animal is a finishing livestock. Feedlot practices often include castration in order to better For administration to mammals, and particularly humans, control the behavior of feedlot animals and to improve the it is expected that the physician will determine the actual quality of the meat (more tender, marbled, and colored). This dosage and duration of treatment, which will be most suitable occurs with a loss of productivity which could be offset using for an individual and can vary with the age, weight and nonsteroidal androgens, representing one embodiment of a response of the particular individual. mechanism whereby the compounds and composition find For administration to mammals, in Some embodiments, the 25 application therein. present invention provides compounds, compositions and In some embodiments, enhancing measures of productiv methods of use thereof for the enhanced meat productivity in ity in feedlot animals may comprise enhancing the number of food animals. In some embodiments, this invention provides animals per litter, litters per breeding animal per year, slaugh compounds, compositions and methods of use thereof for the ter head count per breeding animal per year, meat product modulation of appetite for feedlot animals. In some embodi 30 production (in pounds) per breeding animal per year, average ments, this invention provides compounds, compositions and daily growth in pounds, live weight (in pounds), dressing methods of use thereof for improved feed efficiency. percent (% of live weight), dressed weight in pounds, retail For administration to mammals, in some embodiments, meat in pounds perhead count, retail meat yield (percent of this invention provides compounds, compositions and meth live weight), or any combination thereof. ods of use thereof for decreased time to market for feedlot 35 In one embodiment, the compounds, compositions or animals. In some embodiments, this invention provides com methods of use thereof may find application in Stud farm pounds, compositions and methods of use thereof for productivity. Androgens (steroidal and nonsteroidal) are increased terminal weight of feedlot animals. In some known to enhance sex drive in males and females such that, in embodiments, this invention provides compounds, composi Some embodiments, the stud animals are productive in terms tions and methods of use thereof for decreased time to termi 40 of “open mating time or births per mating event. In some nal weight of feedlot animals. In some embodiments, this embodiments, the Support of sex organs and accessory tissues invention provides compounds, compositions and methods of (and health benefits) of the compounds/compositions of this use thereof for increased lean weight of feedlot animals. In invention may increase productive life of a stud animal, Some embodiments, this invention provides compounds, allowing him to 'stand at Stud' (i.e. meaning available for compositions and methods of use thereof for decreased body 45 reproduction) for a longer period of time. Female receptivity fat weight of feedlot animals. In some embodiments, this is enhanced, in Some embodiments, in terms of frequency, in invention provides compounds, compositions and methods of response to contact with/administration of a compound/com use thereof for decreased percent body fat weight of feedlot position of this invention. animals. In some embodiments, this invention provides com In some embodiments, this invention comprises applica pounds, compositions and methods of use thereof for the 50 tion of any method as herein described for veterinary use, in modulation of meat quality in feedlot animals. In some any animal as described herein. In some embodiments, treat embodiments, this invention provides compounds, composi ment of Such conditions or diseases in animals may find tions and methods of use thereof for increased is meat pro application for pleasure and/or profit animals, may increase duction. the size of game animals by Supplementation, etc. as will be In some embodiments, the term “feedlot animals' refers to, 55 appreciated by one skilled in the art. interalia, any animal the meat of which is considered edible In some embodiments, the compounds/compositions may in a given culture or country. In some embodiments, such term be administered to any animal as herein described, for may include without limitation Swine (domestic pig, wild example to livestock. Such administration, in Some embodi boars), bovine (bison, cattle, yaks), cervids (deer, elk, ments, is accomplished via, inter alia, Supplementation in moose), Ovine (sheep/lamb), caprine (goats), lagomorphs 60 feeds, formulation into feeds, controlled release implants, (rabbit, pika), avian (chicken, turkey, duck, game birds, emu/ dissolution in drinking water, rumen-stable formulations to ostrich), fish (catfish, tilapia, Salmon, red drum), shellfish include coatings and derivatives, repeated injection, and other (crustaceans such as crab, lobster, shrimp; and mollusks Such means as will be known to the skilled artisan. as clams, octopus, Squid), roe (caviar), amphibians (frogs, In some embodiments, dosages as described herein for salamanders), reptiles (Snakes, turtle, alligator), canids (dog, 65 humans will be adjusted to accommodate the varying size of foX), felines (cat), equines (horse, donkey, Zebras), marsupi animals. Such modification of dosage is well known in the als (kangaroo, opossum), insects (grasshopper, beetles, lar field of veterinary art, and is available in common veterinary US 8,309,603 B2 37 38 manuals, and may vary on a scale ranging from milligrams to comprising administering a compound of formula XXIII its grams as a function of Such varying size. isomer, pharmaceutically acceptable salt, pharmaceutical In some embodiments, the compounds/compositions may product, crystal, N-oxide, hydrate or any combination be administered to any animal as herein described, in combi thereof. In one embodiment, this invention is directed to a nation with any other agent as described herein, befitting the method of affecting the carcass composition of an animal particular animal and condition in the animal, which is being comprising administering a compound of formula XXIV or treated. In some embodiments, such combination therapy its isomer, pharmaceutically acceptable salt, pharmaceutical may comprise administration of the compounds/composi product, crystal, N-oxide, hydrate or any combination tions with high fat diets such as Supplemented with fatty acids thereof. In one embodiment, this invention is directed to a or oils to improve the meat quality; various combinations 10 method of affecting the carcass composition of an animal with androgens, progestins, anti-glucocorticoids, estrogens, comprising administering a compound of formula XXV or its growth hormone, etc. can be tailored to produce maximum isomer, pharmaceutically acceptable salt, pharmaceutical weight gain performance in different types of animals (cows product, crystal, N-oxide, hydrate or any combination vs. pigs; intact vs. castrated) the specifics of which are known thereof. In another embodiment, the carcass composition is by those skilled in the art (see for example, Environ Qual Saf 15 affected by increasing the lean mass, reducing the fat mass, or Suppl. 1976; (5):89-98). reducing percent fat mass. In another embodiment, the car In some embodiments, the compounds/compositions may cass composition comprises increasing the growth perfor be administered to any animal as herein described, which is a mance in said animal. food source for humans, and in Some embodiments, the tis In one embodiment, this invention is directed to a method sue-selectivity and shorter half-lives of the compounds as of increasinglean mass of an animal comprising administer herein described significantly lowers anticipated environ ing a compound of this invention. In one embodiment, this mental effects. In some embodiments, the risk to human invention is directed to a method of increasinglean mass of an consumption thereby, as compared to agricultural use of Ste animal comprising administering a compound of formula roidal androgens such as trenbolone acetate whose half-life is IIIA or its isomer, pharmaceutically acceptable salt, pharma 3 days, is much reduced, and comprises therefore an embodi 25 ceutical product, crystal, N-oxide, hydrate or any combina ment of an advantage of the compounds of this invention. tion thereof. In one embodiment, this invention is directed to In some embodiments, an advantage of the compounds/ a method of increasing lean mass of an animal comprising compositions of this invention may comprise the anabolic administering a compound of formula I or its isomer, phar activity of the compound thereby producing larger animals maceutically acceptable salt, pharmaceutical product, crys and affecting carcass composition in less time. Factors con 30 tal, N-oxide, hydrate or any combination thereof. In one tributing to the increasing productivity may include, in some embodiment, this invention is directed to a method of increas embodiments, enhanced mineral (and other nutrient) absorp ing lean mass of an animal comprising administering a com tion in the gut, enhanced body protein accretion and metabo pound of formula II or its isomer, pharmaceutically accept lism of fat stores resulting in increased lean growth rates; able salt, pharmaceutical product, crystal, N-oxide, hydrate increasing nitrogen uptake by muscles, leading to an increase 35 or any combination thereof. In one embodiment, this inven in the rate of protein synthesis and muscle/bone growth. tion is directed to a method of increasing lean mass of an In some embodiments, the present invention provides a animal comprising administering a compound of formula method for enhanced production Such as milk, sperm, or egg. XXIII its isomer, pharmaceutically acceptable salt, pharma In some embodiments, the present invention provides a ceutical product, crystal, N-oxide, hydrate or any combina method for enhanced production of lean meats or eggs. In 40 tion thereof. In one embodiment, this invention is directed to Some embodiments, the present invention provides a method a method of increasing lean mass of an animal comprising for increased productivity of feeds or stud livestock, for administering a compound of to formula XXIV or its isomer, example, increased sperm count, improved morphology of pharmaceutically acceptable salt, pharmaceutical product, sperm, etc. In some embodiments, the present invention pro crystal, N-oxide, hydrate or any combination thereof. In one vides a method for expanding the productive life of farm 45 embodiment, this invention is directed to a method of increas animals, for example, egg-laying hens, milk-producing cows, ing lean mass of an animal comprising administering a com etc., and/or enhanced herd health, for example, improved pound of formula XXV or its isomer, pharmaceutically immune clearance, stronger animals. acceptable salt, pharmaceutical product, crystal, N-oxide, In one embodiment, this invention is directed to a method hydrate or any combination thereof. of affecting the carcass composition of an animal comprising 50 In one embodiment, this invention is directed to a method administering a compound of this invention. In one embodi of reducing fat mass of an animal comprising administering a ment, this invention is directed to a method of affecting the compound of this invention. In one embodiment, this inven carcass composition of an animal comprising administering a tion is directed to a method of reducing fat mass of an animal compound of formula IIIA or its isomer, pharmaceutically comprising administering a compound of formula IIIA or its acceptable salt, pharmaceutical product, crystal, N-oxide, 55 isomer, pharmaceutically acceptable salt, pharmaceutical hydrate or any combination thereof. In one embodiment, this product, crystal, N-oxide, hydrate or any combination invention is directed to a method of affecting the carcass thereof. In one embodiment, this invention is directed to a composition of an animal comprising administering a to com method of reducing fat mass of an animal comprising admin pound of formula I or its isomer, pharmaceutically acceptable istering a compound of formula I or its isomer, pharmaceuti salt, pharmaceutical product, crystal, N-oxide, hydrate or any 60 cally acceptable salt, pharmaceutical product, crystal, N-ox combination thereof. In one embodiment, this invention is ide, hydrate or any combination thereof. In one embodiment, directed to a method of affecting the carcass composition of this invention is directed to a method of reducing fat mass of an animal comprising administering a compound of formula an animal comprising administering a compound of formula II or its isomer, pharmaceutically acceptable salt, pharmaceu II or its isomer, pharmaceutically acceptable salt, pharmaceu tical product, crystal, N-oxide, hydrate or any combination 65 tical product, crystal, N-oxide, hydrate or any combination thereof. In one embodiment, this invention is directed to a thereof. In one embodiment, this invention is directed to a method of affecting the carcass composition of an animal method of reducing fat mass of an animal comprising admin US 8,309,603 B2 39 40 istering a compound of formula XXIII its isomer, pharma invention is directed to a method of increasing feed efficiency ceutically acceptable salt, pharmaceutical product, crystal, of an animal comprising administering a compound of for N-oxide, hydrate or any combination thereof. In one embodi mula XXIV or its isomer, pharmaceutically acceptable salt, ment, this invention is directed to a method of reducing fat pharmaceutical product, crystal, N-oxide, hydrate or any mass of an animal comprising administering a compound of 5 combination thereof. In one embodiment, this invention is formula XXIV or its isomer, pharmaceutically acceptable directed to a method of increasing feed efficiency of an animal salt, pharmaceutical product, crystal, N-oxide, hydrate or any comprising administering a compound of formula XXV or its combination thereof. In one embodiment, this invention is isomer, pharmaceutically acceptable salt, pharmaceutical directed to a method of reducing fat mass of an animal com product, crystal, N-oxide, hydrate or any combination prising administering a compound of formula XXV or its 10 thereof. isomer, pharmaceutically acceptable salt, pharmaceutical In one embodiment, this invention is directed to a method product, crystal, N-oxide, hydrate or any combination of increasing average to daily gain (ADG) of an animal com thereof. prising administering a compound of this invention. In one In one embodiment, this invention is directed to a method embodiment, this invention is directed to a method of increas of reducing percent fat mass of an animal comprising admin 15 ing average daily gain (ADG) of an animal comprising istering a compound of this invention. In one embodiment, administering a compound of formula IIIA or its isomer, this invention is directed to a method of reducing percent fat pharmaceutically acceptable salt, pharmaceutical product, mass of an animal comprising administering a compound of crystal, N-oxide, hydrate or any combination thereof. In one formula IIIA or its isomer, pharmaceutically acceptable salt, embodiment, this invention is directed to a method of is pharmaceutical product, crystal, N-oxide, hydrate or any increasing average daily gain (ADG) of an animal comprising combination thereof. In one embodiment, this invention is administering a compound of formula I or its isomer, phar directed to a method of reducing percent fat mass of an animal maceutically acceptable salt, pharmaceutical product, crys comprising administering a compound of formula I or its tal, N-oxide, hydrate or any combination thereof. In one isomer, pharmaceutically acceptable salt, pharmaceutical embodiment, this invention is directed to a method of increas product, crystal, N-oxide, hydrate or any combination 25 ing average daily gain (ADG) of an animal comprising thereof. In one embodiment, this invention is directed to a administering a compound of formula II or its isomer, phar method of reducing to percent fat mass of an animal compris maceutically acceptable salt, pharmaceutical product, crys ing administering a compound of formula II or its isomer, tal, N-oxide, hydrate or any combination thereof. In one pharmaceutically acceptable salt, pharmaceutical product, embodiment, this invention is directed to a method of increas crystal, N-oxide, hydrate or any combination thereof. In one 30 ing average daily gain (ADG) of an animal comprising embodiment, this invention is directed to a method of reduc administering a compound of formula XXIII its isomer, phar ing percent fat mass of an animal comprising administering a maceutically acceptable salt, pharmaceutical product, crys compound of formula XXIII its isomer, pharmaceutically tal, N-oxide, hydrate or any combination thereof. In one acceptable salt, pharmaceutical product, crystal, N-oxide, is embodiment, this invention is directed to a method of increas hydrate or any combination thereof. In one embodiment, this 35 ing average daily gain (ADG) of an animal comprising invention is directed to a method of reducing percent fat mass administering a compound of formula XXIV or its isomer, of an animal comprising administering a compound of for pharmaceutically acceptable salt, pharmaceutical product, mula XXIV or its isomer, pharmaceutically acceptable salt, crystal, N-oxide, hydrate or any combination thereof. In one pharmaceutical product, crystal, N-oxide, hydrate or any embodiment, this invention is directed to a method of increas combination thereof. In one embodiment, this invention is 40 ing average daily gain (ADG) of an animal comprising directed to a method of reducing percent fat mass of an animal administering a compound of formula XXV or its isomer, comprising administering a compound of formula XXV or its pharmaceutically acceptable salt, pharmaceutical product, isomer, pharmaceutically acceptable salt, pharmaceutical crystal, N-oxide, hydrate or any combination thereof. product, crystal, N-oxide, hydrate or any combination In one embodiment, this invention is directed to a method thereof. 45 of decreasing feed to gain ratio (F:G) of an animal comprising In one embodiment, this invention is directed to a method administering a compound of this invention. In one embodi of increasing feed efficiency of an animal comprising admin ment, this invention is directed to a method of decreasing feed istering a compound of this invention. In one embodiment, to gain ratio (F:G) of an animal comprising administering a this invention is directed to a method of increasing feed effi compound of formula IIIA or its isomer, pharmaceutically ciency of an animal comprising administering a compound of 50 acceptable salt, pharmaceutical product, crystal, N-oxide, formula IIIA or its isomer, pharmaceutically acceptable salt, hydrate or any combination thereof. In one embodiment, this pharmaceutical product, crystal, N-oxide, hydrate or any invention is directed to a method of decreasing feed to gain combination thereof. In one embodiment, this invention is ratio (F:G) of an animal comprising administering a com directed to a method of increasing feed efficiency of an animal pound of formula I or its isomer, pharmaceutically acceptable comprising administering a compound of formula I or its 55 salt, pharmaceutical product, crystal, N-oxide, hydrate or any isomer, pharmaceutically acceptable salt, pharmaceutical combination thereof. In one embodiment, this invention is product, crystal, N-oxide, hydrate or any combination directed to a method of decreasing feed to gain ratio (F:G) of thereof. In one embodiment, this invention is directed to a an animal comprising administering a compound of formula method of increasing feed efficiency of an animal comprising II or its isomer, pharmaceutically acceptable salt, pharmaceu administering a compound of formula II or its isomer, phar 60 tical product, crystal, N-oxide, hydrate or any combination maceutically acceptable salt, pharmaceutical product, crys thereof. In one embodiment, this invention is directed to a tal, N-oxide, hydrate or any combination thereof. In one method of decreasing feed to gain ratio (F:G) of an animal embodiment, this invention is directed to a method of increas comprising administering a compound of formula XXIII its ing feed efficiency of an animal comprising administering a isomer, pharmaceutically to acceptable salt, pharmaceutical compound of formula XXIII its isomer, pharmaceutically 65 product, crystal, N-oxide, hydrate or any combination acceptable salt, pharmaceutical product, crystal, N-oxide, thereof. In one embodiment, this invention is directed to a hydrate or any combination thereof. In one embodiment, this method of decreasing feed to gain ratio (F:G) of an animal US 8,309,603 B2 41 42 comprising administering a compound of formula XXIV or of the compounds of this invention for increasinglean mass of its isomer, pharmaceutically acceptable salt, pharmaceutical an animal by 15-30% after 14-21 days. In another embodi product, crystal, N-oxide, hydrate or any combination ment the methods and/or compositions of this invention make thereof. In one embodiment, this invention is directed to a use of the compounds of this invention for increasing lean method of decreasing feed to gain ratio (F:G) of an animal mass of an animal by 15-30% after 21-28 days. In another comprising administering a compound of formula XXV or its embodiment the methods and/or compositions of this inven isomer, pharmaceutically acceptable salt, pharmaceutical tion make use of the compounds of this invention for increas product, crystal, N-oxide, hydrate or any combination ing lean mass of an animal by 15-30% after 28-60 days. thereof. In one embodiment, the methods of this invention include In one embodiment the compounds, compositions and 10 administering a compound and/or feeding composition to an methods of this invention decrease the fat mass of an animal animal. In another embodiment, the compound and/or feed by 2-15%. In another embodiment, decrease the fat mass of an composition is provided in the daily feed to the animal. In animal by 2-10%. In another embodiment, decrease the fat another embodiment, the feed composition comprises a com mass of an animal by 5-10%. In another embodiment, pound of this invention. In another embodiment, the feed decrease the fat mass of an animal by 5-15%. In another 15 composition comprises a combination of a compound of this embodiment, the animal is a pig. In another embodiment the invention and a beta-agonist. In another embodiment, the animal is a beef cattle. In another embodiment, the animal is beta-agonist is Ractopamine hydrochloride (PayleanR). a finishing livestock. In another embodiment the animal is a The pharmaceutical compositions and feed composition feedlot animal. containing the compounds of this invention can be adminis In another embodiment the methods and/or compositions tered to a Subject by any method known to a person skilled in of this invention make use of the compounds of this invention the art, Such as orally, parenterally, intravascularly, paracan for decreasing the fat mass of an animal by 5-15% after 7-28 cerally, transmucosally, transdermally, intramuscularly, days. In another embodiment the methods and/or composi intranasally, intravenously, intradermally, Subcutaneously, tions of this invention make use of the compounds of this Sublingually, intraperitoneally, intraventricularly, intracrani invention for decreasing the fat mass of an animal by 5-15% 25 ally, intravaginally, by inhalation, rectally, intratumorally, or after 7-14 days. In another embodiment the methods and/or by any means in which the recombinant virus/composition compositions of this invention make use of the compounds of can be delivered to tissue (e.g., needle or catheter). Alterna this invention for decreasing the fat mass of an animal by tively, topical administration may be desired for application 5-15% after 14-21 days. In another embodiment the methods to mucosal cells, for skin or ocular application. Another and/or compositions of this invention make use of the com 30 method of administration is via aspiration or aerosol formu pounds of this invention for decreasing the fat mass of an lation. animal by 5-15% after 21-28 days. In another embodiment In one embodiment, the pharmaceutical compositions are the methods and/or compositions of this invention make use administered orally, and are thus formulated in a form Suit of the compounds of this invention for decreasing the fat mass able for oral administration, i.e. as a solid or a liquid prepa of an animal by 5-15% after 28-60 days. 35 ration. Suitable solid oral formulations include tablets, cap In one embodiment the compounds, compositions and Sules, pills, granules, pellets, powders, and the like. Suitable methods of this invention increase lean mass of an animal by liquid oral formulations include Solutions, Suspensions, dis 5-15%. In another embodiment, increase lean mass of an persions, emulsions, oils and the like. In one embodiment of animal by 5-10%. In another embodiment, increase lean mass the present invention, the SARM compounds are formulated of an animal by 8-10%. In another embodiment, increase lean 40 in a capsule. In accordance with this embodiment, the com mass of an animal by 15-30%. In another embodiment, the positions of the present invention comprise in addition to a animal is a pig. In another embodiment the animal is beef compound of this invention and the inert carrier or diluent, a cattle. In another embodiment, the animal is a finishing live hard gelatin capsule. stock. In another embodiment the animal is a feedlot animal. In one embodiment, the micronized capsules comprise par In another embodiment the methods and/or compositions 45 ticles containing a compound of this invention, wherein the of this invention make use of the compounds of this invention term “micronized' used herein refers to particles having a for increasing lean mass of an animal by 5-15% after 7-28 particle size is of less than 100 microns, or in another embodi days. In another embodiment the methods and/or composi ment, less than 60 microns, or in another embodiment, less tions of this invention make use of the compounds of this than 36 microns, or in another embodiment, less than 16 invention for increasing lean mass of an animal by 5-15% 50 microns, or in another embodiment, less than 10 microns, or after 7-14 days. In another embodiment the methods and/or in another embodiment, less than 6 microns. compositions of this invention make use of the compounds of Further, in another embodiment, the pharmaceutical com this invention for increasinglean mass of an animal by 5-15% positions are administered by intravenous, intraarterial, or after 14-21 days. In another embodiment the methods and/or intramuscular injection of a liquid preparation. Suitable liq is compositions of this invention make use of the compounds 55 uid formulations include solutions, Suspensions, dispersions, of this invention for increasing lean mass of an animal by emulsions, oils and the like. In one embodiment, the pharma 5-15% after 21-28 days. In another embodiment the methods ceutical compositions are administered intravenously, and are and/or compositions of this invention make use of the com thus formulated in a form suitable for intravenous adminis pounds of this invention for increasinglean mass of an animal tration. In another embodiment, the pharmaceutical compo by 5-15% after 28-60 days. In another embodiment the meth 60 sitions are administered intraarterially, and are thus formu ods and/or compositions of this invention make use of the lated in a form suitable for intraarterial administration. In compounds of this invention for increasing lean mass of an another embodiment, the pharmaceutical compositions are animal by 15-30% after 7-28 days. In another embodiment administered intramuscularly, and are thus formulated in a the methods and/or compositions of this invention make use form suitable for intramuscular administration. of the compounds of this invention for increasinglean mass of 65 Further, in another embodiment, the pharmaceutical com an animal by 15-30% after 7-14 days. In another embodiment positions are administered topically to body Surfaces, and are the methods and/or compositions of this invention make use thus formulated in a form suitable for topical administration. US 8,309,603 B2 43 44 Suitable topical formulations include gels, ointments, origin, for example, peanut oil, soybean oil, mineral oil, olive creams, lotions, drops and the like. For topical administration, oil, sunflower oil, and fish-liver oil. the compounds of this invention or their physiologically tol Parenteral vehicles (for Subcutaneous, intravenous, intraar erated derivatives such as salts, esters, N-oxides, and the like terial, or intramuscular injection) include Sodium chloride are prepared and applied as solutions, Suspensions, or emul Solution, Ringers dextrose, dextrose and sodium chloride, sions in a physiologically acceptable diluent with or without lactated Ringer's and fixed oils. Intravenous vehicles include a pharmaceutical carrier. fluid and nutrient replenishers, electrolyte replenishers such Further, in another embodiment, the pharmaceutical com as those based on Ringer's dextrose, and the like. Examples positions are to administered as a Suppository, for example a are sterile liquids such as water and oils, with or without the 10 addition of a surfactant and other pharmaceutically accept rectal Suppository or a urethral Suppository. Further, in able adjuvants. In general, water, Saline, aqueous dextrose another embodiment, the pharmaceutical compositions are and related Sugar Solutions, and glycols such as propylene administered by Subcutaneous implantation of a pellet. In a glycols or polyethylene glycol are preferred liquid carriers, further embodiment, the pelletprovides for controlled release particularly for injectable solutions. Examples of oils are of a compound as herein described over a period of time. In a 15 those of petroleum, animal, vegetable, or synthetic origin, for further embodiment, the pharmaceutical compositions are example, peanut oil, soybean oil, mineral oil, olive oil, Sun administered intravaginally. flower oil, and fish-liver oil. In another embodiment, the active compound can be deliv In addition, the compositions may further comprise bind ered in a vesicle, in particular a liposome (see Langer, Science ers (e.g. acacia, cornstarch, gelatin, carbomer, ethyl cellulose, 249:1627-1633 (1990); Treat et al., in Liposomes in the guar gum, hydroxypropyl cellulose, hydroxypropyl methyl Therapy of Infectious Disease and Cancer, Lopez-Berestein cellulose, povidone), disintegrating agents (e.g. cornstarch, and Fidler (eds.), Liss, New York, pp. 363-366 (1989); Lopez potato starch, alginic acid, silicon dioxide, croScarmelose Berestein, ibid., pp. 317-327; see generally ibid). Sodium, crospovidone, guar gum, Sodium starch glycolate), As used herein “pharmaceutically acceptable carriers or buffers (e.g., Tris-HCl acetate, phosphate) of various pH and diluents' are well knownto those skilled in theart. The carrier 25 ionic strength, additives such as albuminor gelatin to prevent or diluent may be a solid carrier or diluent for solid formula absorption to Surfaces, detergents (e.g., Tween 20, Tween 80, tions, a liquid carrier or diluent for liquid formulations, or Pluronic F68, bile acid salts), protease inhibitors, surfactants mixtures thereof. (e.g. sodium lauryl Sulfate), permeation enhancers, Solubiliz Solid carriers/diluents include, but are not limited to, a ing agents (e.g., cremophor, glycerol, polyethylene glycerol, gum, a starch (e.g. corn starch, pregeletanized starch), a Sugar 30 benzlkonium chloride, benzyl benzoate, cyclodextrins, sobi (e.g., lactose, mannitol. Sucrose, dextrose), a cellulosic mate tan esters, Stearic acids), anti-oxidants (e.g., ascorbic acid, rial (e.g. microcrystalline cellulose), an acrylate (e.g. polym sodium metabisulfite, butylated hydroxyanisole), stabilizers ethylacrylate), calcium carbonate, magnesium oxide, talc, or (e.g. hydroxypropyl cellulose, hyroxypropylmethyl cellu mixtures thereof. lose), viscosity increasing agents (e.g. carbomer, colloidal In one embodiment, the compositions of this invention 35 silicon dioxide, ethyl cellulose, guar gum), Sweetners (e.g. may include, a compound of this invention or any combina aspartame, citric acid), preservatives (e.g., Thimerosal, ben tion thereof, together with one or more pharmaceutically Zyl alcohol, parabens), coloring agents, lubricants (e.g. acceptable excipients. Stearic acid, magnesium Stearate, polyethylene glycol, It is to be understood that this invention encompasses any Sodium lauryl Sulfate), flow-aids (e.g. colloidal silicon diox embodiment of a compound as described herein, which in 40 ide), plasticizers (e.g. diethyl phthalate, triethyl citrate), some embodiments is referred to as “a compound of this emulsifiers (e.g. carbomer, hydroxypropyl cellulose, sodium invention'. Such reference will include any compound, lauryl Sulfate), polymer coatings (e.g., poloxamers or poloX which is characterized by a structure of the formulas I-XXV. amines), coating and film forming agents (e.g. ethylcellulose, or any embodiment thereof, as described herein. acrylates, polymethacrylates), and/or adjuvants. Suitable excipients and carriers may be, according to 45 In one embodiment, the pharmaceutical compositions pro embodiments of the invention, solid or liquid and the type is vided herein are controlled release compositions, i.e. compo generally chosen based on the type of administration being sitions in which the compound of this is invention is released used. Liposomes may also be used to deliver the composition. over a period of time after administration. Controlled or sus Examples of Suitable Solid carriers include lactose, Sucrose, tained release compositions include formulation in lipophilic gelatin and agar. Oral dosage forms may contain Suitable 50 depots (e.g. fatty acids, waxes, oils). In another embodiment, binders, lubricants, diluents, disintegrating agents, coloring the composition is an immediate release composition, i.e. a agents, flavoring agents, flow-inducing agents, and melting composition in which all of the compound is released imme agents. Liquid dosage forms may contain, for example, Suit diately after administration. able solvents, preservatives, emulsifying agents, Suspending In yet another embodiment, the pharmaceutical composi agents, diluents, Sweeteners, thickeners, and melting agents. 