US009266828B2

(12) United States Patent (io) Patent No.: US 9,266,828 B2 Strongin et al. (45) Date of Patent: Feb. 23, 2016

(54) INHIBITORS OF FURIN AND OTHER Merck Manual Colorectal Cancer accessed Aug. 21, 2014 at URL PRO-PROTEIN CONVERTASES merckmanuals.com/home/digestive_disorders/tumors_of_the_ digestive_system/colorec tal_cancer.html).* (71) Applicant: Sanford-BurnhamMedicalResearch Merck Manual Prostate Cancer accessed Aug. 21, 2014 at URL: Institute, La Jolla, CA (US) merckmanuals.com/home/kidney_and_urinary_tract_disorders/ cancers_of_the_kidney_and_genitourinary_tract/prostate_can- (72) Inventors: Alex Strongin, La Jolla, CA (US); cer.html?qt=prostate cancer&alt=sh.* Maurizio Pellecchia, La Jolla, CA (US); Merck Manual Breast Cancer accessed Aug. 21, 2014 at URL: Elisa Barile, La Jolla, CA (US) merckmanuals.com/home/womens_health_issues/breast_disor- ders/breast_cancer.html). * (73) Assignee: SANFORD-BURNHAM MEDICAL Merck Manual Bladder Cancer accessed Aug. 21, 2014 at URL: RESEARCH INSTITUTE, La Jolla, merckmanuals.com/home/kidney_and_urinary_tract_disorders/ CA(US) cancers of the kidney and genitourinary tract/bladder can- cer.html.* ( * ) Notice: Subject to any disclaimer, the term of this National Institute of Cancer—understanding and related topics, patent is extended or adjusted under 35 accessed Aug. 21, 2014 at URL: cancer.gov/cancertopics/ understandingcancer. * U.S.C. 154(b) by 0 days. (Merck Manuals Brain Tumors accessed Aug. 21, 2014 at URL merckmanuals.com/home/brain_spinal_cord_and_nerve_disor- (21) Appl. No.: 14/385,714 ders/tumors_of_the_nervous_system/brain_tumors.html).* Merck Manual Skin Cancer accessed Jun. 9, 2015 at URL: (22) PCT Filed: Mar. 14, 2013 merckmanuals.com/professionial/dermatologic-disorders/cancers- of-the-skin/overview-of-skin-cancer.* (86) PCT No.: PCT/US2013/031733 Merck Manual Skin Cancer accessed Jun. 9, 2015 at URL: § 371 (c)(1), merckmanuals.com/professionial/dermatologic-disorders/cancers- (2) Date: Sep. 16, 2014 of-the-skin/squamous-cell-carcinoma.* Merck Manual Lung Cancer accessed Jun. 9, 2015 at URL: (87) PCT Pub. No.: WO2013/138665 merckmanuals.com/home/ pulmonary-disorders/tumors-of-the- hmgs/lung-carc inoma.html.* PCT Pub. Date: Sep. 19, 2013 Medline Plus—Autoimmune disorders—accessed Jun. 22, 2015 at URL: nlm.nih.gov/medlineplus/ency/article/000816.htm.* (65) Prior Publication Data Medzhitov, “Origin and physiological roles of inflammation,” Nature US 2015/0073054 A l Mar. 12, 2015 454:428-435 (2008).* Bennett et al., “A Furin-Iike Convertase Mediates Propeptide Cleav­ age of BACE, the Alzheimer’s b-Secretase,” J. Biol. Chem. Related U.S. Application Data 275:37712-37717 (2000).* Stawowy et al., “Proprotein convertases furin and PC5: targeting (60) Provisional application No. 61/611,967, filed on Mar. atherosclerosis and restenosis at multiple levels,” J. Mol. Med. 16, 2012. 83:865-875 (2005).* Ma et al., “Effect of Furin inhibitor on lung adenocarcinoma cell Int. Cl. growth and metastasis,” Cane. Cell Intl 14(43):1-6 (2014).* C07C 279/04 (2006.01) Remade et al., “Selective and potent furin inhibitors protect cells A61K 31/155 (2006.01) from anthrax without significant toxicity,” Int J Biochem Cell Biol. A6 IP 35/00 (2006.01) 42(6):987-995 (2010).* C07C 317/50 (2006.01) Becker et al., “Highly Potent Inhibitors of Proprotein Convertase A 6 IK 38/00 (2006.01) Furin as Potential Drugs for Treatment of Infectious Diseases,” J. CO 7K 5/09 (2006.01) Biol. Chem. 287:21992-22003 (2012).* Mercapide et al., “Inhibition of Furin-mediated Processing Results in C07K5/062 (2006.01) Suppression of Astrocytoma Cell Growth and Invasiveness,” Clin. (52) U.S. Cl. Cane. Res. 8:1740-1746 (2002).* CPC ...... C07C 317/50 (2013.01); A 6 1 K 38/00 (Continued) (2013.01); C07K5/06034 (2013.01); C07K 5/0817 (2013.01) (58) Field of Classification Search Primary Examiner — Julie Ha None Assistant Examiner — Kristina M Heilman See application file for complete search history. (74) Attorney, Agent, or Firm — Wilson Sonsini Goodrich & Rosati (56) References Cited (57) ABSTRACT FOREIGN PATENT DOCUMENTS Disclosed herein are Furin/PC inhibitors for inhibiting Furin and other Propprotein Convertases. Method of making the WO WO2013/138665 9/2013 Furin/PC inhibitors, chemical and biological characterization WO WO2013/138666 9/2013 of the Furin/PC inhibitors, and the use of the Furin/PC inhibi­ OTHER PUBLICATIONS tors to treat infectious diseases, cancers, and inflammatory/ Becker et al., “Potent Inhibitors of Furin and Furin-Iike Proprotein autoimmune disorders, are also disclosed. Convertases Containing Decarboxylated Pl Arginine Mimetics,” J. Med. Chem. 53: 1067-1075 (2009).* 11 Claims, 14 Drawing Sheets US 9,266,828 B2 Page 2

(56) References Cited PCT/US2013/031733 International Preliminary Report on Patent­ ability dated Sep. 14, 2014. OTHER PUBLICATIONS PCT/US2013/031733 International Search Report and Written Opin­ Khatib et al., “Inhibition of Proprotein Convertases is Associated ion dated Mar. 14, 2013. with Loss of Growth and Tumorigenicity of HT-29 Human Colon PCT/US2013/031737 International Search Report and Written Opin­ Carcinoma Cells: Importance of Insulin-Like Growth Factor-1 (IGF- ion dated Mar. 14, 2013. 1) Receptor Processing in IGF-I-Mediated Functions,” J. Biol. Chem. 276(33):30686-30693 (2001).* PCT/US2013/031737 International Preliminary Report on Patent­ Bassi et al., “Furin inhibition results in absent or decreased invasive­ ability dated Sep. 16, 2014. ness and tumorigenicity of human cancer cells,” PNAS 98(18):10326-10331 (2001).* * cited by examiner U.S. Patent Feb.23, 2016 Sheet I of 14 US 9,266,828 B2 FIG. 1 FIG.

[-ire] -sud)u| U.S. Patent Feb.23, 2016 Sheet 2 of 14 US 9,266,828 B2

FIG. 2

SAMPLE N F ORMATfON

Sampte Name: '9687 Asquited: Byr Systeav Sampte Type: IM m m BateAequlfed: 3/3Q;20n&i2:5SPFA VsaP £5 Acq. Melftod: 595¾?: AGM 229 Li. TF I Bate AocesseO: 3i1&2013 3:53 10 RM SijsdSca' Voteme: 19:05 at Changes Name; 24$7Chanfi« 2 Rail; TaSK- 20 m MiftOSes Sspipfe Set Name: a

idas ' 1 vied ' lies

RT ,Rrsa HeigM % % Area (lysec) O') HegftS I 10403 3S'1587B: 99.84 587018 93.73 2 11,173 5SB7 O tS 1203 8.2:1 U.S. Patent Feb.23, 2016 Sheet 3 of 14 US 9,266,828 B2

FIG. 3

j A_

11 Hi 3 3 7 S 5 4 3 ? ppm U.S. Patent Feb.23, 2016 Sheet 4 of 14 US 9,266,828 B2

FIG. 4

SAMPLE INFORMATION SarotRe Hmte: 3SS? ,a?„AS«SA„p4C &msma Bv: SSy sfessv Bmpe Type: Un^rsmT. Oass tepseH: VKIOiBOiB S::>7vS7 PtS VVfi •S3 A«; RSetacst OJtOSSB ACtfi JOBBStJSSoS issseekssi s: ■s Safe Pj«csss*S: KSvSK S3 S 33 S 5 BSS SaAvsissi VoAssse 33 .SS (A Ciianss-S !SSftse 3-fiS VAatitiit: 2 •A I I I SSjA Tissse 53:SS !SitUfites I I 3¾

8.08-] i

p so-j

3.084 I

5.SS-j 5.OSH I &$£H I! il a J „ _____ , ' ’ i.scf ’ ' SfiSt fi.cd ®!S) 1 ' sfifis? ' ' sisfit 1 ‘ ties? ' ' Asxi

I RT I AiTtS „ . : Ha

FIG. 5

J

Ij1 I

8.5 8.0 7.5 7.0 6,5 6.0 5,5 5.0 4,5 4.0 3,5 3.0 2,5 2.0 1,5 1.0 ppm O O o -§ US 9,266,828 B2 Sheet 6 of6 14 Sheet Feb.23, 2016 [r>6] 'suajui [r>6]

DM U.S. Patent U.S. U.S. Patent Feb.23, 2016 Sheet 7 of 14 US 9,266,828 B2

FIG. 7

Sample Name: S6B21 Acquired By: System Sample Type: Unknown Date Acquired: 11/26/2012 3:16:00 PM Vial: 28 Acq. Method: 5_95%B ACN_EBMethod lejeciion # 1 Date Processed: 11/26/2012 3:53:58 PM Injection Volume: 30.00 u! Channei Name: 2487Channel 2 Run Time: 17.00 Minutes Sample Set Name: a

1,20-

0.40-

0. 20-

----' -

2001 4.Go' 6 .001 kao' iW 12.001 ' ' 14.00 ■iO.OO1 Minutes

RT Area Height % (min) (SAsee) % Area m Height 1 9.832 7652214 too. 00 1360120 100.00 U.S. Patent Feb.23, 2016 Sheet 8 of 14 US 9,266,828 B2

FIG. 8

9.5 9.0 a.5 6.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 ppm U.S. Patent Feb.23, 2016 Sheet 9 of 14 US 9,266,828 B2

FIG. 9

52.¾ S4.'0D Missies

RT Area Height lit Araa % Smirvj ( rrs s c j (?¥j Height Ii 2.450 277¾ 0 . » 530 1.10 2. 1T.-238 440S i .55 580 I 21 3 11517 224 8.08 92 0.19 .4' 11710 :7701': 97.48 45853 97.50 U.S. Patent Feb.23, 2016 Sheet 10 of 14 US 9,266,828 B2

- O CD

O LO LO CNJ

"=t- O FIG. 10 FIG.

O CTj OO CO

- C D CO

S CNJ

- O

OO CNI

CD CO CO 0 5 U.S. Patent Feb.23, 2016 Sheetll of 14 US 9,266,828 B2

FIG. 11

12 11 10 9 8 7 6 5 4 3 2 1 ppm U.S. Patent Feb.23, 2016 Sheet 12 of 14 US 9,266,828 B2

FIG. 12

Log [inhibitor] pM 96B7■ HiilSIope 0.8806 IC50 0.006948 U.S. Patent Feb.23, 2016 Sheet 13 of 14 US 9,266,828 B2

FIG. 13

h>- 96B22 96B23

Log [Inhibitor] \xM

96B22 96B23 HifiSIope 1.622 1.068 IC50 0.003686 0.009096 U.S. Patent Feb.23, 2016 Sheet 14 of 14 US 9,266,828 B2

FIG. 14

Log [96B15J p M

96B15 HillSfope 1842 IC50 0.3381 US 9,266,828 B2 I 2 INHIBITORS OF FURIN AND OTHER embodiments of the compound of Formula I, R3 is —Z- PRO-PROTEIN CONVERTASES guanidine. In some embodiments of the compound of For­ mula I, Z is CROSS-REFERENCE 5 The present application is the National Phase entry of International Application No. PCT/US 2013/031733, filed Mar. 14, 2013, which claims the benefit of U.S. Provisional Application No. 61/611,967, filed on Mar. 16, 2012, both of 10 which are incorporated herein by reference in their entireties. In some embodiments of the compound of Formula I, X is — CH2— and R3 is BACKGROUND H2N Furin belongs to the subtilisin-like proprotein convertase family. Furin is a proprotein convertase that processes latent 15 precursor proteins into their biologically active products. Itis a calcium-dependent serine endoprotease that cleaves precur­ sor proteins at their paired basic amino acid processing sites. In some embodiments of the compound of Formula I, Y is Some of the Furin substrates are: proparathyroid hormone, —CH2—. In some embodiments of the compound of For­ transforming growth factor beta I precursor, proalbumin, pro- 20 mula I, W is beta-secretase, membrane type-1 matrix metalloproteinase, beta subunit of pro-nerve growth factor and von Willebrand (R7),, factor.

SUMMARY OF THE DISCLOSURE 25

Disclosed herein, in certain embodiments, are compounds R7 is —F, —CF3, —OCF3, —OCH3, or alkyl; and n is 0, I, having the general structure I or pharmaceutically acceptable or 2. In a refinement, R7 is —F. In a further refinement, n is I . salts thereof: A compound of Formula I, or a pharmaceuti­ In some embodiments, the compound of Formula I is selected cally acceptable salt, stereoisomer, tautomer, or prodrug 30 from the group consisting of: thereof:

