WO2005021558A2 - Proteasome inhibitors and methods of using the same - Google Patents

Proteasome inhibitors and methods of using the same Download PDF

Info

Publication number
WO2005021558A2
WO2005021558A2 PCT/US2004/026407 US2004026407W WO2005021558A2 WO 2005021558 A2 WO2005021558 A2 WO 2005021558A2 US 2004026407 W US2004026407 W US 2004026407W WO 2005021558 A2 WO2005021558 A2 WO 2005021558A2
Authority
WO
WIPO (PCT)
Prior art keywords
amino
alkyl
methylbutyl
methyl
nitroamino
Prior art date
Application number
PCT/US2004/026407
Other languages
French (fr)
Other versions
WO2005021558A3 (en
Inventor
Germano D'arasmo
Raffaella Bernardini
Sergio De Munari
Alberto Bernareggi
Paolo G. Cassara
Sankar Chatterjee
Edmondo Ferretti
Mohamed Iqbal
Ernesto Menta
Patricia A. Messina Mclaughlin
Ambrogio Oliva
Original Assignee
Cephalon, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=34278521&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2005021558(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to AU2004268946A priority Critical patent/AU2004268946B2/en
Priority to CA2535686A priority patent/CA2535686C/en
Priority to DK04786507.6T priority patent/DK1660507T4/en
Priority to KR1020067003068A priority patent/KR101093880B1/en
Priority to DE602004022424T priority patent/DE602004022424D1/en
Priority to PL04786507T priority patent/PL1660507T5/en
Priority to AT04786507T priority patent/ATE438650T2/en
Priority to ES04786507.6T priority patent/ES2330008T5/en
Priority to JP2006523419A priority patent/JP4917431B2/en
Priority to EA200600414A priority patent/EA010804B1/en
Priority to BRPI0413582A priority patent/BRPI0413582B8/en
Application filed by Cephalon, Inc. filed Critical Cephalon, Inc.
Priority to MXPA06001687A priority patent/MXPA06001687A/en
Priority to EP04786507.6A priority patent/EP1660507B2/en
Priority to CN2004800301520A priority patent/CN1867572B/en
Priority to SI200431273T priority patent/SI1660507T1/en
Priority to NZ545775A priority patent/NZ545775A/en
Publication of WO2005021558A2 publication Critical patent/WO2005021558A2/en
Publication of WO2005021558A3 publication Critical patent/WO2005021558A3/en
Priority to IL173722A priority patent/IL173722A/en
Priority to IS8343A priority patent/IS3013B/en
Priority to NO20061197A priority patent/NO20061197L/en
Priority to HK06112369.2A priority patent/HK1091839A1/en
Priority to HRP20090590TT priority patent/HRP20090590T4/en
Priority to IL217623A priority patent/IL217623A/en
Priority to NO20150303A priority patent/NO20150303L/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic System
    • C07F5/02Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic System
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic System
    • C07F5/02Boron compounds
    • C07F5/025Boronic and borinic acid compounds

Definitions

  • the present invention relates to boronic acid and boronic ester compounds useful as proteasome inhibitors and modulation of apoptosis.
  • the proteasome (also refered to as multicatalytic protease (MCP), multicatalytic proteinase, multicatalytic proteinase complex, multicatalytic endopeptidase complex, 20S, 26S, or ingensin) is a large, multiprotein complex present in both the cytoplasm and the nucleus of all eukaryotic cells. It is a highly conserved cellular structure that is responsible for the ATP-dependent proteolysis of most cellular proteins (Tanaka, Biochem Biophy. Res. Comrnun., 1998, 247, 537).
  • the 26S proteasome consists of a 20S core catalytic complex that is capped at each end by a 19S regulatory subunit.
  • the archaebacterial 20S proteasome contains fourteen copies of two distinct types of subunits, ⁇ and ⁇ , which form a cylindrical structure consisting of four stacked rings.
  • the top and bottom rings contain seven ⁇ -subunits each, while the inner rings contain seven ⁇ -subunits.
  • the more complex eukaryotic 20S proteasome is composed of about 15 distinct 20-30 kDa subunits and is characterized by three major activities with respect to peptide substrates.
  • the proteasome displays tryptic-, chymotryptic-, and peptidylglutamyl peptide-hydrolytic activities ( ivett, Biochem. J., 1993, 291, 1 and Orlowski, Biochemistry, 1990, 29, 10289).
  • the proteasome has a unique active site mechanism which is believed to utilize a threonine residue as the catalytic nucleophile (Seemuller, et al., Science, 1995, 268, 579).
  • the 26S proteasome is able to degrade proteins that have been marked by the addition of ubiquitin molecules.
  • ubiquitin is attached to the ⁇ -amino groups of Iysines in a multistep process utilizing ATP and El (ubiquitin activating) and E2 (ubiquitin-conjugating) enzymes. Multi-ubiquitinated substrate proteins are recognized by the 26S proteasome and are degraded.
  • the multi-ubiquitin chains are generally released from the complex and ubiquitin is recycled (Goldberg, et al., Nature, 1992, 357, 375).
  • Numerous regulatory proteins are substrates for ubiquitin dependent proteolysis. Many of these proteins function as regulators of physiological as well as pathophysiological cellular processes. Alterations in proteasome activity have been implicated in a number of pathologies including neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, as well as occlusion/ischaemia reperfusion injuries, and aging of the central nervous system.
  • the ubiquitin-proteasome pathway also plays a role in neoplastic growth.
  • Tumor suppressor p53 A known substrate of the proteasome is the tumor suppressor p53 which is involved in several cellular processes (see, e.g., Ko, L. J. Genes Dev., 1996, 10, 1054). Tumor suppressor p53 has been shown to induce apoptosis in several haematopoietic cell lines (Oren, M., Semin. Cancer Biol., 1994, 5, 221). Induction of p53 leads to cell growth arrest in the Gl phase of the cell cycle as well as cell death by apoptosis. Tumor suppressor p53 degradation is known to be carried out via the ubiquitin-proteasome pathway, and disrupting p53 degradation by inhibition of the proteasome is a possible mode of inducing apoptosis.
  • proteasome is also required for activation of the transcription factor
  • NF- ⁇ B by degradation of its inhibitory protein, I ⁇ B (Palombella, et al., Cell, 1994, 78, 773).
  • I ⁇ B inhibitory protein
  • NF- ⁇ B has a role in maintaining cell viability through the transcription of inhibitors of apoptosis. Blockade of NF- ⁇ B activity has been demonstrated to make cells more susceptible to apoptosis.
  • Lactacystin is a Streptomyces metabolite that specifically inhibits the proteolytic activity of the proteasome complex (Fenteany, et al., Science, 1995, 268, 726). This molecule is capable of inhibiting the proliferation of several cell types (Fenteany, et al., Proc. Natl. Acad. Sci. USA, 1994, 91, 3358). It has been shown that lactacystin binds irreversibly, through its ⁇ -lactone moiety, to a threonine residue located at the amino terminus of the ⁇ - subunit of the proteasome.
  • Peptide aldehydes have been reported to inhibit the chymotrypsin-like activity associated with the proteasome (Vinitsky, et al., Biochemistry, 1992, 31, 9421; Tsubuki, et al., Biochem. Biophys. Res. Commun., 1993, 196, 1195; and Rock, et al., Cell, 1994, 78, 761).
  • Dipeptidyl aldehyde inhibitors that have IC 50 values in the 10-100 nM range in vitro (Iqbal, M., et al., J. MedChem., 1995, 38, 2276) have also been reported.
  • N-terminal tri-peptide boronic ester and acid compounds have been shown to inhibit the growth of cancer cells (U.S. Pat. No. 5,106,948).
  • a broad class of N-terminal tri-peptide boronic ester and acid compounds and analogs thereof has been shown to inhibit renin (U.S. Pat. No. 5,169,841).
  • Various inhibitors of the peptidase activities of the proteasome have also been reported.
  • 5,693,617 report peptidyl aldehyde compounds as proteasome inhibitors useful for reducing the rate of degradation of protein in an animal. Inhibition of the 26S and 20S proteasome by indanone derivatives and a method for inhibiting cell proliferation using indanone derivatives are reported by Lum et al., U.S. Pat. No. 5,834,487. Alpha-ketoamide compounds useful for treating disorders mediated by 20S proteasome in mammals are reported in Wang et al., U.S. Pat. No. 6,075,150. France, et al., WO 00/64863, report the use of 2,4-diamino-3-hydroxycarboxylic acid derivatives as proteasome inhibitors.
  • Carboxylic acid derivatives as proteasome inhibitors are reported by Yamaguchi et al., EP 1166781. Ditzel, et al., EP 0 995 757 report bivalent inhibitors of the proteasome.
  • 2-Aminobenzylstatine derivatives that inhibit noncovalently the chymotrypsin-like activity of the 20S proteasome have been reported by Garcia-Echeverria, et al., Bioorg. Med. Chem. Lett., 2001, 11, 1317.
  • Some further proteasome inhibitors can contain boron moieties.
  • Drexler et al., WO 00/64467 report a method of selectively inducing apoptosis in activated endothelial cells or leukemic cells having a high expression level of c-myc by using tetrapeptidic boronate containing proteasome inhibitors.
  • Furet et al., WO 02/096933 report 2-[[N-(2-amino-3-(heteroaryl or aryl)propionyl)aminoacyl]amino]alkylboronic acids and esters for the therapeutic treatment of proliferative diseases in warm-blooded animals.
  • the present invention is directed to novel boronic acid and boronic ester compounds useful as proteasome inhibitors and modulation of apoptosis.
  • the subject invention also comprises methods for inhibition of multicatalytic protease ("MCP") associated with certain disorders, including the treatment of muscle wasting disorders.
  • MCP multicatalytic protease
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula (I) and a pharmaceutically acceptable carrier.
  • the present invention provides a method of inhibiting activity of proteasome comprising contacting a compound of Formula (I) with said proteasome.
  • the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to said cancer a therapeutically effective amount of a compound of Formula (I).
  • the present invention provides a method of treating cancer treating cancer comprising administering to a mammal having or predisposed to said cancer a therapeutically effective amount of a compound of Formula (I), and wherein said cancer is selected from skin, prostate, colorectal, pancreas, kidney, ovary, mammary, liver, tongue, lung, and smooth muscle tissue.
  • the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to said cancer a therapeutically effective amount of a compound of Formula (I), and wherein said cancer is selected from leukemia, lymphoma, non-Hodgkin lymphoma, myeloma, and multiple myeloma.
  • the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to said cancer a therapeutically effective amount of a compound of Formula (I) in combination with one or more antitumor or anticancer agent and/or radiotherapy.
  • the present invention provides a method of inhibiting activity of transcription factor NF- ⁇ B comprising contacting I ⁇ B, the inhibitor of transcription factor NF- ⁇ B, with a compound of Formula (I).
  • the present invention provides a compound of
  • the present invention provides use of a compound of Formula (I) for the manufacture of a medicament for the treatment of cancer.
  • the present invention provides processes for preparing a compound of Formula (II):
  • the present invention provides, inter alia, compounds that can inhibit proteasome activity and be used for the treatment of diseases or disorders related to proteasome activity.
  • Compounds of the invention include compounds of Formula (I)
  • R 1 is Ci-Cg alkyl, C 2 -C 8 alkenyl, C -C 8 alkynyl, or C 3 -C 7 cycloalkyl
  • a, b, and c are each, independently, 0, 1, 2, 3, 4, 5, or 6
  • d and e are each, independently, 0, 1, 2, 3, or 4;
  • R s and R 6 are each, independently, H, Ci-Cio alkyl, carbocyclyl, heterocarbocyclyl, or an amino protecting group; alternatively, R 5 and R 6 together with the N atom to which they are attached form a heterocarbocyclyl group;
  • R 7 is H or Ci-Cio alkyl
  • R 9 is H, - o alkyl, carbocyclyl, or heterocarbocyclyl
  • R 11 is Ci-C ⁇ alkyl, aryl, or NR 12 R 13 ;
  • R 21a is H, C ⁇ -C 20 alkyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, carbocyclyl or heterocarbocyclyl;
  • R ⁇ is Hor -Ce alkyl; alternatively, two R 23a may be combined, together with the N atom to which they are attached, to form a 5 to 7 membered heterocyclic group; and
  • R 1 can be C ⁇ -C 4 alkyl, and in further embodiments, R 1 can be propyl, such as 2-propyl.
  • R 2 s -(CH 2 ) d CH(R 7 )NR 9 R 10 and d is 0, 1, or 2.
  • R 2 s -(CH 2 ) d CH(R 7 )NR 9 R 10 and d is 0.
  • R 2 s -(CH 2 ) d CH(R 7 )NR 9 R 10 and R 9 is H.
  • R 2 s -(CH 2 ) d CH(R 7 )NR 9 R 10 are examples of R 2 s -(CH 2 ) d CH(R 7 )NR 9 R 10 .
  • R 2 s -CH(R 7 )NR 9 R 10 are examples of R 2 s -CH(R 7 )NR 9 R 10 .
  • R 2 s -(CH 2 ) e CH(R 7 )ZR 8 and e is 0, 1, or 2.
  • R 2 s -(CH 2 ) e CH(R 7 )ZR 8 and e is 0.
  • R 2 s -(CH 2 ) e CH(R 7 )ZR 8 are examples of R 2 s -(CH 2 ) e CH(R 7 )ZR 8 .
  • R 2 s -CH(R 7 )ZR 8 .
  • Q has Formula (II-a): (II-a) wherein D, R 15a , R 15b , R 15c , R 15d , p and q are defined herein below.
  • Q is B(OH) 2 or a cyclic boronic ester wherein said cyclic boronic ester contains from 6 to 10 carbon atoms and contains at least one cycloalkyl moiety.
  • Q is B(OH) 2 .
  • Q is pinanediol boronic ester.
  • Q is bicyclohexyl-l,l'-diol boronic ester.
  • Q is l,2-dicyclohexyl-ethane-l,2-diol boronic ester.
  • Q is -B(OH) 2 , -B(OR 14 ) 2 ,
  • R 14 , R 15a , R 15b , R 15c , R lsd , W, W 1 , p and q are as defined hereinbelow.
  • Q is: W is a substituted or unsubstituted C 4 -C ⁇ o cycloalkyl ring.
  • X is R A S(O) 2 -.
  • R A is C ⁇ -C 14 alkyl substituted by -(0-alkyl) r -OH or
  • R A is -C ⁇ alkyl substituted by -(O-alkyl) r -OH or
  • R A comprises at least one -CH 2 CH 2 O- group.
  • R A is -CH 2 (OCH 2 CH 2 ) r OCH 3 .
  • R A is -CH 2 OCH 2 CH OCH 2 CH 2 OCH 3 or
  • R A is aryl or heteroaryl each optionally substituted with 1-5 R 21 .
  • R A is cycloalkyl or heterocycloalkyl each optionally substituted with 1-5 R 21 .
  • R A is C ⁇ -C 2 o alkyl; C 2 -C 20 alkenyl; or C 2 -C 20 alkynyl, each optionally substituted with R 20 .
  • R A is C ⁇ -C 2 o alkyl; C 2 -C 20 alkenyl; or C -C 0 alkynyl, each substituted with a carbocyclyl group or a heterocarbocyclyl group wherein said carbocyclyl group or heterocarbocyclyl group is optionally substituted with 1, 2 or 3 R 21 .
  • R A is C1-C 2 0 alkyl; C 2 -C 20 alkenyl; or C 2 -C 20 alkynyl, each substituted with an aryl group wherein said aryl group is optionally substituted with 1, 2 or 3 R 21 .
  • R A is C ⁇ -C 2 o alkyl; C 2 -C 2 o alkenyl; or C 2 -C 20 alkynyl, each substituted with an heteroaryl group wherein said heteroaryl group is optionally substituted with 1, 2 or 3 R 21 .
  • R A is C ⁇ -C 2 o alkyl; C 2 -C 20 alkenyl; or C 2 -C 20 alkynyl, each substituted with an cycloalkyl group wherein said cycloalkyl group is optionally substituted with 1, 2 or 3 R 21 .
  • R A is C C 2 o alkyl; C -C 20 alkenyl; or C 2 -C 20 alkynyl, each substituted with an heterocycloalkyl group wherein said heterocycloalkyl group is optionally substituted with 1, 2 or 3 R 21 .
  • R 21 is phenoxy.
  • R 20 , and R 20 is aryl optionally substituted with 1-3 R 21 ; and in yet further embodiments aryl is substituted by at least one halo.
  • R 20 is -OR 20a or-OR 20c .
  • R 20 and R 20 is heterocarbocyclyl optionally substituted with 1-3 R 21 .
  • X is R A S(O) 2 - and R A is C 3 -C ⁇ 6 alkyl.
  • the present invention provides compounds of Formula (I) wherein the stereochemistry is of Formula (I-s):
  • the present invention provides compounds of Formula (I)
  • R 1 is d-Cg alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, or C 3 -C 7 cycloalkyl
  • a, b, and c are each, independently, 0, 1, 2, 3, 4, 5, or 6
  • d and e are each, independently, 0, 1, 2, 3, or 4
  • R 4 is H or Ci-do alkyl
  • R 5 and R 6 are each, independently, H, Ci-Qo alkyl
  • R 11 is C ⁇ -C 6 alkyl, aryl, or NR 12 R 13 ;
  • R 12 and R 13 are, independently, H, Ci-Cio alkyl, carbocyclyl, heterocarbocyclyl, or an amino protecting group; alternatively, R 12 and R 13 together with the N atom to which they are attached form a heterocarbocyclyl group;
  • Z is O, S, Se, or Te
  • Q is -B(OH) 2 , -B(OR 14 ) 2 , or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R 14 is H, d-C 4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • R A is C ⁇ -C 20 alkyl optionally substituted with R 20 ; C 2 -C 20 alkenyl optionally substituted with R 20 ; C 2 -C 20 alkynyl optionally substituted with R 20 ; carbocyclyl optionally substituted with 1-5 R 21 ; or heterocarbocyclyl optionally substituted with 1-5 R 21 ;
  • R 20a is C ⁇ -C o alkyl, C -C 20 alkenyl, or C 2 -C 20 alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, d-C 4 alkyl, aryl, heteroaryl or -NHR 0b ;
  • R 20b is an amino protecting group
  • R 20c is carbocyclyl optionally substituted with 1-5 R 22 ; or heterocarbocyclyl optionally substituted with 1-5 R 22 ;
  • R 1 is d-Cg alkyl
  • a is 1, 2, 3, 4, or 5;
  • c is 1, 2, 3, 4, or 5;
  • d is O, 1, or 2;
  • e is O, l, or 2;
  • R 7 is H or methyl
  • R 9 is H, d-do alkyl, carbocyclyl, or heterocarbocyclyl;
  • R A is d-C 2 o alkyl optionally substituted with R 20 ; C 2 -C 2 o alkenyl optionally substituted with R 20 ; C -C 0 alkynyl optionally substituted with R 20 ; carbocyclyl optionally substituted with 1-5 R 21 ; or heterocarbocyclyl optionally substituted with 1-5 R 21 ;
  • R 0a is d-C 2 o alkyl, C 2 -C 2 o alkenyl, or C 2 -C 2 o alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, C)-C 4 alkyl, aryl, heteroaryl or -NHR 20b ;
  • R 20 is an amino protecting group
  • R 20c is carbocyclyl optionally substituted with 1-5 R 22 ; or heterocarbocyclyl optionally substituted with 1-5 R 22 ;
  • the present invention provides compounds of
  • R 1 is C 1 -C 4 alkyl
  • R 7 is H or methyl
  • R 9 is H or Ci-do alkyl
  • R 10 is H, Ci-do alkyl, or an amino protecting group
  • Y is H, CN, orNO 2 ;
  • Q is -B(OH) 2 , pinanediol boronic ester, bicyclohexyl-l,l'-diol boronic ester, or 1,2- dicyclohexyl-ethane-l,2-diol boronic ester;
  • R A is CpC 2 o alkyl optionally substituted with R 20 ; C 2 -C o alkenyl optionally substituted with R 20 ; C 2 -C 20 alkynyl optionally substituted with R 20 ; carbocyclyl optionally substituted with 1-5 R 21 ; or heterocarbocyclyl optionally substituted with 1-5 R 21 ;
  • R 20 is selected from the
  • the present invention provides compound of
  • R 1 is C ⁇ -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, or C 3 -C 7 cycloalkyl;
  • R 2 is -CH 2 NH or -CH 2 NR 9 R 10 ;
  • R 9 is H or Cj-Cio alkyl
  • R 23a is H or C ⁇ -C 6 alkyl; , alternatively, two R 23a may be combined, together with the N atom to which they are attached, to form a 5 to 7 membered heterocyclic group; and r is 2, 3, 4, or 5.
  • the present invention provides compounds of
  • R 1 is Ci-Cg alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, or C 3 -C 7 cycloalkyl;
  • R 2 is H
  • Q is -B(OH) 2 , -B(OR 14 ) 2 , or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R 14 is H, C ⁇ -C 4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • R A is C ⁇ -C 2 o alkyl optionally substituted with R 20 ; C 2 -C 2 o alkenyl optionally substituted with R 20 ; C 2 -C 2 o alkynyl optionally substituted with R 20 ; carbocyclyl optionally substituted with 1
  • R A is d-C ⁇ alkyl optionally substituted with R 20 ;
  • R 20 is -(O-alkyl) r -OH or -(O-alkyl) r -(O-alkyl); and r is 1, 2, 3, 4, or 1 5.
  • the O-alkyl is methoxy, ethoxy, or propoxy.
  • the present invention provides compounds of
  • R 1 is 2-propyl
  • R 9 is H;
  • Q is -B(OH) 2 , pinanediol boronic ester, bicyclohexyl-l,l'-diol boronic ester, or 1,2- dicyclohexyl-ethane-l,2-diol boronic ester;
  • R is CH 3 -, C 2 H 5 -, C 3 H 7 -, C H -, C5H11-, C 6 H ⁇ 3 -, C 7 H ⁇ 5 -, C 8 H ⁇ 7 -, C 9 H ⁇ 9 -, C ⁇ 0 H 2 r, C ⁇ H 23 -, C ⁇ 2 H 2 5-, C I3 H 27 -, adamantyl-, bicycloheptanyl-, C ⁇ -3 alkyl substituted with R 20 ; C 2-10 alkenyl substituted with R 20 ; cyclopropyl substituted with 0-3 R 21 ; cyclopentyl substituted with 0-2 R 21 ; cyclohexyl substituted with 0-2 R 21 ; phenyl substituted with 0-3 R 21 ; naphthyl- substituted with 0-2 R 21 ; pyrazinyl substituted with 0-1 R 21 ; quinolinyl substituted with 0-1 R 21 ; imidazoly
  • boronic acid refers to a compound containing a B(OH) 2 moiety.
  • boronic acid compounds can form oligomeric anhydrides by dehydration of the boronic moiety.
  • Snyder, et al., J. Am. Chem. Soc, 1958, SO, 3611 report oligomeric arylboronic acids.
  • “boronic acid”, or a chemical formula containing a -B(OH) 2 moiety is intended to encompass free boronic acids, oligomeric anhydrides, including but not limited to, dimers, trimers, tetramers, and mixtures thereof.
  • boronic acid anhydride or “boronic anhydride” refers to a compound formed by the combination of two or more molecules of a boronic acid compound of Formula (I), with loss of one or more water molecules from the boronic acid moieties. When contacted with water, the boronic acid anhydride compound can be hydrated to release free boronic acid compound.
  • the boronic acid anhydride structure can contain two, three, four, or more boronic acid units and can have a cyclic or linear configuration.
  • the boronic acid anhydride compound exists substantially in a single oligomeric form; however, boronic acid anhydrides also encompass mixtures of different oligomeric boronic acid anhydride as well as free boronic acids.
  • Non-limiting examples of boronic acid anhydrides of the invention include compounds of Formula (II) and (III) where G is a moiety of Formula (IV) and t is O to 10 or 1, 2, 3, or 4.
  • At least about 80% of boronic acid present in a boronic acid anhydride compound exists in a single oligomeric anhydride form. In further embodiments, at least about 85, about 90, about 95, or about 99 % of the boronic acid present in the boronic acid anhydride exists in a single oligomeric anhydride form.
  • the boronic acid anhydride compound consists essentially of a single oligomeric boronic acid anhydride.
  • the boronic acid anhydride compound consists of a single oligomeric boronic acid anhydride.
  • the boronic acid anhydride compound contains a boroxine of Formula (III), wherein t is 1.
  • Boronic acid anhydride compounds can be prepared from the corresponding boronic acid compound by exposure to dehydrating conditions, including, for example, crystallization, lyophilization, exposure to heat, and/or exposure to a drying agent.
  • Some suitable crystallization solvents include ethyl acetate, dichloromethane, hexanes, ether, benzene, acetonitrile, ethanol, and mixtures thereof.
  • the phrase "boronic ester” or “boronic acid ester” refers to an ester derivative of a boronic acid compound.
  • cyclic boronic ester is intended to mean a stable cyclic boronic moiety of general formula -B(OR)(OR) wherein the two R substituents are linked together forming a cyclic moiety (e.g., 3- to 10-membered cycloalkyl group) optionally further substituted with one or more substituents or fused with (sharing at least one bond) one or more further carbocyclyl or heterocarbocyclyl groups.
  • the cyclic boronic ester can contain from 2 to 20 carbon atoms, and optionally, a heteroatom which can be N, S, or O. Cyclic boronic esters are well known in the art.
  • cyclic boronic esters include, but are not limited to, pinanediol boronic ester, pinacol boronic ester, 1,2-ethanediol boronic ester, 1,3- propanediol boronic ester, 1,2- ⁇ ropanediol boronic ester, 2,3-butanediol boronic ester, 1,1,2,2-tetramethylethanediol boronic ester, 1,2-diisopropylethanediol boronic ester, 5,6-decanediol boronic ester, 1,2-dicyclohexylethanediol boronic ester, bicyclohexyl- l,l'-diol, diethanolamine boronic ester, and 1,2-diphenyl- 1,2-ethanediol boronic ester.
  • the "cyclic boronic ester" has Formula (II
  • R 15a , R 15b , R 15c , R 15d , R 15e , R 15f are each, independently, H, d-C l0 alkyl, C 3 -C 7 cycloalkyl, aryl or heteroaryl, wherein said d-Cio alkyl, d-do cycloalkyl, aryl or heteroaryl are each optionally substituted by 1, 2, 3 or 4 halo, Cj-C 4 alkyl, C ⁇ -C 4 alkoxy, C ⁇ -C 4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; or R 15a and R 15b together with the C atoms to which they are attached form C 3 - Cio cycloalkyl or a 3- to 10-membered heterocycloalkyl group,
  • D is NR 16 .
  • D is NH
  • D is CH 2 .
  • R 15a and R I5b together with the C atoms to which they are attached form C 3 -do cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, d-C 4 alkyl, d-C 4 alkoxy, C ⁇ -C haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; and R 15c and R 15d together with the C atoms to which they are attached form C3-C1 0 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C ⁇ -C alkyl, d-C 4 alkoxy, C ⁇ -C haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl.
  • R I5a and R I5b together with the C atoms to which they are attached form cyclopropyl, cyclobutyl, cyclopenytyl, cyclohexyl or cycloheptyl; and R 15c and R 15d together with the C atoms to which they are attached form cyclopropyl, cyclobutyl, cyclopenytyl, cyclohexyl or cycloheptyl.
  • D is absent and R ,5b and R 15c together with the C atoms to which they are attached form aryl, heteroaryl, C 3 -C ⁇ o cycloalkyl or a 3- to 10- membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, Ci- C 4 alkyl, C ⁇ -C alkoxy, C ⁇ -C 4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl.
  • D is absent and R 15b and R 15c together with the C atoms to which they are attached form C3-C10 cycloalkyl optionally substituted by 1, 2, 3 or 4 halo, C ⁇ -C alkyl, C ⁇ -C 4 alkoxy, C ⁇ -C 4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl.
  • D is absent and R 15b and R 15c together with the C atoms to which they are attached form C 3 -C10 cycloalkyl optionally substituted by 1, 2,
  • D is absent and R 15b and R 15c together with the C atoms to which they are attached form a C 7 -C ⁇ o bicyclic cycloalkyl group optionally substituted by 1, 2, 3 or 4 halo or d-C 4 alkyl.
  • p and q are each 1.
  • At least one of R 15 , R 15b , R 150 , R 15d is other than
  • cyclic boronic esters include, boronic esters with the following structures:
  • W is a substituted or unsubstituted C 4 -C 1 o cycloalkyl ring or a substituted or unsubstituted phenyl ring; W 1 is, independently at each occurrence, a substituted or unsubstituted C 3 -C 6 cycloalkyl ring.
  • Groups R 15a , R 15b , R 15c , R 15d , R 15e , R 15f , p and q are, defined as provided above.
  • alkyl or “alkylene” is meant to refer to a saturated hydrocarbon group which is straight-chained or branched.
  • Example alkyl groups include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, s-butyl, t-butyl), pentyl (e.g., n-pentyl, isopentyl, neopentyl) and the like.
  • alkyl group can contain from 1 to about 20, from 2 to about 20, from 1 to about 10, from 1 to about 8, from 1 to about 6, from 1 to about 4, or from 1 to about 3 carbon atoms.
  • alkenyl refers to an alkyl group having one or more double carbon-carbon bonds.
  • Example alkenyl groups include ethenyl, propenyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl, and the like.
  • alkynyl refers to an alkyl group having one or more triple carbon-carbon bonds.
  • Example alkynyl groups include ethynyl, propynyl, butynyl, pentynyl, and the like.
  • haloalkyl refers to an alkyl group having one or more halogen substituents.
  • Example haloalkyl groups include CF 3 , C 2 F 5 , CHF 2 , CC1 3 , CHC1 2 , C 2 Cl5, and the like.
  • An alkyl group in which all of the hydrogen atoms are replaced with halogen atoms can be referred to as "perhaloalkyl.”
  • perhaloalkyl groups include CF 3 and C 2 F 5 .
  • Carbocyclyl groups are saturated (i.e., containing no double or triple bonds) or unsaturated (i.e., containing one or more double or triple bonds) cyclic hydrocarbon moieties. Carbocyclyl groups can be mono- or polycyclic.
  • Example carbocyclyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, 1, 3-cyclopentadienyl, cyclohexenyl, norbornyl, norpinyl, norcarnyl, adamantyl, phenyl, and the like.
  • Carbocyclyl groups can be aromatic (e.g., "aryl") or non-aromatic (e.g., "cycloalkyl”). In some embodiments, carbocyclyl groups can have from 3 to about 20, 3 to about 10, or 3 to about 7 carbon atoms.
  • aryl refers to aromatic carbocyclyl groups including monocyclic or polycyclic aromatic hydrocarbons such as, for example, phenyl, naphthyl, anthracenyl, phenanthrenyl, indanyl, indenyl, and the like. In some embodiments, aryl groups have from 6 to about 18 ring-forming carbon atoms.
  • cycloalkyl refers to non-aromatic carbocyclyl groups including cyelized alkyl, alkenyl, and alkynyl groups. Cycloalkyl groups can include bi- or poly-cyclic ring systems. Example cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptatrienyl, norbornyl, norpinyl, norcarnyl, adamantyl, and the like.
  • cycloalkyl moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the cycloalkyl ring, for example, benzo derivatives of cyclopentane (indanyl), cyclohexane (tetrahydronaphthyl), and the like.
  • cycloalkyl groups can have 3, 4, 5, 6, or 7 ring forming carbon atoms.
  • cycloalkyl groups can have 0, 1, or 2 double or triple ring-forming bonds.
  • heterocarbocyclyl groups can be saturated or unsaturated carbocyclyl groups wherein one or more of the ring-forming carbon atoms of the carbocyclyl group is replaced with a heteroatom such as O, S, or N.
  • Heterocarbocyclyl groups can be aromatic (e.g., "heteroaryl") or non-aromatic (e.g., "heterocycloalkyl”).
  • Heterocarbocyclyl groups can correspond to hydrogenated and partially hydrogenated heteroaryl groups.
  • Heterocarbocyclyl groups can contain, in addition to at least one heteroatom, from about 1 to about 20, about 2 to about 10, or about 2 to about 7 carbon atoms and can be attached through a carbon atom or heteroatom.
  • heterocarbocyclyl groups include morpholino, thiomorpholino, piperazinyl, tetrahydrofuranyl, tetrahydrothienyl, 2,3-dihydrobenzofuryl, 1,3- benzodioxole, benzo- 1,4-dioxane, piperidinyl, pyrrolidinyl, isoxazolidinyl, isothiazolidinyl, pyrazolidinyl, oxazolidinyl, thiazolidinyl, imidazolidinyl, and the like.
  • heteroaryl groups are aromatic heterocarbocyclyl groups and include monocyclic and polycyclic aromatic hydrocarbons that have at least one heteroatom ring member such as sulfur, oxygen, or nitrogen.
  • Heteroaryl groups include, without limitation, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrrolyl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl indazolyl, 1,2,4-thiadiazolyl, isothiazolyl, benzothienyl, purinyl, carbazolyl, benzimidazolyl, and the like.
  • heteroaryl groups can have from 3 to about 20 ring-forming carbon atoms, and in further embodiments from about 3 to about 12 ring forming carbon atoms. In some embodiments, heteroaryl groups have 1 to about 4, 1 to about 3, or 1 to 2 heteroatoms.
  • heterocycloalkyl refers to a non-aromatic heterocarbocyclyl group including cyelized alkyl, alkenyl, and alkynyl groups where one or more of the ring-forming carbon atoms is replaced by a heteroatom such as an O, N, or S atom.
  • Ring-forming carbon and heteroatoms such as S and N can further be oxidized in a heterocycloalkyl moeity.
  • heterocycloalkyl moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the nonaromatic heterocyclic ring, for example phthalimidyl, naphthalimidyl pyromellitic diimidyl, phthalanyl, and benzo derivatives of saturated heterocycles such as indolene and isoindolene groups.
  • heterocycloalkyl groups have 3 to about
  • heterocycloalkyl groups have 3, 4, 5, 6, or 7 ring-forming atoms. In some embodiments, heterocycloalkyl groups have 0, 1, or 2 double or triple ring-forming bonds.
  • halo or halogen includes fluoro, chloro, bromo, and iodo.
  • alkoxy refers to an -O-alkyl group.
  • Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the like. In some embodiments, alkoxy groups have from 1 to 20, 1 to 12, 1 to 8, 1 to 6, 1 to 4 or 1 to3 carbon atoms.
  • alkoxyalkoxy refers to an -O-alkyl-O-alkyl group.
  • thioalkoxy refers to an alkoxy group in which the O atom is replaced by an S atom.
  • aryloxy refers to an -O-aryl group.
  • An example aryloxy group is phenoxy.
  • thioaryloxy refers to an aryloxy group in which the O atom is replaced by an S atom.
  • aralkyl refers to an alkyl moiety substituted by an aryl group.
  • Example aralkyl groups include benzyl and naphthylmethyl groups. In some embodiments, aralkyl groups have from 7 to 11 carbon atoms.
  • amino refers to an -NH 2 group.
  • Alkylamino refers to an amino group substituted by an alkyl group and “dialkylamino” refers to an amino group substituted by two alkyl groups.
  • aminoalkyl refers to an alkyl group substituted by an amino group.
  • hydroxy refers to -OH.
  • mercapto refers to -SH.
  • ureido refers to -NHCONH 2 .
  • sulfonyl refers to >SO 2 .
  • oxy refers to -O-.
  • alkylcarbonylalkenyl refers to an alkenyl group substituted by a carbonyl group which in turn is substituted by an alkyl group.
  • alkylcarbonylalkenyl refers to an alkenyl group substituted by a carbonyl group which in turn is substituted by an alkyl group.
  • carbocyclylalkyl refers to an alkyl moiety substituted by a carbocyclyl group.
  • Example carbocyclylalkyl groups include “aralkyl” (alkyl substituted by aryl) and “cycloalkylalkyl” (alkyl substituted by cycloalkyl).
  • carbocyclylalkenyl refers to an alkenyl moiety substituted by a carbocyclyl group.
  • Example carbocyclylalkenyl groups include
  • aralkenyl alkenyl substituted by aryl
  • cycloalkylalkenyl alkenyl substituted by cycloalkyl
  • carbocyclylalkynyl refers to an alkynyl moiety substituted by a carbocyclyl group.
  • Example carbocyclylalkynyl groups include
  • aralkynyl alkynyl substituted by aryl
  • cycloalkylalkynyl alkynyl substituted by cycloalkyl
  • heterocarbocyclylalkyl refers to an alkyl moiety substituted by a heterocarbocyclyl group.
  • Example heterocarbocyclylalkyl groups include “heteroarylalkyl” (alkyl substituted by heteroaryl) and “heterocycloalkylalkyl”
  • heterocarbocyclylalkenyl refers to an alkenyl moiety substituted by a heterocarbocyclyl group.
  • Example heterocarbocyclylalkenyl groups include “heteroarylalkenyl” (alkenyl substituted by heteroaryl) and
  • heterocycloalkylalkenyl (alkenyl substituted by heterocycloalkyl).
  • heterocarbocyclylalkynyl refers to an alkynyl moiety substituted by a heterocarbocyclyl group.
  • Example heterocarbocyclylalkynyl groups include “heteroarylalkynyl” (alkynyl substituted by heteroaryl) and
  • heterocycloalkynylalkyl (alkynyl substituted by heterocycloalkyl).
  • protecting group refers to a chemical functional group that can be selectively appended to and removed from functionalities, such as hydroxyl groups, amino groups, and carboxyl groups. Protecting groups are usually introduced into a chemical compound to render such functionality inert to chemical reaction conditions to which the compound is exposed. Any of a variety of protecting groups can be employed with the present invention.
  • a protecting group of an amino moiety can be referred to as an "amino protecting group” and a protecting group of a guanidino moiety can be referred to as a "guanidino protecting group.”
  • Example amino and guanidino protecting groups can also include t-butyloxycarbonyl (BOC), fluorenylmethoxycarbonyl (Fmoc), benzyloxycarbonyl (Cbz), and a phthalimido group.
  • BOC t-butyloxycarbonyl
  • Fmoc fluorenylmethoxycarbonyl
  • Cbz benzyloxycarbonyl
  • a phthalimido group include the following moieties:
  • R E and R F may be combined, with the nitrogen to which they are attached, to form a 5 to 7 membered heterocyclic ring, for example pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, and N-methylpiperazinyl.
  • a chemical group herein is "substituted" it may have up to the full valance of substitution, provided the resulting compound is a stable compound or stable structure; for example, a methyl group may be substituted by 1, 2, or 3 substituents, a methylene group may be substituted by 1 or 2 substituents, a phenyl group may be substituted by 1, 2, 3, 4, or 5 substituents, and the like.
  • leaving group refers to any group that can be replaced by a nucleophile upon nucleophilic substitution.
  • Example leaving groups include, halo (F, CI, Br, I), hydroxyl, alkoxy, mercapto, thioalkoxy, triflate, alkylsulfonyl, substituted alkylsulfonate, arylsulfonate, substituted arylsulfonate, heterocyclosulfonate or trichloroacetimidate.
  • stable compound or “stable structure” refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and preferably capable of formulation into an efficacious therapeutic agent.
  • the present invention is directed only to stable compounds.
  • the compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated.
  • the compounds herein described may have asymmetric centers which result in one enantiomer of a compound of Formula (I) demonstrating superior biological activity over the opposite enantiomer. Both of the configurations are considered part of the invention.
  • the R2 substituent of a compound of Formula (I) may exist in either an S or R configuration.
  • An example of a preferred enantiomeric configuration of the invention is a compound of Formula (I-s):
  • Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • the present invention also includes pharmaceutically acceptable salts of the compounds described herein.
  • pharmaceutically acceptable salts refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form.
  • Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like
  • organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic,
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
  • Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and in the Journal of Pharmaceutical Science, 66, 2 (1977), the disclosures of each of which are hereby incorporated by reference.
  • the reactions for preparing compounds of the invention can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis.
  • suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected.
  • Preparation of compounds of the invention can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3rd. Ed., Wiley & Sons, Inc., New York (1999), which is incorporated herein by reference in its entirety.
  • Reactions can be monitored according to any suitable method known in the art.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., ] H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., ] H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., ] H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or
  • Synthesis of compounds of the invention can involve a boron-containing fragment (FI) having a structure indicated by Formula (A).
  • the boronic ester moiety of FI can include, for example, a diol ester such as is indicated by the loop connecting oxygen atoms in Formula (A).
  • Stereochemistry at the carbon atom alpha to the boron atom in Formula (A) is indicated by the loop connecting oxygen atoms in Formula (A).
  • (A) can be controlled using an asymmetric boronic ester group in the preparation of FI.
  • pinanediol esters of boronic acid can facilitate the preparation or stereochemically pure, or substantially stereochemically pure, FI fragment.
  • the FI fragment can be prepared by reacting a compound of Formula (B) (showing a pinanediol boronic ester obtained from (+)-pinanediol) with a strong base (e.g., lithium diisopropylamide or lithium dicyclohexylamide) in the presence of dichloromethane or dibromomethane, followed by addition of a Lewis acid, (e.g., ZnCl 2 , ZnBr 2 , or FeCl 3 ) to yield a compound of Formula (C) (where L is halo) having a newly introduced stereocenter at the carbon alpha to the boron.
  • B a Lewis acid
  • the compound of Formula (C) can, in turn, be reacted with an alkali amide (e.g., lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide, and potassium bis(trimethylsilyl)amide) or other nucleophile that effectively inverts the newly formed stereocenter (such as by an SN2 type mechanism) and introduces an amine group (NR 2 ) in place of the halo group (e.g., chloro), forming a compound of Formula (D) (where R can be, e.g., alkyl, Si(alkyl) 3 , aryl, or aralkyl).
  • an alkali amide e.g., lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide, and potassium bis(trimethylsilyl)amide
  • R 2 amine group
  • R can be, e.g., alkyl, Si(alkyl) 3 , aryl, or aralkyl
  • the compound of Formula (D) can be further reacted with an agent capable of converting the NR 2 group to NH 2 , or salt thereof, to form an FI fragment substantially capable of coupling with a further fragment through the amine.
  • a suitable agent for converting the NR 2 ' group to NH 2 can be a protic acid such as HCl such as when R is a silyl group (e.g., trimethylsilyl).
  • the compound of Formula (B) can also be prepared according to a two step procedure involving reaction of a trialkoxyborane, preferably triisopropoxyborane, with (IS, 2S, 3R, 5S)-(+) pinanediol, to give a mono-alkoxy [(IS, 2S, 3R, 5S)-(+) pinanediol] borane intermediate wherein two of the alkoxy groups of the trialkoxy borane have been replaced by (IS, 2S, 3R, 5S)-(+) pinanediol.
  • This mixed pinanediol alkoxy borane upon reaction with the appropriate organometallic derivative, e.g.
  • Example A.2 herein.
  • the present invention is further directed to methods of preparing compounds of Formula (II):
  • each R 17 is, independently, Ci-do alkyl or C 3 -C ⁇ o cycloalkyl; for a time and under conditions suitable for forming a mixed trialkoxyborane intermediate of Formula (II-c):
  • R 17 is d-C 4 alkyl.
  • R 17 is isopropyl
  • the diol of Formula (Il-b) is pinanediol, pinacol, bicyclohexyl-l,l'-diol, 1,2-ethanediol, 1,3-propanediol, 1,2-propanediol, 2,3-butanediol, 1 , 1 ,2,2-tetramethylethanediol, 1 ,2-diisopropylethanediol, 5,6-decanediol, 1,2-dicycIohexylethanediol, bicyclohexyl-l,l'-diol, diethanolamine, or 1 ,2-di ⁇ henyl- 1 ,2-ethanediol.
  • the diol of Formula (Il-b) is pinanediol.
  • the Formula R'C ⁇ MX 1131 is a Grignard reagent.
  • the Formula R'CH ⁇ -MX 1131 is R ! CH 2 MgBr.
  • R 1 is isopropyl
  • the present invention provides a process for preparing a compound of Formula (Il-i):
  • ⁇ -0 i comprising: a) reacting (IS, 2S, 3R, 5S)-(+)-pinanediol with triisopropoxy borane for a time and under conditions suitable for forming an intermediate of Formula (Il-ii):
  • the present invention provides a compound of
  • the reacting steps can be carried out in any suitable solvent that is non- reactive with the reagents and products and allows combining of reagents at lowered temperatures (e.g., temperatures colder than room temperature).
  • suitable solvents include ethers such as dimethoxymethane, tetrahydrofuran, 1,3-dioxane, 1,4-dioxane, furan, diethyl ether, ethylene glycol dimethyl ether, ethylene glycol diethyl ether, diethylene glycol dimethyl ether, diethylene glycol diethyl ether, triethylene glycol dimethyl ether, anisole, or t-butyl methyl ether.
  • the ether solvent contains tetrahydrofuran and/or diethyl ether,
  • reaction temperatures will depend on, for example, the melting and boiling points of the reagents and solvent, if present; the thermodynamics of the reaction (e.g., vigorously exothermic reactions may need to be carried out at reduced temperatures); and the kinetics of the reaction (e.g., a high activation energy barrier may need elevated temperatures).
  • Elevated temperature refers to temperatures above room temperature (about 22 °C) and “reduced temperature” refers to temperatures below room temperature.
  • suitable temperatures are reduced temperatures.
  • the reaction of the trialkoxyborane and diol to prepare a mixed trialkoxyborane intermediate can be carried out, for example, at a temperature of about -20 to about 10 °C. In some embodiments, the reaction of the trialkoxyborane and diol can be carried out at about 0 °C.
  • the reaction of the mixed trialkoxyborane intermediate with the organometallic reagent R'CTyVTX 1181 or the alkyl lithium reagent R ! CH 2 Li can be carried out, for example, at temperature from about -100 to about -20 °C. In some embodiments, the reaction of the mixed trialkoxyborane intermediate and R ⁇ B ⁇ MX 1 " 1 is carried out at about -78 °C.
  • reactions of the processes described herein can be carried out in air or under an inert atomosphere.
  • reactions containing reagents or products that are_ substantially reactive with air can be carried out using air-sensitive synthetic techniques that are well known to the skilled artisan.
  • the mid-section of compounds of the present invention can be represented by fragment F2 which couples to fragment FI by peptide bond formation for form an F2-F1 intermediate.
  • Methods for coupling compounds through peptide bonds, or amide bonds, are well known in the art and described, for example, in The Peptides: Analysis, Synthesis, Biology, Vol. I., eds. Gross, et al., Academic Press, 1979.
  • An example F2 fragment is provided in Formula (E) (Pg is an amino protecting group, R 2 is defined herein). Additionally, protection of the amino group of amino acids using Boc or other amino protecting groups is well known in the art.
  • aza-serines can be prepared generally by the Hoffman Rearrangement (Hoffman's Reaction) using, for example, asparagine where the amide of the asparagine side chain is converted to an amine (which can be subsequently protected). Methods for carrying out Hoffman Rearrangements, such as for amino acids, are known in the art and also provided in the Examples below. Additionally aza-serines can be prepared as disclosed in Zhang, et al. J Org. Chem., 1997, 62, 6918-6920.
  • a further fragment (F3) can be coupled to the F2 fragment of the F2-F1 intermediate by any of various means such as by nucleophilic substitution or addition reactions where, for example, F2 contains a nucleophile (e.g., amine) and F3 contains an electrophile (e.g., CO, SO 2 , and the like) and optionally a leaving group (e.g., halo, hydroxy, alkoxy, alkylsulfonyl, arylsulfonyl, and the like).
  • a nucleophile e.g., amine
  • F3 contains an electrophile (e.g., CO, SO 2 , and the like) and optionally a leaving group (e.g., halo, hydroxy, alkoxy, alkylsulfonyl, arylsulfonyl, and the like).
  • Example F3 fragments can have the formula R x COX L , R x SO 2 X L , R x NCO, or R x HCO, (e.g., R can be R A , R B , or R c as defined herein and X can be a leaving group).
  • Coupling of R x COX L (such as when X L is OH) to the F2-F1 intermediate can be carried out according to standard procedures for peptide bond formation to prepare compounds having the formula F3-F2- Fl where the F3 and F2 fragments are coupled via an amide bond.
  • F3 and F2 can be coupled by a sulfonylamino linkage prepared by reacting R x SO 2 X L with the F2-F1 intermediate in which an amino moiety on the F2-F1 intermediate displaces the X L leaving group of R SO 2 X L .
  • reaction of R x NCO with an amino moiety of the F2-F1 intermediate can result in a urea linkage (- HNCONH-), while reaction of R x HCO with an amino moiety of the F2-F1 intermediate followed by reduction of the resulting imine moiety can form an amine linkage.
  • Other coupling means are known in the art and are also suitable.
  • F3 fragments can be obtained from commercial sources or made by methods known in the art.
  • R 2 is -(CH 2 ) d CH(R 7 )NR 9 R 10
  • R 10 can be prepared by removal of an R 1 amino protecting group to form the corresponding deprotected compound wherein R 10 is H.
  • This deprotected compound can be reacted with a reagent having the formula R IOa X L , wherein R 10 has the same meaning as R 10 with the exception of H and X L is a leaving group such as halo or a sulfonic acid derivative, or wherein R 10a and X taken together represent, for example, a reactive alkyl, carbocyclyl or heterocarbocyclyl isocyanate, or an alkyl, carbocyclyl, heterocarbocyclyl sulphonylisocyanate.
  • Example D.26 can be prepared by the deprotection of the benzyloxycarbonyl group of Example D.16.6 to give Example D.17, from which the azaserine nitrogen can be subsequently acylated.
  • the present invention further provides methods for preparing azaserine
  • a reducing agent such as a hydrogenation reagent.
  • the hydrogenation reagent contains H 2 which is optionally used in the presence of a metal catalyst (e.g., Pd/C 10%). Hydrogenation can be further carried out in the presence of a protic acid such as HCl and in a suitable hydrogenation solvent containing, for example, an alcohol and/or an ether solvent.
  • the hydrogenation solvent contains an ether such as 1,4-dioxane.
  • the hydrogenation solvent cotains an alcohol such as methanol.
  • the hydrogenation solvent contains a mixture of alcohol and ether.
  • An example preparation of an azaserine compound according to this process is provided, for example, in Example D.17. Reaction parameters including temperature, pressure, atomosphere and the like are readily dete ⁇ nined by one skilled in the art of chemical synthesis and reaction progress can be monitored by routine methods including, e.g., NMR.
  • the present invention provides a process for the preparation of compounds of Formula (I):
  • R 1 is Ci-C ⁇ alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, or C 3 -C 7 cycloalkyl;
  • R 2 is -CH 2 NH 2 ;
  • Q is -B(OR I4 ) 2 , or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R 14 is C1-C 4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • R A is Ci -C 20 alkyl optionally substituted with R 20 ; C 2 -C 2 o alkenyl optionally substituted with R 20 ; C 2 -C 2 o alkynyl optionally substituted with R 20 ; carbocyclyl optionally substituted with 1-5 R 21 ; or heterocarbocyclyl optionally substituted with 1-5 R 21 ;
  • R 20 is selected from the group consisting of: -CN, halo, haloalkyl-, d-C
  • the hydrogenation agent is H 2 in the presence of
  • Hydrolysis conditions can include contacting a boronic ester with excess acid, such as a protic acid like HCl.
  • boronic acids can be esterified by contacting the acid compound
  • esterification reaction can be acid or base catalyzed.
  • Step 1 2-(2-methylpropyl)-(3aS, 4S, 6S, 7aR)-hexahydro-3a, 5, 5-trimethyl-4, 6-methano- 1, 3,2-benzodioxaborole
  • Step 2 2-[(lS)-l-chloro-3-methylbutyl]-(3aS,4S,6S, 7aR)-hexahydro-3a,5,5-trimethyl- 4, 6-methano-l, 3,2-benzodioxaborole
  • the reaction mixture was stirred at -78°C for 3 hours, then allowed to warm to room temperature. After removal of the solvents by rotary evaporation the residue was partitioned between petroleum ether (1000 ml) and a 10% aqueous solution of ammonium chloride (800 ml). The aqueous layer was further extracted with petroleum ether (300 ml). The combined organic phases were dried over anhydrous sodium sulfate and concentrated. The product was obtained as a brown oil (59.0 g, 98% yield) containing about 9% mol/mol of starting material ( ⁇ -NMR), and was used in the subsequent step without further purification.
  • Step 3 N,N-Bis(trimethylsilyl)-(lR)-l-[(3aS,4S,6S, 7aR)-hexahydro-3a,5,5-trimethyl- 4, 6-methano-l, 3, 2-benzodioxaborol-2-yl]-3-methylbutylamine
  • Step 4 (lR)-l-[(3aS,4S,6S, 7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt
  • Step 1 2-(l-methylethoxy)-(3aS,4S,6S, 7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1, 3, 2-benzodioxaborole.
  • Step 2 2-(2-methylpropyl)-(3aS,4S,6S, 7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1, 3, 2-benzodioxaborole.
  • the aqueous phases were further extracted with ethyl acetate (150 ml).
  • the combined organic phases were dried over sodium sulfate and concentrated.
  • the resulting oily residue was redissolved in ethyl acetate (500 ml) and the solution was washed with cold water (200 ml).
  • the aqueous phases were further extracted with ethyl acetate (500 ml).
  • the combined organic phases were dried over sodium sulfate and concentrated.
  • the residue was dissolved in diethyl ether (100 ml) an the solution was slowly added to hexane (600 ml) while stirring.
  • the white solid was collected by filtration (43.4 g) and purified by column chromatography eluting initially with 50:50 hexane:ethyl acetate mixture and then with ethyl acetate. The fractions containing the product were concentrated, the residue was dissolved in diethyl ether (100 ml) and the resulting solution was slowly added to hexane (600 ml) while stirring. The white solid was collected by filtration (15.2 g, 66% yield).
  • Boc-L-threonine (870 mg, 3.97 mmol, 1.2eq.) was dissolved in DMF dry
  • Example B.9 Synthesis of Further Compounds [00205] Following the procedures of Examples B.4-B.8, the following compounds can be prepared by reaction of (lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt of Example A.l and intermediates of Examples G.l 1, G.12 and G.13.
  • Example B 10 Carbamic acid 1,1-dimethylethyl ester, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-l- methylbutyl] amino] carbonyl] -methyl
  • Example B.l (4.04 g, 7.06 mmol) in a mixture of dioxane (40 ml) and diethyl ether (7 ml), while cooling at 0°C.
  • the reaction mixture was allowed to warm to room temperature and stirred for further 4 hours.
  • the solvent was removed by rotary evaporation, the residue was treated with diethyl ether (50 ml) and the mixture was stirred at r.t. for three days.
  • the resulting solid was collected by filtration affording 3.18 g of pure product (90% yield)
  • Example B.l (3.1 g, 5.48 mmol) was carefully dissolved, under nitrogen at 0°C, in 20 mL of HCl 37%; the resultant mixture was allowed to warm to room temperature and to stir overnight. The reaction mixture was washed with Et 2 O until complete removal of pinanediol; the aqueous solution was concentrated to dryness and dried in vacuo to afford 1.82 g (4.93 mmol, yield 90%) of the title compound, used without further purification.
  • Example C.9 Synthesis of further compounds [00151] Following the procedures of Examples C.4-C.8, the following compounds can be prepared starting from intermediates of Example B.9.
  • Example D.2 10-(l,3-Dioxo-l,3-dihydro-isoindol-2-yI)-decanamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaboroI- 2-yl]-3-methylbutyl]amino]carbonyI]-4-[[imino(nitroamino)methyl]amino]butyl]-;
  • the reaction mixture was stirred at -15 to 10 °C for 4h, then concentrated to small volume and partitioned between ethyl acetate (20 ml) and water (10 ml). The aqueous phase was further extracted with ethyl acetate (10 ml). The combined organic phases were dried over sodium sulfate and concentrated. The residue was taken up with ethyl acetate (3 ml) and the solution was dropwise added to hexane (120 ml) while stirring at room temperature. The solid was collected by decantation and dried under vacuum (730 mg, 94%).
  • Example D.2.6 Further compounds prepared according to the above reported procedure in Example D.2 are reported in Table D-2A
  • the compound of Example D.2.6 was prepared starting from 2-aminoacetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a, 5,5-trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 -methylbutyl] - amino]carbonyl]-4-[[imino(nitroamino)methyl]-amino]-butyl], hydrochloride salt of Example D.14.
  • Examples 2.9 and 2.10 were prepared from (2S)-2- amino-5-ureidopentanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- i 4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]; hydrochloride salt of Example C.3
  • PS-Carbodiimide N-cyclohexylcarbodiimide-N'-propyloxymethyl polystyrene, 769 mg, 1 mmol, loading 1.31 mmol/g
  • HOAt l-Hydroxy-7- azabenzotriazole, 115 mg, 0.85 mmol
  • Example D.6 N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyI-4,6-methano- l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]-N'-(l-naphthyI) urea.
  • Example D.8 Decanamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a s 5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methyIbutyI]amino]carbonyI]-2- hydroxypropyl]- Chiral
  • Example D.8, D.8.1 and D.8.2 are reported in Table D-8.
  • D.8.7, D.8.11, D.8.12 and D.8.13 were prepared according to literature procedures.
  • Compound 2,2-dimethyldecanoic acid was prepared as described by Roth et al. in J Med. Chem. 1992, 35, 1609-1617.
  • Compounds 4-(3-pyridyl)benzoic acid, 3-(3- Pyridyl)benzoic acid and 6-phenyl-2-pyridinecarboxilic acid were prepared according the procedure described by Gong et al. in Synlett, 2000, (6), 829-831.
  • Compound 3- propoxybenzoic acid was prepared according the procedure described by Jones in J. Chem. Soc. 1943, 430-432.
  • Example D.8, D.8.1 and D.8.2 This compound has been prepared essentially according to the above procedures for Example D.8, D.8.1 and D.8.2 starting from (2S)-2-amino-3- carbamoylpropanamide, N-[(1R)- 1 -[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyI-4,6- methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]; hydrochloride salt of Example C.3.
  • Example D.8.20 This compound has been prepared essentially according to the above procedures for Example D.8, D.8.1 and D.8.2 starting from (2S)-2-amino-3-carbamoylpropanamide, N- [(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]; hydrochloride salt of Example C.3.
  • Example D.8.20 Example D.8.20
  • Example D.8, D.8.1 and D.8.2 This compound has been prepared essentially according to the above procedures for Example D.8, D.8.1 and D.8.2 starting from (2S)-2-amino-3- carbamoylpropanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-l,3,2-benzodioxaborol-2-yI]-3-methylbutyl]; hydrochloride salt of Example C.3.
  • Example D.16 Synthesis of Further Compounds [00253] Following the procedures of Examples D.9-D.13, the following compounds can be prepared by reaction of decanoic acid with the intermediates of Example C.9.
  • Example D.17 4-Butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaboroI-2-yl]-3- methylbutyl]amino]carbonyl]-2-(aminoethyI)-hydrochloride salt.
  • the mixture was stirred at 0 -5 °C for 30', then at 10 °C for 1 h.
  • the solvent was removed under reduced pressure, the crude was dissolved in Ethyl acetate and washed with a solution of citric acid 2% (50 ml) then with a solution of sodium bicarbonate 2% (50ml) and a solution of sodium chloride 2% (50 ml).
  • the solution was dried over anhydrous sodium sulfate and the solvent evaporated under reduced pressure.
  • Triethylamine (0,4 ml, l,8mmol, 2,2 eq.) and (4-methylphenyl)-ureido- sulfonylchloride (0,34g, l,38mmol, l,09eq.) of example G.1X have been added.
  • the suspension was stirred at 25 °C for lh, then was poured in a citric acid 1% solution (30ml) and extracted with Ethyl acetate (50ml).
  • Example D.l 8 - D.23 Following the procedures of Examples D.l 8 - D.23, the following compounds can be prepared by reaction of the intermediates of Example D.17 or D.25 with the appropriate commercially available carboxylic acids, acyl halides, sulphonyl halides, isocyanates, sulphonylisocyanates, or with the compounds of Examples G.14,
  • Example D.25 Synthesis of Further Compounds [00262] Following the procedures of Example D17, the following compounds can be prepared starting from the compounds of Example D.16.8 and D.16.9.
  • Example D.26 4-Butylbenzamide, N-[(lR)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyI-4,6-methano-l,3,2-benzodioxaborol-2-yI]-3- methylbutyl]amino]carbonyl]-2-[(4-methylbenzoyl)amino]ethyI]-
  • Example D.1.1 (564 mg, 0.90 mmol), 2-methylpropylboronic acid (222 mg, 2.19 mmol) and 4N hydrogen chloride dioxane solution (225 ⁇ l) in a 40:60 heterogeneous mixture of methanol:hexane (10 ml) was stirred at room temperature for 4 hours. Hexane (4 ml) was added, the mixture was stirred for a while, then the hexane layer was removed. Fresh hexane (5 ml) and 2-methylpropylboronic acid (100 mg, 0.99 mmol) were added and the mixture was stirred at room temperature for 3 hours.
  • Example D.8.3 (155 mg, 0.283 mmol), 2-methylpropylboronic acid (81 mg, 0.793 mmol) and 2N aqueous hydrochloric acid (0.3 ml) in a heterogeneous mixture of methanol (3 ml) and hexane (3 ml) was stirred at room temperature for 24 hours.
  • the solution was washed with a mixture of NaCl saturated solution (7 ml) and 10% NaHCO 3 (2 ml). The layers were separated and the aqueous phase was further washed with ethyl acetate (2 x 20 ml). The combined organic phases were dried over sodium sulfate and concentrated. The residue was taken up with ethyl acetate (about 20 ml) and the minimum amount of methanol, and then concentrated to small volume (about 5 ml). The resulting white was collected by filtration and dried under vacuum at 50°C (160 mg, 65% overall yield).
  • Example E.4 are reported in Table E-4. Table E-4
  • Example F using the appropriate boronic acid starting material and bicyclohexyl-1,1'- diol.
  • Step 1 2-undec-l 0-enyl-l ,3-dioxo-l ,3 -dihydro isoindole
  • Step 2 10-(l ,3-Dioxo-l ,3-dihydroisoindol-2-yl)decanoic acid.
  • Step 1 N-tert-butoxycarbonyl-L-asparagine [Commercially available] o ⁇
  • Step 2 N-[(l,l-dimethylethoxycarbonyl)amino] -L-asparagine, benzyl ester.
  • Step 3 3-Amino-2-S-[(l,l-dimethylethoxycarbonyl)amino]-propionic acid, benzyl ester.
  • Step 1 2-S-[(l, l-dimethylethoxycarbonyl)amino]-3-(4-methylbenzoylamino)propionic acid, benzyl ester.
  • Step 2 (2S)-2-[(l,l-dimethylethoxycarbonyl)amino]-3-(4- methylbenzoylaminojpropionic acid.
  • Example G.7 2-S-[(l,l-dimethylethoxycarbonyl)ammo]-3-(hexanoyIamino)propionic acid.
  • Stepl 2-S-[(l, l-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid, benzyl ester.
  • Step 2 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid.
  • Example G.8 2-S-tert-butoxycarbonylamino-3-(4-fluorosulfonylamino)propionic acid.
  • Step 1 2-S-[(l, 1-dimethylethoxycarbonyl) amino] -3-(4-fluorosulfonylamino)propionic acid, benzyl ester.
  • Step 2 2-S-[(l, l-dimethylethoxycarbonyl)amino]-3-(4-fluorosulfonylamino)propionic acid.
  • Step 1 2-S-[(l, l-dimethylethoxycarbonyl)amino]-3-(3, 4-dimethoxyphenylacetamido)- propionic acid, benzyl ester.
  • Step 2 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(3,4-dimethoxyphenylacetamido)- propionic acid.
  • Example G 10 2-[(l,l-dimethyIethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid.
  • Step 1 2-[(l,l-dimethy/ethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid, benzyl ester.
  • Step 2 2- [(l,l-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid.
  • Example G.ll Synthesis of Further Compounds [00299]
  • the following compounds can be prepared starting from 3-amino-2-S- [(l,l-dimethylethoxycarbonyl)amino]propionic acid, benzyl ester of Example G.5, with the methods described in Step 1 and Step 2 of Examples G.6-G.10.
  • Example G.12 2-[(l,l-Dimethylethoxycarbonyl)amino]-3-(3-benzyloxycarbonylamino)propionic acid.
  • N-tert-butoxycarbonyl-L-asparagine from step 1 of Example G.5 or commercially available, (8 g, 0.034 mol, leq.) was suspended in ethyl acetate (72 ml), acetonitrile (72ml) and water (36ml), and lodobenzenediacetate (13,3 g, 0.041 mol, 1,2 eq.) was added at 5°C. The mixture was stirred at 10°-25°C for 3-4h, then a white solid came off. The solid was filtered, washed with diethyl ether and dried under vacuum to give a white powder . Yield 57%. 4g.
  • Step 2 2-[(l , 1-dimethylethoxy carbonyl) amino) '-3-(3- benzyloxycarbonylamino)propionic acid.
  • benzylchloroformate 3ml, 0,020 mol, 1,1 eq.
  • the mixture was poured in water (100ml) and washed with diethyl ether (100ml).
  • Step 1 l-[(l,l-Dimethylethoxycarbonyl)amino]-piperidine-4-carboxylic acid
  • Piperidine-4-carboxylic acid (5g, 38.7 mmol, leq.) was dissolved in sodium carbonate solution (4.5g, 42.61mmol, 2.2 eq.), 70ml, and 1,4-dioxane (30ml).
  • a solution of di-tert-butyldicarbonate (9.3g, 42.61mmol, l.leq.) in 1,4-dioxane (40ml) was added dropwise and the resulting mixture was stirred overnight at room temperature.
  • the organic solvent was removed under reduced pressure and the resulting solution was acidified with HCl 37% till pH 2.
  • the obtained suspension was filtered , the white solid washed with diethyl ether (5ml).
  • Step 2 l-[(l,l-Dirnethylethoxycarbonyl)amino]-piperidine-4-carboxylic acid benzyl ester
  • Step 3 Piperidine-4-carboxylic acid benzyl ester, hydrochloride salt.
  • Step 4 l-Methanesulfonyl-piperidine-4-carboxylic acid benzyl ester.
  • Example G.1 Isoxazole-5-carboxylic acid.
  • the present compounds can inhibit proteasome activity.
  • Table F-l below provides data related to several Example compounds of the invention with respect to, for example, ability to inhibit proteasome activity.
  • proteasome inhibitors can modulate, such as induce, apoptosis in a cell.
  • the compounds herein can kill tumor cells by induction of apoptosis.
  • the present compounds can be used to treat cancer, tumors or other proliferative disorders.
  • inhibition of proteasome function by compounds of the invention can inhibit the activation or processing of transcription factor NF- ⁇ B. This protein plays a role in the regulation of genes involved in the immune and inflammatory responses as well as in cell viability.
  • Inhibition of proteasome function can also inhibit the ubiquitination/proteolysis pathway.
  • This pathway catalyzes, inter alia, selective degradation of highly abnormal proteins and short-lived regulatory proteins.
  • compounds of the invention can prevent the degradation of p53 which is typically degraded by the ubiquitin-dependent pathway.
  • the ubiquitination/proteolysis pathway also is involved in the processing of internalized cellular or viral antigens into antigenic peptides that bind to MHC-I molecules.
  • the compounds of the invention can be used to reduce the activity of the cytosolic ATP- ubiquitin-dependent proteolytic system in a number of cell types.
  • the usefulness of such compounds can include therapeutics, such as the treatment of various diseases or disorders associated with proteasome.
  • the methods include administering a therapeutically effective amount of a compound of the invention, or composition thereof, to a mammal, such as a human having a disease or disorder associated with ⁇ proteasome.
  • therapeutically effective amount refers to an amount sufficient to prevent, alleviate, or ameliorate any phenomenon, such as a cause or symptom, known in the art to be associated with the disease or disorder.
  • Treatable diseases or disorders can be associated with either normal or abnormal activities of proteasome, such as the regulation of apoptosis.
  • Numerous diseases or disorders that are associated with proteasome, or that are desirably treated by induction of apoptosis are known and include, for example, various cancers and tumors including those associated with skin, prostate, colorectal, pancreas, kidney, ovary, mammary, liver, tongue, lung, and smooth muscle tissues.
  • Preferred tumors that can be treated with proteasome inhibitors include, but are not limited to hematological tumors, such as, for example, leukemias, lymphomas, non-Hodgkin lymphoma, myeloma, multiple myeloma, as well as solid tumors such as, for example, colorectal, mammary, prostate, lung, and pancreas tumors.
  • the proteasome inhibitors can be administered to patients as single agents or in combination with one or more antitumor or anticancer agent and/or radiotherapy.
  • anti-tumor or anti-cancer agents which can be advantageously administered concomitantly with a proteasome inhibitor include but are not limited to, adriamycin, daunomycin, methotrexate, vincristin, 6-mercaptopurine, cytosine arabinoside, cyclophosphamide, 5-FU, hexamethylmelamine, carboplatin, cisplatin, idarubycin, paclitaxel, docetaxel, topotecan, irinotecam, gemcitabine, L-PAM, BCNU and VP-16.
  • Further diseases or disorders associated with the proteasome include accelerated or enhanced proteolysis that occurs in atrophying muscles, such as is often associated with activation of a nonlysomal ATP-requiring process involving ubiquitin. Accelerated or enhanced proteolysis can be the result of any of numerous causes including sepsis, burns, trauma, cancer, infection, neurodegenerative diseases such as muscular dystrophy, acidosis, or spinal nerve injuries, corticosteroid use, fever, stress, and starvation.
  • Compounds of the invention can be tested for inhibition of muscle wastage by any various procedures known in the art such as by measuring urinary excretion of modified amino acid 3-methylhistidine (see, e.g., Young, et al., Federation Proc, 1978, 37, 229).
  • Compounds of the present invention can be further used to treat or prevent diseases or disorders associated with activity of NF- ⁇ B including for example, human immunodeficiency virus (HIV) infection and inflammatory disorders resulting from, for example, transplantation rejection, arthritis, infection, inflammatory bowel disease, asthma, osteoporosis, osteoarthritis, psoriasis, restenosis, and autoimmune diseases.
  • HIV human immunodeficiency virus
  • inflammatory disorders resulting from, for example, transplantation rejection, arthritis, infection, inflammatory bowel disease, asthma, osteoporosis, osteoarthritis, psoriasis, restenosis, and autoimmune diseases Accordingly, a process that prevents activation of NF- ⁇ B in patients suffering from such a disease would be therapeutically beneficial. Inhibition of the NF- ⁇ B activity can be measured by using a DNA binding assay such a described in Palombella, et al., Cell, 1994, 78, 112,.
  • HEP Human Erythrocyte Proteasome
  • the concentration of free AMC (aminomethylcoumarin) was determined on a Perkin Elmer HTS 7000 Plus microplate reader, excitation 370 nM and emission 465nM. Proteasome activity was determined under conditions in which substrate hydrolysis increased linearly with time and the change in fluorescence signal was proportional to the concentration of free AMC.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Communicable Diseases (AREA)
  • Rheumatology (AREA)
  • Hematology (AREA)
  • Pulmonology (AREA)
  • Dermatology (AREA)
  • Neurology (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Neurosurgery (AREA)
  • Diabetes (AREA)
  • Obesity (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • AIDS & HIV (AREA)
  • Vascular Medicine (AREA)
  • Biomedical Technology (AREA)
  • Transplantation (AREA)
  • Urology & Nephrology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides boronic acid compounds, boronic esters, and compositions thereof that can modulate apoptosis such as by inhibition of proteasome activity. The compounds and compositions can be used in methods of inducing apoptosis and treating diseases such as cancer and other disorders associated directly of indirectly with proteasome activity.