55 tion can be delivered in a controlled release system. For Parenteral and intravenous forms should also include miner example, the agent may be administered using intravenous als and other materials to make them compatible with the type infusion, an implantable osmotic pump, a transdermal patch, of injection or delivery system chosen. Of course, to other liposomes, or other modes of administration. In one embodi excipients may also be used. ment, a pump may be used (see Langer, Supra; Sefton, CRC For liquid formulations, pharmaceutically acceptable car 60 Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., riers may be aqueous or non-aqueous Solutions, Suspensions, Surgery 88:607 (1980); Saudek et al., N. Engl. J. Med. 321: emulsions or oils. Examples of non-aqueous solvents are 674 (1989). In another embodiment, polymeric materials can propylene glycol, polyethylene glycol, and injectable organic be used. In yet another embodiment, a controlled release esters such as ethyl oleate. Aqueous carriers include water, system can be placed in proximity to the therapeutic target, alcoholic/aqueous solutions, cyclodextrins, emulsions or Sus 65 i.e., the brain, thus requiring only a fraction of the systemic pensions, including saline and buffered media. Examples of dose (see, e.g., Goodson, in Medical Applications of Con oils are those of petroleum, animal, vegetable, or synthetic trolled Release, supra, Vol. 2, pp. 116-138 (1984). Other US 8,309,603 B2 45 46 controlled release systems are discussed in the review by of this invention include acid addition salts which may, for Langer (Science 249:1627-1633 (1990). example, be formed by mixing a solution of the compound The compositions may also include incorporation of the according to the invention with a solution of a pharmaceuti active material into or onto particulate preparations of poly cally acceptable to acid such as hydrochloric acid, Sulphuric meric compounds such as polylactic acid, polglycolic acid, acid, methaneSulphonic acid, fumaric acid, maleic acid, Suc hydrogels, etc, or onto liposomes, microemulsions, micelles, cinic acid, acetic acid, benzoic: acid, oxalic acid, citric acid, unilamellar or multilamellar vesicles, erythrocyte ghosts, or tartaric acid, carbonic acid or phosphoric acid. spheroplasts.) Such compositions will influence the physical In one embodiment, this invention provides pharmaceuti state, solubility, stability, rate of in vivo release, and rate of in cal compositions comprising a compound of this invention. In Vivo clearance. 10 Also comprehended by the invention are particulate com one embodiment, such compositions are useful for oral test positions coated with polymers (e.g. poloxamers or poloxam osterone replacement therapy. ines) and the compound coupled to antibodies directed In one embodiment, this invention also provides a compo against tissue-specific receptors, ligands or antigens or sition comprising two or more compounds of this invention, coupled to ligands of tissue-specific receptors. 15 or polymorphs, isomers, hydrates, salts, N-oxides, etc., Also comprehended by the invention are compounds modi thereof. The present invention also relates to compositions fied by the covalent attachment of water-soluble polymers and pharmaceutical compositions which comprise a com Such as polyethylene glycol, copolymers of polyethylene gly pound of this invention alone or in combination with a proges col and polypropylene glycol, carboxymethyl cellulose, dex tin or estrogen, or in another embodiment, chemotherapeutic tran, polyvinyl alcohol, polyvinylpyrrolidone or polyproline. compound, osteogenic or myogenic compound, or other The modified compounds are known to exhibit substantially agents suitable for the applications as herein described. In one longer half-lives in blood following intravenous injection embodiment, the compositions of this invention will com than do the corresponding unmodified compounds (Abu prise a suitable carrier, diluent or salt. chowski et al., 1981; Newmarket al., 1982; and Katre et al., In one embodiment, the methods of this invention may 1987). Such modifications may also increase the compounds 25 comprise administration of a compound of this invention at solubility in aqueous Solution, eliminate aggregation, various dosages. In another embodiment, the methods of this enhance the physical and chemical stability of the compound, invention may comprise administration of a compound of and greatly reduce the immunogenicity and reactivity of the formula II of this invention at various dosages. In one embodi compound. As a result, the desired in vivo biological activity ment, the compound of this invention is administered at a may be achieved by the is administration of Such polymer 30 dosage of 0.1-200 mg per day. In one embodiment, the com compound abducts less frequently or in lowerdoses than with pound of this invention is administered at a dose of 0.1-10 mg. the unmodified compound. or in another embodiment, 0.1-26 mg, or in another embodi The preparation of pharmaceutical compositions which ment, 0.1-60 mg, or in another embodiment, 0.3-16 mg, or in contain an active component is well understood in the art, for another embodiment, 0.3-30 mg. or in another embodiment, example by mixing, granulating, or tablet-forming processes. 35 0.6-26 mg, or in another embodiment, 0.6-60 mg, or in The active therapeutic ingredient is often mixed with excipi another embodiment, 0.76-16 mg, or in another embodiment, ents which are pharmaceutically acceptable and compatible 0.76-60 mg. or in another embodiment, 1-6 mg, or in another with the active ingredient. For oral administration, the com embodiment, 1-20 mg, or in another embodiment, 3-16 mg, pounds of this invention or their physiologically tolerated or in another embodiment, 30-60 mg. or in another embodi derivatives such as salts, esters, N-oxides, and the like are 40 ment, 30-76 mg, or in another embodiment, 100-2000 mg. or mixed with additives customary for this purpose. Such as in another embodiment, 1000-20,000 mg. vehicles, stabilizers, or inert diluents, and converted by cus In one embodiment, the methods of this invention may tomary methods into Suitable forms for administration, Such comprise administration of a compound of this invention at as tablets, coated tablets, hard or soft gelatin capsules, aque various dosages. In another embodiment, the methods of this ous, alcoholic or oily solutions. For parenteral administration, 45 invention may comprise administration of a compound of the compounds of this invention or their physiologically tol formula II of this invention at various dosages. In one embodi erated derivatives such as salts, esters, N-oxides, and the like ment, the compound of this invention is administered at a are converted into a solution, Suspension, or emulsion, if dosage of 1 mg. In another embodiment the compound of this desired with the substances customary and suitable for this invention is administered at a dosage of 6 mg, 10 mg, 16 mg, purpose, for example, solubilizers or other. 50 20 mg, 26 mg, 30 mg, 36 mg, 40 mg., 46 mg, 50 mg, 56 mg, 60 An active component can be formulated into the composi mg, 66 mg, 70 mg, 76 mg, 80 mg, 86 mg, 90 mg., 96 mg, 100 tion as neutralized pharmaceutically acceptable salt forms. mg, 200 mg, 500 mg, 1000 mg, 2000 mg, 10,000 mg. or Pharmaceutically acceptable salts include the acid addition 20,000 mg. salts (formed with the free amino groups of the polypeptide or In one embodiment, the present invention provides meth antibody molecule), which are formed with inorganic acids 55 ods of use comprising the administration of a composition Such as, for example, hydrochloric or phosphoric acids, or comprising a) any embodiment of a compound as described Such organic acids as acetic, oxalic, tartaric, mandelic, and the herein; and b) additives, a pharmaceutically acceptable car like. Salts formed from the free carboxyl groups can also be rier or diluent; which is to be understood to include an analog, derived from inorganic bases Such as, for example, Sodium, isomer, metabolite, derivative, pharmaceutically acceptable potassium, ammonium, calcium, or ferric hydroxides, and 60 salt, N-oxide, hydrate or any combination thereof of a com Such organic bases as isopropylamine, trimethylamine, pound as herein described, and may comprise compounds of 2-ethylamino ethanol, histidine, procaine, and the like. formulas I-XXV. For use in medicine, the salts of the compound will be In some embodiments, the present invention provides pharmaceutically acceptable salts. Other salts may, however, methods of use of a composition comprising a) any embodi be useful in the preparation of the compounds according to 65 ment of the compounds as described herein, including an the invention or of their pharmaceutically acceptable salts. analog, isomer, metabolite, derivative, pharmaceutically Suitable pharmaceutically acceptable salts of the compounds acceptable salt, pharmaceutical product, N-oxide, hydrate US 8,309,603 B2 47 48 thereof or any combination thereof; b) a pharmaceutically RU-56279, WS9761A and B, RU-59063, RU-58841, bexlos acceptable carrier or diluent; c) a flow-aid; and d) a lubricant. teride, LG-2293, L-245976, LG-121071, LG-121091, In another embodiment, the present invention provides LG-121104, LGD-2226, LGD-2941, YM-92088, methods of use of a composition comprising a) any embodi YM-175735, LGD-1331, BMS-357597, BMS-391197, ment of the compounds as described herein, including an S-40503, BMS-482404, EM-4283, EM-4977, BMS-564929, analog, isomer, metabolite, derivative, pharmaceutically BMS-391197, BMS-434588, BMS-487745, BMS-501949, acceptable salt, pharmaceutical product, N-oxide, hydrate SA-766, YM-92088, YM-580, LG-123303, LG-123129, thereof or any combination thereof; b) lactose monohydrate; PMCol, YM-175735, BMS-591305, BMS-591309, BMS c) microcrystalline cellulose; d) magnesium Stearate; e) addi 665139, BMS-665539, CE-590, 116BG33, 154BG31, arcar tives and f) colloidal silicon dioxide. 10 ine, ACP-105: SERMs, such as tamoxifene, 4-hydroxyta In some embodiments, the methods of this invention make moxifene, , , ospennifene, , use of compositions comprising compounds of this invention, , , , PPT (1,3,5-tris(4-hy which offer the advantage that the compounds are nonsteroi droxyphenyl)-4-propyl-1H-pyrazole), DPN, , dal ligands for the androgen receptor, and exhibit anabolic , EM-800, EM-652, , , activity in vivo. According to this aspect, such compounds are 15 tesmilifene, phosphate, RU 58,688, EM139, ICI unaccompanied by serious side effects, provide convenient 164,384, ICI 182,780, clomiphene, MER-25, diethyl modes of administration, and lower production costs and are stibestrol, , , GW5638, LY353581, orally bioavailable, lack of significant cross-reactivity with Zuclomiphene, enclomiphene, delmadinone acetate, DPPE, other undesired receptors, and may possess long bio (N,N-diethyl-2-4-(phenylmethyl)-phenoxyethanamine), logical half-lives. TSE-424, WAY-070, WAY-292, WAY-818, cyclocommunol, In one embodiment, the compositions for administration , ERB-041, WAY-397, WAY-244, ERB-196, WAY may be sterile solutions, or in other embodiments, aqueous or 169122, MF-101, ERb-002, ERB-037, ERB-017, BE-1060, non-aqueous, Suspensions or emulsions. In one embodiment, BE-380, BE-381, WAY-358, FIFEDNP, LSN-500307, the compositions may comprise propylene glycol, polyethyl AA-102, Ban Zhilian, CT-101, CT-102, VG-101; GnRHago ene glycol, injectable organic esters, for example ethyl oleate, 25 nists orantagonists, such as, leuprolide, goserelin, triptorelin, or cyclodextrins. In another embodiment, compositions may alfaprostol, histrelin, detirelix, ganirelix, antide iturelix, also comprise wetting, emulsifying and/or dispersing agents. cetrorelix, ramorelix, ganirelix, antarelix, teverelix, abarelix, In another embodiment, the compositions may also comprise ozarelix, Sufugolix, prazarelix, degarelix, NBI-56418, TAK sterile water or any other sterile injectable medium. 810, acyline; FSH agonist/antagonist, LH agonist/antago In one embodiment, the invention provides compounds 30 nists, aromatase inhibitors, such as, letrozole, anastrazole, and compositions, including any embodiment described atameStane, fadrozole, minamestane, exemestane, plomes herein, for use in any of the methods of this invention, as tane, liarozole, NKS-01, vorozole, YM-511, finrozole, 4-hy described herein. In one embodiment, use of a compound of droxyandrostenedione, aminogluethimide, rogletimide; Ste this invention or a composition comprising the same, will roidal or nonsteroidal glucocorticoid receptor ligands. Such have utility in inhibiting, Suppressing, enhancing or stimulat 35 as, ZK-216348, ZK-243149, ZK-243185, LGD-5552, mife ing a desired response in a Subject, as will be understood by pristone, RPR-106541, ORG-34517, GW-215864x, Sesqui one skilled in the art. In another embodiment, the composi cillin, CP-472555, CP-394531, A-222977, AL-438, tions may further comprise additional active ingredients, A-216054, A-276575, CP-394531, CP-409069, UGR-07; whose activity is useful for the particular application for Steroidal or nonsterodial progesterone receptor ligands; Ste which the compound of this invention is being administered. 40 roidal or nonsteroidal AR antagonists Such as flutamide, In some embodiments, the methods of this invention make hydroxyflutamide, bicalutamide, nilutamide, hydroxysteroid use of compositions comprising compounds of this invention, dehydrogenase inhibitors, PPARC. ligand such as bezafibrate, which offer the advantage that the compounds are nonsteroi fenofibrate, gemfibrozil; PPAR ligands such as dargilitazone, dal ligands for the androgen receptor, and exhibit anabolic pioglitaZone, rosiglitaZone, isaglitaZone, rivoglitaZone, activity in vivo. According to this aspect, such compounds are 45 netoglitaZone; Dual acting PPAR ligands, such as navegli unaccompanied by serious side effects, provide convenient tazar, fargilitazar, tesaglitazar, ragaglitazar, Oxeglitazar, modes of administration, and lower production costs and are PN-2034, PPAR 8; an anti-glucocorticoid such as RU-486; a orally bioavailable, lack significant cross-reactivity with 17-ketoreductase inhibitors, 3?-AHA4.6-isomerase inhibi other undesired steroid receptors, and may possess long bio tors, 3 B-AHA4.5-isomerase inhibitors, 17.20-desmolase logical half-lives. 50 inhibitors, p450c17 inhibitors, p450ssc inhibitors, 17.20 In some embodiments, the compositions will further com lyase inhibitors, or combinations thereof. prise a 5C.-reductase inhibitors (5ARI), a beta-agonist, a In some embodiments, the compositions will further com SARM or SARMs, a selective estrogen receptor modulator prise Ghrelin receptor ligand or growth hormone analogues (SERM), an , such as but not limited to and secretagogues, IGF-1, IGF-1 analogues and secreta anastraZole, exemestane, or letrozole; a GnRH agonist or 55 gogues, myostatin analogues, proteasome inhibitors, andro antagonist, a steroidal or nonsteroidal GR ligand, a steroidal genic-anabolic steroids, Enbrel, melanocortin 4 receptorago or nonsterodial PR ligand, a steroidal or nonsteroidal AR nist, insulins, or combinations thereof. Such compositions antagonist, a 17-aldoketoreductase inhibitor or 17 B-hydrox may be used, in Some embodiments, for promoting growth in ysteroid dehydrogenase inhibitor. Such compositions may be feedlot animals. used, in some embodiments, for treating a hormone depen 60 In some embodiments, the composition will comprise the dent condition, Such as, for example, infertility, neoplasia of compounds as described herein, as well as another therapeu a hormone-responsive cancer, for example, a gonadalcancer, tic compound, including interalia, ghrelin receptor ligand or or a urogenital cancer. growth hormone analogues and secretagogues, such as, pral In some embodiments, the composition will comprise the morelin, examorelin, tabimorelin, capimorelin, capromore compounds as described herein, as well as another therapeu 65 lin, ipamorelin, EP-01572, EP-1572, JMV-1843, an andro tic compound, including interalia, a 5ARI Such as finasteride, genic Such as testosterone or Oxandrolone; a dutasteride, izonsteride; other SARMs, such as, RU-58642, melanocortin 4 receptor agonist, Such as bremelanotide, a US 8,309,603 B2 49 50 ghrelin or analogue thereof. Such as human ghrelin, CYT megestrol acetate, cyproheptadine, hydrazine Sulfate, pen 009-GhrOb, L-692429, GHRP-6, SK&F-1 10679, toxifylline, thalidomide, anticytokine antibodies, cytokine U-75799E), leptin (metreleptin, pegylated leptin; a leptin inhibitors, eicosapentaenoic acid, indomethacin, ibuprofen, receptor agonist, such as LEP(116-130), OB3, D-Leu-4- melatonin, insulin, growth hormone, clenbuterol, porcine OB3, ra AV-leptin, AAV-hCB, ra AVhCB; an insulin (short pancreas extract, IGF-1, IGF-1 analogue and secretagogue, intermediate-, and long acting formulations); a cortisol or myostatin analogue, proteasome inhibitor, testosterone, corticosteroid, or a combination thereof. Oxandrolone, Enbrel, melanocortin 4 receptor agonist, or a The invention contemplates, in Some embodiments, combination thereof. administration of compositions comprising the individual In one embodiment, the agent used to treat a wasting dis agents, administered separately and by similar or alternative 10 ease is a ghrelin receptor ligand, growth hormone analogue, routes, formulated as appropriately for the route of adminis or a secretagogue. In some embodiments, ghrelin receptor tration. The invention contemplates, in Some embodiments, ligands, growth hormone analogues, or secretagogues administration of compositions comprising the individual include but are not limited to prailmorelin, examorelin, tabi agents, administered in the same formulation. The invention morelin, capimorelin, capromorelin, ipamorelin, EP-01572, contemplates, in some embodiments, staggered administra 15 EP-1572, or JMV-1843. tion, concurrent administration, of administration of the Vari In one embodiment, growth promoting agents such as but ous agents over a course of time, however, their effects are not limited to TRH. diethylstilbesterol, theophylline, synergistic in the Subject. enkephalins, E series prostaglandins, compounds disclosed in It is to be understood that any of the above means, timings, U.S. Pat. No. 3.239,345, e.g., , and compounds dis routes, or combinations thereof, of administration of two or closed in U.S. Pat. No. 4,036.979, e.g., sulbenox or peptides more agents is to be considered as being encompassed by the disclosed in U.S. Pat. No. 4,411,890 are utilized as agents phrase “administered in combination', as described herein. used to treat a wasting disease. It is to be understood that reference to “a compound of this In other embodiments, agents treating a wasting disease invention' or a use thereof is to be considered to encompass may comprise growth hormone secretagogues such as use of any compound as herein described, including any 25 GHRP-6, GHRP-1 (as described in U.S. Pat. No. 4,411,890 embodiment thereof. It is to be considered to encompass all of and publications WO 89/07110 and WO 89/07111), GHRP-2 compounds which may be characterized by the structure of (as described in WO 93/04081), NN703 (Novo Nordisk), formulas I-XXV. LY444711 (Lilly), MK-677 (Merck), CP424391 (Pfizer) and In one embodiment, the compound is administered in com B-HT920, or, in other embodiments, with growth hormone bination with an agent, which treats bone diseases, disorders 30 releasing factor and its analogs or growth hormone and its or conditions, such as osteoporosis, bone fractures, etc., and analogs, or with alpha-adrenergic agonists, such as clonidine this invention comprises methods of treating the same, by or serotinin 5-HTDagonists, such as Sumatriptan, or agents administering the compounds as herein described, alone or in which is inhibit somatostatin or its release, such as physos combination with other agents. tigmine and pyridostigmine. In some embodiments, agents Such agents for combined use may comprise a SERM, as 35 treating a wasting disease may comprise parathyroid hor herein described, a bisphosphonate, for example, alendr mone, PTH (1-34) or bisphosphonates, such as MK-217 (al onate, tiludroate, clodroniate, pamidronate, etidronate, allen endronate). In other embodiments, agents treating wasting dronate, Zolendronate, cimadronate, neridronate, minodronic disease may further comprise estrogen, a selective estrogen acid, ibandronate, risedronate, homoresidronate, a calcitonin, receptor modulator, Such as tamoxifene or raloxifene, or other for example, salmon, Elcatonin, SUN-8577, TJN-135; a Vita 40 androgen receptor modulators, such as those disclosed in min D or derivative (ZK-156979); a Vitamin D receptor Edwards, J. P. et. al., Bio. Med. Chem. Let., 9, 1003-1008 ligand or analogues thereof. Such as calcitriol, topitriol, (1999) and Hamann, L. G. et. al., J.Med. Chem., 42, 210-212 ZK-15O123, TEI-9647, BXL-628, Ro-26-9228, BAL-2299, (1999). In some embodiments, agents treating a wasting dis Ro-65-2299, DP-035, an estrogen, estrogen derivative, or ease may further comprise a progesterone receptor agonists conjugated estrogen; an antiestrogen, progestin, Synthetic 45 (“PRA), such as levonorgestrel, medroxyprogesterone estrogen/progestin; a RANK ligand mAb, for example, deno acetate (MPA). In some embodiments, agents treating a wast sumab or AMG162 (Amgen); a beta 3 integrin receptor ing disease may include nutritional Supplements, such as antagonist; an osteoclast vacuolar ATPase inhibitor; an those described in U.S. Pat. No. 5,179,080, which, in other antagonist of VEGF binding to osteoclast receptors; a cal embodiments are in combination with whey protein or casein, cium receptor antagonist; PTh (parathyroid hormone) orana 50 amino acids (such as leucine, branched amino acids and logues thereof, PTHrP analogues (parathyroid hormone-re hydroxymethylbutyrate), triglycerides, vitamins (e.g., A, B6. lated peptide), Cathepsin K inhibitors (AAE581); Strontium B 12, folate, C, D and E), minerals (e.g., selenium, magne ranelate; : HCT-1026, PSK3471; Gallium maltolate: sium, Zinc, chromium, calcium and potassium), camitine, Nutropin AQ: Prostaglandins, p38 protein kinase inhibitor; a lipoic acid, creatinine, B-hyroxy-B-methylbutyriate (Juven) bone morphogenetic protein; an inhibitor of BMP antago 55 and coenzyme Q. In one embodiment, agents treating a wast nism, an. HMG-CoA reductase inhibitor, a Vitamin K or ing disease may further comprise antiresorptive agents, Vita derivative, an antiresorptive, an Ipriflavone, a fluoride salt, min D analogues, elemental calcium and calcium Supple dietary calcium Supplement, Osteoprotegerin, or any combi ments, cathepsin K inhibitors, MMP inhibitors, vitronectin nation thereof. In one embodiment, the combined adminis receptor antagonists, Src SH2 antagonists, Vacular-H+-AT tration of a SARM as herein described, Osteoprotegerin and 60 Pase inhibitors, ipriflavone, fluoride, tibolone, prostanoids, parathyroid hormone is contemplated for treating any dis 17-beta hydroxysteroid dehydrogenase inhibitors and Src ease, disorder or condition of the bone. kinase inhibitors. In one embodiment, the compound is administered with an In one embodiment, the SARM compound is administered agent used to treat a wasting disease. In some embodiments, with an agent treating osteoporosis. In some embodiments, agents used to treat a wasting disease include but are not 65 agents treating osteoporosis include but are not limited to limited to corticosteroids, anabolic steroids, cannabinoids, SERMs, calcitonin, vitamin D, vitamin D derivatives, vita metoclopramide, cisapride, medroxyprogesterone acetate, min D receptor ligand, Vitamin D receptor ligand analogue, US 8,309,603 B2 51 52 estrogen, estrogen derivative, conjugated estrogen, antiestro severity of a disease, age, sex, or body weight as can be gen, progestin, Synthetic estrogen, Synthetic progestin, readily made by a person skilled in the art. RANK ligand monoclonal antibody, integrin receptor It is to be understood that this invention is directed to antagonist, osteoclast vacuolar ATPase inhibitor, antagonist compositions and combined therapies as described herein, for of VEGF binding to osteoclast receptors, calcium receptor any disease, disorder or condition, as appropriate, as will be antagonist, parathyroid hormone, parathyroid hormone ana appreciated by one skilled in the art. Certain applications of logue, parathyroid hormone-related peptide, cathepsin K Such compositions and combined therapies have been inhibitor, strontium ranelate, tibolone, HCT-1026, PSK3471, described hereinabove, for specific diseases, disorders and gallium maltolate, nutropin AQ, prostaglandin, p38 protein conditions, representing embodiments of this invention, and 10 methods of treating Such diseases, disorders and conditions in kinase inhibitor, bone morphogenetic protein (BMP), inhibi a Subject by administering a compound as herein described, tor of BMP antagonism, HMG-CoA reductase inhibitor, vita alone or as part of the combined therapy or using the compo min K. vitamin K derivative, ipriflavone, fluoride salts, sitions of this invention represent additional embodiments of dietary calcium Supplement, or osteoprotegerin. this invention. In one embodiment, the agent treating osteoporosis is a 15 Biological Activity of Selective Androgen Modulator calcitonin. In some embodiments, calcitonins include but are Compounds not limited to salmon, elcatonin, SUN-8577, or TJN-135. In some embodiments, the compounds of this invention In one embodiment, the agent treating osteoporosis is a possess in Vivo tissue selective androgenic and anabolic activ Vitamin D receptor ligand or analogue. In some embodi ity, which is accordingly utilized for particular applications, ments, vitamin D receptor ligands or analogues include but as will be appreciated by one skilled in the art. are not limited to calcitriol, topitriol, ZK-150123, TEI-9647, In one embodiment, the methods of this invention are use BXL-628, Ro-26-9228, BAL-2299, Ro-65-2299, or DP-035. ful a subject, which is a human. In another embodiment, the In one embodiment, the compound of this invention is Subject is a mammal. In another embodiment the Subject is an administered with a vitamin. In some embodiments, vitamins animal. In another embodiment the subject is an invertebrate. includebutare not limited to vitamin D, vitamin E, vitamin K, 25 In another embodiment the subject is a vertebrate. In one vitamin B, Vitamin C, or a combination thereof. embodiment, the subject is a feedlot animal, a beef cattle In some embodiments, any of the compositions of this and/or a finishing livestock. invention will comprise a compound of formula I-XXV, in In one embodiment, the subject is male. In another embodi any form or embodiment as described herein. In some ment, the subject is female. In some embodiments, while the embodiments, any of the compositions of this invention will 30 methods as described herein may be useful for treating either consist of a compound of formula I-XXV, in any form or males or females, females may respond more advantageously embodiment as described herein. In some embodiments, of to administration of certain compounds, for certain methods, the compositions of this invention will consistessentially of a as described and exemplified herein. compound of I-XXV, in any form or embodiment as In another embodiment of the present invention, a method described herein. In some embodiments, the term “comprise' 35 is provided for hormonal therapy in a patient (i.e., one Suffer refers to the inclusion of the indicated active agent, such as the ing from an androgen-dependent condition) which includes compound of formula I-XXV, as well as inclusion of other contacting an androgen receptor of a patient with a compound active agents, and pharmaceutically acceptable carriers, and/or a non steroidal agonist of the present invention and/or excipients, emollients, stabilizers, etc., as are known in the its analog, derivative, isomer, metabolite, pharmaceutically pharmaceutical industry. In some embodiments, the term 40 acceptable salt, pharmaceutical product, polymorph, crystal, “consisting essentially of refers to a composition, whose impurity, hydrate, N-oxide or any combination thereof, in an only active ingredient is the indicated active ingredient, how amount effective to bind the compound to the androgen ever, other compounds may be included which are for stabi receptor and effect a change in an androgen-dependent con lizing, preserving, etc. the formulation, but are not involved dition. directly in the therapeutic effect of the indicated active ingre 45 In one embodiment of this invention, a method is provided dient. In some embodiments, the term "consisting essentially for hormone replacement therapy in a patient (i.e., one Suf of may refer to components which facilitate the release of the fering from an androgen-dependent condition) which active ingredient. In some embodiments, the term “consist includes administering a compound as herein described and/ ing refers to a composition, which contains the active ingre or its analog, derivative, isomer, metabolite, pharmaceuti dient and a pharmaceutically acceptable carrier or excipient. 