(I) 35

40 R3

wherein R1 is alkyl, cycloalkyl, heteroalicyclyl, aryl, or het­ eroaryl; 45 R2 is alkyl, cycloalkyl, or heteroalicyclyl; R3 is —Z-guanidine or —Z—C(NH2)=N H , wherein Z is aryl or heteroaryl; R4 is — W— C(NH2)= N R ', wherein W is aryl, thiophe- nyl, furanyl, oxazolyl, pyrrolyl, or picolinyl; and 50 wherein R' is hydrogen or hydroxyl; R5 is —U-guanidine, wherein U is alkyl, cycloalkyl, heteroalicyclyl, aryl, or heteroaryl; X =-CH 2- , -C H 2-C H 2- , —CH2NHC(=0)—, —CH2CH2C(=0)NH—, or —CH2C(=0)NH—; 55 Y is -C H 2- , —S(=0)2—, or— C(=0)—. In some embodiments of the compound of Formula I, R' is hydrogen. In some embodiments of the compound of For­ mula I, R' is hydroxyl. In some embodiments of the com­ pound of Formula I, R1 is a C1-C6 alkyl. In some embodi­ 60 ments of the compound of Formula I, R1 is methyl. In some embodiments of the compound of Formula I, R2 is a C1-C6 alkyl. In some embodiments of the compound of Formula I, R2 is isopropyl. In some embodiments of the compound of Formula I, U is C1-C6 alkyl. In some embodiments of the 65 compound of Formula I, U is — (CH2)3— . In some embodi­ ments of the compound of Formula I, X is —CH2— . In some US 9,266,828 B2 3 4 ■continued Also disclosed herein, in certain embodiments, are phar­ maceutical compositions comprising a Furin/PC inhibitor disclosed herein. Additionally disclosed herein, in certain embodiments, are 5 methods of treating an infectious disease in a subject in need of such treatment. In some embodiments, the methods com­ prise administering a therapeutically effective amount of a Furin/PC inhibitor disclosed herein. In some embodiments, the infection disease is associated with influenza virus, 10 human immunodeficiency virus I, Ebola, measles, cytome­ galovirus, and flaviviruses (Dengue, Yellow fever, West Nile, Japanese encephalitis and multiple additional related flavivi­ ruses) and parasitic nematodes. In some embodiments, the Further disclosed herein, in certain embodiments, are com­ Furin/PC inhibitor neutralizes an exotoxin selected from the pounds of Formula II, or a pharmaceutically acceptable salt, 15 group consisting of anthrax toxin, pseudomonas exotoxin A, stereoisomer, tautomer, or prodrug thereof: Shiga toxin, diphtheria toxin, tetanus and botulism neurotox­ ins, and combinations thereof. In some embodiments, the Furin/PC inhibitor neutralizes virulence of bacteria carrying (H) the exotoxin. 20 Further disclosed herein, in certain embodiments, are NH2 methods of treating a cancer in a subject in need thereof. In some embodiments, the methods comprise administering a therapeutically effective amount of a Furin/PC inhibitor dis­ closed herein. In some embodiments, the cancer is skin 25 tumors, head and neck squamous cell carcinomas, astrocy­ toma, lung non-small cell carcinoma, or metastasis of col­ orectal cancer. Also disclosed herein, in certain embodiments, are meth­ ods of treating an autoimmune or inflammatory disease, dis­ wherein: 30 order or condition in a subject in need thereof. In some R1 is alkyl, cycloalkyl, or heteroalicyclyl; embodiments, the methods comprise administering a thera­ R2 is —U-guanidine, wherein U is alkyl, cycloalkyl, het­ peutically effective amount of a Furin/PC inhibitor disclosed eroalicyclyl, aryl, or heteroaryl; herein. In some embodiments, the autoimmune or inflamma­ Y is —CONH-, -S O 2N H -, —O—, -C H 2- , —S—, tory disease is atherosclerosis, arthritis, or Alzheimer’s Dis­ -SO 2- , Or-COSO2NH-; 35 ease. Z is —CONH-, -S O 2N H -, —O—, -C H 2- , —S—, -SO 2- , Or-COSO2NH-; BRIEF DESCRIPTION OF THE DRAWINGS R3 and R4 are each independently —F, —CF3, —OCF3, —OCH3, or alkyl; The technical features of the present disclosure are set forth a and b are each independently 0, I, or 2; and 40 with particularity in the appended claims. A better under­ m and n are each independently 0, 1, 2, or 3. standing of the features and advantages of the present disclo­ In some embodiments of the compound of Formula II, R1 sure will be obtained by reference to the following detailed is C1-C6 alkyl. In some embodiments of the compound of description that sets forth illustrative embodiments, in which Formula II, R1 is isopropyl. In some embodiments of the the principles of the disclosure are utilized, and the accom­ compound of Formula II, U is C1-C6 alkyl. In some embodi­ 45 panying drawings of which: ments of the compound of Formula II, U is —(CH2)3— . In FIG. I exemplifies the HPLC profile of Compound A; some embodiments of the compound of Formula II, Y is FIG. 2 exemplifies the MS profile of Compound A 2; —CONH— . In some embodiments of the compound of For­ FIG. 3 exemplifies the 1H NMR spectrum of CompoundA mula II, Z is — SO2NH— . In some embodiments of the com­ in DMSO-d6; pound of Formula II, m is I and n is I. In some embodiments 50 FIG. 4 exemplifies the HPLC profile Compound B; of the compound of Formula II, a and b are 0. In some FIG. 5 exemplifies the 1H NMR spectrum of Compound B embodiments, the compound of Formula II is in deuterated PBS; US 9,266,828 B2 5 6 FIG. 6 exemplifies the MS (MALDI) profile of Compound closed herein required to provide a clinically significant D; decrease in disease symptoms without undue adverse side FIG. 7 exemplifies the HPLC profile of Compound D; effects. An appropriate “effective amount” in any individual FIG. 8 exemplifies the 1H NMR spectrum of Compound D case may be determined using techniques, such as a dose in deuterated PBS; 5 escalation study. The term “therapeutically effective amount” FIG. 9 exemplifies the HPLC profile of Compound E. includes, for example, a prophylactically effective amount. FIG. 10 exemplifies the MS profile of Compound E; An “effective amount” of a compound disclosed herein is an FIG. 11 exemplifies 1H NMR spectrum o f Compound E in amount effective to achieve a desired pharmacologic effect or DMSO-d6; therapeutic improvement without undue adverse side effects. FIG. 12 exemplifies the biochemical assay dose response 10 It is understood that “an effect amount” or “a therapeutically for Compound A; effective amount” can vary from subject to subject, due to FIG. 13 exemplifies the biochemical assay dose response variation in metabolism of Ibrutinib, age, weight, general for Compound B and Compound C; and condition of the subject, the condition being treated, the FIG. 14 exemplifies the biochemical assay dose response severity of the condition being treated, and the judgment of for Compound E. 15 the prescribing physician. By way of example only, therapeu­ tically effective amounts may be determined by routine DETAILED DESCRIPTION OF THE experimentation, including but not limited to a dose escala­ DISCLOSURE tion clinical trial. The terms “subject”, “patient” and “individual” are used Proprotein convertases (PCs), such as Furin, plays an 20 interchangeably. As used herein, they refer to an animal. By important role in diseases such as Alzheimer’s disease, can­ way o f example only, a subject may be, but is not limited to, cer, and viral and bacterial infections. Many pathogens a mammal including, but not limited to, a human. The terms depend on the human pro-protein convertase Furin to process do not require the supervision (whether continuous or inter­ their toxins or cell adhesion factors. Accordingly, Furin mittent) of a medical professional. inhibitors that inactivate these mechanisms in host-pathogen 25 The terms “isolated” and “purified” refer to a material that interactions provides an effective route to prevent the initia­ is substantially or essentially removed from or concentrated tion or propagation of the infection. Thus, inhibition of furin in its natural environment. For example, an isolated nucleic may provide a feasible and promising approach for therapeu­ acid is one that is separated from the nucleic acids that nor­ tic intervention of furin-mediated disease mechanisms. mally flank it or other nucleic acids or components (proteins, There is a need for identifying potent and selective agents 30 lipids, etc.) in a sample. In another example, a polypeptide is for the treatment of various diseases, disorders and patholo­ purified if it is substantially removed from or concentrated in gies, such as infectious diseases (e.g. influenza and Anthrax), its natural environment. Methods for purification and isola­ cancers, and neurodegenerative diseases (e.g. Alzheimer). tion of nucleic acids and proteins are documented method­ Proteolysis data have revealed detailed information on ologies. Furin and related Furin-Iike PCs cleavage preferences for 35 The term “optionally substituted” or “substituted” means their substrates, consisting of multibasic consensus that the referenced group substituted with one or more addi­ sequences, preferentially located after an aiginine residue. tional group(s). In certain embodiments, the one or more Because no small-molecule inhibitors of Furin are currently additional group(s) are individually and independently available, d-Arg-based peptides, a l -antitrypsin Portland, and selected from amide, ester, alkyl, cycloalkyl, heteroalkyl, the synthetic inhibitor decanoyl-Arg-Val-Lys-Arg-chlorom- 40 aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, ethylketone (DEC-RVKR-CMK) have been used to validate alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, ester, alkyl- the role of Furin in a variety of cellular models. sulfone, arylsulfone, cyano, halogen, alkoyl, alkoyloxo, iso- However, Arg-based peptides such as hexa- and nona-d- cyanato, thiocyanato, isothiocyanato, nitro, haloalkyl, Arg have either low or no therapeutic potential because of haloalkoxy, fluoroalkyl, amino, alkyl-amino, dialkyl-amino, their intrinsic ability to cross-react with multiple, pathogen 45 amido. and host, proteinase and non-proteinase targets, which are An “alkyl” group refers to an aliphatic hydrocarbon group. unrelated to Furin. Reference to an alkyl group includes “saturated alkyl” and/or The terms “treat,” “treating” or “treatment”, as used herein, “unsaturated alkyl”. The alkyl group, whether saturated or include alleviating, abating or ameliorating a disease or con­ unsaturated, includes branched, straight chain, or cyclic dition symptoms, preventing additional symptoms, amelio­ 50 groups. By way of example only, alkyl includes methyl, ethyl, rating or preventing the underlying metabolic causes of propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, t-butyl, pen­ symptoms, inhibiting the disease or condition, e.g., arresting tyl, iso-pentyl, neo-pentyl, and hexyl. In some embodiments, the development of the disease or condition, relieving the alkyl groups include, but are in no way limited to, methyl, disease or condition, causing regression of the disease or ethyl, propyl, isopropyl, butyl, isobufyl, tertiary butyl, pentyl, condition, relieving a condition caused by the disease or 55 hexyl, ethenyl, propenyl, butenyl, cyclopropyl, cyclobutyl, condition, or stopping the symptoms of the disease or condi­ cyclopentyl, cyclohexyl, and the like. A “lower alkyl” is a tion. The terms “treat,” “treating” or “treatment”, include, but C1-C6 alkyl. A “heteroalkyl” group substitutes any one of the are not limited to, prophylactic and/or therapeutic treatments. carbons of the alkyl group with a heteroatom having the The terms “effective amount” or “therapeutically effective appropriate number of hydrogen atoms attached (e.g., a CH2 amount,” as used herein, refer to a sufficient amount of an 60 group to an NH group or an O group). agent or a compound being administered which will relieve to An “alkoxy” group refers to a (alkyl)O— group, where some extent one or more of the symptoms of the disease or alkyl is as defined herein. condition being treated. The result can be reduction and/or The term “alkylamine” refers to the N(Blkyl)xHy group, alleviation of the signs, symptoms, or causes of a disease, or wherein alkyl is as defined herein and x and y are selected any other desired alteration of a biological system. For 65 from the group x=l, y=l and x=2, y=0. When x=2, the alkyl example, an “effective amount” for therapeutic uses is the groups, taken together with the nitrogen to which they are amount of the composition including a compound as dis­ attached, optionally form a cyclic ring system. US 9,266,828 B2 7 8 An “amide” is a chemical moiety with formula C(O)NHR its ring system, and with the proviso that the ring of said group OrNHC(O)R, where R is selected from alkyl, cycloalkyl, aryl, does not contain two adjacent O or S atoms. Non-aromatic heteroaryl (bonded through a ring carbon) and heteroalicyclic heterocyclic groups include groups having 3 atoms in their (bonded through a ring carbon). ring system, but aromatic heterocyclic groups must have at The term “ester” refers to a chemical moiety with formula 5 least 5 atoms in their ring system. The heterocyclic groups —C (=0)0R , where R is selected from the group consisting include benzo-fused ring systems. An example of a 3-mem- of alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic. bered heterocyclic group is aziridinyl (derived from aziri- As used herein, the term “aryl” refers to an aromatic ring dine). An example of a 4-membered heterocyclic group is wherein each of the atoms forming the ring is a carbon atom. azetidinyl (derived from azetidine). An example of a 5-mem- 10 Aryl rings described herein include rings having five, six, bered heterocyclic group is thiazolyl. An example of a seven, eight, nine, or more than nine carbon atoms. Aryl 6-membered heterocyclic group is pyridyl, and an example of groups are optionally substituted. Examples of aryl groups a 10-membered heterocyclic group is quinolinyl. Examples include, but are not limited to phenyl, and naphthalenyl. of non-aromatic heterocyclic groups are pyrrolidinyl, tetrahy- The term “cycloalkyl” refers to a monocyclic or polycyclic drofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropy- non-aromatic radical, wherein each of the atoms forming the 15 ranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, ring (i.e. skeletal atoms) is a carbon atom. In various embodi­ morpholino, thiomorpholino, thioxanyl, piperazinyl, aziridi­ ments, cycloalkyls are saturated, or partially unsaturated. In nyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepa- some embodiments, cycloalkyls are fused with an aromatic nyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1,2,3,6- ring. Cycloalkyl groups include groups having from 3 to 10 tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, ring atoms. Illustrative examples of cycloalkyl groups 20 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazoli- include, but are not limited to, the following moieties: nyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3-azabicyclo[3.1 .Ojhexanyl, 3-azabicyclo[4.1 .Ojheptanyl, 25 3H-indolyl and quinolizinyl. Examples of aromatic heterocy­ clic groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thia­ zolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinoli- nyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, inda- 30 zolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl. 35 The terms “heteroaryl” or, alternatively, “heteroaromatic” D o - o o refers to an aryl group that includes one or more ring heteroa­ toms selected from nitrogen, oxygen and sulfur. An N-con- taining “heteroaromatic” or “heteroaryl” moiety refers to an aromatic group in which at least one of the skeletal atoms of 40 the ring is a nitrogen atom. In certain embodiments, het­ eroaryl groups are optionally substituted. In certain embodi­ ments, heteroaryl groups are monocyclic or polycyclic. Examples of monocyclic heteroaryl groups include and are not limited to: 45 o CD CO H O O O 6 50 CO pyrrole fliran thiophene pyrazole (pyrrolyl) (furanyl) (thiophenyl) (pyrazolyl H and the like. Monocyclic cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Dicylclic cycloalkyls include, 55 O *-----N O O*-----N O but are not limited to tetrahydronaphthyl, indanyl, tetrahydro- pentalene or the like. Polycyclic cycloalkyls include adaman- imidazole isoxazole oxazole isothiazole tane, norbomane or the like. The term cycloalkyl includes (imidazolyl) (isoxazolyl) (oxazolyl (isothiazolyl) “unsaturated nonaromatic carbocyclyl” or “nonaromatic H H unsaturated carbocyclyl” groups both of which refer to a 60 nonaromatic carbocycle, as defined herein, that contains at O A O least one carbon carbon double bond or one carbon carbon O^-----N V-----N N—N *-----N triple bond. thiazolyl 1,2,3-triazole 1,3,4-triazole l-oxa-2,3- The term “heterocyclic” or “heterocyclyl” refers to het­ (thiazolyl) (1,2,3- (1,3,4- diazole eroaromatic and heteroalicyclic groups containing one to four 65 triazolyl) triazolyl) (l-oxa-2,3- ring heteroatoms each selected from O, S and N. In certain diazolyl) instances, each heterocyclic group has from 4 to 10 atoms in US 9,266,828 B2 9 10 -continued -continued 0 % A CN ---- J \JI N—N Y"^---- N

I -oxa-2,4- l-oxa-2,5- l-oxa-3,4- l-thia-2,3- diazole diazole diazole diazole pyrazolo[4,3-d]pyridine pyrazolo[4,3-d]pyridine (l-oxa-2,4- (l-oxa-2,5- (l-oxa-3,4- (l-thia-2,3- (pyrazolo[4,3-d]pyridinyl) (pyrazolo[4,3-d]pyridinyl) diazolyl) diazolyl) diazolyl) diazolyl) H / s \ O x N

25

indolizine imidazo[l,2-a]pyridine (indolininyl) (imidazo [1,2-a]pyridinyl) 1,3,5-triazine (triazinyl) 30 ,L Examples of bicyclic heteroaryl groups include and are not CO / limited to: imidazo[l,5-a]pyridine pyrazolo [1,5-a]pyri dine (imidazo [ 1,5 -ajpyridiny I) (pyrazolo [ 1,5 -ajpyridiny I)

pyrrolo[l,2-b]pyridazine imidazo [1,2-c]pyrimidine benzofuran benzothiophene indole (pyrrolo[l ,2-b]pyridazinyl) (imidazo[l ,2-c]pyrimidinyl) (benzofuranyl) (benzothiaphenyl) (indolyl)

thienopyrimidine thienopyrimidine quinoline benzimidazole indazole benzotriazole (thienopyrimidinyl) (thienopyrimidinyl) (quinolinyl) (benzimidazolyl)a z o ^ (indazolyl) (benzotriazolyl) SC r Y Y i Y r CC n JC CO H isoquinoline cinnoline quinazoline pyrrolo[2,3-b]pyridine pyrrolo[2,3-c]pyridine (isoquinolinyl) (cinnolinyl) (azaquinazoline) (pyrrolo[2,3-b]pyridinyl) (pyrrolo[2,3-c]pyridinyl)

quinoxaline phthalazine 1,6-naphthyridine pyrrolo[3,2-c]pyridine pyrrolo [3,2-b]pyridine (quinoxalinyl) (phthalazinyl) (1,6-naphthyridinyl) (pyrrolo[3,2-c]pyridinyl) (pyrrolo[3,2-b]pyridinyl)

1,7-naphthyridine 1,8-naphthyridine 1,5-naphthyridine imidazo[4,5-b]pyridine imidazo[4,5-c]pyridine 65 (1,7-naphthyridinyl) (1,8-naphthyridinyl) (1,5-naphthyridinyl) imidazo[4,5-b]pyridinyl) imidazo [4,5-c]pyridinyl) US 9,266,828 B2 1 1 12 -continued -continued

2,6-naphthyridine 2,7-naphthyridine pyrido[3,2-d]pyrim.idine (2,6-naphthyridinyl) (2,7-naphthyridinyl) (pyrido[3,2-d]pyrimidinyl) 1,4- morpholine 1,4- piperazine oxathiane (morpholinyl) dithiane (piperazinyl) OA (1,4- oxathianyl) dithianyl)

pyrido[4,3-d]pyrim.idine pyrido[3,4-d]pyrimidine (pyrido[4,3-d]pyrimidinyl) (pyrido [3,4-d]pyrim.idinyl)

thiepane azepane 1,4- (thiepanyl) (azepanyl) azathiane (1,4- pyrido[2,3-d]pyrimidine pyrido[2,3-b]pyrazine azathianyl) (pyrido[2,3-d]pyrimidinyl) (pyrido[2,3-b]pyrazinyl)

1,4- 1,4- 1,4- 1,4- pyrido [3,4-b]pyrazine pyrimido [5,4-d]pyrimidine dioxepane oxathiepane oxaaepane dithiepane (pyrido[3,4-b]pyrazinyl) (pyrido[5,4-d]pyrimidinyl) (1,4- (1,4- (1,4- (1,4- dioxypanyl) oxathiepanyl) oxaazepanyl) dithiepanyl)

pyrazino[2,3-b]pyrazine pyrimido[4,5-d]pyrimidine (pyrazino[2,3-b]pyrazinyl) (pyrido [4,5-d]pyrimidinyl) NH NH NH 1,4- 1,4- tropane thieazapane diazepane (tropanyl) or the like. ¢1,4- (1,4- thieazapanyl) diazepanyl) A “heteroalicyclic” group or “heterocycloalkyl” group refers to a cycloalkyl group, wherein at least one skeletal ring atom is a heteroatom selected from nitrogen, oxygen and Examples of partially unsaturated heterocycloalkyl groups sulfur. In some embodiments, the radicals are fused with an include aryl or heteroaryl. Example of saturated heterocyloalkyl groups include

O ZN □° a 3,4-dihydro-2H-pyran 5,6-dihydro-2H-pyran 2H-pyran oxirane thiarane aziridine oxetane thiatane (3,4,-dihydro-2H-pyranyl) (5,6-dihydro-2H-pyranyl) (2H-pyranyl) (oxiranyl) )thiaranyl) (aziridinyl) (oxetanyl) (thiatanyl) 50 H S. I----- NH □ azetidine tetrahydro- tetrahydro- pyrrolidine 1,2,5,6-tetrahydropyridine (azetidinyl) furan thiaphene (pyrro- (1,2,5,6-tetrahydropyridinyl) (tetrahydro- (tetrahydro- lidinyl) furanyl) thiaphenyl) Other illustrative examples of heterocycloalkyl groups, also referred to as non-aromatic heterocycles, include:

tetrahydro- tetrahydro- piperidine 1,4- pyran thiopyran (piperidinyl) dioxane (tetrahydro- (tetrahydro- 65 pyranyl) thiopyranyl) (1,4- dioxanyl) US 9,266,828 B2 13 14 An “isothiocyanato” group refers to a NCS group. “Alkoyloxy” refers to a R C (= 0)0— group. “Alkoyl” refers to a R C (=0)— group. 5 Compounds Provided herein are compounds having the general struc­ ture I or pharmaceutically acceptable salts stereoisomers, tautomers, or prodrugs thereof: 10