Description

PROTEASOME INHIBITORS AND METHODS OF USING THE SAME
FIELD OF THE INVENTION
[0001] The present invention relates to boronic acid and boronic ester compounds useful as proteasome inhibitors and modulation of apoptosis.
BACKGROUND OF THE INVENTION
[0002] The proteasome, (also refered to as multicatalytic protease (MCP), multicatalytic proteinase, multicatalytic proteinase complex, multicatalytic endopeptidase complex, 20S, 26S, or ingensin) is a large, multiprotein complex present in both the cytoplasm and the nucleus of all eukaryotic cells. It is a highly conserved cellular structure that is responsible for the ATP-dependent proteolysis of most cellular proteins (Tanaka, Biochem Biophy. Res. Comrnun., 1998, 247, 537). The 26S proteasome consists of a 20S core catalytic complex that is capped at each end by a 19S regulatory subunit. The archaebacterial 20S proteasome contains fourteen copies of two distinct types of subunits, α and β, which form a cylindrical structure consisting of four stacked rings. The top and bottom rings contain seven α-subunits each, while the inner rings contain seven β-subunits. The more complex eukaryotic 20S proteasome is composed of about 15 distinct 20-30 kDa subunits and is characterized by three major activities with respect to peptide substrates. For example, the proteasome displays tryptic-, chymotryptic-, and peptidylglutamyl peptide-hydrolytic activities ( ivett, Biochem. J., 1993, 291, 1 and Orlowski, Biochemistry, 1990, 29, 10289). Further, the proteasome has a unique active site mechanism which is believed to utilize a threonine residue as the catalytic nucleophile (Seemuller, et al., Science, 1995, 268, 579). [0003] The 26S proteasome is able to degrade proteins that have been marked by the addition of ubiquitin molecules. Typically, ubiquitin is attached to the ε-amino groups of Iysines in a multistep process utilizing ATP and El (ubiquitin activating) and E2 (ubiquitin-conjugating) enzymes. Multi-ubiquitinated substrate proteins are recognized by the 26S proteasome and are degraded. The multi-ubiquitin chains are generally released from the complex and ubiquitin is recycled (Goldberg, et al., Nature, 1992, 357, 375). [0004] Numerous regulatory proteins are substrates for ubiquitin dependent proteolysis. Many of these proteins function as regulators of physiological as well as pathophysiological cellular processes. Alterations in proteasome activity have been implicated in a number of pathologies including neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, as well as occlusion/ischaemia reperfusion injuries, and aging of the central nervous system.
[0005] The ubiquitin-proteasome pathway also plays a role in neoplastic growth.
The regulated degradation of proteins such as cyclins, CDK2 inhibitors, and tumor suppressors is believed to be important in cell cycle progression and mitosis. A known substrate of the proteasome is the tumor suppressor p53 which is involved in several cellular processes (see, e.g., Ko, L. J. Genes Dev., 1996, 10, 1054). Tumor suppressor p53 has been shown to induce apoptosis in several haematopoietic cell lines (Oren, M., Semin. Cancer Biol., 1994, 5, 221). Induction of p53 leads to cell growth arrest in the Gl phase of the cell cycle as well as cell death by apoptosis. Tumor suppressor p53 degradation is known to be carried out via the ubiquitin-proteasome pathway, and disrupting p53 degradation by inhibition of the proteasome is a possible mode of inducing apoptosis.
[0006] The proteasome is also required for activation of the transcription factor
NF-κB by degradation of its inhibitory protein, IκB (Palombella, et al., Cell, 1994, 78, 773). NF-κB has a role in maintaining cell viability through the transcription of inhibitors of apoptosis. Blockade of NF-κB activity has been demonstrated to make cells more susceptible to apoptosis.
[0007] Several inhibitors of the proteolytic activity of the proteasome have been reported. See, for example, Kisselev, et al, Chemistry & Biology, 2001, 8, 739. Lactacystin is a Streptomyces metabolite that specifically inhibits the proteolytic activity of the proteasome complex (Fenteany, et al., Science, 1995, 268, 726). This molecule is capable of inhibiting the proliferation of several cell types (Fenteany, et al., Proc. Natl. Acad. Sci. USA, 1994, 91, 3358). It has been shown that lactacystin binds irreversibly, through its β-lactone moiety, to a threonine residue located at the amino terminus of the β- subunit of the proteasome.
[0008] Peptide aldehydes have been reported to inhibit the chymotrypsin-like activity associated with the proteasome (Vinitsky, et al., Biochemistry, 1992, 31, 9421; Tsubuki, et al., Biochem. Biophys. Res. Commun., 1993, 196, 1195; and Rock, et al., Cell, 1994, 78, 761). Dipeptidyl aldehyde inhibitors that have IC50 values in the 10-100 nM range in vitro (Iqbal, M., et al., J. MedChem., 1995, 38, 2276) have also been reported. A series of similarly potent in vitro inhibitors from α.-ketocarbonyl and boronic ester derived dipeptides has also been reported (Iqbal, et al., Bioorg. Med. Chem. Lett., 1996, 6, 287, U.S. Pat. Nos. 5,614,649; 5,830,870; 5,990,083; 6,096,778; 6,310,057; U.S. Pat. App. Pub. No. 2001/0012854, and WO 99/30707). [0009] N-terminal peptidyl boronic ester and acid compounds have been reported previously (U.S. Pat. Nos. 4,499,082 and 4,537,773; WO 91/13904; Kettner, et al., J. Biol. Chem., 1984, 259(24), 15106). These compounds are reported to be inhibitors of certain proteolytic enzymes. N-terminal tri-peptide boronic ester and acid compounds have been shown to inhibit the growth of cancer cells (U.S. Pat. No. 5,106,948). A broad class of N-terminal tri-peptide boronic ester and acid compounds and analogs thereof has been shown to inhibit renin (U.S. Pat. No. 5,169,841). [0010] Various inhibitors of the peptidase activities of the proteasome have also been reported. See, e.g., Dick, et al., Biochemistry, 1991, 30, 2725; Goldberg, et al., Nature, 1992, 357, 375; Goldberg, Eur. J. Biochem., 1992, 203, 9; Orlowski, Biochemistry, 1990, 29, 10289; Rivett, et al., Archs. Biochem. Biophys., 1989, 218, 1; Rivett, et al., J. Biol. Chem., 1989, 264, 12215; Tanaka, et al., New Biol, 1992, 4, 1; Murakami, et al., Proc. Natl. Acad Sci. USA, 1986, 83, 7588; Li et al., Biochemistry, 1991, 30, 9709; Goldberg, Eur. J. Biochem., 1992, 203, 9; and Aoyagi, et al., Proteases and Biological Control, Cold Spring Harbor Laboratory Press (1975), pp.429-454. [0011] Stein et al., U.S. patent application Ser. No. 08/212,909, filed March 15,
1994, report peptide aldehydes useful for reducing in an animal both the rate of loss of muscle mass and the rate of intracellular protein breakdown. The compounds are also said to reduce the rate of degradation of p53 protein in an animal. Palombella, et al., WO 95/25533, report the use of peptide aldehydes to reduce the cellular content and activity of NF-κB in an animal by contacting cells of the animal with a peptide aldehyde inhibitor of proteasome function or ubiquitin conjugation. Goldberg and Rock, WO 94/17816, report the use of proteasome inhibitors to inhibit MHC-I antigen presentation. Stein, et al., U.S. Pat. No. 5,693,617 report peptidyl aldehyde compounds as proteasome inhibitors useful for reducing the rate of degradation of protein in an animal. Inhibition of the 26S and 20S proteasome by indanone derivatives and a method for inhibiting cell proliferation using indanone derivatives are reported by Lum et al., U.S. Pat. No. 5,834,487. Alpha-ketoamide compounds useful for treating disorders mediated by 20S proteasome in mammals are reported in Wang et al., U.S. Pat. No. 6,075,150. France, et al., WO 00/64863, report the use of 2,4-diamino-3-hydroxycarboxylic acid derivatives as proteasome inhibitors. Carboxylic acid derivatives as proteasome inhibitors are reported by Yamaguchi et al., EP 1166781. Ditzel, et al., EP 0 995 757 report bivalent inhibitors of the proteasome. 2-Aminobenzylstatine derivatives that inhibit noncovalently the chymotrypsin-like activity of the 20S proteasome have been reported by Garcia-Echeverria, et al., Bioorg. Med. Chem. Lett., 2001, 11, 1317. [0012] Some further proteasome inhibitors can contain boron moieties. For example, Drexler et al., WO 00/64467, report a method of selectively inducing apoptosis in activated endothelial cells or leukemic cells having a high expression level of c-myc by using tetrapeptidic boronate containing proteasome inhibitors. Furet et al., WO 02/096933 report 2-[[N-(2-amino-3-(heteroaryl or aryl)propionyl)aminoacyl]amino]alkylboronic acids and esters for the therapeutic treatment of proliferative diseases in warm-blooded animals. U.S. Pat. Nos. 6,083,903; 6,297,217; 5,780454; 6,066,730; 6,297,217; 6,548,668; U.S. Patent Application Pub. No. 2002/0173488; and WO 96/13266 report boronic ester and acid compounds and a method for reducing the rate of degradation of proteins. A method for inhibiting viral replication using certain boronic acids and esters is also reported in U.S. Pat. No. 6,465,433 and WO 01/02424. Pharmaceutically acceptable compositions of boronic acids and novel boronic acid anhydrides and boronate ester compounds are reported by Plamondon, et al., U.S. Patent Application Pub. No. 2002/0188100. A series of di- and tripeptidyl boronic acids are shown to be inhibitors of 20S and 26S proteasome in Gardner, et al., Biochem. J., 2000, 346, 447.
[0013] Other boron-containing peptidyl and related compounds are reported in
U.S. Pat. Nos. 5,250,720; 5,242,904; 5,187,157; 5,159,060; 5,106,948; 4,963,655; 4,499,082; and WO 89/09225, WO/98/17679, WO 98/22496, WO 00/66557, WO 02/059130, WO 03/15706, WO 96/12499, WO 95/20603, WO 95/09838, WO 94/25051, WO 94/25049, WO 94/04653, WO 02/08187, EP 632026, and EP 354522. [0014] A great interest exists, as evidenced by the above references, in drugs which can modulate proteasome activity. For example, molecules capable of inhibiting proteasome activity can arrest or delay cancer progression by interfering with the ordered degradation of cell cycle proteins or tumor suppressors. Accordingly, there is an ongoing need for new and/or improved inhibitors of proteasome.
SUMMARY OF THE INVENTION
[0015] The present invention is directed to novel boronic acid and boronic ester compounds useful as proteasome inhibitors and modulation of apoptosis. The subject invention also comprises methods for inhibition of multicatalytic protease ("MCP") associated with certain disorders, including the treatment of muscle wasting disorders. [0016] In one embodiment are provided compounds having Formula (I):
Figure imgf000006_0001
(I) wherein constituent members are defined infra, as well as preferred constituent members.
[0017] In another embodiment the present invention provides a pharmaceutical composition comprising a compound of Formula (I) and a pharmaceutically acceptable carrier.
[0018] In another embodiment the present invention provides a method of inhibiting activity of proteasome comprising contacting a compound of Formula (I) with said proteasome.
[0019] In another embodiment the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to said cancer a therapeutically effective amount of a compound of Formula (I). [0020] In another embodiment the present invention provides a method of treating cancer treating cancer comprising administering to a mammal having or predisposed to said cancer a therapeutically effective amount of a compound of Formula (I), and wherein said cancer is selected from skin, prostate, colorectal, pancreas, kidney, ovary, mammary, liver, tongue, lung, and smooth muscle tissue. [0021] In another embodiment the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to said cancer a therapeutically effective amount of a compound of Formula (I), and wherein said cancer is selected from leukemia, lymphoma, non-Hodgkin lymphoma, myeloma, and multiple myeloma.
[0022] In another embodiment the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to said cancer a therapeutically effective amount of a compound of Formula (I) in combination with one or more antitumor or anticancer agent and/or radiotherapy.
[0023] In another embodiment the present invention provides a method of inhibiting activity of transcription factor NF-κB comprising contacting IκB, the inhibitor of transcription factor NF-κB, with a compound of Formula (I).
[0024] In another embodiment, the present invention provides a compound of
Formula (I) for use in therapy.
[0025] In another embodiment, the present invention provides use of a compound of Formula (I) for the manufacture of a medicament for the treatment of cancer.
[0026] In another embodiment, the present invention provides processes for preparing a compound of Formula (II):
Figure imgf000007_0001
(II) wherein constituent members are defined herein, by reacting a diol of Formula (Il-b):
Figure imgf000007_0002
(H-b) with an appropriate trialkoxyborane of Formula (II-a): R17Cv ^OR17
OR17 (II-a) wherein constituent members are defined herein; for a time and under conditions suitable for forming an intermediate of Formula (II-c):
Figure imgf000008_0001
(II-c) and reacting the intermediate of Formula (II-c) with either i) a reagent of formula R'CFfeMX1131, wherein M is a metal and XhaI is a halogen atom, or ii) a reagent of formula
Figure imgf000008_0002
, for a time and under conditions suitable for forming the compound of Formula (II).
[0027] These and other features of the compounds will be set forth in expanded form as the disclosure continues.
DESCRD7TION OF EMBODIMENTS OF THE INVENTION
[0028] The present invention provides, inter alia, compounds that can inhibit proteasome activity and be used for the treatment of diseases or disorders related to proteasome activity. Compounds of the invention include compounds of Formula (I)
Figure imgf000008_0003
or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein: R1 is Ci-Cg alkyl, C2-C8 alkenyl, C -C8 alkynyl, or C3-C7 cycloalkyl; R2 is H, -(CH2)aCH2NHC(=NR4)NH-Y, -(CH2)bCH2CONR5R6, - (CH2)cCH2N(R4)CONH2, -(CH2)dCH(R7)NR9R10, or -(CH2)eCH(R7)ZR8; a, b, and c are each, independently, 0, 1, 2, 3, 4, 5, or 6; d and e are each, independently, 0, 1, 2, 3, or 4;
R^s H or Q-Cio alkyl;
Rs and R6are each, independently, H, Ci-Cio alkyl, carbocyclyl, heterocarbocyclyl, or an amino protecting group; alternatively, R5 and R6 together with the N atom to which they are attached form a heterocarbocyclyl group;
R7 is H or Ci-Cio alkyl;
R8 is H, Ci-Cio alkyl, alkyl-S(=O)2-, aryI-S(=O)2-, H2NS(=O)2-, -SO3H, or a protecting group;
R9 is H, - o alkyl, carbocyclyl, or heterocarbocyclyl;
R10is H, Ci-Cio alkyl, carbocyclyl, heterocarbocyclyl, Ci-Cio alkyl-C(=O)-, C2-Cιo alkenyl-C(=0)-, C2-Cιo alkynyl-C(=O)-, carbocyclyl-C(=O)-, heterocarbocyclyl-C(=0)-, carbocyclylalkyl-C(=O)-, heterocarbocyclylalkyl-C(=O)-, Ci-Cio alkyl-S(=O)2-, carbocyclyl-S(=O)2-, heterocarbocyclyl-S(=0)2-, carbocyclylalkyl-S(=O)2-, heterocarbocyclylalkyl-S(=O)2-, CrC10 alkyl-NHC(=O)-, carbocyclyl-NHC(=0)-, heterocarbocyclyl-NHC(=0)-, carbocyclylalkyl-NHC(=0)-, heterocarbocycIylaIkyl-NF£C(=0)-, Ci-Cio alkyl-OC(=0)-, carbocyclyl-OC(=O)-, heterocarbocyclyl-OC(=O)-, carbocyclylalkyl-OC(=0)-, heterocarbocyclylalkyl-OC(=O)-, Ci-Cio alkyl-NH-C(=0)-NHS(=0)2-, carbocyclyl-NH-C(=0)-NHS(=O)2-, heterocarbocyclyl-NH-C(=0)-NHS(=0)2-, -Cio alkyl-S(=0)2-NH-C(=O)-, carbocyclyl-S(=O)2-NH-C(=0)-, heterocarbocyclyl-S(=O)2-NH-C(=0)-, or an amino protecting group; wherem R10 is optionally substituted with 1, 2 or 3, R23; alternatively, R9 and R10 together with the N atom to which they are attached form a heterocarbocyclyl group optionally substituted with 1, 2 or 3 R23;
Y is H, -CN, -NO2, -S(=O)2Rn, or a guanidino protecting group;
R11 is Ci-Cβ alkyl, aryl, or NR12R13;
R12 and R13 are, independently, H, Cι-C10 alkyl, carbocyclyl, heterocarbocyclyl, or an amino protecting group; alternatively, Ri2 and R13 together with the N atom to which they are attached form a heterocarbocyclyl group; Z is O, S, Se, or Te; Q is -B(OH)2, -B(OR14)2, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O; R14 is H, Cι-C4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl; X is RAC(=O)-, RANHC(=O)-, RAS(=O)2-, RAOC(=O)-, RASC(=0)-, or RA; RA is Cι-C2o alkyl optionally substituted with R20; C2-C2o alkenyl optionally substituted with R20; C2-C20 alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R21; or heterocarbocyclyl optionally substituted with 1-5 R21; R20 is selected from the group consisting of: -CN, halo, haloalkyl-, Ct-C4 alkyl, C2-C alkenyl, C2-C4 alkynyl, -CO2H, -C(=O)C02H, -C(=0)NH2, -C(=O)H, -S(=O)NH2, -S(=O)2NH2, -OH, -SH, -NH2, -NH(aIkyl), -N(alkyl)2, -NHC(=0)NH2, -NHC(=O)R20a, -NHC(=O)OR20a, -OR20a, -SR20a, -S(=O)R20a, -S(=O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(=O)2R20a, -NHR20b, phthalimido, -(0-alkyl)r-OH, -(0-alkyl)r-(0-alkyl), -OR20c, -SR20c, -O-alkyl-R20c, -S-alkyl-R20c, -S(=O)-R20c, -S(=O)2-R20c, -S(=O)2-NHR20c, -SC(O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR20c, carbocyclyl optionally substituted with 1-5 R21; and heterocarbocyclyl optionally substituted with 1-5 R21; R20a is Cι-C20 alkyl, C2-C20 alkenyl, or C2-C20 alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, OH, CN, -C4 alkyl, Cι-C4 alkoxy, C2-C8 alkoxyalkoxy, aryl, heteroaryl or -NHR20b; R20b is an amino protecting group; R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22; R21 is selected from the group consisting of: Cι-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, -OR21a, -SR21a, -CN, halo, haloalkyl, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)O-alkyl, -NHC(=O)alkyl, -COOH, -C(=0)O-alkyl, -C(=0)alkyl, -C(O)H, -S(=O)-alkyl, -S(=O)2-alkyl, -S(=O)-aryl, -S(=O)2-aryl, carbocyclyl optionally substituted with 1-5 R22, and heterocarbocyclyl optionally substituted with 1-5 R22;
R21a is H, Cι-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, carbocyclyl or heterocarbocyclyl;
R22 is selected from the group consisting of: -Cio alkyl, C2-C10 alkenyl, C2-Clo alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, aIkyI-OC(=0)-, alkyl-C(=0)-, aryl-OC(=0)-, alkyl-OC(=O)NH-, aryl-OC(=O)NH-, alkyl-C(=O)NH-, alkyl-C(=O)0-, (alkyl-0)r-alkyl, HO-(aIkyl-O)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=O)-, alkyl-S(=O)2-, H2NS(=O)-, and H2NS(=0)2-;
R23 is selected from the group consisting of: Ci-Cβ alkyl, C2-C6 alkenyl, C2-C6 alkynyl, F, CI, Br, I, haloalkyl, -NH2, -NHR23a, -N(R23a)2, -N3, -NO2, -CN, -CNO, -CNS, -C(=O)OR23a, -C(=O)R23a, -OC(=0)R23a, -N(R23a)C(=O)R 3a, -N(R23a)C(=0)OR23a, -C(=O)N(R23a)2, ureido, -OR23a, -SR23 , -S(=O)-(C C6 alkyl), -S(=O)2-(C1-C6 alkyl), -S(=0)-aryl, -S(=0)2-aryl, -S(=O)2-N(R 3a)2; carbocyclyl optionally substituted with 1-5 R24; and heterocarbocyclyl optionally substituted with 1-5 R24;
R^ is Hor -Ce alkyl; alternatively, two R23a may be combined, together with the N atom to which they are attached, to form a 5 to 7 membered heterocyclic group; and
R24 is selected from the group consisting of: Cι-C alkyl, C2-C4 alkenyl, C2-C4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=0)-, alkyl-C(=O)-, aryl-OC(=0)-, alkyl-OC(=0)NH-, aryl-OC(=0)NH-, alkyl-C(=0)NH-, alkyl-C(=O)0-, (alkyl-O)r-alkyl, HO-(alkyl-0)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=0)-, alkyl-S(-O)2-, H2NS(=O)-, and H2NS(=0)2-; and r is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; with the proviso that when Q is a 1,1,2,2-tetramethylethanediol boronic ester, then X is not aralkyloxycarbonyl; with the proviso that when Q is a 1,1,2,2-tetramethylethanediol boronic ester, and R1 is cycloalkyl, then R2 is not -CH2CONH2; and with the proviso that when X is RAC(=O)-, RA is a C4-Cιs straight-chained alkyl substituted with R20, and R20 is -CN, -CO2H, -C(=O)O-R20a, -NHS(=O)2R20a, - NHC(=O)R20a, -NHR20b, or phthalimido; then R2 is not - (CH2)aCH2NHC(=NR4)NH-Y, wherein Y is H, -CN, -NO2, or a guanidino protecting group. [0029] In further embodiments, when R2 is -(CH2)eCH(R7)ZR8, e is 0, R7 is H,
R8 is Ci-Cto alkyl and X is RAC(=O)-, then RA is not aminoalkyl-, alkylaminoalkyl-, dialkylaminoalkyl-, or ureidoalkyl-.
[0030] In some embodiments, R1 can be Cι-C4 alkyl, and in further embodiments, R1 can be propyl, such as 2-propyl.
[0031] In some embodiments, R2 can be -(CH2)aCH2NHC(=NR4)NH-Y,
-(CH2)bCH2CONR5R6, -(CH2)cCH2N(R4)CONH2, -(CH2)dCH(R7)NR9R10, or
-(CH2)eCH(R7)ZRs.
[0032] In some
Figure imgf000012_0001
and a is 1, 2
3, 4, or 5.
[0033] In some embodiments, R2 s -(CH2)aCH2NHC(=NR4)NH-Y and a is 2.
[0034] In some embodiments, R2 s -CH2CH2CH2NHC(=NR4)NH-Y.
[0035] In some embodiments, R2 s -(CH2)dCH(R7)NR9R10 and d is 0, 1, or 2.
[0036] In some embodiments, R2 s -(CH2)dCH(R7)NR9R10 and d is 0.
[0037] In some embodiments, R2 s -(CH2)dCH(R7)NR9R10 and R9 is H.
[0038] In some embodiments, R2 s -(CH2)dCH(R7)NR9R10.
[0039] In some embodiments, R2 s -CH(R7)NR9R10.
[0040] In some embodiments, R2 s -CH2NH-C(-O)OCH2(C6H5).
[0041] In some embodiments, R2 s -(CH2)eCH(R7)ZR8 and e is 0, 1, or 2.
[0042] In some embodiments, R2 s -(CH2)eCH(R7)ZR8 and e is 0.
[0043] In some embodiments, R2 s -(CH2)eCH(R7)ZR8.
[0044] In some embodiments, R2 s -CH(R7)ZR8.
[0045] In further embodiments, Z s O.
[0046] In further embodiments, Q has Formula (II-a):
Figure imgf000013_0001
(II-a) wherein D, R15a, R15b, R15c, R15d, p and q are defined herein below. [0047] In further embodiments, Q is B(OH)2 or a cyclic boronic ester wherein said cyclic boronic ester contains from 6 to 10 carbon atoms and contains at least one cycloalkyl moiety. [0048] In further embodiments Q is B(OH)2.
[0049] In further embodiments Q is pinanediol boronic ester.
[0050] In further embodiments Q is bicyclohexyl-l,l'-diol boronic ester.
[0051] In further embodiments, Q is l,2-dicyclohexyl-ethane-l,2-diol boronic ester.
[0052] Alternatively, in some embodiments, Q is -B(OH)2, -B(OR14)2,
Figure imgf000013_0002
wherein: R14, R15a, R15b, R15c, Rlsd , W, W1, p and q are as defined hereinbelow.
[0053] In further embodiments Q is:
Figure imgf000014_0001
W is a substituted or unsubstituted C4-Cιo cycloalkyl ring. [0054] In some embodiments, X is RAC(=O)-.
[0055] In some embodiments, X is RANHC(=0)-.
[0056] In some embodiments, X is RAS(O)2-.
[0057] In some embodiments, RA is Cι-C14 alkyl substituted by -(0-alkyl)r-OH or
-(O-alkyl)r(0-alkyl), wherein r is 1, 2, 3, 4, or 5.
[0058] In some embodiments, RA is -Cπ alkyl substituted by -(O-alkyl)r-OH or
-(O-alkyl)r-(O-alkyl), wherein r is 1, 2 or 3.
[0059] In some embodiments, RA comprises at least one -CH2CH2O- group.
[0060] In some embodiments, RA is -CH2(OCH2CH2)rOCH3.
[0061] In some embodiments, RA is -CH2OCH2CH OCH2CH2OCH3 or
-CH2OCH2CH2OCH3.
[0062] In some embodiments, RA is aryl or heteroaryl each optionally substituted with 1-5 R21.
[0063] In some embodiments, RA is cycloalkyl or heterocycloalkyl each optionally substituted with 1-5 R21.
[0064] In some embodiments, RA is Cι-C2o alkyl; C2-C20 alkenyl; or C2-C20 alkynyl, each optionally substituted with R20.
[0065] In some embodiments, RA is Cι-C2o alkyl; C2-C20 alkenyl; or C -C 0 alkynyl, each substituted with a carbocyclyl group or a heterocarbocyclyl group wherein said carbocyclyl group or heterocarbocyclyl group is optionally substituted with 1, 2 or 3 R21.
[0066] In some embodiments, RA is C1-C20 alkyl; C2-C20 alkenyl; or C2-C20 alkynyl, each substituted with an aryl group wherein said aryl group is optionally substituted with 1, 2 or 3 R21.
[0067] In some embodiments, RA is Cι-C2o alkyl; C2-C2o alkenyl; or C2-C20 alkynyl, each substituted with an heteroaryl group wherein said heteroaryl group is optionally substituted with 1, 2 or 3 R21. [0068] In some embodiments, RA is Cι-C2o alkyl; C2-C20 alkenyl; or C2-C20 alkynyl, each substituted with an cycloalkyl group wherein said cycloalkyl group is optionally substituted with 1, 2 or 3 R21.
[0069] In some embodiments, RA is C C2o alkyl; C -C20 alkenyl; or C2-C20 alkynyl, each substituted with an heterocycloalkyl group wherein said heterocycloalkyl group is optionally substituted with 1, 2 or 3 R21.
[0070] In some embodiments, RA is Cj-C2o alkyl; C2-C20 alkenyl; or C2-C20 alkynyl, each optionally substituted with R20, wherein R20 is selected from CN, halo, haloalkyl, -CO2H, -C(=O)C02H, -C(=0)NH2, -C(=0)H, -S(O)NH2, -S(O)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=0)NH2, -NHC(=O)R20a, -NHC(=O)OR20a, -OR20a, -SR20a, -S(O)R20a, -S(O)2R20a, -S(O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(O)2R20a, -NHR20b, phthalimido, -(O-alkyl), -(O-alkyl)r-OH, -(O-alkyl)r-(O-alkyl), -OR 0c, -SR20c, -O-alkyl-R20c, -S-alkyl-R20c, -S(O)-R20c, -S(O)2-R20c, -S(O)2-NHR20c, -SC(=O)R20c, -C(-O)R20c, -C(=O)OR20c, and -C(=O)NHR20c.
[0071] In some embodiments, R2 is H and X is (O-alkyl)-(O-alkyl)r-(C Cι4 alkyl)-C(=0)- or HO-(alkyl-O)r-(Cι-C14 alkyl)-C(=0)-. [0072] In some embodiments X is RAC(=0 and RA is C4-d6 alkyl.
[0073] In some embodiments X is RAC(=0)- and RA is aryl optionally substituted with 1-3 R21.
[0074] In some embodiments X is RAC(=0)- and RA is heterocarbocyclyl group optionally substituted with 1-3 R21.
[0075] In some embodiments X is RAC(=0)-; RA is phenyl substituted with one
R21; and R21 is phenoxy.
[0076] In some embodiments X is RAC(=0)-, RA is Cι-C4 alkyl substituted with
R20, and R20 is aryl optionally substituted with 1-3 R21; and in yet further embodiments aryl is substituted by at least one halo.
[0077] In some embodiments X is RAC(=0)-; RA is C Cι alkyl substituted with
R20; and R20 is -OR20a or-OR20c.
[0078] In some embodiments X is RAC(=0 ; RA is Cι-C14 alkyl substituted with
R20; and R20 is heterocarbocyclyl optionally substituted with 1-3 R21. [0079] In some embodiments X is RAS(O)2- and RA is C3-Cι6 alkyl. [0080] In some embodiments the present invention provides compounds of Formula (I) wherein the stereochemistry is of Formula (I-s):
Figure imgf000016_0001
(I-s) or pharmaceutically acceptable salt form thereof.
[0081] In some embodiments, the present invention provides compounds of Formula (I)
Figure imgf000016_0002
(I) or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein: R1 is d-Cg alkyl, C2-C8 alkenyl, C2-C8 alkynyl, or C3-C7 cycloalkyl; R2 is H, -(CH2)aCH2NHC(=NR4)NH-Y, -(CH2)bCH2CONR5R6, -(CH2)cCH2N(R4)CONH2, -(CH2)dCH(R7)NR9R10, or -(CH2)eCH(R7)ZR8; a, b, and c are each, independently, 0, 1, 2, 3, 4, 5, or 6; d and e are each, independently, 0, 1, 2, 3, or 4; R4 is H or Ci-do alkyl; R5 and R6 are each, independently, H, Ci-Qo alkyl, carbocyclyl, heterocarbocyclyl, or an amino protecting group; alternatively, R5 and R6 together with the N atom to which they are attached form a heterocarbocyclyl group; R7 is H or Ci-Cio alkyl; R8 is H, Ci-Cio alkyl, alkyl-S(=0)2-, aryl-S(=O)2-, H2NS(=O)2-, -SO3H, or a protecting group; R9 is H, Q-Cio alkyl, carbocyclyl, or heterocarbocyclyl; R10is H, Ci-Cio alkyl, carbocyclyl, heterocarbocyclyl, d-do alkyl-C(=O)-, carbocyclyl-C(=O)-, heterocarbocyclyl-C(=O)-, carbocyclylalkyl-C(=O)-, heterocarbocyclylalkyl-C(=O)-, - o alkyl-S(=O)2-, carbocyclyl-S(=O)2-, heterocarbocyclyl-S(=O)2-, carbocyclylalkyl-S(=O)2-, heterocarbocyclylalkyl-S(=O)2-, C Cι0 alkyl-NHC(=O)-, carbocyclyl-NHC(=O)-, heterocarbocyclyl-NHC(=O)-, carbocyclylalkyl-NHC(=0)-, heterocarbocyclylalkyl-NHC(=O)-, Crdo alkyl-OC(=0 , carbocyclyl-OC(=O)-, heterocarbocyclyl-OC(=O)-, carbocyclylalkyl-OC(=O)-, heterocarbocyclylalkyl-OC(=O)-, or an amino protecting group; wherein R10 is optionally substituted with 1, 2, or 3 R23; alternatively, R9 and R10 together with the N atom to which they are attached form a heterocarbocyclyl group;
Y is -H, -CN, -NO2, -S(=O)2Rn, or a guanidino protecting group;
R11 is Cι-C6 alkyl, aryl, or NR12R13;
R12 and R13 are, independently, H, Ci-Cio alkyl, carbocyclyl, heterocarbocyclyl, or an amino protecting group; alternatively, R12 and R13 together with the N atom to which they are attached form a heterocarbocyclyl group;
Z is O, S, Se, or Te;
Q is -B(OH)2, -B(OR14)2, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
R14 is H, d-C4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
X is RAC(=O)-, RANHC(=0)-, RAS(=O)2-, RAOC(=O)-, RASC(=O)-, or RA;
RA is Cι-C20 alkyl optionally substituted with R20; C2-C20 alkenyl optionally substituted with R20; C2-C20 alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R21; or heterocarbocyclyl optionally substituted with 1-5 R21;
R20 is selected from the group consisting of: -CN, halo, haloalkyl-, Cι-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, -C02H, -C(=0)CO2H, -C(=O)NH2, -C(=O)H, -S(=0)NH2, -S(=0)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=0)NH2, -NHC(=O)R 0a, -NHC(=O)OR20a, -OR20a, -SR20a, -S(=O)R20a, -S(=O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(=O)2R20a, -NHR20b, phthalimido, -(-O-alkyl)r, -O-alkyl-OH, -(O-alkyl)r-OH, -OR20c, -SR20c, -O-alkyl-R20c, -S-alkyl-R20c, -S(=O)-R20c, -S(=O)2-R20°, -S(=O)2-NHR20c, -SC(=O)R 0c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR20c, carbocyclyl optionally substituted with 1-5 R21; and heterocarbocyclyl optionally substituted with 1-5 R21;
R20a is Cι-C o alkyl, C -C20 alkenyl, or C2-C20 alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, d-C4 alkyl, aryl, heteroaryl or -NHR 0b;
R20b is an amino protecting group;
R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22;
R21 is selected from the group consisting of: d-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, Cι-C20 alkoxy, d-C20 thialkoxy, -OH, -CN, halo, haloalkyl, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)O-alkyl, -NHC(=0)alkyl, -C(=O)O-alkyl, -C(=O)alkyl, -S(=O)-alkyl, -S(=O)2-alkyl, -S(=O)-aryl, -S(=0)2-aryl, carbocyclyl optionally substituted with 1-5 R22; and heterocarbocyclyl optionally substituted with 1-5 R22;
R22 is selected from the group consisting of: Ci- o alkyl, C2-Cιo alkenyl, C2-Cιo alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=0)-, alkyl-C(=0)-, aryI-OC(=O)-, alkyl-OC(=O)NH-, aryl-OC(=0)NH-, alkyl-C(=O)NH-, alkyl-C(=O)0-, (alkyl-0)r-alkyl, HO-(alkyl-O)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=0)-, alkyl-S(=O)2-, H2NS(=O>, and H2NS(=O)2-;
R23 is selected from the group consisting of: d-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, F, CI, Br, I, haloalkyl, -NH2, -NHR23a, -N(R23a)2, -N3, -NO2, -CN, -CNO, -CNS, -C(=0)OR23a, -C(=0)R23a, -OC(=0)R23a, -N(R23a)C(=0) R23a, -C(=O)N(R23a)2, ureido, -OR23a, -SR23a, -S(=O)2-(d-C6 alkyl), -S(=O)2-aryl, and -S(=O)2-N(R23a)2; R23a is H or Cι-C6 alkyl; alternatively, two R23a may be combined, together with the N atom to which they are attached, to form a 5 to 7 membered heterocyclic group; and r is 2, 3, 4, 5, 6, 7, 8, 9, or 10; and with the proviso that when Q is a 1,1,2,2-tetramethylethanediol boronic ester, then X is not aralkyloxycarbonyl; with the proviso that when when Q is a 1,1,2,2-tetramethylethanediol boronic ester, and R1 is cycloalkyl, then R2 is not -CH2CONH2; and with the proviso that when X is RAC(=0)-, RA is a C4-Cι5 straight-chained alkyl substituted with R20, and R20 is -CN, -CO2H, -C(=O)O-R20a, -NHS(=O)2R20a, - NHC(=O)R 0a, -NHR20b, or phthalimido; then R2 is not - (CH2)aCH2NHC(=NR4)NH-Y, wherein Y is H, -CN, -NO2, or a guanidino protecting group. [0082] In some embodiments, R1 is 2-propyl; R2 is H, -
(CH2)aCH2NHC(=NR4)NH-Y, -(CH2)bCH2CONR5R6, -(CH2)cCH2N(R4)CONH2, - (CH2)dCH(R7)NR9R10, or -(CH2)eCH(R7)ZR8; Q is -B(OH)2 or pinanediol boronic ester; X is RAC(=O)-; and RA is C -Cι6 alkyl; aryl optionally substituted with 1-3 R21; or heterocarbocyclyl group optionally substituted with 1-3 R21. [0083] In some embodiments, the present invention provides compounds of
Formula (I)
Figure imgf000019_0001
(I) or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein:
R1 is d-Cg alkyl;
R2 is -(CH2)aCH2NHC(=NH)NH-Y, -(CH2)cCH2NHCONH2, -(CH2)dCH(R7)NR9R10, or -(CH2)eCH(R7)ZR8; a is 1, 2, 3, 4, or 5; c is 1, 2, 3, 4, or 5; d is O, 1, or 2; e is O, l, or 2;
R7 is H or methyl;
R8 is H, Ci-Cio alkyl, -S(=O)2-alkyl, -S(=O)2-aryl, -S(=O)2-NH2, -SO3H, or a protecting group; Y is -H, -CN, -N02, -S(=0) R!1, or a guanidino protecting group; R9 is H, d-do alkyl, carbocyclyl, or heterocarbocyclyl; R10is H, d-do alkyl, carbocyclyl, heterocarbocyclyl, Ci-do alkyl-C(=0)-, carbocyclyl-C(=O)-, heterocarbocyclyl-C(=O)-, carbocyclylalkyl-C(=O)-, heterocarbocyclylalkyl-C(=0)-, d-do alkyl-S(=O)2-, carbocyclyl-S(=O)2-, heterocarbocyclyl-S(=O)2-, carbocyclylalkyl-S(=O)2-, heterocarbocydylalkyl-S(=0)2-, Ci-Cio alkyl-NHC(=0)-, carbocyclyl-NHC(=O)-, heterocarbocyclyl-NHC(=0)-, carbocyclylalkyl-NHC(=0)-, heterocarbocyclylalkyl-NHC(=0)-, d-do alkyl-OC(=O)-, carbocyclyl-OC(=O)-, heterocarbocyclyl-OC(=O)-, carbocyclylalkyl-OC(=O)-, heterocarbocyclyIalkyl-OC(=O)-, or an amino protecting group; wherein R10 is optionally substituted with 1, 2 or 3 R23; alternatively, R9 and R10 together with the N atom to which they are attached form a heterocarbocyclyl group; R11 is Ci-Gs alkyl, aryl, or NR12R13; R12and R13are, independently, H, Ci-do alkyl, carbocyclyl, heterocarbocyclyl, or an amino protecting group; alternatively, R12 and R13 together with the N atom to which they are attached form a heterocarbocyclyl group; Z is O or S; Q is -B(OH)2, -B(OR14)2, or a cyclic boronic ester wherein said cyclic boronic ester contains from 6 to 20 carbon atoms and contains at least one cycloalkyl moiety; R14 is H, Cι-C4 alkyl, or cycloalkyl;
X is RAC(=O)-, RANHC(=O)-, RAS(=0)2-, RAOC(=O)-, RASC(=O)-, or RA; RA is d-C2o alkyl optionally substituted with R20; C2-C2o alkenyl optionally substituted with R20; C -C 0 alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R21; or heterocarbocyclyl optionally substituted with 1-5 R21; R20 is selected from the group consisting of: -CN, halo, haloalkyl-, d-C alkyl, C2-C4 alkenyl, C2-C4 alkynyl, -CO2H, -C(=O)CO2H, -C(=O)NH2, -C(=O)H, -S(=O)NH2, -S(=O)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=0)NH2, -NHC(=O)R20a, -NHC(=O)OR20a, -OR20a, -SR20a, -S(=O)R20a, -S(=O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(-O)2R20a, -NHR20b, phthalimido, -(O-alkyl)r, -O-alkyl-OH, -(0-alkyl)r-OH, -OR20c, -SR20c, -O-alkyl-R20c, -S-alkyl-R20c, -S(-O)-R20c, -S(=O)2-R20c, -S(=O)2-NHR20c, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR20c, carbocyclyl optionally substituted with 1-5 R21; and heterocarbocyclyl optionally substituted with 1-5 R21;
R 0a is d-C2o alkyl, C2-C2o alkenyl, or C2-C2o alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, C)-C4 alkyl, aryl, heteroaryl or -NHR20b;
R20 is an amino protecting group;
R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22;
R21 is selected from the group consisting of: Cι-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, Cι-C20 alkoxy, C C20 thialkoxy, -OH, -CN, halo, haloalkyl, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)O-alkyl, -NHC(=0)alkyl, -C(=O)O-alkyl, -C(=0)alkyl, -S(-0)-aIkyl, -S(=O)2-alkyl, -S(=O)-aryl, -S(=0)2-aryI, carbocyclyl optionally substituted with 1-5 R22, and heterocarbocyclyl optionally substituted with 1-5 R22;
R22 is selected from the group consisting of: Ci-do alkyl, C2-Cιo alkenyl, C2-Cιo alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=0 , alkyl-C(=O)-, aryl-OC(=0)-, alkyl-OC(=O)NH-, aryl-OC(=0)NH-, alkyl-C(=0)NH-, alkyl-C(=0)0-, (alkyl-O)r-alkyl, HO-(alkyl-0)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=0)-, alkyl-S(=0)2-, H2NS(=O)-, and H2NS(=O)2-;
R23 is selected from the group consisting of: Ci-Cβ alkyl, C2-C6 alkenyl, C2-C6 alkynyl, F, CI, Br, I, haloalkyl, -NH2, -NHR23a, -N(R23a)2, -N3, -N02, -CN, -CNO, -CNS, -C(=O)OR23a, -C(=O)R23a, -OC(=0)R23a, -N(R23a)C(=0) R23a, -C(=O)N(R23a)2, ureido, -OR23a, -SR23a, -S(=O)2-(d-C6 alkyl), -S(=0)2-aryl, and -S(=0)2-N(R23a)2;
R >2^3jaa . is H or d-C6 alkyl; alternatively, two R23a may be combined, together with the N atom to which they are attached, to form a 5 to 7 membered heterocyclic group; and r is 2, 3, 4, 5, 6, 1, 8, 9, or 10; with the proviso that when X is RAC(=O)-, RA is a C4-Cι5 straight-chained alkyl substituted with R ,2^0υ, and R >20υ is -CN, -CO2H, -C(=O)O-R 20uaa, -NHS(=O)2R >20a NHC(=O)R20a, -NHR20b, or phthalimido; then R2 is not - (CH2)aCH2NHC(=NR4)NH-Y, wherein Y is H, -CN, -NO2, or a guanidino protecting group.
[0084] In further embodiments, the present invention provides compounds of
Formula (I)
Figure imgf000022_0001
(I) or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein:
R1 is C1-C4 alkyl;
R2 is -(CH2)aCH2NHC(=NH)NH-Y, -(CH2)cCH2NHCONH2, or -(CH2)dCH(R7)NR9R10; a is 1, 2, or 3; c is 1, 2, or 3; d is O or 1;
R7 is H or methyl;
R9 is H or Ci-do alkyl;
R10 is H, Ci-do alkyl, or an amino protecting group;
Y is H, CN, orNO2; Q is -B(OH)2, pinanediol boronic ester, bicyclohexyl-l,l'-diol boronic ester, or 1,2- dicyclohexyl-ethane-l,2-diol boronic ester; X is RAC(=O)-, RANHC(=O , RAS(=O)2-, RAOC(=O)-, RASC(=O)-, or RA; RA is CpC2o alkyl optionally substituted with R20; C2-C o alkenyl optionally substituted with R20; C2-C20 alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R21; or heterocarbocyclyl optionally substituted with 1-5 R21; R20 is selected from the group consisting of: -CN, halo, haloalkyl-, Cι-C alkyl, C2-C4 alkenyl, C2-C4 alkynyl, -CO2H, -C(=0)CO2H, -C(=O)NH2, -C(=O)H, -S(=O)NH2, -S(=O)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)NH2, -NHC(=O)R20a, -NHC(=O)OR20a, -OR20a, -SR20a, -S(=O)R 0a, -S(=O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(=O)2R20a, -NHR20b, phthalimido, -(O-alkyl)r, -O-alkyl-OH, -(O-alkyl)r-OH, -OR20c, -SR20c, -O-alkyl-R20c, -S-aikyl-R20c 5 -S(=O)-R20c, -S(=O)2-R20c, -S(=O)2-NHR20c, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR 0c, carbocyclyl optionally substituted with 1-5 R21; and heterocarbocyclyl optionally substituted with 1-5 R21; R20a is Ci-C20 alkyl, C2-C2o alkenyl, or C2-C2o alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, C1-C4 alkyl, aryl, heteroaryl or -NHR20b; R20b is an amino protecting group; R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22; R21 is selected from the group consisting of: Cι-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, Cι-C20 alkoxy, C1-C20 thialkoxy, -OH -CN, halo, haloalkyl, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)0-alkyl, -NHC(=0)alkyl, -C(=0)O-alkyl, -C(=O)alkyl, -S(=0)-alkyl, -S(=O)2-alkyl, -S(=O)-aryl, -S(=0)2-aryl, carbocyclyl optionally substituted with 1-5 R22, and heterocarbocyclyl optionally substituted with 1-5 R22; R is selected from the group consisting of: d-do alkyl, C2-do alkenyl, C2-Cι0 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=0)-, alkyl-C(=0)-, aryl-OC(=0)-, alkyI-OC(=0)NH-, aryl-OC(=O)NH-, alkyl-C(=0)NH-, alkyl-C(=O)O-, (alkyl-O)r-alkyl, HO-(alkyl-O)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=O)-, alkyl-S(=O)2-, H2NS(=O)-, and H2NS(=O)2-; and r is 2, 3, 4, or 5; with the proviso that when X is RAC(=O)-, RA is a C4-Cι5 straight-chained alkyl substituted with R20, and R20 is -CN, -CO2H, -C(=O)O-R20a, -NHS(=O)2R20a, - NHC(=O)R 0a, -NHR 0 , or phthalimido; then R2 is not - (CH2)aCH2NHC(=NR4)NH-Y, wherein Y is H, -CN, or -NO2.
[0085] In yet further embodiments, the present invention provides compound of
Formula (I) or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein:
R1 is Cι-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or C3-C7 cycloalkyl;
R2 is -CH2NH or -CH2NR9R10;
R9 is H or Cj-Cio alkyl;
R10 is H, Ci-do alkyl, carbocyclyl, heterocarbocyclyl, d-Cio alkyl-C(=O)-, carbocyclyl-C(=0)-, heterocarbocyclyl-C(=O)-, carbocyclylalkyl-C(=O)-, heterocarbocyclylalkyl-C(=O)-, Ci-Cio alkyl-S(=0)2-, carbocyclyl-S(=O)2-, heterocarbocyclyl-S(=0)2-, carbocyclylalkyl-S(=0)2-, heterocarbocyclylalkyl-S(=O)2-, d-Cio alkyl-NHC(=O)-, carbocyclyl-NHC(=0)-, heterocarbocyclyl-NHC(=0)-, carbocyclylalkyl-NHC(=O)-, heterocarbocyclylalkyl-NHC(=0)-, Ci-Cio alkyl-OC(=O)-, carbocyclyl-OC(=O)-, heterocarbocyclyl-OC(=O)-, carbocyclylalkyl-OC(=O)-, heterocarbocyclylalkyl-OC(=O)-, or an amino protecting group; wherein R10 is optionally substituted with 1, 2 or 3, R23; alternatively, R9 and R10 together with the N atom to which they are attached form a heterocarbocyclyl group; Q is -B(OH)2, -B(OR14)2, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O; R14 is H, Cj-C4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl; X is RAC(=O)-, RANHC(=O)-, RAS(=0)2-, RAOC(=0)-, RASC(=O)-, or RA; RA is C1-C20 alkyl optionally substituted with R20; C2-C2o alkenyl optionally substituted with R20; C2-C2o alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R21; or heterocarbocyclyl optionally substituted with 1-5 R21; R20 is selected from the group consisting of: -CN, halo, haloalkyl-, d-C alkyl, C2-C alkenyl, C2-C alkynyl, -C02H, -C(=O)CO2H, -C(=0)NH2, -C(=O)H, -S(=O)NH2, -S(=O)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)NH2, -NHC(=O)R20a, -NHC(=O)OR 0a, -OR20a, -SR20a, -S(=O)R20a, -S(=O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20 , -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(=O)2R20a, -NHR20 , phthalimido, -(O-alkyl)r, -O-alkyl-OH, -(O-alkyl)r-OH, -OR20c, -SR20c, -O-alkyl-R20c, -S-alkyl-R20c, -S(=O)-R20c, -S(=O)2-R20°, -S(=O)2-NHR20c, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR20c, carbocyclyl optionally substituted with 1-5 R21; and heterocarbocyclyl optionally substituted with 1-5 R21; R20a is Cι-C2o alkyl, C2-C2o alkenyl, or C2-C2o alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, Cι-C alkyl, aryl, heteroaryl or -NHR20b; R20b is an amino protecting group; R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22; R21 is selected from the group consisting of: Cι-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, Cι-C20 alkoxy, Cι-C20 thialkoxy, -OH -CN, halo, haloalkyl, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=0)0-alkyl, -NHC(=O)alkyl, -C(=0)0-alkyl, -C(=O)alkyl, -S(=0)-alkyl, -S(=O)2-alkyl, -S(=0)-aryl, -S(=0)2-aryl, carbocyclyl optionally substituted with 1-5 R22, and heterocarbocyclyl optionally substituted with 1-5 R22;
R22 is selected from the group consisting of: Ci-Cjo alkyl, C2-Cι0 alkenyl, C2-Cι0 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=O)-, alkyl-C(=O)-, aryl-OC(=O)-, alkyl-OC(=O)NH-, aryl-OC(=O)NH-, alkyl-C(=O)NH-, alkyl-C(=O)0-, (alkyl-O)r-alkyl, HO-(alkyl-O)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=O)-, alkyl-S(=O)2-, H2NS(=O)-, and H2NS(=O)2-;
R23 is selected from the group consisting of: d-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, F, CI, Br, I, haloalkyl, -NH2, -NHR23a, -N(R23a)2, -N3, -NO2, -CN, -CNO, -CNS, -C(=O)OR23a, -C(=O)R23a, -OC(=0)R23a, -N(R23a)C(=O) R23a, -C(=O)N(R2 a)2, ureido, -OR23a, -SR23a, -S(=O)2-(d-C6 alkyl), -S(=O)2-aryl, and -S(=O)2-N(R23a)2;
R23a is H or Cι-C6 alkyl; , alternatively, two R23a may be combined, together with the N atom to which they are attached, to form a 5 to 7 membered heterocyclic group; and r is 2, 3, 4, or 5.
[0086] In yet further embodiments, the present invention provides compounds of
Formula (I) or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein:
R1 is Ci-Cg alkyl, C2-C8 alkenyl, C2-C8 alkynyl, or C3-C7 cycloalkyl;
R2 is H;
Q is -B(OH)2, -B(OR14)2, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O; R14 is H, Cι-C4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl; X is RAC(=O)-, RANHC(=O)-, RAS(=O)2-, RAOC(=O)-, RASC(=O)-, or RA; RA is Cι-C2o alkyl optionally substituted with R20; C2-C2o alkenyl optionally substituted with R20; C2-C2o alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22; R20 is selected from the group consisting of: -OR20a, -SR20a, -S(=O)R20a, -S(=O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, phthalimido, -(0-alkyI)r, -O-alkyl-OH, -(O-alkyl)r-OH, -OR20c, -SR20c, -O-alkyl-R 0c, -S-alkyl-R20c, -S(=O)-R20c, -S(=O)2-R20c, -S(=O)2-NHR20c, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR20c, carbocyclyl optionally substituted with 1-5 R22; and heterocarbocyclyl optionally substituted with 1-5 R22; R20a is d-C20 alkyl, C2-C2o alkenyl, or C2-C2o alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, Cι-C4 alkyl, aryl, heteroaryl or -NHR20b; R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22; R22 is selected from the group consisting of: Ci-do alkyl, C2-Cιo alkenyl, C2-Cιo alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=O)-, alkyl-C(=0)-, aryl-OC(=0)-, alkyl-OC(=0)NH-, aryl-OC(=O)NH-, alkyl-C(=O)NH-, alkyl-C(=0)O-, (alkyl-O)r-alkyl, HO-(alkyl-O)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=O)-, alkyI-S(=0)2-, H2NS(=0)-, and H2NS(=O)2-; and r is 2, 3, 4, 5, 6, 7, 8, 9, or 10. [0087] In yet further embodiments:
X is RAC(=O)-, RANHC(=O)-, RAS(O)2-, or RA; RA is d-Cπ alkyl optionally substituted with R20; R20 is -(O-alkyl)r-OH or -(O-alkyl)r-(O-alkyl); and r is 1, 2, 3, 4, or1 5. In further embodiments, the O-alkyl is methoxy, ethoxy, or propoxy.
[0088] In yet further embodiments, the present invention provides compounds of
Formula (I) or pharmaceutically acceptable salts, stereoisomeric or tautomeric forms thereof, wherein: R1 is 2-propyl;
R2 is -CH2CH2CH2NHC(=NH)NH-NO2, -CH2CH2CH2NHC(=O)NH2, -CH(CH3)OH, -CH2CONH2, -CH2NH2, or -CH2NR9R10; R9 is H;
R10 is methyl-C(=O)-, ethyl-C(=O)-, propyl-C(=O)-, butyl-C(=O)-, pentyl-C(-O)-, 2-(ethoxycarbonyl)ethyl-C(=O)-, 4-methyl-phenyl-C(=O)-, cyclopropyl-C(=O)-, 4-fluoro-phenyl-C(=O)-, 4-H2NS02-phenyl-C(=O)-, 4-H3CS02-phenyl-C(=O)-, 4-phenyl-phenyl-C(=0)-, 3,4-dimethoxy-benzyl-C(=O)-, 3-pyridinyl-C(=0)-, 2-(hydroxy)-pyridin-3-yl-C(=0)-, 6-(morpholino)-pyridin-3-yl-C(=0)-, 2-(pyridin-4-yl)thiazol-4-yl-C(=0)-, 2-pyrazinyl-C(=O)-, 2,5-dimethyl-pyrazolyl-C(=0)-, N-methyl-2-pyrrolyl-C(=O)-, 2-pyrroIidinyl-C(=0)-, 2-thiophenyl-C(=O)-, 5-isoxazolyl-C(=0)-, 4-(tetrazol-5-yl) phenyl-C(=0)-, (5-tetrazolyl)CH2-C(=O)-, N-H3CSO2-piperidinyl-C(=0)-, butyl-OC(=O)-, (benzyl)-OC(=O)-, (9-fluorenylmethyl)-OC(=0)-, pentyl-NHC(=0 , propyl-NHC(=O)-, phenyl-NHC(=O)-, 4-methyI-phenyl-NHC(=O)-, methyl-S(=0)2-, 4-fluoro-phenyl-S(=0)2-, 4-cyano-phenyl-S(=O)2-, 1 -methyl-imidazol-4-yl-S(=0)2-, 2-thiophenyl-S(=O)2-, (4-methyl-phenyl)-NHC(=0)NH-S(=O)2-, and (4-methyl-phenyl)-S(=O)2NHC(=0)-, alternatively, R9 and R10 together with the N atom to which they are attached form pyrrolyl or pyrazolyl;
Q is -B(OH)2, pinanediol boronic ester, bicyclohexyl-l,l'-diol boronic ester, or 1,2- dicyclohexyl-ethane-l,2-diol boronic ester;
X is RAC(=O)-, RANHC(=0)-, RAS(=0)2-, or RAOC(=O)-;
R is CH3-, C2H5-, C3H7-, C H -, C5H11-, C63-, C75-, C87-, C99-, Cι0H2r, CπH23-, Cι2H25-, CI3H27-, adamantyl-, bicycloheptanyl-, Cι-3 alkyl substituted with R20; C2-10 alkenyl substituted with R20; cyclopropyl substituted with 0-3 R21; cyclopentyl substituted with 0-2 R21; cyclohexyl substituted with 0-2 R21; phenyl substituted with 0-3 R21; naphthyl- substituted with 0-2 R21; pyrazinyl substituted with 0-1 R21; quinolinyl substituted with 0-1 R21; imidazolyl substituted with 0-1 R21; tetrahydrofuranyl substituted with 0-1 R21; oxothiazolidinyl substituted with 0-1 R21; benzothiazolyl substituted with 0-1 R21; thiazolyl substituted with 0-2 R21; furanyl substituted with 0-2 R21; pyrrolidinyl substituted with 0-1 R21; piperidinyl substituted with 0-1 R21; piperazinyl substituted with 0-1 R21; or pyridinyl substituted with 0-1 R21;
R 20 is selected from the group co 'nsisting of: hydroxy-, methoxy-, ethoxy-, propoxy-, butoxy-, pentoxy-, hexyloxy-, heptyloxy-, octyloxy-, methoxyethoxy-, methoxyethoxyethoxy-, methyl-S-, ethyl-S-, octyl-S-, methyl-C(=O)S-, (acetylamino)methyl-S-, amino-, methylamino-, dimethylamino-, methyl-C(=O)-, phenyl-C(=0)-, (H3CS02)phenyl-C(=0)-, thiophenyl-C(=O)-, methyl-OC(=O)-, ethyl-OC(=0)-, butyl-OC(=0)NH-, methyl-C(0)NH-, methoxyethoxy-methyl-C(=0)NH~, H2NC(=0)-, methyl-NHC(=O)-, ethyl-NHC(=0)-, propyl-NHC(=0)-, phenyl-NHC(=O)-, H2NC(=0)NH-, H2NS(=O)2-, octyl-S(=0)2-, phenyl-S(=O)2-, methylphenyl-S(=0)2-, thiophenyl-S(=0)2-, cyclopentyl-, cyclohexyl-, cycloheptyl-, adamantyl-, bicycloheptanyl-, cyclopentenyl-, phenyl-, methoxy-phenyl-, methyl-phenyl-, dimethyl-phenyl-, ethyl-phenyl-, propyl-phenyl-, butyl-phenyl-, fluoro-phenyl-, difluoro-phenyl-, chloro-phenyl-, bromo-phenyl-, iodo-phenyl-, dimethylamino-phenyl-, cyclohexyloxy-, 2-isopropyl-5-methyl-cyclohexyloxy-, naphthyl-, methoxynaphthyl-, naphthyloxy-, phenoxy-, (methyl-phenyl)oxy-, (ethyl-phenyl)oxy-, (propyl-phenyl)oxy-, (butyl-phenyl)oxy-, (fluoro-phenyl)oxy-, (chloro-phenyl)oxy-, (bromo-phenyl)oxy-, naphthyl-S-, benzyl-S-, ( ethyl-phenyl)methyl-S-, pyrimidinyl-S-, piperidinyl-, N-methyl-piperidinyl-, N-propyl-piperidinyl-, phthalimido-, thiophenyl-, methyl-thiophenyl-, imidazolyl-, furnayl-, tetrazolyl-, oxopyrrolidinyl-, indolyl-, and methyl-indolyl-; and
R21 is selected from the group consisting of: methyl-, ethyl-, propyl-, butyl-, pentyl-, hexyl-, heptyl-, ethenyl-, propenyl-, butenyl-, methoxy-, ethoxy-, propoxy-, phenoxy-, fluoro-, chloro-, bromo-, methyI-C(=O)-, butyl-OC(=0)-, butyl-OC(=O)NH-, phenyl-, methoxyphenyl-, fluorophenyl-, chlorophenyl-, bromophenyl-, pyrrolyl-, and pyridinyl-.
[0089] It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.