50 cally acceptable salt, pharmaceutical product, polymorph, In one embodiment, the present invention provides com crystal, impurity, hydrate, N-oxide or any combination bined preparations. In one embodiment, the term "a com thereof, to a subject, in an amount Sufficient to effect a change bined preparation' defines especially a “kit of parts” in the in a hormone-dependent condition in the Subject. sense that the combination partners as defined above can be In one embodiment, this invention provides for the use of a dosed independently or by use of different fixed combinations 55 compound as herein described, or its prodrug, analog, isomer, with distinguished amounts of the combination partners i.e., metabolite, derivative, pharmaceutically acceptable salt, simultaneously, concurrently, separately or sequentially. In pharmaceutical product, polymorph, crystal, impurity, N-OX Some embodiments, the parts of the kit of parts can then, e.g., ide, hydrate or any combination thereof, for a) accelerate be administered simultaneously or chronologically stag bone repair; b) treating bone disorders; c) treating bone den gered, that is at different time points and with equal or differ 60 sity loss; d) treating low bone mineral density (BMD); e) ent time intervals for any part of the kit of parts. The ratio of treating reduced bone mass: f) treating metabolic bone dis the total amounts of the combination partners, in some ease; g) promoting bone growth or regrowth; h) promoting embodiments, can be administered in the combined prepara bone restoration: i) promoting bone fracture repair, j) promot tion. In one embodiment, the combined preparation can be ing bone remodeling, k) treating bone damage following varied, e.g., in order to cope with the needs of a patient 65 reconstructive Surgery including of the face, hip, or joints; 1) subpopulation to be treated or the needs of the single patient enhancing of bone strength and function; m) increasing cor which different needs can be due to a particular disease, tical bone mass; n) increasing trabecular connectivity. US 8,309,603 B2 53 54 In one embodiment, the bone related disorder is a genetic ment, the hormonal disorder, disruption or imbalance is asso disorder, or in another embodiment, is induced as a result of ciated with menopause. In another embodiment, the hor a treatment regimen for a given disease. For example, and in monal disorder, disruption or imbalance is associated with one embodiment, the compounds as herein described are andropause, andropausal vasomotor symptoms, andropausal useful in treating a bone-related disorder that arises as a result gynecomastia, muscle strength and/or function, bone of cancer metastasis to bone, or in another embodiment, as a strength and/or function and anger. In another embodiment, result of androgen-deprivation therapy, for example, given in hormone deficiency is a result of specific manipulation, as a response to prostate carcinogenesis in the Subject. byproduct of treating a disease or disorder in the subject. For In one embodiment, the bone-related disorder is example, the hormone deficiency may be a result of androgen osteoporosis. In another embodiment, the bone-related disor 10 depletion in a subject, as a therapy for prostate cancer in the der is osteopenia. In another embodiment, the bone-related Subject. Each possibility represents a separate embodiment of disorder is increased bone resorption. In another embodi the present invention. ment, the bone-related disorder is bone fracture. In another In another embodiment the invention is directed to treating embodiment, the bone-related disorder is bone frailty. sarcopenia or cachexia, and associated conditions related In another embodiment, the bone-related disorder is a loss 15 thereto, for example diseases or disorders of the bone. of bone mineral density (BMD). In another embodiment, the In one embodiment, this invention provides for the use of a bone-related disorder is any combination of osteoporosis, compound as herein described, or its prodrug, analog, isomer, osteopenia, increased bone resorption, bone fracture, bone metabolite, derivative, pharmaceutically acceptable salt, frailty and loss of BMD. Each disorder represents a separate pharmaceutical product, polymorph, crystal, impurity, N-OX embodiment of the present invention. ide, hydrate or any combination thereof, for 1) treating a “Osteoporosis” refers, in one embodiment, to a thinning of muscle wasting disorder; 2) preventing a muscle wasting the bones with reduction in bone mass due to depletion of disorder; 3) treating, preventing, Suppressing, inhibiting or calcium and bone protein. In another embodiment, reducing muscle loss due to a muscle wasting disorder; 4) osteoporosis is a systemic skeletal disease, characterized by treating, preventing, inhibiting, reducing or Suppressing low bone mass and deterioration of bone tissue, with a con 25 muscle wasting due to a muscle wasting disorder, and/or 5) sequent increase in bone fragility and Susceptibility to frac treating, preventing, inhibiting, reducing or Suppressing ture. In osteoporotic patients, bone strength is abnormal, in muscle protein catabolism due to a muscle wasting disorder; one embodiment, with a resulting increase in the risk of and/or treating, preventing, inhibiting, reducing or Suppress fracture. In another embodiment, osteoporosis depletes both ing end stage renal disease; and/or 6) treating, preventing, the calcium and the protein collagen normally found in the 30 inhibiting, reducing or Suppressing frailty. bone, in one embodiment, resulting in either abnormal bone In another embodiment, the use of a compound for treating quality or decreased bone density. In another embodiment, a subject having a muscle wasting disorder, or any of the bones that are affected by osteoporosis can fracture with only disorders described herein, includes administering a pharma a minor fall or injury that normally would not cause a bone ceutical composition including a compound as herein fracture. The fracture can be, in one embodiment, either in the 35 described. In another embodiment, the administering step form of cracking (as in a hip fracture) or collapsing (as in a includes intravenously, intraarterially, or intramuscularly compression fracture of the spine). The spine, hips, and wrists injecting to said Subject said pharmaceutical composition in are common areas of osteoporosis-induced bone fractures, liquid form; Subcutaneously implanting in said subject a pel although fractures can also occur in other skeletal areas. let containing said pharmaceutical composition; orally Unchecked osteoporosis can lead, in another embodiment, to 40 administering to said Subject said pharmaceutical composi changes in posture, physical abnormality, and decreased tion in a liquid or Solid form; or topically applying to the skin mobility. Surface of said subject said pharmaceutical composition. In one embodiment, the osteoporosis results from andro A muscle is a tissue of the body that primarily functions as gen deprivation. In another embodiment, the osteoporosis a source of power. There are three types of muscles in the follows androgen deprivation. In another embodiment, the 45 body: a) skeletal muscle—the muscle responsible for moving osteoporosis is primary osteoporosis. In another embodi extremities and external areas of the bodies; b) cardiac ment, the osteoporosis is secondary osteoporosis. In another muscle—the heart muscle; and c) Smooth muscle—the embodiment, the osteoporosis is postmenopausal osteoporo muscle that is in the walls of arteries and bowel. sis. In another embodiment, the osteoporosis is juvenile A wasting condition or disorder is defined herein as a osteoporosis. In another embodiment, the osteoporosis is 50 condition or disorder that is characterized, at least in part, by idiopathic osteoporosis. In another embodiment, the an abnormal, progressive loss of body, organ or tissue mass. A osteoporosis is senile osteoporosis. wasting condition can occuras a result of a pathology Such as, In one embodiment, the methods of this invention are use for example, cancer, or an infection, or it can be due to a ful in treating diseases or disorders caused by, or associated physiologic or metabolic state, such as disuse deconditioning with a hormonal disorder, disruption or imbalance. In one 55 that can occur, for example, due to prolonged bed restor when embodiment, the hormonal disorder, disruption or imbalance a limb is immobilized. Such as in a cast. A wasting condition comprises an excess of a hormone. In another embodiment, can also be age associated. The loss of body mass that occurs the hormonal disorder, disruption or imbalance comprises a during a wasting condition can be characterized by a loss of deficiency of a hormone. In one embodiment, the hormone is total body weight, or a loss of organ weight such as a loss of a steroid hormone. In another embodiment, the hormone is an 60 bone or muscle mass due to a decrease in tissue protein. estrogen. In another embodiment, the hormone is an andro In one embodiment, "muscle wasting or “muscular wast gen. In another embodiment, the hormone is a glucocorticoid. ing, used herein interchangeably, refer to the progressive In another embodiment, the hormone is a cortico-steroid. In loss of muscle mass and/or to the progressive weakening and another embodiment, the hormone is Luteinizing Hormone degeneration of muscles, including the skeletal or Voluntary (LH). In another embodiment, the hormone is Follicle Stimu 65 muscles which control movement, cardiac muscles which lating Hormone (FSH). In another embodiment, the hormone control the heart, and Smooth muscles. In one embodiment, is any other hormone known in the art. In another embodi the muscle wasting condition or disorder is a chronic muscle US 8,309,603 B2 55 56 wasting condition or disorder. "Chronic muscle wasting is muscle mass and function. Further, increased lean body mass defined hereinas the chronic (i.e. persisting overalong period is associated with decreased morbidity and mortality for cer of time) progressive loss of muscle mass and/or to the chronic tain muscle-wasting disorders. In addition, other circum progressive weakening and degeneration of muscle. stances and conditions are linked to, and can cause muscle The loss of muscle mass that occurs during muscle wasting 5 wasting disorders. For example, studies have shown that in can be characterized by a muscle protein breakdown or deg severe cases of to chronic lowerback pain, there is paraspinal radation, by muscle protein catabolism. Protein catabolism muscle wasting. occurs because of an unusually high rate of protein degrada Muscle wasting and other tissue wasting is also associated tion, an unusually low rate of protein synthesis, or a combi with advanced age. It is believed that general weakness in old nation of both. Protein catabolism or depletion, whether 10 age is due to muscle wasting. As the body ages, an increasing caused by a high degree of protein degradation or a low proportion of skeletal muscle is replaced by fibrous tissue. degree of protein synthesis, leads to a decrease in muscle The result is a significant reduction in muscle power, perfor mass and to muscle wasting. The term “catabolism” has its mance and endurance. commonly known meaning in the art, specifically an energy In another embodiment, muscle wasting or other tissue burning form of metabolism. 15 wasting may be a result of alcoholism, and may be treated Muscle wasting can occur as a result of a pathology, dis with the compounds and compositions of the invention, rep ease, condition or disorder. In one embodiment, the pathol resenting embodiments thereof. ogy, illness, disease or condition is chronic. In another In one embodiment, the invention provides a use of SARM embodiment, the pathology, illness, disease or condition is compound as described herein or its prodrug, analog, isomer, genetic. In another embodiment, the pathology, illness, dis metabolite, derivative, pharmaceutically acceptable salt, ease or condition is neurological. In another embodiment, the pharmaceutical product, polymorph, crystal, impurity, N-OX pathology, illness, disease or condition is infectious. As ide, hydrate or any combination thereof for the treatment of a described herein, the pathologies, diseases, conditions or dis wasting disease, disorder or condition in a Subject. orders for which the compounds and compositions of the In one embodiment, the wasting disease, disorder or con present invention are administered are those that directly or 25 dition being treated is associated with chronic illness indirectly produce a wasting (i.e. loss) of muscle mass, that is This invention is directed to treating, in Some embodi a muscle wasting disorder. ments, any wasting disorder, which may be reflected in In one embodiment, muscle wasting in a subject is a result muscle wasting, weight loss, malnutrition, starvation, or any of the Subject having a muscular dystrophy; muscle atrophy; wasting or loss of functioning due to a loss of tissue mass. X-linked spinal-bulbar muscular atrophy (SBMA). 30 In some embodiments, wasting diseases or disorders, such The muscular dystrophies are genetic diseases character as cachexia; malnutrition, tuberculosis, leprosy, diabetes, ized by progressive weakness and degeneration of the skeletal renal disease, chronic obstructive pulmonary disease or Voluntary muscles that control movement. The muscles of (COPD), cancer, end stage renal failure, sarcopenia, emphy the heart and some other involuntary muscles are also affected sema, osteomalacia, or cardiomyopathy, may be treated by in some forms of muscular dystrophy. The major forms of 35 the methods of this invention, via the administration of a muscular dystrophy (MD) are: duchenne muscular dystro SARM compound as herein described, compositions com phy, myotonic dystrophy, becker muscular dystrophy, limb prising the same, with or without additional drugs, com girdle muscular dystrophy, facioscapulhumeral muscular pounds, or agents, which provide a therapeutic effect for the dystrophy, congenital muscular dystrophy, oculopharyngeal condition being treated. muscular dystrophy, distal muscular dystrophy and emery 40 In some embodiments, wasting is due to infection with dreifuss muscular dystrophy. enterovirus, Epstein-Barr virus, herpes Zoster, HIV, trypano Muscular dystrophy can affect people of all ages. Although Somes, influenze, coxsackie, rickettsia, trichinella, Schisto Some forms first become apparent in infancy or childhood, Soma or mycobacteria, and this invention, in Some embodi others may not appear until middle age or later. Duchenne ments, provides methods of treatment thereof. MD is the most common form, typically affecting children. 45 Cachexia is weakness and a loss of weight caused by a Myotonic dystrophy is the most common of these diseases in disease or as a side effect of illness. Cardiac cachexia, i.e. a adults. muscle protein wasting of both the cardiac and skeletal Muscle atrophy (MA) is characterized by wasting away or muscle, is a characteristic of congestive heart failure. Cancer diminution of muscle and a decrease in muscle mass. For cachexia is a syndrome that occurs in patients with Solid example, Post-Polio MA is a muscle wasting that occurs as 50 tumors and hematological malignancies and is manifested by part of the post-polio syndrome (PPS). The atrophy includes weight loss with massive depletion of both adipose tissue and weakness, muscle fatigue, and pain. lean muscle to mass. Another type of MA is X-linked spinal-bulbar muscular Cachexia is also seen in acquired immunodeficiency syn atrophy (SBMA also known as Kennedy's Disease). This drome (AIDS), human immunodeficiency virus (HIV)-asso disease arises from a defect in the androgen receptor gene on 55 ciated myopathy and/or muscle weakness/wasting is a rela the X chromosome, affects only males, and its onset is in tively common clinical manifestation of AIDS. Individuals adulthood. Because the primary disease cause is an androgen with HIV-associated myopathy or muscle weakness or wast receptor mutation, androgen replacement is not a current ing typically experience significant weight loss, generalized therapeutic strategy. There are some investigational studies or is proximal muscle weakness, tenderness, and muscle atro where exogenous testosterone propionate is being given to 60 phy. boost the levels of androgen with hopes of overcoming andro In one embodiment, "Hypogonadism’ is a condition gen insensitivity and perhaps provide an anabolic effect. Still, resulting from or characterised by abnormally decreased use of Supraphysiological levels of testosterone for Supple functional activity of the gonads, with retardation of growth mentation will have limitations and other potentially serious and sexual development. complications. 65 In some embodiments, the present invention provides a Sarcopenia is a debilitating disease that afflicts the elderly method for treating, reducing the incidence, delaying the and chronically ill patients and is characterized by loss of onset or progression, or reducing and/or abrogating the Symp US 8,309,603 B2 57 58 toms associated with a wasting disease in a subject. In one In one embodiment, the wasting disease is cachexia or embodiment, the method comprises administering to a Sub involuntary weight loss in a Subject. In another embodiment, ject a composition comprising a compound of this invention the present invention provides a method of treating, prevent and anti-cancer agent, an immunomodulating agent, an ing, inhibiting, reducing or Suppressing muscle wasting in a antidiabetic agent, an agent treating the cardiovascular sys Subject Suffering from a kidney disease. In one embodiment, tem, an agent treating the gastrointestinal system, an agent the present invention provides a method of treating, prevent treating the central nervous system, an agent treating a meta ing, inhibiting, reducing or Suppressing protein catabolism in bolic disease, an agent treating a wasting disease, a gene a Subject Suffering from a kidney disease or disorder, therapy agent, an agent treating the endocrine system, Vita In some embodiments, the present invention provides a 10 method for treating, reducing the incidence, delaying the mins, or a combination thereof. In some embodiments, wast onset or progression, or reducing and/or abrogating the Symp ing diseases comprise muscle injury, bed rest, immobility, toms associated with a hypogonadal state in a Subject. In one nerve injury, neuropathy, diabetic neuropathy, alcoholic neu embodiment, the present invention provides a method for ropathy, Subacute combined degeneration of the spinal cord, treating, reducing the incidence, delaying the onset or pro diabetes, rheumatoid arthritis, motor neurone diseases, Duch 15 gression, or reducing and/or abrogating the symptoms asso enne muscular dystrophy, carpal tunnel syndrome, chronic ciated with a pharmacotherapy induced hypogonadal state in infection, tuberculosis, Addison's disease, adult Sma, limb a subject. In some embodiments, hypogonadism is caused by muscle atrophy, alcoholic neuropathy, anorexia, anorexia treatments which alter the secretion of hormones from the sex nervosa, anorexia associated with cachexia, anorexia associ glands in both women and men. In some embodiments, ated with aging, back tumour, dermatomyositis, hip cancer, hypogonadism may be "primary or “central'. In primary inclusion body myositis, incontinentia pigmenti, intercostal hypogonadism, the ovaries or testes themselves do not func neuralgia, juvenile rheumatoid arthritis, Legg-Calve-Perthes tion properly. In some embodiments, hypogonadism may be disease, muscle atrophy, multifocal motor neuropathy, neph induced by Surgery, radiation, genetic and developmental rotic syndrome, osteogenesis imperfecta, post-polio Syn disorders, liver and kidney disease, infection, or certain drome, rib tumor, spinal muscularatrophy, reflex sympathetic 25 autoimmune disorders. In some embodiments, menopause is dystrophy syndrome, or Tay-Sachs. a form of hypogonadism. A wasting condition or disorder is defined herein as a In some embodiments, the present invention provides a condition or disorder that is characterized, at least in part, by method for treating, reducing the incidence, delaying the an abnormal, progressive loss of body, organ or tissue mass. A onset or progression, or reducing and/or abrogating the Symp wasting condition can occuras a result of a pathology Such as, 30 toms associated with a combination of diseases and/or disor for example, cancer, or it can be due to a physiologic or ders in a subject as described hereinabove. In one embodi metabolic state, such as disuse deconditioning that can occur, ment, the method comprises administering to a subject a for example, due to prolonged bed rest or when a limb is composition comprising a compound of this invention and an immobilized, such as in a cast, or with the occurrence of anti-cancer agent, an immunomodulating agent, an antidia multiple wounds, including, for example, amputation, as 35 betic agent, an agent treating the cardiovascular system, an occurs in diabetics, and other conditions, as will be appreci agent treating the gastrointestinal system, an agent treating ated by one skilled in the art. A wasting condition can also be the central nervous system, an agent treating a metabolic age associated. The loss of body mass that occurs during a disease, an agent treating a wasting disease, a gene therapy wasting condition can be characterized by a loss of total body agent, an agent treating the endocrine system, an agent treat weight, or a loss of organ weight Such as a loss of bone or 40 ing a dermatological disorder, an anti-infective agent, an muscle mass due to a decrease in tissue protein. agent treating the liver, an agent treating the kidney, Vitamins, In one embodiment, the terms “muscle wasting or “mus or a combination thereof. cular wasting, refer to the progressive loss of muscle mass It is to be understood that any method of this invention, as and/or to the progressive weakening and degeneration of herein described, encompasses the administration of a com muscles, including the skeletal or Voluntary muscles which 45 pound as herein described, or a composition comprising the control movement, cardiac muscles which control the heart, same, to the Subject, in order to treat the indicated disease, and Smooth muscles. In one embodiment, the muscle wasting disorder or condition. The methods as herein described each condition or disorder is a chronic muscle wasting condition or and/or all may further comprise administration of an addi disorder. “Chronic muscle wasting is defined herein as the tional therapeutic agent as herein described, and as will be chronic (i.e. persisting over along period of time) progressive 50 appreciated by one skilled in the art. loss of muscle mass and/or to the chronic progressive weak In some embodiments, the present invention provides a ening and degeneration of muscle. method for enhanced production Such as milk, sperm, or egg. The loss of muscle mass that occurs during muscle wasting In some embodiments, the present invention provides a can be characterized by a muscle protein breakdown or deg method for enhanced production of lean meats or eggs. In radation, by muscle protein catabolism. Protein catabolism 55 Some embodiments, the present invention provides a method occurs because of an unusually high rate of protein degrada for increased productivity of feeds or stud livestock, for tion, an unusually low rate of protein synthesis, or a combi example, increased sperm count, improved morphology of nation of both. Protein catabolism or depletion, whether sperm, etc. In some embodiments, the present invention pro caused by a high degree of protein degradation or a low vides a method for expanding the productive life of farm degree of protein synthesis, leads to a decrease in muscle 60 animals, for example, egg-laying hens, milk-producing cows, mass and to muscle wasting. The term “catabolism” has its etc., and/or enhanced herd health, for example, improved commonly known meaning in the art, specifically an energy immune clearance, stronger animals. burning form of metabolism. In another embodiment, the compounds of this invention Muscle wasting can occur as a result of pathology, disease, and compositions as described herein are useful in promoting condition or disorders, including disorders for treatment via 65 or speeding recovery following a Surgical procedure. the methods of this invention, Such as, for example, end stage In one embodiment, the present invention provides a use of renal failure. a compound as to described herein for reducing a fat mass in US 8,309,603 B2 59 60 a subject. In another embodiment the invention provides Such Accordingly, the compounds can be useful in methods of methods for use of the compound as described herein or its blocking adipogenesis, and/or altering stem cell differentia prodrug, analog, isomer, metabolite, derivative, pharmaceu tion, as described herein. tically acceptable salt, pharmaceutical product, polymorph, In another embodiment, this invention relates to a method crystal, impurity, N-oxide, hydrate or any combination of decreasing, Suppressing, inhibiting or reducing appetite of thereof, or a composition comprising the same. a Subject, comprising the step of administering to the Subject In one embodiment, the present invention provides a use of a compound as herein described and/or its analog, derivative, a compound as described herein for increasing a lean mass in isomer, metabolite, pharmaceutically acceptable salt, phar a subject. In another embodiment Such use comprises admin 10 maceutical product, hydrate, N-oxide, prodrug, polymorph, istration of a compound as described herein or its prodrug, crystal, or any combination thereof, in an amount effective to analog, isomer, metabolite, derivative, pharmaceutically decrease, Suppress, inhibit or reduce the appetite of the Sub acceptable salt, pharmaceutical product, polymorph, crystal, ject. impurity, N-oxide, hydrate or any combination thereof. In another embodiment, this invention relates to a method In one embodiment the Subject has a hormonal imbalance, 15 disorder, or disease. In another embodiment the Subject has of altering the body composition of a subject, comprising the menopause. step of administering to the Subject a compound as herein Example 4 demonstrates that a compound of formula (S-II) described and/or its analog, derivative, isomer, metabolite, is anabolic yet minimally androgenic, thus such compounds pharmaceutically acceptable salt, pharmaceutical product, may be useful in treating patient groups in which androgens hydrate, N-oxide, prodrug, polymorph, crystal, or any com were contraindicated in the past. Compound of formula (S-II) bination thereof, in an amount effective to alter the body was shown to stimulate muscle growth, whether in the pres composition of the Subject. In one embodiment, altering the ence or absence of testosterone while exerting anti-prolifera body composition comprises altering the lean body mass, the tive effects on the prostate, thus, in one embodiment, the fat free body mass of the subject, or a combination thereof. methods of this invention provide for restoring lost muscle 25 In another embodiment, this invention relates to a method mass in patients with sarcopenia or cachexia. of altering lean body mass or fat free body mass of a Subject, In one embodiment, the compounds as herein described comprising the step of administering to the Subject a com alter the levels of leptin in a subject. In another embodiment, pound as herein described and/or its analog, derivative, iso the compounds as herein described decrease the levels of mer, metabolite, pharmaceutically acceptable salt, pharma leptin. In another embodiment, the compounds as herein 30 ceutical product, hydrate, N-oxide, prodrug, polymorph, described increase the levels of leptin in a subject. Leptin is crystal, or any combination thereof, in an amount effective to known to have an effect on appetite on weight loss in obese alter the lean body mass or fat free body mass of the subject. mice, and thus has been implicated in obesity. In another embodiment, this invention relates to a method The compounds as herein described, in one embodiment, 35 of converting fat to lean muscle in a subject, comprising the affect circulating, or in another embodiment, tissue levels of step of administering to the Subject a compound as herein leptin. In one embodiment, the term level/s of leptin refers to described and/or its analog, derivative, isomer, metabolite, the serum level of leptin. As contemplated herein, the com pharmaceutically acceptable salt, pharmaceutical product, pounds of the present invention have an effect on leptin in hydrate, N-oxide, prodrug, polymorph, crystal, or any com vitro and in vivo. Leptin levels can be measured by methods 40 bination thereof, in an amount effective to convert fat to lean known to one skilled in the art, for example by commercially muscle in the Subject. available ELISA kits. In addition, Leptin levels may be deter It is to be understood that any use of any of the compounds mined in in vitro assays, or in in Vivo assays, by any method as herein is described may be used in the treatment of any known to a person skilled in the art. disease, disorder or condition as described herein, and repre Since leptin is implicated in controlling appetite, weight 45 loss, food intake, and energy expenditure, modulating and/or sents an embodiment of this invention. controlling the levels of leptin is a useful therapeutic The following examples are presented in order to more approach in treating preventing, inhibiting or reducing the fully illustrate the preferred embodiments of the invention. incidence of obesity in subjects suffering from obesity. They should in no way, however, be construed as limiting the Modulating the level of leptin can result in a loss of appetite, 50 broad scope of the invention. a reduction of food intake, and an increase in energy expen diture in the subject, and thus may contribute to the control EXAMPLES and treatment of obesity. The term “obesity' is defined, in one embodiment, as an increase in body weight beyond the limitation of skeletal and 55 Example 1 physical requirement, as the result of excessive accumulation offat in the body. Synthesis of (S) Enantiomer of Compound of The term “obesity-associated metabolic disorder” refers, Formula II in one embodiment, to a disorder which results from, is a consequence of, is exacerbated by or is secondary to obesity. 60 Non-limiting examples of Such a disorder are osteoarthritis, Type II diabetes mellitus, increased blood pressure, stroke, C and heart disease. 2NNaOHacetone In addition, androgens have recently been shown to be -- CO2H --- 65 O X involved in commitment of mesenchymal pluripotent cells N H into myogenic lineage and to block differentiation into adi H pogenic lineage (Singh et al., Endocrinology, 2003, Jul. 24). US 8,309,603 B2 61 62 -continued HZ, J=6.7 Hz, 1H, CH at the chiral center), 4.02 (d. J=11.4 Hz, Cy" 1H, CHH), 3.86 (d. J=11.4 Hz, 1H, CHH), 3.53-3.24 (m, v 4H, CH), 2.30-2.20 (m, 1H, CH), 2.04-1.72 (m,3H, CH, and N H CH), 1.56 (s. 2H, Me); C NMR (75 MHz, DMSO-d) & 5 167.3, 163.1,83.9, 57.2, 45.4, 37.8, 29.0, 22.9, 21.6; IR (KBr) O 3474, 1745 (C=O), 1687 (C=O), 1448, 1377, 1360, 1308, 1227, 1159, 1062 cm; C+124.5° (c=1.3, chloroform): Anal. Calcd. for CHBrNO: C, 41.24; H, 4.61; N, 5.34. (2R)-1-Methacryloylpyrrolidin-2-carboxylic Acid. D-Pro- 9 Found: C, 41.46; H, 4.64; N, 5.32. line, 14.93 g, 0.13 mol) was dissolved in 71 mL of 2N NaOH and cooled in an ice bath; the resulting alkaline solution was H diluted with acetone (71 mL). An acetone solution (71 mL) of O methacryloyl chloride (13.56 g., 0.13 mol) and 2N NaOH N 24% HBir solution (71 mL) were simultaneously added over 40 minto - - the aqueous solution of D-proline in an ice bath. The pH of the O HC OH mixture was kept at 10-11° C. during the addition of the S. Br (R)-3-bromo-2-hydroxy-2-ydroxy methacryloyl chloride. After stirring (3h, room temperature), H3C methylpropanoic acid the mixture was evaporated in vacuo at a temperature at 35-45° C. to remove acetone. The resulting solution was 20 washed with ethyl ether and was acidified to pH 2 with con- (2R)-3-Bromo-2-hydroxy-2-methylpropanoic Acid. A centrated HC1. The acidic mixture was saturated with NaCl mixture of bromolactone (18.5g, 71 mmol) in 300 mL of 24% and was extracted with EtOAc (100 mLX3). The combined HBr was heated at reflux for 1 h. The resulting solution was extracts were dried over NaSO filtered through Celite, and diluted with brine (200 mL), and was extracted with ethyl evaporated in vacuo to give the crude product as a colorless 2 acetate (100 mLX4). The combined extracts were washed oil. Recrystallization of the oil from ethyl ether and hexanes with saturated NaHCO to (100 mLx4). The aqueous solution afforded 16.2 g (68%) of the desired compound as colorless was acidified with concentrated HCl to pH=1, which, in turn, crystals: mp 102-103°C. (lit. 214 mp 102.5-103.5°C.); the was extracted with ethyl acetate (100 mLX4). The combined NMR spectrum of this compound demonstrated the existence organic Solution was dried over Na2SO4, filtered through of two rotamers of the title compound. "H NMR (300 MHz, Celite, and evaporated in vacuo to dryness. Recrystallization DMSO-d) & 5.28 (s) and 5.15 (s) for the first rotamer, 5.15 (s) from toluene afforded 10.2 g (86%) of the desired compound and 5.03 (s) for the second rotamer (totally 2H for both as colorless crystals: mp 107-109°C. (lit. 214 mp 109-1130 rotamers, vinyl CH2), 4.48-4.44 for the first rotamer, 4.24- C. for the S-isomer); H NMR (300 MHz, DMSO-d) & 3.63 4.20 (m) for the second rotamer (totally 1H for both rotamers, (d. J=10.1 Hz, 1H, CHH), 3.52 (d. J=10.1 Hz, 1H, CHH), CH at the chiral canter), 3.57-3.38 (m, 2H, CH,), 2.27-2.12 35 (s.3H, Me); IR (KBr)3434 (OH), 3300-2500 (COOH), (1H, CH), 1.97-1.72 (m, 6H, CH, CH, Me); C NMR (75 1730 (C=O), 1449, 1421, 1380, 1292, 1193, 1085 cm; MHz, DMSO-da) 8 for major rotamer 173.3, 169.1, 140.9, c.26+10.50 (c-2.6, MeOH); Anal. Calcd. for CHBrO, C, 116.4, 58.3, 48.7, 28.9, 24.7, 19.5: for minor rotamer 174.0, 26.25; H, 3.86. Found: C, 26.28; H, 3.75. 170.0, 1416, 115.2, 60.3, 45.9, 31.0, 22.3, 19.7: IR (KBr)