(I)

15 HHH

R3

20 wherein R1 is alkyl, cycloalkyl, heteroalicyclyl, aryl, or het­ eroaryl; R2 is alkyl, cycloalkyl, or heteroalicyclyl; 25 R3 is —Z-guanidine or —Z—C(NH2)=N H , wherein Z is aryl or heteroaryl; R4 is —W—C(NH2)=N R', wherein W is aryl, thiophe- nyl, furanyl, oxazolyl, pyrrolyl, or picolinyl; and 30 wherein R' is hydrogen or hydroxyl; R5 is —U-guanidine, wherein U is alkyl, cycloalkyl, heteroalicyclyl, aryl, or heteroaryl; or the like. X =-CH 2- , -C H 2-C H 2- , —CH2NHC(=0)—, —CH2CH2C(=0)NH—, or —CH2C(=0)NH—; The term heteroalicyclic also includes all ring forms of the 35 carbohydrates, including but not limited to the monosaccha­ Y is -C H 2- , —S(=0)2—, or —C(=0)—. rides, the disaccharides and the oligosaccharides. In some embodiments of the compound of Formula I, R' is The term “halo” or, alternatively, “halogen” means fluoro, hydrogen. In some embodiments of the compound of For­ chloro, bromo and iodo. 40 mula I, R' is hydroxyl. Without wishing to be bound by any The terms “haloalkyl,” and “haloalkoxy” include alkyl and particular theory, it is contemplated in the present disclosure alkoxy structures that are substituted with one or more halo­ that replacement of one or more imino hydrogen (such as in gens. In embodiments, where more than one halogen is the amidine and/or guanidine moiety) with hydroxyl included in the group, the halogens are the same or they are improves bioavailability in some embodiment. For example, different. The terms “fluoroalkyl” and “fluoroalkoxy” include 45 replacement of the carboxylmidamide in position R4 with a haloalkyl and haloalkoxy groups, respectively, in which the N'-hydroxyimidamide improves bioavailability in some halo is fluorine. embodiments. The term “heteroalkyl” include optionally substituted In some embodiments of the compound of Formula I, R1 is alkyl, alkenyl and alkynyl radicals which have one or more a C1-C6 alkyl. In some embodiments of the compound of skeletal chain atoms selected from an atom other than carbon, 50 Formula I, R1 is methyl. In some embodiments of the com­ e.g., oxygen, nitrogen, sulfur, phosphorus, silicon, or combi­ pound of Formula I, R2 is a C1-C6 alkyl. In some embodi­ nations thereof. In certain embodiments, the heteroatom(s) is ments of the compound of Formula I, R2 is isopropyl. placed at any interior position of the heteroalkyl group. In some embodiments of the compound of Formula I, U is Examples include, but are not limited to, —CH2—O—CH3, C1-C6 alkyl. In some embodiments of the compound of For­ -CH2-CH2-O -C H 3, -CH2-NH-CH3, -CH2- 55 mula I, U is — (CH2)3— . CH2-NH-CH3, -CH2-N(CH3)-CH3, -CH2- In some embodiments of the compound of Formula I, X is CH2-NH-CH3, -CH2-CH2-N(CH3)-CH3, —CH2—. In some embodiments of the compound of For­ -CH2-S-C H 2-CH3, -CH2-CH2, -S(O)-CH3, mula I, R3 is —Z-guanidine. In some embodiments of the -CH 2-CH 2-S(O)2-CH 3, -C H -C H -O -C H 3, —Si compound o f Formula I, Z is (CH3)3, -CH 2-CH -N-OCH 3, and -CH=CH-N 60 (CH3)—CH3. In some embodiments, up to two heteroatoms are consecutive, such as, by way of example, —CH2—NH— OCH3 and—CH2-O -S i(C H 3)3. A “cyano” group refers to a CN group. 65 An “isocyanato” group refers to a NCO group. A “thiocyanato” group refers to a CNS group. US 9,266,828 B2 15 16 In some embodiments of the compound of Formula I, X is -continued — CH2— and R3 is

H2N 5

10

In some embodiments of the compound of Formula I, Y is - C H 2- . In some embodiments o f the compound of Formula I, W is 15 Also disclosed herein, in certain embodiments, are com­ pounds of formula II, or pharmaceutically acceptable salts, stereoisomers, tautomers, or prodrugs thereof: ( R 7 )„

20 (H)

R7 is — F, — CF3, —OCF3, —OCH3, or alkyl; and n is 0 , 1, or 2. In a refinement, R7 is —F. In a further refinement, nisi. 25 In some embodiments, the compound of Formula I is selected from the group consisting of:

30 wherein: R1 is alkyl, cycloalkyl, or heteroalicyclyl; R2 is —U-guanidine, wherein U is alkyl, cycloalkyl, het­ 35 eroalicyclyl, aryl, or heteroaryl; Y is —CO N H -, —S02NH—, —O—, —CH2-, —S—, —S02-, or—C0S02NH—; Z is —CONH-, -S O 2N H -, —O—, -C H 2- , —S—, - S O 2- , or -C O S O 2N H - ; 40 R3 and R4 are each independently —F, —CF3, —OCF3, —OCH3, or alkyl; a and b are each independently 0, I, or 2; and m and n are each independently 0, I, 2, or 3. 45 In some embodiments, the prodrug of the compound in Formula II is formed by replacing one or more imino hydro­ gen (such as in the ami dine and/or guanidine moiety) with hydroxyl. In some embodiments, replacement of one or more imino hydrogen with hydroxyl improves bioavailability. In 50 some embodiments, replacement of the carboxy lmidamide in position R4 with a N'-hydroxyimidamide improves bioavail­ ability. In some embodiments of the compound of Formula II, R1 is C1-C6 alkyl. In some embodiments of the compound of 55 Formula II, R1 is isopropyl. In some embodiments of the compound of Formula II, U is C1-C6 alkyl. In some embodiments of the compound of For­ mula II, U is — (CH2)3— . In some embodiments o f the compound o f Formula II, Y is 60 —CONH— . In some embodiments of the compound of For­ mula II, Z is — SO2NH— . In some embodiments, Y is —CONH- and Z is -S O 2N H -. In some embodiments of the compound of Formula II, m is 65 I andn is I. In some embodiments of the compound of Formula II, a is O and b is 0. US 9,266,828 B2 17 18 In some embodiments, the compound of Formula II is

In some embodiments, compounds o f Formula I and II are:

Compound No. Chemical Structure IC50 against Furin

H2N NH2 US 9,266,828 B2 19 20 -continued

Compound No. Chemical Structure IC50 against Furin

In certain embodiments, compounds described herein have 30 art, for example by reacting a sufficiently basic compound one or more chiral centers. As such, all stereoisomers are such as an amine with a suitable acid affording a physiologi­ envisioned herein. In various embodiments, compounds cally acceptable anion. Alkali metal (for example, sodium, described herein are present in optically active or racemic potassium or lithium) or alkaline earth metal (for example forms. It is to be understood that the compounds described calcium) salts of carboxylic acids can also be made. herein encompass racemic, optically-active, regioisomeric 35 Compounds described herein also include isotopically-la- and stereoisomeric forms, or combinations thereof that pos­ beled compounds wherein one or more atoms is replaced by sess the therapeutically useful properties described herein. an atom having the same atomic number, but an atomic mass Preparation of optically active forms is achieve in any suitable or mass number different from the atomic mass or mass manner, including by way of non-limiting example, by reso­ 40 number usually found in nature. Examples of isotopes suit­ lution of the racemic form by recrystallization techniques, by able for inclusion in the compounds described herein include synthesis from optically-active starting materials, by chiral and are not limited to 2H, 3H, 11C, 13C, 14C, 36Cl, 18F, 123I, synthesis, or by chromatographic separation using a chiral 125I, 13N, 15N, 15O, 17O, 18O, 32P, 35S or the like. In some stationary phase. In some embodiments, mixtures of one or embodiments, isotopically-labeled compounds are useful in more isomer is utilized as the therapeutic compound 45 drug and/or substrate tissue distribution studies. In some described herein. In certain embodiments, compounds embodiments, substitution with heavier isotopes such as deu­ described herein contains one or more chiral centers. These terium affords certain therapeutic advantages resulting from compounds are prepared by any means, including enantiose- greater metabolic stability (for example, increased in vivo lective synthesis and/or separation of a mixture of enanti­ half-life or reduced dosage requirements). In some embodi- omers and/or diastereomers. Resolution of compounds and 50 ments, substitution with positron emitting isotopes, such as isomers thereof is achieved by any means including, by way 11C, 18F, 15O and 13N, is useful in Positron Emission Topog­ of non-limiting example, chemical processes, enzymatic pro­ raphy (PET) studies for examining substrate receptor occu­ cesses, fractional crystallization, distillation, chromatogra­ pancy. Isotopically-labeled compounds are prepared by any phy, and the like. suitable method or by processes using an appropriate isoto- In various embodiments, pharmaceutically acceptable 55 pically-labeled reagent in place of the non-labeled reagent salts described herein include, by way of non-limiting otherwise employed. example, a nitrate, chloride, bromide, phosphate, sulfate, In some embodiments, Furin/PC inhibitors disclosed acetate, hexafluorophosphate, citrate, gluconate, benzoate, herein reduce or inhibit the binding between Furin and/or PCs propionate, butyrate, sulfosalicylate, maleate, laurate, and at least one of its natural binding partners (e.g., Cdc42 or malate, fumarate, succinate, tartrate, amsonate, pamoate, 60 Rac). In some instances, binding between Furin and/or PCs p-toluenenesulfonate, mesylate and the like. Furthermore, and at least one of its natural binding partners is stronger in pharmaceutically acceptable salts include, by way of non­ the absence of a Furin/PC inhibitors (by e.g., 90%, 80%, 70%, limiting example, alkaline earth metal salts (e.g., calcium or 60%, 50%, 40%, 30% or 20%) than in the presence of a magnesium), alkali metal salts (e.g., sodium-dependent or Furin/PC inhibitors. potassium), ammonium salts and the like. Pharmaceutically 65 In some embodiments, a Furin/PC inhibitors suitable for acceptable salts of the compounds of the present invention the methods described herein is a direct Furin/PC inhibitors. may be obtained using standard procedures well known in the In some embodiments, a Furin/PC inhibitors suitable for the US 9,266,828 B2 21 22 methods described herein is an indirect Furin/PC inhibitors. groups which yield the covalent linkages. Table A is used as In some embodiments, a Furin/PC inhibitors suitable for the guidance toward the variety of electrophiles and nucleophiles methods described herein decreases Furin and/or PCs activity combinations available that provide covalent linkages. Pre­ relative to a basal level of Furin and/or PCs activity by about cursor functional groups are shown as electrophilic groups 1.1 fold to about 100 fold, e.g., to about 1.2 fold, 1.5 fold, 1.6 5 and nucleophilic groups. fold, 1.7 fold, 2.0 fold, 3.0 fold, 5.0 fold, 6.0 fold, 7.0 fold, 8.5 fold, 9.7 fold, 10 fold, 12 fold, 14 fold, 15 fold, 20 fold, 30 fold, 40 fold, 50 fold, 60 fold, 70 fold, 90 fold, 95 fold, or by Covalent Linkage Product Electrophile Nucleophile any other amount from about 1.1 fold to about 100 fold Carboxamides Activated esters amines/anilines relative to basal Furin and/or PCs activity. In some embodi­ 10 Carboxamides acyl azides amines/anilines ments, the Furin/PC inhibitors is a reversible Furin/PC inhibi­ Carboxamides acyl halides amines/anilines tors. In other embodiments, the Furin/PC inhibitors is an Esters acyl halides alcohols/phenols irreversible Furin/PC inhibitors. Esters acyl nitriles alcohols/phenols In some embodiments, a Furin/PC inhibitors used for the Carboxamides acyl nitriles amines/anilines Imines Aldehydes amines/anilines methods described herein has in vitro ED50 for Furin and/or 15 Hydrazones aldehydes or ketones Hydrazines PCs activation of less than 100 pM (e.g., less than 10 pM, less Oximes aldehydes or ketones Hydroxylamines than 5 (xM, less than 4 (xM, less than 3 (xM, less than I (xM, less Alkyl amines alkyl halides amines/anilines than 0.8 (xM, less than 0.6 (xM, less than 0.5 (xM, less than 0.4 Esters alkyl halides carboxylic acids Thioethers alkyl halides Thiols (xM, less than 0.3 (xM, lessthanless than 0.2 (xM, less than 0.1 Ethers alkyl halides alcohols/phenols (xM, less than 0.08 (xM, less than 0.06 (xM, less than 0.05 (xM, 20 Thioethers alkyl sulfonates Thiols less than 0.04 (xM, less than 0.03 (xM, less than less than 0.02 Esters alkyl sulfonates carboxylic acids (xM, less than 0.01 (xM, less than 0.0099 (xM, less than 0.0098 Ethers alkyl sulfonates alcohols/phenols Esters Anhydrides alcohols/phenols (xM, less than 0.0097 (xM, less than 0.0096 (xM, less than Carboxamides Anhydrides amines/anilines 0.0095 (xM, less than 0.0094 (xM, less than 0.0093 (xM, less Thiophenols aryl halides Thiols than 0.00092 (xM, or less than 0.0090 (xM). 25 Aryl amines aryl halides Amines In some embodiments, a Furin/PC inhibitors used for the Thioethers Azindines Thiols Boronate esters Boronates Glycols methods described herein has in vitro ED50 for Furin and/or Carboxamides carboxylic acids amines/anilines PCs activation of less than 100 (xM (e.g., less than 10 (xM, less Esters carboxylic acids Alcohols than 5 (xM, less than 4 (xM, less than 3 (xM, less than I (xM, less hydrazines Hydrazides carboxylic acids N-acylureas or Anhydrides carbodiimides carboxylic acids than 0.8 (xM, less than 0.6 (xM, less than 0.5 (xM, less than 0.4 30 Esters diazoalkanes carboxylic acids (xM, less than 0.3 (xM, lessthanless than 0.2 (xM, less than 0.1 Thioethers Epoxides Thiols (xM, less than 0.08 (xM, less than 0.06 (xM, less than 0.05 (xM, Thioethers haloacetamides Thiols less than 0.04 (xM, less than 0.03 (xM, less than less than 0.02 Ammotriazines halotriazines amines/anilines (xM, less than 0.01 (xM, less than 0.0099 (xM, less than 0.0098 Triazinyl ethers halotriazines alcohols/phenols Amidines imido esters amines/anilines (xM, less than 0.0097 (xM, less than 0.0096 (xM, less than 35 Ureas Isocyanates amines/anilines 0.0095 (xM, less than 0.0094 (xM, less than 0.0093 (xM, less Urethanes Isocyanates alcohols/phenols than 0.00092 (xM, or less than 0.0090 (xM). Thioureas isothiocyanates amines/anilines Synthesis and Characterization Thioethers Maleimides Thiols Phosphite esters phosphoramidites Alcohols The compounds described herein, and other related com­ Silyl ethers silyl halides Alcohols pounds having different substituents are synthesized using 40 Alkyl amines sulfonate esters amines/anilines techniques and materials described herein and as described, Thioethers sulfonate esters Thiols for example, in Fieser and Fieser’s Reagents for Oiganic Esters sulfonate esters carboxylic acids Ethers sulfonate esters Alcohols Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Sulfonamides sulfonyl halides amines/anilines Rodd’s Chemistry of Carbon Compounds, Volumes 1-5 and Sulfonate esters sulfonyl halides phenols/alcohols Supplemental (Elsevier Science Publishers, 1989); Oiganic 45 Reactions, Volumes 1-40 (John Wiley and Sons, 1991), Use of Protecting Groups Larock’s Comprehensive Organic Transformations (VCH In the reactions described, it is necessary to protect reactive Publishers Inc., 1989), March, A dvanced O rganic C hemistry functional groups, for example hydroxy, amino, imino, thio or 4th Ed., (Wiley 1992); Carey and Sundberg, A dvanced carboxy groups, where these are desired in the final product, O rganic C hemistry 4th Ed., Vols. A and B (Plenum 2000, 50 in order to avoid their unwanted participation in reactions. 2001), and Green and Wuts, P rotective G roups in O rganic Synthesis Y d Ed., (Wiley 1999) (all of which are incorporated Protecting groups are used to block some or all of the reactive by reference for such disclosure). General methods for the moieties and prevent such groups from participating in preparation of compound as described herein are modified by chemical reactions until the protective group is removed. In the use of appropriate reagents and conditions, for the intro­ 55 some embodiments it is contemplated that each protective duction of the various moieties found in the formula as pro­ group be removable by a different means. Protective groups vided herein. As a guide the following synthetic methods are that are cleaved under totally disparate reaction conditions utilized. Compounds described herein are synthesized using fulfill the requirement of differential removal. any suitable procedures starting from compounds that are In some embodiments, protective groups are removed by available from commercial sources, or are prepared using 60 acid, base, reducing conditions (such as, for example, hydro- procedures described herein. genolysis), and/or oxidative conditions. Groups such as trityl, Formation of Covalent Linkages by Reaction of an Electro­ dimethoxytrityl, acetal and t-butyldimethylsilyl are acid phile with a Nucleophile labile and are used to protect carboxy and hydroxy reactive The compounds described herein are modified using vari­ moieties in the presence of amino groups protected with Cbz ous electrophiles and/or nucleophiles to form new functional 65 groups, which are removable by hydrogenolysis, and Fmoc groups or substituents. Table below lists selected non-limit­ groups, which are base labile. Carboxylic acid and hydroxy ing examples of covalent linkages and precursor functional reactive moieties are blocked with base labile groups such as, US 9,266,828 B2 23 24 but not limited to, methyl, ethyl, and acetyl in the presence of -continued amines blocked with acid labile groups such as t-butyl car­ H3C CH3 bamate or with carbamates that are both acid and base stable \/ but hydrolytically removable. (H3C)3C In some embodiments carboxylic acid and hydroxy reac- 5 '■S (CH3)3C tive moieties are blocked with hydrolytically removable pro­ tective groups such as the benzyl group, while amine groups TBDMS Teoc capable of hydrogen bonding with acids are blocked with base labile groups such as Fmoc. Carboxylic acid reactive moieties are protected by conversion to simple ester com- to pounds as exemplified herein, which include conversion to alkyl esters, or are blocked with oxidatively-removable pro­ tective groups such as 2,4-dimethoxybenzyl, while co-exist- ing amino groups are blocked with fluoride labile silyl car­ bamates. 15 Allyl blocking groups are useful in the presence of acid- and base-protecting groups since the former are stable and are subsequently removed by metal or pi-acid catalysts. For example, an allyl-blocked carboxylic acid is deprotected with a Pd0-Catalyzed reaction in the presence of acid labile t-butyl 20 carbamate or base-labile acetate amine protecting groups. Yet another form of protecting group is a resin to which a com­ pound or intermediate is attached. As long as the residue is attached to the resin, that functional group is blocked and does not react. Once released from the resin, the functional 25 Fmoc group is available to react. Typically blocking/protecting groups are selected from:

Other protecting groups, plus a detailed description of techniques applicable to the creation of protecting groups and their removal are described in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, N.Y., 1999, and Kocienski, Protective Groups, 35 Thieme Verlag, New York, N.Y., 1994, which are incorpo­ rated herein by reference for such disclosure. Synthesis Example Compound A 40 CompoundA was prepared by a combination of solid phase and solution synthesis. Briefly, theN-acylated segment P2-P4 protected in P2 and P4 positions, after weak acidic cleavage from the 2-chloro-tritylchloride resin was coupled to unpro- 45 tected 4-amidinobenzylamine and derivatives, followed by side chains deprotection. All the final compounds were puri­ fied by both preparative and semipreparative reverse phase F1PLC, lyophilized and obtained as TFA or FlCl salts. US 9,266,828 B2 25 26

J l I" I l

.S ES O “ > K ES -¾ b i . s a 8 IJ 1 1 9 S- 0I -o S B &H ^ 1U -oC -u -¾ ^ S3 > III i %M 3a (L>S.S ,_ t I &Q I S Q > Tt O W Q S .g 3 " O & 8 & S 5 I < Q Q Uh O (n 6

O

ffi 2 I US 9,266,828 B2 27 Purity of all compounds was obtained by analytical HPLC on a Breeze system from Waters Co. using a 5 dm , 4.6x150 mm symmetry reverse phase column with a linear gradient of acetonitrile containing 0.1% TFA at a flow rate of I mL/min and by 1H NMR spectra recorded on a Bruker 600 MHz 5 instrument. The HPLC and MS profile of CompoundA is presented in FIG. I and FIG. 2. 1H NMR spectrum of Compound A in DMSO-d6 is pre­ sented in FIG. 3. to Compound B: Compound B was prepared similar to Compound A according to the reaction scheme below. All the final com­ pounds were purified by both preparative and semiprepara­ tive reverse phase HPLC, lyophilized and obtained as TFA or 15 HCl salts. US 9,266,828 B2 29 30