[0090] As used herein, the phrase "boronic acid" refers to a compound containing a B(OH)2 moiety. In some embodiments, boronic acid compounds can form oligomeric anhydrides by dehydration of the boronic moiety. For example, Snyder, et al., J. Am. Chem. Soc, 1958, SO, 3611 report oligomeric arylboronic acids. Thus, unless otherwise indicated, "boronic acid", or a chemical formula containing a -B(OH)2 moiety, is intended to encompass free boronic acids, oligomeric anhydrides, including but not limited to, dimers, trimers, tetramers, and mixtures thereof. [0091] As used herein, "boronic acid anhydride" or "boronic anhydride" refers to a compound formed by the combination of two or more molecules of a boronic acid compound of Formula (I), with loss of one or more water molecules from the boronic acid moieties. When contacted with water, the boronic acid anhydride compound can be hydrated to release free boronic acid compound. In some embodiments, the boronic acid anhydride structure can contain two, three, four, or more boronic acid units and can have a cyclic or linear configuration. In some embodiments, the boronic acid anhydride compound exists substantially in a single oligomeric form; however, boronic acid anhydrides also encompass mixtures of different oligomeric boronic acid anhydride as well as free boronic acids.
[0092] Non-limiting examples of boronic acid anhydrides of the invention include compounds of Formula (II) and (III) where G is a moiety of Formula (IV) and t is O to 10 or 1, 2, 3, or 4.
Figure imgf000031_0001
(IV) [0093] In some embodiments, at least about 80% of boronic acid present in a boronic acid anhydride compound exists in a single oligomeric anhydride form. In further embodiments, at least about 85, about 90, about 95, or about 99 % of the boronic acid present in the boronic acid anhydride exists in a single oligomeric anhydride form. In some embodiments, the boronic acid anhydride compound consists essentially of a single oligomeric boronic acid anhydride. In yet further embodiments, the boronic acid anhydride compound consists of a single oligomeric boronic acid anhydride. In further embodiments, the boronic acid anhydride compound contains a boroxine of Formula (III), wherein t is 1.
[0094] Boronic acid anhydride compounds can be prepared from the corresponding boronic acid compound by exposure to dehydrating conditions, including, for example, crystallization, lyophilization, exposure to heat, and/or exposure to a drying agent. Some suitable crystallization solvents include ethyl acetate, dichloromethane, hexanes, ether, benzene, acetonitrile, ethanol, and mixtures thereof. [0095] As used herein, the phrase "boronic ester" or "boronic acid ester" refers to an ester derivative of a boronic acid compound. As used herein, "cyclic boronic ester" is intended to mean a stable cyclic boronic moiety of general formula -B(OR)(OR) wherein the two R substituents are linked together forming a cyclic moiety (e.g., 3- to 10-membered cycloalkyl group) optionally further substituted with one or more substituents or fused with (sharing at least one bond) one or more further carbocyclyl or heterocarbocyclyl groups. The cyclic boronic ester can contain from 2 to 20 carbon atoms, and optionally, a heteroatom which can be N, S, or O. Cyclic boronic esters are well known in the art. Examples of cyclic boronic esters include, but are not limited to, pinanediol boronic ester, pinacol boronic ester, 1,2-ethanediol boronic ester, 1,3- propanediol boronic ester, 1,2-ρropanediol boronic ester, 2,3-butanediol boronic ester, 1,1,2,2-tetramethylethanediol boronic ester, 1,2-diisopropylethanediol boronic ester, 5,6-decanediol boronic ester, 1,2-dicyclohexylethanediol boronic ester, bicyclohexyl- l,l'-diol, diethanolamine boronic ester, and 1,2-diphenyl- 1,2-ethanediol boronic ester. [0096] In some embodiments, the "cyclic boronic ester" has Formula (II-a):
Figure imgf000032_0001
(II-a) wherein: D is absent, O, S, NR16, or CRi5eR15f; R15a, R15b, R15c, R15d, R15e, R15f are each, independently, H, d-Cl0 alkyl, C3-C7 cycloalkyl, aryl or heteroaryl, wherein said d-Cio alkyl, d-do cycloalkyl, aryl or heteroaryl are each optionally substituted by 1, 2, 3 or 4 halo, Cj-C4 alkyl, Cι-C4 alkoxy, Cι-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; or R15a and R15b together with the C atoms to which they are attached form C3- Cio cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C1-C4 alkyl, d-C4 alkoxy, C1-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; or RISc and R15d together with the C atoms to which they are attached form C3- C10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C C4 alkyl, d-Q alkoxy, Cι-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; or R15b and R15c together with the C atoms to which they are attached and the intevening D moiety form aryl, heteroaryl, C3-C10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, d-C4 alkyl, d- C4 alkoxy, Cι-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; R16 is H or Ci-Cδ alkyl; and p and q are each, independently, 1, 2 or 3. [0097] In some embodiments, D is absent.
[0098] In some embodiments, D is NR16.
[0099] In some embodiments, D is NH.
[00100] In some embodiments, D is CH2.
[00101] In some embodiments, R15a and RI5b together with the C atoms to which they are attached form C3-do cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, d-C4 alkyl, d-C4 alkoxy, Cι-C haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; and R15c and R15d together with the C atoms to which they are attached form C3-C10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, Cι-C alkyl, d-C4 alkoxy, Cι-C haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl.
[00102] In some embodiments, RI5a and RI5b together with the C atoms to which they are attached form cyclopropyl, cyclobutyl, cyclopenytyl, cyclohexyl or cycloheptyl; and R15c and R15d together with the C atoms to which they are attached form cyclopropyl, cyclobutyl, cyclopenytyl, cyclohexyl or cycloheptyl. [00103] In some embodiments, D is absent and R,5b and R15c together with the C atoms to which they are attached form aryl, heteroaryl, C3-Cιo cycloalkyl or a 3- to 10- membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, Ci- C4 alkyl, Cι-C alkoxy, Cι-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl.
[00104] In some embodiments, D is absent and R15b and R15c together with the C atoms to which they are attached form C3-C10 cycloalkyl optionally substituted by 1, 2, 3 or 4 halo, Cι-C alkyl, Cι-C4 alkoxy, Cι-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl. [00105] In some embodiments, D is absent and R15b and R15c together with the C atoms to which they are attached form C3-C10 cycloalkyl optionally substituted by 1, 2,
3 or 4 halo or C1-C4 alkyl.
[00106] In some embodiments, D is absent and R15b and R15c together with the C atoms to which they are attached form a C7-Cιo bicyclic cycloalkyl group optionally substituted by 1, 2, 3 or 4 halo or d-C4 alkyl.
[00107] In some embodiments, p and q are each 1.
[00108] In some embodiments, at least one of R15 , R15b, R150, R15d is other than
H.
[00109] Further examples of "cyclic boronic esters", as defined herein, include, boronic esters with the following structures:
Figure imgf000034_0001
wherein: W is a substituted or unsubstituted C4-C1o cycloalkyl ring or a substituted or unsubstituted phenyl ring; W1 is, independently at each occurrence, a substituted or unsubstituted C3-C6 cycloalkyl ring. Groups R15a, R15b, R15c, R15d, R15e, R15f, p and q are, defined as provided above.
[00110] As used herein, the term "alkyl" or "alkylene" is meant to refer to a saturated hydrocarbon group which is straight-chained or branched. Example alkyl groups include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, s-butyl, t-butyl), pentyl (e.g., n-pentyl, isopentyl, neopentyl) and the like. An alkyl group can contain from 1 to about 20, from 2 to about 20, from 1 to about 10, from 1 to about 8, from 1 to about 6, from 1 to about 4, or from 1 to about 3 carbon atoms. [00111] As used herein, "alkenyl" refers to an alkyl group having one or more double carbon-carbon bonds. Example alkenyl groups include ethenyl, propenyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl, and the like. [00112] As used herein, "alkynyl" refers to an alkyl group having one or more triple carbon-carbon bonds. Example alkynyl groups include ethynyl, propynyl, butynyl, pentynyl, and the like.
[00113] As used herein, "haloalkyl" refers to an alkyl group having one or more halogen substituents. Example haloalkyl groups include CF3, C2F5, CHF2, CC13, CHC12, C2Cl5, and the like. An alkyl group in which all of the hydrogen atoms are replaced with halogen atoms can be referred to as "perhaloalkyl." Examples perhaloalkyl groups include CF3 and C2F5.
[00114] As used herein, "carbocyclyl" groups are saturated (i.e., containing no double or triple bonds) or unsaturated (i.e., containing one or more double or triple bonds) cyclic hydrocarbon moieties. Carbocyclyl groups can be mono- or polycyclic. Example carbocyclyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, 1, 3-cyclopentadienyl, cyclohexenyl, norbornyl, norpinyl, norcarnyl, adamantyl, phenyl, and the like. Carbocyclyl groups can be aromatic (e.g., "aryl") or non-aromatic (e.g., "cycloalkyl"). In some embodiments, carbocyclyl groups can have from 3 to about 20, 3 to about 10, or 3 to about 7 carbon atoms. [00115] As used herein, "aryl" refers to aromatic carbocyclyl groups including monocyclic or polycyclic aromatic hydrocarbons such as, for example, phenyl, naphthyl, anthracenyl, phenanthrenyl, indanyl, indenyl, and the like. In some embodiments, aryl groups have from 6 to about 18 ring-forming carbon atoms. [00116] As used herein, "cycloalkyl" refers to non-aromatic carbocyclyl groups including cyelized alkyl, alkenyl, and alkynyl groups. Cycloalkyl groups can include bi- or poly-cyclic ring systems. Example cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptatrienyl, norbornyl, norpinyl, norcarnyl, adamantyl, and the like. Also included in the definition of cycloalkyl are moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the cycloalkyl ring, for example, benzo derivatives of cyclopentane (indanyl), cyclohexane (tetrahydronaphthyl), and the like. In some embodiments, cycloalkyl groups can have 3, 4, 5, 6, or 7 ring forming carbon atoms. In some embodiments, cycloalkyl groups can have 0, 1, or 2 double or triple ring-forming bonds.
[00117] As used herein, "heterocarbocyclyl" groups can be saturated or unsaturated carbocyclyl groups wherein one or more of the ring-forming carbon atoms of the carbocyclyl group is replaced with a heteroatom such as O, S, or N. Heterocarbocyclyl groups can be aromatic (e.g., "heteroaryl") or non-aromatic (e.g., "heterocycloalkyl"). Heterocarbocyclyl groups can correspond to hydrogenated and partially hydrogenated heteroaryl groups. Heterocarbocyclyl groups can contain, in addition to at least one heteroatom, from about 1 to about 20, about 2 to about 10, or about 2 to about 7 carbon atoms and can be attached through a carbon atom or heteroatom. Examples of heterocarbocyclyl groups include morpholino, thiomorpholino, piperazinyl, tetrahydrofuranyl, tetrahydrothienyl, 2,3-dihydrobenzofuryl, 1,3- benzodioxole, benzo- 1,4-dioxane, piperidinyl, pyrrolidinyl, isoxazolidinyl, isothiazolidinyl, pyrazolidinyl, oxazolidinyl, thiazolidinyl, imidazolidinyl, and the like. [00118] As used herein, "heteroaryl" groups are aromatic heterocarbocyclyl groups and include monocyclic and polycyclic aromatic hydrocarbons that have at least one heteroatom ring member such as sulfur, oxygen, or nitrogen. Heteroaryl groups include, without limitation, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrrolyl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl indazolyl, 1,2,4-thiadiazolyl, isothiazolyl, benzothienyl, purinyl, carbazolyl, benzimidazolyl, and the like. In some embodiments, heteroaryl groups can have from 3 to about 20 ring-forming carbon atoms, and in further embodiments from about 3 to about 12 ring forming carbon atoms. In some embodiments, heteroaryl groups have 1 to about 4, 1 to about 3, or 1 to 2 heteroatoms.
[00119] As used herein, "heterocycloalkyl" refers to a non-aromatic heterocarbocyclyl group including cyelized alkyl, alkenyl, and alkynyl groups where one or more of the ring-forming carbon atoms is replaced by a heteroatom such as an O, N, or S atom. Ring-forming carbon and heteroatoms such as S and N can further be oxidized in a heterocycloalkyl moeity. For example, the ring-forming carbon or heteroatom can bear one or two oxo or sufido moieties (e.g., >C=0, >S=0, >S(=O)2, N→O, etc.). Also included in the definition of heterocycloalkyl are moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the nonaromatic heterocyclic ring, for example phthalimidyl, naphthalimidyl pyromellitic diimidyl, phthalanyl, and benzo derivatives of saturated heterocycles such as indolene and isoindolene groups. In some embodiments, heterocycloalkyl groups have 3 to about
20 ring-forming atoms. In some embodiments, heterocycloalkyl groups have 3, 4, 5, 6, or 7 ring-forming atoms. In some embodiments, heterocycloalkyl groups have 0, 1, or 2 double or triple ring-forming bonds.
[00120] As used herein, "halo" or "halogen" includes fluoro, chloro, bromo, and iodo.
[00121] As used herein, "alkoxy" refers to an -O-alkyl group. Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the like. In some embodiments, alkoxy groups have from 1 to 20, 1 to 12, 1 to 8, 1 to 6, 1 to 4 or 1 to3 carbon atoms.
[00122] As used herein, "alkoxyalkoxy" refers to an -O-alkyl-O-alkyl group.
[00123] As used herein, "thioalkoxy" refers to an alkoxy group in which the O atom is replaced by an S atom.
[00124] As used herein, "aryloxy" refers to an -O-aryl group. An example aryloxy group is phenoxy.
[00125] As used herein, "thioaryloxy" refers to an aryloxy group in which the O atom is replaced by an S atom.
[00126] As used herein, "aralkyl" refers to an alkyl moiety substituted by an aryl group. Example aralkyl groups include benzyl and naphthylmethyl groups. In some embodiments, aralkyl groups have from 7 to 11 carbon atoms.
[00127] As used herein, "amino" refers to an -NH2 group. "Alkylamino" refers to an amino group substituted by an alkyl group and "dialkylamino" refers to an amino group substituted by two alkyl groups. On the contrary, "aminoalkyl" refers to an alkyl group substituted by an amino group.
[00128] As used herein, "carbonyl" refers to >C=O.
[00129] As used herein, "carboxy" or "carboxyl" refers to -COOH.
[00130] As used herein, "hydroxy" refers to -OH.
[00131] As used herein, "mercapto" refers to -SH.
[00132] As used herein, "ureido" refers to -NHCONH2.
[00133] As used herein, "sulfinyl" refers to >SO.
[00134] As used herein, "sulfonyl" refers to >SO2. [00135] As used herein, "oxy" refers to -O-.
[00136] The above chemical terms can be combined to refer to moieties containing a combination of chemical groups. This combination term is generally read such that a recited term is understood to be a substituent of a following term. For example, "alkylcarbonylalkenyl" refers to an alkenyl group substituted by a carbonyl group which in turn is substituted by an alkyl group. The following terms can also exemplify such combinations.
[00137] As used herein, "carbocyclylalkyl" refers to an alkyl moiety substituted by a carbocyclyl group. Example carbocyclylalkyl groups include "aralkyl" (alkyl substituted by aryl) and "cycloalkylalkyl" (alkyl substituted by cycloalkyl).
[00138] As used herein, "carbocyclylalkenyl" refers to an alkenyl moiety substituted by a carbocyclyl group. Example carbocyclylalkenyl groups include
"aralkenyl" (alkenyl substituted by aryl) and "cycloalkylalkenyl" (alkenyl substituted by cycloalkyl).
[00139] As used herein, "carbocyclylalkynyl" refers to an alkynyl moiety substituted by a carbocyclyl group. Example carbocyclylalkynyl groups include
"aralkynyl" (alkynyl substituted by aryl) and "cycloalkylalkynyl" (alkynyl substituted by cycloalkyl).
[00140] As used herein, "heterocarbocyclylalkyl" refers to an alkyl moiety substituted by a heterocarbocyclyl group. Example heterocarbocyclylalkyl groups include "heteroarylalkyl" (alkyl substituted by heteroaryl) and "heterocycloalkylalkyl"
(alkyl substituted by heterocycloalkyl).
[00141] As used herein, "heterocarbocyclylalkenyl" refers to an alkenyl moiety substituted by a heterocarbocyclyl group. Example heterocarbocyclylalkenyl groups include "heteroarylalkenyl" (alkenyl substituted by heteroaryl) and
"heterocycloalkylalkenyl" (alkenyl substituted by heterocycloalkyl).
[00142] As used herein, "heterocarbocyclylalkynyl" refers to an alkynyl moiety substituted by a heterocarbocyclyl group. Example heterocarbocyclylalkynyl groups include "heteroarylalkynyl" (alkynyl substituted by heteroaryl) and
"heterocycloalkynylalkyl" (alkynyl substituted by heterocycloalkyl).
[00143] As used herein, the phrase "protecting group" refers to a chemical functional group that can be selectively appended to and removed from functionalities, such as hydroxyl groups, amino groups, and carboxyl groups. Protecting groups are usually introduced into a chemical compound to render such functionality inert to chemical reaction conditions to which the compound is exposed. Any of a variety of protecting groups can be employed with the present invention. A protecting group of an amino moiety can be referred to as an "amino protecting group" and a protecting group of a guanidino moiety can be referred to as a "guanidino protecting group." Amino and guanidino protecting groups can have the formulas aryl-SO2-, alkyl-SO2-, aryl-C(=0)-, aralkyl-C(0)-, alkyl-C(=0)-, aryl-OC(=O)-, aralkyl-OC(=O)-, alkyl-OC(=O)-, aryl- NHC(=O)-, alkyl-NHC(=0)-, and the like, wherein said alkyl, aryl and aralkyl groups may be substituted or unsubstituted. Example amino and guanidino protecting groups can also include t-butyloxycarbonyl (BOC), fluorenylmethoxycarbonyl (Fmoc), benzyloxycarbonyl (Cbz), and a phthalimido group. Other protecting groups include the following moieties:
Figure imgf000039_0001
MTR MTS
Figure imgf000039_0002
PMC and TOS
Further representative protecting groups can be found in T.W. Green and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3rd. Ed., Wiley & Sons, Inc., New York (1999), which is incorporated, herein by reference in its entirety. [00144] As used herein, "substituted" indicates that at least one hydrogen atom of a chemical group is replaced by a non-hydrogen moiety. Example substituents include F, CI, Br, I, d-C6 alkyl, d-C6 alkenyl, d-C6, alkynyl, haloalkyl, NRERF, N3, NO2, CN, CNO, CNS, C(=O)ORE, RECO, REC(=O)O, RECONRE, RERFNCO, ureido, ORE, SRE, SO2-alkyl, S02-aryl, and S02-NRERF, wherein RE and RF are each, independently, H or Cι-C6 alkyl. Alternatively, RE and RF may be combined, with the nitrogen to which they are attached, to form a 5 to 7 membered heterocyclic ring, for example pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, and N-methylpiperazinyl. When a chemical group herein is "substituted" it may have up to the full valance of substitution, provided the resulting compound is a stable compound or stable structure; for example, a methyl group may be substituted by 1, 2, or 3 substituents, a methylene group may be substituted by 1 or 2 substituents, a phenyl group may be substituted by 1, 2, 3, 4, or 5 substituents, and the like.
[00145] As used herein, "leaving group" refers to any group that can be replaced by a nucleophile upon nucleophilic substitution. Example leaving groups include, halo (F, CI, Br, I), hydroxyl, alkoxy, mercapto, thioalkoxy, triflate, alkylsulfonyl, substituted alkylsulfonate, arylsulfonate, substituted arylsulfonate, heterocyclosulfonate or trichloroacetimidate. Representative examples include p-(2,4- dinitroaniIino)benzenesulfonate, benzenesulfonate, methylsulfonate, p- methylbenzenesulfonate, p-bromobenzenesulfonate, trichloroacetimidate, acyloxy, 2,2,2-trifluoroethanesulfonate, imidazolesulfonyl and 2,4,6-trichlorophenyl. [00146] As used herein "stable compound" or "stable structure" refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and preferably capable of formulation into an efficacious therapeutic agent. The present invention is directed only to stable compounds. [00147] The compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present invention that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. Cis and trans geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms.
[00148] In addition to the above, the compounds herein described may have asymmetric centers which result in one enantiomer of a compound of Formula (I) demonstrating superior biological activity over the opposite enantiomer. Both of the configurations are considered part of the invention. For example, the R2 substituent of a compound of Formula (I) may exist in either an S or R configuration. An example of a preferred enantiomeric configuration of the invention is a compound of Formula (I-s):
Figure imgf000041_0001
(I-s) but is not intended to be limited to this example. When required, separation of the racemic material can be achieved by methods known in the art. [00149] Compounds of the invention can also include tautomeric forms, such as keto-enol tautomers. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
[00150] Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium.
[00151] The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. [00152] The present invention also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, "pharmaceutically acceptable salts" refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and in the Journal of Pharmaceutical Science, 66, 2 (1977), the disclosures of each of which are hereby incorporated by reference.
Synthesis
[00153] Compounds of the invention, including salts and solvates thereof, can be prepared using known organic synthesis techniques and can be synthesized according to any of numerous possible synthetic routes.
[00154] The reactions for preparing compounds of the invention can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected.
[00155] Preparation of compounds of the invention can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3rd. Ed., Wiley & Sons, Inc., New York (1999), which is incorporated herein by reference in its entirety. [00156] Reactions can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., ]H or 13C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography. [00157] Compounds of the invention can be prepared according to methods for preparing aminoboronic acids, esters thereof, and related compounds described in the art, such as in U.S. Pat. No. 4,537,773, and in U.S. Pat. No. 5,614,649, each of which is incorporated herein by reference in its entirety. In some embodiments, the present compounds can be prepared by the sequential coupling of three fragment components (FI, F2, and F3).
FI Fragment
[00158] Synthesis of compounds of the invention can involve a boron-containing fragment (FI) having a structure indicated by Formula (A).
Figure imgf000043_0001
(A) [00159] The boronic ester moiety of FI can include, for example, a diol ester such as is indicated by the loop connecting oxygen atoms in Formula (A). [00160] Stereochemistry at the carbon atom alpha to the boron atom in Formula
(A) can be controlled using an asymmetric boronic ester group in the preparation of FI. For example, pinanediol esters of boronic acid can facilitate the preparation or stereochemically pure, or substantially stereochemically pure, FI fragment. As an example, the FI fragment can be prepared by reacting a compound of Formula (B) (showing a pinanediol boronic ester obtained from (+)-pinanediol) with a strong base (e.g., lithium diisopropylamide or lithium dicyclohexylamide) in the presence of dichloromethane or dibromomethane, followed by addition of a Lewis acid, (e.g., ZnCl2, ZnBr2, or FeCl3) to yield a compound of Formula (C) (where L is halo) having a newly introduced stereocenter at the carbon alpha to the boron.
Figure imgf000044_0001
(B)
Figure imgf000044_0002
(C) [00161] The compound of Formula (C) can, in turn, be reacted with an alkali amide (e.g., lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide, and potassium bis(trimethylsilyl)amide) or other nucleophile that effectively inverts the newly formed stereocenter (such as by an SN2 type mechanism) and introduces an amine group (NR2) in place of the halo group (e.g., chloro), forming a compound of Formula (D) (where R can be, e.g., alkyl, Si(alkyl)3, aryl, or aralkyl).
Figure imgf000044_0003
CD) [00162] The compound of Formula (D) can be further reacted with an agent capable of converting the NR2 group to NH2, or salt thereof, to form an FI fragment substantially capable of coupling with a further fragment through the amine. A suitable agent for converting the NR2' group to NH2 can be a protic acid such as HCl such as when R is a silyl group (e.g., trimethylsilyl).
[00163] The compound of Formula (B) can also be prepared according to a two step procedure involving reaction of a trialkoxyborane, preferably triisopropoxyborane, with (IS, 2S, 3R, 5S)-(+) pinanediol, to give a mono-alkoxy [(IS, 2S, 3R, 5S)-(+) pinanediol] borane intermediate wherein two of the alkoxy groups of the trialkoxy borane have been replaced by (IS, 2S, 3R, 5S)-(+) pinanediol. This mixed pinanediol alkoxy borane, upon reaction with the appropriate organometallic derivative, e.g. the Grignard reagent R!CH2MgBr or the alkyl lithium R'CH^i, gives compound (B) in good yields and purities. The process starting from triisopropoxyborane to give the intermediate mixed pinanediol isopropoxy borane (F) and the compounds of formula (B) is depicted in the following scheme:
Figure imgf000045_0001
and exemplified in Example A.2, herein.
[00164] Accordingly, the present invention is further directed to methods of preparing compounds of Formula (II):
Figure imgf000045_0002
(II) wherein the variable constituents are defined hereinabove, by the process of a) reacting a diol of Formula (Il-b):
Figure imgf000045_0003
(Il-b) with an appropriate trialkoxyborane of Formula (II-a):
R17Cv .OR17
OR17 (Il-a) wherein each R17 is, independently, Ci-do alkyl or C3-Cιo cycloalkyl; for a time and under conditions suitable for forming a mixed trialkoxyborane intermediate of Formula (II-c):
Figure imgf000046_0001
(II-c) and reacting the intermediate of Formula (II-c) with either i) a reagent of formula R1CH2MXhal, wherein M is a metal and Xhal is a halogen atom, or ii) a reagent of formula R'CH^Li , for a time and under conditions suitable for forming the compound of Formula (II).
[00165] In some embodiments, R17 is d-C4 alkyl.
[00166] In some embodiments, R17 is isopropyl.
[00167] In some embodiments, the diol of Formula (Il-b) is pinanediol, pinacol, bicyclohexyl-l,l'-diol, 1,2-ethanediol, 1,3-propanediol, 1,2-propanediol, 2,3-butanediol, 1 , 1 ,2,2-tetramethylethanediol, 1 ,2-diisopropylethanediol, 5,6-decanediol, 1,2-dicycIohexylethanediol, bicyclohexyl-l,l'-diol, diethanolamine, or 1 ,2-diρhenyl- 1 ,2-ethanediol.
[00168] In some embodiments, the diol of Formula (Il-b) is pinanediol.
[00169] In some embodiments, the Formula R'C^MX1131 is a Grignard reagent.
[00170] In some embodiments, the Formula R'CH^-MX1131 is R!CH2MgBr.
[00171] In some embodiments, R1 is isopropyl.
[00172] In some embodiments, the present invention provides a process for preparing a compound of Formula (Il-i):
Figure imgf000046_0002
αι-0 i comprising: a) reacting (IS, 2S, 3R, 5S)-(+)-pinanediol with triisopropoxy borane for a time and under conditions suitable for forming an intermediate of Formula (Il-ii):
Figure imgf000047_0001
(Il-ii) and b) reacting the intermediate of Formula (Il-ii) with isobutyl magnesium bromide for a time and under conditions suitable for forming the compound of Formula (Il-i). [00173] In some embodiments, the present invention provides a compound of
Formula (Il-ii):
Figure imgf000047_0002
(Il-ii). [00174] The reacting steps can be carried out in any suitable solvent that is non- reactive with the reagents and products and allows combining of reagents at lowered temperatures (e.g., temperatures colder than room temperature). Suitable solvents include ethers such as dimethoxymethane, tetrahydrofuran, 1,3-dioxane, 1,4-dioxane, furan, diethyl ether, ethylene glycol dimethyl ether, ethylene glycol diethyl ether, diethylene glycol dimethyl ether, diethylene glycol diethyl ether, triethylene glycol dimethyl ether, anisole, or t-butyl methyl ether. In some embodiments, the ether solvent contains tetrahydrofuran and/or diethyl ether,
[00175] The reactions of the processes described herein can be carried out at appropriate temperatures which can be readily determined by the skilled artisan. Reaction temperatures will depend on, for example, the melting and boiling points of the reagents and solvent, if present; the thermodynamics of the reaction (e.g., vigorously exothermic reactions may need to be carried out at reduced temperatures); and the kinetics of the reaction (e.g., a high activation energy barrier may need elevated temperatures). "Elevated temperature" refers to temperatures above room temperature (about 22 °C) and "reduced temperature" refers to temperatures below room temperature. [00176] In some embodiments, suitable temperatures are reduced temperatures.
The reaction of the trialkoxyborane and diol to prepare a mixed trialkoxyborane intermediate can be carried out, for example, at a temperature of about -20 to about 10 °C. In some embodiments, the reaction of the trialkoxyborane and diol can be carried out at about 0 °C. The reaction of the mixed trialkoxyborane intermediate with the organometallic reagent R'CTyVTX1181 or the alkyl lithium reagent R!CH2Li can be carried out, for example, at temperature from about -100 to about -20 °C. In some embodiments, the reaction of the mixed trialkoxyborane intermediate and R^BMX1"1 is carried out at about -78 °C.
[00177] The reactions of the processes described herein can be carried out in air or under an inert atomosphere. Typically, reactions containing reagents or products that are_ substantially reactive with air can be carried out using air-sensitive synthetic techniques that are well known to the skilled artisan.
B. F2 Fragment
[00178] The mid-section of compounds of the present invention can be represented by fragment F2 which couples to fragment FI by peptide bond formation for form an F2-F1 intermediate. Methods for coupling compounds through peptide bonds, or amide bonds, are well known in the art and described, for example, in The Peptides: Analysis, Synthesis, Biology, Vol. I., eds. Gross, et al., Academic Press, 1979. An example F2 fragment is provided in Formula (E) (Pg is an amino protecting group, R2 is defined herein). Additionally, protection of the amino group of amino acids using Boc or other amino protecting groups is well known in the art.
Figure imgf000048_0001
(E) [00179] Compounds of Formula (E) that are amino acids or amino acid derivatives are available commercially or prepared by routine methods. For example, aza-serines can be prepared generally by the Hoffman Rearrangement (Hoffman's Reaction) using, for example, asparagine where the amide of the asparagine side chain is converted to an amine (which can be subsequently protected). Methods for carrying out Hoffman Rearrangements, such as for amino acids, are known in the art and also provided in the Examples below. Additionally aza-serines can be prepared as disclosed in Zhang, et al. J Org. Chem., 1997, 62, 6918-6920. Boc-cyanoarginine derivatives can be prepared as disclosed in Wagenaar et al., J. Org. Chem. 1993, 58, 4331-4338. Synthesis of F2 fragments wherein R2 is -CH2CH2CH2NHC(=NR4)NH-Y, - CH2CONR5R6, -CH2NHCONR5R6, -CH2NR9R10, or -CH(R7)ZR8 are further disclosed herein. F2 fragments can be obtained from commercial sources or made by methods known to one skilled in the art.
C. F3 Fragments
[00180] A further fragment (F3) can be coupled to the F2 fragment of the F2-F1 intermediate by any of various means such as by nucleophilic substitution or addition reactions where, for example, F2 contains a nucleophile (e.g., amine) and F3 contains an electrophile (e.g., CO, SO2, and the like) and optionally a leaving group (e.g., halo, hydroxy, alkoxy, alkylsulfonyl, arylsulfonyl, and the like). Example F3 fragments can have the formula RxCOXL, RxSO2XL, RxNCO, or RxHCO, (e.g., R can be RA, RB, or Rc as defined herein and X can be a leaving group). Coupling of RxCOXL (such as when XL is OH) to the F2-F1 intermediate can be carried out according to standard procedures for peptide bond formation to prepare compounds having the formula F3-F2- Fl where the F3 and F2 fragments are coupled via an amide bond. In other embodiments, F3 and F2 can be coupled by a sulfonylamino linkage prepared by reacting RxSO2XL with the F2-F1 intermediate in which an amino moiety on the F2-F1 intermediate displaces the XL leaving group of R SO2XL. Additionally, reaction of RxNCO with an amino moiety of the F2-F1 intermediate can result in a urea linkage (- HNCONH-), while reaction of RxHCO with an amino moiety of the F2-F1 intermediate followed by reduction of the resulting imine moiety can form an amine linkage. Other coupling means are known in the art and are also suitable. F3 fragments can be obtained from commercial sources or made by methods known in the art. [00181] Certain compounds of the invention wherein R2 is -(CH2)dCH(R7)NR9R10 can be prepared by removal of an R1 amino protecting group to form the corresponding deprotected compound wherein R10 is H. This deprotected compound can be reacted with a reagent having the formula RIOaXL, wherein R10 has the same meaning as R10 with the exception of H and XL is a leaving group such as halo or a sulfonic acid derivative, or wherein R10a and X taken together represent, for example, a reactive alkyl, carbocyclyl or heterocarbocyclyl isocyanate, or an alkyl, carbocyclyl, heterocarbocyclyl sulphonylisocyanate. For example, the compound of Example D.26 can be prepared by the deprotection of the benzyloxycarbonyl group of Example D.16.6 to give Example D.17, from which the azaserine nitrogen can be subsequently acylated. [00182] The present invention further provides methods for preparing azaserine
(e.g., where R2 is -CH2NH2) compounds of Formula I. In general, the azaserine group can be generated by removal of a benzyloxycarbonyl group
Figure imgf000050_0001
which is attached to one of the nitrogens of the azaserine group (e.g., compounds of Formula I where R2 is -CH2NR9R10 and R9 is H and Ri0 is -C(=0)OCH2(C6H5)). Removal of the benzyloxycarbonyl group can be carried out by treatment with a reducing agent, such as a hydrogenation reagent. In some embodiments, the hydrogenation reagent contains H2 which is optionally used in the presence of a metal catalyst (e.g., Pd/C 10%). Hydrogenation can be further carried out in the presence of a protic acid such as HCl and in a suitable hydrogenation solvent containing, for example, an alcohol and/or an ether solvent. In some embodiments, the hydrogenation solvent contains an ether such as 1,4-dioxane. In further embodiments, the hydrogenation solvent cotains an alcohol such as methanol. In further embodiments, the hydrogenation solvent contains a mixture of alcohol and ether. An example preparation of an azaserine compound according to this process is provided, for example, in Example D.17. Reaction parameters including temperature, pressure, atomosphere and the like are readily deteπnined by one skilled in the art of chemical synthesis and reaction progress can be monitored by routine methods including, e.g., NMR.
[00183] Accordingly, the present invention provides a process for the preparation of compounds of Formula (I):
Figure imgf000050_0002
(I) wherein: R1 is Ci-Cβ alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or C3-C7 cycloalkyl;
R2 is -CH2NH2;
Q is -B(ORI4)2, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O; R14 is C1-C4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl; X is RAC(=O)-; RA is Ci -C20 alkyl optionally substituted with R20; C2-C2o alkenyl optionally substituted with R20; C2-C2o alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R21; or heterocarbocyclyl optionally substituted with 1-5 R21; R20 is selected from the group consisting of: -CN, halo, haloalkyl-, d-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, -CO2H, -C(=O)CO2H, -C(=0)NH2, -C(=0)H, -S(=O)NH2, -S(=O)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)NH2, -NHC(=O)R20a, -NHC(=O)OR20a, -OR 0a, -SR20a, -S(=O)R20a, -S(=O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(=O)2R20a, -NHR20b, phthalimido, -(O-alkyl)r, -O-alkyl-OH, -(O-alkyl)r-OH, -OR20c, -SR20c, -O-alkyl-R20c, -S-alkyl-R20c, -S(=O)-R20c, -S(=O)2-R20c, -S(=O)2-NHR20c, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR20c, carbocyclyl optionally substituted with 1-5 R21; and heterocarbocyclyl optionally substituted with 1-5 R21; R20 is Cι-C20 alkyl, C2-C2o alkenyl, or C2-C2o alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, d-C4 alkyl, aryl, heteroaryl or -NHR20b; R20b is an amino protecting group; R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22; R21 is selected from the group consisting of: d-C20 alkyl, C2-C2o alkenyl, C2-C2o alkynyl, Cι-C2o alkoxy, Ci- o thialkoxy, -OH -CN, halo, haloalkyl, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=0)0-alkyl, -NHC(=0)alkyl, -C(=0)0-alkyl, -C(=0)alkyl, -S(=O)-alkyl, -S(=O)2-alkyl, -S(=O)-aryl, -S(=O)2-aryl, carbocyclyl optionally substituted with 1-5 R22, and heterocarbocyclyl optionally substituted with 1-5 R22;
R22 is selected from the group consisting of: Ci-do alkyl, C2-C10 alkenyl, C2-Cι0 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=0)-, alkyl-C(=0)-, aryl-OC(=O)-, alkyl-OC(=O)NH-, aryl-OC(=0)NH-, alkyl-C(=O)NH-, alkyl-C(=0)0-, (alkyl-O)r-alkyl, HO-(alkyl-O)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=O)-, alkyl-S(=0)2-, H2NS(=0)-, and H2NS(=0)2-; and r is 2, 3, 4, or 5; comprising: reacting a compound of Formula (I) wherein R2 is -CH2NH-C(=O)OCH2(C6H5); with a suitable hydrogenation reagent for a time and under conditions suitable for forming the compound of Formula (I) wherein R2 is -CH2NH2, provided the hydrogenation agent is selective for the benzyloxycarbonyl group of R2.
[00184] In some embodiments, the hydrogenation agent is H2 in the presence of
Pd/C 10% and HCl in 1,4-dioxane.
Boronic Ester/Boronic Acid Conversion
[00185] Compounds of the invention containing boronic esters, such as pinanediol esters, can be hydrolyzed by any suitable means to prepare corresponding boronic acid
(-B(OH)2) derivatives. Hydrolysis conditions can include contacting a boronic ester with excess acid, such as a protic acid like HCl.
[00186] Conversely, boronic acids can be esterified by contacting the acid compound
(-B(OH)2) with an alcohol such as a diol for sufficient time to produce the corresponding ester. The esterification reaction can be acid or base catalyzed.
[00187] The invention will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of noncritical parameters which can be changed or modified to yield essentially the same results.
EXAMPLES
Example A.1
Synthesis of (lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyI-4,6-methano- l,352-benzodioxaborol-2-yl]-3-methyIbutylamine hydrochloride salt
Step 1 : 2-(2-methylpropyl)-(3aS, 4S, 6S, 7aR)-hexahydro-3a, 5, 5-trimethyl-4, 6-methano- 1, 3,2-benzodioxaborole
Figure imgf000053_0001
[00188] A mixture of (+)-pinanediol (23.9 g, 0.140 mol) and 2- methylpropylboronic acid (15 g, 0.147 mol) in diethyl ether (300 ml) was stirred at room temperature for 24h. The mixture was dried over anhydrous sodium sulfate and purified by column chromatography (Silica gel 230-400 mesh), eluting with hexane:ethyl acetate 90:10 mixture. The product was obtained as a clear oil (32.6 g, 94% yield).
1H NMR (DMSO-d6): 4.28 (IH, dd, J=8.8 Hz, 2.0); 2.30 (IH, m); 2.18 (IH, m); 1.96 (IH, t, J=5.3); 1.86 (IH, m); 1.78 (IH, set, J=6.8); 1.68 (IH, m); 1.30 (3H, s); 1.25 (3H, s); 1.01 (IH, d); 0.9 (6H, d, J=6.6 ); 0.81 (3H, s); 0.69 (2H, m).
Step 2: 2-[(lS)-l-chloro-3-methylbutyl]-(3aS,4S,6S, 7aR)-hexahydro-3a,5,5-trimethyl- 4, 6-methano-l, 3,2-benzodioxaborole
Figure imgf000053_0002
[00189] A solution of lithium diisopropylamide was prepared by addition of 10.0
M butyl lithium solution in hexane (25.4 ml, 0.254 mol) to a solution of diisopropylamine (35.7 ml, 0.254 mol) in dry tetrahydrofuran (60 ml), at -50 °C, and allowing the temperature to rise to -30 °C. This solution was transferred via canula into a solution of 2-(2-methylproρyl)-(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-l,3,2-benzodioxaborole of Step 1 (50 g, 0.212 mol) and CH2C12 (50 ml, 0.848 mol) in dry tetrahydrofuran (700 ml), while keeping the temperature below -70°C. A 1.0 M solution of dry zinc chloride in diethyl ether (339 ml, 0.339 mol) was then added over a 30 minutes period while keeping the internal temperature below -70°C. The reaction mixture was stirred at -78°C for 3 hours, then allowed to warm to room temperature. After removal of the solvents by rotary evaporation the residue was partitioned between petroleum ether (1000 ml) and a 10% aqueous solution of ammonium chloride (800 ml). The aqueous layer was further extracted with petroleum ether (300 ml). The combined organic phases were dried over anhydrous sodium sulfate and concentrated. The product was obtained as a brown oil (59.0 g, 98% yield) containing about 9% mol/mol of starting material (Η-NMR), and was used in the subsequent step without further purification.
'H NMR (DMSO-d6): 4.43 (IH, dd, J=8.8, 1.8 ); 3.59 (IH, m); 2.33 (IH, m); 2.21 (IH, m); 2.01 (IH, m); 1.88 (IH, m); 1.84-1.55 (5H, m); 1.34 (3H, s); 1.26 (3H, s); 1.09 (IH, , J=10.1); 0.9 (3H, d5 J=6.8); 0.87 (3H, d, J=6.4); 0.82 (3H, s).
Step 3: N,N-Bis(trimethylsilyl)-(lR)-l-[(3aS,4S,6S, 7aR)-hexahydro-3a,5,5-trimethyl- 4, 6-methano-l, 3, 2-benzodioxaborol-2-yl]-3-methylbutylamine
Figure imgf000054_0001
[00190] A 1.0 M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran
(189 ml, 0.189 mol) was added, over 30 minutes, to a solution of crude 2-[(lS)-l- chloro-3-methylbutyl]-(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborole of Step 2 (59.0 g, 91% purity, 0.189 mol) in tetrahydrofuran (580 ml) while cooling at -78 °C. The reaction mixture was allowed to slowly warm to room temperature overnight. The solvent was removed by rotary evaporation and the residue taken up with dry hexane (800 ml). The resulting suspension was stirred at room temperature for 2 hours, then the solid was removed by filtration on a celite cake, which was washed with dry hexane (3 x 100 ml). The filtrate was concentrated giving a satisfactorily pure product as a brown oil (79 g) in practically quantitative yield. The product was used for the subsequent step without further purification. 1H NMR (DMSO-de): 4.33 (IH, dd, J=1.5 Hz, 8.6); 2.58 (IH, m); 2.29 (IH, m); 2.18 (IH, m); 1.95 (IH, t, J=5.9); 1.85 (IH, m); 1.9-1.55 (3H, m); 1.31 (3H, s); 1.24 (3H, s); 1.17 (IH, m); 1.01 (IH, d, J-10.6); 0.85 (3H, d, J=6.6), 0.83 (3H, d, J=6.6); 0.80 (3H, s); 0.08 (18H, s).
Step 4: (lR)-l-[(3aS,4S,6S, 7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt
Figure imgf000055_0001
[00191] To a solution of crude N,N-Bis(trimethylsilyl)-(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutylamine of Step 3 (79 g, 0.193 mol) in a mixture of dioxane (100 ml) and diethyl ether (200 ml), a 4 N solution of hydrogen chloride in dioxane (193 ml, 0.772 mol) was added, while cooling at 0°C. The mixture was then stirred at room temperature for 4 hours and concentrated. The residue was taken up with anhydrous hexane (500 ml) and a 2 M solution of hydrogen chloride in diethyl ether (48 ml, 0.096 mol) was added. The mixture was stirred at 0°C for 1 hour, then concentrated. The residue was taken up with anhydrous hexane and the resulting suspension was stirred at room temperature overnight. The solid was collected by filtration and dried under vacuum affording 38.1 g of product (66% yield). A second crop (4.13 g, 7% yield) was obtained from the mother liquors.
1H NMR (DMSO-de): 7.85 (3H, br); 4.45 (IH, dd, J= 9.2 Hz); 2.78 (IH, m); 2.34 (IH, m); 2.21 (IH, m); 2.01 (IH, t, J=5.3); 1.89 (IH, m); 1.82-1.65 (2H, m); 1.49 (IH, m); 1.38 (3H, s); 1.27 (3H, s); 1.12 (IH, d, J=1.12); 0.87 (6H, d, J=6.6); 0.83 (3H, s). Example A.2
Alternate synthesis of 2-(2-methylpropyl)-(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyI-4,6-methano-l,3,2-benzodioxaboroIe.
Step 1: 2-(l-methylethoxy)-(3aS,4S,6S, 7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1, 3, 2-benzodioxaborole.
Figure imgf000056_0001
[00192] To a solution of (IS, 2S, 3R, 5S)-(+)-Pinanediol (50.0 g, 0.293 mol) in anhydrous tetrahydrofuran (350 ml) triisopropoxy borane was slowly added while stirring at 0°C under nitrogen. After 2h the solvent was removed by rotary evaporation. The oily residue was redissolved in hexane (150 ml) and the solution was filtered to remove a very small amount of a white solid. The filtrate was concentrated by rotary evaporation affording the product as a clear oil (62.6 g, 90% yield). Η NMR (DMSO-d6): 4.31-4.20 (2H, m); 2.34-2.16 (2H, m); 1.96 (IH, t, J=5.5); 1.90- 1.85 (IH, m); 1.74-1.67 (IH, m); 1.32 (3H, s); 1.31 (IH, d, J=7.6); 1.25 (3H, s); 1.14 (3H, d, J=6.1); 1.13 (3H, d, J=6.1); 0.81 (3H, s).
Step 2: 2-(2-methylpropyl)-(3aS,4S,6S, 7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1, 3, 2-benzodioxaborole.
Figure imgf000056_0002
[00193] A 2M solution of isobutyl magnesium bromide in diethyl ether (131.5 ml, 0.263 mol) was added dropwise, in 1 hour, to a solution of 2-(l-methylethoxy)- (3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborole obtained in Step 1 (62.6 g, 0.263 mol), in anhydrous tetrahydrofuran (330 ml) while stirring at -78°C, under nitrogen. The mixture was then allowed to warm to room temperature, then transferred in a mixture of 2N sulfuric acid (150 ml) and diisopropyl ether (250 ml). After stirring for 10 minutes, a saturated solution of NaCl was added (100 ml) and the layers were separated. The organic phase was washed with brine (100 ml), dried over sodium sulfate and concentrated. The residue was purified by column chromatography (silica gel) eluting with 5% diethyl ether in hexane. The product was obtained as a clear oil (38.45 g, 62% yield).
[00194]
Example B.l
Carbamic acid 1,1-dimethylethyl ester, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaboroI-2-yI]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]-.
Figure imgf000057_0001
Method A: HOAt/HATU
[00195] To a solution of BocNH(NO2)ArgOH (15.7 g, 49.3 mmol) in anhydrous
DMF (100 ml), HATU (0-(7-azabenzotriazol-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate; 18.7 g, 49.3 mmol) and HOAt (l-hydroxy-7-azabenzotriazole; 6.71 g, 49.3 mmol) were added. The mixture was cooled to 0°C and N- methylmo holine was added (13.6 ml, 0.123 mol). After 10 minutes (1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutylamine hydrochloride salt of Example A.l (12.4 g, 41.1 mmol) was added. The cooling bath was removed and the mixture was stirred at r.t. for 4.5 hours. The mixture was diluted with ethyl acetate (800 ml), washed with a 2% solution of citric acid (2x 150 ml), 2% solution of NaHCO3 (2x150 ml) and 2% solution of NaCl (2x150 ml). The aqueous phases were further extracted with ethyl acetate (150 ml). The combined organic phases were dried over sodium sulfate and concentrated. The resulting oily residue was redissolved in ethyl acetate (500 ml) and the solution was washed with cold water (200 ml). The aqueous phases were further extracted with ethyl acetate (500 ml). The combined organic phases were dried over sodium sulfate and concentrated. The residue was dissolved in diethyl ether (100 ml) an the solution was slowly added to hexane (600 ml) while stirring. The white solid was collected by filtration (43.4 g) and purified by column chromatography eluting initially with 50:50 hexane:ethyl acetate mixture and then with ethyl acetate. The fractions containing the product were concentrated, the residue was dissolved in diethyl ether (100 ml) and the resulting solution was slowly added to hexane (600 ml) while stirring. The white solid was collected by filtration (15.2 g, 66% yield).
Method BCF
[00196] To a suspension of BocNH(NO2)ArgOH (5.82 g, 18.2 mmol) in anhydrous dichloromethane (100 ml) N-methylmorpholine (2.0 ml, 18.2 mmol) was added. The mixture was cooled to -15 °C then isobutyl chloroformate was added (2.37 ml, 18.2 mmol). The mixture was stirred at -15°C for 10 minutes then (1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yI]-3-methylbutyIamine hydrochloride salt obtained as in Example A.l was added (5.0 g, 16.6 mmol), immediately followed by further N-methylmorpholine (2.0 ml, 18.2 mmol). The reaction mixture was stirred for 1.5 hours at -15°C, then allowed to warm to room temperature and partitioned between ethyl acetate (150 ml), water (150 ml) and 0.1N hydrochloric acid (10 ml). The organic phase was washed with a saturated solution of NaHCO3, dried over anhydrous sodium sulphate and concentrated. The oily residue (9.25 g) was purified by crystallization from ethyl acetate affording three crops of satisfactorily pure product (5.03 g, 54% yield).
Η NMR (DMSO-d6): 8.80 (IH, br); 8.50 (IH, br), 7.87 (2H, br);7.01 (IH, d, J=7.9), 4.07 (IH, dd, J=7.9); 4.0 (IH, m); 3.12 (2H, m); 2.55 (IH, m); 2.2 (IH, m); 2.01 (IH, m); 1.83 (IH, t, J=5.1); 1.78 (IH, m); 1.74-1.44 (7H, m); 1.38 (9H, s); 1.33 (IH, d, J=10.3); 1.24 (5H, s); 1.22 (3H, s); 0.84 (6H, d, J=6.6); 0.81 (3H, s)
Example B.2
Carbamic acid 1,1-dimethylethyl ester, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyI]amino]carbonyl]-2-hydroxypropyl]-
Figure imgf000059_0001
[00197] Boc-L-threonine (870 mg, 3.97 mmol, 1.2eq.) was dissolved in DMF dry