3437 (OH), 1737 (C=O), 1647 (CO, COOH), 1584, 1508, 40 1459, 1369, 1348, 1178 cm; C+80.8° (c=1, MeOH): Anal. Calcd. for CHNO: C, 59.00; H, 7.15: N, 7.65. SOCI THF/ Found: C, 59.13; H, 7.19; N, 7.61. O-5°C. 45 HC OH H (R)-3-bromo-2-hydroxy-2- O methylpropanoic acid

N H NBS DMF FC NH2 RT EtN/RT -- He O NC NC 55 O

(3R,8aR)-3-Bromomethyl-3-methyl-tetrahydro-pyrrolo 2.1-c. 14 oxazine-1,4-dione. A solution of NBS (23.5 g. FC N 2. Br 0.132 mol) in 100 mL of DMF was added dropwise to a stirred HC oH solution of the (methyl-acryloyl)-pyrrolidine (16.1 g, 88 mmol) in 70 mL of DMF under argon at room temperature, 60 R-19 and the resulting mixture was stirred 3 days. The solvent was removed in vacuo, and a yellow solid was precipitated. The Synthesis of (2R)-3-Bromo-N-4-cyano-3-(trifluorom Solid was suspended in water, stirred overnight at room tem- ethyl)phenyl-2-hydroxy-2-methylpropanamide. Thionyl perature, filtered, and dried to give 18.6 g (81%) (smaller chloride (46.02 g, 0.39 mol) was added dropwise to a cooled weight when dried ~34%) of the title compound as a yellow 65 solution (less than 4°C.) of R-18 (51.13 g, 0.28 mol) in 300 solid: mp 152-154° C. (lit. 214 mp 107-109° C. for the mL of THF under an argon atmosphere. The resulting mixture S-isomer); H NMR (300 MHz, DMSO-d) & 4.69 (dd, J=9.6 was stirred for 3 h under the same condition. To this was US 8,309,603 B2 63 64 added EtN (39.14g, 0.39 mol) and stirred for 20 min under Example 2 the same condition. After 20 min, 5-amino-2-cyanobenzotri fluoride (40.0 g, 0.21 mol), 400 mL of THF were added and Metabolic Stability of the Compounds of this then the mixture was allowed to stir overnight at room tem Invention perature. The solvent was removed under reduced pressure to give a solid which was treated with 300 mL of HO, extracted Metabolic stability assays were performed in order to with EtOAc (2x400 mL). The combined organic extracts assess the in vitro half-life of compounds of formula II when were washed with saturated NaHCO, solution (2x300 mL) incubated with human liver microsomes. The data generated and brine (300 mL). The organic layer was dried over MgSO was transformed to determine intrinsic clearance values. In a and concentrated under reduced pressure to give a solid, 10 separate experiment, permeability across human, intestinal which was purified from column chromatography using epithelial monolayers (Caco-2 cells) was used as a measure of CHCl/EtOAc (80:20) to give a solid. This solid was recrys intestinal permeability as well as an indicator of efflux poten tallized from CHC1/hexane to give 55.8 g (73.9%) of (2R)- tial. Caco-2 cells are often used as an early Screening Surro 3-Bromo-N-4-cyano-3-(trifluoromethyl)phenyl-2-hy gate for oral bioavailability. Microsomal half-life can be con 15 Verted to in vitro clearance values as a means to predict droxy-2-methylpropanamide (R-19) as a light-yellow solid. hepatic intrinsic clearance. Intrinsic clearance is defined as "H NMR (CDC1/TMS) & 1.66 (s.3H, CH,), 3.11 (s, 1H, the functional ability of the liver to metabolize a drug or other OH, 3.63 (d. J=10.8 Hz, 1H, CH), 4.05 (d. J=10.8 Hz, 1H, compound. CH), 7.85 (d. J–8.4 Hz, 1H, ArH), 7.99 (dd, J=2.1, 8.4 Hz, Materials and Methods: 1H, ArH), 8.12 (d. J=2.1 Hz, 1H, ArH), 9.04 (bs, 1H, NH). Metabolic Stability Measured in Human Liver Microsomes: Calculated Mass: 349.99, M-H 349.0. M. p. 124-126° C. Compound of formula S-II in this study was incubated at a final concentration of 0.6 uM. Microsome reactions were NC performed under either Phase I or “Phase I and II' conditions, where indicated. Compound stocks (10 mMACN) were ini 25 tially diluted to a concentration of 60LM (in 60% ACN/HO) -- resulting in a “working stock” solution of 100x. Human liver FC microsomes were utilized at a final concentration of 0.6 mg/ml. Duplicate wells were used for each time point (0, 6, R-19 10, 30, and 60 minutes). Reactions were carried out at 37°C. CN 30 in a shaking water bath, and the final concentration of solvent K2CO3 was kept constant at 0.6%. The final volume for each reaction Her2-propanol was 600 ul, comprised of 368 ul of 100 mMKPO, buffer, (pH HO F 7.4); 12.6 ul of HLM (from a 20 mg/ml stock); 6 Jul of 100x NC C “working stock' drug compound, and 126 ul of NRS “master O mix' solution. At each time point, 100 ul of reaction was