5 > > e 11 I,I'll 1U I Tf

rs|P

£ S I ^ s S s .a x

Z & ES S s l s SE S Q a

ffi% US 9,266,828 B2 31 HPLC of Compound B is presented in FIG. 4. 1H NMR spectrum of Compound B in deuterated PBS is presented in FIG. 5. Compound D Compound D was prepared similar to Compound A 5 according to the reaction scheme below. All the final com­ pounds were purified by both preparative and semiprepara­ tive reverse phase HPLC, lyophilized and obtained as TFA or HCl salts. US 9,266,828 B2 33 34 US 9,266,828 B2 35 MS (MALDI) and HPLC profile of Compound D is pre­ sented in FIG. 6 and FIG. 7. 1H NMR spectrum of Compound D in deuterated PBS is presented in FIG. 8. Compound E 5 Compound E was prepared by a combination of solid phase and solution synthesis. Briefly, the segment P2-P4 protected in P2, after weak acidic cleavage from the 2-chloro-tritylchlo- ride resin was coupled to unprotected 4-amidinobenzy- lamine, followed by final side chain deprotection. The final to products were purified by both preparative and semiprepara­ tive reversed phase HPLC and obtained Compound E as lyophilized powder. US 9,266,828 B2 37 38 US 9,266,828 B2 39 40 Purity of Compound E was obtained by analytical HPLC the number of viable cells present in the wells. The dye on a Breeze system from Waters Co. using a 5 pm, 4.6x150 content in each well is quantified using a 96-well spectropho­ mm symmetry reverse phase column with a linear gradient of tometer at 540 nm wavelength. The dye content in each set of acetonitrile containing 0.1% TFA at a flow rate of I mL/min wells is converted to a percentage of dye present in untreated and by 1H NMR spectra recorded on a Bruker 600 MHz 5 control wells using a Microsoft Excel computer-based instrument spreadsheet. The 50% effective (EC50, virus-inhibitory) con­ HPLC and MS profile of Compound E is presented in FIG. centrations and 50% cytotoxic (CC50, cell-inhibitory) con­ 9 and FIG. 10. centrations are then calculated by linear regression analysis. 1H NMR spectrum of Compound E in DMSO-d6 is pre­ The quotient of CC50 divided by EC50 gives the selectivity sented in FIG. 11. 10 index (SI) value. Other compounds according to the present disclosure can Secondary CPE/Virus yield reduction (VYR) assay. This be similarly prepared in light of the present disclosure and assay involves similar methodology to what is described in examples provided herein. the previous paragraphs using 96-well microplates of cells. Bioassay Example The differences are noted in this section. Eight half-log 10 Compound A 15 concentrations of inhibitor are tested for antiviral activity and Macrophage Cytotoxicity Assay. cytotoxicity. After sufficient virus replication occurs, a RAW 264.7 Murine monocyte macrophages (4.5xl04 sample of supernatant is taken from each infected well (three cells/well) were plated into 96-well tissue culture plates in replicate wells are pooled) and held for the VYR portion of Hyclone DMEM (4500 mg/L Glucose, 110 g/L Sodium Pyru­ this test, if needed. Alternately, a separate plate may be pre­ vate) supplemented with 5% fetal bovine serum, 2 mM 20 pared and the plate may be frozen for the VYR assay. After Glutamax (Invitrogen, Carlsbad, Calif.), 1% penicillin/strep­ maximum CPE is observed, the viable plates are stained with tomycin (Omega Scientific). Cells were cultured overnight at neutral red dye. The incorporated dye content is quantified as 37° C. in a humidified incubator containing 5% CO2. They described above. The data generated from this portion of the were replenished with fresh serum-free medium (0.1 ml/well) test are neutral red EC50, CC50, and SI values. Compounds and exposed to a pre-incubated solution of test compounds 25 observed to be active above are further evaluated by VYR (increasing concentrations from 0.015 to 33.3 uM), PA83 (5 00 assay. TheVYRtest is a direct determination of how much the ng/mL) and LF (37.5 ng/mL). The analyzed inhibitors were test compound inhibits virus replication. Virus that was rep­ dissolved in DMSO reaching a final DMSO concentration of licated in the presence of test compound is titrated and com­ 1%. Controls included untreated cells and LF/PA-only treated pared to virus from untreated, infected controls. Titration of cells. After incubation for 3.5 hours at 37° C., cell viability 30 pooled viral samples (collected as described above) is per­ was assessed using ATPlite assay from Perkin Elmer formed by endpoint dilution. This is accomplished by titrat­ (Waltham, Mass.). Each datum point represents triplicates of ing log 10 dilutions of virus using 3 or 4 microwells per each concentration in one experiment. Viability was normal­ dilution on fresh monolayers of cells by endpoint dilution. ized to control cells which were treated with the vehicle, Wells are scored for presence or absence of virus after distinct DMSO. Biochemical assay dose response for CompoundA is 35 CPE (measured by neutral red uptake) is observed. Plotting presented in FIG. 12. the log 10 of the inhibitor concentration versus log 10 of virus Dengue viral replication assay. Primary cytopathic effect produced at each concentration allows calculation of the 90% (CPE) reduction assay. Four-concentration CPE inhibition (one log 10) effective concentration by linear regression. assays are performed. Confluent or near-confluent cell culture Dividing EC90 by the CC50 obtained in part I of the assay monolayers in 96-well disposable microplates are prepared. 40 gives the SI value for this test. Compound A has a cellular Cells are maintained in MEM or DMEM supplemented with activity against Dengue (EC90) of 14 pm. FBS as required for each cell line. For antiviral assays the Compound B and Compound C same medium is used but with FBS reduced to 2% or less and Macrophage cytotoxicity assay. RAW 264.7 Murine supplemented with 50 pg/ml gentamicin. The test compound monocyte macrophages (4.5xl04 cells/well) were plated into is prepared at four log 10 final concentrations, usually 0.1, 45 96-well tissue culture plates in Hyclone DMEM (4500 mg/L 1.0, 10, and 100 pg/ml or pM. The virus control and cell Glucose, 110 g/L Sodium Pyruvate) supplemented with 5% control wells are on every microplate. In parallel, a known fetal bovine serum, 2 mM Glutamax (Invitrogen, Carlsbad, active drug is tested as a positive control drug using the same Calif.), 1% penicillin/streptomycin (Omega Scientific). Cells method as is applied for test compounds. The positive control were cultured overnight at 37° C. in a humidified incubator is tested with each test run. The assay is set up by first 50 containing 5% CO2. They were replenished with fresh serum- removing growth media from the 96-well plates of cells. Then free medium (0.1 ml/well) and exposed to a pre-incubated the test compound is applied in 0.1 ml volume to wells at 2x solution of test compounds (increasing concentrations from concentration. Virus, normally at <100 50% cell culture 0.015 to 33.3 uM), PA83 (500 ng/mL) and LF (37.5 ng/mL). infectious doses (CCID50) in 0.1 ml volume, is placed in The analyzed inhibitors were dissolved in DMSO reaching a those wells designated for virus infection. Medium devoid of 55 final DMSO concentration of 1%. Controls included virus is placed in toxicity control wells and cell control wells. untreated cells and LF/PA-only treated cells. After incubation Virus control wells are treated similarly with virus. Plates are for 3.5 hours at 37° C., cell viability was assessed using incubated at 37° C. with 5% C02 until maximum CPE is ATPlite assay from Perkin Elmer (Waltham, Mass.). Each observed in virus control wells. The plates are then stained datum point represents triplicates of each concentration in with 0.011% neutral red for approximately two hours at 37° 60 one experiment. Viability was normalized to control cells C. in a 5% CO2 incubator. The neutral red medium is removed which were treated with the vehicle, DMSO. by complete aspiration, and the cells may be rinsed I x with Biochemical assay dose response for Compound B and phosphate buffered solution (PBS) to remove residual dye. Compound C (with one chiral center unresolved) is presented The PBS is completely removed and the incorporated neutral in FIG. 13. red is eluted with 50% Sorensen’s citrate buffer/50% ethanol 65 Compound E (pH 4.2) for at least 30 minutes. Neutral red dye penetrates Macrophage cytotoxicity. RAW 264.7 Murine monocyte into living cells, thus, the more intense the red color, the laiger macrophages (4.5xl04 cells/well) were plated into 96-well US 9,266,828 B2 41 42 tissue culture plates in Hyclone DMEM (4500 mg/L Glucose, Biochemical assay dose response for Compound E is pre­ HO g/L Sodium Pyruvate) supplemented with 5% fetal sented in FIG. 14. bovine serum, 2 mM Glutamax (Invitrogen, Carlsbad, Calif.), Diseases and Conditions 1% penicillin/streptomycin (Omega Scientific). Cells were Viral and/or Infectious Diseases cultured overnight at 37° C. in a humidified incubator con­ 5 Disclosed herein, in certain embodiments, are methods of taining 5% CO2. They were replenished with fresh serum- neutralizing an exotoxin in a subject in need thereof compris­ free medium (0.1 ml/well) and exposed to a pre-incubated ing administering a Furin/PC inhibitors disclosed herein. In solution of test compounds (increasing concentrations from some embodiments, the exotoxin is anthrax toxin, pseudomo­ nas exotoxin A, Shiga toxin, diphtheria toxin, tetanus and 0.015 to 33.3 uM), PA83 (500 ng/mL) and LF (37.5 ng/mL). 10 botulism neurotoxins, and combinations thereof. In addition, The analyzed inhibitors were dissolved in DMSO reaching a some Furin/PC inhibitors, including those disclosed herein, final DMSO concentration of 1%. Controls included are capable of neutralizing virulence of bacteria carrying untreated cells and LF/PA-only treated cells. After incubation those exotoxin. for 3.5 hours at 37° C., cell viability was assessed using PCs, including furin, are involved in many pathogenic ATPlite assay from Perkin Elmer (Waltham, Mass.). Each 15 states as they process to maturity membrane fusion proteins datum point represents triplicates of each concentration in and toxins of a variety of both bacteria and viruses, including one experiment. Viability was normalized to control cells anthrax and botulinum toxins, influenza A H5N1 (bird flu), which were treated with the vehicle, DMSO. flaviviruses, Marbuig, influenza virus, human immunodefi­ Purification of soluble furin. Furin-overexpressing MDCK ciency virus I, Ebola, measles, cytomegalovirus, and flavivi­ cells were grown using 15 cm plates (Falcon) in DMEM/High 20 ruses (Dengue5Yellow fever, WestNile, Japaneseencephalitis modified synthetic media (Thermo Scientific) supplemented and multiple additional related flaviviruses) and parasitic with Gentamicin (10 pg/ml). Eachtwo days the medium was nematodes. collected. Cells were replenished with fresh medium. To After processing by furin and the subsequent endocytic remove cell debris, the collected samples were spun at internalization in the complex with the respective cell surface 3000xg and the supernatant fraction was filtered through a 25 receptor followed by acidification of the endosomal compart­ 0.22 pm filter (Coming) and then 100-fold concentrated using ment, the processed, partially denatured, infectious proteins a PelliconXL Biomax 10 concentrator (Millipore). Furinwas expose their membrane-penetrating peptide region and isolated from the concentrated medium samples using Ni2+- escape into the cytoplasm. The intact toxins and viral pro­ chelating chromatography on a HiTrap Chelating HP 1.6x2.5 teins, however, are incapable of accomplishing these pro­ cm column (Amersham Biosciences) equilibrated with 20 30 cesses, because they cannot penetrate the membrane and mM Tris-HCl buffer, pH 8.0, containing IM NaCl. To remove escape into the cytoplasm. the impurities, the column was washed with 50 ml of the same Cancer buffer containing 25 mM imidazole. Furin was eluted using Disclosed herein, in certain embodiments, are methods of 500 mM imidazole. The purified fractions were pooled, con­ treating cancer in a subject in need thereof comprising admin­ centrated using an Amicon Ultra 50K-cutoffmembrane (Mil­ 35 istering a Furin/PC inhibitors disclosed herein. In some lipore), dialyzed against PBS and stored at -80° C. According embodiments, the cancer is lung cancer, colon cancer, squa­ to SDS-PAGE followed by Coomassie staining of the gel, mous cell carcinoma, SCC Head and neck, skin cancer, astro­ Western blotting and enzyme activity assays the purity of the cytoma, or any combinations thereof. isolated furin samples was >95%. The typical yield of puri­ Furin and other PC family members (furin/PCs) activate fied furin was 0.8-1 mg/liter of cell culture medium. Specific 40 proteins vital to proper physiological functioning, including activity of purified furin was >120 units where one unit is the growth factors and hormones, receptors, plasma proteins, and amount of furin that will cleave 1.0 pmol methyl-coumaryl- matrix metalloproteases (MMPs). Some of the PC substrates, 7-amide (AMC) from the Pyr-Arg-Thr-Ly s-Arg-methyl-cou- such as growth factors and their receptors, matrix metallopro- maryl-7-amide (Pyr-RTKR-AMC) substrate per min at ambi­ teinases and adhesion molecules, are involved in the neoplas­ ent temperature, pH 7.5. 45 tic transformation, proliferation, invasion and metastasis for­ Enzymatic assay. Furin activity was measured in triplicate mation. In certain instances, the expression and activity of in wells of a 96-well plate in 0.2 ml 50 mM HEPES, pH 7.5, furin/PC are necessary for processing substrates important containing I mM CaCl2, 0.005% Brij-35 and 20% glycerol. for cell transformation and tumor progression, metastasis, Pyr-RTKR-AMC (25 pM) was used as a substrate. The con­ and angiogenesis. Furin processing of the remodeling pro­ centration of furin in the reactions was 50 nM. The steady- 50 tease membrane type-1 matrix metalloproteinase (MTl- state rate of substrate hydrolysis was monitored continuously MMP) enhances cellular motility and invasiveness, contrib­ (Xex=360 nm and Xem=465 nm) at 37° C. using a Spectramax uting to aggression and metastatic potential cancer cells. In Gemini EM fluorescent spectrophotometer (Molecular certain instances, overexpression and activity of furin/PC Devices). exacerbate a cancer phenotype. In certain instances, inhibi­ Determination of the IC50 values of the compounds.Furin 55 tion of furin/PC activity decreases or nullifies furin/PC-me- (50 nM) was preincubated for 30 min at 20° C. with increas­ diated effects on cancers. ing concentrations of the individual compounds in 0.1 ml of The expression of furin is higher in squamous cell carci­ 50 mM HEPES, pH 7.5, containing I mM CaCl2, 20% glyc­ nomas and adenocarcinomas, than in small-cell lung carci­ erol and 0.005% Brij 35. The Pyr-RTKR-AMC substrate (25 nomas (SCLCs). Opposite results were found for PCI and XM) was added in 0.1 ml of the same buffer. Reaction velocity 60 PC2 expression where mRNAs are absent in normal lung was monitored continuously at Xex=360 nm and Xem=465 nm epithelium and non-small cell lung cancers but over-ex- on a Spectramax Gemini EM fluorescence spectrophotom­ pressed in multiple lung cancer cell lines. eter. All assays were performed in triplicate in wells of a The expression of PCI and PC2 is altered in liver colorectal 96-well plate. IC50 values were calculated by determining the metastasis when compared to a normal liver. Moreover, PC2 concentrations of the compounds needed to inhibit 50% of the 65 overexpression was found to correlate with the expression of furin activity against Pyr-RTKR-AMC substrate. GraphPad its specific binding protein 7B2. Inhibition of PC decreased Prism was used as fitting software. proliferation and invasive ability in HT-29 human colon car­ US 9,266,828 B2 43 44 cinoma cells and tumorigenicity in xenografts. This effect reduces cleavage of the BACE propeptide. Moreover, the was linked to inhibition of IGFlR processing by furin and BACE propeptide is not processed in the furin-deficient LoVo PC5 downregulation. cell line; however, processing is restored upon furin transfec­ The up-regulation of VEGF-C by furin is associated with tion. Finally, in vitro digestion of recombinant soluble BACE squamous carcinogenesis development. Further, furin inhibi­ 5 with recombinant furin results in complete cleavage only at tion reduces of invasiveness and tumorigenicity in a FiNSCC the established E46 site. model due to decrease in processing of TGF (3 and MTl- In some embodiments, the autoimmune or inflammatory MMR disorder is: Acute disseminated encephalomyelitis; Addi­ In human primary melanoma cells, inhibition of PC leads son’s disease; Ankylosing spondylitis; Antiphospholipid to a decrease in invasiveness which correlates with the inabil­ 10 antibody syndrome; Autoimmune hemolytic anemia; ity of the cells to process PC substrates such as pro-IGFIR, Autoimmune hepatitis; Autoimmune inner ear disease; pro-PDGFA orpro-MMPs. Moreover, inhibition of PACE4 in Bullous pemphigoid; Chagas disease; Chronic obstructive skin carcinoma cells, characterized by high PACE4 activity, pulmonary disease; Coeliac disease; Dermatomyositis; Dia­ causes a decrease in both cell proliferation in vitro and tumor betes mellitus type I; Diabetes mellitus type 2; Endometrio­ development in vivo via disruption of IGFlR signaling. 15 sis; Goodpasture’s syndrome; Graves’ disease; Guillain- Furin is expressed in primary glial cell cultures and Barre syndrome; Hashimoto’s disease; Idiopathic elevated expression is seen in tumorigenic astrocytoma cell thrombocytopenic purpura; Interstitial cystitis; Systemic lines. Inhibition of furin by a I-PDX results in a decrease in lupus erythematosus (SLE); Metabolic syndrome, Multiple cell growth, an inhibition of tumorigenicity and invasion sclerosis; Myasthenia gravis; Myocarditis, Narcolepsy; Obe­ caused by inability of the cells to activate MTl-MMP and, 20 sity; Pemphigus Vulgaris; Pernicious anaemia; Polymyositis; consequently, to activate MMP-2. Furthermore, in vivo inva­ Primary biliary cirrhosis; Rheumatoid arthritis; Schizophre­ siveness is also reduced. nia; Scleroderma; Sjogren’s syndrome; Vasculitis; Vitiligo; “Cancer” includes any malignant growth or tumor caused Wegener’s granulomatosis; Allergic rhinitis; Ulcerative coli­ by abnormal and uncontrolled cell division. “Cancer” tis; Crohn’s disorder; Collagenous colitis; Lymphocytic coli­ includes solid tumors and non-solid tumors. Examples of 25 tis; Ischaemic colitis; Diversion colitis; Behfet’s syndrome; cancers include CML, CNS cancer, Flodgkin’s Disease, Infective colitis; Indeterminate colitis; Inflammatory liver NSCLC, a T-cell lymphoma, a B-cell lymphoma, adenocar­ disorder, Endotoxin shock, Rheumatoid spondylitis, Anky­ cinoma, adenocarcinoma, bladder cancer, bone cancer, brain losing spondylitis, Gouty arthritis, Polymyalgia rheumatica, cancer, brain stem glioma, breast cancer, cancer of the adrenal Alzheimer’s disorder, Parkinson’s disorder, Epilepsy, AIDS gland, cancer of the anal region, cancer of the bladder, cancer 30 dementia, Asthma, Adult respiratory distress syndrome, of the endocrine system, cancer of the esophagus, cancer of Bronchitis, Cystic fibrosis, Acute leukocyte-mediated lung the parathyroid gland, cancer of the penis, cancer of the small injury, Distal proctitis, Wegener’s granulomatosis, Fibromy­ intestine, cancer of the thyroid gland, cancer of the urethra, algia, Bronchitis, Cystic fibrosis, Uveitis, Conjunctivitis, carcinoma of the cervix, carcinoma of the endometrium, car­ Psoriasis, Eczema, Dermatitis, Smooth muscle proliferation cinoma of the fallopian tubes, carcinoma of the renal pelvis, 35 disorders, Meningitis, Shingles, Encephalitis, Nephritis, carcinoma of the vagina, carcinoma of the vulva, colon can­ Tuberculosis, Retinitis, Atopic dermatitis, Pancreatitis, Peri­ cer, cutaneous or intraocular melanoma, gastric cancer, gas­ odontal gingivitis, Coagulative Necrosis, Liquefactive trointestinal stromal tumors, gastrointestinal stromal tumors, Necrosis, Fibrinoid Necrosis, Hyperacute transplant rejec­ glioblastoma, head and neck cancer, hepatocellular cancer, tion, Acute transplant rejection, Chronic transplant rejection, kidney cancer, leukemia, lung cancer, lymphocytic lympho­ 40 Acute graft-versus-host disease, Chronic graft-versus-host mas, lymphoma, melanoma, meningiomas, myeloma, neu­ disease, or combinations thereof. rofibromatosis, renal cell carcinoma, ovarian cancer, pancre­ Pharmaceutical Compositions and Methods of Administra­ atic cancer, pituitary adenoma, primary CNS lymphoma, tion prostate cancer, rectal cancer, renal cell carcinoma, sarcoma Provided herein, in certain embodiments, are