(30 ml) at r.t.. To this solution, TBTU (N,N,N',N'-tetramethyl-0-(benzotriazol-l- yl)uronium tetrafluoroborate; 1270 mg, 3.97 mmol, 1.2eq.) was added and the mixture was cooled at 0°-5°C. Then NMM (0.9 ml, 8.27 mmol, 2.5eq.) and (1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutylamine hydrochloride salt of Example A.l (1000 mg, 3.3 mmol, 1 eq.) were added. The mixture was stirred at r.t. for 16h, then was extracted with ethyl acetate (100 ml) washed with the following solutions: citric acid 2% (50 ml), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated under reduced pressure to give 1290 mg of glassy solid. Yield 84.3%. M.p. 25°-30°C
1H NMR (DMSO-d6): 8.88 (IH, br); 6.49 (IH, d, J=8.4 Hz); 4.88 (IH, d, J=5.8); 4.05 (IH, dd); 3.93 (IH, m); (IH, m); 2.51 (IH, m); 2.19 (IH, m); 2.01 (IH, m); 1.83 (IH, t, J=5.9), 1.78 (IH, m); 1.68 (IH, m); 1.62 (IH, m); 1.39 (9H, s); 1.34 (IH, d, J=10.0); 1.24 (3H, s); 1.22 (3H, s); 1.06 (3H, d, J=6.4); 0.85 (6H, d, J=6.4); 0.80 (3H, s)
Example B.3
Further intermediate compounds
[00198] Starting from the appropriate intermediate and following either of the procedures described in the Example B.l and B.2, the intermediates reported below were prepared.
(2S)-2-[(l,l-dimethylethoxycarbonyl)amino]-5-ureidopentanamide, N-[(1R)-1- [(3aS, 4S, 6S, 7aR)-hexahydro-3a, 5, 5-trimethyl-4, 6-methano-l , 3, 2-benzodioxaborol-2- ylJ-3-methylbutylJ Chiral
Figure imgf000060_0001
'H NMR (DMSO-d6): 8.85 (IH, br); 7.01 (IH, d, J=8.0 Hz);5.9 (IH, t, 3=5.1); 5.36 (2H, br); 4.03 (2H, m); 2.93 (2H, m); 2.19 (IH, m); 2.0 (IH, m); 1.83 (IH, t, J=5.3); 1.78 (IH, m); 1.68 (IH, m); 1.62 (IH, m); 1.52 (2H, m); 1.38 (9H, s); 1.33 (IH, d, J=9.9); 1.24 (3H, s); 1.22 (2H, s); 0.86 (3H, d, J=6.6); 0.84 (3H, d, J=6.6); 0.80 (3H, s).
(2S)-3-(Aminocarbonyl)-2-[(l,l-dimethyleihoxycarbonyl)amino]propanamide, N-[(1R)- l-[(3aS, 4S, 6S, 7aR)-hexahydro-3a, 5, 5-trimethyl-4, 6-methano-l, 3,2-benzodioxaborol-2- yl]-3-methyϊbutyl]
Figure imgf000060_0002
lH NMR (DMSO-dό): 8.74 (IH, br); 7.28 (IH, br);6.95 (2H, m); 4.36 (IH, m); 4.07 (IH, m); 2.55 (IH, m); 2.38 (2H, m); 2.2 (IH, m); 2.02 (2H, m); 1.84 (IH, t, J=5.5); (IH, m); 1.79 (IH, m); 1.68 (IH, m); 1.63 (IH, m); 1.38 (9H, s); 1.33 (IH, d, J=10); 1.24 (3H, s); 1.22 (2H, s); 0.85 (3H, d, J=6.4); 0.83 (3H, d, J=6.4); 0.81 (3H, s).
Carbamic acid benzyl ester, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a, 5, 5-trimethyl-4, 6-methano-l, 3, 2-benzodioxaborol-2-yl]-3- methylbutyl] amino] carbonyl] -2-hydroxypropyl].
Figure imgf000061_0001
[00199] M.p. 57-60 °C. Η NMR (DMSO-d6): 8.66 (IH, s); 7.40-7.29 (5H, m);
7.09 (IH, d, J=8.75); 5.06 (2H, s); 4.90 (IH, J=5.68); 4.11-3.99 (2H, m); 3.91-3.77 (IH, m); 2.58-2.53 (IH, m); 2.26-2.14 (IH, m); 2.07-1.97 (IH, s); 1.84 (IH, t, J=5.52); 1.81- 1.75 (IH, m); 1.73-1.58 (2H, m);1.33 (2H, d, J=10.1); 1.27-1.20 (7H, m); 1.06 (3H, t, J=6.27); 0.91-0.79 (9H, m).
Example B.4
(2S)-2-[(l,l-Dimethylethoxycarbonyl)amino]-3-[(4- methylbenzoyl)amino]propanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyI-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]-
Figure imgf000061_0002
[00200] (2S)-2-[(l,l-Dimethylethoxycarbonyl)amino]-3-[(4- methylbenzoyl)amino]-propanoic acid, (650 mg, 2 mmol, 1.2 eq.) of Example G.6, was dissolved in DMF dry (15 ml), under nitrogen, and TBTU (640 mg, 2 mmol, 1.2 eq.) was added at r.t.. The mixture was cooled at 0°-5°C with ice bath and NMM (0.55 ml, 5 mmol, 2.5eq.) and (lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- l,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt, (500 mg, 1.65 mmol, 1 eq.) of Example A.l, were added. The mixture was stirred overnight, poured in water (200 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (20 mL), sodium bicarbonate 2% (20 ml), NaCl 2% (20 ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated to give 740 mg of glassy solid (quantitative yield). ]H NMR (DMSO-de) 8.76 (IH, br); 8.28 (IH, t, J=5.31 Hz);7.71 (2H, d, J=7.9); 7.26 (2H, d, J=7.9); 6.97 (IH, d, J=8.0); 4.27 (IH, m); 4.07 (IH, dd, J=8.2, 1.5); 3.48 (2H, m), 2.58 (IH, m); 2.35 (3H, s); 2.19 (IH, m); 2.02 (IH, m); 1.83 (IH, t, J=4.9); 1.78 (IH, m); 1.62 (2H, m); 1.35 (12H, m); 1.24 (3H, s); 1.23 (3H, s); 0.82 (3H, d); 0.80 (3H, d); 0.78 (3H, s).
Example B.5
2-S-[(l,l-DimethyIethoxycarbonyl)amino]-3-(hexanoylamino)-propionamide, N- [(lS)-l-[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyI-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]
Figure imgf000062_0001
[00201] 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid, (300 mg, 1 mmol, 1.2 eq.) of Example G.7 was dissolved in DMF dry (25 ml), under nitrogen, and TBTU (318 mg, 1 mmol, 1.2 eq.) was added at r.t.. The mixture was cooled at 0°-5°C with ice bath and NMM (0.27 ml, 2.47 mmol, 2.47eq.) and (1R)-1-
[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutylamine hydrochloride salt, (250 mg, 0.82 mmol, 1 eq.) of Example A.l, were added. The mixture was stirred 3h, poured in water (150 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (50 mL), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated to give 450 mg of glassy solid. Yield quantitative.
Analytical data:
'H NMR (DMSO-de). δH: 8.71 (IH, br d, J==2.6 Hz); 7.73 (IH, br t, J=5.9 Hz); 6.81 (IH, d, J= 8.2); 4.10 (2H, m); 3.24 (2H, m); 2.56 (IH, m); 2.19 (IH, ra); 2.03 (3H, m); 1.83 (IH, t, J=5.5); 1.78 (IH, m); 1.64 (2H, m); 1.47 (2H, m); 1.36 (9H, s);1.4-1.15 (9H, m); 1.24 (3H, s); 1.21 (3H); 0.83 (9H, m); 0.79 (3H, s)
Example B.6
2-S-[(l,l-Dimethylethoxycarbonyl)amino]-3-(4-fluorosulfonyIamino)propionamide, N-[(lS)-l-[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]
Figure imgf000063_0001
[00202] 2-S-[(l, 1 -dimethylethoxycarbonyl)amino]-3-(4- fluorosulfonylamino)propionic acid, (1.39 g, 3.83 mmol, 1.2 eq.) of Example G.8, was dissolved in DMF dry (20 ml), under nitrogen, and TBTU (1.23 g, 3.83 mmol, 1.2 eq.) was added at r.t.. The mixture was cooled at 0°-5°C with ice bath and NMM (1 ml, 9.57 mmol, 3eq.) and (lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- l,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt, (0.96 g, 3.19 mmol, 1 eq.) of Example A.l, were added. The mixture was stirred 2h, poured in water (200 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (50 mL), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated with diethyl ether to give 1.5g of white solid. Yield 77%. Analytical data: lH NMR (DMSO-de). δH: 8.54 (IH, d, J=2.9 Hz); 7.91 (2H, m); 7.75 (IH, t, J=5.9); 7.50 (2H, t, J=8.8); 6.83 (IH, d, J=8.4); 4.19 (IH, br d, J=8.2); 4.14 (IH, m); 3.01 (2H, m); 2.69 (IH, m); 2.25 (IH, m); 2.09 (IH, m); 1.90 (IH, t, J=5.7); 1.85 (IH, m); 1.8-1.6 (2H, m); 1.5-1.2 (5H, m); 1.43 (9H, s); 1.29 (6H, s); 0.89 (6H, d, J=6.4); 0.86 (3H, s). Example B.7
2-S-[(l,l-Dimethylethoxycarbonyϊ)amino]-3-(3,4- dimethoxyphenylacetamido)propionamide, N-[(lS)~l-[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methy lbutyl] amino] carbonyl]
Figure imgf000064_0001
[00203] 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(3,4- dimethoxyphenylacetamido)-propionic acid, (0.73 g, 1.90 mmol, 1.2 eq.) of Example G.9, was dissolved in DMF dry (20 ml), under nitrogen, and TBTU (0.61 g, 1.90 mmol, 1.2 eq.) was added at r.t.. The mixture was cooled at 0°-5°C with ice bath and NMM (0.52 ml, 4.7 mmol, 2.5 eq.) and (lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt, (0.47 g, 1.6 mmol, 1 eq.) of Example A.l, were added. The mixture was stirred 2h, poured in water (200 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (50 mL), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated with diethyl ether to give 0.95g of crude that was purified by silica gel chromatography (eluent ethyl acetate) to give 0.3 g of white foam. Yield 30%. Analytical data: TLC silica gel (eluent ethyl acetate 100%, R.f.=0.50)
Figure imgf000064_0002
δH: 8.69 (IH, d, J=2.6 Hz); 7.90 (IH, t, J=5.7); 6.85 (2H, m); 6.74 (IH, dd, J=1.5, 8.1); 6.85 (3H, m); 4.12 (2H, m); 3.73 (3H, s); 3.72 (3H, s); 3.34 (2H, s); 3.31 (2H, m); 2.58 (IH, m); 2.20 (IH, m); 2.03 (IH, m); 1.85 (IH, t, J=5.3); 1.79 (IH, m); 1.66 (2H, m); 1.38 (9H, s); 1.40-1.15 ( 3H, m); 1.25 (3H, s); 1.23 (3H, s); 0.83 (6H, d, J=6.6); 0.81 (3H, s).
Example B.8
2-S-[(l,l-DimethyIethoxycarbonyl)amino]-3-(3-phenylureido)propionamide, N- [(lS)-l-[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaboroI-2-yl]-3-methylbutyl]amino]carbonyl]
Figure imgf000065_0001
[00204] 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid, (0.41 g, 1.26 mmol, 1.2 eq.) of Example G.10, was dissolved in DMF dry (20 ml), under nitrogen, and TBTU (0.40 g, 1.26 mmol, 1.2 eq.) was added at r.t.. The mixture was cooled at 0°-5°C with ice bath and NMM (0.346 ml, 3.15 mmol, 2.5 eq.) and (1R)- l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutyIamine hydrochloride salt, (0.31 g, 1 mmol, 1 eq.) of Example A.l, were added. The mixture was stirred 2h, poured in water (200 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (50 mL), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated with diethyl ether (50 ml) to give 0.58g of white solid. Yield 96.6%.
Analytical data: TLC silica gel (eluent ethyl acetate 100%, R.f.=0.47), m.p. 128°-130°C. 1H NMR (DMSO-d6). δH: 8.79 (IH, d, J=2.7 Hz); 8.69 (IH, s); 7.38 (2H, d, J= 7.9); 7.22 (2H, t, J= 8.1); 7.00 (IH, d, J= 8.1); 6.90 (IH, t, J=7.3); 6.16 (IH, t, J=5.7); 4.12 (2H, m); 3.45 (IH, m); 3.17 (IH, m); 2.60 (IH, m); 2.21 (IH, m); 2.04 (IH, m); 1.85 (IH, t, J=5.3); 1.79 (IH, m); 1.66 (2H, m); 1.38 (9H, s); 1.40-1.15 (3H, m); 1.26 (3H, s); 1.23 (3H, s); 0.84 (6H, d, J=6.6); 0.81 (3H, s). Example B.9 Synthesis of Further Compounds [00205] Following the procedures of Examples B.4-B.8, the following compounds can be prepared by reaction of (lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt of Example A.l and intermediates of Examples G.l 1, G.12 and G.13.
Figure imgf000066_0001
Figure imgf000067_0002
Example B.10 Carbamic acid 1,1-dimethylethyl ester, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-l- methylbutyl] amino] carbonyl] -methyl
Figure imgf000067_0001
[00206] This compound has been prepared following the procedure of Exampe B.l Method B starting from (lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt of Example A.l and commercially available N-(l,l-dimethylethoxycarbonyl)glycine. Η-NMR (DMSO-d6): 8.84 (IH, s); 7.08 (IH, t, J=5.93 Hz); 4.06 (IH, d, J=7.48 Hz); 3.67 (2H, t, J=5.32 Hz); 2.60-2.48 (IH, m); 2.24-2.16 (IH, m); 2.06-1.96 (IH, m); 1.84 (IH, t, J=5.50 Hz); 1.82-1.76 (IH, m); 1.74-1.58 (2H, m); 1.39 (10H, bs); 1.23 (9H, d, J 8.18 Hz); 0.87-0.83 (6H, m); 0.82 (3H, bs).
Example C.l
(2S)-2-amino-5-[[imino(nitroamino)methyl]amino]pentanamide, N-[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol- 2-yI]-3-methyIbutyI]; hydrochloride salt
Figure imgf000068_0001
Method A
[00207] A 4 N solution of hydrogen chloride in dioxane (15 ml) was added to a solution of carbamic acid 1,1-dimethylethyl ester, N-[(1S)-1-[[[(1R)-1- [(3 aS,4S,6S, 7aR)-hexahydro-3 a, 5, 5 -trimethyl-4, 6-methano- 1 , 3 ,2-benzodioxaborol-2- yl] -3 -methylbutyl] amino] carbonyl] -4-[[imino(nitroamino)methyl] amino]butyl] - of
Example B.l, (4.04 g, 7.06 mmol) in a mixture of dioxane (40 ml) and diethyl ether (7 ml), while cooling at 0°C. The reaction mixture was allowed to warm to room temperature and stirred for further 4 hours. The solvent was removed by rotary evaporation, the residue was treated with diethyl ether (50 ml) and the mixture was stirred at r.t. for three days. The resulting solid was collected by filtration affording 3.18 g of pure product (90% yield)
Method B
[00208] Carbamic acid 1,1-dimethylethyl ester, N-[(1S)-1-[[[(1R)-1-
[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl] -3 -methylbutyl] -amino] -carbonyl] -4-[[imino(nitroamino)-methyl] -amino]butyl] - of Example B.l, (3 g, 5.3 mmol) was dissolved in Et2O (40 mL) and a solution of about 10%) HCl in Et2O (20 mL) was added dropwise at 0°C under nitrogen. The reaction mixture was allowed to warm to room temperature and to stir for further 5 hours. The solvent was decanted and the residue, washed twice with Et2O (20 mL), was dried in vacuo to give the title compound as a white powder (2.43 g, yield 91%). 1H NMR (DMSO-de): 8.56 (2H, br); 8.22 (3H, br); 7.97 (2H, br); 4.28 (IH, dd, J=8.6 Hz, 2.01); 3.77 (IH, m); 3.04 (IH, m); 2.28 (IH, m); 2.11 (2H, m), 1.92 (IH, t, J=5.5); 1.83 (IH, m); 1.79-1.59 (4H, m); 1.59-1.37 (3H, m); 1.31 (4H, s); 1.24 (3H, s); 1.19 (IH, d, J=10.4); 0.88 (3H, d, J=6.0); 0.86 (3H, d, J=6.0); 0.81 (3H, s).
Example C.2
Boronic acid, [(lR)-l-[[(2S)-2-amino-5-[[imino(nitroamino)methyl]amino]-l- oxopentyl]amino]-3-methyIbutyI], hydrochloride salt
Figure imgf000069_0001
[00209] Carbamic acid 1,1-dimethylethyl ester, N-[(1S)-1-[[[(1R)-1-
[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]- of
Example B.l, (3.1 g, 5.48 mmol) was carefully dissolved, under nitrogen at 0°C, in 20 mL of HCl 37%; the resultant mixture was allowed to warm to room temperature and to stir overnight. The reaction mixture was washed with Et2O until complete removal of pinanediol; the aqueous solution was concentrated to dryness and dried in vacuo to afford 1.82 g (4.93 mmol, yield 90%) of the title compound, used without further purification. H NMR (DMSO + D2O+TFA): 3.78 (m, IH); 3.19 (m, 2H); 3.09 (m, IH); 1.71 (m, 2H); 1.70-1.48 (m, 3H); 1.49-1.23 (m, 2H); 0.89 (d, J=5.8 Hz, 3H); 0.88 (d, J=5.8 Hz, 3H).
Example C.3
Synthesis of further intermediates
[00210] Starting from the appropriate intermediate and following either the procedures described in the Example C.l the intermediates reported below were prepared:
(2S, 3R)-2-Amino-3-hydroxybutanamide, N-[(lR)-l-[(3aS, 4S, 6S, 7aR)-hexahydro-3a, 5, 5- trimethyl-4, 6-methano-l ,3, 2-benzodioxaborol-2-yl]-3-methylbutyl]-, hydrochloride salt
Figure imgf000070_0001
Η NMR (DMSO-dg) δH: 8.62 (IH, d, J=5.0 Hz); 8.17 (3H, d, J=3.5); 4.28 (IH, dd, J=8.8, 1.8); 3.78 (IH, m); 3.52 (IH, m); 3.00 (IH, m); 2.28 (IH, m); 2.10 (IH, m); 1.92 (IH, t, J=5.7); 1.84 (IH, m); 1.75-1.62 (2H, m); 1.43 (IH, m); 1.31 (3H, s) 1.25 (3H, s); 1.22 (IH, d, J=10.6); 1.14 (3H, d, J=6.2); 0.88 (3H, d, J=6.4); 0.86 (3H, d, J=6.4); 0.81 (3H, s)
(2S)-2-Amino-5-ureidopentanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4, 6-methano-l ,3, 2-benzodioxaborol-2-yl] -3-methylbutyl] ; hydrochloride salt
Chiral
Figure imgf000071_0001
[00211] 1H NMR (DMSO-de) 8.51(1H, d, J= 5.1Hz); 8.17 (3H, br); 6.1 (IH, br);
4.27 (IH, dd, J=8.6 Hz, 1.8); 3.73 (IH, m); 2.99 (IH, m); 2.94 (2H, t); 2.27 (IH, ); 2.10 (IH, m), 1.92 (IH, t, J=5.5); 1.82 (IH, m); 1.75-1.15 (9H, m); 1.30 (3H, s); 1.23 (3H, m); 0.87 (3H, d, J=6.0); 0.85 (3H, d, J=6.0); 0.80 (3H, s).
(2S)-2-Amino-3-carbamoylpropanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a, 5, 5-trimethyl-4, 6-methano-l, 3, 2-benzodioxaborol-2-yl]-3-methylbutyl]; hydrochloride salt
Chiral
Figure imgf000071_0002
[00212] 1H-NMR (DMSO-d6): 8.46-8.41 (IH, m); 8.06 (3H, bs); 7.67 (IH, s);
7.26 (IH, s); 4.30-4.25 (IH, ); 4.08-4.02 (IH, m); 2.96 (IH, m); 2.60-2.52 (IH, m); 2.36-2.24 (IH, m); 2.20-2.10 (IH, m); 1.95 (IH, t, J=5.5); 1.88-1.83 (IH, m); 1.75-1.60 (2H, m); 1.46-1.36 (IH, m); 1.32 (3H, s); 1.30-1.18 (6H, m); 0.86 (6H, t, J=6.7); 0.82 (3H, s).
2-Aminoacetamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4, 6-methano-l ,3,2-benzodioxaborol-2-yl]-l -methylbutyl] '; Hydrochloride salt Chiral
Figure imgf000072_0001
1H-NMR (DMSO-d6): 8.50 (IH, s ); 8.20 (3H, bs); 4.29 (IH, d, J=7.70 Hz); 3.15 (2H, bs); 3.05 (IH, s); 2.36-2.24 (IH, m) 2.20-2.10 (IH, m); 1.95 (IH, t, J=5.38 Hz); 1.85 (IH, s); 1.75-1.60 (2H, m); 1.50-1.38 (IH, m); 1.35-1.30 (3H, m); 1.28-1.25 (4H, m); 1.24-1.17 (IH, m); 0.86 (6H, t, J=5.94 Hz); 0.84 (3H, s).
Example C4
(2S)-2-Amino-3-[(4-methylbenzoyI)amino]propanamide, N-[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyI-4,6-methano-l,3,2-benzodioxaborol- 2-yI]-3-methylbutyI]-, hydrochloride salt.
Figure imgf000072_0002
[00213] (2S)-2-[(l , 1 -Dimethylethoxycarbonyl)amino]-3-[(4-methylbenzoyl)- amino]-proρanamide, N-[(1R)- 1 -[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]-, Example B.4, (740 mg, 1.65 mmol, 1 eq.), was dissolved in 1,4-dioxane (20 ml). To this solution, HCl 4N in 1,4- dioxane (5 ml, 19.8 mmol, 12 eq.) was added and the solution stirred overnight at r.t.. The solvent was removed under reduced pressure to give 800 mg of a glassy solid (quantitative yield). lH NMR (DMSO-d6) 8.63 (IH, d, J=5.5 Hz); 8.38 (IH, t, J=8.4 Hz);8.34 (3H, br); 7.80 (2H, t, J=8.2); 7.28 (2H, d, J=8.2 Hz); 4.15 (IH, dd, J=8.8, 1.8); 4.02 (IH, br); 3.66 (IH, m); 3.55 (IH, m); 2.99 (IH, m); 2.35 (3H, s); 2.19 (IH, m); 2.06 (IH, m); 1.86 (IH, t, J=5.7); 1.80 (IH, m); 1.64 (2H, m); 1.41 (IH, m); 1.33-1.19 (2H, m); 1.27 (3H, s), 1.21 (3H, s); 1.16 (IH, d, J=10.6); 0.82 (3H, d); 0.80 (3H, d); 0.78 (3H, s). Example C.5
2-S-amino-3-(hexanoyIamino)-propionamide, N-[(lS)-l-[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methyIbutyI]amino]carbonyl], hydrochloride salt.
Figure imgf000073_0001
[00214] 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-
(hexanoylamino)propionamide, N-[(lS)-l-[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 -methylbutyl] amino]carbonyl] , of Example B.5, (450 mg, 0.8 mmol, 1 eq.), was dissolved in 1,4-dioxane (15 ml). To this solution, HCl 4N in 1,4-dioxane (2.45 ml, 0.98 mmol, 12 eq.) was added and the solution stirred overnight at r.t.. The solvent was removed under reduced pressure to give 400 mg of a glassy solid. Yield quantitative. Analytical data: !H NMR (DMSO-de). δH: 8.54 (IH, d, J=5.3 Hz); 8.18 (3H, br); 7.74 (IH, t, J=5.7); 4.29 (IH, dd, J=1.8, 8.8); 3.83 (IH, m); 3.40 (2H, m); 3.00 (IH, m); 2.29 (IH, m); 2.11 (IH, m); 2.08 (2H, t, J=7.5); 1.93 (IH, t, J=5.5); 1.84 (IH, m); 1.75-1.15 (11H, m); 1.32 (3H, s); 1.24 (3H, s); 0.86 (3H, d, J=6.6); 0.84 (3H, d, J=6.6); 0.81 (3H, s).
Example C.6
2-S-amino-3-(4-fluorosulfonylamino)propionamide, N-[(1S)-1-[[(1R)-1-
[(3aS,4S,6S,7aR)-hexahydro~3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-
2-yl]-3-methylbutyl]amino]carbonyl], hydrochloride salt.
Figure imgf000074_0001
[00215] 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(4- fluorosulfonylamino)ρropionamide, N-[(lS)-l-[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl], of Example B.6, (0.7 g, 1.14 mmol, 1 eq.), was dissolved in 1,4-dioxane (20 ml). To this solution, HCl 4N in 1,4-dioxane (3.4 ml, 13.68 mmol, 12 eq.) was added and the solution stirred overnight at r.t.. The solvent was removed under reduced pressure to give 440 mg of a white solid. Yield 71%. Analytical data: 1H NMR (DMSO-de). δH: 8.54 (IH, d, J=5.5 Hz); 8.26 (3H, br); 7.89 (3H, m); 7.48 (3H, t, JM8.8); 4.26 (IH, dd, J=1.3, 8.6); 3.84 (IH, m); 3.06 (2H, m); 2.97 (IH, m); 2.25 (IH, m); 2.03 (IH, m); 1.83 (2H, m); 1.64 (2H, m); 1.42 (IH, m); 1.35-1.15 (3H, m); 1.28 (3H, s); 1.22 (3H, s); 1.11 (IH, d, J=10.8); 0.85 (6H, m); 0.80 (3H, s).
Example C.7
2-S-amino-3-(3,4-dimethoxyphenylacetamido)propionamide, N-[(1S)-1-[[(1R)-1-
[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyI-4,6-methano-l,3,2-benzodioxaboroI-
2-yl]-3-methylbutyl]amino]carbonyl], hydrochloride salt.
Figure imgf000074_0002
[00216] 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(3,4- dimethoxyphenylacetamido)-ρropionamide, N-[(lS)-l-[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl], of Example B.7, (0.3 g, 0.47 mmol, 1 eq.), was dissolved in 1,4-dioxane (20 ml). To this solution, HCl 4N in 1,4-dioxane (1.43 ml, 5.71 mmol, 12 eq.) was added and the solution stirred overnight at r.t.. The solvent was removed under reduced pressure, diethyl ether was added and evaporated to give 230 mg of a white solid. Yield 85%.
Analytical data:
1H NMR (DMSO-de). δH: 8.57 (IH, br); 8.12 (3H, br); 7.91 (IH, t, J=5.7 Hz); 6.86 (2H, m); 6.76 (IH, dd,
J=1.8, 8.2); 4.26 (IH, br d, J=7.3); 3.82 (IH, m); 3.72 (3H, s); 3.71 (3H, s); 3.36 (2H, s);
3.34 (2H, m); 2.99 (IH, m); 2.26 (IH, m); 2.10 (IH, m); 1.92 (IH, t, J=5.3); 1.83 (IH, m); 1.67 (2H, m); 1.45-1.15 ( 3H, m); 1.31 (3H, s); 1.23 (3H, s); 0.86 (3H, d, J=6.6);
0.84 (3H, d, J=6.6); 0.80 (3H, s).
Example C.8
2-S-amino-3-(3-phenyl-ureido)-propionamide, N-[(lS)-l-[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methyIbutyI]amino]carbonyI], hydrochloride salt.
Figure imgf000075_0001
2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionamide, N-[(1S)-1- [[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl], of Example B.8, (0.58 g, 0.1 mmol, 1 eq.), was dissolved in 1,4-dioxane (25 ml). To this solution, HCl 4N in 1,4- dioxane (3 ml, 12.1 mmol, 12 eq.) was added and the solution stirred overnight at r.t.. The solvent was removed under reduced pressure, diethyl ether was added and evaporated to give 0.52 g of desired product. Yield 100%. Analytical data: ]H NMR (DMSO-d6). δH: 8.82 (IH, s); 8.59 (IH, d, J=5.7 Hz); 8.18 (3H, br); 7.40 (2H, d, J= 7.9); 7.22 (2H, t, J= 8.1); 6.90 (IH, t, J=7.3); 6.31 (IH, t, J=5.7); 4.26 (IH, dd, J=1.5, 8.6); 3.89 (IH, m); 3.48 (IH, m); 3.36 (IH, m); 3.01 (IH, m); 2.24 (IH, m); 2.10 (IH, m); 1.92 (IH, t, J=5.3); 1.82 (IH, m); 1.67 (2H, m); 1.50-1.15 (3H, m); 1.31 (3H, s); 1.21 (3H, s); 0.85 (3H, d, J=6.6); 0.84 (3H, d, J=6.6); 0.79 (3H, s).
Example C.9 Synthesis of further compounds [00151] Following the procedures of Examples C.4-C.8, the following compounds can be prepared starting from intermediates of Example B.9.
Figure imgf000076_0001
Figure imgf000077_0002
Example D.l Decanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1 ,3,2-benzodioxaborol-2-yl] -3-methylbutyl] amino] carbonyl] -4- [[imino(nitroamino)methyl]amino]butyl]-
Figure imgf000077_0001
[00217] To a solution of decanoic acid (0.84 g, 4.83 mmol) in anhydrous DMF (30 ml) HATU (1.84 g, 4.83 mmol) and HOAt (0.66 g, 4.83 mmol) were added. After stirring at room temperature for 15 minutes the mixture was cooled at 0°C and N- methylmorpholine (1.33 ml, 12.1 mmol) was added. After further 20 minutes (2S)-2- amino-5-[[imino(nitroamino)methyl]amino]pentanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]- hydrochloride salt of Example C.l (2.2 g, 4.03 mmol) was added. The mixture was allowed to warm to room temperature and stirred for 5 hours, then diluted with ethyl acetate (150 ml), washed with a 2% solution of citric acid (2x 100 ml), 2% solution of NaHCO3 (2x 100 ml), and 2% solution of NaCl (2x 100 ml). The organic phases were dried over sodium sulfate and concentrated. The residue was purified by column chromatography eluting with AcOEt/n-Hexane mixtures from 80/20 to 100/0. The resulting solid was triturated with diethyl ether, collected by filtration and dried under vacuum giving 1.8 g of product (72% yield). M.P. 89-94 °C
El. Anal. Calculated: C 59.99% H 9.26% N 13.54% Found C 59.47% H 9.51% N 13.42%
lH NMR (DMSO-de): 8.82 (IH, d, J=2.7 Hz); 8.53 (IH, br); 7.99 (IH, d, J=8.05); 7.88 (2H, br); 4.33 (IH, m); 4.08 (IH, dd, J=1.6, 8.6); 3.14 (2H, m); 2.56 (IH, m); 2.20 (IH, m); 2.11 (2H, m); 2.01 (IH, m); 1.84 (IH, t, J=5.7); 1.79 (IH, m); 1.74-1.58 (3H, m); 1.57-1.39 (5H, m); 1.32 (IH, d, J=9.9); 1.24 (19H, m); 0.85 (9H, m); 0.80 (3H, s). [00218] Starting with the (2S)-2-amino-5-[[imino(nitroamino)methyl]amino]- pentanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- l,3,2-benzodioxaborol-2-yl]-3-methylbutyl] hydrochloride salt of Example C.l and the appropriate carboxylic acids, further compounds prepared fundamentally in accordance with the above experimental procedures are reported in Table D-l.
Table D-l
Ex # Structure Chemical Name and Analytical Data
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
[00219] Following the above described procedure for Example D.l and using as starting material the (2S)-2-amino-5-[[imino(nitroamino)methyl]amino]pentanamide, N- [(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl] -3 -methylbutyl]- hydrochloride salt of Example C.l and the appropriate carboxylic acids, the compounds reported in Table D-IA are prepared.
Table D-IA
Figure imgf000082_0002
Figure imgf000083_0001
Example D.2 10-(l,3-Dioxo-l,3-dihydro-isoindol-2-yI)-decanamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaboroI- 2-yl]-3-methylbutyl]amino]carbonyI]-4-[[imino(nitroamino)methyl]amino]butyl]-;
Figure imgf000083_0002
[00220] To a solution of 10-(l,3-Dioxo-l,3-dihydro-isoindol-2-yl)-decanoic acid (353 mg, 1.11 mmol), prepared according to Example G.l, in anhydrous dichloromethane (10 ml), N-methylmorpholine was added (122 μl, 1.11 mmol). The mixture was cooled to -15°C, then isobutyl chloroformate (144 μl, 1.11 mmol) was slowly added. After 15 minutes (2S)-2-amino-5- [[imino(nitroamino)methyl]amino]pentanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]- hydrochloride salt of Example C.l (508 mg, 1.01 mmol) and further N- methylmorpholine (122 μl, 1.11 mmol) were added. The reaction mixture was stirred at -15 to 10 °C for 4h, then concentrated to small volume and partitioned between ethyl acetate (20 ml) and water (10 ml). The aqueous phase was further extracted with ethyl acetate (10 ml). The combined organic phases were dried over sodium sulfate and concentrated. The residue was taken up with ethyl acetate (3 ml) and the solution was dropwise added to hexane (120 ml) while stirring at room temperature. The solid was collected by decantation and dried under vacuum (730 mg, 94%). ]H NMR (DMSO-de): 8.81 (IH, d, J=2.7 Hz); 8.52 (IH, br); 7.98 (IH, d, J=8.05); 7.88 (2H, br); 7.85 (4H, m); 4.34 (IH, m); 4.06 (IH, dd, J=7.1); 3.56 (2H, t, J=7.14); 3.14 (2H, m); 2.55 (IH, m); 2.19 (IH, m); 2.10 (2H, t, J=7.14); 2.0 (IH, m); 1.82 (IH, t, J=5.7); 1.78 (IH, m); 1.73-1.35 (10H, m); 1.31 (IH, d, J=9.9); 1.24 (19H, m); 0.84 (9H, m); 0.79 (3H, s). [00221] Further compounds prepared fundamentally in accordance with the above experimental procedures are reported in Table D-2.
Table D-2
Figure imgf000084_0001
Figure imgf000085_0002
[00222] Further compounds prepared according to the above reported procedure in Example D.2 are reported in Table D-2A The compound of Example D.2.6, was prepared starting from 2-aminoacetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a, 5,5-trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 -methylbutyl] - amino]carbonyl]-4-[[imino(nitroamino)methyl]-amino]-butyl], hydrochloride salt of Example D.14. The compounds of example D.2.7 and D.2.8 were prepared from 2- aminoacetamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaboroI-2-yl]-l-methylbutyl]; hydrochloride salt of Example C.3. The compounds of Examples 2.9 and 2.10 were prepared from (2S)-2- amino-5-ureidopentanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- i 4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]; hydrochloride salt of Example C.3
Table D-2A
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0002
Example D.3 H-Cyanoundecanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyI-4,6-methano-l,3,2-benzodioxaborol-2-yI]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]-
Chira]
Figure imgf000087_0001
[00223] PS-Carbodiimide (N-cyclohexylcarbodiimide-N'-propyloxymethyl polystyrene, 769 mg, 1 mmol, loading 1.31 mmol/g) and HOAt (l-Hydroxy-7- azabenzotriazole, 115 mg, 0.85 mmol) were added to a solution of 11-cyanoundecanoic acid (115 mg, 0.54 mmol) in dichloromethane (DCM) (9 mL). After stirring for 10 minutes (2S)-2-amino-5-[[imino(nitroamino)methyl]amino]pentanamide, N-[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutyl]-, hydrochloride salt of Example C.l (251 mg, 0.50 mmol) and DIPEA (0.128 ml, 0.75 mmol) were added. The suspension was shaken overnight at room temperature and then the PS-Carbodiimide was filtered off and washed several times with DCM (4 x 6 mL). [00224] The organic phase was passed through a VARIAN CHEM ELUT cartridge for liquid-liquid extraction pre-conditioned with saturated aqueous NaHCO3 and finally washed with DCM (15 mL). The solvent was evaporated and the crude reaction was purified with normal-phase ISOLUTE SPE-SI column (DCM 9, MeOH 1) to afford 200 mg of the desired compound (yield 61%).
NMR (CDCi3): 7.53 (s, br, 2H); 7.36 (d, br, J=4.7 Hz, IH); 6.88 (d, J=8.2 Hz, IH); 4.46 (m, IH); 4.15 (dd, J=8.5, 1.9 Hz, IH); 3.19 (m, 2H); 2.93 (m, IH); 2.23 (t, J=7.2 Hz,
2H); 2.21 (m, IH); 2.09 (t, J=7.5, 2H); 2.04 (m, IH); 1.88 (t, J=5.4 Hz, IH); 1.77 (m, IH); 1.69 (m, IH); 1.64-1.43 (m, 9H); 1.40-1.26 (m, 4H); 1.26 (s, 3H); 1.244.12 (m, 16H); 0.80 (d, J=6.6, 3H); 0.79 (d, J= 6.6, 3H); 0.73 (s, 3H). LC-MS 659.7, MH+. ESI POS; AQA; spray 4 kV/skimmer: 20V/probe 250 C. [00225] Further compounds prepared fundamentally in accordance with the above experimental procedures are reported in Table D-3.
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
N-[(lS)-
N-
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
[00226] Further compounds prepared according to the above Example D.3 are reported in Table D-3A. Table D-3A
Figure imgf000122_0001
Figure imgf000123_0001
Example D.4 NaphthaIene-2-sulfonamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyI-4,6-methano-l,3,2-benzodioxaboroI-2-yI]-3- methylbutyl] amino] carbonyl] -4-[[imino(nitroamino)methyl] amino] butyl] -.
Chiral
Figure imgf000123_0002
[00227] To a solution of (2S)-2-amino-5-[[imino(nitroamino)methyl]-amino]- pentanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]-, hydrochloride salt of Example C.l (70 mg, 0.14 mmol) in DCM (4 mL), TEA (0.04 mL, 0.31 mmol) and naρhthalene-2- sulfonyl chloride (35.1 mg, 0.16 mmol) were added at room temperature. After stirring overnight a second portion of TEA (0.04 mL, 0.31 mmol) and naphthalene-2-sulfonyl chloride (35.1 mg, 0.16 mmol) was added and the reaction was allowed to stir for a further night. The reaction mixture was then washed with saturated aqueous K2CO3 and the separated organic phase was concentrated to dryness. The reaction crude was purified on SPE-SI normal phase cartridge to afford the title compound (64 mg, yield
NMR (CDC13): 8.42 (s, br, IH); 7.96 (dd, J=7.5, 2.2 Hz, IH); 7.95 (d, J=8.5 Hz, IH); 7.89 (d, br, J=7.9 Hz, IH); 7.81 (dd, J=8.8, 1.9 Hz, IH); 7.68-7.57 (m, 2H); 7.23 (s br, 2H); 6.23 (s br, IH); 6.03 (d, J=8.5 Hz, IH); 4.19 (dd, J=9.1, 2.2 Hz, IH); 3.92 (s, br, IH); 3.31 (m, 2H); 2.97 (m, IH); 2.26 (m, IH); 2.12 (m, IH); 1.93 (t, J=5.7 Hz, IH); 1.90-1.68 (m, 6H); 1.30 (s, 3H); 1.28 (m, IH); 1.25 (s, 3H); 1.06 (m, 4H); 0.79 (s, 3H); 0.58 (d, J=9.4 Hz, 3H); 0.56 (d, J=9.4 Hz, 3H).
LC-MS 657.3, MH+, ESI POS; AQA ; spray 4 kV/skimmer: 20 V/ probe 250 C. [00228] Further compounds prepared fundamentally in accordance with the above experimental procedures are reported in Table D-4.
Table D-4
Figure imgf000124_0001
Figure imgf000125_0001
Example D.4.9 NaphthaIene-2-suIfonamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl].
Figure imgf000126_0001
[00229] Naphthalene-2-sulfonyl chloride (144 mg, 0.637 mmol) was added to a solution of (2S)-amino-(3R)-hydroxy-butyric amide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutyl]amino]-carbonyl] hydrochloride salt, of Example C.3, and NMM (0.175 ml, 1.59 mmol) in anhydrous dichloromethane, while stirring at 0°C under nitrogen. After 6 hours the mixture was allowed to warm to room temperature and stirred overnight. A 10% solution of NaHCO3 (10 ml) was added and the layers were separated. The aqueous phase was further extracted with dichloromethane (5 ml). The organic phases were washed with a 20% solution of NaH2PO4, dried over sodium sulfate and concentrated. The residue was purified by column chromatography (Silica gel, 25 g) eluting with a 1:1 (v/v) mixture of hexane and ethyl acetate. The product was obtained as a white glassy solid (219 mg, 74% yield) but still containing some pinanediol. A sample of that product (160 mg) was triturated with a mixture of diethyl ether (3 ml) and hexane (3 ml) affording the pure product as a white solid (80 mg, 27% yield). M.p. 147- 149°C
JH MR (DMSO-d6): 8.40 (IH, s); 8.28-8.22 (IH, m); 8.11 (IH, d, J=7.7); 8.05 (IH, d, J=8.7); 8.01 (IH, d, J=7.8); 7.81 (IH, dd, J=8.7, 1.7); 7.75 (IH, s br.); 7.72-7.61 (2H, m); 4.84 (IH, s br.); 4.03 (IH, dd, J=8.5, 1,.7); 3.82-3.72 (2H, m); 2.41-2.33 (IH, m); 2.20-2.10 (IH, m); 2.02-1.93 (IH, m); 1.82-1.72 (2H, m); 1.58-1.50 (IH, m); 1.36-1.24 (IH, m); 1.20 (3H, s); 1.18 (3H, s); 0.99 (3H, d, J=6.1); 0.94-0.82 (2H, m); 0.77 (3H, s); 0.63 (3H, d, J=7.1); 0.61 (3H, d, J=7.1).
Example D.5
(2S)-4-[[imino(nitroamino)methyl]amino]-2-[(2-naphthylmethyl)-amino]- pentanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]. Chiral
Figure imgf000127_0001
[00230] A solution of (2S)-2-amino-5-
[[imino(nitroamino)methyl]amino]pentanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]-, hydrochloride salt of Example C.l (88 mg, 0.175 mmol) in MeOH (4 mL) was passed through a ISOLUTE PSA cartridge in order to obtain the starting material as a free base. To a solution of the free base in MeOH (4 mL), 2-naphtaldehyde (45 mg, 0.28 mmol) and NaCNBH3 (18 mg, 0.28 mmol) were added at room temperature; AcOH was added until the pH of the solution was 4-5. The reaction mixture was stirred overnight, then H20 (1 mL) was added and the resulting solution was concentrated; the residue, dissolved in AcOEt, was washed with brine and the organic phase was concentrated to dryness. Purification by silica gel flash chromatography (DCM/MeOH/NH4OH, 97.5/2.5/0.25) of the reaction crude, afforded the desired compound (30 mg, yield 28%). NMR (CDCl3+D2O): 7.81 (m, 3H); 7.71 (s, br, IH); 7.52-7.38 (m, 3H); 4.66 (s, br, IH); 4.27 (dd, J=8.8, 1.9 Hz, IH); 3.91 and 3.83 (ABq, 2H); 3.39-3.11 (m, 3H); 2.30 (m, IH); 2.13 (m, IH); 1.98-1.45 (m, 8H); 1.45 (m, 2H); 1.38 (s, 3H); 1.23 (s, 3H); 1.22 (m, IH); 0.91 (d, J=6.3 Hz, 6H); 0.81 (s, 3H).
LC-MS 607.1, MH+. ESI POS; AQA ; spray 4 kV/skimmer: 20 V/probe 250 C. [00231] Further compounds prepared fundamentally in accordance with the above experimental procedures are reported in Table D-5.
Table D-5
Ex # Structure Chemical Name and Analytical Data
Figure imgf000128_0001
Example D.6 N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyI-4,6-methano- l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]-N'-(l-naphthyI) urea.
Chiral
Figure imgf000129_0001
[00232] To a solution of (2S)-2-amino-5-[[imino(nitroamino)methyl]aminoj- pentanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]-, hydrochloride salt of Example C.l (50 mg, 0.10 mmol) in CH3CN (4 mL), TEA (0.014 mL, 0.10 mmol) and naphthalene- 1- isocyanate (0.014 mL, 0.10 mmol) were added at room temperature. The reaction mixture was stirred for 4 hours and then concentrated to dryness. The residue, dissolved in DCM, was washed with H20: the organic layer was separated and the solvent removed under vacuum. Purification by silica gel flash chromatography (DCM 95, MeOH 5) gave the title compound as a white powder (60 mg, yield 94%). NMR (CDCU): 8.08 (s, br, IH); 7.98 (m, IH); 7.79 (m, 2H); 7.57 (d, J=8.2 Hz, IH) 7.51-7.35 (m, 4H); 7.36 (d, J=7.5 Hz, IH); 7.17 (s, br, IH); 6.67 (d, br, J=6.6 Hz, IH) 4.49 (m, IH); 4.20 (dd, J=8.5, 1.9 Hz, IH); 3.39 (m, IH); 3.20 (m, IH); 3.04 (m, IH) 2.26 (m, IH); 2.08 (m, 2H); 1.93 (t, J=5.6 Hz, IH); 1.89-1.55 (m, 7H); 1.39 (m, IH) 1.32 (s, 3H); 1.31 (m, IH); 1.21 (s, 3H); 1.20 (m, IH); 0.85 (d, J=6.0 Hz, 6H); 0.79 (s, 3H).
LC-MS 636.3, MH+. ESI POS; AQA; spray 4 kV/skimmer: 20 V/probe 250 °C. [00233] Further compounds prepared fundamentally in accordance with the above experimental procedures are reported in Table D-6. Table D-6
Figure imgf000129_0002
Figure imgf000130_0001
Figure imgf000131_0002
Example D.7 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[(E)-3- (naphthaIen-2-yl)prop-2-enoyl]amino]-l-oxopentyI]amino]-3-methyIbutyl]-
Figure imgf000131_0001
[00234] To a suspension of PS-HOBT (l-hydroxybenzotriazole-6- sulfonamidomethyl polystyrene, 277 mg, 0.31 mmol, loading 1.12 mmol/g) in DCM (6 mL) and DMF (0.6 mL), 3-naphthalen-2-yl-acrylic acid (91.2 mg, 0.46 mmol), DIC (Diisopropylcarbodiimide, 0.22 mL, 1.40 mmol) and DIPEA (0.05 mL, 0.19 mmol) were added. The suspension was shaken for 3 hours at room temperature and then the resin was filtered under nitrogen and washed several times with DMF (3x5 mL), DCM (3x5 mL), DMF (3x5 mL) and THF (3x5 mL). The well dried resin was suspended in DCM (6 mL) and DMF (0.6 mL) and [(lR)-l-[[(2S)-2-amino-5- [[imino(nitroamino)methyl]amino]-l-oxopentyl]amino]-3-methylbutyl]-boronic acid hydrochloride salt of Example C.2 (50 mg, 0.14 mmol) and DIPEA (0.06 mL, 0.20 mmol) were added. The reaction mixture was shaken overnight at room temperature. The resin was filtered off and washed with DMF (10 mL) and DCM (2 mL) and the solvent was concentrated to dryness. Purification of the crude compound by ISOLUTE SPE-SI normal phase cartridge (DCM 1, MeOH 1), afforded the title compound (25 mg, yield 35%).
NMR (DMSO+D2O, 343 K): 8.06 (s, IH); 7.95 (d, J=9.0 Hz, IH); 7.94 (m, 2H); 7.72 (d, IH); 7.61 (d, J=14.9 Hz, IH); 7.55 (d, J=9.0 Hz, IH); 7.55 (m, 2H); 6.89 (d, J=14.9 Hz, IH); 4.40 (m, IH); 3.30-3.10 (m, 3H); 1.82 (m, IH); 1.73-1.53 (m, 4H); 1.50-1.32 (m, 2H); 0.87 (d, J=6.1 Hz, 3H); 0.86 (d, J=6.1 Hz, 3H). LC-MS 495.0, [M-18JH+. ESI POS; AQA ; spray 5 kV/skimmer: 15 V/ probe 250 C. [00235] Further compounds prepared fundamentally in accordance with the above experimental procedures are reported in Table D-7.
Table D-7
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
[00236] Further compounds prepared according to the above procedure for Example D.7 are reported in Table D-7A.
Table D-7A
Figure imgf000141_0002
Example D.8 Decanamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3as5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methyIbutyI]amino]carbonyI]-2- hydroxypropyl]- Chiral
Figure imgf000142_0001
[00237] Decanoic acid (220 mg, 1.28 mmol, 1.2eq.) was dissolved in DMF dry
(15 ml) at r.t., TBTU (410 mg, 1.28 mmol, 1.2eq.) was added and the resulting solution was stirred for 10'. The mixture was cooled at 0°-5°C, NMM (0.35 ml, 3.2 mmol, 3eq.) was added and then (2S)-amino-(3R)-hydroxy-butyric amide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutyl]amino]carbonyl] hydrochloride salt, of Example C.3 , (430 mg, 1.067 mmol, leq.) was added. The solution was stirred for 2h, then was poured in water (200ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (20 ml), sodium bicarbonate 2% (20ml), NaCl 2% (25ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated under reduced pressure to give 600 mg of oil that was purified by silica gel chromatography (ethyl acetate / n-hexane 1/1) to give 540 mg of white solid that was suspended overnight in diethyl ether (5 ml) and n-hexane (20 ml). The suspension was filtered to give 110 mg of white solid. Yield 20%.
Analytical data: m.p. 108°-110°C, TLC silica gel (n-hexane / ethyl acetate 1/1 r.f. 0.33). E.A. calculated C (66.91%), H (10.26%), N (5.38%), B (2.08%); found C (66.82%), H (10.61%), N ( 5.35%), B (1.93%).
1H-NMR (DMSO-de) δH: 8.81 (IH, br); 7.68 (IH, d, J=8.80 Hz);4.93 (IH, d, J=5.2); 4.28 (IH, dd, J=8.8, 4.3); 4.05 (IH, dd, J=8.6, 1.8); 3.92 (IH, m); 2.52 (IH, m); 2.20 (IH, m), 2.17 (2H, t, J=7.1); 2.00 (IH, m); 1.83 (IH, t, J=5.8); 1.78 (IH, m); 1.64 (IH, m); 1.62 (IH, m); 1.49 (2H, r l.34 (IH, d, J=10.0); 1.31-1.17 (21H, m); 1.04 (3H, d, J=6.4); 0.91-0.83 (9H, m); 0.81 (3H, s).
[00238] Further compounds prepared according to the above procedure include the following:
Example D.8.1
(2S)-2-[(BenzyloxycarbonyI)amino]-4-methyIpentanamide, N-[(lS,2R)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyI-4,6-methano-l,3,2-benzodioxaborol- 2-yl]-3-methylbutyl]amino]carbonyI]-2-hydroxypropyI]-
Figure imgf000143_0001
[00239] Analytical data: TLC (CHC13 9 / MeOH 1, R.f. 0.63), .p. 38°-40°C,
E.A. calculated C (64.60%), H (8.54%), N (6.85%); found C (62.44%), H (8.24%), N (7.47%).
1H NMR (DMSO-d6) δH: 8.78 (IH, br); 7.82 (IH, d, J=8.60 Hz); 7.52 (IH, d, J=8.1); 7.40-7.27 (6H, m); 5.02 (2H, br s); 5.00 (IH, d, J=5.1); 4.28 (IH, dd, J=8.6, J=4.2); 4.12 (IH, q, J 7.8); 4.05 (IH, dd, J=8.6, J=1.8); 3.94 (IH, m); 2.52 (IH, m); 2.19 (IH, m); 2.01 (IH, m); 1.83 (IH, t, J=5.8); 1.78 (IH, m); 1.74-1.55 (5H, m); 1.46 (2H, m); 1.32 (IH, d, J=10.1); 1.24 (3H, s); 1.22 (3H, s); 1.04 (3H, d, J=6.2); 0.91-0.82 (12H, m); 0.80 (3H, s).
Example D.8.2
10-(1 ,3-dioxo-l ,3-dihyd ro-isoindoI-2-yl)-decanoic -amide-N- [(1 S),(2R)-2-hydroxy , l-[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyI] aminocarbonyU-propyl]-
Figure imgf000143_0002
[00240] Analytical data: TLC (CHC13 9 / MeOH 1 R.f. 0.83), E.A. calculated C
(66.52%), H (8.43%), N (6.37%); found C (66.76%), H (8.48%), N (6.31 1HNMR (DMSO-d6) δH: 8.80 (IH, br); 7.85 (4H, m), 7.67 (IH, d, J=8.80 Hz);4.93 (IH, d, J=5.5), 4.28 (IH, dd, J=8.6, 4.0); 4.04 (IH, dd); 3.92 (IH, m); 3.56 (2H, t, J=8.1); 2.49 (IH, m); 2.23-2.12 (3H, m); 2.00 (IH, m); 1.82 (IH, t, J=6.6); 1.78 (IH, m); 1.73- 1.53 (5H, m); 1.48 (2H, m); 1.33 (IH, d, J=10.1); 1.31-1.17 (20H, m); 1.03 (3H, d, J=6.2); 0.84 (6H, d, J=6.6); 0.80 (3H, s). [00241] Further compounds prepared according to the above procedures for
Example D.8, D.8.1 and D.8.2 are reported in Table D-8.
Table D-8
Ex # Structure Chemical Name
D.8.3 Chemical Name: 4-(Pyridin-3-yl)benzamide, N-[(lS,2R)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3-
Figure imgf000144_0001
methylbutyl]amino]carbonyl]-2-hydroxypropyl]. Analytical Data: 'HNMR (DMSO-d6): 9.02 (IH, s); 8.99 (IH, s); 8.63 (IH, d, J=4.7); 8.22 (IH, d, J=8.4); 8.17 (IH, d, J=8.1); 8.04 (2H, d, J=8.3); 7.89 (2H, d, J=8.3); 7.53 (IH, dd, J=7.8, 4.8); 5.18 (IH, d, J=5.1); 4.53 (IH, dd, J=8.3, 5.1); 4.11-4.01 (2H, m); 2.60-2.53 (IH, m); 2.25-2.15 (IH, m); 2.05-1.97 (IH, m); 1.86-1.75 (2H, m); 1.73- 1.58 (2H, m); 1.37-1.24 (3H, m); 1.25 (3H, s); 1.22 (3H, s); 1.13 (3H, d, J=6.2); 0.85 (6H, d, J=6.4); 0.81 (3H, s). D.8.4 Chemical Name: 2-Pyrazinecarbossamide, N-[(lS,2R)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trirnethyl-4,6- methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl] amino] carbonyl]-2-hy droxypropyl] . D.8.5 Chemical Name: Tridecanamide, N-[(lS,2R)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-
Figure imgf000144_0002
methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]. D.8.6 Chemical Name: 4-Phenylbenzamide, N-[(lS,2R)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3-
Figure imgf000144_0003
methylbutyI]amino]carbonyl]-2-hydroxypropyl]. Analytical Data: Η -NMR (DMSO-d6): 9.04 (IH, bs); 8.18 (IH, d, J=8.5); 8.00 (2H, d, J=8.5); 7.81 (2H, d, J=8.4); 7.77- 7.73 (2H, m); 7.51 (2H, t, J=7.5); 7.43 (IH, t, J=7.3); 5.07 (IH, d, J=6.2); 4.55-4.50 (IH, m); 4.10-4.01 (2H, m); 2.60-2.54 (IH, m); 2.25-2.16 (IH, m); 2.06-1.98 (IH, m); 1.84 (IH, t, J=5.6); 1.82-1.76 (IH, m); 1.74- 1.60 (2H, m); 1.35 (IH, d, J=10); 1.30-1.26 (2H, m); 1.25 (3H, s); 1.22 (3H, s); 1.13 (3H, d, J=6.2); 0.87-0.83 (6H, m); 0.81 (3H, s). D.8.7 Chemical Name: " 1 1 " 2,2-Dimethydecanamide, N-[(lS,2R)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]. Analytical Data: Η -NMR (DMSO-d6): 8.93 (IH, bs); 7.03 (IH, d, J=8.6); 5.06 (IH, d, J=5.9); 4.36-4.31 (IH, m); 4.06- 4.01 (2H, m); 3.99-3.92 (IH, m); 2.24-2.14 (IH, m); 1.90-1.76 (2H, m); 1.70-1.58 (2H, m); 1.50-1.42 (2H, m); 1.38-1.32 (IH, m); 1.28-1.20 (15H, m); 1.19-1.12 (6H, m); 1.12-1.08 (6H, m); 1.03 (3H, d, J=6.3); 0.87- 0.83 (9H, m); 0.81 (3H, s).
D.8.8 Chemical Name: (4-phenoxy)benzamide, N-[(lS,2R)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-
Figure imgf000145_0001
methano- 1 ,3,2-benzodioxaborol-2-yl]-3 - methylbutyl]amino]carbonyl]-2-hydroxyρropyl]. Analytical Data: Η -NMR (DMSO-d6): 9.01 (IH, bs); 8.07 (IH, d, J=8.5); 7.96-7.92 (2H, m); 7.47-7.42 (2H, m); 7.22 (IH, t, J=7.4); 7.11-7.06 (4H, m); 5.04 (IH, d, J=6.2); 4.52- 4.47 (IH, m); 4.10-3.98 (2H, m); 2.60-2.52 (IH, m); 2.24-2.16 (IH, m); 2.08-1.98 (IH, m); 1.86-1.74 (2H, m); 1.62-1.58 (2H, m); 1.35 (IH, t, J=10.0); 1.30-1.24 (2H, m); 1.23 (3H, s); 1.22 (3H, s); 1.10 (3H, d, J=6.3); 0.86-0.84 (6H, m); 0.80 (3H, s).
D.8.9 Chemical Name: 5-Butyl-2-ρyridinecarboxamide, N-[(lS,2R)-l-[[[(lR l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-
Figure imgf000145_0002
methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl].
D.8.10 Chemical Name: 4-propoxybenzamide, N-[(lS,2R)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- ""W ςzx methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]. Analytical Data: Η NMR (DMSO-d6): 9.02 (IH, s); 7.95 (IH, d, J=8.6); 7.87 (2H, d, J=8.8); 7.02 (2H, d, J=8.8); 5.03 (IH, d, J=6.2); 4.49 (IH, dd, J=8.4, 4.9); 4.03-3.98 (4H, m); 2.58-2.50 (IH, m); 2.24-2.15 (IH, ); 2.04-1.97 (IH, m); 1.85-1.59 (7H, m); 1.23 (3H, s); 1.22 (3H, s); 1.18 (2H, t, J=7.1); 1.10 (3H, d, J=6.3); 0.99 (3H, t, J=7.4); 0.85 (3H, d, J=6.4); 0.84 (3H, d, J=6.4); 0.81 (3H, s).
D.8.11 Chemical Name: 3-(3-Pyridyl)benzamide, N-[(lS,2R)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-l,3,2-benzodioxaboroI-2-yI]-3-
Figure imgf000146_0001
methylbutyl]amino]carbonyl]-2-hydroxypropyl]. Analytical Data: Η -NMR (DMSO-d6): 9.05-8.95 (2H, m); 8.63 (IH, dd, J= 1.53 Hz, J= 4.76 Hz); 8.39 (IH, J= 8.51 Hz); 8.25 (IH, m); 8.19-8.14 (IH, m); 7.96-7.90 (2H, m); 7.64 (IH, t, J= 7.74 Hz); 7.57-7.51 (IH, m); 5.053 (IH, d, J= 6.06 Hz); 4.54 (IH, dd, J= 5.36 Hz, J= 8.43 Hz); 4.12- 4.00 (2H, m); 2.61-2.54 (IH, m); 2.25-2.14 (IH, m); 2.05-1.95 (2H, m); 1.82 (IH, t, J= 5.55 Hz); 1.80-1.74 IH, m); 1.73-1.56 (IH, m); 1.34 (IH, d, J= 10.04 Hz); 1.31-1.25 (2H, m); 1.22 (6H, d, J= 9.04 Hz); 1.14 (3H, d, J= 6.33 Hz); 0.87-0.83 (6H, m); 0.79 (3H, bs).
D.8.12 Chemical Name: 6-Phenyl-2-pyridinecarboxamide,N-[(lS,2R)-l-[[[(lR)- l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-
Figure imgf000146_0002
methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]. Analytical Data: Η -NMR (DMSO-d6): 9.20-8.95 (IH, m); 8.76 (IH, d, J=8.55 Hz); 8.26-8.16 (4H, m); 8.12 (IH, t, J= 7.77 Hz); 8.02 (IH, d, J= 7.56 Hz); 7.60-7.47 (4H, m); 5.27 (IH, d, J= 4.97 Hz); 4.50 (IH, dd, J= 4.22 Hz, J= 8.50 Hz); 4.16-4.07 (2H, m); 2.65-2.56 (IH, m); 2.25-2.15 (IH, m); 2.09-1.98 (IH, m); 1.84 (IH, t, J= 5.62 Hz); 1.79- 1.73 (IH, m); 1.73-1.66 (IH, m); 1.66-1.59 (IH, m); 1.40-1.26 (4H, m); 1.23 (7H, d, J= 10.89 Hz); 1.15-1.10 (4H, m); 0.85 (7H, d, J= 6.56 Hz); 0.79 (IH, bs).
D.8.13 Chemical Name: 3-propoxybenzamide, N-[(lS,2R)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]. Analytical Data: Η -NMR (DMSO-d6): 9.05-9.0 (IH, ); 8.11 (IH, d, J= 8.49 Hz); 7.48-7.43 (2H, m); 7.40 (IH, t, J= 7.80 Hz); 7.15-7.10 (IH, m); 5.04 (IH, d, J= 6.26 Hz); 4.49 (IH, dd, J= 5.15, J= 8.43 Hz); 4.10-4.05 (IH, m); 4.05- 4.01 (IH, m); 3.99 (2H, t, J= 6.50 Hz); 2.25-2.15 (IH, m); 2.05-1.96 (IH, m); 1.83 (IH, t, J= 5.56 Hz); 1.81- 1.72 (3H, m); 1.72-1.57 (2H, m); 1.34 (IH, d, J= 10.06 Hz); 1.31-1.25 (2H, m); 1.24 (4H, bs); 1.22 (3H, bs); 1.10 (3H, d, J= 6.31 Hz); 1.02 (3H, t, J= 7.40 Hz); 0.84 (6H, dd, J= 1.84 Hz, J= 6.56 Hz), 0.81 (3H, bs).
D.8.14 Chemical Name: 1 -Bromonaphthalene-2-carboxamide, N-[( 1 S,2R)- 1 - [[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-
Figure imgf000146_0003
methylbutyl]amino]carbonyl]-2-hydroxypropyl].
Figure imgf000147_0001
[00242] The intermediate carboxylic acids for the synthesis of examples D.8.3,
D.8.7, D.8.11, D.8.12 and D.8.13 were prepared according to literature procedures. Compound 2,2-dimethyldecanoic acid was prepared as described by Roth et al. in J Med. Chem. 1992, 35, 1609-1617. Compounds 4-(3-pyridyl)benzoic acid, 3-(3- Pyridyl)benzoic acid and 6-phenyl-2-pyridinecarboxilic acid were prepared according the procedure described by Gong et al. in Synlett, 2000, (6), 829-831. Compound 3- propoxybenzoic acid was prepared according the procedure described by Jones in J. Chem. Soc. 1943, 430-432.
Example D.8.18
2-Pyrazinecarboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]ammo]carbonyl]-2-carbamoylethyl]
Figure imgf000148_0001
[00243] This compound has been prepared essentially according to the above procedures for Example D.8, D.8.1 and D.8.2 starting from (2S)-2-amino-3- carbamoylpropanamide, N-[(1R)- 1 -[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyI-4,6- methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]; hydrochloride salt of Example C.3.
IH-NMR (DMSO-d6): 9.20 (IH, d, J=1.29 Hz); 9.02 (IH, d, J=8.52 Hz); 8.91 (iH, d, J=2.45 Hz); 8.81-8.76 (2H, m); 7.42 (IH, s); 6.95 (IH, s); 5.00-4.80 (IH, m); 4.30-4.08 (IH, m); 2.85-2.72 (IH, m); 2.62-2.56 (2H, m); 2.25-2.15 (IH, m); 2.06-1.98 (IH, m); 1.84 (IH, t, J=5.54 Hz); 1.81-1.76 (IH, m); 1.72-1.58 (2H, m); 1.32-1.26 (IH, m); 1.23 (8H, d, J=5.36 Hz); 0.85-0.79 (9H, m).
Example D.8.19
Decanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-l,3,2-benzodioxaborol-2-yl]-3-methylburyI]amino]carboπyl]-2- carbamoylethyl]
Figure imgf000148_0002
This compound has been prepared essentially according to the above procedures for Example D.8, D.8.1 and D.8.2 starting from (2S)-2-amino-3-carbamoylpropanamide, N- [(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]; hydrochloride salt of Example C.3. Example D.8.20
4-Butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyI-4,6-methano-l ,3,2-benzodioxaborol-2-yI] -3- methyIbutyl]amino]carbonyl]-2-carbamoylethyl] Chiral
Figure imgf000149_0001
[00244] This compound has been prepared essentially according to the above procedures for Example D.8, D.8.1 and D.8.2 starting from (2S)-2-amino-3- carbamoylpropanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-l,3,2-benzodioxaborol-2-yI]-3-methylbutyl]; hydrochloride salt of Example C.3.
Example D.9
Decanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyI-4,6- methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2-[(4- methy lbenzoyI)amino] ethyl] -