removed and added to a sample well containing 100 ul of FC NH O F ice-cold, 100% ACN (plus internal standards), to stop the OH reaction. The NRS “master mix' is a solution of glucose-6- phosphate dehydrogenase, NADP, MgCl2, and glucose-6- 40 phosphate, prepared per manufacturer's instructions (BD Synthesis of (S)-3-(4-chloro-3-fluorophenoxy)-N-(4-cy Biosciences, Waltham, Mass.). Each 6.0 ml stock of NRS ano-3-(trifluoromethyl)phenyl)-2-hydroxy-2-methylpro “master mix” solution contains 3.8 ml H0, 1.0 ml solution panamide. A mixture of bromoamide (2R)-3-bromo-N-4- “A” (Cat. #461220), and 0.2 ml solution “B” (Cat. #461200). cyano-3-(trifluoromethyl)phenyl-2-hydroxy-2- Human liver microsomes (lot #0610279, Xenotech Corp.) methylpropanamide, (R-19)2.0g, 5.70 mmol) and anhydrous 45 represented a pool of 60 donors. KCO (2.4g, 17.1 mmol) was heated to reflux for 2 hand Samples were centrifuged at 3,000 rpm for 10 minutes at 4 then concentrated under reduced pressure to give a solid. The C. to remove debris and precipitate protein. Approximately resulting solid was treated with 4-chloro-3-fluorophenol (1.3 160 ul of Supernatant was Subsequently transferred to a new g, 8.5 mmol) and anhydrous KCO (1.6 g., 11.4 mmol) in 50 sample block for analysis. The concentration of parent drug mL of 2-propanol and was heated to reflux for 3 h, then 50 remaining in each well (expressed as percent remaining ver concentrated under reduced pressure to give a solid. The sus Time 0, at the beginning of the reaction) was measured residue was treated with 100 mL of H2O and then extracted by LC/MS, as detailed below. The intrinsic clearance rates with EtOAc (2x100 mL). The combined EtOAc extracts were (CLint) were calculated from 0-60 minutes based on first washed with 10% NaOH (4x100 mL) and brine, successively. order decay kinetics as a is function of microsomal protein The organic layer was dried over MgSO and then concen 55 concentration. trated under reduced pressure to give an oil which was puri Permeability Across Human, Intestinal Epithelial Monolay fied by column chromatography using EtOAc/hexane (50:50) CS to give a solid which was recrystallized from CH2Cl2/hexane Permeability was measured in the Apical (pH 6.6) to Baso to give 1.7 g (70.5%) of (S)-3-(4-chloro-3-fluorophenoxy)- lateral (pH 7.4) and Basolateral (pH 7.4) to Apical (pH 6.6) N-(4-cyano-3-(trifluoromethyl)phenyl)-2-hydroxy-2-meth 60 directions across polarized, Caco-2 epithelial monolayers. ylpropanamide as a colorless Solid. Compound stocks (10 mM acetonitrile) were tested in the "H NMR (CDC1/TMS) & 1.60 (s.3H, CH), 3.28 (s, 1H, study at a final concentration of 10 uM. The concentration of OH), 3.98 (d. J=9.05 Hz, 1H, CH), 6.64-6.76 (m, 2H, ArH), drug in the receiver well was measured by LC/MS/MS using 7.30 (d. J=8.67 Hz, 1H, ArH), 7.81 (d. J=8.52 Hz, 1H, ArH), a standard curve. The apparent permeability (Papp) for each 7.96 (q, J=2.07, 8.52 Hz, 1H, ArH), 8.10 (d. J=2.07 Hz, 1H, 65 compound was calculated, and values (A-B) were classified ArH), 9.10 (s, 1H, NH). Calculated Mass: M-H 414.9. Mp: as: Poor (Papp: <1), Low (Papp 1-2), Medium (Papp 2-10) or 132-1349 C. High (Papp. 10). US 8,309,603 B2 65 66 Papp (x10 cm/sec)=Amount transported/ binding assay with 17o-methyl-H-Mibolerone (HMIB, (Area. Initial concentration*Time) PerkinElmer), a high affinity AR ligand. Recombinant andro gen receptor ligand binding domain (AR LBD) was com bined with HIMIB in buffer A (10 mM Tris, pH 7.4, 1.6 mM V-volume of the receptor chamber (ml, or cm) disodium EDTA, 0.26 M sucrose, 10 mM sodium molybdate, A=area of the membrane insert (cm) 1 mM PMSF) to determine the equilibrium dissociation con Ci-initial concentration of drug (uM) stant(K) of HIMIB. Protein was incubated with increasing Cf-final concentration of drug (uM) concentrations of HIMIB with and without a high concen Tassay time (seconds) tration of unlabeled MIB in order to determine total and Analytical Methods: 10 non-specific binding. Non-specific binding was then Sub All samples were analyzed on the MDS/Sciex API4000 Q tracted from total binding to determine specific binding and Trap system with electrospray ionization (ESI) in the positive graphed using SigmaPlot and non-linear regression for ligand or negative SIM mode, depending on the compounds. The binding curve with one site Saturation to determine the K of mobile phases were isocratic at 30% A (0.1% formic acid in MIB (1.84 nM). In addition, the concentration of HMIB water) and 70% B (0.1% formic acid in acetonitrile) with a 15 required to saturate ARLBD was determined to be 4 nM. flow rate of 0.4 mL/min. A Phenomenex Luna Phenyl-Hexyl Compound of formula S-II was tested in a range of con column (60x2.0 mm ID, 6L) was used. The injection volume centrations from 10' to 10 M using the conditions was 10 uL. The total run time per sample was 1.6 to 3.0 described above. Following incubation, plates were harvested minutes. and diclofenac were used as internal with GF/B filters on the Unifilter-96 Harvester (PerkinElmer) standards for the positive and negative mode, respectively. and washed three times with ice-cold buffer B (60 mM Tris, The percentage of parent drug compound remaining after pH 7.2). The filter plates were dried at RT, then 36 ul Micros each time point was determined relative to the initial mea cint-O cocktail was added to each well and sealed with sured concentration at the beginning of the reaction (To min). TopSeal-A. The receptor bound radioligand was then deter Data Analysis: mined with the TopCount(R) NXT Microplate Scintillation For half-life determination, data was fitted using GraphPad 25 Counter (PerkinElmer). Prism, v 4.03 with the non-linear regression equation “one The specific binding of HIMIB at each concentration of phase exponential decay defined as: SARM was determined by subtracting the nonspecific bind Y=Spanexp(-KX)+Plateau (decays to Plateau with a ing of HIMIB (determined by incubating with 10 Munla first-order rate constant, K). beled MIB), and expressed as a percentage of the specific “-K” is the slope of the curve. The halflife (minutes), T = 30 binding in the absence of each SARM. The concentration of ln 2/-K and is therefore defined as -0.693/K, a?k/a -0.693/ SARM required to decrease the HIMIB binding by 60%, slope). Intrinsic Clearance (ul/min/mg protein) is defined as: IC value, was determined by computer-fitting the data with CLint=0.693*(1/T)*(ml incubation/mg protein)*1000; SigmaPlot and non-linear regression with the standard curve This equation can also be expressed as (K*1000)/microsome four parameter logistic curve. The equilibrium binding con concentration. 35 stant (K) of each compound was then determined with the Results: following equation: TABLE 1. where K is the equilibrium dissociation constant of HIMIB Metabolic Stability Measured in Human Liver MicroSones: 40 (1.84 nM), and L is the concentration of HIMIB (4 nM). Results: HalfLife CL in HalfLife CL in Compound (minutes) (ulminimg) (minutes) (ulmin/mg) The binding affinity for compound of formula S-II was tested having Phase I Phase I Phase I+ Phase I+ in the radioligand binding assay with ARLBD as the receptor formula only only II II with K, (nM)=8.1. 45 S-II Stable <1 Stable <1 Example 4

The results had shown that in vitro half-life as determined Preclinical Anabolic and Androgenic Pharmacology from the microsomal assays demonstrated that compound of of Compound of Formula (S-II) in Intact and formula S-II under both phase I and phase I/II metabolic 50 Castrate Male Rats conditions. As shown in Table 1, the compound didn't exhibit an intrinsic clearance (CLint) value greater than 10 ul/min/ Anabolic and androgenic efficacy of compound of formula mg. It is generally accepted that an in vitro CLint value of less S-II administered by daily oral gavage were tested. The S-iso than 10 ul/min/mg protein represents favorable metabolic mer of compound of formula II was synthesized and tested as stability of the test compound. Compound of formula S-II 55 described herein exhibited low clearance in human liver microsomes. In con Materials and Methods: clusion, based on the data reported herein, compound of Male Sprague-Dawley rats weighing approximately 200 g formula S-II exhibited favorable metabolic stability profiles were purchased from Harlan Bioproducts for Science (India in vivo studies. napolis, Ind.). The animals were maintained on a 12-hlight/ 60 dark cycle with food (7012C LM-485 Mouse/Rat Sterilizable Example 3 Diet, Harlan Teklad, Madison, Wis.) and water available ad libitum. The animal protocol was reviewed and approved by Androgen Receptor Binding Affinity of SARMs the Institutional Animal Care and Use Committee of the Uni versity of Tennessee. The anabolic and androgenic activity of Materials and Methods: 65 the compound of formula S-II was studied in intact animals, The androgen receptor (AR) binding affinity of SARMs acutely orchidectomized (ORX) animals and chronically (9 was determined by using an in vitro competitive radioligand days) ORX rats. US 8,309,603 B2 67 68 The test article for this study was weighed and dissolved in Testosterone propionate (TP) and S-3-(4-acetylaminophe 10% DMSO (Fisher) diluted with PEG 300 (Acros Organics, noxy)-2-hydroxy-2-methyl-N-(4-nitro-3-trifluorometh NJ) for preparation of the appropriate dosage concentrations. ylphenyl) propionamide (S-4), maximally stimulated the The animals were housed in groups of 2 to 3 animals percage. levator ani muscle weight to 104% and 10.1%, respectively. Animals were randomly assigned to one of seven groups 5 These data show that compound of formula S-II exhibited consisting of 4 to 5 animals per group. Control groups (intact significantly greater efficacy and potency than either TP or and ORX) were administered vehicle daily. Compound of S-4. As a whole, these data show that compound of formula formula S-II was administered via oral gavage at doses of S-II is able to stimulate muscle growth in the presence or 0.01, 0.03, 0.1, 0.3, 0.75, and 1 mg/day to both intact and absence of testosterone while exerting anti-proliferative ORX groups. Where appropriate, animals were castrated on 10 day one of the study. Treatment with compound of formula effects on the prostate. These data show that that compound of S-II began nine days post ORX and was administered daily formula S-II restores lost muscle mass in patients with Sar via oral gavage for fourteen days. copenia or cachexia. Additionally, the antiproliferative The animals were sacrificed under anesthesia (ketamine? effects of compound of formula S-II on the prostate may Xyalzine, 87:13 mg/kg) and body weights were recorded. In 15 allow some to patient populations, in which androgens are addition, Ventral prostate, seminal vesicles, and levator ani currently contraindicated, access to anabolic agents. muscle were removed, individually weighed, normalized to Compound of formula S-II exhibited anabolic muscle/ body weight, and expressed as a percentage of intact control. prostate ratio in castrated rats of 4.10, 2.39, 2.28, 1.97, 1.53, Students T-test was used to compare individual dose groups 1.05 following doses of 0.01, 0.03, 0.1, 0.3, 0.75 and 1 to the intact control group. Significance was defined a priori mg/day, respectively. as a P-values 0.05. Ventral prostate and seminal vesicle weights were evaluated as a measure of androgenic activity, Pharmacology results following 1 mg/day of compound of whereas levator ani muscle weight was evaluated as a mea formula S-II exhibited that prostate weight was 110%+14% sure of anabolic activity. Blood was collected from the of intact control and levator ani muscle weight was abdominal aorta, centrifuged, and sera were frozen at-80°C. 25 129%+10% of intact control. Compound of formula S-II prior to determination of serum hormone levels. Serum maintained prostate weight following orchidectomy at luteinizing hormone (LH) and follicle stimulating hormone 123+22% of intact controls and levator ani muscle weight at (FSH) concentrations were determined. 129-14% of intact controls. A range of between 0.1 mg/day to Results: 0.3 mg/day of compound of formula S-II restored 100% of A series of dose-response studies in intact and castrated 30 levator ani muscle weight, while between 39 to 60% prostate rats in order to evaluate the potency and efficacy of compound weight was restored. of formula S-II in both androgenic (prostate and seminal vesicles) and anabolic (levator ani muscle) tissue was con ducted. In intact animals, compound of formula S-II treat Example 5 ment resulted in decreases in the weight of both prostate and 35 seminal vesicles while the levator ani muscle weight was significantly increased. Levator ani muscle weight following In Vitro CYP Inhibition Assay compound of formula S-II treatment were 100%+10%, 98%+7%, 110%-5%, 110%-5%, 125%+10%, and Materials and Methods: 129%+10% of intact controls following doses of 0.01, 0.03, 40 P450 enzyme inhibition was measured using human 0.1, 0.3, 0.75, and 1 mg/day, respectively. The prostate cDNA-expressed CYP3A4, 2D6, 2019, 209, and 1A2 weights were 117%+20%, 98%+15%, 82%+20%. 62%+5%, recombinant enzymes and fluorogenic Substrates (coumarin 107%+30%, and 110%+14% of intact controls following analogues) that are converted to fluorescent products. The doses of 0.01, 0.03, 0.1, 0.3, 0.75, and 1 mg/day, respectively. analogues utilized for each isoenzyme are as follows: 7-ben These results are significant since current androgen therapies 45 are contraindicated in some patient populations due to the Zyloxy-trifluoromethylcoumarin, (BFC) for 3A4; 3-2-(N.N- proliferative androgenic effects in prostate and breast tissues. diethyl-N-methyl amino) ethyl 7-methoxy-4-methylcou However, many patients in these populations could benefit marin, (AMMC) for 2D6; 3-cyano-7-ethoxycoumarin, from the anabolic actions of androgens in muscle and bone. (CEC) for 2C19 and 1A2; and 7-methoxy-4-trifluoro-meth Since compound of formula (S-II) exhibited tissue selective 50 ylcoumarin, (MFC) for 2C9. These substrates were utilized at anabolic effects, it may be possible to treat patient groups in a single concentration (either 50 uM or 75uM) at or near the which androgens were contraindicated in the past. apparent K, for each Substrate. Fluorescence intensity was In castrated, ORX animals, prostate weights following measured using a Wallac 1420 Victor Multi-label Counter compound of formula S-II treatment were 10%+3%, Model (Perkin-Elmer, Wellesley, Mass.), with an excitation 12%+3%, 26%.7%, 39%.6%, 60%.14%, 88%.16%, and 55 wavelength filter of 405 nm, and an emission filter of 460 nm 123%22% of intact controls following doses of 0, 0.01, (535 nm for the 3A4 and 2C9 substrates). Compound stocks 0.03, 0.1, 0.3, 0.75, and 1 mg/day, respectively (FIG. 2). (10 mM in a 4:1 ratio of acetonitrile. DMSO) were tested in Similarly, seminal vesicle weights were 11%+1%, 11%+1%, this study using an 8-point dose response curve in duplicate 11%+1%, 27%.14%, 58%+18%, 86%+12%, and 100%-8% (ranging from 0.15uM-20.0 uM). The concentration of aceto of intact controls following doses of 0, 0.01, 0.03, 0.1, 0.3, 60 nitrile was kept constant at 0.4%, and the reaction was carried 0.75, and 1 mg/day, respectively (FIG. 2). Significant out at 37° C. for 30 minutes. Averages (minus background) increases were seen in levator ani muscle weights in all dose groups, when compared to intact controls. The levator ani and ICso values were calculated. muscle weights were 48%+8%, 50%+5%, 62%+6%, Results: 89%+10%, 118%+6%. 134%-8% and 129%+14% of intact 65 The in vitro Screening results for potential drug-drug inter controls corresponding to 0, 0.01, 0.03, 0.1.0.3, 0.75, and 1.0 actions (DDI) of SARM compound of formula S-II is pre mg/day dose groups, respectively (FIG. 2). sented in Table 2: US 8,309,603 B2 69 70 TABLE 2 TABLE 4 CYP (P450) Inhibition. ICsa (IIM

Compound 3A4 2D6 2C19 2C9 1A2 ON S-II >2O 17.7 2.4 1.3 >2O

Example 6 10 Pharmacokinetics of Compound of Formula (S-II) in Compound R Dogs S-1 C-1 C-2 C 3 In order to determine the pharmacokinetics of compound 15 C-3 of formula S-II, the compound was administered to beagle C-4 dogs per orally, and circulating plasma levels, terminal elimi C-6 C-8 nation half-life (t), total body clearance (CL), terminal C-10 volume distribution (VZ) and absolute bioavailability (F 96) C-11 C-12 (Table 3) were determined. Table 3: C-13 C-14 C-17 C-18 Compound C-22 C S-II C-23 25 T12 (hr) 37 - 26.8 CL (mL/min kg) O36 0.12 VZ (mL/kg) 1266 352 F9% 72.59% Example 8

30 Effects of SARM Compounds on Growth Performance and Carcass Composition of Finishing Example 7 Pigs

Anabolic and Androgenic Activity of SARM Materials and Methods: Compounds in Intact and Castrated Male Rats 35 The effects of SARM as represented by compound of for mula S-II on growth performance and carcass composition of The in vivo pharmacological activity of each synthetic AR finishing pigs was examined. Forty crossbred barrows, (TR4x ligand (listed in Table 4, below) was examined in five male C22) with an initial weight of 209.4 lb were used for this 28-d Sprague-Dawley rats weighing approximately 200 g. Ani experiment. Pigs were blocked by weight and allotted to one mals were castrated via a scrotal incision under anesthesia 24 40 of four treatments with ten replicate pens per treatment. Pigs h before drug treatments and received daily Sc injections of were housed with one pig per pen in an environmentally the compound of interest at a dose rate of 1 mg/d for 14 d. All controlled finishing barn with 4 ftx4 ft slatted pens. compounds of interest were freshly dissolved in vehicle con All animals were fed a corn-soybean meal diet with 1% taining dimethylsulfoxide (5%, vol/vol) in polyethylene gly corn oil. For the is treated groups, appropriate quantities of a col300 before dose administration. An additional two groups 45 compound of formula S-II (referred to as SARM in the text of animals with or without castration received vehicle only and figures/tables of this example) were dissolved in 100 mL and served as castrated or intact control groups, respectively. of polysorbate (tween) 80 and diluted with 20 lbs of corn oil Animals were killed at the end of the treatment. Plasma prior to incorporation into the test diet. Final SARM concen samples were collected and stored at -80°C. for future use. trations were 1, 3, and 10 ppm. All animals were fed their 50 respective diets for the duration of the study. The test diets The to Ventral prostate, seminal vesicles, and levator ani contained 1.07% TID lysine. Prior to being placed on study, muscle were removed, cleared of extraneous tissue, and all pigs were fed a common corn-Soybean meal diet formu weighed. All organ weights were normalized to body weight lated to 0.75% TID lysine. and compared. The weights of prostate and seminal vesicles Pigs were allowed ad libitum access to feed and water. Pigs were used to evaluate androgenic activity, whereas the levator 55 and feeders were weighed on day 7, 14, 21, and 28 to calculate ani muscle weight was used as a measure of anabolic activity. average daily gain (ADG), average daily feed intake (ADFI), Ventral prostate weights in SARM treated castrated rats were and feed-to-gain ratio (F/G or F:G). Each pen served as an all (except C-6) statistically lowered than intact control (FIG. experimental unit for all statistical analysis. 4). Whereas levator ani weights in SARM castrated rats Pigs were slaughtered at the Kansas State University Meats treated with C-3, C-6, C-8, C-10, C-11, or C-18 demonstrated 60 Laboratory at the end of the study for collection of individual Support of muscle weight same as or in excess of intact carcass data. At 24 hours postmortem, the right side of the control (FIG.3). Further S-1, C-1, C-4, C-22, and C-23 dem carcass was frozen at -40°C. for approximately 1 h. After onstrated levator aniagonism of 75% of intact control (FIG. freezing, sides were ground once through a grinder equipped 3) vs. <25% of intact control in all of these cases for ventral with a 19 mm die, then mixed and ground through a second prostate (FIG. 4). This demonstrated tissue-selective anabo 65 grinder equipped with a 9.5 mm die. A Sub Sample of ground lism for a variety of SARMs of this invention. The results are carcass was then chemically analyzed to determine percent graphically depicted in FIGS. 3 and 4. ages of crude protein, moisture/dry matter, lipid, and ash. US 8,309,603 B2 71 72 Carcass measurements were done on the left side of the car demonstrated up to a 2% improvement in lean percent and cass, and a sample of lean and fat was taken from the longis pounds offat free lean. However, due to the variability in this simus at approximately the 10" rib. study, none of these measurements reached significance.