pharmaceu­ of soft tissue, spinal axis tumors, spontaneous schwannomas, 45 tical compositions comprising a therapeutically effective stomach cancer, uterine cancer, or any combinations thereof. amount of a Furin/PC inhibitor disclosed herein and a phar- Inflammatory and Autoimmune Disorders maceutically-acceptable excipient. Disclosed herein, in certain embodiments, are methods of Pharmaceutical compositions are formulated using one or treating an inflammatory or autoimmune disorder in a subject more physiologically acceptable excipients. Proper formula­ in need thereof comprising administering a furin/PC inhibitor 50 tion is dependent upon the route of administration chosen. A disclosed herein. In some embodiments, the inflammatory or summary of pharmaceutical compositions is found, for autoimmune disorder is Alzheimer’s Disease, arthritis, ath­ example, in Remington: The Science and Practice of Phar­ erosclerosis, or any combinations thereof. macy, Nineteenth Ed (Ea hston, Pa.: Mack Publishing Com­ The novel transmembrane aspartic protease BACE (for pany, 1995); Hoover, John E., Remington’s Pharmaceutical Beta-site APP CleavingEnzyme) is the (3-secretase that 55 Sciences, MackPublishingCo., Easton, Pa. 1975; Liberman, cleaves amyloid precursor protein to initiate (3-amyloid for­ H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, mation. As such, BACE is a prime therapeutic target for the Marcel Decker, New York, N.Y., 1980; and Pharmaceutical treatment of Alzheimer’s disease. BACE, like other aspartic Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lip- proteases, has a propeptide domain that is removed to form pincott Williams & Wilkins, 1999). the mature enzyme. BACE propeptide cleavage occurs at the 60 In some embodiments, the pharmaceutically compositions sequence RLPRjE. BACE and furin co-localize within the further comprise a pharmaceutically acceptable diluent(s), Golgi apparatus, and propeptide cleavage is inhibited by excipient(s), or carrier(s). In addition, the Furin/PC inhibitors brefeldin A and monensin, drugs that disrupt trafficking is optionally administered as pharmaceutical compositions in through the Golgi. Treatment of cells with the calcium iono- which it is mixed with other active ingredients, as in combi­ phore A23187, leading to inhibition of calcium-dependent 65 nation therapy. In some embodiments, the pharmaceutical proteases including furin, or transfection with the a l -antit­ compositions includes other medicinal or pharmaceutical rypsin variant a l -PDX, a potent furin inhibitor, dramatically agents, carriers, adjuvants, such as preserving, stabilizing, US 9,266,828 B2 45 46 wetting or emulsifying agents, solution promoters, salts for the like) and spray drying the solution. The solvent rapidly regulating the osmotic pressure, and/or buffers. In addition, evaporates from droplets which rapidly solidifies the polymer the pharmaceutical compositions also contain other therapeu­ and drug mixture trapping the drug in amorphous form as an tically valuable substances. amorphous molecular dispersion. In some embodiments, The pharmaceutical formulations described herein are 5 such amorphous dispersions are filled in capsules and/or con­ optionally administered to an individual by any suitable stituted into oral powders for reconstitution. Solubility of an administration route, including but not limited to, oral, SDD comprising a drug is higher than the solubility of a parenteral (e.g., intravenous, subcutaneous, intramuscular), crystalline form of a drug or a non-SDD amorphous form of intranasal, buccal, topical, rectal, or transdermal administra­ a drug. tion routes. The pharmaceutical formulations described 10 Pharmaceutical preparations for oral use are optionally herein include, but are not limited to, aqueous liquid disper­ obtained by mixing one or more solid excipients with a Furin/ sions, self-emulsifying dispersions, solid solutions, liposo­ PC inhibitor disclosed herein, optionally grinding the result­ mal dispersions, aerosols, solid dosage forms, powders, ing mixture, and processing the mixture of granules, after immediate release formulations, controlled release formula­ adding suitable auxiliaries, if desired, to obtain tablets or tions, fast melt formulations, tablets, capsules, pills, delayed 15 dragee cores. Suitable excipients include, for example, fillers release formulations, extended release formulations, pulsatile such as sugars, including lactose, sucrose, mannitol, or sor­ release formulations, multiparticulate formulations, and bitol; cellulose preparations such as, for example, maize mixed immediate and controlled release formulations. starch, wheat starch, rice starch, potato starch, gelatin, gum In some embodiments, the pharmaceutical compositions tragacanth, methylcellulose, microcrystalline cellulose, comprise at least one Furin/PC inhibitor disclosed herein, as 20 hydroxypropylmethylcellulose, sodium carboxymethylcel- an active ingredient in free-acid or free-base form, or in a lulose; or others such as: polyvinylpyrrolidone (PVP or povi­ pharmaceutically acceptable salt form. In addition, the meth­ done) or calcium phosphate. If desired, disintegrating agents ods and pharmaceutical compositions described herein are added, such as the cross linked croscarmellose sodium, include the use of N-oxides, crystalline forms (also known as polyvinylpyrrolidone, agar, or alginic acid or a salt thereof polymorphs), as well as active metabolites of these Furin/PC 25 such as sodium alginate. inhibitors having the same type of activity. In some embodi­ Dragee cores are provided with suitable coatings. For this ments, Furin/PC inhibitors disclosed herein exist as tau­ purpose, concentrated sugar solutions are generally used, tomers. All tautomers are included within the scope of the which optionally contain gum arabic, talc, polyvinylpyrroli­ Furin/PC inhibitors disclosed herein. Additionally, the Furin/ done, carbopol gel, polyethylene glycol, and/or titanium PC inhibitors exist in unsolvated as well as solvated forms 30 dioxide, lacquer solutions, and suitable organic solvents or with pharmaceutically acceptable solvents such as water, solvent mixtures. Dyestuffs or pigments are optionally added ethanol, and the like. The solvated forms of the Furin/PC to the tablets or dragee coatings for identification. inhibitors presented herein are also considered to be disclosed In some embodiments, a composition disclosed herein is herein. formulated as a solid dosage form. In some embodiments, a “Carrier materials” include any commonly used excipients 35 Furin/PC inhibitor disclosed here is a tablet, (including a in pharmaceutics and should be selected on the basis of com­ suspension tablet, a fast-melt tablet, a bite-disintegration tab­ patibility with a Furin/PC inhibitor disclosed herein and the let, a rapid-disintegration tablet, an effervescent tablet, or a release profile properties of the desired dosage form. Exem­ caplet), a pill, a powder (including a sterile packaged powder, plary carrier materials include, e.g., binders, suspending a dispensable powder, or an effervescent powder) a capsule agents, disintegration agents, filling agents, surfactants, solu­ 40 (including both soft or hard capsules, e.g., capsules made bilizers, stabilizers, lubricants, wetting agents, diluents, and from animal-derived gelatin or plant-derived HPMC, or the like. “sprinkle capsules”), solid dispersion, solid solution, bio- Moreover, the pharmaceutical compositions described erodible dosage form, controlled release formulations, pulsa­ herein are formulated into any suitable dosage form, includ­ tile release dosage forms, multiparticulate dosage forms, pel­ ing but not limited to, aqueous oral dispersions, liquids, gels, 45 lets, granules, or an aerosol. In some embodiments, a Furin/ syrups, elixirs, slurries, suspensions and the like, for oral PC inhibitor disclosed here is a capsule. In some ingestion by a patient to be treated, solid oral dosage forms, embodiments, a Furin/PC inhibitor disclosed here is a pow­ aerosols, controlled release formulations, fast melt formula­ der. tions, effervescent formulations, lyophilized formulations, In some embodiments, a pharmaceutical composition dis­ tablets, powders, pills, dragees, capsules, delayed release for­ 50 closed herein is a microencapsulated formulation. In some mulations, extended release formulations, pulsatile release embodiments, one or more other compatible materials are formulations, multiparticulate formulations, and mixed present in the microencapsulation material. Exemplary mate­ immediate release and controlled release formulations. rials include, but are not limited to, pH modifiers, erosion In some embodiments, the pharmaceutical compositions facilitators, anti-foaming agents, antioxidants, flavoring disclosed herein are solid drug dispersions. A solid dispersion 55 agents, and carrier materials such as binders, suspending is a dispersion of one or more active ingredients in an inert agents, disintegration agents, filling agents, surfactants, solu­ carrier or matrix at solid state prepared by the melting (or bilizers, stabilizers, lubricants, wetting agents, and diluents. fusion), solvent, or melting-solvent methods (Chiou and Exemplary microencapsulation materials useful for delay­ Riegelman, Journal of Pharmaceutical Sciences, 60, 1281 ing the release of the formulations including a Furin/PC (1971)). The dispersion of one or more active agents in a solid 60 inhibitors, include, but are not limited to, hydroxypropyl cel­ diluent is achieved without mechanical mixing. Solid disper­ lulose ethers (HPC) such as Klucel® or Nisso HPC, low- sions are also called solid-state dispersions. substituted hydroxypropyl cellulose ethers (L-HPC), hydrox­ In some embodiments, the pharmaceutical compositions ypropyl methyl cellulose ethers (HPMC) such as Seppifilm- disclosed herein are spray dried dispersions (SDD). An SDD LC, Pharmacoat®, Metolose SR, Methocel®-E, Opadry YS, is a single phase amorphous molecular dispersion of a drug in 65 PrimaFlo, Benecel MP824, and Benecel MP843, methylcel­ a polymer matrix. It is a solid solution prepared by dissolving lulose polymers such as Methocel®-A, hydroxypropylmeth­ the drug and a polymer in a solvent (e.g., acetone, methanol or ylcellulose acetate stearate Aqoat (HF-LS, HF-LG, HF-MS) US 9,266,828 B2 47 48 and Metolose®, Ethylcelluloses (EC) and mixtures thereof In some embodiments, a pharmaceutical composition such as E461, Ethocel®, Aqualon®-EC, Surelease®, Poly­ described herein is formulated for nasal administration. Nasal vinyl alcohol (PVA) such as Opadry AMB, hydroxyethylcel- dosage forms generally contain large amounts of water in luloses such as Natrosol®, carboxymethylcelluloses and salts addition to the active ingredient. Minor amounts of other of carboxymethylcelluloses (CMC) such as Aqualon®- 5 ingredients such as pFl adjusters, emulsifiers or dispersing CMC, polyvinyl alcohol and polyethylene glycol co-poly- agents, preservatives, surfactants, gelling agents, or buffering mers such as Kollicoat IR®, monoglycerides (Myverol), trig­ and other stabilizing and solubilizing agents are optionally lycerides (KLX), polyethylene glycols, modified food starch, present. acrylic polymers and mixtures of acrylic polymers with cel­ In some embodiments, a pharmaceutical compositions dis­ lulose ethers such as Eudragit® EPO, Eudragit® L30D-55, 10 closed herein is an aerosol, a mist or a powder. In some Eudragit® FS 30D Eudragit® LlOO-55, Eudragit® LlOO, embodiments, a pharmaceutical composition described Eudragit® S 100, Eudragit® RD 100, Eudragit® E100, herein is delivered in the form of an aerosol spray presenta­ Eudragite® L12.5, Eudragit® 512.5, Eudragit® NE30D, and Eudragit® NE 40D, cellulose acetate phthalate, sepifilms tion from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorof- such as mixtures of ElPMC and stearic acid, cyclodextrins, 15 and mixtures of these materials. luoromethane, dichlorotetrafluoroethane, carbon dioxide or In some embodiments, a pharmaceutical composition dis­ other suitable gas. In the case of a pressurized aerosol, the closed herein is formulated to provide controlled release of a dosage unit is determined by providing a valve to deliver a Furin/PC inhibitor disclosed herein. Controlled release pro­ metered amount. Capsules and cartridges of, such as, by way files include, for example, sustained release, prolonged 20 of example only, gelatin for use in an inhaler or insufflator are release, pulsatile release, and delayed release profiles. In con­ formulated containing a powder mix of a Furin/PC inhibitor trast to immediate release compositions, controlled release disclosed herein and a suitable powder base such as lactose or compositions allow delivery of an agent to an individual over starch. an extended period of time according to a predetermined In some embodiments, a pharmaceutical composition profile. Such release rates provide therapeutically effective 25 described herein is formulated for buccal administration. levels of agent for an extended period of time and thereby Buccal formulations that include a Furin/PC inhibitor dis­ provide a longer period of pharmacologic response while closed herein include, but are not limited to, U.S. Pat. Nos. minimizing side effects as compared to conventional rapid 4,229,447,4,596,795, 4,755,386, and 5,739,136. In addition, release dosage forms. Such longer periods of response pro­ the buccal dosage forms described herein optionally further vide for many inherent benefits that are not achieved with the 30 include a bioerodible (hydrolysable) polymeric carrier that corresponding short acting, immediate release preparations. also serves to adhere the dosage form to the buccal mucosa. In some embodiments, a composition disclosed herein is The buccal dosage form is fabricated so as to erode gradually formulated as a pulsatile dosage form. over a predetermined time period, wherein the delivery of the In some embodiments, a composition disclosed herein is Furin/PC inhibitors, is provided essentially throughout. Buc­ formulated as a liquid dosage form. In some embodiments, a 35 cal drug delivery avoids the disadvantages encountered with pharmaceutical composition disclosed herein is an aqueous oral drug administration, e.g., slow absorption, degradation suspension selected from the group including, but not limited of the active agent by fluids present in the gastrointestinal to, pharmaceutically acceptable aqueous oral dispersions, tract and/or first-pass inactivation in the liver. The bioerodible emulsions, solutions, elixirs, gels, and syrups. See, e.g., Singh (hydrolysable) polymeric carrier generally comprises hydro­ et ah, Encyclopedia of Pharmaceutical Technology, 2nd Ed., 40 philic (water-soluble and water-swellable) polymers that pp. 754-757 (2002). In addition, the liquid dosage forms adhere to the wet surface of the buccal mucosa. Examples of optionally include additives, such as: (a) disintegrating polymeric carriers useful herein include acrylic acid poly­ agents; (b) dispersing agents; (c) wetting agents; (d) at least mers and co, e.g., those known as “carbomers” (Carbopol®, one preservative, (e) viscosity enhancing agents, (f) at least which may be obtained from B.F. Goodrich, is one such one sweetening agent, and (g) at least one flavoring agent. In 45 polymer). Other components also be incorporated into the some embodiments, the aqueous dispersions further includes buccal dosage forms described herein include, but are not a crystal-forming inhibitor. limited to, disintegrants, diluents, binders, lubricants, flavor­ In some embodiments, a pharmaceutical formulation ing, colorants, preservatives, and the like. For buccal or sub­ described herein is a self-emulsifying drug delivery systems lingual administration, the compositions optionally take the (SEDDS). Emulsions are dispersions of one immiscible 50 form of tablets, lozenges, or gels formulated in a conventional phase in another, usually in the form of droplets. Generally, manner. By way of example, Examples 26c and 26d describe emulsions are created by vigorous mechanical dispersion. sublingual formulations. SEDDS, as opposed to emulsions or microemulsions, spon­ In some embodiments, a pharmaceutical composition taneously form emulsions when added to an excess of water described herein is formulated for transdermal administra­ without any external mechanical dispersion or agitation. An 55 tion. advantage of SEDDS is that only gentle mixing is required to In some embodiments, a transdermal formulation distribute the droplets throughout the solution. Additionally, described herein comprises: (I) a Furin/PC inhibitor dis­ water or the aqueous phase is optionally added just prior to closed herein; (2) a penetration enhancer; and (3) an aqueous administration, which ensures stability of an unstable or adjuvant. In addition, transdermal formulations include com­ hydrophobic active ingredient. Thus, the SEDDS provides an 60 ponents such as, but not limited to, gelling agents, creams and effective delivery system for oral and parenteral delivery of ointment bases, and the like. In some embodiments, the trans­ hydrophobic active ingredients. In some embodiments, dermal formulation further includes a woven or non-woven SEDDS provides improvements in the bioavailability of backing material to enhance absorption and prevent the hydrophobic active ingredients. Methods of producing self- removal of the transdermal formulation from the skin. In emulsifying dosage forms include, but are not limited to, for 65 other embodiments, the transdermal formulations described example, U.S. Pat. Nos. 5,858,401, 6,667,048, and 6,960, herein maintain a saturated or supersaturated state to promote 563. diffusion into the skin. US 9,266,828 B2 49 50 In some embodiments, formulations suitable for transder- lotions, gels, pastes, medicated sticks, balms, creams or oint­ mal administration of a Furin/PC inhibitor disclosed herein ments. Suchpharmaceutical compositions optionally contain employ transdermal delivery devices and transdermal deliv­ solubilizers, stabilizers, tonicity enhancing agents, buffers ery patches and are lipophilic emulsions or buffered, aqueous and preservatives. solutions, dissolved and/or dispersed in a polymer or an adhe­ 5 The Furin/PC inhibitors is also optionally formulated in sive. Such patches are optionally constructed for continuous, rectal compositions such as enemas, rectal gels, rectal foams, pulsatile, or on demand delivery of pharmaceutical agents. rectal aerosols, suppositories, jelly suppositories, orretention Still further, transdermal delivery of a Furin/PC inhibitor enemas, containing conventional suppository bases such as disclosed herein is optionally accomplished by means of ion- cocoa butter or other glycerides, as well as synthetic polymers tophoretic patches and the like. Additionally, transdermal 10 such as polyvinylpyrrolidone, PEG, and the like. In supposi­ patches provide controlled delivery of a Furin/PC inhibitor. tory forms of the compositions, a low-melting wax such as, The rate of absorption is optionally slowed by using rate­ but not limited to, a mixture of fatty acid glycerides, option­ controlling membranes or by trapping a Furin/PC inhibitor ally in combination with cocoa butter is first melted. within a polymer matrix or gel. Conversely, absorption Combinations enhancers are used to increase absorption. An absorption 15 In certain instances, it is appropriate to a Furin/PC inhibitor enhancer or carrier includes absorbable pharmaceutically disclosed herein in combination with an additional therapeu­ acceptable solvents to assist passage through the skin. For tic agent. Additional therapeutic agents are selected for their example, transdermal devices are in the form of a bandage particular usefulness against the condition that is being comprising a backing member, a reservoir containing a Furin/ treated. In general, the additional therapeutic agent does not PC inhibitor optionally with carriers, optionally a rate con­ 20 need to be administered in the same pharmaceutical compo­ trolling barrier to deliver the Furin/PC inhibitors to the skin of sition, at the same time or via the same route and the Furin/PC the host at a controlled and predetermined rate over a pro­ inhibitor disclosed herein. In one embodiment, the initial longed period of time, and means to secure the device to the administration is made according to established protocols, skin. and then, based upon the observed effects, the dosage, modes Formulations that include a Furin/PC inhibitor disclosed 25 of administration and times of administration, further modi­ herein suitable for intramuscular, subcutaneous, or intrave­ fied. nous injection include physiologically acceptable sterile In some embodiments, the additional therapeutic agent is aqueous or non-aqueous solutions, dispersions, suspensions administered concurrently (e.g., simultaneously, essentially or emulsions, and sterile powders for reconstitution into ster­ simultaneously or within the same treatment protocol) or ile injectable solutions or dispersions. Examples of suitable 30 sequentially, depending upon the nature of the disease, the aqueous and non-aqueous carriers, diluents, solvents, or condition of the patient, and the actual choice of compounds vehicles including water, ethanol, polyols (propyleneglycol, used. In certain embodiments, the determination of the order polyethylene-glycol, glycerol, cremophor and the like), suit­ of administration, and the number of repetitions of adminis­ able mixtures thereof, vegetable oils (such as olive oil) and tration of each therapeutic agent during a treatment protocol, injectable organic esters such as ethyl oleate. Proper fluidity 35 is based upon evaluation of the disease being treated and the is maintained, for example, by the use of a coating such as condition of the patient. lecithin, by the maintenance of the required particle size in the The dose of the additional therapeutic agent varies depend­ case of dispersions, and by the use of surfactants. Formula­ ing on the additional therapeutic agent, the disease or condi­ tions suitable for subcutaneous injection also contain optional tion being treated and so forth. additives such as preserving, wetting, emulsifying, and dis­ 40 In some embodiments, the additional therapeutic agent is a pensing agents. chemotherapeutic agent, a steroid, an immunotherapeutic For intravenous injections, a Furin/PC inhibitor disclosed agent, a targeted therapy, or a combination thereof. In some herein is optionally formulated in aqueous solutions, prefer­ embodiments, the additional therapeutic agent is a B cell ably in physiologically compatible buffers such as Hank’s receptor pathway inhibitor. In some embodiments, the B cell solution, Ringer’s solution, or physiological saline buffer. For 45 receptor pathway inhibitor is a CD79A inhibitor, a CD79B transmucosal administration, penetrants appropriate to the inhibitor, a CDl 9 inhibitor, a Lyn inhibitor, a Syk inhibitor, a barrier to be permeated are used in the formulation. For other PI3K inhibitor, a Blnk inhibitor, a PLCy inhibitor, a PKC|3 parenteral injections, appropriate formulations include aque­ inhibitor, or a combination thereof. In some embodiments, ous ornonaqueous solutions, preferably with physiologically the additional therapeutic agent is an antibody, B cell receptor compatible buffers or excipients. 