Figure imgf000149_0002
[00245] Decanoic acid (330 mg, 1,95 mmol, 1.2 eq.) was dissolved in DMF dry,
(20 ml) and TBTU (620 mg, 1.95 mmol, 1.2 eq.) was added at r.t. under nitrogen . The solution was stirred for 10', cooled at 0°-5°C and NMM (0.53 ml, 4.9 mmol, 3 eq.) and (2S)-2-amino-3-[(4-methylbenzoyl)amino]propanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]-, hydrochloride salt (800 mg, 1.58 mmol, 1 eq.) of Example C.4, were added and the resulting mixture was stirred at r.t. for 3h. The solution was poured in water (200 ml) extracted with ethyl acetate (100 ml), washed with solutions of citric acid 2% (50 ml), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered, evaporated and suspended in diethyl ether (20 ml) for 30'. The suspension was filtered and dried to give 330 mg of white solid. Yield 33%. M.P.: 134°C-136°C, TLC, silica gel, (eluent n-hexane/ethyl acetate, r.f. 0.5). E.A. calculated C (69.33%), H (9.37%), N (6.74%), B (1.73%); found C (%), H (%), N (23%), B (%).
1H NMR (DMSO-de) 8.74 (IH, d, J=3.5 Hz); 8.25 (IH, t, J=5.6);7.95 (IH, d, J=7.9); 7.71 (2H, d, J=8.1); 7.25 (2H, t, J=8.1); 4.59 (IH, m); 4.1 (IH, dd, J=1.8, 8.8); 3.49 (2H, m); 2.59 (IH, m); 2.35 (3H, s); 2.20 (IH, m); 2.09 (IH, t, J=7.3); 2.02 (IH, m); 1.83 (IH, t, J=5.5); 1.78 (IH, m); 1.62 (2H, m); 1.44 (2H, m); 1.36-1.21 (17H, m); 1.25 (3H, s), 1.22 (3H, s); 0.85 (3H, t, J=6.8); 0.80 (9H, m).
Example D.10
2-S-decanoylamino-3-(hexanoylamino)-propionamide, N-[(1S)-1-[[(1R)-1-
[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyI-4,6-methano-l,3,2-benzodioxaboroI-
2-yl]-3-methyIbutyl]amino]carbonyl].
Figure imgf000150_0001
[00246] Decanoic acid (170 mg, 0.98 mmol, 1.2 eq.) was dissolved in DMF dry,
(15 ml) and TBTU (310 mg, 0.98 mmol, 1.2 eq.) was added at r.t. under nitrogen . The solution was stirred for 20', cooled at 0°-5°C and NMM (0.271 ml, 2.46 mmol, 2.5 eq.) and 2-S-amino-3-(hexanoylamino)-propionamide, N-[(1S)-1-[[(1R)-1-
[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutyl]amino]carbonyl], hydrochloride salt, (400 mg, 0.82 mmol, 1 eq.) of Example C.5, were added and the resulting mixture was stirred at r.t. for 3h. The solution was poured in water (150 ml) extracted with ethyl acetate (100 ml), washed with solutions of citric acid 2% (50 ml), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered, evaporated and suspended in ethyl acetate (20 ml) for 30'. The suspension was filtered and dried to give 230 mg of white solid. Yield 47%. Analytical data: m.p. 135°-137°C, TLC silica gel (eluent hexane /ethyl acetate 2/1, R.f.= 0.27). E.A. calculated C (67.64%), H (10.35%), N (6.96%); found C (66.93%), H (10.29%), N (7.14%).
JH NMR (DMSO-de) δH: 8.67 (IH, d, J=2.9 Hz); 7.83 (IH, d, J=8.2); 7.67 (IH, t, J=5.5); 4.41 (IH, m); 4.10 (IH, dd, J=1.5, 8.6); 3.25 (2H, m); 2.56 (IH, m); 2.20 (IH, m); 2.13-1.95 (5H, m); 1.84 (IH, t, J=5.5); 1.78 (IH, m); 1.64 (2H, m); 1.46 (4H, m); 1.35-1.15 (27H, m); 0.84 (9H, m); 0.79 (3H, s).
Example D.ll
2-S-decanoylamino-3-(4-fluorosuIfonyIamino)propionamide, N-[(1S)-1-[[(1R)-1-
[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-
2-yl]-3-methylbutyI] amino] carbonyl] .
Figure imgf000151_0001
[00247] Decanoic acid (160 mg, 0.94 mmol, 1.2 eq.) was dissolved in DMF dry,
(20 ml) and TBTU (300 mg, 0.94 mmol, 1.2 eq.) was added at r.t. under nitrogen . The solution was stirred for 20', cooled at 0°-5°C and NMM (0.259 ml, 2.36 mmol, 2.5 eq.) and 2-S-amino-3-(4-fluorosulfonylamino)propionamide, N-[(1S)-1-[[(1R)-1-
[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutyl]amino]carbonyl], hydrochloride salt, (430 mg, 0.78 mmol, 1 eq.) of Example C.6, were added and the resulting mixture was stirred at r.t. for 2h. The solution was poured in water (200 ml) extracted with ethyl acetate (100 ml), washed with the following solutions: citric acid 2% (50 ml), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered, evaporated and purified by silica gel chromatography (eluent n-hexane/ ethyl acetate 2/1). The solvent was evaporated and n-hexane was added to give 100 mg of solid. Yield 19%.
Analytical data: m.p. 83°-85°C, TLC silica gel (eluent hexane /ethyl acetate 2/1, R.f.= 0.53). ]H NMR (DMSO-de) δH: 8.45 (IH, d, J=3.8 Hz); 7.83 (3H, m); 7.63 (IH, t, J=6.2); 7.42 (2H, t, J= 8.8); 4.40 (IH, m); 4.12 (IH, dd, J=1.5, 8.6); 2.95 (2H, m); 2.64 (IH, m); 2.21 (IH, m); 2.17 (2H, t, J=7.3); 2.01 (IH, m); 1.83(1H, t, J=5.5); 1.78 (IH, m); 1.62 (2H, m); 1.45 (2H, m); 1.4-1.1 (23H, m); 0.87-0.8 (9H, m); 0.79 (3H, s).
Example D.12
2-S-decanoylamino-3-(3,4-dimethoxyphenylacetamido)propionamide, N-[(1S)-1- [[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl].
Figure imgf000152_0001
[00248] Decanoic acid (80 mg, 0.48 mmol, 1.2 eq.) was dissolved in DMF dry,
(20 ml) and TBTU (150 mg, 0.48 mmol, 1.2 eq.) was added at r.t. under nitrogen . The solution was stirred for 20', cooled at 0°-5°C and NMM (0.13 ml, 1.2 mmol, 2.5 eq.) and 2-S-amino-3-(3,4-dimethoxyphenylacetamido)propionamide, N-[(1S)-1-[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutyl]amino]carbonyl], hydrochloride salt, (230 mg, 0.4 mmol, 1 eq.) of Example C.7, were added and the resulting mixture was stirred at r.t. for 2h. The solution was poured in water (200 ml) extracted with ethyl acetate (100 ml), washed with the following solutions: citric acid 2% (50 ml), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered, evaporated and purified by silica gel chromatography (eluent n-hexane/ ethyl acetate 1/1). The solvent was evaporated to give 100 mg of glassy solid. Yield 35.7%. Analytical data: TLC silica gel (eluent hexane /ethyl acetate 1/1, R.f.= 0.53). E.A. calculated C (67.13%), H (9.25%), N (6.02%); found C (65.38%), H (9.20%), N (5.49). 1H NMR (DMSO-d6) δH: 8.65 (IH, d, J=3.5 Hz); 7.84 (2H, m); 6.83 (2H, m); 6.72 (IH, dd, J=1.7, 8.1); 4.43 (IH, m); 4.10 (IH, dd, J=1.8, 8.6); 3.72 (3H, s); 3.70 (3H, s); 3.30 (2H, s); 3.27 (2H, m); 2.58 (IH, m); 2.19 (IH, m); 2.02 (3H, m); 1.84 (IH, t, J=5.5); 1.78 (IH, m); 1.63 (2H, m); 1.43 (2H, m); 1.35-1.15 (23H, m); 0.87-0.8 (9H, m); 0.79 (3H, s).
Example D.13
2-S-decanoylamino-3-(phenylureido)propionamide, N-[(1S)-1-[[(1R)-1-
[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-
2-yI]-3-methylbutyl]amino]carbonyI].
Figure imgf000153_0001
[00249] Decanoic acid (170 mg, 0.99 mmol, 1.2 eq.) was dissolved in DMF dry,
(20 ml) and TBTU (310mg, 0.99 mmol, 1.2 eq.) was added at r.t. under nitrogen . The solution was stirred for 20', cooled at 0°-5°C and NMM (0.27 ml, 2.4 mmol, 2.5 eq.) and 2-S-amino-3-(phenylureido)propionamide, N-[(lS)-l-[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl], hydrochloride salt, (420 mg, 0.82mmol, 1 eq.) of Example C.8, were added and the resulting mixture was stirred at 0°C for 2h. The solution was poured in water (200 ml) extracted with ethyl acetate (100 ml), washed with the following solutions: citric acid 2% (50 ml), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered, evaporated and suspended in diethyl ether (20 ml) for lh, filtered and dried under vacuum to give 140 mg of white solid that was purified by silica gel chromatography (n-hexane/ethyl acetate 1/1). Yield 25%. Analytical data: TLC silica gel (eluent hexane /ethyl acetate 1/1, R.f.= 0.4). lH NMR (DMSO-d6) δH: 8.73 (IH, d, J=3.1 Hz); 8.64 (IH, br s); 7.97 (IH, d, J=8.2); 7.36 (2H, d, J=8.1); 7.19 (2H, t, J=8.1); 6.87 (IH, t, J=8.1); 6.1 (IH, t, J=6.0); 4.44 (IH, m); 4.10 (IH, dd, J=1.8, 8.6); 3.41 (IH, m); 3.22 (IH, m); 2.59 (IH, m); 2.19 (IH, m); 2.10 (2H, t, J=7.3); 2.02 (IH, m); 1.84 (IH, t, J=5.5); 1.78 (IH, m); 1.64 (2H, m); 1.46 (2H, m); 1.35-1.15 (23H, m); 0.87-0.8 (9H, m); 0.79 (3H, s). Example D.14
2-Aminoacetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyI-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyI]amino]butyl], Hydrochloride salt
Figure imgf000154_0001
[00250] To a solution of N-Boc-Glycine (383 mg, 2.18 mmol), in anhydrous dichloromethane (20 ml), N-methylmorpholine was added (275 μl, 2.5 mmol). The mixture was cooled to -15°C, then isobutyl chloroformate (286 μl, 1.2 mmol) was slowly added. After 15 minutes (2S)-2-amino-5-
[[imino(nitroamino)methyl]amino]pentanamide, N-[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]- hydrochloride salt of Example C.l (1.00 g, 2.0 mmol) and further N-methylmorpholine (275 μl, 2.5 mmol) were added. The reaction mixture was stirred at -15°C-10°C for 4h, then concentrated to small volume and partitioned between ethyl acetate (100 ml) and water (50 ml). The aqueous phase was further extracted with ethyl acetate (20 ml). The combined organic phases were dried over sodium sufate and concentrated. The residue was taken up with ethyl acetate (5 ml) and the solution was dropwise added to hexane (120 ml) while stirring at room temperature. The solid was collected by decantation and dried under vacuum (1.18 g, 95%). Part of this Boc-protected intermediate (1.08 g, 1.73 mmol) was dissolved in THF (15 ml), then a 4N solution of HCl in dioxane was added. After stirring for 5 hours at room temperature the mixture was concentrated and the residue was triturated with diethyl ether (50 ml). The resulting white solid was collected by filtration, washed with diethyl ether and dried under vacuum, yelding 856 mg of the title compound (88% yield).
Η NMR (DMSO-d6): 8.76 (IH, d, J=3.1Hz); 8.68 (IH, d, J=8.1); 8.56 (IH, br); 8.06 (3H, m); 7.91 (2H, br); 4.43 (IH, m); 4.14 (IH, dd, J=8.6, 3=1.6); 3.60 (2H, m); 3.15 (2H, br); 2.67 (IH, m); 2.23 (IH, m); 2.04 (IH, m); 1.87 (IH, t, J=5.8); 1.81 (IH, m); 1.75-1.60 (3H, m); 1.52 (3H, m); 1.41-1.28 (3H, m); 1.27 (3H, s); 1.23 (3H, s); 0.86 (3H, d, J=6.4); 0.84 (3H, d, 3=6.4); 0.81 (3H, s).
Example D.l 5
3-Aminopropanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyI]amino]butyl]; hydrochloride salt
Chiral
Figure imgf000155_0001
[00251] To a solution of 3-[[(l,l-dimethylethoxy)carbonyl]amino]propanamide,
N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl- ]amino]butyl]-, of Example D.3.118 (42 mg, 0.075 mmol) in diethyl ether (1.0 ml), cooled at 0°C, a 10% v/v solution of hydrogen chloride in diethyl ether (2 ml) was added. The mixture was stirred for 5 hours while allowing to warm to room temperature. The resulting solid was collected by filtration, washed with diethyl ether (3x3 ml) and dried under vacuum, giving 33 mg of the title compound (76% yield). LC-MS 538.7, MH+. ESI POS; AQA; spray 4 kV/skimmer: 20V/probe 250 C. [00252] Further compounds prepared according to the above Example, starting from the corresponding Boc protected compound of Table D.3, are reported in the following Table D-l 5.
Table D-15
Ex # Structure Chemical Name and Analytical Data
Figure imgf000156_0001
Example D.16 Synthesis of Further Compounds [00253] Following the procedures of Examples D.9-D.13, the following compounds can be prepared by reaction of decanoic acid with the intermediates of Example C.9.
Figure imgf000157_0001
Figure imgf000158_0001
Example D.17 4-Butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaboroI-2-yl]-3- methylbutyl]amino]carbonyl]-2-(aminoethyI)-hydrochloride salt.
Figure imgf000158_0002
[00254] 4-Butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(benzyloxycarbonylamide)ethyl]-, of Example D.l 6.6, (400 mg, 0,62 mmol, leq.), was dissolved in 1,4-dioxane (10 ml) and methanol (5 ml). To this solution, Pd/C 10% (40 mg) and HCl 4N 1,4-dioxane (1.1 eq.) were added. The mixture was hydrogenated at 1 bar. At the end of the reaction, Pd/C was filtered over celite, the solvent removed under reduced pressure to give a white foam. Yield 95%, 320mg. Analytical data:
Η NMR (DMSO-d6): 8.76 (IH, d); 8.55 (IH, d); 8.15 (3H, br s); 7.95 (2H, d); 7.25 (2H, d); 4.8 (IH, m); 4.2 (IH, d); 2.80 (IH, m); 2.62 (2H, t); 2.23 (IH, m); 2.04 (IH, m); 1.87 (IH, t); 1.80 (IH, m); 1.75-1.50 (2H, m), (2H, m); 1.41-1.20 (6H, d), (6H, m); 1.0-0.80 (3H, d); (3H, d); (3H, s), (3H t).
Example D.18
2-S-(4-ButylbenzoyIamino)-3-(2-pyrazinocarbonylamino)-N~[(lS)-l-[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol- 2-yI]-3-methylbutyI]amino]carbonyl].
Figure imgf000159_0001
[00255] 2-Pyrazine carboxylic acid, (76 mg, 0.61 mmol, 1.1 eq.) was dissolved in
DMF dry, (5 ml) and TBTU (200mg, 0.61 mmol, 1.1 eq.) was added at r.t. under nitrogen . The solution was stirred for 15', cooled at 0°-5°C and NMM (0.20 ml, 1.85 mmol, 3.3 eq.) and 4-butylbenzamide,N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a, 5,5-trimethyl-4, 6-methano- 1 ,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-(aminoethyl)-hydrochloride salt, from Example D.17, (310 mg, 0,56 mmol, leq.) were added and the resulting mixture was stirred at 25°C for 4h. The solution was poured in water (100 ml) extracted with ethyl acetate (50 ml), washed with the following solutions: citric acid 2% (50 ml), NaCl 2% (50 ml), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered, evaporated and suspended in diethyl ether- n-hexane for lh, to give a white solid that was filtered and dried under vacuum to give a white powder. Yield 52%. 180 mg. Analytical data: M.p. 70°-72°C. Η NMR (DMSO-d6): 9.20 (IH, s); 9.0 (IH, t); 8.85 (IH, d); 8.8 (IH, d); 8.78 (IH, d); 8.60 (IH, d); 7.82 (2H, d); 7.35 (2H, d); 4.8 (IH, m); 4.1 (IH, d); 3.80 (IH, m); 3.62 (IH, m); 2.82 (IH, b); 2.65 (2H, m); 2.2-2.0 (2H, m); 1.80 (IH, m); 1.75-1.50 (2H, m), (2H, m); 1.41-1.20 (6H, d), (6H, m); 1.0-0.80 (3H, d); (3H, d); (3H, s), (3H t).
Example D.19
4-Butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[4-fluoro-benzenesulfonammide ]ethyl]-
Figure imgf000160_0001
[00256] 4-Butylbenzamide,N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-
3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(benzyloxycarbonylamide)ethyl]-, of Example D.17, (2,75 g, 5,02 mmol, leq.), was dissolved in dry methylene chloride at 0°-5°C. To this solution 4-fluorobenzenesulfonyl chloride (l,07g, 5,52mmol, l,leq.) was added and N- methylmorpholine (NMM) (1,1 lg, ll,04mmol, 2,2 eq.) was added dropwise, after few minutes. The mixture was stirred at 0 -5 °C for 30', then at 10 °C for 1 h. The solvent was removed under reduced pressure, the crude was dissolved in Ethyl acetate and washed with a solution of citric acid 2% (50 ml) then with a solution of sodium bicarbonate 2% (50ml) and a solution of sodium chloride 2% (50 ml). The solution was dried over anhydrous sodium sulfate and the solvent evaporated under reduced pressure. The crude was purified by silica gel chromatography (eluent ethyl acetate / n-hexane 1 / 2), the collected fractions have been evaporated under reduced pressure and the white solid was suspended in diethyl ether, filtered and dried under vacuum to give a white wax. Yield 60%, 2g. Analytical data:
1H NMR (DMSO-d6): 8.60 (IH, d); 8.30 (IH, d); 7.85 (3H, m); 7.8 (2H, d); 7.38 (2H, d); 7.30 (2H, d); 4.62 (IH, m); 4.15 (IH, d); 3.25 (2H, br); 2.61 (3H, m); 2.3-2.0 (IH, m); (IH, m); 1.80 (IH, m); 1.75-1.50 (2H, m), (2H, m); 1.41-1.20 (6H, d), (6H, m); 1.0- 0.80 (3H, d); (3H, d); (3H, s), (3H t).
Example D.20
4-Butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaboroI-2-yI-]-
3-methylbutyl]amino]carbonyl]-2-[(2,5-dimethyI-2H-pyrazole) carbonylamino] ethyl] -
Figure imgf000161_0001
[00257] 4-Butylbenzamide,N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-
3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(benzyIoxycarbonylamide)ethyl]-, of Example D.17, (0,9 g, l,64mmol, leq.), was dissolved in dry dichloromethane (10ml). The resulting solution was cooled to 0° <øT< 5°C and N-methyl-morpholine (0.381g, 3.78mmol, 2.3eq.) was added. To the mixture, l,3-dimethyl-lH-pyrazole-5-carbonyl chloride (Rn [55458-67-8]) ( 0.286mg, l,8mmol, l,leq.) was added. The mixture was stirred for lh, then the temperature was raised to 20°C. The mixture was evaporated under reduced pressure, suspended in ethyl acetate (50ml), twashed with 2% citric acid solution (30ml), 2% sodium bicarbonate (30ml), 2% sodium chloride (30ml). The organic layer was dried over anhydrous sodium sulfate and evaporated under reduced pressure. The crude was purified by silica gel chromatography (eluent Ethyl acetate / n-hexane 8/2). The collected fractions were evaporated to give a white powder, that was suspended in diethyl ether and filtered to give the desired compound. Yield 65%, 650mg. Rf. 0.62. Analytical data: M.p. 62°-64°C.
*H NMR (DMSO-d6): 8.82 (IH, d); 8.40 (2H, m); 7.85 (2H, d); 7.3 (2H, d); 6.5 (IH, s); 4.8 (IH, m); 4.15 (IH, d); 3.9 (3H, s); 3.61 (2H, m); 2.65 (3H, m); 2.25 (IH, m); 2.15 (3H, s); 2.0 (IH, m); 1.80 (IH, m); 1.75-1.50 (4H, m), 1.41-1.20 (5H, m), (6H, m); 0.90 (3H, t); 0.8 (9H, m); Example D.21
4-Butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyI-4,6-methano-l,3,2-benzodioxaborol-2-yl-]-
3-methyIbutyI]amino]carbonyl]-2-(4-methylphenyIuriedosulfonylamino)ethyl]-
Figure imgf000162_0001
[00258] 4-Butylbenzamide,N-[(l S)-l -[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-
3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(benzyloxycarbonylamide)ethyl]-, of Example D.17, (0,7 g, l,27mmol, leq.), was dissolved in dry THF (10ml), the the solution was cooled at 0°-5°C. Triethylamine (0,4 ml, l,8mmol, 2,2 eq.) and (4-methylphenyl)-ureido- sulfonylchloride (0,34g, l,38mmol, l,09eq.) of example G.1X have been added. The suspension was stirred at 25 °C for lh, then was poured in a citric acid 1% solution (30ml) and extracted with Ethyl acetate (50ml). The organic solution was washed with sodium chloride 2% solution, dried over anhydrous sodium sulfate, filtered and evaporated under reduced pressure to give acrude thata was purified by silica gel chromatography (eluent Ethyl acetate / n-hexane 1/1) Rf 0.64. The collected fractions have been evaporated and the oil was coevaporated with diethyl ether to give a white foam. Yield 31%, 280mg. Analytical data: M.p. 115°-120°C.
1HNMR (DMSO-d6): 8.80 (IH, s); 8.40 (IH, d); 7.82 (2H, d); 7.3 (2H, d); 7.25 (2H, d); 7.00 (2H, d); 4.62 (IH, m); 4.15 (IH, d); 2.61 (3H, m); 2.3-2.0 (3H, s); 1.80 (IH, m); 1.75 (2H, m), 1.6 (4H, m), 1.2 (13H, m); 0.9 (3H, s), 0.8 (9H m).
Example D.22
4-Phenoxybenzamide,N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyI-4,6-methano-l,3,2-benzodioxaborol-2-yI]-3- methylbutyI]amino]carbonyl]-2-(3-phenyl-ureido)ethyI]-
Figure imgf000163_0001
[00259] 4-Phenoxybenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-
3 a,5 , 5 -trimethyl-4, 6-methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3 - methylbutyl]amino]carbonyl]-2-(amino)ethyl]- hydrochloride salt, from example D.25.2, (lg, 17mmol, leq,), was dissolved in dry dichloromethane (30ml) and N- methyl-morpholine (0.2g, 18.8 mmol, l.leq.) was added. The solution was cooled at 0°- 5°C and phenylisocyanate (0.22g, 17.7mmol, l.leq.) in dichloromethane (ml) was added. The mixture was stirred for lh at 0°-5°C. The solution was washed with sodium chloride 2% solution (50ml), dried over anhydrous sodium sulfate and evaporated under vacuum. The crude was suspended in diethyl ether (20ml), stirred for 2h, filtered and dried under vacuum at 50°C to give a white powder. Yield 74.3%, 0.84g. Analytical data: M.p. 143°-145°C.
1H NMR (DMSO-d6): 8.9 (IH, d); 8.75 (IH, s); 8.59 (IH, d); 7.95 (2H, d); 7.45 (2H, t); 7.35 (2H, d); 7.2 (3H, m); 7.1 (4H,m); 6.9 (IH, m); 6.25 (IH, t); 4.65 (IH, m); 4.10 (IH, d); 3.65 (IH, m); 3.4 (IH, m); 2.6 (IH, m); 2.2 (IH, m); 2.1 (IH, m); 1.85 (2H, m); 1.65 (2H, m), 1.3 (3H, m); (6H, d); 0.80 (9H, t).
Example D.23
4-Burylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl-]-
3-methyIbutyl]amino]carbonyl]-2-(4-methylphenylsulfonylureido)ethyl]-
Figure imgf000164_0001
[00260] 4-Butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-
3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methyIbutyl]amino]carbonyl]-2-(aminoethyl)-hydrochloride salt, from Example D.17, (560 mg, 1.07 mmol, leq.) was dissolved in dichloromethane dry (20ml), and the solution was cooled at 0°-5°C. N-methyl-morpholine (0.125ml, 1.129mmol, l.leq,); and 4-toluenesulfonylisocyanate (0.22g, 1.12mmol, l.leq,) were added and the mixture was stirred at room temperature for 2h. The mixture was washed with a solution of citric acid 2% (20ml) and a sodium chloride 2% solution (25ml). The organic layer was dried over anhydrous sodium sulfate, filtered and evaporated under reduced pressure. The crude was dissolved in diethyl ether (40ml) and the solvent was evaporated. The crude was suspended in n-hexane (20ml), stirred for lh at room temperature, filtered and dried under vacuum at 50°C to give a white powder. Yield 75.6%, 0.55g. Analytical data: M.p. 168°-170°C.
1H NMR (DMSO-d6): 10.8 (IH, s); 8.75 (IH, d); 8.35 (IH, d); 7.75 (4H, m); 7.35 (5H, m); 6.65 (IH, t); 4.5 (IH, t); 4.1 (IH, d); 3.5 (IH, m); 3.25 (IH, m); 2.65 (3H, m); 2.3 (3H, d); 2.2 (IH, m); 2.1 (IH, m); 1.80 (2H, m); 1.65 (4H, m), 1.3 (12H, m); 0.80 (12H, m).
Example D.24
Synthesis of Further Compounds
[00261] Following the procedures of Examples D.l 8 - D.23, the following compounds can be prepared by reaction of the intermediates of Example D.17 or D.25 with the appropriate commercially available carboxylic acids, acyl halides, sulphonyl halides, isocyanates, sulphonylisocyanates, or with the compounds of Examples G.14,
G.15 and G.16. All the obtained compounds have been charcterized by !H-NMR. Table D-24
Figure imgf000165_0001
D.24.5 Chemical Name: 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l- [(3aS,aS,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl-]- 3-methylbutyl]amino]carbonyl]-2- (propylureido)ethyl]- Analytical Data: !H NMR (DMSO-d6): 8.9 (IH, s); 8.6 (IH, d); 7.8 (2H, d); 7.25 (2H, d); 6.2 (IH, t); 6.05 (IH, t); 4.5 (IH, t); 4.05 (IH, t); 3.4 (IH, m); 2.9
Figure imgf000166_0001
(IH, m); 2.65 (2H, t); 2.2 (IH, m); 2.0 (IH, m); 1.8 (2H, m); 1.65 (4H, m); 1.2 (15H, m), 0.80 (16H, m).
D.24.6 Chemical Name: 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l- [(3aS,aS,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl-]-3- methylbutyl]amino]carbonyl]-2-[(4- methylphenyl)carbonylamino]ethyl]- Analytical Data: Η NMR (DMSO-d6): 8.8 (IH, d); 8.5 (2H, m), 7.9 (2H, d), 7.8 (2H, d);7.3 (2H, d) 7.25 (2H, d); 4.7 (IH, q); 4.1 (IH, d), 3.7-3.4 (2H, m); 2.6 (3H, m), 2.2 (IH, m), 2.0 (IH, m), 1.7-
Figure imgf000166_0002
1.5 (4H, m); 1.4-1.1 (12H, m) 1.1 (IH, t), 0.95 (3H, t), 0.8 (12H, m) . M.p. 150°-152°C.
D.24.7 Chemical Name: 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l- [(3aS,aS,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl-]- 3 -methylbuty l]amino]carbony 1 ]-2- [(1,1- dimethylethoxycarbonyl)amino]ethyl]- Analytical Data: 'H MR (DMSO-d6): 8.8 (IH, s); 8.25 (IH, d); 7.8 (2H, d); 7.3 (2H, d); 6.9 (IH, t); 4.65 (IH, t); 4.1 (IH, d); 2.65 (2H, m); 2.2 (IH,
Figure imgf000166_0003
m); 2.1 (IH, m); 1.8 (2H, m); 1.6 (4H, m); 1.3 (20H, m); 0.9-0.80 (12H, m).
D.24.8 Chemical Name: 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyI- 4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(thien-2- ylcarbonyl)amino]ethyl]- Analytical Data: 'HNMR (DMSO-d6): 8.8 (IH, d); 8.5 (IH, m), 8.0 (IH, t), 7.80 (2H, d); 7.7 (2H, m); 7.3 (2H, d); 7.2 (IH, t); 4.7 (IH, q); 4.1 (IH,
Figure imgf000166_0004
d),2.2 (IH, m), 2.0 (IH, m), 1.9 (IH, t); 1.7- 1.5 (4H, m); 1.4-1.1 (10H, m) 1.1 (IH, t), 0.95 (3H, t), 0.8 (9H, m) . D.24.9 Chemical Name: Decanamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(thien-2- ylcarbonyl)amino]ethyl]- Analytical Data: Η NMR (DMSO-d6): 8.8 (IH, d); 8.5 (IH, m), 8.0 (IH, t), 7.80 (2H, d); 7.7 (2H, m); 7.3 (2H, d); 7.2 (IH, t); 4.7 (IH, q); 4.1 (IH, d), 3.5 (2H, t), 2.9 (IH, m); 2.8 (IH, m); 2.4 (4H,
Figure imgf000167_0001
m);2.2 (IH, m), 2.0 (IH, m), 1.9 (IH, t); 1.7- 1.5 (4H, m); 1.4-1.1 (10H, m) 1.1 (IH, t), 0.95 (3H, t), 0.8 (9H, m) .2.9 (IH, m); 2.8 (IH, m); 2.4 (4H, m); 1.9 (IH, m); 1.85 (IH, m); 1.65 (2H, m); 1.50 (2H, m); 1.35 (IH, m); 0.85 (12H, m). M.p. 110°C.
D.24.10 Chemical Name: 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2- (hexanonylamino)ethyl]- Analytical Data: 'H NMR (DMSO-d6): 8.8 (IH, d); 8.5 (IH, d), 8.0 (IH, t), 7.80 (2H, d); 7.3 (2H, d); 4.7 (IH,
Figure imgf000167_0002
q); 4.1 (IH, d), 3.5 (2H, t), 2.6 (3H, m), 2.2 (IH, m), 2.0 (3H, t), 1.9-1.75 (2H, m); 1.7-1.5 (4H, m); 1.5 (2H,m), 1.4-1.1 (16H, m), 0.95- 0.8 (16H, m)
D.24.11 Chemical Name: 4-butylbenzamide,N-[(lS)-l-[[[(lR)-l- [3aS,aS,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano,l,3,2-benzodioxaborol-2-yl-]- 3-methylbutyl]amino]carbonyl]-2- (cyclopropancarbonylamino)ethyl]- Analytical Data: Η NMR (DMSO-d6): 8.8 (IH, d); 8.5 (IH, d), 8.0 (IH, t), 7.80 (2H, d); 7.3 (2H, d); 4.7 (IH, q); 4.1 (IH, d), 3.5 (2H, t), 2.6 (3H, m), ), 2.2 (IH, m), 2.0 (IH, m), 1.9 (IH, t); 1.7-1.5 (4H,
Figure imgf000167_0003
m); 1.4-1.1 (10H, m) 1.1 (IH, t), 0.95 (3H, t), 0.8 (9H, ), 0.7 (4H, m).
D.24.12 Chemical Name: 4-Butylbenzamide,N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-(3-phenyl- ureido)ethyl]- Analytical Data: 'H NMR (DMSO-d6): 8.9 (IH, m);8.8 (IH, s), 8.5 (IH, s), 7.9 (2H, d), 7.5 (2H, d); 7.4 (2H, d); 7.3 (2H, d), 6.9 (IH, t); 4.7 (IH, q); 4.1
Figure imgf000167_0004
(IH, d), 3.7-3.4 (2H, m); 2.6 (3H, m), 2.2 (IH, m), 2.0 (IH, m), 1.7-1.5 (4H, m); 1.4-1.1 (12H, m) 1.1 (IH, t), 0.95 (3H, t), 0.8 (9H, m) D.24.13 Chemical Name: 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(N-methyl-2- pyrrolylcarbonylamide)ethyl]- Analytical Data: Η NMR (DMSO-d6): 8.9 (IH, d); 8.45 (IH, d), 8.05 (IH, t), 7.8 (2H, d), 7.3 (2H, d); 6.9 (IH, s); 6.7 (IH, t), 5.95 (IH, t); 4.7 (IH, q);
Figure imgf000168_0001
4.1 (IH, d), 3.8 (3H, s); 3.6 (2H, m); 2.6 (3H, m), 2.2 (IH, m), 2.05 (IH, m), 1.8 (4H, m); 1.3 (12H, m) 0.91 (3H, t), 0.8 (9H, m) . M.p. 88°-92°C.
D.24.14 Chemical Name: 4-butylbenzamide,N-[(lS)-l-[[[(lR)-l- [3aS,aS,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano,l,3,2-benzodioxaborol-2-yl-]- 3-methylb tyl]amino]carbonyl]-2-[(3,4- dimethoxyphenyl)acetylamino]ethyl]- Analytical Data: Η NMR (DMSO-d6): 8.8 (IH, m); 8.4 (IH, d), 8.1 (IH, t), 7.9 (2H, d), 7.3 (2H, d), 6.8 (IH, s); 6.6 (2H, t), 4.7 (IH, q); 4.1 (IH, d), 3.7-3.4 (2H, m); 2.6 (3H, m), 2.2 (IH, m), 2.0 (IH, m), 1.7-1.5 (4H, m); 1.4-1.1 (12H, m),
Figure imgf000168_0002
1.1 (IH, t), 0.95 (3H, t), 0.8 (9H, m) .
D.24.15 Chemical Name: 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2- (nicotinonylamino)ethyl]- Analytical Data: !H NMR (DMSO-d6): 9.1 (lH,s) 8.9 (IH, m); 8.7 (IH, t), 8.6 (IH, d), 8.5 (IH, d), 8.1 (IH, d), 7.9 (2H, d), 7.5 (IH, m), 7.3 (2H, d), 4.7 (IH, ; 4.1 (IH, d), 3.7-3.4 (2H, m); 2.6 (3H,
Figure imgf000168_0003
m), 2.2 (IH, m), 2.0 (IH, m), 1.7-1.5 (4H, m); 1.4-1.1 (12H, m) 1.1 (IH, t), 0.95 (3H, t), 0.8 (9H, m) .
D.24.16 Chemical Name: 4-butylbenzamide,N-[(lS)-l-[[[(lR)-l- [3aS,aS,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano,l,3,2-benzodioxaborol-2-yl-]- 3-methylbutyl]amino]carbonyl]-2-[(4- sulfonylamino)benzoylamino]ethyl]- Analytical Data: Η NMR (DMSO-d6): 8.9 (IH, m); 8.7 (IH, t), 8.6 (IH, d), 8.0 (2H, d), 7.9 (2H, d), 7.8 (2H, d) ,7.5 (2H, s), 7.3 (2H, d), 4.7 (IH, q); 4.1 (IH, d), 3.7-3.4 (2H, m); 2.6 (3H, ), 2.2
Figure imgf000168_0004
(IH, m), 2.0 (IH, m), 1.7-1.5 (4H, m); 1.4-1.1 (12H, m) 1.1 (IH, t), 0.95 (3H, t), 0.8 (9H, m) . M.p.l45°-147°C.
Figure imgf000169_0001
Figure imgf000170_0001
(IH, s); 7.25 (2H, 2.2 (4H,
8.4 (1H, m); 7.21 4.7 (IH, t); 2.2 (4H, m); 100°-
(IH, m);
Figure imgf000171_0001
3.1 (2H, m); 2.6 (IH, m); 2.2 (4H, m); 2.0 (IH, m); U (2H, m); 1.65 (2H, m); 1.3 (9H, m); 0.9-0.80 (9H, m). M.p. 90°-93°C.
D.24.30 Chemical Name: 4-phenoxybenzamide, N-[(lS)-l-[[[(lR)-l- [(3aS,aS,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl-]- 3-methylbutyl]amino]carbonyl]-2-[(2,5- dimethyl-2H-pyrazole)carbonylamino]ethyl]- Analytical Data: IH NMR (DMSO-d6): 8.9 (IH, br); 8.55 (IH, d); 8.48 (IH, m); 7.9 (2H, m); 7.48 (2H, m); 7.2 (IH, m); 7.05 (4H, m); 6.55 (IH, s); 4.75
Figure imgf000172_0001
(IH, q) 4.1(1H, d); 3.6 (2H, m); 2.2 (4H, m); 2.1 (3H, s); 2.0 (IH, m); 1.8 (2H, m); 1.65 (2H, m); 1.25 (9H, m); 0.8 (9H, m). M.p. 100°-103°C.
D.24.31 Chemical Name: 4-phenoxybenzamide,N-[( 1 S)- 1 -[[[( 1R)- 1 - [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-(4- phenylbenzoylamino)ethyl]- Analytical Data: IH NMR (DMSO-d6): 8.85 (IH, br); 8.55 (2H, m); 7.9 (4H, d); 7.75 (4H, m); 7.48 (5H, m); 7.2 (IH, t); 7.05 (4H, m); 4.8 (IH, q); 4.1 (IH, d); 3.7 (2H, m); 2.65 (IH, m); 2.2 (IH,
Figure imgf000172_0002
m); 2.0 (IH, m); 1.8 (2H, m); 1.6 (2H, m); 1.25 (9H, m); 0.8 (9H, m). M.p. 150°-152°C.
D.24.32 Chemical Name: 4-butylbenzamide,N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-(4- phenylbenzoylamino)ethyl]- Analytical Data: 1HNMR (DMSO-d6): 8.85 (IH, br); 8.6 (IH, m); 8.5 (IH, d); 7.9 (2H, m); 7.75 (5H, m); 7.5 (2H, t); 7.4 (IH, ); 7.3 (2H, m); 4.8 (IH, q); 4.1 (IH, d); 3.7 (2H, m); 2.6 (3H, m); 2.2 (IH, m); 2.0 (IH, m); 1.8 (2H, m); 1.6 (4H,
Figure imgf000172_0003
m); 1.25 (9H, m); 0.8 (12H, m). M.p. 195°- 198°C. -phenyl -
Figure imgf000174_0001
Example D.25 Synthesis of Further Compounds [00262] Following the procedures of Example D17, the following compounds can be prepared starting from the compounds of Example D.16.8 and D.16.9.
Table D-25
Figure imgf000174_0002
Figure imgf000175_0002
Example D.26 4-Butylbenzamide, N-[(lR)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyI-4,6-methano-l,3,2-benzodioxaborol-2-yI]-3- methylbutyl]amino]carbonyl]-2-[(4-methylbenzoyl)amino]ethyI]-
Figure imgf000175_0001
[00263] Following the same procedures used for the preparation of the compound of Exanple D.17, the intermediate 4-butylbenzamide, N-[(1R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutyl]amino]carbonyl]-2-(aminoethyl)-hydrochloride salt is prepared using D-asparagine as starting material. This latter intermediate is then reacted with 4- methylbenzoic acid following the procedure described in Example D.l 8 to give the title compound.
!H NMR (MeOD-d4): 8.88 (2H, d); 8.45 (2H, m); 7.8 (2H, d); 7.7 (2H, d); 7.35 (2H, rn); 7.25 (2H, d); 4.75 (IH, ); 4.1 (IH, d); 3.8 (IH, m); 3.65 (2H, m); 2.65 (3H, m); 2.2 (lH,m); 2.1 (IH, m); 1.8 (2H, m); 1.6 (4H, m); 1.3-1.1 (2H, m); 0.9-0.80 (14H, m).
Example E.l Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(2- naphthoyl)amino]-l-oxopentyl]amino]-3-methylbutyI]-.
Figure imgf000176_0001
[00264] A mixture of naρhthalene-2-carboxamide, N-[(1S)-1-[[[(1R)-1-
[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutyl]-amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]- of
Example D.1.1 (564 mg, 0.90 mmol), 2-methylpropylboronic acid (222 mg, 2.19 mmol) and 4N hydrogen chloride dioxane solution (225 μl) in a 40:60 heterogeneous mixture of methanol:hexane (10 ml) was stirred at room temperature for 4 hours. Hexane (4 ml) was added, the mixture was stirred for a while, then the hexane layer was removed. Fresh hexane (5 ml) and 2-methylpropylboronic acid (100 mg, 0.99 mmol) were added and the mixture was stirred at room temperature for 3 hours. The hexane layer was removed and the methanol phase was washed with hexane (2 x 5 ml). The residue obtained upon concentration of the methanol phase was purified by silica gel column chromatography eluting with ethyl acetate first, then with 40:40:20 acetone:methanol:hexane mixture. The product was redissolved in a mixture of ethyl acetate (250 ml) and methanol (6 ml) and the organic phase was washed with water (2 x 25 ml), dried over sodium sulfate and concentrated. The residue was dried under vacuum at 80 °C for 3 hours affording the product as a white solid (280 mg, 64% yield). M.p. 170-190°C
1H NMR (DMSO-de): 8.76 (IH, m); 8.51 (2H, br); 8.09-7.09 (5H, m); 7.88 (2H, br); 7.60 (2H, br); 4.67 (IH, m); 3.17 (2H, m); 2.58 (IH, m); 1.81 (2H, m); 1.56 (3H, m); 1.38-1.11 (4H, m); 0.83 (IH, m); 0.81 (IH, m); 0.74 (3H, d, J-6.4); 0.74 (3H, d, J=6.4).
El. Anal. Calculated: C 54.33% H 6.43% N 17.28% B 2.22% Found C 54.87% H 6.64% N 17.00% B 2.12% [00265] Further compounds prepared fundamentally in accordance with the above experimental procedures are reported in Table E-1.
Table E-1
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
[00266] Further compounds prepared according to the above procedure for Example E.l are reported in Table E-1A.
Table E-1A
Figure imgf000181_0001
[00267] Further compounds prepared according to the above procedure for Example E.l are reported in Table E-1B. Table E-1B
Figure imgf000182_0001
[00268] Further compounds prepared according to the above procedure for Example E.l are reported in Table E-IC. starting from the compounds of Example D.8.19 and D.8.20. Table E-IC
Figure imgf000183_0001
[00269] Further compounds prepared according to the above procedure for Example E.l are reported in Table E-ID. starting from the compounds of Example D.2.9 and D.2.10.
Table E-ID
Figure imgf000183_0002
Example E.2
Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-
[(decanoyl)amino]-l-oxopentyl]amino]-3-methyIbutyI]-.
Figure imgf000184_0001
[00270] Decanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]- of Example D.l (77 mg, 0.12 mmol), was dissolved in Et2O (1 mL) and HCl 37% (2 mL) was added carefully at 0°C. The reaction mixture was allowed to warm to room temperature and to shake overnight. The mixture was concentrated to dryness and the residue, dissolved in MeOH (1 mL), was passed through ISOLUTE PSA cartridge, and washed with MeOH. The solvent was evaporated and the reaction crude product was purified with ISOLUTE SPE-DIOL cartridges (DCM:MeOH 1:1) to afford the title compound (19 mg, yield 33%). NMR (DMSO+D2O, 343 K): 4.20 (m, IH); 3.13 (m, 2H); 3.05 (m, IH); 2.10 (t, J=6.2 Hz, 2H); 1-69 (m, IH); 1.53-1.40 (m, 4H); 1.39-1.20 (m, 14H); 0.84 (m, 9H). LC-MS 468.9, MH+. ESI POS; AQA; spray 4kV/skimmer: 20V/probe 250 °C. [00271] Further compounds prepared fundamentally in accordance with the above experimental procedures are reported in Table E-2. ι
Table E-2
Ex # Structure Chemical Name and Analytical Data
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
[00272] Further compounds prepared according to the above procedure for Example E.2 are reported in Table E-2A.
Table E-2A
Figure imgf000195_0002
Figure imgf000196_0001
Example E.3 Boronic acid, [(lR)-l-[[(2S,3R)-3-hydroxy-2-[(4-buty!benzoyI)amino]-l- oxobutyI]amino]-3-methylbutyI].
Figure imgf000197_0001
[00273] A mixture of 4-butylbenzamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutyl]amino]-carbonyl]-2-hydroxypropyl]- of Example D.3.179 (1.38 g, 2.63 mmol), 2-methylpropylboronic acid (0.75 g, 7.37 mmol) and 2N aqueous hydrochloric acid (2 ml) in a heterogeneous mixture of methanol (20 ml) and hexane (20 ml) was stirred at room temperature for 16 hours. The mixture was diluted with methanol (20 ml) and hexane (20 ml) then the hexane layer was removed. Ethyl acetate (50 ml) was added to the methanol layer which was then concentrated. The residue was taken up with ethyl acetate and the mixture was concentrated. This step was repeated (2- 3 times) until an amorphous white solid was obtained. The solid was then triturated with diethyl ether (10-15 ml) and the surnatant was removed by decantation. This step was repeated 4 times. After a further trituration with diethyl ether (15 ml) the white solid was collected by filtration and dried under vacuum at room temperature (0.724 g, 70% yield). >H NMR (MeOH-d4): 7.83 (2H, d, 3=8.2); 7.34 (2H, d, J=8.2); 4.77 (IH, d, J=6.4); 4.36-4.28 (IH, m); 2.77 (IH, t, J=7.6); 2.71 (2H,t, J=7.6); 1.72-1.58 (3H, m); 1.46-1.32 (4H, m); 1.29 (3H, d, J=6.4); 0.97 (3H, t, J=7.34); 0.94 (6H, dd, J=l.l, 6.6)
[00274] Further compounds prepared according to the above procedure for Example E.3 are reported in Table E-3. Table E-3
Figure imgf000197_0002
Figure imgf000198_0001
Figure imgf000199_0001
Example E.4 Boronic acid, [(lR)-l-[[(2S,3R)-3-hydroxy-2-[[4-(3-pyridyl)benzoyl]amino]-l- oxobutyI]amino]-3-methyIbutyl].
Figure imgf000200_0001
[00275] A mixture of 4-(pyridin-3-yl)benzamide, N-[(1S,2R)-1-[[[(1R)-1-
[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yl]-3-methylbutyl]amino]carbonyl]-2-hydroxypropyl]- of Example D.8.3 (155 mg, 0.283 mmol), 2-methylpropylboronic acid (81 mg, 0.793 mmol) and 2N aqueous hydrochloric acid (0.3 ml) in a heterogeneous mixture of methanol (3 ml) and hexane (3 ml) was stirred at room temperature for 24 hours. The hexane layer was removed and the methanolic layer was washed with fresh hexane (about 5 ml). Ethyl acetate (10 ml) was added to the methanol layer which was then concentrated. The residue was taken up with ethyl acetate and the mixture was concentrated. This step was repeated (2-3 times) until an amorphous white solid was obtained. The solid was then triturated with diethyl ether (5 ml) and the surnatant was removed by decantation. This step was repeated. The residue (126 mg) was combined with the product of a similar preparation (140 mg) and dissolved in ethyl acetate (about 40 ml) and a small amount of methanol (2-3 ml). The solution was washed with a mixture of NaCl saturated solution (7 ml) and 10% NaHCO3 (2 ml). The layers were separated and the aqueous phase was further washed with ethyl acetate (2 x 20 ml). The combined organic phases were dried over sodium sulfate and concentrated. The residue was taken up with ethyl acetate (about 20 ml) and the minimum amount of methanol, and then concentrated to small volume (about 5 ml). The resulting white was collected by filtration and dried under vacuum at 50°C (160 mg, 65% overall yield).
1H NMR (MeOH-d4): 8.90 (IH, s); 8.49 (IH, d, J=4.0); 8.20 (IH, d, J=8.1); 8.06 (2H, d, J=8.1); 7.85 (2H, d, J=8.1); 7.58 (IH, t br., J=6.0); 4.80 (IH, d, J=3.9); 4.40-4.29 (IH, m); 2.78 (IH, t, J=7.5); 1.73-1.61 (IH, m); 1.38 (2H, t, J=6.9); 1.31 (3H, d, J=6.3); 0.94 (6H, d, J=6.31). [00276] Further compounds prepared according to the above procedure for
Example E.4 are reported in Table E-4. Table E-4
Figure imgf000201_0001
Example E.5
Boronic acid, [(lR)-l-[[(2S)-3-(2-pyrazincarbonylamino)-2-[(4- butyIbenzoylamino)]-l-oxopropyI]amino]-3-methylbutyl]
Figure imgf000201_0002
2-S-(4-Butylbenzoylamino)-3-(2-pyrazinocarbonylamino)-N-[(lS)-l-[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2- yI]-3-methylbutyl]amino]carbonyι], from example D.18, (120mg, 0.19rnmol, leq.), was dissolved in methanol (2ml), and n-hexane (2ml). The this solution, Isobutylboronic acid ( 60mg, 0.57mmol, 3eq,) and HCl 4N 1,4-dioxane (0.07ml, 0.28mmol, 1.5eq.) have been added. The resulting bifasic mixture was stirred at room temperature for 20h, the n-hexane was removed, the methanolic solution was washed with n-hexane (2ml) and evaporated under reduced pressure. The crude was suspended in diethyl ether/ n-hexane /4ml), stirred at room temperature and filtered , to give a white powder. Yield 65%, 69 mg. Analytical data: M.p. 145°-150°C. 1H NMR (MeOD-d4): 9.3 (IH, s); 8.85 (IH, s); 8.75 (IH, s); 7.8 (2H, d); 7.3 (2H, d); 5.1 (2H, t); 4 (2H, dd); 2.8 (IH, t); 2.75 (2H, t); 1.65 (3H, m); 1.4 (4H, m); 1.0 (3H, t) 0.9 (6H, dd).
[00277] Further compounds prepared according to the above procedure for Example E.5 are reported in Table E-5.
Table E-5
Figure imgf000202_0001
(7H, m); 1.65 (3H,
(IH, d); 5.06 (3H, m), d).
(2H, d); dd); 2.8 m);
-3 - (7H, m); (2H, t); M.p. 92°-
(2H, m); (2H, dd); (4H,
d); 4.8 (2H, t); 2 (3H,
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
(IH,
(2H, t); 6.55 (IH, 2.8 (IH, m); 0.9
(2H, t); d); 7.05 (IH, m);
7.35 (IH, 2.8 m); 1.0
(2H, d); 7.05 (4H, 1.6 (IH,
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Example F.l Decanamide, N-[(lS)-l-[[[(lR)-l-[(4R,5R)-4,5-dicyclohexyl-[l,3,2]dioxaborolan-2- yl]-3-methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]-
Chiral
Figure imgf000210_0002
[00278] To a suspension of boronic acid, [(lR)-l-[[(2S)-5-
[[imino(nitroamino)methyl]amino]-2-[(decanoyl)amino]-l-oxopentyl]amino]-3- methylbutyl]-, (125 mg, 0.26 mmol) obtained as in Example E.2, in a mixture of diethyl ether (0.5 ml) and dichloromethane (1 ml), a few drops of methanol were added until complete dissolution of the solid. (lR,2R)-l,2-dicyclohexyl-l,2-ethanediol (61 mg, 0.26 mmol) was added and the mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated to dryness and the residue was purified by column chromatography (silica gel) eluting with a 50:50 ethyl acetaterrhexane mixture. The product was then triturated with hexane and the solvent was removed by decantation. The trituration was repeated two further times. The product was obtained as a waxy solid (65 mg, 37% yield). M.p. 75-100°C
!H NMR (DMSO-d6): 8.99 (IH, d, J=2.5 Hz); 8.52 (IH, br); 7.98 (IH, d, J=8.05); 7.88 (2H, br); 3.48 (2H, d, J=5.7); 3.14 (2H, m); 2.55 (IH, m); 2.19 (IH, m); 2.10 (2H, m); 1.79 (2H, m); 1.74-1.35 (16H, m); 1.24 (22H, m); 1.12 (5H, m); 0.89 (4H, m); 0.84 (9H, m).
Example F.2
4-Phenylbutanamide, N-[(lS)-l-[[[(lR)-l-[13,15-dioxa-14-bora-dispiro[5.0.5.3]- pentadee-14-yI]-3-methylbutyI]amino]carbonyl]-4-[[imino(nitroaιnino)methyl]- aminojbutyl]-
Figure imgf000211_0001
[00279] The title compound was prepared according to the above procedure for
Example F.l using the appropriate boronic acid starting material and bicyclohexyl-1,1'- diol. Analytical results: lH NMR (DMSO-de): 8.79 (IH, d, J=2.5 Hz); 8.52 (IH, br); 8.00 (IH, d, J=7.94); 7.85 (2H, br); 7.31-7.23 (2H, m); 7.20-7.14 (3H, m); 4.40-4.30 (IH, m); 3.15 (2H, m); 2.55 (3H, m); 2.14 (2H, t, J=7.3 Hz); 1.78 (2H, q, J=7.3 Hz); 1.70-0.97 (27H, m); 0.84 (3H, t, J=6.7 Hz); 0.83 (3H, t, J=6.7 Hz).
Example F.2.1
4-ButyIbenzamide, N-[(lS,2R)-l-[[[(lR)-l-[13,15-dioxa-14-bora- dispiro[5.0.5.3]pentadec-14-yl]-3-methylbutyl]amino]carbonyl]-2-hydroxypropyI]-.
Figure imgf000212_0001
[00280] The title compound was prepared according to the above procedure for
Example F.l using the appropriate boronic acid starting material and bicyclohexyl-1,1'- diol.
Analytical results: !H MR (DMSO-d6): 8.98 (IH, s br.); 8.00 (IH, d, J^8.5); 7.81 (2H, d, J=8.2); 7.31 (2H, d, J=8.2); 5.03 (IH, d, J=6.2); 4.49 (IH , dd, J=8.5, 5.0); 4.07-3.98
(IH, m); 2.64 (IH, t, J=7.6); 2.57-2.50 (IH, m); 1.65-1.21 (21H, m); 1.14-1.00 (9H, m);
0.90 (3H, t, J=7.4); 0.85 (6H, d, 6.5).
Example G.l 10-(l,3-Dioxo-l,3-dihydro-isoindol-2-yl)-decanoic acid.
Figure imgf000212_0002
Step 1: 2-undec-l 0-enyl-l ,3-dioxo-l ,3 -dihydro isoindole
[00281] To a mixture of 10-undecen-l-ol (4.23 g, 24.8 mmol), phthalimide (3.65 g, 24.8 mmol) and triphenylphosphme (6.51 g, 24.8 mmol) in anhydrous tetrahydrofuran (30 ml), a solution of DEAD (3.9 ml, 24.8 mmol) in anhydrous tetrahydrofuran (10 ml) was slowly added while keeping the temperature below 8-10°C. After 2 hours further DEAD (1.0 ml, 6.37 mmol) and triphenylphosphine (1.3 g, 4.96 mmol) were added and the mixture was stirred at room temperature overnight. The reaction mixture was concentrated and the residue was triturated with diethyl ether (50 ml). The solid was removed by filtration and washed with diethyl ether (2 x 50 ml). The combined filtrates were concentrated and the residue was triturated with hexane (50 ml) at 40°C. The resulting solid was removed by filtration and washed with hexane (2 x 50 ml). The combined filtrates were concentrated and the residue was purified by column chromatography eluting with 10:2 hexane -.ethyl acetate mixture. The product was obtained as a low-melting white solid (4.9 g, 66% yield). M.p. 25-30°C
1H NMR (DMSO-de) 7.83 (4H, m); 5.76 (IH, m); 4.96 (IH, dq, J= 17.2, 1.6 Hz); 4.90 (IH, ddt, J= 10.2, 2.2, 1.1); 3.54 (2H, t, J=7.1), 1.97 (2H, q, J= 6.7); 1.56 (2H, m); 1.35- 1.15 (14H, m).
Step 2: 10-(l ,3-Dioxo-l ,3-dihydroisoindol-2-yl)decanoic acid.
[00282] A solution of 2-undec-10-enyI-l,3-dioxo-l,3-dihydroisoindole (2 g, 6.68 mmol) of Step 1 and Aliquat® 336 (0.2 g) in a mixture of hexane (20 ml) and acetic acid (6 ml) was added dropwise to a solution of potassium permanganate (2.76 g, 20 mmol) in water (28 ml) while cooling at 0°C. The reaction mixture was stirred at room temperature for 7 hours, then an aqueous solution of sodium bisulfite was added until disappearance of the purple colour. The mixture was then extracted with ethyl acetate and the organic phase was dried over sodium sulfate and concentrated. The residue was purified by silica gel column chromatography eluting with 2:1 hexane:ethyl acetate mixture. The product was obtained as a white solid (1.29 g, 61% yield). M.p. 58-60°C
1H NMR (DMSO-d6) 11.95 (IH, br); 7.85 (4H, m); 3.55 (2H, t, J=7.2 Hz); 2.17 (2H, t, J=7.2 Hz); 1.7-1.4 (4H, m); 1.22 (10H, m).
Example G.2
6-(Benzenesulfonylamino)hexanoic cid
[00283] Benzenesulfonyl chloride (2.5 ml, 19 mmol) was added to a solution of
6-aminohexanoic acid (1 g, 7.62 mmol) in 2N NaOH (22 ml) and dioxane (3 ml), while stirring at 0°C-5°C. The mixture was allowed to warm to room temperature and stirred for 1 hour. The reaction mixture was washed with ethyl acetate (50 ml), then acidified to pH 2 with 37% hydrochloric acid and extracted with ethyl acetate (2 x 50 ml). The combined organic layers were dried over sodium sulfate and concentrated. The residue was triturated with hexane. The solid was collected by filtration and dried under vacuum at 50°C affording 1.1 g of the title compound (55% yield). M.p. 113-115°C
1H NMR (DMSO-de): 11.96 (IH, s); 7.79 (2H, m); 7.60 (4H, m); 2.71 (2H, m); 2.13 (2H, t, J= 7.14Hz); 1.38 (4H, m); 1.21 (2H, m).
Example G.3
6-(EthylsuIfonylamino)he\anoic acid
[00284] A solution of ethanesulfonyl chloride (3.9 ml, 41.1 mmol) in dioxane (10 ml) was added to a solution of 6-aminohexanoic acid (2 g, 15.2 mmol) in IN NaOH (56 ml) and dioxane (10 ml), while stirring at 0°C-5°C. The pH of the reaction mixture was adjusted to 8-9 by addition of 25% sodium hydroxide solution. The mixture was allowed to warm to room temperature and stirred for 30 minutes. Further 25% NaOH solution was added to adjust the pH to about 11. After 3.5h IN hydrochloric acid (15 ml) and ethyl acetate (60 ml) were added. The organic layer was dried over sodium sulfate and concentrated. The residue was triturated with a mixture of diethyl ether (5 ml) and hexane (15 ml). The solid was collected by filtration and dried affording 1.3 g of the title compound (40% yield).
!H NMR (DMSO-d6): 11.9 (IH, s); 6.97 (IH, t, J=5.7 Hz); 2.97 (2H, q, J=7.1); 2.88 (2H, q, J=6.6); 2.2 (2H, t, J=7.3); 1.47 (4H, m); 1.29 (2H, m); 1.18 (3H, t, J=7.3).
Example G.4
8-(EthyIsuIfonylam o)octanoic acid
[00285] A solution of ethanesulfonyl chloride (1.5 ml, 15.7 mmol) in dioxane
(5ml) was added to a solution of 8-aminooctanoic acid (1 g, 6.28 mmol) in IN NaOH (22 ml) and dioxane (5 ml), while stirring at 0°C-5°C. The mixture was allowed to warm to room temperature and stirred for 3.5 minutes. During this period, at 1 hour intervals, the pH was adjusted to 7-8 by addition of 25% NaOH solution. The reaction mixture was washed with diethyl ether (30 ml). The pH was adjusted to 1-2 by addition of IN HCl and the mixture was extracted with ethyl acetate (70 ml). The organic layer was dried over sodium sulfate and concentrated. The residue was triturated with a mixture of diethyl ether. The solid was collected by filtration and dried under vacuum affording 600 mg of the title compound (38% yield).
1H NMR (DMSO-de): 11.9 (IH, s); 6.96 (IH, t, J=6 Hz); 2.96 (2H, q, J=7.1); 2.88 (2H, q, J=6.6); 2.2 (2H, t, J=7.3); 1.45 (4H, m); 1.26 (6H, m); 1.18 (3H, t, J=7.3).
Example G.5
3-Amino-2-S-[(l,l-dimethylethoxycarbonyl)amino]-propionic acid, benzyl ester.
Figure imgf000215_0001
Step 1: N-tert-butoxycarbonyl-L-asparagine [Commercially available] o ^Λ
[00286] L-asparagine (15 g, 0.113 mol, leq.) and sodium carbonate (12 g, 0.113 mol) were dissolved in water (225 ml) and 1,4-dioxane (225 ml) at r.t.. To this solution, di-tert-butyl-dicarbonate (30 g, 0.137 mol, 1.2 eq.) was added and the mixture was stirred overnight. The solvent was evaporated under reduced pressure till 1,4-dioxane was distilled and the pH adjusted to 2 with HCl 37% to give a white solid that was filtered, washed with water and dried. Yield 91%. 24g. Analytical data: m.p. 175°C-180°C (lit. 175°C).
!H NMR (DMSO-de) 12.5 (IH, br); 7.31 (IH, br);6.91 (IH, br); 6.87 (IH, d, J=8.4 Hz); 4.23 (IH, q, J=7.7 Hz); 2.56-2.36 (2H, m); 1.38 (9H, s).
Step 2: N-[(l,l-dimethylethoxycarbonyl)amino] -L-asparagine, benzyl ester.
Figure imgf000215_0002
[00287] The compound was prepared according to Bioorg.Med.Chem.,
6(1998)1185-1208. N-[(l,l-dimethylethoxycarbonyl)amino]-L-asparagine (20.7 g, 89.1 mmol, 1 eq.), of Step 1, was dissolved in methanol (500 ml) and cesium carbonate (15.97 g, 49 mmol, 0.55 eq.) was added. The solvent was evaporated to give a white solid that was dissolved in N,N-dimethylformamide (200 ml). To the suspension, benzyl bromide (11.6 ml, 98 mmol, 1.1 eq.) was added dropwise and the mixture was stirred overnight. The solvent was reduced under reduced pressure, water (300 ml) was added and the mixture extracted with ethyl acetate (200 ml), washed with brine (50ml) and the solvent removed under reduced pressure to give a crude that was suspended in n-hexane (160 ml), filtered and dried under vacuum to give 14.68 g of white solid. Yield 51%. Analytical data: m.p. 1130-115°C.
1H NMR (DMSO-de) 7.35 (6H, m); 7.13 (IH, d, J=7.9 Hz);6.94 (IH, br s); 5.10 (2H, s); 4.39 (IH, q, J=7.4 Hz); 2.6-2.4 (2H, m); 2.03 (2H, t, J=7.3); 1.37 (9H, s).
Step 3: 3-Amino-2-S-[(l,l-dimethylethoxycarbonyl)amino]-propionic acid, benzyl ester.
Figure imgf000216_0001
[00288] N-[(l,l-dimethylethoxycarbonyl)amino]-L-asparagine, benzyl ester, (2 g,