TABLE 6 Vehicle SARM, ppm Probability, Ps Parameter Control 1 3 10 Linear Quadratic Heart Wt, Ib O.99 1.04 1.07 1.01 O.92 O.08 Liver Wt, Ib 4.19 4.58 4.95 4.84 O.OS O.O2 Kidney Wt, lb 1.08 1.2 1.18 1.16 O.38 O.3 Dress, 9% 70.3 69.6 69.3 70 O.S2 O.23 1 rib fat, in 1.59 1.47 148 1.43 O.21 O.31 Last rib fat, in O.94 O.9 O.91 O.86 0.57 0.79 O.84 O.77 O.82 O.74 O16 0.75 10" rib fat, in O.86 O.78 O.8 O.74 O.35 0.7 LEA, in 7.27 7.26 7.22 7.59 O.09 O41 Lean, % 52.4 53.2 52.9 S4.3 O16 1 Fat free lean, Ib 104.2 106.9 105.5 106.7 O46 0.79 * A total of 40 barrows were used (carcass weight of 1991b)

The data were analyzed as a randomized complete-block Table 7 shows the complete data set for all parameters design. Analysis of variance was performed by using the which were collected in this Example. MIXED procedure of SAS. Linear and quadratic contrasts were used to evaluate the effects of increasing the level of the 25 TABLE 7 SARM on growth and carcass performance. SARM SARM SARM Results: Control (1 PPM) (3 PPM) (10 PPM) Although there were few statistical differences observed in ADG (Days 0-7) 2.51 2.89 2.37 2.17 the measured parameters due to the Small group sizes and 30 ADG (Days 8-14) 2.74 2.59 2.65 2.37 individual housing of experimental animals, we observed ADG (Days 15-21) 2.25 2.33 2.65 2.43 positive trends in several key parameters as shown in FIG. 5. ADG (Days 22-28) 2.77 3.15 3.15 3.03 Raw data are also summarized below. SARM increased aver ADG Total 2.57 2.74 2.70 2.50 ADFI (Days 0-7) 7.66 7.56 7.54 7.40 age daily gain (ADG) over the course of the study (FIG. 5A), ADFI (Days 8-14) 8.55 8.74 8.50 7.89 decreased feed to gain ratio (F:G) (FIG. 5B), increased fat 35 ADFI (Days 15-21) 7.99 8.33 8.61 7.78 free lean gain per day (FIG.5C), and dramatically increased ADFI (Days 22-28) 8.19 8.51 8.89 8.33 F/G (Days 0-7) 3.24 2.67 3.27 3.85 ADG for days 21-28 (FIG. 5D). F/G (Days 8-14) 3.54 3.52 3.30 3.46 Further, SARM treatment resulted in significantly F/G (Days 15-21) 3.70 3.88 3.40 3.33 increased Day 0-7F:G and decreased Day 8-14 average daily F/G (Days 22-28) 3.12 2.72 2.87 2.83 feed intake (ADFI). Table 5 shows the weekly as well as the FIG Total 3.2O 3.04 3.11 3.17 40 Gain (Days 0-7) 17.5 20.3 16.6 15.2 overall ADG, ADFI, and F:G data from the study. Gain (Days 8-14) 19.2 18.2 18.6 16.6 Gain (Days 15-21) 15.7 16.3 18.5 17.0 TABLE 5 Gain (Days 22-28) 19.4 22.0 22.0 21.2 initial Wt 209 209 209 209 Vehicle SARM, ppm Probability, Ps Live Wt 284 289 288 281 45 L side 97.6 98.0 97.7 96.8 Parameter Control 1 3 10 Linear Quadratic Rside 101 103 102 99.8 Carcass Wt 199 2O1 2OO 197 Day 0-7 ADG 2.51 2.89 2.37 2.17 O.O7 O.87 Dress % 70% 70% 69% 70% ADFI 7.66 7.56 7.54 7.4 0.37 O.8 strib fat 1.59 1.47 148 1.43 F:G 3.24 2.67 3.27 3.85 O.OS O.62 Last rib fat O.935 O.900 O.905 0.855 Day 8-14 ADG 2.74 2.59 2.65 2.37 0.14 O.94 50 Last lumbar fat O.835 0.770 O.815 O.740 ADFI 8. SS 8.74 8.5 7.89 0.04 O.67 Oth rib fat 0.855 O.78O O.800 0.735 F:G 3.54 3.52 3.3 3.46 0.83 0.57 LEA 7.27 7.26 7.22 7.59 Day 15-21 ADG 2.25 2.33 2.65 2.43 O.39 O.21 b FFL 104 107 106 107 ADFI 7.99 8.33 8.61 7.78 O.45 O.09 bs FFLD O.727 O822 0.774 O.814 F:G 3.7 3.88 3.4 3.33 O.19 O.71 Heart Wt O.988 1.04 1.07 1.01 Day 22-28 ADG 2.77 3.15 3.15 3.03 0.92 O.19 55 Liver Wt 4.19 4.58 4.95 4.84 ADFI 8.19 8.51 8.89 8.33 O.79 O.26 Kidney Wt 1.08 1.2O 1.18 1.16 F:G 3.12 2.72 2.87 2.83 0.95 O.24 Day 0-28 ADG 2.57 2.74 2.7 2.5 O.28 O.31 ADFI 8.1 8.28 8.38 7.85 0.29 0.27 The potential for using SARM (compound of formula S-II) F:G 3.2 3.04 3.11 3.17 O.S6 O45 to improve finishing characteristics in food-animals was dem 60 onstrated. Feeding the SARM greatly improved ADG and At the time of sacrifice, a carcass composition analysis was F:G while reducing carcass crude fat. Some of the greatest performed. The data from this analysis are presented in Table improvements in these parameters were noted as the lowest 6. Trends towards increased lean mass and decreased fat were dose (1 ppm). Lesser effects at high dose have been observed observed. SARM-treated pigs showed a 7 to 10% decrease in with other SARMs. Taken as a whole, these data support that first rib fat, 3 to 8% decrease in last rib fat, 2 to 11% decrease 65 SARM treatment would improve carcass composition and in last lumbar fat, 6 to 14% decrease in 10" rib fat, and up to growth performance which are key factors in the economics a 4% increase in loin eye area (LEA). Treated animals also of Swine production. US 8,309,603 B2 73 74 Example 9 170.0, 1416, 115.2, 60.3, 45.9, 31.0, 22.3, 19.7: IR (KBr) 3437 (OH), 1737 (C=O), 1647 (CO, COOH), 1584, 1508, Effects of SARM Compounds on Growth 1459, 1369, 1348, 1178 cm; C+80.8° (c=1, MeOH): Performance and Carcass Composition of Finishing Anal. Calcd. for CHNO: C, 59.00; H, 7.15: N, 7.65. Pigs Compared with Paylean R. 5 Found: C, 59.13; H, 7.19; N, 7.61. Further studies with lower doses, larger group sizes and a direct comparison to ractopamine are conducted. As a direct CO2H H competitor to ractopamine, the SARM treated animals dem XX O onstrate the highest ADGs in the fourth week of treatment. By 10 N H NBSDMF N the fourth week of treatment with ractopamine the animals RT are desensitized to the beta-agonist and the producers are seeing diminished returns in lean mass gain. Therefore, O Br longer treatment periods (>28 days) may be advantageous to H3C the producers when feeding a SARM than when feeding 15 ractopamine. (3R,8aR)-3-Bromomethyl-3-methyl-tetrahydro-pyrrolo Example 10 2.1-c. 14 oxazine-1,4-dione. A solution of NBS (23.5 g. 0.132 mol) in 100 mL of DMF was added dropwise to a stirred Synthesis of Compound of Formula S-XXIII solution of the (methyl-acryloyl)-pyrrolidine (16.1 g, 88 mmol) in 70 mL of DMF under argon at room temperature, Synthesis of (S) Enantiomer of Compound of Formula XXIII and the resulting mixture was stirred 3 days. The solvent was (FIG. 6) removed in vacuo, and a yellow solid was precipitated. The Solid was suspended in water, stirred overnight at room tem 25 perature, filtered, and dried to give 18.6 g (81%) (smaller C weight when dried ~34%) of the title compound as a yellow 2NNaOHacetone --- solid: mp 152-154° C. (lit. 214 mp 107-109° C. for the -- CO2H O-5° C. RT3 hrs O w S-isomer); H NMR (300 MHz, DMSO-d) & 4.69 (dd, J=9.6 N H HZ, J=6.7 Hz, 1H, CH at the chiral center), 4.02 (d. J=11.4 Hz, H 30 1H, CHH), 3.86 (d. J=11.4 Hz, 1H, CHH), 3.53-3.24 (m, 4H, CH), 2.30-2.20 (m, 1H, CH), 2.04-1.72 (m,3H, CH, and N H CH), 1.56 (s. 2H, Me); 'C NMR (75 MHz, DMSO-d) & 167.3, 163.1,83.9, 57.2, 45.4, 37.8, 29.0, 22.9, 21.6; IR (KBr) 3474, 1745 (C=O), 1687 (C=O), 1448, 1377, 1360, 1308, 35 1227, 1159, 1062 cm; C+124.5° (c=1.3, chloroform): Anal. Calcd. for CHBrNO: C, 41.24; H, 4.61; N, 5.34. Found: C, 41.46; H, 4.64; N, 5.32. (2R)-1-Methacryloylpyrrolidin-2-carboxylic Acid. D-Pro line, 14.93 g, 0.13 mol) was dissolved in 71 mL of 2N NaOH 40 and cooled in an ice bath; the resulting alkaline solution was H diluted with acetone (71 mL). An acetone solution (71 mL) of O methacryloyl chloride (13.56 g., 0.13 mol) and 2N NaOH N 24% HBir solution (71 mL) were simultaneously added over 40 minto O Reflux HC OHa. the aqueous solution of D-proline in an ice bath. The pH of the 45 mixture was kept at 10-11° C. during the addition of the O S. Br (R)-3-bromo-2-hydroxy-2- methacryloyl chloride. After stirring (3h, room temperature), H3C methylpropanoic acid the mixture was evaporated in vacuo at a temperature at 35-45° C. to remove acetone. The resulting solution was (2R)-3-Bromo-2-hydroxy-2-methylpropanoic Acid. A washed with ethyl ether and was acidified to pH 2 with con 50 mixture of bromolactone (18.5g, 71 mmol) in 300 mL of 24% centrated HC1. The acidic mixture was saturated with NaCl HBr was heated at reflux for 1 h. The resulting solution was and was extracted with EtOAc (100 mLX3). The combined diluted with brine (200 mL), and was extracted with ethyl extracts were dried over NaSO, filtered through Celite, and acetate (100 mLx4). The combined extracts were washed evaporated in vacuo to give the crude product as a colorless with saturated NaHCO (100 mLx4). The aqueous solution oil. Recrystallization of the oil from ethyl ether and hexanes 55 afforded 16.2 g (68%) of the desired compound as colorless was acidified with concentrated HCl to pH=1, which, in turn, crystals: mp 102-103°C. (lit. 214 mp 102.5-103.5°C.); the was extracted with ethyl acetate (100 mLx4). The combined NMR spectrum of this compound demonstrated the existence organic Solution was dried over Na2SO4, filtered through of two rotamers of the title compound. "H NMR (300 MHz, Celite, and evaporated in vacuo to dryness. Recrystallization DMSO-d) & 5.28 (s) and 5.15 (s) for the first rotamer, 5.15 (s) 60 from toluene afforded 10.2 g (86%) of the desired compound and 5.03 (s) for the second rotamer (totally 2H for both as colorless crystals: mp 107-109° C. (lit. 214 mp 109-113° rotamers, vinyl CH2), 4.48-4.44 for the first rotamer, 4.24 C. for the S-isomer); H NMR (300 MHz, DMSO-d) & 3.63 4.20 (m) for the second rotamer (totally 1H for both rotamers, (d. J=10.1 Hz, 1H, CHH), 3.52 (d. J=10.1 Hz, 1H, CHH), CH at the chiral center), 3.57-3.38 (m, 2H, CH), 2.27-2.12 1.35 (s.3H, Me); IR (KBr)3434 (OH), 3300-2500 (COOH), (1H, CH), 1.97-1.72 (m, 6H, CH, CH, Me); C NMR (75 65 1730 (0–0), 1449, 1421, 1380, 1292, 1193, 1085 cm; MHz, DMSO-d) & for major rotamer 173.3, 169.1, 140.9, C+10.5° (c=2.6, MeOH); Anal. Calcd. for CHBrO: C, 116.4, 58.3, 48.7, 28.9, 24.7, 19.5: for minor rotamer 174.0, 26.25; H, 3.86. Found: C, 26.28; H, 3.75. US 8,309,603 B2 75 -continued NC CN O

SOCI THF/O-59 C. FC NH O F (R)-3-bromo-2-hydroxy-2- Hd oH methylpropanoic acid

Synthesis of (S)-N-(4-cyano-3-(trifluoromethyl)phenyl)- 10 3-(4-cyano-3-fluorophenoxy)-2-hydroxy-2-methylpropana C Br mide. A mixture of bromoamide (2R)-3-bromo-N-4-cyano Hd OH 3-(trifluoromethyl)phenyl-2-hydroxy-2- R-18 methylpropanamide, R-19 (2.0 g, 5.70 mmol) and anhydrous KCO (2.4g, 17.1 mmol) in 50 mL of acetone was heated to FC NH2 15 EtN/RT reflux for 2 hand then concentrated under reduced pressure to -- Hs give a solid. The resulting solid was treated with 2-fluoro-4- hydroxybenzonitrile (1.2g, 8.5 mmol) and anhydrous KCO NC (1.6 g., 11.4 mmol) in 50 mL of 2-propanol and was heated to NC reflux for 3 h, then concentrated under reduced pressure to O give a solid. The residue was treated with 100 mL of HO and then extracted with EtOAc (2x100 mL). The combined FC N Br EtOAc extracts were washed with 10% NaOH (4x100 mL) H 2. HC OH and brine, Successively. The organic layer was dried over R-19 MgSO and then concentrated under reduced pressure to give 25 an oil which was crystallized from CHCl2/hexane to give 0.5 g (23%) of (S)-N-(4-cyano-3-(trifluoromethyl)phenyl)-3-(4- Synthesis of (2R)-3-Bromo-N-4-cyano-3-(trifluorom cyano-3-fluorophenoxy)-2-hydroxy-2-methylpropanamide ethyl)phenyl-2-hydroxy-2-methylpropanamide. Thionyl as a colorless Solid. chloride (46.02 g, 0.39 mol) was added dropwise to a cooled H NMR (CDC1/TMS) & 1.63 (s, 3H, CH), 3.34 (bs, solution (less than 4°C.) of R-18 (51.13 g, 0.28 mol) in 300 30 1H-OH), 4.08 (d. J=9.17 Hz, 1H, CH), 4.50 (d. J=9.17 Hz, mL of THF under an argon atmosphere. The resulting mixture 1H, CH), 6.74-6.82 (m, 2H, ArH), 7.50-7.55 (m, 1H, ArH), was stirred for 3 h under the same condition. To this was 7.81 (d. J=8.50 Hz, 1H, ArH), 7.97 (q, J=2.03, 8.50 Hz, 1H, added EtN (39.14g, 0.39 mol) and stirred for 20 min under ArH), 8.11 (d. J–2.03 Hz, 1H, ArH), 9.12 (s, 1H, NH). Cal the same condition. After 20 min, 5-amino-2-cyanobenzotri culated Mass: 407.1, M+Na" 430.0. Mp: 124-125° C. fluoride (40.0 g, 0.21 mol), 400 mL of THF were added and 35 FIG. 6 schematically depicts some embodiments of syn then the mixture was allowed to stir overnight at room tem thetic processes to obtain compound of formula S-XXIII. perature. The solvent was removed under reduced pressure to Example 11 give a solid which was treated with 300 mL of HO, extracted with EtOAc (2x400 mL). The combined organic extracts 40 Preclinical Anabolic and Androgenic Pharmacology were washed with saturated NaHCO, solution (2x300 mL) of Compound for Formula (S-XXIII) in Intact and and brine (300 mL). The organic layer was dried over MgSO Castrate Male Rats and concentrated under reduced pressure to give a solid which was purified from column chromatography using CH-Cl/ Anabolic and androgenic efficacy of compound of formula EtOAc (80:20) to give a solid. This solid was recrystallized 45 S-XXIII administered by daily oral gavage were tested. The from CHC1/hexane to give 55.8 g (73.9%) of (2R)-3- S-isomer of compound (XXIII) was synthesized and tested as bromo-N-4-cyano-3-(trifluoromethyl)phenyl-2-hydroxy described herein. 2-methylpropanamide (R-19) as a light-yellow solid. Materials and Methods: "H NMR (CDC1/TMS) & 1.66 (s.3H, CH), 3.11 (s, 1H, Male Sprague-Dawley rats weighing approximately 200 g OH), 3.63 (d. J=10.8 Hz, 1H, CH,), 4.05 (d. J=10.8 Hz, 1H, 50 were purchased from Harlan Bioproducts for Science (India CH), 7.85 (d. J=8.4 Hz, 1H, ArH), 7.99 (dd, J=2.1, 8.4 Hz, napolis, Ind.). The animals were maintained on a 12-hlight/ 1H, ArH), 8.12 (d. J=2.1 Hz, 1H, ArH), 9.04 (bs, 1H, NH). dark cycle with food (7012C LM-485 Mouse/Rat Sterilizable Calculated Mass: 349.99, M-H 349.0. M. p. 124-126° C. Diet, Harlan Teklad, Madison, Wis.) and water available ad libitum. The animal protocol was reviewed and approved by 55 the Institutional Animal Care and Use Committee of the Uni NC versity of Tennessee. The testarticle for this study was weighed and dissolved in 10% DMSO (Fisher) diluted with PEG 300 (Acros Organics, NJ) for preparation of the appropriate dosage concentrations. 60 The animals were housed in groups of 2 to 3 animals percage. Animals were randomly assigned to one of seven groups consisting of 4 to 5 animals per group. Control groups (intact and ORX) were administered vehicle daily. Compound of formula S-XXIII was administered via oral gavage at doses of 65 0.01, 0.03, 0.1, 0.3, 0.75, and 1 mg/day to both intact and ORX groups. Where appropriate, animals were castrated on day one of the study. Treatment with compound of formula US 8,309,603 B2 77 78 S-XXIII began nine days post ORX and was administered mally stimulated the levator ani muscle weight to 104% and daily via oral gavage for fourteen days. 101%, respectively, indicating the significantly enhanced The animals were sacrificed under anesthesia (ketamine? efficacy and potency of compound of formula S-XXIII. Taken Xyalzine, 87:13 mg/kg) and body weights were recorded. In together, these data show that compound of formula S-XXIII addition, Ventral prostate, seminal vesicles, and levator ani restores lost muscle mass, which in Some embodiments, finds muscle were removed, individually weighed, normalized to valuable application in patients with sarcopenia or cachexia, body weight, and expressed as a percentage of intact control. or other wasting diseases or disorders. Additionally, the anti Students T-test was used to compare individual dose groups proliferative effects of compound of formula S-XXIII on the to the intact control group. Significance was defined a priori prostate may allow some patient populations, in which andro as a P-values 0.05. Ventral prostate and seminal vesicle 10 gens are currently contraindicated, access to anabolic agents. weights were evaluated as a measure of androgenic activity, Emax values were obtained and were 147%+10%, whereas levator ani muscle weight was evaluated as a mea 188%+135%, and 147%+10% for prostate, seminal vesicles, sure of anabolic activity. Blood was collected from the and levator ani, respectively. The EDs in prostate, seminal abdominal aorta, centrifuged, and sera were frozen at-80°C. vesicles, and levator ani was 0.21+0.04, 0.2+0.04, and prior to determination of serum hormone levels. Serum 15 0.03+0.01 mg/day, respectively. These results are graphically luteinizing hormone (LH) and follicle stimulating hormone depicted in FIG. 9. (FSH) concentrations were determined. Results: Example 12 A series of dose-response studies in intact and castrated rats in order to evaluate the potency and efficacy of compound Synthesis of Compound XXIV of formula S-XXIII in both androgenic (prostate and seminal vesicles) and anabolic (levator ani muscle) tissue was con Synthesis of (S) Enantiomer of Compound of ducted. In intact animals, compound of formula S-XXIII Formula (XXIV) (FIG. 10) treatment resulted in decreases in the weight of both prostate and seminal vesicles while the levator ani muscle weight was 25 significantly increased. Levator ani muscle weight following compound of formula S-XXIII treatment were 116%+7%, 134%-8%, 134%-21%, 134%+11%, 142%+10%, and -- Cy" --2NNaOHacetone 147%+10% of intact controls, following treatment with 0.01, O N H O-5°C. RT3 hrs 0.03, 0.1, 0.3, 0.75, and 1.0 mg/day dose groups, respectively. 30 H The prostate weights were 98%+21%, 99%+8%, 85%+18%, 98%+22%, 126%-17%, and 126%-17% of intact controls, following treatment with 0.01, 0.03, 0.1, 0.3, 0.75, and 1 mg/day, respectively. Similarly seminal vesicle weight was N H 11.5%+12%, 109%-17%, 106%-13%, 121%.11%, 35 157%+5%, and 13.6%+3% of intact controls following treat ment with 0.01, 0.03, 0.1, 0.3, 0.75, and 1 mg/day, respec tively. The results are graphically presented in FIG. 7. These results are significant since current androgen therapies are contraindicated in Some patient populations due to the prolif 40 (2R)-1-Methacryloylpyrrolidin-2-carboxylic Acid. D-Pro erative androgenic effects in prostate and breast tissues. How line, 14.93 g, 0.13 mol) was dissolved in 71 mL of 2N NaOH ever, many patients in these populations could benefit from and cooled in an ice bath; the resulting alkaline solution was the anabolic actions of androgens in muscle and bone. Since diluted with acetone (71 mL). An acetone solution (71 mL) of compound of formula S-XXIII exhibited tissue selective ana methacryloyl chloride (13.56 g., 0.13 mol) and 2N NaOH bolic effects, it may be possible to treat patient groups in 45 solution (71 mL) were simultaneously added over 40 minto which androgens were contraindicated in the past. the aqueous solution of D-proline in an ice bath. The pH of the In castrated (ORX) animals, prostate weights following mixture was kept at 10-11° C. during the addition of the compound of formula S-XXIII treatment were 24%+4%, methacryloyl chloride. After stirring (3h, room temperature), 37%.9%, 50%.11%, 88%.16%, 132%.16%, and 118-12% the mixture was evaporated in vacuo at a temperature at of intact controls following doses of 0, 0.01, 0.03, 0.1, 0.3, 50 35-45° C. to remove acetone. The resulting solution was 0.75, and 1 mg/day, respectively. Similarly, seminal vesicle washed with ethyl ether and was acidified to pH 2 with con weights were 15%+2%, 25%+9%. 67%+20%, 113%+6%, centrated HC1. The acidic mixture was saturated with NaCl 155%+16%, and 160%+7% of intact controls, following and was extracted with EtOAc (100 mLX3). The combined doses of 0, 0.01, 0.03, 0.1, 0.3, 0.75, and 1 mg/day, respec extracts were dried over NaSO filtered through Celite, and tively. Significant increases were seen in levator ani muscle 55 evaporated in vacuo to give the crude product as a colorless weights of in all dose groups, when compared to intact con oil. Recrystallization of the oil from ethyl ether and hexanes trols. The levator ani muscle weights were 71%+4%, afforded 16.2 (68%) of the desired compound as colorless 101%-15%, 125%-20%, 126%.14%, 151.9%, and crystals: mp 102-103°C. (lit. 214 mp 102.5-103.5°C.); the 143-17% of intact controls corresponding to 0, 0.01, 0.03, NMR spectrum of this compound demonstrated the existence 0.1.0.3, 0.75, and 1.0 mg/day dose groups, respectively. The 60 of two rotamers of the title compound. "H NMR (300 MHz, results are graphically presented in FIG. 8. DMSO-d) & 5.28(s) and 5.15 (s) for the first rotamer, 5.15 (s) One unexpected finding was that administration of only and 5.03 (s) for the second rotamer (totally 2H for both 0.03 mg/day was able to fully restore levator ani muscle rotamers, vinyl CH), 4.48-4.44 for the first rotamer, 4.24 weight. 4.20 (m) for the second rotamer (totally 1H for both rotamers, Comparable administration of testosterone propionate 65 CH at the chiral canter), 3.57-3.38 (m, 2H, CH), 2.27-2.12 (TP) and S-3-(4-acetylaminophenoxy)-2-hydroxy-2-methyl (1H, CH), 1.97-1.72 (m, 6H, CH, CH, Me); C NMR (75 N-(4-nitro-3-trifluoromethylphenyl) propionamide, maxi MHz, DMSO-d) & for major rotamer 173.3, 169.1, 140.9, US 8,309,603 B2 79 80 116.4, 58.3, 48.7, 28.9, 24.7, 19.5: for minor rotamer 174.0, 170.0, 1416, 115.2, 60.3, 45.9, 31.0, 22.3, 19.7: IR (KBr) 3437 (OH), 1737 (C=O), 1647 (CO, COOH), 1584, 1508, 1459, 1369, 1348, 1178 cm; C+80.8° (c=1, MeOH): Anal. Calcd. for CHNO: C, 59.00; H, 7.15; N, 7.65. B SOCI THF/O-5°C. Found: C, 59.13; H, 7.19; N, 7.61. (R)-3-bromo-2-hydroxy-2- methylpropanoic acid R-18 CO2H H 10 s O N H NBS DMF N RT O C 2. Br O O w Hd oH 15 NC EtN/RT (3R,8aR)-3-Bromomethyl-3-methyl-tetrahydro-pyrrolo -- Ho 2.1-c. 14 oxazine-1,4-dione. A solution of NBS (23.5 g. C NH2 0.132 mol) in 100 mL of DMF was added dropwise to a stirred solution of the (methyl-acryloyl)-pyrrolidine (16.1 g, 88 NC mmol) in 70 mL of DMF under argon at room temperature, O and the resulting mixture was stirred 3 days. The solvent was removed in vacuo, and a yellow solid was precipitated. The C NH Br Solid was suspended in water, stirred overnight at room tem 25 OH perature, filtered, and dried to give 18.6 g (81%) (smaller weight when dried ~34%) of the title compound as a yellow solid: mp 152-154° C. (lit. 214 mp 107-109° C. for the Synthesis of (2R)-3-bromo-N-(3-chloro-4-cyanophenyl)- S-isomer); H NMR (300 MHz, DMSO-d) & 4.69 (dd, J=9.6 2-hydroxy-2-methylpropanamide. Thionyl chloride (7.8 g. HZ, J=6.7 Hz, 1H, CH at the chiral center), 4.02 (d. J=11.4 Hz, 30 65.5 mmol) was added dropwise to a cooled solution (less 1H, CHH), 3.86 (d. J=11.4 Hz, 1H, CHH), 3.53-3.24 (m, than 4° C.) of (R)-3-bromo-2-hydroxy-2-methylpropanoic 4H, CH,), 2.30-2.20 (m. 1H, CH), 2.04-1.72 (m,3H, CH, and acid (9.0 g, 49.2 mol) in 50 mL of THF under an argon CH), 1.56 (s. 2H, Me); C NMR (75 MHz, DMSO-d) & atmosphere. The resulting mixture was stirred for 3 hunder 167.3, 163.1,83.9, 57.2, 45.4, 37.8, 29.0, 22.9, 21.6; IR (KBr) the same condition. To this was added EtN (6.6 g. 65.5 mol) 3474, 1745 (C=O), 1687 (C=O), 1448, 1377, 1360, 1308, 35 and stirred for 20 minunder the same condition. After 20 min, 1227, 1159, 1062 cm; C+124.5° (c=1.3, chloroform): 4-amino-2-chlorobenzonitrile (5.0g, 32.8 mmol) and 100 mL Anal. Calcd. for CHBrNO: C, 41.24; H, 4.61; N, 5.34. of THF were added and then the mixture was allowed to stir Found: C, 41.46; H, 4.64; N, 5.32. overnight at room temperature. The solvent was removed 40 under reduced pressure to give a solid which was treated with