50 signaling inhibitor, a PI3K inhibitor, an IAP inhibitor, an Parenteral injections optionally involve bolus injection or mTOR inhibitor, a radioimmunotherapeutic, a DNA damag­ continuous infusion. Formulations for injection are option­ ing agent, a proteosome inhibitor, a histone deacefylase ally presented in unit dosage form, e.g., in ampoules or in inhibitor, a protein kinase inhibitor, a hedgehog inhibitor, an multi dose containers, with an added preservative. In some Hsp90 inhibitor, a telomerase inhibitor, a Jakl/2 inhibitor, a embodiments, a pharmaceutical composition described 55 protease inhibitor, a PKC inhibitor, a PARP inhibitor, or a herein is in a form suitable for parenteral injection as a sterile combination thereof. suspensions, solutions or emulsions in oily or aqueous Additional therapeutic agents that maybe administered in vehicles, and contain formulatory agents such as suspending, conjunction with a Furin/PC inhibitor disclosed herein stabilizing and/or dispersing agents. Pharmaceutical formu­ include, but are not limited to, Nitrogen Mustards such as for lations for parenteral administration include aqueous solu­ 60 example, bendamustine, chlorambucil, chlormethine, cyclo­ tions of a Furin/PC inhibitor disclosed herein in water soluble phosphamide, ifosfamide, melphalan, prednimustine, trofos- form. Additionally, suspensions of a Furin/PC inhibitor dis­ famide; Alkyl Sulfonates like busulfan, mannosulfan, treo- closed herein is optionally prepared as appropriate oily injec­ sulfan; Ethylene Imines like carboquone, thiotepa, tion suspensions. triaziquone; Nitrosoureas like carmustine, fotemustine, In some embodiments, the Furin/PC inhibitors is adminis­ 65 lomustine, nimustine, ranimustine, semustine, streptozocin; tered topically and formulated into a variety of topically Epoxides such as for example, etoglucid; Other Alkylating administrable compositions, such as solutions, suspensions, Agents such as for example dacarbazine, mitobronitol, pipo- US 9,266,828 B2 51 52 broman, temozolomide; Folic Acid Analogues such as for melanoma , , pegademase, , example methotrexate, permetrexed, pralatrexate, raltitrexed; , poly TC, poly ICLC, , tasonermin, thy­ Purine Analogs such as for example cladribine, clofarabine, mopentin; Immunosuppressants such as for example abata- fludarabine, mercaptopurine, nelarabine, tioguanine; Pyrimi­ cept, abetimus, alefacept, antilymphocyte immunoglobulin dine Analogs such as for example azacitidine, capecitabine, 5 (horse), antithymocyte immunoglobulin (rabbit), eculi- carmofur, cytarabine, decitabine, fluorouracil, gemcitabine, zumab, efalizumab, everolimus, gusperimus, leflunomide, tegafur; Vinca Alkaloids such as for example vinblastine, muromab-CD3, mycophenolic acid, natalizumab, sirolimus; vincristine, vindesine, vinflunine, vinorelbine; Podophyllo- TNF alpha Inhibitors such as for example adalimumab, afe- toxin Derivatives such as for example etoposide, teniposide; limomab, certolizumab pegol, etanercept, golimumab, inflix­ Colchicine derivatives such as for example demecolcine; 10 imab; Inhibitors such as for example anakinra, Taxanes such as for example docetaxel, paclitaxel, paclitaxel basiliximab, canakinumab, daclizumab, mepolizumab, poliglumex; OtherPlantAlkaloids andNatural Products such rilonacept, tocilizumab, ustekinumab; Calcineurin Inhibitors as for example trabectedin; Actinomycines such as for such as for example ciclosporin, tacrolimus; Other Immuno­ example dactinomycin; Antracyclines such as for example suppressants such as for example azathioprine, lenalidomide, aclambicin, daunorubicin, doxorubicin, epirubicin, idarubi- 15 methotrexate, thalidomide. cin, mitoxantrone, pirambicin, valrabicin, zorubincin; Other Further therapeutic agents that maybe administered in con­ Cytotoxic Antibiotics such as for example bleomycin, ixa- junction with a Furin/PC inhibitor disclosed herein include, bepilone, mitomycin, plicamycin; Platinum Compounds but are not limited to, Adalimumab, Alemtuzumab, Basilix­ such as for example carboplatin, cisplatin, oxaliplatin, satra- imab, Bevacizumab, Cetuximab, Certolizumab pegol, Dacli­ platin; Methylhydrazines such as for example procarbazine; 20 zumab, Eculizumab, Efalizumab, Gemtuzumab, Ibritumo- Sensitizers such as for example aminolevulinic acid, efaprox- mab tiuxetan, Infliximab, Muromonab-CD3, Natalizumab, iral, methyl aminolevulinate, porfimer sodium, temoporfin; Panitumumab, Ranibizumab, Rituximab, Tositumomab, Protein Kinase Inhibitors such as for example dasatinib, erlo- Trastuzumab, or the like, or a combination thereof. tinib, everolimus, gefitinib, imatinib, lapatinib, nilotinib, Additional therapeutic agents that maybe administered in pazonanib, sorafenib, sunitinib, temsirolimus; Other Antine­ 25 conjunction with a Furin/PC inhibitor disclosed herein oplastic Agents such as for example alitretinoin, altretamine, include, but are not limited to, Monoclonal Antibodies such as amzacrine, anagrelide, arsenic trioxide, asparaginase, bex- for example alemtuzumab, bevacizumab, catumaxomab, arotene, bortezomib, celecoxib, denileukin diftitox, estra- cetuximab, edrecolomab, gemtuzumab, ofatumumab, panitu­ mustine, hydroxycarbamide, irinotecan, lonidamine, maso- mumab, rituximab, trastuzumab, Immunosuppressants, ecu­ procol, miltefosein, mitoguazone, mitotane, oblimersen, 30 lizumab, efalizumab, muromab-CD3, natalizumab; TNF pegaspargase, pentostatin, romidepsin, sitimagene ceraden- alpha Inhibitors such as for example adalimumab, afelimo- ovec, tiazofurine, topotecan, tretinoin, vorinostat; Estrogens mab, certolizumab pegol, golimumab, infliximab, Interleukin such as for example diethylstilbenol, ethinylestradiol, fos- Inhibitors, basiliximab, canakinumab, daclizumab, mepoli­ festrol, polyestradiol phosphate; Progestagens such as for zumab, tocilizumab, ustekinuma, Radiopharmaceuticals, example gestonorone, medroxyprogesterone, megestrol; 35 ibritumomab tiuxetan, tositumomab; Others Monoclonal Gonadotropin Releasing Flormone Analogs such as for Antibodies such as for example abagovomab, adecatu- example buserelin, goserelin, leuprorelin, triptorelin; Anti- mumab, alemtuzumab, anti-CD30 monoclonal antibody Estrogens such as for example fulvestrant, tamoxifen, Xmab2513, anti-MET monoclonal antibody MetMab, apoli- toremifene; Anti-Androgens such as for example bicaluta- zumab, apomab, arcitumomab, basiliximab, bispecific anti­ mide, flutamide, nilutamide, Enzyme Inhibitors, aminoglute- 40 body 2B1, blinatumomab, brentuximab vedotin, capromab thimide, anastrozole, exemestane, formestane, letrozole, pendetide, cixutumumab, claudiximab, conatumumab, vorozole; Other Flormone Antagonists such as for example dacetuzumab, denosumab, eculizumab, epratuzumab, abarelix, degarelix; such as for example epratuzumab, ertumaxomab, etaracizumab, figitumumab, histamine dihydrochloride, mifamurtide, pidotimod, plerix- fresolimumab, galiximab, ganitumab, gemtuzumab ozo- afor, roquinimex, ; Immunosuppressants such as 45 gamicin, glembatumumab, ibritumomab, inotuzumab ozo- for example everolimus, gusperimus, leflunomide, mycophe- gamicin, ipilimumab, lexatumumab, lintuzumab, lintu- nolic acid, sirolimus; Calcineurin Inhibitors such as for zumab, lucatumumab, mapatumumab, matuzumab, example ciclosporin, tacrolimus; Other Immunosuppressants milatuzumab, monoclonal antibody CC49, necitumumab, such as for example azathioprine, lenalidomide, methotrex­ nimotuzumab, ofatumumab, oregovomab, pertuzumab, ate, thalidomide; and Radiopharmaceuticals such as for 50 ramacurimab, ranibizumab, siplizumab, sonepcizumab, tan- example, iobenguane. ezumab, tositumomab, trastuzumab, tremelimumab, tucotu- Further therapeutic agents that maybe administered in con­ zumab celmoleukin, veltuzumab, visilizumab, volociximab, junction with a Furin/PC inhibitor disclosed herein include, zalutumumab. but are not limited to , , Tumor Necro­ Further therapeutic agents that maybe administered in con­ sis Factors, Growth Factors, or the like. 55 junction with a Furin/PC inhibitor disclosed herein include, Additional therapeutic agents that maybe administered in but are not limited to, agents that affect the tumor micro­ conjunction with a Furin/PC inhibitor disclosed herein environment such as cellular signaling network (e.g. phos- include, but are not limited to, Immunostimulants such as for phatidylinositol 3-kinase (PI3K) signaling pathway, signal­ example , , , , ing from the B-cell receptor and the IgE receptor). In some , ; Interferons such as for example 60 embodiments, the second agent is a PI3K signaling inhibitor alfa natural, -2a, interferon alfa-2b, or a syc kinase inhibitor. In one embodiment, the syk inhibitor interferon alfacon-1, interferon alfa-nl, interferon beta natu­ is R788. In another embodiment is a PKCy inhibitor such as ral, interferon beta-1 a, interferon beta-lb, , by way of example only, enzastaurin. peginterferon alfa-2a, peginterferon alfa-2b; Interleukins Examples of agents that affect the tumor micro-environ­ such as for example aldesleukin, ; Other Immuno- 65 ment include PI3K signaling inhibitor, syc kinase inhibitor, stimulants such as for example BCG vaccine, glatiramer Protein Kinase Inhibitors such as for example dasatinib, erlo- acetate, histamine dihydrochloride, immunocyanin, , tinib, everolimus, gefitinib, imatinib, lapatinib, nilotinib, US 9,266,828 B2 53 54 pazonanib, sorafenib, sunitinib, temsirolimus; Other Angio­ pegaspargase; peliomycin; pentamustine; peplomycin sul­ genesis Inhibitors such as for example GT-111, JI-101, fate; perfosfamide; pipobroman; piposulfan; piroxantrone Rl 530; Other Kinase Inhibitors such as for example AC220, hydrochloride; plicamycin; plomestane; porfimer sodium; AC480, ACE-041, AMG 900, AP24534, Arry-614, AT7519, porfiromycin; prednimustine; procarbazine hydrochloride; AT9283, AV-951, axitinib, AZDl 152, AZD7762, AZD8055, 5 puromycin; puromycin hydrochloride; pyrazofurin; ribo- AZD8931, bafetinib, BAY 73-4506, BGJ398, BGT226, BI prine; rogletimide; safingol; safingol hydrochloride; semus- 811283, BI6727, BIBF 1120, BIBW 2992, BMS-690154, tine; simtrazene; sparfosate sodium; sparsomycin; spiroger- BMS-777607, BMS-863233, BSK-461364, CAL-101, CEP- manium hydrochloride; spiromustine; spiroplatin; 11981, CYCl 16, DCC-2036, dinaciclib, dovitinib lactate, streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan E7050, EMD 1214063, ENMD-2076, fostamatinib diso­ 10 sodium; tegafur; teloxantrone hydrochloride; temoporfin; dium, GSK2256098, GSK690693, INCB18424, INNO-406, teniposide; teroxirone; testolactone; thiamiprine; thiogua- JNJ-26483327, JX-594, KX2-391, linifanib, LY2603618, nine; thiotepa; tiazofurin; tirapazamine; toremifene citrate; MGCD265, MK-0457, MKl 496, MLN8054, MLN8237, trestolone acetate; triciribine phosphate; trimetrexate; trime- MP470, NMS-1116354, NMS-1286937, ON 01919.Na, OSI- trexate glucuronate; triptorelin; tubulozole hydrochloride; 027, OSI-930, Btk inhibitor, PF-00562271, PF-02341066, 15 uracil mustard; uredepa; vapreotide; verteporfin; vinblastine PF-03814735, PF-04217903, PF-04554878, PF-04691502, sulfate; vincristine sulfate; vindesine; vindesine sulfate; vine- PF-3758309, PHA-739358, PLC3397, progenipoietin, R547, pidine sulfate; vinglycinate sulfate; vinleurosine sulfate; R763, ramucirumab, regorafenib, R05185426, SAR103168, vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; SCH 727965, SGI-1176, SGX523, SNS-314, TAK-593, vorozole; zeniplatin; zinostatin; zorubicin hydrochloride. TAK-901, TKI258, TLN-232, TTP607, XL147, XL228, 20 Further therapeutic agents that maybe administered in con­ XL281R05126766, XL418, XL765. junction with a Furin/PC inhibitor disclosed herein include, Further examples of therapeutic agents for use in combi­ but are not limited to, 20-epi-l, 25 dihydroxyvitamin D3; nation with a Furin/PC inhibitor disclosed herein include, but 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; ade- are not limited to, inhibitors of mitogen-activated protein cypenol; adozelesin; aldesleukin; ALL-TK antagonists; altre­ kinase signaling, e.g., U0126, PD98059, PD184352, 25 tamine; ambamustine; amidox; amifostine; aminolevulinic PD0325901, ARRY-142886, SB239063, SP600125, BAY acid; amrubicin; amsacrine; anagrelide; anastrozole; 43-9006, wortmannin, or LY294002; Syk inhibitors; mTOR andrographolide; angiogenesis inhibitors; antagonist D; inhibitors; and antibodies (e.g., rituxan). antagonist G; antarelix; anti-dorsalizing morphogenetic pro­ Other agents that may be employed in combination with a tein-1; antiandrogen, prostatic carcinoma; antiestrogen; anti- Furin/PC inhibitor disclosed herein include, but are not lim­ 30 neoplaston; antisense oligonucleotides; aphidicolin glyci- ited to, Adriamycin, Dactinomycin, Bleomycin, Vinblastine, nate; apoptosis gene modulators; apoptosis regulators; Cisplatin, acivicin; aclarubicin; acodazole hydrochloride; apurinic acid; ara-CDP-DL-PTBA; arginine deaminase; asu- acronine; adozelesin; aldesleukin; altretamine; ambomycin; lacrine; atamestane; atrimustine; axinastatin I ; axinastatin 2; ametantrone acetate; aminoglutethimide; amsacrine; anastro- axinastatin 3; azasetron; azatoxin; aza osine; baccatin III zole; anthramycin; asparaginase; asperlin; azacitidine; 35 derivatives; balanol; batimastat; BCRMBL antagonists; ben- azetepa; azotomycin; batimastat; benzodepa; bicalutamide; zochlorins; benzoylstaurosporine; beta lactam derivatives; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; beta-alethine; betaclamycin B; betulinic acid; bFGF inhibi­ bleomycin sulfate; brequinar sodium; bropirimine; busulfan; tor; bicalutamide; bisantrene; bisaziridinylspermine; bisna­ cactinomycin; calusterone; caracemide; carbetimer; carbopl- fide; bistratene A; bizelesin; breflate; bropirimine; budoti- atin; carmustine; cambicin hydrochloride; carzelesin; cedef- 40 tane; buthionine sulfoximine; calcipotriol; calphostin C; ingol; chlorambucil; cirolemycin; cladribine; crisnatol mesy­ camptothecin derivatives; canarypox IL-2; capecitabine; car- late; cyclophosphamide; cytarabine; dacarbazine; boxamide-amino-triazole; carboxyamidotriazole; CaRest daunorubicin hydrochloride; decitabine; dexormaplatin; M3; CARN 700; cartilage derived inhibitor; carzelesin; dezaguanine; dezaguanine mesylate; diaziquone; doxorubi­ casein kinase inhibitors (ICOS); castanospermine; cecropin cin; doxorubicin hydrochloride; droloxifene; droloxifene cit­ 45 B; cetrorelix; chlorins; chloroquinoxaline sulfonamide; cica- rate; dromostanolone propionate; duazomycin; edatrexate; prost; cis-porphyrin; cladribine; clomifene analogues; clotri­ eflornithine hydrochloride; elsamitrucin; enloplatin; enpro- mazole; collismycin A; collismycin B; combretastatin A4; mate; epipropidine; epirubicin hydrochloride; erbulozole; combretastatin analogue; conagenin; crambescidin 816; cri­ esorubicin hydrochloride; estramustine; estramustine phos­ snatol; cryptophycin 8; cryptophycin A derivatives; curacin phate sodium; etanidazole; etoposide; etoposide phosphate; 50 A; cyclopentanthraquinones; cycloplatam; cypemycin; cyt­ etoprine; fadrozole hydrochloride; fazarabine; fenretinide; arabine ocfosfate; cytolytic factor; cytostatin; dacliximab; floxuridine; fludarabine phosphate; fluorouracil; fluorocitab- decitabine; dehydrodidemnin B; deslorelin; dexamethasone; ine; fosquidone; fostriecin sodium; gemcitabine; gemcitab- dexifosfamide; dexrazoxane; dexverapamil; diaziquone; ine hydrochloride; hydroxyurea; idarubicin hydrochloride; didemnin B; didox; diethylnorspermine; dihydro-5-azacyti- ifosfamide; iimofosine; interleukin Il (including recombinant 55 dine; 9-dioxamycin; diphenyl spiromustine; docosanol; dola- interleukin II, or rlL2), interferon alfa-2a; interferon alfa-2b; setron; doxifluridine; droloxifene; dronabinol; duocarmycin interferon alfa-nl; interferon alfa-n3; interferon beta-la; SA; ebselen; ecomustine; edelfosine; edrecolomab; eflorni­ interferon gamma-lb; iproplatin; irinotecan hydrochloride; thine; elemene; emitefur; epirubicin; epristeride; estramus­ lameotide acetate; letrozole; leuprolide acetate; liarozole tine analogue; estrogen agonists; estrogen antagonists; hydrochloride; lometrexol sodium; lomustine; losoxantrone 60 etanidazole; etoposide phosphate; exemestane; fadrozole; hydrochloride; masoprocol; maytansine; mechlorethamine fazarabine; fenretinide; filgrastim; finasteride; flavopiridol; hydrochloride; megestrol acetate; melengestrol acetate; mel- flezelastine; fluasterone; fludarabine; fluorodaunorunicin phalan; menogaril; mercaptopurine; methotrexate; methotr­ hydrochloride; forfenimex; formestane; fostriecin; fotemus- exate sodium; metoprine; meturedepa; mitindomide; mito- tine; gadolinium texaphyrin; gallium nitrate; galocitabine; carcin; mitocromin; mitogillin; mitomalcin; mitomycin; 65 ganirelix; gelatinase inhibitors; gemcitabine; glutathione mitosper; mitotane; mitoxantrone hydrochloride; mycophe- inhibitors; hepsulfam; heregulin; hexamethylene bisaceta- nolic acid; nocodazoie; nogalamycin; ormaplatin; oxisuran; mide; hypericin; ibandronic acid; idarubicin; idoxifene; idra- US 9,266,828 B2 55 56 mantone; ilmofosine; ilomastat; imidazoacridones; imiqui- lomide; teniposide; tetrachlorodecaoxide; tetrazomine; thali- mod; irmnunostimulant peptides; insulin-such as for example blastine; thiocoraline; thrombopoietin; thrombopoietin growth factor-1 receptor inhibitor; interferon agonists; inter­ mimetic; thymalfasin; thymopoietin receptor agonist; thy- ferons; interleukins; iobenguane; iododoxorubicin; motrinan; thyroid stimulating hormone; tin ethyl etiopurpu- ipomeanol, 4-; iroplact; irsogladine; isobengazole; isohomo- 5 rin; tirapazamine; titanocene bichloride; topsentin; halicondrin B; itasetron; jasplakinolide; kahalalide F; lamel- toremifene; totipotent ; translation inhibitors; Iarin-N triacetate; lameotide; leinamycin; lenograstim; Ien- tretinoin; triacetyluridine; triciribine; trimetrexate; triptore- tinan sulfate; leptolstatin; letrozole; leukemia inhibiting lin; tropisetron; turosteride; tyrosine kinase inhibitors; tyr- factor; leukocyte alpha interferon; leuprolide+estrogen+ phostins; UBC inhibitors; ubenimex; urogenital sinus-de­ progesterone; leuprorelin; levamisole; liarozole; linear 10 rived growth inhibitory factor; urokinase receptor polyamine analogue; lipophilic disaccharide peptide; lipo­ antagonists; vapreotide; variolin B; vector system, erythro­ philic platinum compounds; lissoclinamide 7; lobaplatin; cyte gene therapy; velaresol; veramine; verdins; verteporfin; lombricine; lometrexol; lonidamine; losoxantrone; lovasta- vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeni- tin; loxoribine; lurtotecan; lutetium texaphyrin; lysofylline; platin; zilascorb; and zinostatin stimalamer. lytic peptides; maitansine; mannostatin A; marimastat; maso- 15 Other therapeutic agents that maybe administered in con­ procol; maspin; matrilysin inhibitors; matrix metalloprotein­ junction a Furin/PC inhibitor disclosed herein include, but are ase inhibitors; menogaril; merbarone; meterelin; methioni- not limited to, alkylating agents, antimetabolites, natural nase; metoclopramide; MIF inhibitor; mifepristone; products, or hormones, e.g., nitrogen mustards (e.g., mechlo- miltefosine; mirimostim; mismatched double stranded RNA; roethamine, cyclophosphamide, chlorambucil, etc.), alkyl mitoguazone; mitolactol; mitomycin analogues; mitonafide; 20 sulfonates (e.g., busulfan), nitrosoureas (e.g., carmustine, mitotoxin fibroblast growth factor-saporin; mitoxantrone; lomusitne, ete.), ortriazenes (decarbazine, etc.). Examples of mofarotene; molgramostim; monoclonal antibody, human antimetabolites include but are not limited to folic acid analog chorionic gonadotrophin; monophosphoryl lipid A+myobac- (e.g., methotrexate), orpyrimidine analogs (e.g., Cytarabine), terium cell wall sk; mopidamol; multiple drug resistance gene purine analogs (e.g., mercaptopurine, thioguanine, pentosta­ inhibitor; multiple tumor suppressor I-based therapy; mus­ 25 tin). tard anticancer agent; mycaperoxide B; mycobacterial cell Examples of alkylating agents that include, but are not wall extract; myriaporone; N-acetyldinaline; N-substituted limited to, nitrogen mustards (e.g., mechloroethamine, cyclo­ benzamides; nafarelin; nagrestip; naloxone+pentazocine; phosphamide, chlorambucil, meiphalan, etc.), ethylenimine napavin; naphterpin; nartograstim; nedaplatin; nemorubicin; and methylmelamines (e.g., hexamethlymelamine, thiotepa), neridronic acid; neutral endopeptidase; nilutamide; nisamy- 30 alkyl sulfonates (e.g., busulfan), nitrosoureas (e.g., carmus­ cin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; tine, lomusitne, semustine, streptozocin, etc.), or triazenes 06-benzylguanine; octreotide; okicenone; oligonucleotides; (decarbazine, ete.). Examples of antimetabolites include, but onapristone; ondansetron; ondansetron; oracin; oral are not limited to folic acid analog (e.g., methotrexate), or inducer; ormaplatin; osaterone; oxaliplatin; oxaunomycin; pyrimidine analogs (e.g., fluorouracil, floxouridine, Cytara­ palauamine; palmitoylrhizoxin; pamidronic acid; panax- 35 bine), purine analogs (e.g., mercaptopurine, thioguanine, ytriol; panomifene; parabactin; pazelliptine; pegaspargase; pentostatin. peldesine; pentosan polysulfate sodium; pentostatin; pentro- Additional therapeutic agents that maybe administered in zole; perflubron; perfosfamide; perillyl alcohol; phenazino- conjunction with a Furin/PC inhibitor disclosed herein mycin; phenylacetate; phosphatase inhibitors; picibanil; pilo­ include, but are not limited to: Erbulozole (also known as carpine hydrochloride; pirarubicin; piritrexim; placetin A; 40 R-55104), Dolastatin 10 (also known as DLS-IO and NSC- placetin B; plasminogen activator inhibitor; platinum com­ 376128), Mivobulin isethionate (also known as CI-980), Vin­ plex; platinum compounds; platinum-triamine complex; por- cristine, NSC-639829, Discodermolide (also known as NVP- fimer sodium; porfiromycin; prednisone; propyl bis-acri- XX-A-296), ABT-751 (Abbott, also known as E-7010), done; prostaglandin J2; proteasome inhibitors; protein Altorhyrtins (such as Altorhyrtin A and Altorhyrtin C), A-based immune modulator; protein kinase C inhibitor; pro­ 45 Spongistatins (suchas Spongistatin I, Spongistatin2, Spong­ tein kinase C inhibitors, microalgal; protein tyrosine phos­ istatin 3, Spongistatin 4, Spongistatin 5, Spongistatin 6, phatase inhibitors; purine nucleoside phosphorylase inhibi­ Spongistatin 7, Spongistatin 8, and Spongistatin 9), Cema- tors; purpurins; pyrazoloacridine; pyridoxylated hemoglobin dotinhydrochloride (also known as LU-103793 andNSC-D- polyoxyethylene conjugate; raf antagonists; raltitrexed; 669356), Epothilones (such as Epothilone A, Epothilone B, ramosetron; ras farnesyl protein transferase inhibitors; ras 50 Epothilone C (also known as desoxyepothilone A or dEpoA), inhibitors; ras-GAP inhibitor; retelliptine demethylated; rhe­ Epothilone D (also referred to as KOS-862, dEpoB, and des­ nium Re 186 etidronate; rhizoxin; ribozymes; RH retinamide; oxyepothilone B), Epothilone E, Epothilone F, Epothilone B rogletimide; rohitukine; romurtide; roquinimex; rubiginone N-oxide, Epothilone A N-oxide, 16-aza-epothilone B, BI; ruboxyl; safingol; saintopin; SarCNU; sarcophytol A; 21-aminoepothilone B (also known as BMS-310705), 21-hy- sargramostim; Sdi I mimetics; semustine; senescence 55 droxyepothilone D (also known as Desoxyepothilone F and derived inhibitor I; sense oligonucleotides; signal transduc­ dEpoF), 26-fluoroepothilone), Auristatin PE (also known as tion inhibitors; signal transduction modulators; single chain NSC-654663), Soblidotin (also known as TZT-1027), antigen-binding protein; sizofiran; sobuzoxane; sodiumboro- LS-4559-P (Pharmacia, also known as LS-4577), LS-4578 captate; sodium phenylacetate; solverol; somatomedin bind­ (Pharmacia, also known as LS-477-P), LS-4477 (Pharmacia), ing protein; sonermin; sparfosic acid; spicamycin D; spiro- 60 LS-4559 (Pharmacia), RPR-112378 (Aventis), Vincristine mustine; splenopentin; spongistatin I; squalamine; stem cell sulfate, DZ-3358 (Daiichi), FR-182877 (Fujisawa, also inhibitor; stem-cell division inhibitors; stipiamide; stromel- known as WS-9885B), GS-164 (Takeda), GS-198 (Takeda), ysin inhibitors; sulfinosine; superactive vasoactive intestinal KAR-2 (Fhmgarian Academy of Sciences), BSF-223651 peptide antagonist; suradista; suramin; swainsonine; syn­ (BASF, also known as ILX-651 and LU-223651), thetic glycosaminoglycans; tallimustine; tamoxifen methio- 65 SAFI-49960 (Lilly/Novartis), SDZ-268970 (Lilly/Novartis), dide; tauromustine; tazarotene; tecogalan sodium; tegafur; AM-97 (Armad/Kyowa Hakko), AM-132 (Annad), AM-138 tellurapyrylium; telomerase inhibitors; temoporfin; temozo- (Armad/Kyowa Flakko), IDN-5005 (Indena), Cryptophycin US 9,266,828 B2 57 58 52 (also known as LY-355703), AC-7739 (Ajinomoto, also izes an exotoxin selected from the group consisting of anthrax known as AVE-8063A and CS-39.HC1), AC-7700 (Ajino­ toxin, pseudomonas exotoxin A, Shiga toxin, diphtheria moto, also known as AVE-8062, AVE-8062A, CS-39-L-Ser- toxin, tetanus and botulism neurotoxins, and combinations .ElCl, and RPR-258062A), Vitilevuamide, Tubulysin A, thereof. In some embodiments, a Furin/PC inhibitor disclosed Canadensol, Centaureidin (also known as NSC-106969), 5 herein neutralizes virulence of bacteria carrying the exo toxin. T-138067 (Tularik, also known as T-67, TL-138067 and Further disclosed herein, in certain embodiments, are TI-138067), COBRA-1 (Parker Elughes Institute, also known methods of treating a cancer in a mammal in need thereof. In as DDE-261 and WE1I-261), EllO (Kansas State University), some embodiments, the methods comprise administering a Ell 6 (Kansas State University), OncocidinAl (also known as therapeutically effective amount of a Furin/PC inhibitor dis­ BTO-956 and DIME), DDE-313 (Parker Elughes Institute), 10 closed herein. In some embodiments, the cancer is skin Fijianolide B, Laulimalide, SPA-2 (Parker Elughes Institute), tumors, head and neck squamous cell carcinomas, astrocy­ SPA-1 (Parker Elughes Institute, also known as SPIKET-P), toma, lung non-small cell carcinoma, or metastasis of col­ 3-IAABU (Cytoskeleton/Mt. Sinai School of Medicine, also orectal cancer. known as MF-569), Narcosine (also known as NSC-5366), Also disclosed herein, in certain embodiments, are meth­ Nascapine, D-24851 (Asta Medica), A-105972 (Abbott), 15 ods of treating an autoimmune or inflammatory disease, dis­ Elemiasterlin, 3-BAABU (Cytoskeleton/Mt. Sinai School of order or condition in a mammal in need thereof. In some Medicine, also known as MF-191), TMPN (Arizona State embodiments, the methods comprise administering to the University), Vanadocene acetylacetonate, T-138026 (Tu­ mammal a therapeutically effective amount of a Furin/PC larik), Monsatrol, Inanocine (also known as NSC-698666), inhibitor disclosed herein. In some embodiments, the neuro- 3-1AABE (Cytoskeleton/Mt. Sinai School of Medicine), 20 degenerative disease is arthritis, atherosclerosis, and Alzhe­ A-204197 (Abbott), T-607 (Tuiarik, also known as imer’s disease. T-900607), RPR-115781 (Aventis), Eleutherobins (such as The administration of a Furin/PC inhibitor disclosed herein Desmethyleleutherobin, Desaetyleleutherobin, lsoeleuther- is optionally administered chronically, that is, for an extended obin A, and Z-Eleutherobin), Caribaeoside, Caribaeolin, period of time, including throughout the duration of the Eialichondrin B, D-64131 (Asta Medica), D-68144 (Asta 25 patient’s life in order to ameliorate or otherwise control or Medica), Diazonamide A, A-293620 (Abbott), NPI-2350 limit the symptoms of the patient’s disease or condition. (Nereus), Taccalonolide A, TUB-245 (Aventis), A-259754 In some embodiments, a Furin/PC inhibitor disclosed (Abbott), Diozostatin, (-)-Phenylahistin (also known as herein is given continuously. In some embodiments, admin­ NSCL-96F037), D-68838 (Asta Medica), D-68836 (Asta istration of a Furin/PC inhibitor disclosed herein is tempo­ Medica), Myoseverin B, D-43411 (Zentaris, also known as 30 rarily reduced or temporarily suspended for a certain length D-81862), A-289099 (Abbott), A-318315 (Abbott), HTI-286 of time (Te., a “drug holiday”). The length of the drug holiday (also known as SPA-110, trifluoroacetate salt) (Wyeth), optionally varies between 2 days and I year, including by way D-82317 (Zentaris), D-82318 (Zentaris), SC-12983 (NCI), o f example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, Resverastatin phosphate sodium, BPR-OY-007 (National 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, Health Research Institutes), and SSR-250411 (Sanofi). 35 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, Where the individual is suffering from or at risk of suffer­ 250 days, 280 days, 300 days, 320 days, 350 days, or 365 ing from an autoimmune disease, or an inflammatory disease days. The dose reduction during a drug holiday includes from a Furin/PC inhibitor disclosed herein may be used in combi­ 10%-100%, including, by way of example only, 10%, 15%, nation with: immunosuppressants (e.g., tacrolimus, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, cyclosporin, rapamicin, methotrexate, cyclophosphamide, 40 70%, 75%, 80%, 85%, 90%, 95%, or 100%. azathioprine, mercaptopurine, mycophenolate, or FTY720), In some embodiments, once improvement of a patient’s glucocorticoids (e.g., prednisone, cortisone acetate, pred­ conditions has occurred, maintenance doses of a Furin/PC nisolone, methylprednisolone, dexamethasone, betametha­ inhibitor disclosedherein are administered. Subsequently, the sone, triamcinolone, beclometasone, fludrocortisone acetate, dosage or the frequency of administration, or both, is reduced, deoxycorticosterone acetate, aldosterone), non-steroidal 45 as a function of the symptoms, to a level at which the anti-inflammatory drugs (e.g., salicylates, arylalkanoic acids, improved disease, disorder or condition is retained. In some 2-arylpropionic acids, N-arylanthranilic acids, oxicams, cox- embodiments, patients require intermittent treatment on a ibs, or sulphonanilides), Cox-2-specific inhibitors (e.g., val- long-term basis upon any recurrence of symptoms. decoxib, celecoxib, or rofecoxib), leflunomide, gold thioglu- In some embodiments, a pharmaceutical composition cose, gold thiomalate, aurofin, sulfasalazine, 50 described herein is in unit dosage forms suitable for single hydroxychloroquinine, minocycline, TNF-a binding pro­ administration of precise dosages. In unit dosage form, the teins (e.g., infliximab, etanercept, or adalimumab), abatacept, formulation is divided into unit doses containing appropriate anakinra, interferon-|3, interferon-y, interleukin-2, allergy quantities of one or more Furin/PC inhibitors. In some , antihistamines, antileukotrienes, beta-agonists, embodiments, the unit dosage is in the form of a package theophylline, or anticholineigics. 55 containing discrete quantities of the formulation. Non-limit­ Dosing and Treatment Regimens ing examples are packaged tablets or capsules, and powders Disclosed herein, in certain embodiments, are methods of in vials or ampoules. In some embodiments, aqueous suspen­ treating an infectious disease in a mammal in need of such sion compositions are packaged in single-dose non-reclos- treatment. In some embodiments, the methods comprise able containers. Alternatively, multiple-dose reclosable con­ administering to the mammal a therapeutically effective 60 tainers are used, in which case it is typical to include a amount of a Furin/PC inhibitor disclosed herein. In some preservative in the composition. By way of example only, embodiments, the infection disease is associated with influ­ formulations for parenteral injection are presented in unit enza virus, human immunodeficiency virus I, Ebola, dosage form, which include, but are not limited to ampoules, measles, cytomegalovirus, and flaviviruses (Dengue, Yellow or in multi dose containers, with an added preservative. fever, West Nile, Japanese encephalitis and multiple addi­ 65 In prophylactic applications, a Furin/PC inhibitor dis­ tional related flaviviruses) and parasitic nematodes. In some closed herein or a pharmaceutical composition containing a embodiments, a Furin/PC inhibitor disclosed herein neutral­ Furin/PC inhibitor disclosed herein is administered to an US 9,266,828 B2 59 60 individual susceptible to or otherwise at risk of a particular circulating concentrations that include the ED50 with mini­ disease, disorder or condition. In certain embodiments of this mal toxicity. The dosage optionally varies within this range use, the precise amounts of a Furin/PC inhibitor disclosed depending upon the dosage form employed and the route of herein depend on an individual’s state of health, weight, and administration utilized. the like. Furthermore, in some instances, when a Furin/PC 5 While some embodiments of the present disclosure have inhibitor disclosed herein or a pharmaceutical composition been shown and described herein, such embodiments are comprising a Furin/PC inhibitor described herein is adminis­ provided by way of example only. It is intended that the tered to an individual, effective amounts for this use depend following claims define the scope of the present disclosure on the severity and course of the disease, disorder or condi­ and that methods and structures within the scope of these tion, previous therapy, an individuaPs health status and 10 claims and their equivalents be covered thereby. response to the drugs, and the judgment of the treating phy­ sician. What is claimed is: In certain instances, wherein following administration of a selected dose of a Furin/PC inhibitor disclosed herein or a I . A compound of formula II, or a pharmaceutically accept­ able salt, stereoisomer, tautomer, or prodrug thereof: pharmaceutical composition comprising a Furin/PC inhibitor 15 described herein, an individual’s condition does not improve, upon the doctor’s discretion the administration of the Furin/ PC inhibitor disclosed herein or pharmaceutical composition (H) is optionally administered chronically, that is, for an extended period of time, including throughout the duration of an indi­ 20 vidual’s life in order to ameliorate or otherwise control or limit the symptoms of an individual’s disorder, disease or condition. In certain embodiments, an effective amount of a given agent varies depending upon one or more of a number of 25 factors such as the particular Furin/PC inhibitor disclosed herein, disease or condition and its severity, the identity (e.g., weight) of an individual or host in need of treatment, and is determined according to the particular circumstances sur­ wherein: rounding the case, including, e.g., the specific agent being 30 R1 is alkyl, cycloalkyl, or heteroalicyclyl; administered, the route of administration, the condition being R2 is —U-guanidine, wherein U is alkyl, cycloalkyl, het­ treated, and an individual or host being treated. In some eroalicyclyl, aryl, or heteroaryl; embodiments, doses administered include those up to the Y is —CONH-, -S O 2N H -, —O—, -C H 2- , —s—, maximum tolerable dose. In certain embodiments, about 0.02 - S O 2- , or -C O S O 2N H - ; to about 5000 mg per day, from about I to about 1500 mg per 35 Z is —CONH-, -S O 2N H -, —o—, -C H 2- , —s—, day, about I to about lOOmg/day, about I to about 50mg/day, - S O 2- , or -C O S O 2N H - ; or about I to about 30 mg/day, or about 5 to about 25 mg/day of a Furin/PC inhibitor disclosed herein is administered. In R3 and R4 are each independently —F, —CF3, —OCF3, various embodiments, the desired dose is conveniently be —OCH3, or alkyl; presented in a single dose or in divided doses administered 40 a and b are each independently 0, I, or 2; and simultaneously (or over a short period of time) or at appro­ m and n are each independently 0, I, 2, or 3. priate intervals, for example as two, three, four or more sub­ 2. The compound of claim I, or a pharmaceutically accept­ doses per day. able salt, stereoisomer, tautomer, or prodrug thereof, wherein In certain instances, there are a large number of variables in R1 is C1-C6 alkyl. regard to an individual treatment regime, and considerable 45 3. The compound of claim I , or a pharmaceutically accept­ excursions from these recommended values are considered able salt, stereoisomer, tautomer, or prodrug thereof, wherein within the scope described herein. Dosages described herein R 1 is isopropyl. are optionally altered depending on a number of variables such as, by way of non-limiting example, the activity of a 4. The compound of claim I, or a pharmaceutically accept­ Furin/PC inhibitor disclosed herein, the disease or condition 50 able salt, stereoisomer, tautomer, or prodrug thereof, wherein to be treated, the mode of administration, the requirements of U isC 1-C6 alkyl. an individual, the severity of the disease or condition being 5. The compound of claim 4, or a pharmaceutically accept­ treated, and the judgment of the practitioner. able salt, stereoisomer, tautomer, or prodrug thereof, wherein Toxicity and therapeutic efficacy of such therapeutic regi­ U is-(CH 2)3- . mens are optionally determined by pharmaceutical proce­ 55 6. The compound of claim I, or a pharmaceutically accept­ dures in cell cultures or experimental animals, including, but able salt, stereoisomer, tautomer, or prodrug thereof, wherein not limited to, the determination of the LD50 (the dose lethal Y is —CONH-. to 50% of the population) and the ED50 (the dose therapeuti­ 7. The compound of claim I, or a pharmaceutically accept­ cally effective in 50% of the population). The dose ratio able salt, stereoisomer, tautomer, or prodrug thereof, wherein between the toxic and therapeutic effects is the therapeutic 60 Z is -S O 2N H -. index and it can be expressed as the ratio between LD50 and ED50. In some embodiments, a Furin/PC inhibitor disclosed 8. The compound of claim I, or a pharmaceutically accept­ herein exhibiting high therapeutic indices is preferred. In able salt, stereoisomer, tautomer, or prodrug thereof, wherein certain embodiments, data obtained from cell culture assays m is I an d n is I. and animal studies are used in formulating a range of dosage 65 9. The compound of claim I, or a pharmaceutically accept­ for use in human. In some embodiments, the dosage of a able salt, stereoisomer, tautomer, or prodrug thereof, wherein Furin/PC inhibitor disclosed herein lies within a range of a and b are 0. US 9,266,828 B2 61 62 10. The compound of claim I, or a pharmaceutically acceptable salt, stereoisomer, tautomer, or prodrug thereof, wherein the compound is

11. A pharmaceutical composition comprising a com­ pound of claim I, or a pharmaceutically acceptable salt, ste- 20 reoisomer, tautomer, or prodrug thereof, and a pharmaceuti­ cally acceptable carrier thereof.