6.3 mmol, 1 eq.), of Step 2, was dissolved in acetonitrile (80 ml) and water (80 ml). The solution was cooled to 0°-5°C and iodobenzene diacetate (3 g, 9.3 mmol, 1.5 eq.) was added portionwise. The mixture was stirred at 0°C for 30', then at r.t. for 4h. The organic solvent was removed under vacuum, diethyl ether and HCl IN were added. The acqueous layer was separated and extracted with dichloromethane (100ml) and sodium bicarbonate (3.5g). The organic solvent was dried over sodium sulphate anhydrous, evaporated under reduced pressure to give 0.65g of colourless oil. Yield 36% Analytical data:
1H NMR (DMSO-d6) 7.45-7.20 (7H, m); 7.20 (IH, d, J=7.7 Hz); 5.13 (2H, AB q, J= 12.8); 4.01 (IH, m); 2.80 (2H, m); 1.38 (9H, s).
Example G.6 (2S)-2-[(l,l-dimethylethoxycarbonyI)amino]-3-[(4-methylbenzoyl)amino]propanoic acid.
Figure imgf000217_0001
Step 1 : 2-S-[(l, l-dimethylethoxycarbonyl)amino]-3-(4-methylbenzoylamino)propionic acid, benzyl ester.
Figure imgf000217_0002
[00289] 3-Amino-2-S-[(l,l-dimethylethoxycarbonyl)amino]-propionic acid, benzyl ester, (690 mg, 2.34 mmol, leq.), of Example G.5, was dissolved in DMF dry (20 ml) and TBTU (900 mg, 2.98mmol, 1.2eq.) was added. The mixture was stirred at r.t. for 10', cooled to 0°-5°C with ice bath and NMM (0.51 ml, 4.68 mmol, 2 eq.) and 4- methyl benzoic acid (380 mg, 2.81 mmol, 1.2 eq.) were added. The mixture was stirred at r.t. for 3h, poured in water (100 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with a solution of citric acid 2% (50ml), sodium bicarbonate 2% (50ml), NaCl 2% (50 ml), dried over sodium sulphate anhydrous and evaporated at reduced pressure to give lg of oil. Yield quantitative. Analytical data:
1H NMR (DMSO-d6) 8.46 (IH, br t, J=5.7 Hz ); 7.70 (2H, d, J=8.0); 7.35-7.2 (8H, m); 5.07 (2H, s); 4.29 (IH, m); 3.67 (IH, m); 3.58 (IH, m); 2.36(3H, s); 1.37 (9H, s).
Step 2:(2S)-2-[(l,l-dimethylethoxycarbonyl)amino]-3-(4- methylbenzoylaminojpropionic acid.
Figure imgf000217_0003
[00290] 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(4-methylbenzoylamino)- propionic acid, benzyl ester, (930 mg, 2.25 mmol), of Step 1, was dissolved in methanol (25ml) and Pd/C 10% (90 mg) was added. The mixture was hydrogenated at athmospheric pressure for lh. Pd/C was filtered and the solution was evaporated under reduced pressure to give 650 mg of white foam. Yield 86%. Analytical data: !H NMR (DMSO-d6): 12.5 (IH, br); 8.40 (IH, t, J=5.7 Hz); 7.71 (2H, d, J=8.05 Hz), 7.27 (2H, d, J=8.05 Hz);7.09 (IH, d, J=7.9), 4.17 (IH, m); 3.57 (2H, m); 2.35 (3H, s); 1.37 (9H, m).
Example G.7 2-S-[(l,l-dimethylethoxycarbonyl)ammo]-3-(hexanoyIamino)propionic acid.
Figure imgf000218_0001
Stepl : 2-S-[(l, l-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid, benzyl ester.
Figure imgf000218_0002
[00291] Hexanoie acid (450 mg, 3.87 mmol, 1.2 eq.) was dissolved in DMF dry
(15 ml) and TBTU (1.24 g, 3.87 mmol, 1.2eq.) was added, the mixture was stirred at r.t. for 20', then was cooled to 0°-5°C with ice bath. 3-amino-2-S-[(l,l- dimethylethoxycarbonyl)amino]propionic acid, benzyl ester, (950 mg, 3.22 mmol, leq.), of Example G.5, and NMM (1.06 ml, 9.61 mmol, 2.5 eq.) were added. The mixture was stirred at r.t. overnight, poured in water (150 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with a solution of citric acid 2% (50ml), sodium bicarbonate 2% (50ml), NaCl 2% (50 ml), dried over sodium sulphate anhydrous and evaporated at reduced pressure to give a crude that was purified by silica gel column chromatography (eluent: n-hexane/ethyl acetate 2/1, R.f = 0.52) 0.5g of colourless oil. Yield 40%. Analytical data: 1H NMR (DMSO-de). δH: 7.87 (IH, br t, J=6.2 Hz); 7.35 (5H, m); 7.14 (IH, d, J= 8.2); 5.07 (2H, s); 4.14 (1H, m); 3.37 (2H, m); 2.00 (2H, t, J=7.1); 1.43 (2H, m); 1.36 (9H, s);1.3-l.l (4H, m); 0.83 (3H, t, J=7.1 Hz)
Step 2: 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid.
Figure imgf000219_0001
[00292] 2-S-[(l , 1 -dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid, benzyl ester (500 mg, 1.27 mmol), of Step 1, was dissolved in methanol (15 ml) and Pd/C 10% (50 mg) was added. The mixture was hydrogenated at athmospheric pressure for lh. Pd/C was filtered and the solution was evaporated under reduced pressure to give 300 mg of white solid. Yield 78%. Analytical data: m.p. 123°-125°C. 'HNMR (DMSO-dg). δH: 12.6 (IH, br); 7.84 (IH, br t); 6.87 (IH, d, J= 7.5 Hz); 4.00 (IH, m); 3.32 (2H, m); 2.04 (2H, t, J=7.5); 1.47 (2H, m); 1.38 (9H, s);1.3-l.l (4H, m); 0.85 (3H, t, J=7.1 Hz)
Example G.8 2-S-tert-butoxycarbonylamino-3-(4-fluorosulfonylamino)propionic acid.
Figure imgf000219_0002
Step 1: 2-S-[(l, 1-dimethylethoxycarbonyl) amino] -3-(4-fluorosulfonylamino)propionic acid, benzyl ester.
Figure imgf000219_0003
[00293] 3-Amino-2-S-[(l, l-dimethylethoxycarbonyl)amino]propionic acid, benzyl ester (1.25 g, 4.24 mmol, leq.), of Example G.5, was dissolved in dichloromethane dry (20 ml) and the solution was cooled to 0°-5°C, under nitrogen.
TEA (0.65 ml, 4.67 mmol., l.leq.) and 4-fluoro-sulfonylchloride (0.9 g, 4.67 mmol., l.leq.) in dichloromethane dry (10ml) were added. The mixture was stirred at r.t. for lh, evaporated under reduced pressure and diethyl ether (25 ml) was added and a white solid was obtained that was filtered and dried under vacuum to give 1.89g of product.
Yield 99%.
Analytical data: m.p. 105°-107°C. TLC silica gel (eluent: n-hexane/ethyl acetate 1/1, R.f
= 0.55).
!H NMR (DMSO-de). δH: 7.91 (IH, t, J=6.2 Hz); 7.85 (2H, dd, J=5.3, 8.8); 7.43 (2H, t, J=8.8); 7.35 (5H, m);
7.15 (IH, d, J=8.2); 5.09 (2H, s); 4.14 (IH, m); 3.10 (2H, m); 1.36 (9H, s).
Step 2: 2-S-[(l, l-dimethylethoxycarbonyl)amino]-3-(4-fluorosulfonylamino)propionic acid.
Figure imgf000220_0001
[00294] 2-S-[(l , 1 -dimethylethoxycarbonyl)amino]-3-(4- fluorosulfonylamino)propionic acid, benzyl ester (1.8 g, 3.98 mmol.), of Step 1, was dissolved in methanol (30 ml) and Pd/C 10% (180 mg) was added. The mixture was hydrogenated at athmospheric pressure for lh. Pd/C was filtered and the solution was evaporated under reduced pressure to give 1.39 g of colourless oil. Yield 97%.
Analytical data:
1H NMR (DMSO-de). δH: 12.7 (IH, br); 7.83 (2H, dd, J=5.3, 8.8); 7.78 (IH, br t, J=5.5); 7.42 (2H, t, J=8.8);
6.87 (IH, d, J=8.6); 3.99 (IH, m); 3.03 (2H, m); 1.36 (9H, s).
Example G.9 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(3,4-dimethoxyphenylacetamido)- propionic acid.
Figure imgf000221_0001
Step 1: 2-S-[(l, l-dimethylethoxycarbonyl)amino]-3-(3, 4-dimethoxyphenylacetamido)- propionic acid, benzyl ester.
Figure imgf000221_0002
[00295] 3,4-Dimethoxy-phenylacetic acid (720 mg, 3.66 mmol, 1.2 eq.) was dissolved in DMF dry (20 ml) and TBTU (1.17 g, 3.66 mmol, 1.2eq.) was added, the mixture was stirred at r.t. for 20', then was cooled to 0°-5°C with ice bath. 3-amino-2-S- tert-butoxycarbonylamino-propionic acid, benzyl ester (0.9 g, 3.05 mmol, leq.), of Example G.5, and NMM (1.0 ml, 9.15 mmol, 2.5 eq.) were added. The mixture was stirred at 0°C for 2h, then poured in water (200 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (20ml), sodium bicarbonate 2% (20ml), NaCl 2% (20 ml), dried over sodium sulphate anhydrous and evaporated at reduced pressure to give a crude that was purified by silica gel chromatography (eluent: n-hexane/ethyl acetate 1/1, R.f = 0.57) to give 1 g of colourless oil. Yield 69%.
Analytical data: 1H NMR (DMSO-de). δH: 8.02 (IH, t, J=5.7 Hz); 7.34 (5H, m); 7.17 (IH, d, J=7.7); 6.82 (2H, m); 6.71 (IH, dd, J=1.5, 8.2); 5.03 (2H, s); 4.14 (IH, m); 3.71 (3H, s); 3.69 (3H, s); 3.39 (2H, m); 1.36 (9H, s).
Step 2: 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(3,4-dimethoxyphenylacetamido)- propionic acid.
Figure imgf000222_0001
[00296] 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(3,4- dimethoxyphenylacetamido)-ρropionic acid, benzyl ester (1 g, 2.1 mmol.), of Step 1, was dissolved in methanol (30 ml) and Pd/C 10% (10 mg) was added. The mixture was hydrogenated at athmospheric pressure for lh. Pd/C was filtered and the solution was evaporated under reduced pressure to give 0.73 g of white foam. Yield 91%. Analytical data: Η NMR (DMSO-d6). δH: 12.7 (IH, br); 8.06 (IH, t, J=5.9 Hz); 7.00 (lH,d, J= 8.05); 6.91 (2H, m); 6.80 (IH, dd, J=1.55 8.4); 4.08 (IH, m); 3.80 (3H, s); 3.78 (3H, s); 3.5-3.3 (2H, m); 1.36 (9H, s).
Example G.10 2-[(l,l-dimethyIethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid.
Figure imgf000222_0002
Step 1: 2-[(l,l-dimethy/ethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid, benzyl ester.
Figure imgf000222_0003
[00297] 3-Amino-2-S-[(l,l-dimethylethoxycarbonyl)amino]propionic acid, benzyl ester (1.14 g, 3.87 mmol, leq.), of Example G.5, was dissolved in dichloromethane (20 ml) at r.t., The solution was cooled to 0°-5°C and phenyl isocyanate (0.42 ml, 3.87 mmol, 1 eq.) in dichlorometane (5 ml) was added dropwise. The solution was stirred at r.t. for lh, evaporated under reduced pressure and purified by silica gel chromatography (eluent n-hexane/ethyl acetate 1/1) to give 0.71 g of glassy solid that was suspended in diethyl ether to give a white solid. Yield 44%. Analytical data: TLC silica gel (eluent n-hexane/ethyl acetate 1/1 R.f.= 0.44), m.p. 48°-50°C. Η NMR (DMSO-de). δH: 8.68 (IH, s); 7.4-7.27 (8H, m); 7.22 (2H, t, J=8.2 Hz); 6.90 (IH, t, J= 7.3); 6.26 (IH, t, J=5.7); 5.11 (2H, s); 4.12 (IH, m); 3.58 (IH, m); 3.28 (IH, m); 1.38 (9H, s).
Step 2: 2- [(l,l-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid.
Figure imgf000223_0001
[00298] 2-S-[(l,l-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid, benzyl ester (0.7 g, 1.7 mmol.), of Step 1, was dissolved in methanol (25 ml) and Pd/C 10% (70 mg) was added. The mixture was hydrogenated at athmospheric pressure for lh. Pd/C was filtered and the solution was evaporated under reduced pressure to give 0.47 g of desired product. Yield 87%. Analytical data: 1H NMR (DMSO-d6). δH: 12.6 (IH, br); 8.66 (IH, s); 7.37 (2H, d, J=8.1 Hz); 7.21 (2H, t, J=7.50); 7.08 (IH, d, J=7.9); 6.89 (IH, t, J= 7.3); 6.21 (IH, t, J=5.9); 3.98 (IH, m); 3.54 (IH, m); 3.22 (IH, m); 1.38 (9H, s).
Example G.ll Synthesis of Further Compounds [00299] The following compounds can be prepared starting from 3-amino-2-S- [(l,l-dimethylethoxycarbonyl)amino]propionic acid, benzyl ester of Example G.5, with the methods described in Step 1 and Step 2 of Examples G.6-G.10.
Figure imgf000223_0002
Figure imgf000224_0003
Example G.12 2-[(l,l-Dimethylethoxycarbonyl)amino]-3-(3-benzyloxycarbonylamino)propionic acid.
Figure imgf000224_0001
Step /; N2-(tert-Butoxycarbonyl)-L-2,3-diaminopropionic acid
Figure imgf000224_0002
[00300] N-tert-butoxycarbonyl-L-asparagine, from step 1 of Example G.5 or commercially available, (8 g, 0.034 mol, leq.) was suspended in ethyl acetate (72 ml), acetonitrile (72ml) and water (36ml), and lodobenzenediacetate (13,3 g, 0.041 mol, 1,2 eq.) was added at 5°C. The mixture was stirred at 10°-25°C for 3-4h, then a white solid came off. The solid was filtered, washed with diethyl ether and dried under vacuum to give a white powder . Yield 57%. 4g.
Analytical data: m.p. 210°C-211°C. Silica gel (dicloromethane/methanol/acetic acid 5/3/1) Rf 0,5. 'HNMR (DMSO-de) 4.15 (IH, t); 3.15 (2H, m); 1.45 (9H, s);
Step 2: 2-[(l , 1-dimethylethoxy carbonyl) amino) '-3-(3- benzyloxycarbonylamino)propionic acid.
Figure imgf000225_0001
[00301] N2-(tert-Butoxycarbonyl)-L-2,3-diaminopropionic acid, from step 1, (3,8 g, 0,018 mol, 1 eq.) was dissolved in aqueous sodium carbonate 10% (2,2 eq.) at 25°C and 1,4-dioxane (38ml). To this solution, benzylchloroformate ( 3ml, 0,020 mol, 1,1 eq.) was added dropwise and the solution was stirred at 25°C for 3h. At the end of the reaction, the mixture was poured in water (100ml) and washed with diethyl ether (100ml). To the aqueous solution, HCl 37% (6 ml) was added till pH 2 and the obtained mixture was extracted with Ethyl Acetate (100ml). The organic layer was separated, washed with brine and dried over sodium sulfate anhydrous. The solvent was removed under reduced pressure to give a colourless oil that under vacuum gave a white foam. Yield 93%, 5,9 g.
Analytical data: silica gel (dicloromethane/methanol/acetic acid 5/3/1) Rf 1. 1H NMR (DMSO-de) 12.6 (IH br s); 7.35 (5H m) 6.94 (IH, d); 5 (2H, s); 4.1 (2H, m); 1.4 (9H, s).
Example G.13
2-(tert-Butoxycarbonilamino)-3-py razol-1 -yl-propionic acid.
Figure imgf000225_0002
[00302] The intermediate was prepared according to the procedure described in
Vederas, J. Am. Chem. Soc, 1985, 107, 7105-7109.
Example G.14
1-Methanesulfonyl-piperidine 4- carboxylic acid
3=^=0
Step 1: l-[(l,l-Dimethylethoxycarbonyl)amino]-piperidine-4-carboxylic acid
Figure imgf000226_0001
[00303] Piperidine-4-carboxylic acid (5g, 38.7 mmol, leq.) was dissolved in sodium carbonate solution (4.5g, 42.61mmol, 2.2 eq.), 70ml, and 1,4-dioxane (30ml). A solution of di-tert-butyldicarbonate (9.3g, 42.61mmol, l.leq.) in 1,4-dioxane (40ml) was added dropwise and the resulting mixture was stirred overnight at room temperature. The organic solvent was removed under reduced pressure and the resulting solution was acidified with HCl 37% till pH 2. The obtained suspension was filtered , the white solid washed with diethyl ether (5ml). The mother liquor has been extracted with ethyl acetate (120ml) and the previous solid was added. The organic solution was dried over anhydrous sodium sulfate, evaporated under reduced pressure to give a white solid that was dried at 80°C under vacuum to give the title compound. Yield 93%, 8.2g. Analytical data: m.p. 133°-135°C.
1H NMR (DMSO-de) 12.3 (IH br s); 3.85 (2H, d); 2.8 (2H, br); 2.35 (IH, t); 1.8 (2H, d); 1.4 (llH, m).
Step 2: l-[(l,l-Dirnethylethoxycarbonyl)amino]-piperidine-4-carboxylic acid benzyl ester
Figure imgf000227_0001
[00304] 1 -[(1, l-Dimethylethoxycarbonyl)amino]-piperidine-4-carboxylic acid
(6g,26.16mmol, leq.), from stepl, was dissolved in methanol (150ml) and cesium carbonate (4.26g, 13.08mmol, 0.5eq.) was added. The mixture was stirred at room temperature for 2h, the the solvent was removed under reduced pressure. The crude was dissolved in DMF (100ml) and benzylbromide (5.37g, 31.39mmol, 1.2eq.) was added dropwise. The mixture was stirred overnight at room temperature and poured in water (300ml), extracted with Ethyl Acetate (900ml) The organic layer was dried over anhydrous sodium sulfate and evaporated under reduced pressure to give a white solid. Yield 95%, 7g. I
Analytical data:
1H NMR (DMSO-de) 7.3 (5H m); 5.1 (2H, s); 3.85 (2H, d); 2.8 (2H, br); 2.65 (IH, t);
1.8 (2H, d); 1.4 (llH, m).
Step 3: Piperidine-4-carboxylic acid benzyl ester, hydrochloride salt.
Figure imgf000227_0002
CIH
[00305] 1 -[(1 , 1 -Dimethylethoxycarbonyl)amino]-piperidine-4-carboxylic acid benzyl ester (7g, 21.0 mmol), from step2, was dissolved in 1,4-dioxane (20ml). To this solution, HCl 4N in 1,4-dioxane (7.8ml, 300ml, 12 eq.) was added and the resulting solution was stirredovernight at room temperature. The solid was filtered, suspended in n-hexane (50ml), and filtered to give a white solid. Yield 54%, 2.5g.
Analytical data:
1H NMR (DMSO-de) 8.9 (2H, br); 7.35 (5H, m); 5.1 (2H, s); 3.25 (2H, d); 2.9 (2H, t);
2.75 (IH, m); 2.0 (2H, m); 1.8 (2H, m).
Step 4: l-Methanesulfonyl-piperidine-4-carboxylic acid benzyl ester.
Figure imgf000228_0001
[00306] Piperidine-4-carboxylic acid benzyl ester, hydrochloride salt (lg, 3.9 mmol, le.) from step 3, was dissolved in DMF (15ml), Triethylamine (0.55ml, 4mmol, (1 eq.) and methanesulfonylchloride were added. The mixture was stirred for lh at room temperature, then was poured in water (20ml) . The acqueous solution was extracted with Ethyl Acetate (90ml) and the organic layer was dried over anhydrous sodium sulfate, evaporated under reduced pressure to give a colourless oil. Yield 78%, 0.9g. Analytical data: lH NMR (DMSO-de) 7.35 (5H, m); 5.1 (2H, s); 3.5 (2H, d); 2.8 (5H, m); 2.6 (IH, m); 2.0 (2H, m); 1.6 (2H, m).
Step 5: 1-Methanesulfonyl-piperidine 4- carboxylic acid
Figure imgf000228_0002
[00307] l-Methanesulfonyl-piperidine-4-carboxylic acid benzyl ester (0.8g,
26.7mmol) from step 4, was dissolved in ethyl acetate (100ml) and methanol (10ml), Pd/C 10% (80mg) was added and the resulting mixture was hydrogenated at 1 bar. The catalyst was filtered over celite, the solvent was removed under reduced pressure to give a white solid. Yield 73%, 0.4g Analytical data:
1H NMR (DMSO-d6) 12.4 (IH, br); 3.6 (2H, d); 2.9 (4H, m); 2.4 (IH, m); 2.0 (2H, m); 1.6 (2H, m).
Example G.15 (4-MethyIphenyl)-ureido-sulfonyIchloride
Figure imgf000229_0001
[00308] This compound was prepared according to J Med. Chem. 1996, 39,
1243-1252. Briefly, a solution of chlorosulfonylisocyanate (1.62 g, 11.5 mmol, leq.) was diluted in dry diethylether and the resulting solution was cooled at -50°C<T<-40°C. To this solution, p-toluidine (1.23g, 11.5mmol, leq.) was added. The solution was stirred at -35°C for 10' and a suspension was obtained. The solid was filtered and washed with diethyl ether. Yield 80%, 2.3g. Analytical data: m.p. 127°-129°C. !H NMR (DMSO-de) 9.9 (IH, s); 7.3 (2H, d); 7.1 (2H, d); 2.25 (3H, s);
Example G.1 Isoxazole-5-carboxylic acid.
Figure imgf000229_0002
[00309] The desired carboxylic acid was prepared according to the procedure by
Wolfang et al., Synthesis, 1986, 69-70.
UTILITY
Compound Activity
[00310] The present compounds can inhibit proteasome activity. Table F-l below provides data related to several Example compounds of the invention with respect to, for example, ability to inhibit proteasome activity.
Methods and Compositions
[00311] Compounds of the present invention can inhibit the activity of proteasome leading to the inhibition or blocking of a variety of intracellular functions with which the proteasome is directly or indirectly associated. For example, proteasome inhibitors can modulate, such as induce, apoptosis in a cell. In some embodiments, the compounds herein can kill tumor cells by induction of apoptosis. Thus, the present compounds can be used to treat cancer, tumors or other proliferative disorders. [00312] In further embodiments, inhibition of proteasome function by compounds of the invention can inhibit the activation or processing of transcription factor NF-κB. This protein plays a role in the regulation of genes involved in the immune and inflammatory responses as well as in cell viability. Inhibition of proteasome function can also inhibit the ubiquitination/proteolysis pathway. This pathway catalyzes, inter alia, selective degradation of highly abnormal proteins and short-lived regulatory proteins. In some embodiments, compounds of the invention can prevent the degradation of p53 which is typically degraded by the ubiquitin-dependent pathway. The ubiquitination/proteolysis pathway also is involved in the processing of internalized cellular or viral antigens into antigenic peptides that bind to MHC-I molecules. Thus, the compounds of the invention can be used to reduce the activity of the cytosolic ATP- ubiquitin-dependent proteolytic system in a number of cell types. [00313] Accordingly, the usefulness of such compounds can include therapeutics, such as the treatment of various diseases or disorders associated with proteasome. The methods include administering a therapeutically effective amount of a compound of the invention, or composition thereof, to a mammal, such as a human having a disease or disorder associated with ι proteasome. The phrase "therapeutically effective amount" refers to an amount sufficient to prevent, alleviate, or ameliorate any phenomenon, such as a cause or symptom, known in the art to be associated with the disease or disorder. [00314] Treatable diseases or disorders (abnormal physical conditions) can be associated with either normal or abnormal activities of proteasome, such as the regulation of apoptosis. Numerous diseases or disorders that are associated with proteasome, or that are desirably treated by induction of apoptosis, are known and include, for example, various cancers and tumors including those associated with skin, prostate, colorectal, pancreas, kidney, ovary, mammary, liver, tongue, lung, and smooth muscle tissues. Preferred tumors that can be treated with proteasome inhibitors include, but are not limited to hematological tumors, such as, for example, leukemias, lymphomas, non-Hodgkin lymphoma, myeloma, multiple myeloma, as well as solid tumors such as, for example, colorectal, mammary, prostate, lung, and pancreas tumors. In order to elicit therapeutic effects, the proteasome inhibitors can be administered to patients as single agents or in combination with one or more antitumor or anticancer agent and/or radiotherapy. Examples of other anti-tumor or anti-cancer agents which can be advantageously administered concomitantly with a proteasome inhibitor include but are not limited to, adriamycin, daunomycin, methotrexate, vincristin, 6-mercaptopurine, cytosine arabinoside, cyclophosphamide, 5-FU, hexamethylmelamine, carboplatin, cisplatin, idarubycin, paclitaxel, docetaxel, topotecan, irinotecam, gemcitabine, L-PAM, BCNU and VP-16. Methods for determining apoptosis in vitro are well known in the art and kits are available commercially. See for example the Apo-ONE™ Homogeneous Caspase-3/7 Assay from Promega Corporation, Madison WI, USA (Technical Bulletin No. 295, revised 2/02, Promega Coφoration). (
[00315] Further diseases or disorders associated with the proteasome include accelerated or enhanced proteolysis that occurs in atrophying muscles, such as is often associated with activation of a nonlysomal ATP-requiring process involving ubiquitin. Accelerated or enhanced proteolysis can be the result of any of numerous causes including sepsis, burns, trauma, cancer, infection, neurodegenerative diseases such as muscular dystrophy, acidosis, or spinal nerve injuries, corticosteroid use, fever, stress, and starvation. Compounds of the invention can be tested for inhibition of muscle wastage by any various procedures known in the art such as by measuring urinary excretion of modified amino acid 3-methylhistidine (see, e.g., Young, et al., Federation Proc, 1978, 37, 229).
[00316] Compounds of the present invention can be further used to treat or prevent diseases or disorders associated with activity of NF-κB including for example, human immunodeficiency virus (HIV) infection and inflammatory disorders resulting from, for example, transplantation rejection, arthritis, infection, inflammatory bowel disease, asthma, osteoporosis, osteoarthritis, psoriasis, restenosis, and autoimmune diseases. Accordingly, a process that prevents activation of NF-κB in patients suffering from such a disease would be therapeutically beneficial. Inhibition of the NF-κB activity can be measured by using a DNA binding assay such a described in Palombella, et al., Cell, 1994, 78, 112,.
[00317] Those of ordinary skill in the art can readily identify individuals who are prone to or suspected of suffering from such diseases or disorders using standard diagnostic techniques.
Example A Assay for Chymotrypsin-Iike Activity of 20S Human Erythrocyte Proteasome (HEP)
[00318] Proteasome chymotrypsin-like activity of compounds of the invention was assayed according to the following procedure.
[00319] In 96-well microtiter plates, 20 S Human Erythrocyte Proteasome (HEP), purchased from Immatics Biotechnologies Inc., Tubingen, Germany was plated at 0.2 μg/mL (about 0.6 nM catalytic sites) in 0.04% SDS 20mM Tris buffer. A fluorimetric substrate Suc-LLVY-AMC (succinyl-Leu-Leu-Val-Tyr-7-amido-4-methylcoumarin), purchased from Sigma Inc., St. Louis, MO, USA was added to a final concentration of 100 μM from a stock solution of 10 mM in dimethylsulfoxide. Reaction volumes were 100 μl per well. After incubation for various periods of time at 37 °C, the concentration of free AMC (aminomethylcoumarin) was determined on a Perkin Elmer HTS 7000 Plus microplate reader, excitation 370 nM and emission 465nM. Proteasome activity was determined under conditions in which substrate hydrolysis increased linearly with time and the change in fluorescence signal was proportional to the concentration of free AMC.
Example B
Assay for Activity of α-Chymotrypsin
[00320] In 96-well microtiter plates bovine α-chymotrypsin, purchased from
Sigma Inc., was plated at 10 ng/mL (about 2 pM catalytic sites) in 0.5 M NaCl 50mM Hepes buffer. A fluorimetric substrate Suc-AAPF-AMC (succinyl-Ala-Ala-Pro-Phe-7- arnido-4-methyϊcoumarin), purchased from Sigma Inc., St. Louis, MO, USA was added to a final concentration of 25 μM from a stock solution of 10 mM in dimethylsulfoxide. Reaction volumes were 100 μl per well. After incubation for various periods of time at room temperature, the concentration of free AMC was determined on a Perkin Elmer HTS 7000 Plus microplate reader, excitation 370 nM and emission 465 nM. α- Chymotrypsin activity was determined under conditions in which substrate hydrolysis increased linearly with time and the change in fluorescence signal was proportional to the concentration of free AMC.
Example C
Determination of ICS0 Values for HEP and α-Chymotrypsin Inhibitors [00321] IC50 values are typically defined as the concentration of a compound necessary to produce 50% inhibition of the enzyme's activity. IC50 values are useful indicators of the activity of a compound for its designated use. The proteasome inhibitors of the invention can be considered active if they have IC50 values of less than about 1 micromolar for inhibition of human erythrocyte proteasome (HEP). In some embodiments, the inhibitors show some specificity for HEP and the ratio of the IC50 for inhibition of bovine α-chymotrypsin versus the IC50 for inhibition of HEP, i.e, IC50 (α- Chymotripsin) IC5o (HEP), is greater then about 100.
[00322] Inhibition of the chymotrypsin-like activity of HEP and of bovine α- chymotrypsin was determined by incubating the enzyme with various concentrations of putative inhibitors for 15 minutes at 37 °C (or room temperature for α-chymotrypsin) prior to the addition of substrate. Each experimental condition was evaluated in triplicate, and replicate experiments were performed for the inhibitors described herein. I
[00323] Compounds of the present invention are considered active in the above identified assay if their IC50 values for inhibition of HEP are less than 1000 nanoMolar. Preferably compounds of the present invention will have IC50 values for inhibition of HEP less than 100 nanoMolar. More preferably compounds of the present invention will have IC50 values for inhibition of HEP less than 10 nanoMolar. Compounds of the present invention have demonstrated, in the above identified assay, IC50 values for inhibition of HEP less than 1000 nanoMolar.
Example D
Cellular Assay for Chymotrypsin-like activity of Proteasome in Molt-4 Cell Line [00324] The chymotrypsin-like activity of proteasome in Molt-4 cells (human leukemia) was assayed according to the following procedure. A brief description of the method was published previously (Harding et al., J. Immunol., 1995, 155, 1767). [00325] Molt-4 cells were washed and resuspended in HEPES-buffered Saline
(5.4 mM KC1, 120 mM NaCl, 25 mM Glucose, 1.5 mM MgSO4, 1 mM Na pyruvate, 20 mM Hepes) and plated in 96-well microtiter white plates to a final concentration of 6x106 cells/mL. Then various 5X proteasome inhibitor concentrations (or diluted DMSO for controls), prepared from 250X DMSO solutions by diluting 50-fold using HEPES- buffered saline, were added to the plate to a final IX concentration. After 15 minutes incubation at 37°C, a fluorimetric cell permeable substrate (MeOSuc-FLF-AFC) (methoxysuccinyl-Phe-Leu-Phe-7-amido-4-trifluoromethylcoumarin) purchased from Enzyme Systems Products, catalogue number AFC-88, was added to each well at a final concentration of 25 μM from a stock solution of 20 mM in DMSO. Reaction volumes were 100 μl per well.
[00326] The concentration of free AFC was monitored every 1.5 min for 30 min
(22 cycles) on a Polastar Optima, BMG Labtechnologies microplate reader, using an excitation wavelength of 390 nm and emission wavelength of 520 nm. Proteasome activity was determined under conditions in which substrate hydrolysis increased linearly with time and the change in fluorescent signal was proportional to the concentration of free AFC.
Example E
Determination of ECS0 values for proteasome inhibitors in MOLT-4 Cell Line
[00327] EC5o values are typically defined as the concentration of a compound required to produce an inhibition of the enzyme's activity halfway between the minimum and the maximum response (0% and 85-90% respectively for this assay). EC50 values are useful indicators of the activity of a compound for its designated use. The compounds of the invention can be considered active if they have an EC50 of less than about 10 micromolar.
[00328] Inhibition of chymotrypsin-like activity of proteasome in Molt-4 cells was determined by incubating cells with various concentrations of putative inhibitors for 15 minutes at 37°C prior to the addition of substrate. Each experimental condition was evaluated in triplicate, and replicate experiments were performed for the inhibitors described herein.
[00329] Compounds of the present invention are considered active in the above identified assay if their EC50 values for proteasome inhibition in MOLT-4 are less than 10 microMolar. Preferably compounds of the present invention will have EC50 values for proteasome inhibition in MOLT-4 less than 2 microMolar. More preferably compounds of the present invention will have EC50 values for proteasome inhibition in MOLT-4 less than 200 nanomolar. Compounds of the present invention have demonstrated, in the above identified assay, EC50 values for proteasome inhibition in MOLT-4 cells of less than 10 microMolar. Example F
Assay for Trypsin-Iike Activity of the Proteasome
[00330] The trypsin-like activity of human proteasome can be assayed as described above with the following modifications. Reactions can be carried out in Tris- glycerol buffer (pH 9.5) supplemented with 1 mM 2-mercaptoethanol, and the substrate can be a fluorogenic substrate such as benzyloxycarbonyl~Phe~Arg— AMC (100 μM).
[00331] After incubation for various periods of time at 37 °C, the concentration of free AMC can be determined on a Fluoroskan II spectrofluorimeter with an excitation filter of 390 nm and an emission filter of 460 nm. Protease activity can be determined under conditions in which substrate hydrolysis increases linearly with time and the change in fluorescence is proportional to the concentration of free AMC.
Example G
In vivo Inhibition of Cellular Muscle Breakdown
[00332] The effect of inhibitors on the unweighting atrophy of the soleus muscle in juvenile rats can be determined by, for example, the procedures described in Tischler, Metabolism, 1990, 39, 756. For example, juvenile female Sprague-Dawley rats (80-90 g) can be tail-cast, hind limb suspended as described in Jaspers, et al., J. Appl. Physiol., 1984, 57, 1472. The animal's hind limbs can be elevated above the floor of the cage with each animal housed individually. Animals can have free access to food and water, and can be weighed at the time of suspension and at time of termination. During the suspension period the animals can be checked daily to ensure that their toes are not touching the floor of the cage, and that there is no swelling of the tail due to the cast.
Experimental Design— Part 1
[00333] Each experiment can begin with the suspension of 20 rats which are randomly divided into 4 groups of 5 animals each. Group A can be suspended for 2 days, providing baseline data to approximate the soleus muscle size in other animals suspended for longer times. Average body weights for the groups at the outset of the study can be compared and used as a correction factor for body size differences. Group B can be a second control group which has the soleus of one limb treated with an aqueous solution of mersalyl after two days of unweighting, to demonstrate the ability to slow muscle atrophy during unweighting, for each group of animals. At 2 days after unweighting commences, an aqueous solution of mersalyl (200 nM; 4 μL /100 g initial body wt) can be injected into one soleus. The contralateral muscle can be injected with a similar volume of 0.9% saline ("Vehicle"). The animals can be maintained under Innovar-vet (10 μL/100 g body wt) tranquilization during the in situ injection procedure. After the injections, the animals can be suspended for an additional 24 hours and the soleus can be removed. Groups C and D for each experiment can be used for testing each of two different embodiments of the disclosed compounds. Animals can be treated as in group B, except that 1 mM proteasome inhibitor, contained in dimethysulfoxide (DMSO), can be injected into the soleus of one leg and DMSO only into the contralateral soleus. Thus each experiment consists of two control groups and the testing of proteasome inhibitors of the invention. The completion of five such experiments with different pairs of inhibitors provides for an "n" value of 10 for testing each inhibitor and each can be tested in two different shipments of animals.
Processing of the Soleus Muscle—Part 1
[00334] After the animal is sacrificed, the soleus can be excised, trimmed of fat and connective tissue, and carefully weighed. The muscle can then homogenized in 10% trichloroacetic acid (TCA) and the precipitated protein pelleted by centrifugation. The pellet can then be washed once with 10% TCA and once with ethanohether (1:1). The final pellet can be solubilized in 4 ml of IN sodium hydroxide. The sample can be then analyzed for protein content by the biuret procedure, using albumin as a standard.
Data Analysis— Part 1
[00335] The effect of inhibitors on total muscle protein content can be examined primarily by paired comparison with the untreated contralateral muscle. The ratio of contents can be calculated and then analyzed statistically by analysis of variance ("ANOVA"). The left leg can always be the treated leg so that the protein content ratios can be compared to the non-treated control animals as well. In this way, a significant difference can be shown by comparing the protein content of the two legs, as well as the relative effectiveness of the tested inhibitors. A paired student test can also be performed for the effect of each' separate treatment. The non-treated control data also provide an estimate of protein content of day 2. This allows approximation of the protein changes over the 24 hours of treatment for each of the Groups B, C, and D. Experimental Design— Part 2
[00336] Each experiment can consist of 10 animals with groups of 5 animals being tested with one of the inhibitors for its effect on protein synthesis. Control animals are not needed for this aspect of the study as the contralateral DMSO-treated muscle serves as the paired control for the inhibitor-treated muscle. Each group can be injected as described for groups C and D in part 1. Twenty-four hours after the in situ treatment the fractional rate of protein synthesis can be analyzed in both soleus muscles. Each muscle can be injected with a 0.9% saline solution (3.5 μl/100 g final body wt) containing 3H-phenylalanine (50 mM; 1 μCi/ml). Fifteen minutes later the middle two- thirds of the muscle can be excised and the muscle can be processed as described below.
Processing of the Soleus Muscle— Part 2
[00337] The muscle can be first washed for 10 minutes in 0.84% saline containing 0.5 mM cycloheximide, to terminate protein synthesis, and 20 mM cycloleucine, to trap phenylalanine in the cell. The muscle can then be homogenized in 2.5 mL of ice-cold 2% perchloric acid. The precipitated protein can be pelleted by centrifugation. One aliquot of the supernatant can be taken for liquid scintillation counting and another aliquot can be processed for conversion of phenylalanine to phenethylamine to determine the soluble phenylalanine concentration fluorometrically. See, e.g., Garlick, et al., Biochem. J., 1980, 192, 719. These values can provide the intracellular specific activity. The specific activity of phenylalanine in the muscle protein can be determined after hydrolyzing the protein by heating in 6N HCl. The amino acids released can be solubilized in buffer. One aliquot can be taken for scintillation counting and another for analysis of phenylalanine as for the supernatant fraction. The fractional rate of protein synthesis can be calculated as: protein specific activity/intracellular specific activity .times.time.
Data Analysis— Part 2
[00338] Analyses of protein synthesis can be on a paired basis for each inhibitor.
Student paired t test comparisons of the contralateral muscles can determine whether there is any effect of the inhibitor on protein synthesis. Protein breakdown can be calculated approximately as the fractional rate of protein synthesis (from part 2) plus the fractional rate of protein accretion (from part 1), where protein loss yields a negative value for protein accretion.
[00339] Qualitatively the ability of inhibitors to slow protein loss without affecting protein synthesis indicates a slowing of protein degradation.
Example H
In vivo Investigation of Anti-Tumor Activity
Materials
[00340] The proteasome inhibitors used for in vivo studies can be formulated in an appropriate medium for intravenous (iv) or oral (po) administration. For example, for the iv administration the compounds can be administered dissolved in 0.9% NaCl, or in mixtures of 0.9% NaCl, solutol HS15 and dimethylsulfoxide, for example in the ratio 87:10:3 (v:v:v), respectively.
Cell lines
[00341] The following human and murine tumor cell lines of different histological origine can be used to test the antitumor activity of the compounds of the invention: H460 (human, lung), A2780 (human, ovary), PC-3 (human, prostate), LoVo (human, colon), HCT116 (human, colon), BXPC3 (human, pancreatic), PANC-1 (human, pancreatic), MX-1 (human, mammary), MOLT (human, leukemia), multiple myeloma (human, myeloma), YC8 (murine, lymphoma), L1210 (murine, leukemia), 3LL (murine, lung).
Animal species
[00342] 5-6 weeks immunocompetent or immunodeprived mice are purchased from commercial sources, for example from Harlan (Correzzana, Mi Italy). CD1 nu/nu mice are maintained under sterile conditions; sterilized cages, bedding, food and acidified water are used.
Tumor cell implantation and growth
[00343] Solid tumor models of different hystotype (lung, ovary, breast, prostate, pancreatic, colon) can be transplanted subcutaneously (sc.) into the axillary region of immunocompetent mice (murine models) or in immunodeprived mice (human models). Human tumor cell lines, originally obtained from ATCC, can be adapted to grow "in vivo" as solid tumor from "in vitro culture".
[00344] Hematological human or murine tumor models can be transplanted into different sites (iv, ip , ic or sc) in immunocompetent mice (murine tumors) or in immunodeprived mice (human leukemia, lymphoma and myeloma models), according to their highest tumor take.
Drug Treatment
[00345] Mice bearing solid (staged) or hematological tumors are randomized in experimental groups (10 mice/group). For solid tumors, an average tumor weight of 80-
100 mg for each group is considered to start the treatment; mice with the smallest and largest tumors are discarded.
[00346] Experimental groups are randomly assigned to the drug treatment and to the control group. Animals can be treated iv or orally, depending on the oral bioavailability with the compounds following different treatment schedules: iv weekly or twice weekly, or by daily oral administration .
[00347] On solid tumor models, drug treatment can begin when the tumor size ranges between 80-100 mg after tumor transplantation (Day 0).
[00348] The compounds can be administered in a volume of 10 mL/Kg body weight/mouse in the appropriate solvent.
Parameters of antitumor activity
[00349] The following parameters can be assessed for the evaluation of the antitumor activity: - growth of primary solid tumor; in each mouse is monitored by caliper measurement twice weekly; - survival time of treated mice as compared to control mice - twice weekly body weight evaluation of individual mice.
[00350] The tumor growth inhibition, TW1% (percentage of primary tumor growth inhibition in comparison with vehicle treated control groups) or the Relative tumor growth inhibition, RTWP/o in case of staged tumors, is evaluated one week after the last drug treatment and the Tumor weight (TW) can be calculated as follows: TW = l/2 ab2
where a and b are long and short diameters of the tumor mass in mm.
[00351] The antitumor activity can be determined as tumor weight inhibition
(TWI %), which is calculated according to the formula: mean TW treated TWI% = 100 - x lOO mean TW controls
[00352] The RTWI% (relative percentage of primary tumor growth inhibition in comparison with vehicle treated control groups) is evaluated one week after the last drug treatment, according to the following formula: mean RV of treated mice RTWI% = 100 - x lOO mean RV of controls mice
Vt (tumor weight on day t) where RV= Vo (initial tumor weight at the outset of treatment)
[00353] The Percent of Tumor Regression can be calculated as regressions in terms of relative tumor weight, determined as tumor weight at given day divided by initial tumor weight at the outset the experiment.
[00354] On haematological tumour models the antitumor activity can be determined as percentage increase of the median survival time of mice expressed as the ratio (T/C%) of the median survival time of the treated group (T) to that of the control group (C). Animals which are tumour-free at the end of the experiment (60 days after transplantation) are excluded from the calculation and considered as long term survivors (LTS). Evaluation of toxicity in tumor bearing mice
[00355] Toxicity can be evaluated daily on the basis of the gross autopsy findings and the weight loss. Mice are considered to have died of toxicity when death occurs before the death of vehicle treated control animals, or when significant body weight loss
(> 20%), and/or spleen and liver size reduction are observed.
[00356] The BWC% (Body weight change %) is assessed as follow : 100- (mean body weight of mice at given day/mean body weight at start of treatment) x 100. This value is determined one week after the last treatment with the test compound.
Example K
In vitro Viability of Cells
[00357] The ICso values measuring in vitro viability of cells in the presence of test compounds can be determined according to the following procedure. Cells can be seeded in 96-well plates at varying densities and then assayed using the Calcein-AM viability assay after 24 hours to determine the optimal final density for each cell type. Cells can then be seeded in 96-well plates at the determined density in 100 μL of an appropriate cell media known to one skilled in the art.
[00358] Serial dilutions of test compounds can be made so that the concentrations are twice the desired concentration to be evaluated. When 100 μL of the dilution is then added to the cells plated in 100 μL of media, a final concentration of, for example, 0, 11.7, 46.9, 187.5, 375, and 750 nM can be obtained. Compounds can be added to the plates three to four hours after seeding the cells, then the plates can be incubated at 37 °C for the desired time point (e.g., one, two, or three days).
[00359] Calcein-AM viability assays can be conducted at the desired time points as follows. Media can be aspirated using a manifold and metal plate to leave approximately 50 μL/well. The wells can be washed three times with 200 μL DPBS, aspirating each time with the manifold to leave 50 μL/well. A 8 μM solution of Calcein- AM in DPBS can be prepared and 150 μL can be added to each well. The plates can then be incubated at 37 °C for 30 minutes. After incubation, calcein can be aspirated with the manifold and cells can be washed with 200 μL DPBS as before. After final aspiration, fluorescence can be measured using a Cytofluor 2300 fluorescence plate reader. Negative controls can contain media and no cells, and experiments can be conducted in triplicate.
Example L
Kinetic Experiments in vitro
[00360] Compounds of the invention can be tested for proteasome inhibitory activity using a protocol described in Rock, et al., Cell, 1994, 78, 761. According to this procedure, dissociation constants (K;) for the equilibrium established when proteasome and test compound interact to form a complex. The reactions can be carried out using
SDS-activated 20S proteasome from rabbit muscle, and the proteasome substrate can be
Suc-LLVY-AMC.
Example M
Inhibition of Activation of NF-κB
[00361] Compounds of the invention can be tested for inhibiting the activity of
NF-κB by carrying out the assay described in Palombella, et al., Cell, 1994, 78, 111).
For example, MG63 osteocarcinoma cells can be stimulated by treatment with TNF-α for designated times. Whole cell extracts can be prepared and analyzed by electrophoretic mobility shift assays using the PRDII probe from the human IFN-β gene promoter.
Example N Compound Activity
[00362] Using the assays of Example C and Example E above the following
Table F-l demonstrates the utility of compounds of the invention for proteasome inhibition. In the following Tables, for the inhibition of HEP, Example C, compounds of the present invention with a "+" are less than 1000 nM; compounds of the present invention with a "++" are less than 100 nM; and compounds of the present invention with a "+++" are less than 10 nM in IC50 for HEP inhibition. In the following Tables, for the inhibition of MOLT4, Example E, compounds of the present invention with a "+" are less than 10000 nM; compounds of the present invention with a "++" are less than 2000 nM; and compounds of the present invention with a "+++" are less than 200 nM in EC50 for HEP inhibition. Where ">+" occurs activity was greater than the limits of the assay. Where no IC50 value or EC50 value is represented, data has yet to be determined.
Table F-l
Figure imgf000243_0001
Figure imgf000244_0001
Figure imgf000245_0001
Figure imgf000246_0001
Figure imgf000247_0001
Figure imgf000248_0001
Figure imgf000249_0001
Figure imgf000250_0001
Pharmaceutical Formulations and Dosage Forms
[00363] When employed as pharmaceuticals, the compounds of Formula (I) can be administered in the form of pharmaceutical compositions. These compositions can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal, and can be prepared in a manner well known in the pharmaceutical art.
[00364] This invention also includes pharmaceutical compositions which contain, as the active ingredient, one or more of the compounds of Formula (I) above in combination with one or more pharmaceutically acceptable carriers. In making the compositions of the invention, the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
[00365] In preparing a formulation, the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.
[00366] Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. The compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
[00367] The compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 100 mg, more usually about 10 to about 30 mg, of the active ingredient. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
[00368] The active compound can be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
[00369] For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention. When referring to these preformulation compositions as homogeneous, the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 500 mg of the active ingredient of the present invention.
[00370] The tablets or pills of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
[00371] The liquid forms in which the compounds and compositions of the present invention can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
[00372] Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
[00373] The amount of compound or composition administered to a patient will vary depending upon what is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration, and the like. In therapeutic applications, compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. An amount adequate to accomplish this is referred to as "therapeutically effective amount." Effective doses will depend on the disease condition being treated as well as by the judgement of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
[00374] The compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts. [00375] The therapeutic dosage of the compounds of the present invention can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of a compound of the invention in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration. For example, the compounds of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral adminstration. Some typical dose ranges are from about 1 μg/kg to about 1 g/kg of body weight per day. In some embodiments, the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day. The dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
[00376] The present invention also includes pharmaceutical kits useful, for example, in the treatment or prevention of inflammatory diseases, which comprise one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I). Such kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art. Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
[00377] Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference cited in the present application, including patents, published patent applications, and journal articles, is incorporated herein by reference in its entirety.