100 mL of HO, extracted with EtOAc (2x150 mL). The H combined organic extracts were washed with Saturated O NaHCO solution (2x100 mL) and brine (300 mL), succes N 24% HBir sively. The organic layer was dried over MgSO and concen -- O Reflux 45 trated under reduced pressure to give a solid which was puri w (R)-3-bromo-2-hydroxy-2- fied from column chromatography using EtOAc/hexane (50: methylpropanoic acid 50) to give 7.7 g (49.4%) of target compound as a brown solid. "H NMR (CDC1/TMS) & 1.7 (s, 3H, CH,), 3.0 (s, 1H, OH), 3.7 (d. 1H, CH), 4.0 (d. 1H, CH), 7.5 (d. 1H, ArH), 7.7 (2R)-3-Bromo-2-hydroxy-2-methylpropanoic Acid. A 50 mixture of bromolactone (18.5g, 71 mmol) in 300 mL of 24% (d. 1H, ArH), 8.0 (s, 1H, ArH), 8.8 (s, 1H, NH). MS: 342.1 HBr was heated at reflux for 1 h. The resulting solution was (M+23). Mp 129° C. diluted with brine (200 mL), and was extracted with ethyl acetate (100 mLx4). The combined extracts were washed NC with saturated NaHCO (100 mLx4). The aqueous solution 55 was acidified with concentrated HCl to pH=1, which, in turn, was extracted with ethyl acetate (100 mLx4). The combined -- organic Solution was dried over Na2SO4, filtered through C Celite, and evaporated in vacuo to dryness. Recrystallization from toluene afforded 10.2 g (86%) of the desired compound 60 as colorless crystals: mp 107-109° C. (lit. 214 mp 109-113° CN K2CO3 C. for the S-isomer); H NMR (300 MHz, DMSO-d) & 3.63 --- (d. J=10.1 Hz, 1H, CHH), 3.52 (d. J=10.1 Hz, 1H, CHH), 2-propanol 1.35 (s.3H, Me); IR (KBr)3434 (OH), 3300-2500 (COOH), HO 1730 (C=O), 1449, 1421, 1380, 1292, 1193, 1085 cm; 65 C+10.5° (c=2.6, MeOH); Anal. Calcd. for CHBrO: C, 26.25; H, 3.86. Found: C, 26.28; H, 3.75. US 8,309,603 B2 81 82 -continued S-XXIV began nine days post ORX and was administered NC CN daily via oral gavage for fourteen days. O The animals were sacrificed under anesthesia (ketamine?

Xyalzine, 87: 13 mg/kg) and body weights were recorded. In C NH O addition, Ventral prostate, seminal vesicles, and levator ani muscle were removed, individually weighed, normalized to He on body weight, and expressed as a percentage of intact control. Students T-test was used to compare individual dose groups Synthesis of (S)-N-(3-chloro-4-cyanophenyl)-3-(4-cy to the intact control group. Significance was defined a priori anophenoxy)-2-hydroxy-2-methylpropanamide. A mixture 10 as a P-values 0.05. Ventral prostate and seminal vesicle of bromoamide (2.0 g. 6.3 mmol), anhydrous KCO (2.6 g. weights were evaluated as a measure of androgenic activity, 18.9 mmol) in 50 mL of acetone was heated to reflux for 2 h whereas levator ani muscle weight was evaluated as a mea and then concentrated under reduced pressure to give a solid. sure of anabolic activity. Blood was collected from the The resulting solid was treated with 4-cyanophenol (1.1 g, 9.5 abdominal aorta, centrifuged, and sera were frozen at-80°C. prior to determination of serum hormone levels. Serum mmol) and anhydrous KCO (1.7g, 12.6 mmol) in 50 mL of luteinizing hormone (LH) and follicle stimulating hormone 2-propanol was heated to reflux for 3 hand then concentrated (FSH) concentrations were determined. under reduced pressure to give a solid. The residue was Results: treated with 100 mL of H2O and then extracted with EtOAc A series of dose-response studies in intact and castrated (2x100 mL). The combined EtOAc extracts were washed rats in order to evaluate the potency and efficacy of compound with 10% NaOH (4x100 mL) and brine, successively. The of formula S-XXIV in both androgenic (prostate and seminal organic layer was dried over MgSO and then concentrated vesicles) and anabolic (levator ani muscle) tissue was con under reduced pressure to give an oil which was purified by ducted. In intact animals, compound of formula S-XXIV column chromatography using EtOAc/hexane (50:50) to give treatment resulted in decreases in the weight of both prostate a solid. The solid was recrystallized from CHCl/hexane to 25 and seminal vesicles while the levator ani muscle weight was give 1.4 g (61.6%) of (S)-N-(3-chloro-4-cyanophenyl)-3-(4- significantly increased. Levator ani muscle weight following cyanophenoxy)-2-hydroxy-2-methylpropanamide as a color compound of formula S-XXIV treatment were 107%+5%, less solid. 103%+7%,97%.7%, 103%-5%, 118%+7%, and 118%+7% "H NMR (CDC1/TMS) & 1.61 (s, 3H, CH), 3.25 (s, of intact controls following doses of 0.01, 0.03, 0.1.0.3, 0.75, 1H-OH), 4.06 (d. J=9.15 Hz, 1H, CH), 4.50 (d. J=9.15 Hz, 30 and 1 mg/day, respectively. The prostate weights were 1H, CH), 6.97-6.99 (m, 2H, ArH), 7.53-7.59 (m, 4H, ArH), 103%-10%, 99%+10%, 58%-10%, 58%-15%, 65%-20%, 7.97 (d. J=2.01 Hz, 1H, ArH), 8.96 (s, 1H, NH). Calculated and 77%+23% of intact controls following doses of 0.01, Mass: 355.1, M+Nat 378.0. Mp: 103-105° C. 0.03, 0.1, 0.3, 0.75, and 1 mg/day, respectively. These results are significant since current androgen therapies are contrain Example 13 35 dicated in some patient populations due to the proliferative androgenic effects in prostate and breast tissues. However, Preclinical Anabolic and Androgenic Pharmacology many patients in these populations could benefit from the of S-XXIV in Intact and Castrate Male Rats anabolic actions of androgens in muscle and bone. Since compound of formula S-XXIV exhibited tissue selective ana Anabolic and androgenic efficacy of compound of formula 40 bolic effects, it may be possible to treat patient groups in S-XXIV administered by daily oral gavage were tested. The which androgens were contraindicated in the past. S-isomer of compound S-XXIV was synthesized and tested In castrated, ORX animals, prostate weights following as described herein. compound of formula S-XXIV treatment were 12%+2%, Materials and Methods: 17%6%, 31%+3%, 43%-15%, 54%-17%, 58%-10%, and Male Sprague-Dawley rats weighing approximately 200 g 45 73%+12% of intact controls following doses of 0, 0.01, 0.03, were purchased from Harlan Bioproducts for Science (India 0.1.0.3, 0.75, and 1 mg/day, respectively (FIG.11). Similarly, napolis, Ind.). The animals were maintained on a 12-hlight/ seminal vesicle weights were 10%+2%, 10%+3%, 13%+4%, dark cycle with food (7012C LM-485 Mouse/Rat Sterilizable 21%+6%. 43%-8%, 51%+9%, and 69%-14% of intact con Diet, Harlan Teklad, Madison, Wis.) and water available ad trols following doses of 0, 0.01, 0.03, 0.1, 0.3, 0.75, and 1 libitum. The animal protocol was reviewed and approved by 50 mg/day, respectively (FIG. 11). Significant increases were the Institutional Animal Care and The anabolic and andro seen in levatorani muscle weights of in all dose groups, when genic activity of compound of formula S-XXIV in intact compared to intact controls. The levator ani muscle weights animals was tested, as well as a dose response evaluation in were 40%-5%, 52%-8%, 67%:.9%, 98%+10%, acutely orchidectomized (ORX) animals. Regenerative 103%+12%, 105%+12% and 110%-17% of intact controls effects of the compound of formula S-XXIV in chronically (9 55 corresponding to 0, 0.01, 0.03, 0.1, 0.3, 0.75, and 1.0 mg/day days) ORX rats was similarly evaluated. dose groups, respectively (FIG. 11). The test article for this study was weighed and dissolved in Testosterone propionate (TP) and S-3-(4-acetylaminophe 10% DMSO (Fisher) diluted with PEG 300 (Acros Organics, noxy)-2-hydroxy-2-methyl-N-(4-nitro-3-trifluorometh NJ) for preparation of the appropriate dosage concentrations. ylphenyl) propionamide (S-4), maximally stimulated the The animals were housed in groups of 2 to 3 animals percage. 60 levator ani muscle weight to 104% and 10.1%, respectively. Animals were randomly assigned to one of seven groups These data show that compound of formula S-XXIV exhib consisting of 4 to 5 animals per group. Control groups (intact ited significantly greater efficacy and potency than either TP and ORX) were administered vehicle daily. Compound of or S-4. As a whole, these data show that compound of formula formula S-XXIV was administered via oral gavage at doses of S-XXIV is able to stimulate muscle growth in the presence or 0.01, 0.03, 0.1, 0.3, 0.75, and 1 mg/day to both intact and 65 absence of testosterone while exerting anti-proliferative ORX groups. Where appropriate, animals were castrated on effects on the prostate. These data show that the compound of day one of the study. Treatment with compound of formula formula S-XXIV restores lost muscle mass in patients with US 8,309,603 B2 83 84 sarcopenia or cachexia. Additionally, the antiproliferative MHz, DMSO-d) & for major rotamer 173.3, 169.1, 140.9, effects of the compound of formula S-XXIV on the prostate 116.4, 58.3, 48.7, 28.9, 24.7, 19.5: for minor rotamer 174.0, may allow some patient populations, in which androgens are 170.0, 1416, 115.2, 60.3, 45.9, 31.0, 22.3, 19.7: IR (KBr) currently contraindicated, access to anabolic agents. 3437 (OH), 1737 (0–0), 1647 (CO, COOH), 1584, 1508, Anabolic ratios were derived comparing muscle/prostate 5 1459, 1369, 1348, 1178 cm; C+80.8° (c=1, MeOH): weight in castrated rats. Values obtained were 3.02, 2.13. Anal. Calcd. for CHNO: C, 59.00; H, 7.15: N, 7.65. 2.27, 1.90, 1.83 and 1.51 following doses of 0.01, 0.03, 0.1, Found: C, 59.13; H, 7.19; N, 7.61. 0.3, 0.75 and 1 mg/day, respectively. Animals receiving 1 mg/day of compound of formula 10 S-XXIV exhibited a prostate weight of 77%+23% and levator CO2H H ani muscle weight of 118%+7% of intact control values, O N H NBSDMF N respectively. Compound of formula S-XXIV maintained -e- prostate weight following orchidectomy at 73+12% of intact RT yO controls and levator ani muscle weight at 110+17% of intact O O X Br controls. A derived dose of 0.1 mg/day of compound of for 15 mula S-XXIV would restore levator ani muscle weight to HC 100%, while such dose would only restore 43-15% prostate weight. (3R,8aR)-3-Bromomethyl-3-methyl-tetrahydro-pyrrolo 2.1-c. 14 oxazine-1,4-dione. A solution of NBS (23.5 g. Example 14 2O 0.132 mol) in 100 mL of DMF was added dropwise to a stirred solution of the (methyl-acryloyl)-pyrrolidine (16.1 g, 88 Synthesis of Compound of Formula S-XXV mmol) in 70 mL of DMF under argon at room temperature, and the resulting mixture was stirred 3 days. The solvent was Synthesis of (S) Enantiomer of Compound of Formula XXV removed in vacuo, and a yellow solid was precipitated. The (FIG. 12) 25 Solid was suspended in water, stirred overnight at room tem perature, filtered, and dried to give 18.6 g (81%) (smaller weight when dried ~34%) of the title compound as a yellow solid: mp 152-154° C. (lit. 214 mp 107-109° C. for the

-- Cy" -es2NNaOHacetone 30 S-isomer); H NMR (300 MHz, DMSO-d) & 4.69 (dd, J=9.6 O N H O-5°C. RT3 hrs HZ, J=6.7 Hz, 1H, CH at the chiral center), 4.02 (d. J=11.4 Hz, H 1H, CHH), 3.86 (d. J=11.4 Hz, 1H, CHH), 3.53-3.24 (m, 4H, CH,), 2.30-2.20 (m. 1H, CH), 2.04-1.72 (m,3H, CH, and CH), 1.56 (s. 2H, Me); C NMR (75 MHz, DMSO-d) & N H 35 167.3, 163.1,83.9, 57.2, 45.4, 37.8, 29.0, 22.9, 21.6; IR (KBr) 3474, 1745 (C=O), 1687 (C=O), 1448, 1377, 1360, 1308, 1227, 1159, 1062 cm; C+124.5° (c=1.3, chloroform): Anal. Calcd. for CHBrNO: C, 41.24; H, 4.61; N, 5.34. Found: C, 41.46; H, 4.64; N, 5.32. 40

(2R)-1-Methacryloylpyrrolidin-2-carboxylic Acid. D-Pro line, 14.93 g, 0.13 mol) was dissolved in 71 mL of 2N NaOH H and cooled in an ice bath; the resulting alkaline solution was O N 24% HBir diluted with acetone (71 mL). An acetone solution (71 mL) of He methacryloyl chloride (13.56 g., 0.13 mol) and 2N NaOH 45 O Reflux H3C OH solution (71 mL) were simultaneously added over 40 minto O V Br (R)-3-bromo-2-hydroxy-2- the aqueous solution of D-proline in an ice bath. The pH of the methylpropanoic acid mixture was kept at 10-11° C. during the addition of the HC methacryloyl chloride. After stirring (3h, room temperature), the mixture was evaporated in vacuo at a temperature at 50 (2R)-3-Bromo-2-hydroxy-2-methylpropanoic Acid. A 35-45° C. to remove acetone. The resulting solution was mixture of bromolactone (18.5g, 71 mmol) in 300 mL of 24% washed with ethyl ether and was acidified to pH 2 with con HBr was heated at reflux for 1 h. The resulting solution was centrated HC1. The acidic mixture was saturated with NaCl diluted with brine (200 mL), and was extracted with ethyl and was extracted with EtOAc (100 mLX3). The combined acetate (100 mLx4). The combined extracts were washed extracts were dried over NaSO, filtered through Celite, and 55 with saturated NaHCO (100 mLx4). The aqueous solution evaporated in vacuo to give the crude product as a colorless was acidified with concentrated HCl to pH=1, which, in turn, oil. Recrystallization of the oil from ethyl ether and hexanes was extracted with ethyl acetate (100 mLx4). The combined afforded 16.2 g (68%) of the desired compound as colorless organic Solution was dried over Na2SO4, filtered through crystals: mp 102-103°C. (lit. 214 mp 102.5-103.5°C.); the Celite, and evaporated in vacuo to dryness. Recrystallization NMR spectrum of this compound demonstrated the existence 60 from toluene afforded 10.2 g (86%) of the desired compound of two rotamers of the title compound. "H NMR (300 MHz, as colorless crystals: mp 107-109° C. (lit. 214 mp 109-113° DMSO-d) & 5.28 (s) and 5.15 (s) for the first rotamer, 5.15 (s) C. for the S-isomer); H NMR (300 MHz, DMSO-d) & 3.63 and 5.03 (s) for the second rotamer (totally 2H for both (d. J=10.1 Hz, 1H, CHH), 3.52 (d. J=10.1 Hz, 1H, CHH), rotamers, vinyl CH2), 4.48-4.44 for the first rotamer, 4.24 1.35 (s.3H, Me); IR (KBr)3434 (OH), 3300-2500 (COOH), 4.20 (m) for the second rotamer (totally 1H for both rotamers, 65 1730 (C=O), 1449, 1421, 1380, 1292, 1193, 1085 cm; CH at the chiral canter), 3.57-3.38 (m, 2H, CH), 2.27-2.12 C+10.5° (c=2.6, MeOH); Anal. Calcd. for CHBrO: C, (1H, CH), 1.97-1.72 (m, 6H, CH, CH, Me); C NMR (75 26.25; H, 3.86. Found: C, 26.28; H, 3.75. US 8,309,603 B2 85 86 -continued NC

Br SOCI THF/O-59 C. He HC OH FC (R)-3-bromo-2-hydroxy-2- HC methylpropanoic acid (S)-N-(4-cyano-3-(trifluoromethyl)phenyl)-3-(4- cyanophenoxyl)-2-hydroxy-2-methylpropanamide

10 C 2. Br Synthesis of (S)-N-(4-Cyano-3-(trifluoromethyl)phenyl)- Hd oH 3-(4-cyanophenoxy)-2-hydroxy-2-methylpropanamide. A mixture of bromoamide (2R)-3-Bromo-N-4-cyano-3-(trif FC NH2 luoromethyl)phenyl-2-hydroxy-2-methylpropanamide, 50 EtN/RT 15 -- s g, 0.14 mol), anhydrous KCO (59.04 g., 0.43 mol), 4-cy anophenol (25.44 g., 0.21 mol) in 500 mL of 2-propanol was NC heated to reflux for 3 hand then concentrated under reduced NC pressure to give a solid. The resulting residue was treated with O 500 mL of HO and then extracted with EtOAc (2x300 mL). The combined EtOAc extracts were washed with 10% NaOH FC Br (4x200 mL) and brine. The organic layer was dried over N 2. O MgSO and then concentrated under reduced pressure to give an oil which was treated with 300 mL of ethanol and an 25 activated carbon. The reaction mixture was heated to reflux Synthesis of (2R)-3-Bromo-N-4-cyano-3-(trifluorom for 1 h and then the hot mixture was filtered through Celite. ethyl)phenyl-2-hydroxy-2-methylpropanamide. Thionyl The filtrate was concentrated under reduced pressure to give chloride (46.02 g, 0.39 mol) was added dropwise to a cooled an oil. This oil was purified by column chromatography using solution (less than 4°C.) of (R)-3-bromo-2-hydroxy-2-meth CHC1/EtOAc (80:20) to give an oil which was crystallized ylpropanoic acid (51.13 g, 0.28 mol) in 300 mL of THF under 30 from CHC1/hexane to give 33.2 g (59.9%) of (S)-N-(4- an argon atmosphere. The resulting mixture was stirred for 3 cyano-3-(trifluoromethyl)phenyl)-3-(4-cyanophenoxy)-2- hunder the same condition. To this was added EtsN (39.14g, hydroxy-2-methylpropanamide as a colorless solid (a cotton 0.39 mol) and stirred for 20 min under the same condition. After 20 min, 5-amino-2-cyanobenzotrifluoride (40.0g, 0.21 type). mol), 400 mL of THF were added and then the mixture was H NMR (CDC1/TMS) & 1.63 (s, 3H, CH), 3.35 (s, allowed to stir overnight at room temperature. The solvent 35 1H-OH), 4.07 (d. J=9.04 Hz, 1H, CH), 4.51 (d. J=9.04 Hz, was removed under reduced pressure to give a solid which 1H, CH), 6.97-6.99 (m, 2H, ArH), 7.57-7.60 (m, 2H, ArH), was treated with 300 mL of HO, extracted with EtOAc 7.81 (d. J=8.55 Hz, 1H, ArH), 7.97 (dd, J=1.95, 8.55 Hz, 1H, (2x400 mL). The combined organic extracts were washed ArH), 8.12 (d. J=1.95 Hz, 1H, ArH), 9.13 (bs, 1H, NH). with saturated NaHCO, solution (2x300 mL) and brine (300 Calculated Mass: 389.10, M-H 388.1. Mp: 92-94° C. mL). The organic layer was dried over MgSO and concen 40 trated under reduced pressure to give a solid which was puri Example 15 fied from column chromatography using CHC1/EtOAc (80: 20) to give a solid. This solid was recrystallized from CHCl2/ Androgenic & Anabolic Activity in Intact and ORX hexane to give 55.8 g (73.9%) of (2R)-3-bromo-N-4-cyano Rats of Compound of Formula S-XXV 3-(trifluoromethyl)phenyl-2-hydroxy-2- 45 methylpropanamide as a light-yellow solid. Materials and Methods: "H NMR (CDC1/TMS) & 1.66 (s.3H, CH,), 3.11 (s, 1H, Male Sprague-Dawley rats weighing approximately 200 g OH), 3.63 (d. J=10.8 Hz, 1H, CH,), 4.05 (d. J=10.8 Hz, 1H, were purchased from Harlan Bioproducts for Science (India CH), 7.85 (d. J–8.4 Hz, 1H, ArH), 7.99 (dd, J=2.1, 8.4 Hz, 50 napolis, Ind.). The animals were maintained on a 12-hlight/ 1H, ArH), 8.12 (d. J=2.1 Hz, 1H, ArH), 9.04 (bs, 1H, NH). dark cycle with food (7012C LM-485 Mouse/Rat Sterilizable Calculated Mass: 349.99, M-H 349.0. M. p. 124-126° C. Diet, Harlan Teklad, Madison, Wis.) and water available ad libitum. The animal protocol was reviewed and approved by the Institutional Animal Care and Use Committee of the Uni NC 55 versity of Tennessee. Anabolic and androgenic activity of compound of formula S-XXV in intact animals was evalu ated, and the dose response in acutely orchidectomized FC -- (ORX) animals was evaluated as well. Regenerative effects of HC OH Compound III in chronically (9 days) ORX rats were also (2R)-3-bromo-N-4-cyano-3- 60 assessed. (trifluoromethyl)phenyl]-2-hydroxy-2- The compound was weighed and dissolved in 10% DMSO methylpropanamide (Fisher) diluted with PEG 300 (Acros Organics, NJ) for CN preparation of the appropriate dosage concentrations. The K2CO3 He animals were housed in groups of 2 to 3 animals per cage. 2-propanol 65 Intact and ORX animals were randomly assigned to one of HO seven groups consisting of 4 to 5 animals per group. Control groups (intact and ORX) were administered vehicle daily. US 8,309,603 B2 87 88 Compound of formula S-XXV was administered via oral prostate, seminal vesicles, and levator ani, respectively. The gavage at doses of 0.01, 0.03, 0.1, 0.3, 0.75, and 1 mg/day to EDs in prostate, seminal vesicles, and levator ani was both intact and ORX groups. 0.09+0.07, 0.17+0.05, and 0.02+0.01 mg/day, respectively. Castrated animals (on day one of the study) were randomly It will be appreciated by a person skilled in the art that the assigned to dose groups (4-5 animals/group) of 0.01, 0.03. present invention is not limited by what has been particularly 0.1, 0.3, 0.75, and 1 mg/day, for dose-response evaluation. shown and described hereinabove. Rather, the scope of the Dosing began nine days post ORX and was administered invention is defined by the claims that follow: daily via oral gavage for fourteen days. The animals were What is claimed is: sacrificed under anesthesia (ketamine/xyalzine, 87:13 1. A feed composition for an animal comprising a com mg/kg) after a 14-day dosing regimen, and body weights were 10 pound of formula IIIA: recorded. In addition, Ventral prostate, seminal vesicles, and levator ani muscle were removed, individually weighed, nor malized to body weight, and expressed as a percentage of intact control. Student's T-test was used to compare indi vidual dose groups to the intact control group. Significance was defined a priori as a P-values 0.05. As a measure of androgenic activity, Ventral prostate and seminal vesicle weights were evaluated, whereas levator ani muscle weight was evaluated as a measure of anabolic activity. Blood was collected from the abdominal aorta, centrifuged, and sera were frozen at -80° C. prior to determination of serum hor wherein mone levels. Serum luteinizing hormone (LH) and follicle Z is NO, CN, C1, F, Br, I, H, COR, COOH or CONHR: stimulating hormone (FSH) concentrations were determined. Y is CF, alkoxy, alkyl, hydroxyalkyl, alkylaldehyde, formyl, H, F, Br, Cl, I, CN or Sn(R): RESULTS 25 R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, CHF, CF, CFCF, aryl, phenyl, halogen, alkenyl or Prostate weights following compound of formula S-XXV OH: treatment were 111%+21%, 88%-15%, 77%-17%, R is H, F, Cl, Br, I, CH, CF, OH, CN, NO, alkyl, 71%+16%, 71%-10%, and 87%-13% of intact controls fol arylalkyl, NH, NHR, N(R) or SR; lowing doses of 0.01, 0.03, 0.1, 0.3, 0.75, and 1 mg/day, 30 respectively. Similarly, seminal vesicle weights decreased to R is H, F, Cl, Br, I, CN, NO, COR, COOH, CONHR, CF, 94%.9%, 77%.11%, 80%–9%, 73%+12%, 77%+10%, and or Sn(R): 88%+14% of intact controls following doses of 0.01, 0.03, Q is H, alkyl, halogen, CF, CN, C(R), Sn(R), N(R), 0.1, 0.3, 0.75, and 1 mg/day, respectively. Significant NHCONHR, NHCOOR, OCONHR, CONHR, NHC SCH NHCSCF, NHCSR, OH, OCOR or SR; increases were seen in levator ani muscle weights of sham 35 n is an integer of 1-4; and animals, however, in all dose groups, when compared to intact m is an integer of 1-3: controls. The levator ani muscle weights were 120%+12%, or its optical isomer, pharmaceutically acceptable salt, 116%.7%, 128%.7%, 134%.7%, 125%.9%, and or any combination thereof. 146%-17% of intact controls corresponding to 0.01, 0.03, 2. The feed composition of claim 1, wherein said animal is 0.1, 0.3, 0.75, and 1.0 mg/day dose groups, respectively. 40 a feedlot animal, a beef cattle or a finishing livestock. Compound of formula S-XXV partially maintained pros 3. The feed composition of claim 2, wherein said feedlot tate weight following orchidectomy. Prostate weight in animal is a pig or avain; or wherein said finishing livestock is vehicle treated ORX controls decreased to 5% +1% of intact a pig or poultry. controls. At doses of 0.01, 0.03, 0.1.0.3, 0.75, and 1.0 mg/day, 4. The feed composition of claim 1, wherein said compo compound of formula S-XXV maintained prostate weights at 45 sition comprises between about 0.10-50 ppm of said com 8%+2%, 20%-5%, 51%-19%, 56%.9%, 80%.28%, and pound of formula IIIA. 74+12.5% of intact controls, respectively. In castrated con 5. The feed composition of claim 4, wherein said compo trols, seminal vesicle weight decreased to 13%+2% of intact sition comprises 0.10 ppm, 1 ppm, 3 ppm, or 10 ppm of said controls. Compound of formula S-XXV partially maintained compound of formula IIIA. seminal vesicle weights in ORX animals. Seminal vesicle 50 6. The feed composition of claim 1, wherein said compo weights from drug treated animals were 12%+4%, 17%+5%, sition further comprises a beta-adrenergic agonist. 35%+10%, 61%-15%, 70%-14%, and 80%.6% of intact 7. The feed composition of claim 6, wherein said beta controls, following doses of 0.01, 0.03, 0.1, 0.3, 0.75, and 1.0 adrenergic agonist is Ractopamine hydrochloride (Pay mg/day, respectively. In ORX controls the levator ani muscle weight decreased to 55%+7% of intact controls. We observed 55 leanR). an anabolic effect in the levator ani muscle of compound of 8. The feed composition of claim 1, wherein said com formula S-XXV treated animals. Compound of formula pound is a compound of formula I: S-XXV fully maintained levator ani muscle weights at doses 0.1 mg/day. Doses 0.1 mg/day resulted in significant (I) increases in levator ani weight compared to that observed in 60 intact controls. Levatorani muscle weights as a percentage of intact controls were 59%+6%, 85%-9%, 112%-10%, 122%+16%, 127+12%, and 129.66+2% for the 0.01, 0.03, 0.1.0.3, 0.75, and 1.0 mg/day dose groups, respectively. E. and EDso values were determined in each tissue by nonlinear 65 regression analysis in WinNonlinP) and presented in FIG. 13. E values were 83%-25%, 85%+11%, and 13.1%+2% for US 8,309,603 B2 89

wherein Q is alkyl, F, Cl, Br, I, CF, CN, C(R) Sn(R), N(R), NHCONHR, NHCOOR, OCONHR, CONHR, NHC SCH NHCSCF, NHCSR, NHSOCH, NHSOR, OR, COR, OCOR, OSOR, SOR, SR; and R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, CHF. CF, CFCF, aryl, phenyl, F, Cl, Br, I, alkenyl or OH. 9. The feed composition of claim 8, wherein said com pound is:

10

15

25 12. A method of affecting the carcass composition of an animal comprising administering a compound of formula IIIA:

30

10. A pharmaceutical composition for an animal compris 35 ing a compound of formula IIIA:

(IIIA) wherein 40 Z is NO, CN, C1, F, Br, I, H, COR, COOH or CONHR: Y is CF, alkoxy, alkyl, hydroxyalkyl, alkylaldehyde, formyl, H, F, Br, C1, I, CN or Sn(R): R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, CHF, CF, CFCF, aryl, phenyl, halogen, alkenyl or 45 OH: R is H, F, Cl, Br, I, CH, CF, OH, CN, NO, alkyl, wherein arylalkyl, NH, NHR, N(R), or SR; Z is NO, CN, C1, F, Br, I, H, COR, COOH or CONHR: R is H, F, Cl, Br, I, CN, NO, COR, COOH, CONHR, CF Y is CF, alkoxy, alkyl, hydroxyalkyl, alkylaldehyde, or Sn(R): formyl, H, F, Br, Cl, I, CN or Sn(R): 50 Q is H, alkyl, halogen, CF, CN, C(R), Sn(R), N(R), R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, NHCONHR, NHCOOR, OCONHR, CONHR, NHC SCH NHCSCF, NHCSR, OH, OCOR or SR; CHF, CF, CFCF, aryl, phenyl, halogen, alkenyl or n is an integer of 1-4; and OH: m is an integer of 1-3: R is H, F, Cl, Br, I, CH, CF, OH, CN, NO, alkyl, 55 or its optical isomer, pharmaceutically acceptable salt, arylalkyl, NH, NHR, N(R) or SR; or any combination thereof. R is H, F, Cl, Br, I, CN, NO, COR, COOH,CONHR, CF, 13. The method of claim 12, wherein said animal is a or Sn(R): feedlot animal, a beef cattle or a finishing livestock. Q is H, alkyl, halogen, CF, CN, C(R) Sn(R), N(R), 14. The method of claim 13, wherein said feedlot animal is NHCONHR, NHCOOR, OCONHR, CONHR, NHC 60 a pig or avian; or wherein said finishing livestock is a pig or SCH NHCSCF, NHCSR, OH, OCOR or SR; poultry. n is an integer of 1-4; and 15. The method of claim 12, wherein the carcass compo m is an integer of 1-3: sition is affected by increasing the lean mass, reducing the fat or its optical isomer, pharmaceutically acceptable salt or mass, and/or reducing percent fat mass of said animal. any combination thereof. 65 16. The method of claim 12, wherein affecting the carcass 11. The pharmaceutical composition of claim 10, wherein composition comprises increasing the growth performance in said compound is: said animal. US 8,309,603 B2 91 92 17. The method of claim 12, wherein said method com 27. The method of claim 15, wherein said method com prises feeding said animal a daily feed, wherein said feed prises feeding said animal a daily feed wherein said feed comprises said compound of formula IIIA. comprises said compound of formula IIIA. 18. The method of claim 12, wherein said method com 28. The method of claim 15, wherein said method com prises feeding said animal a daily feed wherein said feed prises feeding said animal a daily feed wherein said feed comprises a compound of formula IIIA and a beta-adrenergic comprises a compound of formula IIIA and a beta-adrenergic agonist. agonist. 19. The method of claim 18, wherein said beta-adrenergic agonist is Ractopamine hydrochloride (PayleanR). 29. The method of claim 28, wherein said beta-adrenergic 20. The method of claim 12, wherein said administering 10 agonist is Ractopamine hydrochloride (PayleanR). comprises topical administration or administering said com 30. The method of claim 15, wherein said administering pound via an implant. comprises topical administration or administering said com 21. The method of claim 20, wherein said implant com pound via an implant. prises a controlled release implant. 15 31. The method of claim 30, wherein said implant com 22. The method of claim 20, wherein said topical admin prises a controlled release implant. istration comprises administering a topical spray, a topical 32. The method of claim 30, wherein said topical admin cream or a topical ointment, or any combination thereof. istration comprises administering a topical spray, a topical 23. The method of claim 12, wherein said compound is a cream or a topical ointment, or any combination thereof. compound of formula I: 33. The method of claim 15, wherein said compound is a compound of formula I: (I) NC (I) 25 NC . NH . C NH 30 wherein Q, is alkyl, F, Cl, Br, I, CF, CN, C(R), Sn(R), N(R), NHCONHR, NHCOOR, OCONHR, CONHR, NHC wherein Q is alkyl, F, Cl, Br, I, CF, CN, C(R) Sn(R), SCH NHCSCF, NHCSR, OCOR or SR; and R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, CHF. N(R), NHCONHR, NHCOOR, OCONHR, CONHR, NHC 35 SCH NHCSCF, NHCSR, OCOR or SR; and CF, CFCF, aryl, phenyl, F, Cl, Br, I, alkenyl or OH. R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, CHF 24. The method of claim 23, wherein said compound is: CF, CFCF, aryl, phenyl, F, Cl, Br, I, alkenyl or OH. 34. The method of claim 33, wherein said compound is: Cl, 40 Cl,

45

CN 50

O

55

60

25. The method of claim 15, wherein said animal is a feedlot animal, a beef cattle or a finishing livestock. 26. The method of claim 25, wherein said feedlot animal is 65 35. A method of increasing feed efficiency of an animal a pig or avian; or wherein said finishing livestock is a pig or comprising administering a compound represented by the poultry. structure of formula IIIA: US 8,309,603 B2 93 94 45. The method of claim 44, wherein said compound is: (IIIA)

Cl,

wherein 10 Z is NO, CN, C1, F, Br, I, H, COR, COOH or CONHR: Y is CF, alkoxy, alkyl, hydroxyalkyl, alkylaldehyde, formyl, H, F, Br, C1, I, CN or Sn(R): R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, CHF, CF, CFCF, aryl, phenyl, halogen, alkenyl or 15 OH: R is H, F, Cl, Br, I, CH, CF, OH, CN, NO, alkyl, arylalkyl, NH, NHR, N(R) or SR; R is H, F, Cl, Br, I, CN, NO, COR, COOH,CONHR, CF, or Sn(R): Q is H, alkyl, halogen, CF, CN, C(R) Sn(R), N(R), NHCONHR, NHCOOR, OCONHR, CONHR, NHC SCH NHCSCF, NHCSR, OH, OCOR or SR; n is an integer of 1-4; and 25 m is an integer of 1-3: or its optical isomer, pharmaceutically acceptable salt, or any combination thereof. 46. The method of claim 35, wherein said increased effi 36. The method of claim 35, wherein said animal is a ciency comprises increasing the average daily gain (ADG) feedlot animal, a beef cattle or a finishing livestock. 30 and/or decreasing the feed to gain (F:G) ratio of said animal. 37. The method of claim 36, wherein said feedlot animal is 47. The method of claim 46, wherein said animal is a a pig or avian, or wherein said finishing livestock is a pig or feedlot animal, a beef cattle or a finishing livestock. poultry. 48. The method of claim 47, wherein said feedlot animal is 38. The method of claim 35, wherein said method com a pig or avian; or wherein said finishing livestock is a pig or prises feeding said animal a daily feed wherein said feed 35 poultry. comprises said compound of formula IIIA. 49. The method of claim 46, wherein said method com 39. The method of claim 35, wherein said method com prises feeding said animal a daily feed wherein said feed prises feeding said animal a daily feed wherein said feed comprises said compound of formula IIIA. comprises a compound of formula IIIA and a beta-adrenergic 50. The method of claim 46, wherein said method com agonist. 40 prises feeding said animal a daily feed wherein said feed 40. The method of claim 39, wherein said beta-adrenergic comprises a compound of formula IIIA and a beta-adrenergic agonist is Ractopamine hydrochloride (PayleanR). agonist. 41. The method of claim 35, wherein said administering 51. The method of claim 50, wherein said beta-adrenergic comprises topical administration or administering said com agonist is Ractopamine hydrochloride (PayleanR). pound via an implant. 45 52. The method of claim 46, wherein said administering 42. The method of claim 41, wherein said implant com comprises topical administration or administering said com prises a controlled release implant. pound via an implant. 43. The method of claim 41, wherein said topical admin 53. The method of claim 52, wherein said implant com istration comprises administering a topical spray, a topical prises a controlled release implant. cream or a topical ointment, or any combination thereof. 50 54. The method of claim 52, wherein said topical admin 44. The method of claim 35, wherein said compound is a istration comprises administering a topical spray, a topical compound of formula I: cream or a topical ointment, or any combination thereof. 55. The method of claim 46, wherein said compound is a compound of formula I: (I) 55 NC (I) NC . NH 60 . C NH wherein Q, is alkyl, F, Cl, Br, I, CF, CN, C(R), Sn(R), N(R), NHCONHR, NHCOOR, OCONHR, CONHR, NHC SCH NHCSCF, NHCSR, OCOR or SR; and 65 wherein Q is alkyl, F, Cl, Br, I, CF, CN, C(R) Sn(R), R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, CHF N(R), NHCONHR, NHCOOR, OCONHR, CONHR, NHC CF, CFCF, aryl, phenyl, F, Cl, Br, I, alkenyl or OH. SCH NHCSCF, NHCSR, OCOR or SR; and US 8,309,603 B2 95 96 R is alkyl, haloalkyl, dihaloalkyl, trihaloalkyl, CHF, CHF -continued CN CF, CFCF, aryl, phenyl, F, Cl, Br, I, alkenyl or OH. 56. The method of claim 55, wherein said compound is: O

NC Cl,

NC CN.

FC F 10 FC NC CN, Ol.

FC F 15 UNITED STATES PATENT AND TRADEMARK OFFICE CERTIFICATE OF CORRECTION PATENT NO. : 8,309,603 B2 Page 1 of 6 APPLICATIONNO. : 12/730094 DATED : November 13, 2012 INVENTOR(S) : James T. Dalton et al. It is certified that error appears in the above-identified patent and that said Letters Patent is hereby corrected as shown below:

In the Claims:

Col. 88, Claim 3 should read: -3. The feed composition of claim 2, wherein said feedlot animal is a pig or avian; or wherein Said finishing livestock is a pig or poultry.--

And not

-3. The feed composition of claim 2, wherein said feedlot animal is a pig or avain; or wherein Said finishing livestock is a pig or poultry.--

Col. 89, Claim 9 should read: --9. The feed composition of claim 1, wherein said compound is: N DO O OC F. exH OH f

NC N

F. sixH or F

NC CN

sixHy

Signed and Sealed this Twenty-third Day of July, 2013 * g -éceez. Yafax-6 * ass/ é« Teresa Stanek Rea Acting Director of the United States Patent and Trademark Office CERTIFICATE OF CORRECTION (continued) Page 2 of 6 U.S. Pat. No. 8,309.603 B2

NC CN O F. I six C1 C HC oH .--

And not

--9. The feed composition of claim 8, wherein said compound is: N. C

F. sixIIC YoHQ F

N N

FC sixHC OEI

N N

N 1 *ott O

DOl O O FC sixH “ol

Col. 91, Claim 24 should read: --24. The method of claim 12, wherein said compound is: N C C Cl FC sixH, OH NC CN

FC H.C.> oil F N N DO O O C sixI O OT NC N DOl O FC N Hyoh Or CERTIFICATE OF CORRECTION (continued) Page 3 of 6 U.S. Pat. No. 8,309.603 B2

And not

--24. The method of claim 23, wherein said compound is: N C

F. sixHC OH NC N . N s |CC H OEI N N O O HCMoH>

Col. 92, Claim 34 should read: --34. The method of claim 15, wherein said compound is: NC C FC: six F tic H N N FC six F a OE N N O C H "of six3. O N CN

FC N 3 c ol

And not

--34. The method of claim 33, wherein said compound is: CERTIFICATE OF CORRECTION (continued) Page 4 of 6 U.S. Pat. No. 8,309.603 B2

N DO O CC C sixHC1 °oH F N CN O |CC FC sixHY ot E

N CN DO O O sixII, oh O NC (N

F. sixH^ oil .--

Col. 94, Claim 45 should read: --45. The method of claim 35, wherein said compound is: N CL

FCD.C.-C N F NC cN

s'.DO-C F. sixH CII F N CN C six C HC oH O N CN F. IC O - JC H OH

And not

--45. The method of claim 44, wherein said compound is: CERTIFICATE OF CORRECTION (continued) Page 5 of 6 U.S. Pat. No. 8,309.603 B2

N

F. sixII, oh F N CN O

C sixrHC oth C N N DO O O sixIIC OI O NC N O

F. laH. *o- C .--

Col. 95, Claim 56 should read: --56. The method of claim 46, wherein said compound is: NC

F,C sixH OH F NC CN O CC F. HC1> oil F N CN CDO six C (C OH O N N O

O --CHe ol

And not

--56. The method of claim 55, wherein said compound is: CERTIFICATE OF CORRECTION (continued) Page 6 of 6 U.S. Pat. No. 8,309.603 B2

NC C

F. sixH O F NC N O CC O la-C,H, °o N N O O C N it c1 on O N N DO () C FC sixHi OH .--