Claims

What is claimed is:
1. A compound of F
Figure imgf000255_0001
or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein: R1 is Ci-Cg alkyl, C2-C8 alkenyl, C2-C8 alkynyl, or C3-C7 cycloalkyl; R2 is H, -(CH2)aCH2NHC(=NR4)NH-Y, -(CH2)bCH2CONR5R6, -(CH2)cCH2N(R4)CONH2, -(CH2)dCH(R7)NR9R10, or -(CH2)eCH(R7)ZRs; a, b, and c are each, independently, 0, 1, 2, 3, 4, 5, or 6; d and e are each, independently, 0, 1, 2, 3, or 4; R4 is H or Ci-Cio alkyl; R5 and R6are each, independently, H, Cι-C10 alkyl, carbocyclyl, heterocarbocyclyl, or an amino protecting group; alternatively, Rs and R6 together with the N atom to which they are attached form a heterocarbocyclyl group; R7 is H or Ci-Cio alkyl; R8 is H, Ci-Cio alkyl, alkyl-S(=O)2-, aryl-S(=O)2-, H2NS(=O)2-, -SO3H, or a protecting group; R9 is H, CI-CΪO alkyl, carbocyclyl, or heterocarbocyclyl; R10is H, Ci-Cio alkyl, carbocyclyl, heterocarbocyclyl, C C10 alkyl-C(=O)-, C2-C10 alkenyl-C(=0)-, C2-C10 alkynyl-C(=O)-, carbocyclyl-C(=O)-, heterocarbocyclyl-C(=O)-, carbocyclylalkyl-C(=0)-, heterocarbocyclylalkyl-C(=0)-, Ci-Cio alkyl-S(=O)2-, carbocyclyl-S(=O)2-, heterocarbocyclyl-S(=0)2-, carbocyclylalkyl-S(=O)2-, heterocarbocyclylalkyl-S(=0)2-, Cι-C10 alkyl-NHC(=O)-, carbocyclyl-NHC(=0)-, heterocarbocyclyl-NHC(=O)-, carbocyclylalkyl-NHC(=O)-, heterocarbocyclylalkyl-NHC(=0)-, Ci-Cio alkyl-OC(=0)-, carbocyclyl-OC(=O)-, heterocarbocyclyl-OC(=O)-, carbocyclylalkyl-OC(=O)-, heterocarbocyclylalkyl-OC(=O)-, -Cio alkyl-NH-C(=O)-NHS(=O)2-, carbocyclyl-NH-C(=O)-NHS(=0)2-, heterocarbocyclyl-NH-C(=0)-NHS(=O)2-, C C10 alkyl-S(=O)2-NH-C(=O)-, carbocyclyl-S(=O)2-NH-C(=0)-, heterocarbocyclyl-S(=O)2-NH-C(=0)-, or an amino protecting group; wherein R10 is optionally substituted with 1, 2 or 3, R23; alternatively, R9 and R10 together with the N atom to which they are attached form a heterocarbocyclyl group optionally substituted with 1, 2 or 3 R23; Y is H, -CN, -NO2, -S(=0)2Rn, or a guanidino protecting group; R11 is Ci-Ce alkyl, aryl, or R12R13; R12and RJ3 are, independently, H, Cι-C10 alkyl, carbocyclyl, heterocarbocyclyl, or an amino protecting group; alternatively, R12 and R13 together with the N atom to which they are attached form a heterocarbocyclyl group; Z is O, S, Se, or Te; Q is -B(OH)2, -B(OR14)2, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O; R14 is H, - alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl; X is RAC(=0)-, RANHC(=0 , RAS(=O)2-, RAOC(=O)-, RASC(=O)-, or RA; RA is Cι-C20 alkyl optionally substituted with R20; C2-C20 alkenyl optionally substituted with R20; C2-C20 alkynyl optionally substituted with R20; carbocyclyl optionally substituted with .1-5 R21; or heterocarbocyclyl optionally substituted with 1-5 R21; R20 is selected from the group consisting of: -CN, halo, haloalkyl-, Cj-C4 alkyl, C2-C alkenyl, C2-C alkynyl, -CO2H, -C(=O)CO2H, -C(=0)NH2, -C(=O)H, -S(=O)NH2, -S(=O)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl) , -NHC(=O)NH2, -NHC(=O)R20a, -NHC(=O)OR20a, -OR20a, -SR20a, -S(=O)R20a, -S(=O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(=O)2R20a, -NHR20b, phthalimido, -(0-alkyl)r-OH, -(0-alkyl)r-(0-alkyl), -OR20c, -SR20c, -O-alkyl-R20c, -S-alkyl-R20c, -S(=O)-R20c, -S(=O)2-R20c, -S(=O)2-NHR20c, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR20c, carbocyclyl optionally substituted with 1-5 R21; and heterocarbocyclyl optionally substituted with 1-5 R21;
R20a is C C20 alkyl, C -C20 alkenyl, or C2-C20 alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, OH, CN, Cι-C alkyl, CrC alkoxy, C2-C8 alkoxyalkoxy, aryl, heteroaryl or -NHR20b;
R20b is an amino protecting group;
R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22;
R21 is selected from the group consisting of: Cι-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, -OR21a, -SR21a, -CN, halo, haloalkyl, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=0)O-alkyl, -NHC(=O)alkyl, -COOH, -C(=O)O-alkyl, -C(=O)alkyl, -C(O)H, -S(=O)-aIkyI, -S(=O)2-alkyl, -S(=O)-aryl, -S(=O)2-aryl, carbocyclyl optionally substituted with 1-5 R22, and heterocarbocyclyl optionally substituted with 1-5 R22;
R21a is H, C!-C20 alkyl, C2-C2o alkenyl, C2-C2o alkynyl, carbocyclyl or heterocarbocyclyl;
R22 is selected from the group consisting of: Ci-Cio alkyl, C2-Cιo alkenyl, C2-Cι0 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, aIkyl-OC(=0)-, alkyl-C(=O)-, aryl-OC(=O)-, alkyl-OC(=O)NH-, aryl-OC(=O)NH-, alkyl-C(=0)NH-, alkyl-C(=0)0-, (alkyl-0)r-alkyl, HO-(aIkyl-0)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=0)-, alkyl-S(=0)2-, H2NS(=O)-, and H2NS(=O)2-;
R23 is selected from the group consisting of: Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, F, CI, Br, 1, haloalkyl, -NH2, -NHR23a, -N(R23a)2, -N3, -NO2, -CN, -CNO, -CNS, -C(=O)OR23a, -C(=O)R23a, -OC(=O)R23a, -N(R23a)C(=0)R23a, -N(R23a)C(=O)OR23a, -C(=0)N(R23a)2, ureido, -OR23a, -SR23a, -S(=0)-(d-C6 alkyl), -S(=O)2-(Cι-C6 alkyl), -S(=0)-aryl, -S(=0)2- ryl, -S(=O)2-N(R23a)2; carbocyclyl optionally substituted with 1-5 R24; and heterocarbocyclyl optionally substituted with 1-5 R24;
R23a is H or C C6 alkyl; alternatively, two R23 may be combined, together with the N atom to which they are attached, to form a 5 to 7 membered heterocyclic group; and
R24 is selected from the group consisting of: d-C4 alkyl, C2-C4 alkenyl, C2-C alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=0)-, alkyl-C(=O)-, aryl-OC(=O)-, alkyl-OC(=O)NH-, aryl-OC(=O)NH-, alkyl-C(=O)NH-, alkyl-C(=O)O-, (alkyl-O)r-alkyl, HO-(alkyl-O)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=O)-, alkyl-S(=0)2-, H2NS(=O)-, and H2NS(=O)2-; and r is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; with the proviso that when Q is a 1,1,2,2-tetramethylethanediol boronic ester, then X is not aralkyloxycarbonyl; with the proviso that when Q is a 1,1,2,2-tetramethylethanediol boronic ester, and R1 is cycloalkyl, then R2 is not -CH2CONH ; and with the proviso that when X is RAC(=0)-, RA is a C4-Cι5 straight-chained alkyl substituted with R20, and R20 is -CN, -CO2H, -C(=O)O-R20a, -NHS(=O)2R20a, - NHC(=O)R20a, -NHR20 , or phthalimido; then R2 is not - (CH2)aCH2NHC(=NR4)NH-Y, wherein Y is H, -CN, -NO2, or a guanidino protecting group.
2. The compound of Claim 1 wherein R1 is -C alkyl.
3. The compound of Claim 1 wherein R1 is 2-propyl.
4. The compound of Claim 1 wherein R2 is -(CH2)aCH2NHC(=NR4)NH-Y, -(CH2)bCH2CONR5R6, -(CH2)0CH2N(R4)CONH2, -(CH2)dCH(R7)NR9R10, or -(CH2)eCH(R7)ZR8.
5. The compound of Claim 1 wherein R2 is -(CH2)aCH2NHC(=NR4)NH-Y.
6. The compound of Claim 5 wherein a is 1, 2, 3, or 4.
7. The compound of Claim 5 wherein a is 2.
8. The compound of Claim 1 wherein R2 is -(CH2)dCH(R7)NR9R10.
9. The compound of Claim 8 wherein d is 0, 1, or 2.
10. The compound of Claim 8 wherein d is 0.
11. The compound of Claim 8 wherein R9 is H.
12. The compound of Claim 1 wherein R2 is -(CH2)eCH(R7)ZR8.
13. The compound of Claim 12 wherein Z is O .
14. The compound of Claim 13 wherein e is 0, 1, or 2.
15. The compound of Claim 13 wherein e is 0.
16. The compound of Claim 1 wherein Q is B(OH)2 or a cyclic boronic ester wherein said cyclic boronic ester contains from 6 to 10 carbon atoms and contains at least one cycloalkyl moiety.
17. The compound of Claim 16 wherein Q is pinanediol boronic ester.
18. The compound of Claim 16 wherein Q is bicyclohexyl- 1 , 1 '-diol boronic ester.
19. The compound of Claim 16 wherein Q is l,2-dicyclohexyl-ethane-l,2-diol boronic ester.
20. The compound of Claim 1 wherein X is RAC(=O)-.
21. The compound of Claim 1 wherein X is RANHC(=0)-.
22. The compound of Claim 1 wherein X is RAS(=0)2-.
23. The compound of Claim 1 wherein RA is Cι- 4 alkyl substituted by -(O-aIkyl)r-OH or -(O-alkyl)r-(0-alkyl), wherein r is 1, 2, 3, 4, or 5.
24. The compound of Claim 23 wherein RA is Cj-Cι alkyl substituted by -(O- alkyl)r-OH, -(O-alkyl)r-(O-alkyl), wherein r is 1, 2 or 3.
25. The compound of Claim 23 wherein RA comprises at least one -CH2CH2O- group.
26. The compound of Claim 23 wherein RA is -CH2(OCH2CH2)rOCH3.
27. The compound of Claim 23 wherein RA is -CH2OCH2CH2OCH2CH2OCH3 or -CH2OCH2CH2OCH3.
28. The compound of Claim 1 wherein R is aryl or heteroaryl each optionally substituted with 1-5 R21.
29. The compound of Claim 1 wherein RA is cycloalkyl or heterocycloalkyl each optionally substituted with 1-5 R21.
30. The compound of Claim 1 wherein R A i. s Cι-C20 alkyl; C2-C20 alkenyl; or C2-C2o alkynyl, each optionally substituted with R20.
31. The compound of Claim 1 wherein RΛ is Cι-C2o alkyl; C2-C20 alkenyl; or C2-C20 alkynyl, each substituted with a carbocyclyl group or a heterocarbocyclyl group wherein said carbocyclyl group or heterocarbocyclyl group is optionally substituted with 1, 2 or 3 R21.
32. The compound of Claim 1 wherein RA is Cι-C20 alkyl; C2-C20 alkenyl; or C2-C2o alkynyl, each substituted with an aryl group wherein said aryl group is optionally substituted with 1, 2 or 3 R21.
33. The compound of Claim 1 wherein RA is Cι-C2o alkyl; C2-C20 alkenyl; or C2-C2o alkynyl, each substituted with an heteroaryl group wherein said heteroaryl group is optionally substituted with 1, 2 or 3 R21.
34. The compound of Claim 1 wherein RA is Cι-C20 alkyl; C2-C20 alkenyl; or C2-C20 alkynyl, each substituted with an cycloalkyl group wherein said cycloalkyl group is optionally substituted with 1, 2 or 3 R21.
35. The compound of Claim 1 wherein RA is Cι-C2o alkyl; C2-C2o alkenyl; or C2-C20 alkynyl, each substituted with an heterocycloalkyl group wherein said heterocycloalkyl group is optionally substituted with 1, 2 or 3 R21.
36. The compound of Claim 1 wherein R2 is -CH2NH-C(=O)OCH2(C6H5).
37. The compound of Claim 1 wherein RA is Cι-C20 alkyl; C2-C20 alkenyl; or C2-C20 alkynyl, each optionally substituted with R20, wherein R20 is selected from CN, halo, haloalkyl, -CO2H, -C(=O)CO2H, -C(=O)NH2, -C(=O)H, -S(=O)NH2, -S(=0)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)NH2, -NHC(=O)R20a, -NHC(=O)OR20a, -OR20a, -SR20a, -S(=O)R20a, -S(=O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(-O)NHR20a, -C(=O)O-R 0a, -NHS(=O)2R20a, -NHR20b, phthalimido, -(O-alkyl), -(0-alkyl)r-OH, -(0-alkyl)r-(O-alkyl), -OR20c, -SR20c, -O-alkyl-R20c, -S-alkyl-R20c, -S(-O)-R20c, -S(=O)2-R20c, -S(=O)2-NHR20c, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, and -C(=O)NHR20c.
38. The compound of Claim 1 wherein R2 is H and X is (O-alkyl)-(0-alkyl)r-(C!-C14 alkyl)-C(=O)- or HO-(alkyl-O)r-(Cι-Cι4 alkyl)- C(=O>.
39. The compound of Claim 1 wherein X is RAC(=0> and RA is C4-Cι6 alkyl.
40. The compound of Claim 1 wherein X is RAC(=0)- and RA is aryl optionally substituted with 1-3 R21.
41 The compound of Claim 1 wherein X
Figure imgf000262_0001
is phenyl substituted with one R21; and R21 is phenoxy.
42. The compound of Claim 1 wherein X is RAC(=O)-, RA is Cι-C alkyl substituted with R20, and R20 is aryl optionally substituted with 1-3 R21.
43. The compound of Claim 40 wherein aryl is substituted by at least one halo.
44. The compound of Claim 1 wherein X is RAC(=O)-; RA is CrC14 alkyl substituted with R20; and R20 is -OR20a or -OR20c.
45. The compound of Claim 1 wherein X is RAC(=O)-; RA is C Cι4 alkyl substituted with R20; and R20 is heterocarbocyclyl optionally substituted with 1-3 R21.
46. The compound of Claim 1 wherein X is RAS(=0)2- and RA is C3-C16 alkyl.
47. A compound of claim 1 of Formula (I):
Figure imgf000262_0002
(I) or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein:
R1 is Cι-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, or C3-C7 cycloalkyl;
R2 is H, -(CH2)aCH2NHC(=NR4)NH-Y, -(CH2)bCH2CONR5R6, -(CH2)0CH2N(R )CONH2, -(CH2)dCH(R7)NR9R10, or -(CH2)6CH(R7)ZR8; a, b, and c are each, independently, 0, 1, 2, 3, 4, 5, or 6; d and e are each, independently, 0, 1, 2, 3, or 4;
R4 is H or Ci-C.o alkyl; R5 and R6are each, independently, H, Ci-Cio alkyl, carbocyclyl, heterocarbocyclyl, or an amino protecting group; alternatively, R5 and R6 together with the N atom to which they are attached form a heterocarbocyclyl group; R7 is H or Cι-C10 alkyl; R8 is H, Ci-Cio alkyl, alkyl-S(=O)2-, aryl-S(=O)2-, H2NS(=O)2-, -SO3H, or a protecting group; R9 is H, Ci-Cio alkyl, carbocyclyl, or heterocarbocyclyl; R10is H, Cj-Cio alkyl, carbocyclyl, heterocarbocyclyl, Ci-Cio alkyl-C(=O)-, carbocyclyl-C(=0)-, heterocarbocyclyl-C(=0)-, carbocyclylalkyl-C(=O)-, heterocarbocyclylalkyl-C^O)-, Ci-Cio alkyl-S(=O)2-, carbocyclyl-S(=O)2-, heterocarbocyclyl-S(=0)2-, carbocyclylalkyl-S(=O)2-, heterocarbocyclylalkyl-S(=O)2-, Ci-Cio alkyl-NHC(=0)-, carbocyclyl-NΗC(=O)-, heterocarbocyclyl-NHC(=O)-, carbocyclylalkyl-NHC(=O)-, heterocarbocyclylalkyl-NHC(=O)-, Ci-Cio alkyl-OC(=0)-, carbocyclyl-OC(=0)-, heterocarbocyclyl-OC(=O)-, carbocyclylalkyl-OC(=O)-, heterocarbocyclylalkyl-OC(=O)-, or an amino protecting group; wherein R10 is optionally substituted with 1, 2, or 3 R23; alternatively, R9 and R10 together with the N atom to which they are attached form a heterocarbocyclyl group; Y is -H, -CN, -NO2, -S(=0)2Rπ, or a guanidino protecting group; R11 is Ci-Ce alkyl, aryl, or NR12R13; R12and R13 re, independently, H, Ci-Cio alkyl, carbocyclyl, heterocarbocyclyl, or an amino protecting group; alternatively, R12 and R13 together with the N atom to which they are attached form a heterocarbocyclyl group; Z is O, S, Se, or Te; Q is -B(OH)2, -B(OR14)2, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O; R14 is H, C1-C4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl; X is RAC(=O)-, RANHC(=O)-, RAS(=O)2-, RAOC(=0)-, RASC(=O)-, or RA; RA is Ci-C20 alkyl optionally substituted with R20; C2-C20 alkenyl optionally substituted with R20; C2-C20 alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R21; or heterocarbocyclyl optionally substituted with 1-5 R21; R20 is selected from the group consisting of: -CN, halo, haloalkyl-, -C alkyl, C2-C4 alkenyl, C2-C4 alkynyl, -CO2H, -C(-O)C02H, -C(=0)NH2, -C(=0)H, -S(=O)NH2, -S(=O)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)NH2, -NHC(=O)R20a, -NHC(=O)OR20a, -OR20a, -SR20a, -S(=O)R20a, -S(=O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(=O)2R20a, -NHR20b, phthalimido, -(-0-alkyl)r, -O-alkyl-OH, -(0-alkyl)r-OH, -OR20c, -SR20c, -O-alkyl-R20c, -S-alkyl-R20c, -S(=O)-R20c, -S(=O)2-R20c, -S(=O)2-NHR20c, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR20c, carbocyclyl optionally substituted with 1-5 R21; and heterocarbocyclyl optionally substituted with 1-5 R21; R20a is Ci-C20 alkyl, C2-C20 alkenyl, or C2-C 0 alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, Cι-C4 alkyl, aryl, heteroaryl or -NHR20b; R20b is an amino protecting group; R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22; R21 is selected from the group consisting of: Cι-C20 alkyl, C2-C2o alkenyl, C2-C20 alkynyl, CpC2o alkoxy, Cι-C20 thialkoxy, -OH, -CN, halo, haloalkyl, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=0)O-alkyl, -NHC(=0)alkyl, -C(=0)O-alkyl, -C(=O)alkyi, -S(=O)-alkyl, -S(=O)2-alkyl, -S(=O)-aryl, -S(=O)2-aryl, carbocyclyl optionally substituted with 1-5 R22; and heterocarbocyclyl optionally substituted with 1-5 R22; R22 is selected from the group consisting of: Cι-C10 alkyl, C2-Cιo alkenyl, C2-Cι0 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=O)-, alkyl-C(=O)-, aryl-OC(=O)-, alkyl-OC(=O)NH-, aryl-OC(=O)NH-, alkyl-C(=O)NH-, alkyl-C(=O)0-, (alkyl-0)r-alkyl, HO-(alkyl-O)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=O)-, alkyl-S(=O)2-, H2NS(=O)-, and H2NS(=O)2-;
R23 is selected from the group consisting of: Ci-Cβ alkyl, C2-C6 alkenyl, C2-C6 alkynyl, F, CI, Br, I, haloalkyl, -NH2, -NHR23a, -N(R23a)2, -N3, -NO2, -CN, -CNO, -CNS, -C(=O)OR23a, -C(=O)R23a, -OC(=0)R23a, -N(R23a)C(=O) R23a, -C(=O)N(R23a)2, ureido, -OR23a, -SR23a, -S(=O)2-(Cι-C6 alkyl), -S(=O)2-aryl, and -S(=0)2-N(R23a)2;
R23a is H or C!-C6 alkyl; alternatively, two R23a may be combined, together with the N atom to which they are attached, to form a 5 to 7 membered heterocyclic group; and r is 2, 3, 4, 5, 6, 7, 8, 9, or 10; and with the proviso that when Q is a 1,1,2,2-tetramethylethanediol boronic ester, then X is not aralkyloxycarbonyl; with the proviso that when when Q is a 1,1,2,2-tetramethylethanediol boronic ester, and R1 is cycloalkyl, then R2 is not -CH2CONH ; and with the proviso that when X is RAC(=0)-, RA is a C4-Cι5 straight-chained alkyl substituted with R20, and R20 is -CN, -CO2H, -C( ))O-R20a, -NHS(=O)2R20a, - NHC(=O)R20a, -NHR20b, or phthalimido; then R2 is not - (CH2)aCH2NHC(=NR4)NH-Y, wherein Y is H, -CN, -N02, or a guanidino protecting group.
48. The compound of Claim 47 wherein R1 is 2-proρyl.
49. The compound of Claim 47 wherein Q is -B(OH)2.
50. The compound of Claim 47 wherein Q is pinanediol boronic ester.
51. The compound of Claim 47 wherein X is RAC(=0)-.
52. The compound of Claim 47 wherein R2 is -CH2NH-C(=0)OCH2(C6H5).
53. The compound of Claim 47 wherein X is RAC(=O)- and RA is C4-C16 alkyl.
54. The compound of Claim 47 wherein X is RAC(=O)- and RA is aryl optionally substituted with 1-3 R21.
55. The compound of Claim 47 wherein X is RAC(=O)- and RA is a heterocarbocyclyl group optionally substituted with 1-3 R21.
56. The compound of Claim 47, or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein:
R1 is 2-propyl;
R2 is H, -(CH2)aCH2NHC(=NR )NH-Y, -(CH2)bCH2CONR5R6, -(CH2)cCH2N(R4)CONH2, -(CH2)dCH(R7)NR9R10, or -(CH2)eCH(R7)ZR8; Q is -B(OH)2 or pinanediol boronic ester; X is RAC(=0)-; and RA is C4-Ci6 alkyl; aryl optionally substituted with 1-3 R21; or heterocarbocyclyl group optionally substituted with 1-3 R21.
57. A compound of claim 1 having Formula (I):
Figure imgf000266_0001
O or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein:
R1 is Ci-Cg alkyl;
R2 is -(CH2)aCH2NHC(=NH)NH-Y, -(CH2)cCH2NHCONH2, -(CH2)dCH(R7)NR9R10, or -(CH2)eCH(R7)ZR8; a is 1, 2, 3, 4, or 5; c is 1, 2, 3, 4, or 5; d is O, 1, or 2; e is 0, l, or 2; R7 is H or methyl;
R8 is H, Ci-Cio alkyl, -S(=0)2-alkyl, -S(=0)2-aryl, -S(=O)2-NH2, -SO3H, or a protecting group; Y is -H, -CN, -NO2, -S(=O)2Ru, or a guanidino protecting group; R9 is H, C1- 0 alkyl, carbocyclyl, or heterocarbocyclyl; R10is H, C1-C10 alkyl, carbocyclyl, heterocarbocyclyl, -C10 alkyl-C(=O)-, carbocydyl-C(=0)-, heterocarbocyclyl-C(=O)-, carbocyclylalkyl-C(=0)-, heterocarbocyclylalkyl-C(=0)-5 C1-C10 alkyl-S(=O)2-, carbocyclyl-S(=0)2-, heterocarbocyclyl-S(=O)2-, carbocyclylalkyl-S(=O)2-, heterocarbocyclylalkyl-S(=O)2-, C1-C10 alkyl-NHC(=O)-, carbocyclyl-NHC(=O)-, heterocarbocyclyl-NHC(=O)-, carbocyclylalkyl-NHC(=O)-, heterocarbocyclylalkyl-NHC(=O)-, C1-C10 aIkyl-OC(=0)-, carbocyclyl-OC(=O)-, heterocarbocyclyl-OC(=0)-, carbocyclylalkyl-OC(=O)-, heterocarbocyclylalkyl-OC(=0)-, or an amino protecting group; wherein R10 is optionally substituted with 1, 2 or 3 R23; alternatively, R9 and R10 together with the N atom to which they are attached form a heterocarbocyclyl group; Rn is Cι-C6 alkyl, aryl, orNR12R13; R12and R13are, independently, H, C1-C10 alkyl, carbocyclyl, heterocarbocyclyl, or an amino protecting group; alternatively, R12 and R13 together with the N atom to which they are attached form a heterocarbocyclyl group; Z is O or S; Q is -B(OH)2, -B(OR14)2, or a cyclic boronic ester wherein said cyclic boronic ester contains from 6 to 20 carbon atoms and contains at least one cycloalkyl moiety; R14 is H, C1-C4 alkyl, or cycloalkyl;
X is RAC(=O)-, RANHC(=0)-, RAS(=O)2-, RAOC(=O)-, RASC(=O)-, or RA; RA is Cι-C20 alkyl optionally substituted with R20; C -C 0 alkenyl optionally substituted with R20; C2-C2o alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R21; or heterocarbocyclyl optionally substituted with 1-5 R21; R20 is selected from the group consisting of: -CN, halo, haloalkyl-, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, -CO2H, -C(=O)C02H, -C(=0)NH2, -C(=O)H, -S(=0)NH2, -S(=O)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)NH2, -NHC(=O)R20a, -NHC(=O)OR20a, -OR 0a, -SR20a, -S(=O)R20a, -S(=O)2R20a, -S(=O)2-NHR20β, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(=O)2R20a, -NHR20b, phthalimido, -(0-alkyl)r, -O-alkyl-OH, -(O-alkyl)r-OH, -OR20c, -SR20c, -O-alkyl-R20c, -S-alkyl-R200, -S(=O)-R20c, -S(=O)2-R20c, -S(=O)2-NHR20c, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR2Qc, carbocyclyl optionally substituted with 1-5 R21; and heterocarbocyclyl optionally substituted with 1-5 R21;
R20a is Ci-C20 alkyl, C2-C20 alkenyl, or C2-C20 alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, C1-C4 alkyl, aryl, heteroaryl or -NHR20b;
R20 is an amino protecting group;
R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22;
R21 is selected from the group consisting of: C1-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, C C2o alkoxy, Cι-C20 thialkoxy, -OH, -CN, halo, haloalkyl, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)O-alkyl, -NHC(=0)alkyl, -C(=O)O-alkyl, -C(=0)alkyl, -S(=0)-alkyl, -S(=0)2-alkyl, -S(=0)-aryl, -S(=O)2-aryl, carbocyclyl optionally substituted with 1-5 R22, and heterocarbocyclyl optionally substituted with 1-5 R22;
R22 is selected from the group consisting of: d-Cio alkyl, C2-Cιo alkenyl, C2-Cι0 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=O)-, alkyl-C(=O)-, aryl-OC(=O)-, alkyl-OC(=O)NH-, aryl-OC(=0)NH-, alkyl-C(=O)NH-, alkyl-C(=0)0-, (alkyl-O)r-alkyl, HO-(alkyl-0)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=O)-, alkyl-S(=O)2-, H2NS(=0)-, and H2NS(=0)2-;
R23 is selected from the group consisting of: Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, F, CI, Br, I, haloalkyl, -NH2, -NHR23a, -N(R23a)2, -N3, -NO2, -CN, -CNO, -CNS, -C(=O)OR23a, -C(=O)R23 , -OC(=O)R23a, -N(R23a)C(=0) R23a, -C(=O)N(R23a)2, ureido, -OR 3a, -SR23a, -S(=O)2-(Cι-C6 alkyl), -S(=0)2-aryl, and -S(=O)2-N(R23a)2;
R23a is H or Ci-Cβ alkyl; alternatively, two R23a may be combined, together with the N atom to which they are attached, to form a 5 to 7 membered heterocyclic group; and r is 2, 3, 4, 5, 6, 7, 8, 9, or 10; with the proviso that when X is RAC(=O)-, RA is a C4-Cι5 straight-chained alkyl substituted with R20, and R20 is -CN, -C02H, -C(=O)O-R20a, -NHS(=O)2R20a, - NHC(=O)R20a, -NHR20b, or phthalimido; then R2 is not -(CH2)aCH2NHC(=NR4)NH-Y, wherein Y is H, -CN, -NO2, or a guanidino protecting group.
58. A compound of claim 1 having Formula (I):
Figure imgf000269_0001
(I) or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein:
R1 is C1-C4 alkyl;
R2 is -(CH2)aCH2NHC(=NH)NH-Y, -(CH2)cCH2NHCONH2, or -(CH )dCH(R7)NR9R10; a is 1, 2, or 3; c is 1, 2, or 3; d is O or 1;
R7 is H or methyl;
R9 is H or Cι-Cιo alkyl;
R10 is H, Ci-do alkyl, or an amino protecting group;
Y is H, CN, orN02;
Q is -B(OH)2, pinanediol boronic ester, bicyclohexyl-1,1 '-diol boronic ester, or 1,2- dicyclohexyl-ethane-l,2-diol boronic ester; X is RAC(=0)-, RANHC(=O)-, RAS(=O)2-, RAOC(=O)-, RASC(=O)-, or RA; RA is Ci-C20 alkyl optionally substituted with R20; C2-C20 alkenyl optionally substituted with R20; C2-C20 alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R21; or heterocarbocyclyl optionally substituted with 1-5 R21; R20 is selected from the group consisting of: -CN, halo, haloalkyl-, C]-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, -CO2H, -C(=0)C02H, -C(=O)NH2, -C(=0)H, -S(=0)NH2, -S(=0)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)NH2, -NHC(=O)R20a, -NHC(=O)OR20a, -OR20a, -SR20a, -S(=O)R20a, -S(=O)2R20a, -S(=O)2-NHR 0a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(=O)2R20a, -NHR20b, phthalimido, -(0-alkyl)r, -O-alkyl-OH, -(O-aIkyl)r-OH, -OR20c, -SR20c, -O-alkyl-R20c, -S-alkyl-R20c, -S(=O)-R20c, -S(=O)2-R20c, -S(=O)2-NHR20c, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR20c, carbocyclyl optionally substituted with 1-5 R21; and heterocarbocyclyl optionally substituted with 1-5 R21; R20a is Ci-C20 alkyl, C2-C20 alkenyl, or C2-C o alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, Cι-C4 alkyl, aryl, heteroaryl or -NHR20b;
R ,20b i :s an amino protecting group;
R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22;
R21 is selected from the group consisting of: Cι-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, C1-C20 alkoxy, Cι-C20 thialkoxy, -OH -CN, halo, haloalkyl, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)0-alkyl, -NHC(=O)alkyl, -C(=0)O-aIkyI, -C(=O)alkyl, -S(=O)-alkyl, -S(=0)2-alkyl, -S(=O)-aryl, -S(=0)2-aryl, carbocyclyl optionally substituted with 1-5 R22, and heterocarbocyclyl optionally substituted with 1-5 R22;
R22 is selected from the group consisting of: C1-Q0 alkyl, C2-Cιo alkenyl, C2-Cιo alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=O)-, alkyl-C(=0)-, aryl-OC(=O)-, alkyl-OC(=O)NH-, aryl-OC(=O)NH-, alkyl-C(=O)NH-, alkyl-C(=O)O-, (alkyl-O)r-alkyl, HO-(alkyl-O)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=O)-, alkyl-S(=O)2-, H2NS(=O , and H2NS(=0)2-; and r is 2, 3, 4, or 5; with the proviso that when X is RAC(=0)-, RA is a C4-Cιs straight-chained alkyl substituted with R20, and R20 is -CN, -C02H, -C(=O)O-R20a, -NHS(=O)2R20a, - NHC(=O)R20a, -NHR20b, or phthalimido; then R2 is not - (CH2)aCH2NHC(=NR4)NH-Y, wherein Y is H, -CN, or -NO2.
59. The compound of Claim 58 wherein R1 is 2-propyl.
60. The compound of Claim 58 wherein Q is -B(OH)2.
61. The compound of Claim 58 wherein Q is pinanediol boronic ester.
62. The compound of Claim 58 wherein X is RAC(=O)-.
63. The compound of Claim 58 wherein R2 is -CH2NH-C(=0)OCH2(C6H5).
64. The compound of Claim 58 wherein X is RAC(=O)- and RA is C4-Cιe alkyl.
65. The compound of Claim 58 wherein X is RAC(=O)- and RA is aryl optionally substituted with 1-3 R21.
66. The compound of Claim 58 wherein X is RAC(=O)- and RA is a heterocarbocyclyl group optionally substituted with 1-3 R21.
67. The compound of Claim 58, or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein:
R1 is 2-propyl;
Q is -B(OH)2 or pinanediol boronic ester;
X is RAC(=O)-; and RA is C -Ci6 alkyl; aryl optionally substituted with 1-3 R21; or heterocarbocyclyl group optionally substituted with 1-3 R21.
68. A compound of claim 1 having Formula (I):
Figure imgf000272_0001
(I) or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein:
R1 is Q-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or C3-C7 cycloalkyl;
R2 is -CH2NH2 or -CH2NR9R10;
R9 is H or Cι-C10 alkyl;
R10 is H, C]-Cιo alkyl, carbocyclyl, heterocarbocyclyl, Ci-Cio alkyl-C(=O)-, carbocyclyl-C(=0)-, heterocarbocyclyl-C(=0)-, carbocyclylalkyl-C(=O)-, heterocarbocyclylalkyl-C(=0)-, Ci-Cio alkyl-S(=0)2-, carbocyclyl-S(=O)2-, heterocarbocyclyl-S(=O)2-, carbocyclylalkyl-S(=O)2-, heterocarbocyclylalkyl-S(=O)2-, Ci-Cio alkyl-NHC(=0)-, carbocyclyl-NHC(=O)-, heterocarbocyclyl-NHC(=O)-, carbocyclylalkyl-NHC(=O)-, heterocarbocyclylalkyl-NHC(=O)-, Ci-Cio alkyl-OC(=O)-, carbocyclyl-OC(=0)-, heterocarbocyclyl-OC(=O)-, carbocyclylalkyl-OC(=0)-, heterocarbocyclylalkyl-OC(=O)-, or an amino protecting group; wherein R10 is optionally substituted with 1, 2 or 3, R23; alternatively, R9 and R10 together with the N atom to which they are attached form a heterocarbocyclyl group;
Q is -B(OH)2, -B(OR14)2, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
R14 is H, Cι-C alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
X is RAC(=O)-, RANHC(=O)-, RAS(=O)2-, RAOC(=0)-, RASC(=O)-, or RA;
RA is Cι-C20 alkyl optionally substituted with R20; C2-C20 alkenyl optionally substituted with R20; C2-C20 alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R21; or heterocarbocyclyl optionally substituted with 1-5 R21;
R20 is selected from the group consisting of: -CN, halo, haloalkyl-, Q-C4 alkyl, C2-C alkenyl, C2-C4 alkynyl, -CO2H, -C(=0)CO2H, -C(=O)NH2, -C(=O)H, -S(=O)NH2, -S(=O)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)NH2, -NHC(=O)R20a, -NHC(=O)OR20a, -OR20a, -SR20a, -S(=O)R20 , -S(=O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(=O)2R20a, -NHR20b, phthalimido, -(0-alkyl)r, -O-alkyl-OH, -(O-alkyl)r-OH, X -OR20c, -SR20c, -O-alkyl-R20c, -S-alkyl-R20c, -S(-O)-R20c, -S(=O)2-R20c, -S(=O)2-NHR 0c, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR20c, carbocyclyl optionally substituted with 1-5 R21; and heterocarbocyclyl optionally substituted with 1-5 R21;
R20a is Cι-C20 alkyl, C -C2o alkenyl, or C -C2o alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, C1-C4 alkyl, aryl, heteroaryl or -NHR20b;
R20b is an amino protecting group;
R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22;
R21 is selected from the group consisting of: Cι-C20 alkyl, C2-C2o alkenyl, C2-C20 alkynyl, Cι-C20 alkoxy, CrC20 thialkoxy, -OH -CN, halo, haloalkyl, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=0)0-alkyl, -NHC(=O)aIkyl, -C(=O)O-alkyl, -C(=O)alkyl, -S(=O)-alkyl, -S(=O)2-alkyl, -S(=O)-aryl, -S(=O)2-aryl, carbocyclyl optionally substituted with 1-5 R22, and heterocarbocyclyl optionally substituted with 1-5 R22;
R22 is selected from the group consisting of: Ci- o alkyl, C2-Cιo alkenyl, C2-Cι0 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=0)-, alkyl-C(=O)-, aryl-OC(=0)-, alkyl-OC(=0)NH-, aryl-OC(=O)NH-, alkyl-C(=0)NH-, alkyI-C(=0)0-, (alkyl-O)r-alkyl, HO-(alkyl-0)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=O)-, alkyl-S(=O)2-, H2NS(=0)-, and H2NS(=O)2-;
R23 is selected from the group consisting of: CrCe alkyl, C2-C6 alkenyl, C2-C6 alkynyl, F, CI, Br, I, haloalkyl, -NH2, -NHR23a, -N(R23a)2, -N3, -N02, -CN, -CNO, -CNS, -C(=O)OR23a, -C(=0)R23a, -OC(=0)R23a, -N(R23a)C(=O) R23a, -C(=O)N(R23a)2, ureido, -OR23a, -SR23 , -S(=0)2-(Cι-C6 alkyl), -S(=0)2-aryl, and -S(=0)2-N(R23a)2;
R23a is H or CrCe alkyl; alternatively, two R23a may be combined, together with the N atom to which they are attached, to form a 5 to 7 membered heterocyclic group; and r is 2, 3, 4, or 5.
69. The compound of Claim 68 wherein R1 is 2-propyl.
70. The compound of Claim 68 wherein Q is -B(OH)2.
71. The compound of Claim 68 wherein Q is pinanediol boronic ester.
72. The compound of Claim 68 wherein X is RAC(=0)-.
73. The compound of Claim 68 wherein R2 is -CH2NH-C(=O)OCH2(C6H5).
74. The compound of Claim 68 wherein X is
Figure imgf000274_0001
and RA is C4-Cι6 alkyl.
75. The compound of Claim 68 wherein X is RAC(=0)- and RA is aryl optionally substituted with 1-3 R21.
76. The compound of Claim 68 wherein X is RAC(=O)- and RA is a heterocarbocyclyl group optionally substituted with 1-3 R21.
77. The compound of Claim 68, or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein: R1 is 2-propyl; Q is pinanediol boronic ester; X is RAC( ))-; and
RA is C4- 6 alkyl; aryl optionally substituted with 1-3 R21; or heterocarbocyclyl group optionally substituted with 1-3 R21.
78. A compound of claim 1 having Formula (1):
Figure imgf000275_0001
or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein:
R1 is Ci-Cs alkyl, C2-C8 alkenyl, C2-C8 alkynyl, or C3-C7 cycloalkyl;
R2 is H;
Q is -B(OH)2, -B(OR14)2, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O; R14 is H, C1-C4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl; X is RAC(=O)-, RANHC(=O , RAS(=0)2-, RAOC(=O)-, RASC(=O)-, or RA; RA is Cι-C o alkyl optionally substituted with R20; C2-C20 alkenyl optionally substituted with R20; C2-C20 alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22; R20 is selected from the group consisting of: -OR20a, -SR20a, -S(=O)R20a, -S(-O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, phthalimido, -(0-alkyl)r, -O-alkyl-OH, -(0-alkyl)r-OH, -OR20c, -SR20c, -O-alkyl-R 0c, -S-alkyl-R20c, -S(=O)-R 0c, -S(=O)2-R20c, -S(=O)2-NHR200, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR20°, carbocyclyl optionally substituted with 1-5 R22; and heterocarbocyclyl optionally substituted with 1-5 R22; R20 is Ct-C^ alkyl, C2-C20 alkenyl, or C2-C20 alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, - alkyl, aryl, heteroaryl or -NHR20b;
R 0c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22;
R22 is selected from the group consisting of: Cι-Cι0 alkyl, C2-C10 alkenyl, C2-Cιo alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=0)-, alkyl-C(=0)-, aryl-OC(=O)-, alkyl-OC(=O)NH-, aryl-OC(=O)NH-, alkyl-C(=O)NH-, alkyl-C(=O)0-, (aIkyl-O)r-alkyl, HO-(alkyl-O)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=O)-, alkyl-S(=0)2-, H2NS(=O)-, and H2NS(=O)2-; and r is 2, 3, 4, 5, 6, 7, 8, 9, or 10.
79. The compound of Claim 78 wherein: X is RAC(=O)-, RANHC(=O)-, RAS(=O)2-, or RA; RA is Cι-C1 alkyl optionally substituted with R20; R20 is -O-alkyl, -(O-alkyl)r, -O-alkyl-OH, or -(O-alkyl)r-OH; and r is 2, 3, 4, or 5.
80. The compound of Claim 78 wherein said O-alkyl is methoxy, ethoxy, or propoxy.
81. A compound of Claim 1 of Formula (I):
Figure imgf000276_0001
(I) or pharmaceutically acceptable salt, stereoisomeric or tautomeric form thereof, wherein:
R1 is 2-propyl;
R2 is -CH2CH2CH2NHC(=NH)NH-N02, -CH2CH2CH2NHC(=O)NH2, -CH(CH3)OH, -CH2CONH2, -CH2NH2, or -CH2NR9R10;
R9 is H;
R10 is methyl-C(=O)-, ethyl-C(=O)-, propyl-C(=O)-, butyl-C(=O)-, pentyl-C(=0)-, 2-(ethoxycarbonyl)ethyl-C(=0)-, 4-methyl-phenyl-C(=0)-, cyclopropyl-C(=O)-, 4-fluoro-phenyl-C(=O)-, 4-H2NSO2-ρhenyl-C(=O)-, 4-H3CSO2-phenyl-C(=O)-, 4-ρhenyl-phenyl-C(=O)-, 3,4-dimethoxy-benzyl-C(=O)-, 3-pyridinyl-C(=O)-, 2-(hydroxy)-pyridin-3-yl-C(=0)-, 6-(morpholino)-pyridin-3-yl-C(=0)-, 2-(pyridin-4-yl)thiazol-4-yl-C(=0)-, 2-pyrazinyl-C(=0)-, 2,5-dimethyl-pyrazolyl-C(=O)-, N-methyl-2-pyrrolyl-C(=O)-, 2-pyrrolidinyl-C(=0)-, 2-thiophenyl-C(=O)-, 5-isoxazolyl-C(=O)-, 4-(tetrazol-5-yl) ρhenyl-C(=O)-, (5-tetrazolyl)CH2-C(=O)-, N-H3CSO2-piperidinyl-C(=O)-, butyl-OC(=O)-, (benzyl)-OC(=O)-, (9-fluorenylmethyl)-OC(=O)-, pentyl-NHC(=O)-, propyl-NHC(=O)-, phenyl-NHC(=O)-, 4-methyl-phenyl-NHC(=O)-, methyl-S(=O)2-, 4-fluoro-phenyl-S(=0)2-, 4-cyano-phenyl-S(=0) -, 1 -methyl-imidazol-4-yl-S(=O)2-, 2-thiophenyl-S(=O)2-, (4-methyl-phenyl)-NHC(=O)NH-S(=O)2-, and (4-methyl-phenyl)-S(=0)2NHC(=0)-, alternatively, R9 and R10 together with the N atom to which they are attached form pyrrolyl or pyrazolyl;
Q is -B(OH)2, pinanediol boronic ester, bicyclohexyl- 1,1 '-diol boronic ester, or 1,2- dicyclohexyl-ethane-l,2-diol boronic ester;
X is RAC(=0)-, RANHC(=O)-, RAS(=O)2-, or RAOC(=O)-;
R is CH3-, C2H5-, C3H7-, C4H -, CsHπ-, C63-, C75-, C8H1 -, C9Hι -, CιoH21-, CιιH23-, C12H25-, C13H27-, adamantyl-, bicycloheptanyl-, Cι-3 alkyl substituted with R20; C2-ιo alkenyl substituted with R20; cyclopropyl substituted with 0-3 R21; cyclopentyl substituted with 0-2 R21; cyclohexyl substituted with 0-2 R21 ; phenyl substituted with 0-3 R21; naphthyl- substituted with 0-2 R21; pyrazinyl substituted with 0-1 R21; quinolinyl substituted with 0-1 R21; imidazolyl substituted with 0-1 R21; tetrahydrofuranyl substituted with 0-1 R21; oxothiazolidinyl substituted with 0-1 R21; benzothiazolyl substituted with 0-1 R21; thiazolyl substituted with 0-2 R21; furanyl substituted with 0-2 R21; pyrrolidinyl substituted with 0-1 R21; piperidinyl substituted with 0-1 R21; piperazinyl substituted with 0-1 R21; or pyridinyl substituted with 0-1 R21;elected from the group consisting of: hydroxy-, methoxy-, ethoxy-, propoxy-, butoxy-, pentoxy-, hexyloxy-, heptyloxy-, octyloxy-, methoxyethoxy-, methoxyethoxyethoxy-, methyl-S-, ethyl-S-, octyl-S-, methyl-C(=O)S-, (acetylamino)methyl-S-, amino-, methylamino-, dimethylamino-, methyl-C(=O)-, phenyl-C(=0)-, (H3CSO2)phenyl-C(=0)-, thiophenyl-C(=0)-, methyl-OC(=0)-, ethyl-OC(=O)-, butyl-OC(=O)NH-, methyl-C(=O)NH-, methoxyethoxy-methyl-C(=0)NH-, H2NC(=O)-, methyI-NHC(=0)-, ethyl-NHC(=O)-, propyl-NHC(=O)-, phenyl-NHC(=0)-, H2NC(=0)NH-, H2NS(=O)2-, octyl-S(=O)2-, phenyl-S(=O)2-, methylphenyI-S(=O)2-, thiophenyl-S(=O)2-, cyclopentyl-, cyclohexyl-, cycloheptyl-, adamantyl-, bicycloheptanyl-, cyclopentenyl-, phenyl-, methoxy-phenyl-, methyl-phenyl-, dimethyl-phenyl-, ethyl-phenyl-, propyl-phenyl-, butyl-phenyl-, fluoro-phenyl-, difluoro-phenyl-, chloro-phenyl-, bromo-phenyl-, iodo-phenyl-, dimethylamino-phenyl-, cyclohexyloxy-, 2-isopropyl-5-methyl-cyclohexyloxy-, naphthyl-, methoxynaphthyl-, naphthyloxy-, phenoxy-, (methyl-phenyl)oxy-, (ethyl-phenyl)oxy-, (propyl-ρhenyl)oxy-, (butyl-phenyl)oxy-, (fluoro-phenyl)oxy-, (chloro-phenyl)oxy-, (bromo-phenyl)oxy-, naphthyl-S-, benzyl-S-, (methyl-phenyl)methyl-S-, pyrimidinyl-S-, piperidinyl-, N-methyl-piperidinyl-, N-propyl-piperidinyl-, phthalimido-, thiophenyl-, methyl-thiophenyl-, imidazolyl-, furnayl-, tetrazolyl-, oxopyrrolidinyl-, indolyl-, and methyl-indolyl-; and R2i is selected from the group consisting of: methyl-, ethyl-, propyl-, butyl-, pentyl-, hexyl-, heptyl-, ethenyl-, propenyl-, butenyl-, methoxy-, ethoxy-, propoxy-, phenoxy-, fluoro-, chloro-, bromo-, methyl-C(=O)-, butyl-OC(=O)-, butyl-OC(=O)NH-, phenyl-, methoxyphenyl- fluorophenyl-, chlorophenyl-, bromophenyl-, pyrrolyl-, and pyridinyl-.
82. A compound selected from:
Figure imgf000279_0001
Figure imgf000280_0001
D.3.6 (2S)-2-Phenylbutanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a, 5, 5 -trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.7 Dodecanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.8 Octanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.9 Acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano- 1 ,3,2-benzodioxaborol-2-yl]-3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.10 4-(l , 1 -Dimethylethyl)cyclohexanecarboxamide, N-[(l S)-l -[[[(1R)- 1 - [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.11 trans-4-Pentylcyclohexanecarboxamide, N-[(l S)- 1 -[[[(1R)- 1 - [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.12 4-Phenylbutanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.13 2-(3-Methoxyphenyl)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methylJamino]butyl]
D.3.14 4-(l,l-Dimethylethyl)benzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.15 Nonanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.16 (RS)-2-Cyclopentylhexanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl] D.3.17 Thiophene-2-carboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.18 2,3-Difluorobenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.19 2-(2-Iodophenyl)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a, 5,5 -trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl
D.3.20 Cyclohexanecarboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.21 2-(4-Bromophenyl)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.22 Benzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.23 2-Methylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- . 3 a, 5, 5-trimethy 1-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.24 4-Bromobenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.25 (2S)-2-Phenylpropanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.26 (E)-2-Methyl-3-phenyl-acrylamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.27 2-[(Naphthalen-2-yl)oxy]acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methyIbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.28 2,2-Dimethylbutanamide, N-[(l S)-l -[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl] D.3.29 2-(2-Chlorophenyl)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.30 5-Methylthioρhene-2-carboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.31 cis-3-(2-Methoxyphenyl)acrylamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S56S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.32 (2-Methylphenoxy)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyljamino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.33 2-(2,5-Dimethylphenyl)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.34 trans-3-(2-Bromophenyl)acrylamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a, 5 ,5 -trimethyl-4, 6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.35 4-Isopropylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a, 5,5 -trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl3
D.3.36 4-(4-methylphenyl)butanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.37 2-(2-Naphthylsulfanyl)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.38 5-Methylhexanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.39 3-Thiophen-2-yl-propanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.40 2,4-Dimethylthiazole-5-carboxamide, N-[(l S)-l -[[[(1R)-1 -[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl] D.3.41 Furan-3-carboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]buryl]
D.3.42 (2R)-2-Phenylpropanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.43 2-Cycloheprylacetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.44 l-Methylcyclopropanecarboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.45 l-Methyl-cyclohexanecarboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.46 2-[(lS,2R,5S)-2-isopropyl-5-methylcyclohexyl]oxyacetamide, N-[(1S)-1- [[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.47 (E)-2-Butenamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.48 3-Methylbutanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.49 3-PhenyIpropanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.50 4-(4-Methoxyphenyl)-butanamide, N-[(l S)- 1 -[[[(1R)- 1 -[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.51 Thiophene-3-carboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.52 2-Thiophen-3-yl-acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl D.3.53 (E)-Penta-2,4-dienoic acid amide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a, 5 ,5 -trimethy 1-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.54 2-(4-Isopropylphenoxy)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.55 2-(4-Ethylphenoxy)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.56 (E)-2-Methylhex-2-enoic acid amide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.57 3-(3-Methylphenyl)acrylamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.58 2-Adamantan-l-ylacetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.59 (RS)-2-Cyclopent-2-enylacetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl
D.3.60 4-Diethylaminobenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.61 (RS)-2-Methylbutanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.62 3-(4-Methylphenyl)acrylamide, N-[( 1 S)-l -[[[( 1R)-1 -[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl] amino]carbonyl] -4- [ [imino(nitroamino)methyl] amino]butyl]
D.3.63 Hexa-2,4-dienoic acid amide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.64 4-Pyrrol-l-yl-benzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a, 5 , 5 -trimethy 1-4,6-methano- 1 , 3,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl] D.3.65 (E)-3-Thiophen-3-yl-acrylamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.66 Hept-2-enamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.67 2-(3,4-Dimethylphenoxy)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a, 5 , 5-trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.68 Dec-9-enamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.69 (E)-Undec-2-enoic acid amide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.70 (E)-Dec-3-enoic acid amide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.71 2,2-Dimethyl-3-(2-methylpropenyl)-cyclopropanecarboxamide, N-[(1S)-1- [[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl] -3 -methylbutyl] aminojcarbonyl] -4- [[imino(nitroamino)methyl]amino]butyl]
D.3.72 2-Methylcyclohexanecarboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a, 5 ,5 -trimethyl-4,6-methano-l ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.73 5-Cyclohexylpentanamide, N-[(l S)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.74 3-Methoxycyclohexanecarboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.75 (3R)-3,7-Dimethyl-oct-6-enoic acid amide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl D.3.76 3-[(4-methylbenzyl)sulfanyl]propanamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.77 (3S)-3,7-Dimethyl-oct-6-enoic acid amide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.78 (RS)-4-Ethyloctanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.79 5-Fluoro-2-methoxybenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.80 2-(4-Bromophenoxy)-acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.81 2-(l-Methyl-lH-indol-3-yl)acetamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.82 Hexahydro-2,5-methanopentalene-3a(lH)-carboxamide, N-[(l S)- 1 -[[[(1R)- l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.83 Bicyclo[2.2. l]heptane-2-carboxamide, N-[(l S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [ [imino(nitroamino)methyl] amino]butyl]
D.3.84 (RS)-2-(4-Chlorophenyl)proρionamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.85 (2S)-2-methylbutanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a, 5 , 5 -trimethy 1-4 ,6-methano- 1 , 3 ,2-benzodioxaborol-2-y 1] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl] D.3.86 (4RS)-l-[(l,l-dimethylethoxy)carbonyl]-piperidine-4-carboxamide, N-[(1S)- l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.87 (RS)-4-Methyloctanamide, N-[(l S)- 1-[[[(1R)-1 -[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.88 2-Fluoro-5-methylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.89 2-(Bicyclo[2.2.1]hept-2-yl)acetamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.90 Cyclopropanecarboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a, 5 ,5 -trimethy 1-4, 6-methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.91 4-Ethoxybenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a, 5 , 5 -trimethyl-4,6-methano- 1 ,3,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.92 (E)-3-(4-Bromophenyl)acrylamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.93 (2S)-2-(6-Methoxynaphthalen-2-yl)-propanamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.94 3-Fluoro-4-methoxybenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.95 4-Fluoro-3-methylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a, 5 , 5-trimethyl-4, 6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.96 Non-2-enoic acid amide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a, 5, 5-trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl] D.3.97 (E)-3-(Naphthalen-2-yl)acrylamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.98 Quinoline-2-carboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.99 l-(4-Methoxyphenyl)-cycloρropanecarboxamide, N-[(l S)-l -[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.101 3-Butenamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.102 Tetradecanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano- 1 ,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.103 3-(lH-Indol-3-yl)-propanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.104 4-Phenoxybutanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.105 5-Oxo-5-phenyl-pentanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a, 5, 5 -trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.106 (2RS)- 1 -(( 1 , l-dimethylethoxy)carbonyl)-piperidine-2-carboxamide, N-[(l S)- l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.107 Pyridine-2-carboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.108 Pyridine-3-carboxamide, N-[(l S)-l -[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a,5 , 5-trimethy 1-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl] D.3.109 Pyridine-4-carboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS54S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.110 (2S)- 1 -((1 , 1 -dimethylethoxy)carbonyl)-piperidine-2-carboxamide, N-[( 1 S)- l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a;5,5-trimethyl-4,6-methano- l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3. Ill (2R)- 1 -((1,1 -dimethylethoxy)carbonyl)-piperidine-2-carboxamide, N-[(l S)- l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.112 3,3-Dimethyl-butanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.113 4-[(Phenylamino)carbonyl]butanamide, N-[(l S)-l -[[[(1R)- 1 - [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.114 2,2-Dimethylpentanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.115 5-Thiophen-2-yl-pentanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a, 5 , 5 -trimethy 1-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butylJ
D.3.116 (3RS)- 1 -((1 , l-dimethylethoxy)carbonyl)-piperidine-3-carboxamide, N-[(l S)- l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- l,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.117 8-Phenyl-octanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl
D.3.118 3-[[(l,l-dimethylethoxy)carbonyl]amino]propanamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.119 Tridecanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl] D.3.120 Succinamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR>hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.121 Pentanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.122 [[[(9H-fluoren-9-yl)methoxy]carbonyl]amino]butanamide, N-[(1S)-1- [[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.123 2-(Dimethylamino)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.124 5-(4-Fluorophenyl)-pentanamide, N-[(l S)-1-[[[(1R)-1 -[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.125 8-Oxo-8-phenyloctanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.126 4-(Thiophen-2-yl)butanamide, N-[(l S)-l -[[[(1R)-1 -[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.127 5-Oxo-5-(thiophen-2-yl)pentanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a, 5, 5 -trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.128 2-(3-Chlorophenyl)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.129 Undecanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.130 4-Heρtylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.131 6-Phenylhexanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]buryl] D.3.132 5-Phenylpentanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.133 10-Hydroxydecanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a, 5 , 5 -trimethy 1-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.134 5-Oxo-5-(4-phenylpiperazin-l-yl)ρentanamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.135 2-(lH-Tetrazol-5-yl)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.136 2-(Tetrazol-l-yl)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.137 2-(Pyrimidin-2-ylsulfanyl)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.138 3-Methylsulfanylpropanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.139 3-(Naphthalen-2-ylsulfanyl)-propanamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.140 2-[(Phenylmethyl)sulfanyl]acetamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.141 6-Oxoheptanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a, 5 ,5 -trimethyl-4,6-methano- 1 ,3,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.142 4-(4-Methanesulfonylphenyl)-4-oxobutanamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl] D.3.143 (2S)-l-Acetylpyrrolidine-2-carboxamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.144 3-Hydroxy-2,2-dimethylproρanamide, N-[( 1 S)- 1 -[[[( 1 R)-l - [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.145 2-Ethylsulfanylacetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.146 3-Ureidopropanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a, 5 , 5 -trimethyl-4,6-methano- 1 , 3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.147 3-Methoxypropanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a, 5, 5-trimethy 1-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.148 2-Methylsulfanylacetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S;6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.149 3H-Imidazole-4-carboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.150 7-Oxo-octanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano- 1 ,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.151 (E)-3-(Imidazol-4'yl)acrylamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.152 (RS)-Tetrahydrofuran-3-carboxamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl] -3 -methylbutyl] amino]carbonyl] -4- [[imino(nitroamino)methyl]amino]butyl]
D.3.153 (E)-3-(2-Methoxyphenyl)acrylamide, N-[(l S)-l -[[[(1R)-1 -[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl] D.3.154 2-Ethoxyacetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a, 5 , 5-trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl] amino] carbonyl] -4-[[imino(nitroamino)methyl] amino]butyl]
D.3.155 -Furan-2-yl-propanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.156 -(Benzenesulfonyl)proρanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.157 -Sulfamoylbutanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a, 5 ,5 -trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.158 (4S)-2-Oxo-l,3-thiazolidine-4-carboxamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [ [imino(nitroamino)methyl] amino]butyl]
D.3.159 (2R)-l-Acetylpyrrolidine-2-carboxamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.160 -[(Acetylamino)methylsulfanyl] -propanamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl]
D.3.161 -(Acetylsulfanyl)hexanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.162 (Thiophene-2-sulfonyl)acetamide,N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.163 -(Acetylamino)butanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a,5 ,5 -trimethy 1-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.164 (2Z)-3-(Propylaminocarbonyl)-2-proρenamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-4- [[imino(nitroamino)methyl]amino]butyl] D.3.165 -(Octylsulfonyl)propanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.166 -(Octylsulfanyl)propanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.167 ,2-Dimethylhexanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a, 5 , 5 -trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.168 -Hydroxyhexanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.169 -Oxopentanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.170 -Oxohexanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.171 Benzothiazole-6-carboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a, 5 , 5 -trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.172 -(Octyloxy)propanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.173 -(2-Oxo-pyrrolidin-l-yl)-acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.174 Benzamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]
D.3.175 -[2-(2-Methoxyethoxy)ethoxy]acetamide, N-[(lS,2R)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2-hydroxypropyl]
D.3.176 -Phenylbutanamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3 a, 5 , 5 -trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-2-hydroxypropyl] D.3.177 (4-Methylphenoxy)acetamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]
D.3.178 Hexanamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]
D.3.179 -Butylbenzamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]
D.3.180 Naphthalene-2-carboxamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]
D.3.181 Hexanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.182 -(4-Methylbenzenesulfonyl)acetamide, N-[(lS)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl] -3 -methylbutyl] amino] carbonyl] -4- [[imino(nitroamino)methyl]amino]butyl]
D.3.183 Heptanamide,N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.184 l-(Carbamoyl)undecanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a, 5 ,5 -trimethyl-4,6-methano- 1 ,3,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.3.185 -(Benzenesulfonyl)acetamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.4.1 Naphthalene-l-sulfonamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.4.2 Naphthalene-2-sulfonamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
D.4.3 Decane-l-sulfonamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-4-[[imino(nitroamino)methyl]amino]butyl]
Figure imgf000297_0001
Figure imgf000298_0001
D.7.15 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[((E)- penta-2,4-dienoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.16 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(3,3- dimethyl-butanoyl)amino] - 1 -oxopentyl] amino] -3 -methylbutyl]
D.7.17 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[5- (2,5-dimethylphenoxy)-2,2-dimethylpentanoyl]amino]-l-oxopentyl]amino]- 3-methylbutyl]
D.7.18 Boronic acid, [(1R)-1 -[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(2,2- dimethylpentanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.19 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[4- (thiophen-2-yl)butanoyl]amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.20 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[5-(4- fluorophenyl)pentanoyl]amino]- 1 -oxopentyl]amino]-3-methylbutyl]
D.7.21 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(2,2- dimethylhexanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.22 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(hex- 2,4-enoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.23 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[3- (thiophen-2-yl)propenoyl]amino] - 1 -oxopentyl]amino]-3 -methylbutyl]
D.7.24 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(5- cyclohexylpentanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.25 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[((3R)- 3,7-dimethyloct-6-enoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.26 Boronic acid, [(IR)- 1 -[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[3-[(4- methylbenzyl)sulfanyl]propanoyl]amino]-l-oxopentyl]amino]-3- methylbutyl]
D.7.27 Boronic acid, [(1R)-1 -[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(4- pyrrol- 1 -ylbenzoyl)amino]- 1 -oxopentyl] amino] -3 -methylbutyl]
D.7.28 Boronic acid, [(1R)-1 -[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(5- fluoro-2-methoxybenzoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.29 Boronic acid, [(1R)-1 -[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[((2S)- 2-methylbutanoyl)amino]- 1 -oxopentylJamino]-3-methylbutyl] D.7.30 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2- [(cyclopropanecarbonyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.31 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(4- ethoxybenzoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.32 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[((E)-3- (4-bromophenyl)prop-2-enoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.33 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[(2S)- 2-(6-methoxynaphthalen-2-yl)-propanoyl] amino] - 1 -oxopentyl] amino] -3 - methylbutyl]
D.7.34 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[l-(4- methoxyphenyl)-cyclopropanecarbonyl]amino]-l-oxopentyl]amino]-3- methylbutyl]
D.7.35 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(3- fluoro-4-methoxybenzoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.36 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[(E)-3- (naphthalen-2-yl)prop-2-enoyl]amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.37 Boronic acid, [(IR)- l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(4- fluoro-3 -methylbenzyl)amino] - 1 -oxopentyl]amino] -3 -methylbutyl]
D.7.38 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2- [[[[[(9H-fluoren-9-yl)methoxy]carbonyl]amino]butanoyl]amino]-l- oxopentyl] amino]-3 -methylbutyl]
D.7.39 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(4- bromobenzoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.40 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(3- butenoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.41 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2- [(undecanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.42 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[4- (acetylamino)butanoyl] amino] - 1 -oxopentyl]amino] -3 -methylbutyl]
D.7.43 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(6- phenylhexanoyl)amino]- 1 -oxopentyl] amino] -3 -methylbutyl] -
D.7.44 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(5- phenylpentanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]- D.7.45 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(3- methoxypropanoyl)amino] - 1 -oxopentyl] amino] -3 -methylbutyl] -
D.7.46 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[2,2- dimethyl-3 -(2-methylpropenyl)-cycloρropanecarbonyl] amino] - 1 - oxopentyl]amino]-3 -methylbutyl] -
D.7.47 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(3- methoxycyclohexanecarbonyl)amino]-l-oxopentyl]amino]-3-methylbutyl]-
D.7.48 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[3- (lH-indol-3-yl)-propanoyl]amino]-l-oxopentyl]amino]-3-methylbutyl]-
D.7.49 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[((RS)- 2-cyclopent-2-enyl-acetyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.50 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(5- thiophen-2-yl-pentanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.51 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(6-oxo- heptanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.52 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(7-oxo- octanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.53 Boronic acid, [(IR)- 1 -[[(2S)-5-[[imino(nitroamino)methyl]amino]-2- [(hexanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.54 Boronic acid, [(1R)-1 -[[(2S)-5-[[imino(nitroamino)methyl]amino]-2- [(heptanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.55 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(3- octyloxy-propanoyl)amino]- 1 -oxopentyl]amino]-3-methylbutyl]
D.7.56 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2- [(benzothiazol-6-carbonyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.57 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2- [(undec-2-enoyl)amino] - 1 -oxopentyl]amino]-3 -methylbutyl]
D.7.58 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(9- decenoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
D.7.59 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2- [(tetradecanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
Figure imgf000302_0001
Figure imgf000303_0001
Figure imgf000304_0001
E.2.4 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[((2R)- 2-phenylbutanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
E.2.5 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[4- (l,l-Dimethylethyl)cyclohexanecarbonyl]amino]-l-oxopentyl]amino]-3- methylbutyl]
E.2.6 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(trans- 4-pentylcyclohexanecarbonyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
E.2.1 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(4- phenylbutanoyl)amino] - 1 -oxopentyl]amino] -3 -methylbutyl]
E.2.8 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(4- (1,1 -dimethylethyl)benzoyl)amino] - 1 -oxopentyl] amino] -3 -methylbutyl]
E.2.9 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2- [(nonanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
E.2.10 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(2- thiophenecarbonyl)amino] - 1 -oxopentyl] amino] -3 -methylbutyl]
E.2.11 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(2,3- difluorobenzoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
E.2.12 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2- [(dodecanoyl)amino]- 1 -oxopentyl]amino]-3-methylbutyl]
E.2.13 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[[2-(2- iodophenyl)acetyl]amino]- 1 -oxopentyl]amino]-3-methylbutyl]
E.2.14 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2- [(cyclohexanecarbonyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
E.2.15 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(2- methylbenzoyl)amino]- 1 -oxopentyl] amino] -3 -methylbutyl]
E.2.16 Boronic acid, [(lR)-l-[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[((2S)- 2-phenylpr opanoy l)amino] - 1 -oxopentyl] amino] -3 -methylbutyl]
E.2.17 Boronic acid, [(IR)- 1 -[[(2S)-5-[[imino(nitroamino)methyl]amino]-2-[(2,2- dimethylbutanoyl)amino]-l-oxopentyl]amino]-3-methylbutyl]
E.2.18 Boronic acid, [(1R)-1 -[[(2S)-5-[[imino(nitroamino)methyl]amino]-2- [(quinoline-2-carbonyl)amino] - 1 -oxopentyl] amino] -3 -methylbutyl]
Figure imgf000306_0001
Figure imgf000307_0001
Figure imgf000308_0001
Figure imgf000309_0001
or a pharmaceutically acceptable salt or free base form thereof.
83. A compound selected from: Example Compound Name No
Figure imgf000310_0001
Figure imgf000311_0001
(D.8.10 4-propoxybenzamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]
P.8.11 3-(3-Pyridyl)benzamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]
P.8.12 6-Phenyl-2-pyridinecarboxamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3 a,5 , 5-trimethyl-4,6-methano- 1 , 3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-2-hydroxypropyl] p.8.13 3-propoxybenzamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]
P.8.14 l-Bromonaphthalene-2-carboxamide, N-[(l S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2-hydroxypropyl] .8.15 6-Bromonaphthalene-2-carboxamide, N-[(lS,2R)-l-[[[(lR)-l- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2-hydroxypropyl].
P.8.16 3-Phenylbenzamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl].
P.8.17 4-(2-Fluorophenyl)benzamide, N-[(lS,2R)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl].
P.8.18 2-Pyrazinecarboxamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a, 5 , 5 -trimethy 1-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-2-carbamoylethyl]
P.8.19 Decanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4, 6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-2-carbamoylethyl]
P.8.20 4-Butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethy 1-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl] amino] carbonyl] -2-carbamoylethyl]
P.16.6 4-Butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4, 6-methano- 1 ,3 ,2-benzodioxaborol-2-y 1] -3 - methylbutyl]amino]carbonyl]-2-[(benzyloxycarbonylamide)ethyl]-
Figure imgf000313_0001
Figure imgf000314_0001
P.24.15 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4, 6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl] amino] carbonyl] -2-(nicotinonylamino)ethyl] -
P.24.16 4-butylbenzamide,N-[(lS)-l-[[[(lR)-l-[3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano,l,3;2-benzodioxaborol-2-yl-]-3- methylbutyl]amino]carbonyl]-2-[(4-sulfonylamino)benzoylamino]ethyl]-
P.24.17 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l-[3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4, 6-methano, 1 ,3,2-benzodioxaborol-2-yl-]-3- methylbutyl]amino]carbonyl]-2-[(lH-tertazol-5-yl-acetylamino]ethyl]- ρ.24.18 4-butylbenzamide,N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl-]-3- methylbutyl]amino]carbonyl]-2-[(4- methysulfonylphenyl)carbonylamino]ethyl]-
P.24.19 Decanamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methy lbutyl] amino] carbonyl] -2-(nicotinonylamino)ethyl] -
P.24.20 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4, 6-methano- 1 ,3,2-benzodioxaborol-2-yl-]-3- methylbutyl]amino]carbonyl]-2-[(4-(2H-tetrazol-5- yl)phenyl)carbonylamino] ethyl] -
P.24.21 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl-]-3- methylbutyl]amino]carbonyl]-2-[(l-isoxazol-5-yl)-carbonylamino]ethyl]- .24.22 4-butylbenzamide,N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl-]-3- methylbutyl]amino]carbonyl]-2-[(4-cyanophenyl)sulfonylamino]ethyl]- .24.23 4-butylbenzamide,N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4, -methano- 1 ,3,2-benzodioxaborol-2-yl-] -3 - methylbutyl]amino]carbonyl]-2-[(l-methyl-lH-imidazole-4-) sulfonylamino]ethyl]-
P.24.24 4-butylbenzamide,N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4, 6-methano- 1 ,3 ,2-benzodioxaborol-2-yl-] -3 - methylbutyl]amino]carbonyl]-2-[(2-thiophene)sulfonylamino]ethyl]-
P.24.25 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethy 1-4, 6-methano- 1 ,3,2-benzodioxaborol-2-yl-] -3 - methylbutyl]amino]carbonyl]-2-(6-morpholin-4-nicotinoylamino)ethyl]- P.24.26 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl-]-3- methylbutyl]amino]carbonyl]-2-(2-pyridin-4-thiazolecarbonylamino)ethyl]-
P.24.27 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl-]-3- methylbutyl]amino]carbonyl]-2-(4-methylphenyluriedosulfonylamino)ethyl]- p.24.28 4-phenoxybenzamide, N-[(l S)-l -[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(benzyloxycarbonylamide)ethyl]-
P.24.29 4-phenoxybenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[4-fluoro-benzenesulfonammide ]ethyl]-
P.24.30 4-phenoxybenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl-]-3- methylbutyl]amino]carbonyl]-2-[(2,5-dimethyl-2H-pyrazole) carbonylamino]ethyl]-
P.24.31 4-phenoxybenzamide,N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-(4-phenylbenzoylamino)ethyl]-
P.24.32 4-butylbenzamide,N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4, 6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-2-(4-phenylbenzoylamino)ethyl]-
P.24.33 4-butylbenzamide,N-[(lS)-l-[[[(lR)-l-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl] -3 - methylbutyl]amino]carbonyl]-2-(3-phenylpropynoylamino)ethyl]- .24.34 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethy 1-4,6-methano- 1 ,3 ,2-benzodioxaborol-2-yl-] -3 - methylbutyl] amino]carbonyl] -2-(2-hydroxy-3 -nicotinoylamino)ethyl] -
P.24.35 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-l,3,2-benzodioxaborol-2-yl-]-3- methylbutyl] amino] carbonyl] -2-(D-piroglutamoylamino)ethyl] -
P .24.36 4-butylbenzamide, N-[(lS)-l-[[[(lR)-l-[(3aS,aS,6S,7aR)-hexahydro-3a,5,5- trimethyl-4, 6-methano- 1 ,3 ,2-benzodioxaborol-2-yl-] -3 - methylbutyl] amino]carbonyl] -2-( 1 -methanesulfonyl-piperidine-4- carbonylamino)ethyl]-
Figure imgf000317_0001
Figure imgf000318_0001
Figure imgf000319_0001
Figure imgf000320_0001
Figure imgf000321_0001
Figure imgf000322_0001
or a pharmaceutically acceptable salt or free base form thereof.
84. A composition comprising a compound according to any one of Claims 1-83 and a pharmaceutically acceptable carrier.
85. A method of inhibiting activity of proteasome comprising contacting a compound according to any one of Claims 1-83 with said proteasome.
86. A method of treating cancer comprising administering to a mammal having or predisposed to said cancer a therapeutically effective amount of a compound according to any one of Claims 1-83.
87. The method of Claim 86 for treating cancer wherein said cancer is selected from skin, prostate, colorectal, pancreas, kidney, ovary, mammary, liver, tongue, lung, and smooth muscle tissue.
88. The method of Claim 86 for treating cancer wherein said cancer is selected from leukemia, lymphoma, non-Hodgkin lymphoma, myeloma, and multiple myeloma.
89. The method of Claim 86 for treating cancer further comprising administering to a mammal having or predisposed to said cancer a therapeutically effective amount of a compound according to any one of Claims 1-82 in combination with one or more antitumor or anticancer agent and/or radiotherapy.
90. A method of inhibiting the degradation of a protein comprising contacting proteasome capable of degrading said protein with a compound according to any one of Claims 1-83.
91. The method of Claim 90 wherein said protein is marked with ubiquitin.
92. The method of Claim 90 wherein said protein is p53.
93. A method of treating accelerated or enhanced proteolysis comprising administering to a mammal having or predisposed to said accelerated or enhanced proteolysis a therapeutically effective amount of a compound according to any one of Claims 1-83.
94. A method of inhibiting activity of transcription factor NF-κB comprising contacting lκB, the inhibitor of transcription factor NF-κB, with a compound according to any one of Claims 1-83.
95. A method of treating a disease or disorder selected from human immunodeficiency virus (HIV) infection or inflammatory disorders resulting from transplantation rejection, arthritis, infection, inflammatory bowel disease, asthma, osteoporosis, osteoarthritis, psoriasis, restenosis, and autoimmune diseases comprising administering to a mammal having or predisposed to said disease or disorder a therapeutically effective amount of a compound according to any one of Claims 1-83.
96. A process for preparing a compound of Formula (II):
Figure imgf000324_0001
(II) wherein: D is absent, O, S, NR16, or CR15eR15f; R1 is Cι-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, or C3-C7 cycloalkyl; R15a, R15b, R15c, Rlsd, R15e, R15f are each, independently, H, Ci-C.o alkyl, C3-C7 cycloalkyl, aryl or heteroaryl, wherein said Ci- o alkyl, C3-Cιo cycloalkyl, aryl or heteroaryl are each optionally substituted by 1, 2, 3 or 4 halo, C]-C4 alkyl, Cι-C4 alkoxy, Cι-C haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; or RI5a and R15b together with the C atoms to which they are attached form C3- CJO cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, - alkyl, Ci-C4 alkoxy, C1-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; or R15c and R15d together with the C atoms to which they are attached form C3- C10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, -C4 alkyl, -C4 alkoxy, -C haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; or R15b and R15c together with the C atoms to which they are attached and the intevening D moiety form aryl, heteroaryl, C3-C10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, Cι-C4 alkyl, Ci- C4 alkoxy, C C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; R16 is H or Cι-C6 alkyl; and p and q are each, independently, 1, 2 or 3; comprising a) reacting a diol of Formula (Il-b):
Figure imgf000325_0001
with an appropriate trialkoxyborane of Formula (II-a):
R170^ ^OR17
OR17 (II-a) wherein each R17 is, independently, Ci-Cio alkyl or C3- 0 cycloalkyl; for a time and under conditions suitable for forming an intermediate of Formula (II-c):
Figure imgf000325_0002
(II-c) and b) reacting the intermediate of Formula (II-c) with either i) a reagent of formula R'C^MX113', wherein M is a metal and XhaI is a halogen, or ii) a reagent of formula R'CH2Li , for a time and under conditions suitable for forming the compound of Formula (II).
97. The process of Claim 96 wherein R17 is C1-C4 alkyl.
98. The process of Claim 96 wherein R!7 is isopropyl.
99. The process of Claim 96 wherein the diol of Formula (Il-b) is pinanediol, pinacol, 1,2-ethanediol, 1,3-propanediol, 1,2-propanediol, 2,3-butanediol, 1,1,2,2-tetramethylethanediol, 1,2-diisopropylethanediol, 5,6-decanediol, 1,2-dicyclohexylethanediol, bicyclohexyl- 1,1 '-diol, diethanolamine, or 1 ,2-diphenyl- 1 ,2-ethanediol.
100. The process of Claim 96 wherein the diol of Formula (11-b) is pinanediol.
101. The process of Claim 96 wherein R1CH2MXhal is R1CH3MgBr.
102. The process of Claim 96 wherein R1 is isopropyl.
103. The process of Claim 96 for preparing a compound of Formula (II-i):
Figure imgf000326_0001
(II-i) comprising a) reacting (IS, 2S, 3R, 5S)-(+)-pinanediol with triisopropoxy borane for a time and under conditions suitable for forming an intermediate of Formula (Il-ii):
Figure imgf000326_0002
(Il-ii) and b) reacting the intermediate of Formula (Il-ii) with isobutyl magnesium bromide for a time and under conditions suitable for forming the compound of Formula (II-i).
104. The compound of Formula (Il-ii) :
Figure imgf000326_0003
(Il-ii).
105. A process for the preparation of compounds of Formula (I):
Figure imgf000327_0001
(I) wherein:
R1 is Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or C3-C7 cycloalkyl;
R2 is -CH2NH2;
Q is -B(OR14) , or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O; R14 is Cι-C4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl; X is RAC(=O)-; RA is Cι-C20 alkyl optionally substituted with R20; C -C20 alkenyl optionally substituted with R20; C2-C20 alkynyl optionally substituted with R20; carbocyclyl optionally substituted with 1-5 R21; or heterocarbocyclyl optionally substituted with 1-5 R21; R20 is selected from the group consisting of: -CN, halo, haloalkyl-, Cι-C4 alkyl, C2-C alkenyl, C2-C4 alkynyl, -CO H, -C(=O)CO2H, -C(=O)NH2, -C(=O)H, -S(=O)NH2, -S(=O)2NH2, -OH, -SH, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)NH2, -NHC(=O)R20a, -NHC(=O)OR20a, -OR20a, -SR20a, -S(=O)R20 , -S(=O)2R20a, -S(=O)2-NHR20a, -SC(=O)R20a, -C(=O)R20a, -C(=O)NHR20a, -C(=O)O-R20a, -NHS(=O)2R20a, -NHR20 , phthalimido, -(O-alkyl)r, -O-alkyl-OH, -(O-alkyl)r-OH, -OR20c, -SR20c, -O-alkyl-R20c, -S-alkyl-R20c, -S(=O)-R20c, -S(=O)2-R20c, -S(=O)2-NHR20c, -SC(=O)R20c, -C(=O)R20c, -C(=O)OR20c, -C(=O)NHR20c, • 91 carbocyclyl optionally substituted with 1-5 R ; and heterocarbocyclyl optionally substituted with 1-5 R21; R20a is Ci-C20 alkyl, C2-C 0 alkenyl, or C2-C20 alkynyl; wherein said alkyl, alkenyl, or alkynyl is optionally substituted by one or more halo, -C4 alkyl, aryl, heteroaryl or -NHR20b; R20b is an amino protecting group;
R20c is carbocyclyl optionally substituted with 1-5 R22; or heterocarbocyclyl optionally substituted with 1-5 R22; R21 is selected from the group consisting of: Cι-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, Cι-C20 alkoxy, C1-C20 thialkoxy, -OH -CN, halo, haloalkyl, -NH2, -NH(alkyl), -N(alkyl)2, -NHC(=O)O-alkyl, -NHC(=O)alkyl, -C(=O)O-alkyl, -C(=O)alkyl, -S(=O)-alkyl, -S(=O)2-alkyl, -S(=O)-aryl, -S(=O)2-aryl, carbocyclyl optionally substituted with 1-5 R22, and heterocarbocyclyl optionally substituted with 1-5 R22; 00
R is selected from the group consisting of: C Cio alkyl, C2-C10 alkenyl, C2-Clo alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(=O)-, alkyl-C(=O)-, aryl-OC(=O)-, alkyl-OC(=O)NH-, aryl-OC(=0)NH-, alkyl-C(=O)NH-, alkyl-C(-O)O-, (alkyl-O)r-alkyl, HO-(alkyl-O)r-alkyl-, -OH, -SH, -CN, -N3, -CNO, -CNS, alkyl-S(=O)-, alkyl-S(=O)2-, H2NS(=0)-, and H2NS(=O)2-; and r is 2, 3, 4, or 5; comprising: reacting a compound of Formula (I) wherein R2 is -CH2NH-C(=O)OCH2(C6H5); with a suitable hydrogenation agent for a time and under conditions suitable for forming the compound of Formula (1) wherein R2 is -CH2NH2, provided the hydrogenation agent is selective for the benzyloxycarbonyl group of R2.
106. The process of claim 105 wherein the hydrogenation agent is Pd/C 10% and HCl in 1,4-dioxane.
PCT/US2004/026407 2003-08-14 2004-08-13 Proteasome inhibitors and methods of using the same WO2005021558A2 (en)

Priority Applications (23)

Application Number Priority Date Filing Date Title
MXPA06001687A MXPA06001687A (en) 2003-08-14 2004-08-13 Proteasome inhibitors and methods of using the same.
EP04786507.6A EP1660507B2 (en) 2003-08-14 2004-08-13 Proteasome inhibitors and methods of using the same
CA2535686A CA2535686C (en) 2003-08-14 2004-08-13 Proteasome inhibitors and methods of using the same
CN2004800301520A CN1867572B (en) 2003-08-14 2004-08-13 Proteasome inhibitors and methods of using the same
DE602004022424T DE602004022424D1 (en) 2003-08-14 2004-08-13 PROTEASOMIN HIBITORS AND METHOD FOR THEIR APPLICATION
PL04786507T PL1660507T5 (en) 2003-08-14 2004-08-13 Proteasome inhibitors and methods of using the same
AT04786507T ATE438650T2 (en) 2003-08-14 2004-08-13 PROTEASOME HIBITORS AND METHOD FOR USE THEREOF
ES04786507.6T ES2330008T5 (en) 2003-08-14 2004-08-13 Proteasome inhibitors and procedures for their use
JP2006523419A JP4917431B2 (en) 2003-08-14 2004-08-13 Proteasome inhibitors and methods of using them
EA200600414A EA010804B1 (en) 2003-08-14 2004-08-13 Boronic acid derivatives (embodiments), processes for preparing thereof, intermediate compounds, composition for inhibiting of proteasome activity, processes for inhibiting of proteasome activity, factor of nf-kb transcription and protein degradation and methods for cancer treatment
BRPI0413582A BRPI0413582B8 (en) 2003-08-14 2004-08-13 compounds, compositions and methods for inhibiting proteasome activity
AU2004268946A AU2004268946B2 (en) 2003-08-14 2004-08-13 Proteasome inhibitors and methods of using the same
NZ545775A NZ545775A (en) 2003-08-14 2004-08-13 Proteasome inhibitors and method of using the same
DK04786507.6T DK1660507T4 (en) 2003-08-14 2004-08-13 Proteasome inhibitors and methods for their use
KR1020067003068A KR101093880B1 (en) 2003-08-14 2004-08-13 Proteasome inhibitors and methods of using the same
SI200431273T SI1660507T1 (en) 2003-08-14 2004-08-13 Proteasome inhibitors and methods of using the same
IL173722A IL173722A (en) 2003-08-14 2006-02-14 Proteasome inhibitors and methods of using the same
IS8343A IS3013B (en) 2003-08-14 2006-03-08 Melting grains and methods of using them
NO20061197A NO20061197L (en) 2003-08-14 2006-03-14 Proteasome inhibitors and methods for using the same
HK06112369.2A HK1091839A1 (en) 2003-08-14 2006-11-10 Proteasome inhibitors and methods of using the same
HRP20090590TT HRP20090590T4 (en) 2003-08-14 2009-11-03 Proteasome inhibitors and methods of using the same
IL217623A IL217623A (en) 2003-08-14 2012-01-19 Substituted amino-carbonyl-boronic compound
NO20150303A NO20150303L (en) 2003-08-14 2015-03-09 proteasome inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US49576403P 2003-08-14 2003-08-14
US60/495,764 2003-08-14
US10/918,664 2004-08-12
US10/918,664 US7576206B2 (en) 2003-08-14 2004-08-12 Proteasome inhibitors and methods of using the same

Publications (2)

Publication Number Publication Date
WO2005021558A2 true WO2005021558A2 (en) 2005-03-10
WO2005021558A3 WO2005021558A3 (en) 2005-05-12

Family

ID=34278521

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/026407 WO2005021558A2 (en) 2003-08-14 2004-08-13 Proteasome inhibitors and methods of using the same

Country Status (25)

Country Link
US (5) US7576206B2 (en)
EP (1) EP1660507B2 (en)
JP (1) JP4917431B2 (en)
KR (1) KR101093880B1 (en)
CN (1) CN102603781A (en)
AT (1) ATE438650T2 (en)
AU (1) AU2004268946B2 (en)
BR (1) BRPI0413582B8 (en)
CA (1) CA2535686C (en)
CY (1) CY1109551T1 (en)
DE (1) DE602004022424D1 (en)
DK (1) DK1660507T4 (en)
EA (1) EA010804B1 (en)
ES (1) ES2330008T5 (en)
HK (1) HK1091839A1 (en)
HR (1) HRP20090590T4 (en)
IL (2) IL173722A (en)
IS (1) IS3013B (en)
MX (1) MXPA06001687A (en)
NO (2) NO20061197L (en)
NZ (1) NZ545775A (en)
PL (1) PL1660507T5 (en)
PT (1) PT1660507E (en)
SI (1) SI1660507T1 (en)
WO (1) WO2005021558A2 (en)

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006086600A1 (en) * 2005-02-11 2006-08-17 Cephalon, Inc. Proteasome inhibitors and methods of using the same
WO2009020448A1 (en) * 2007-08-06 2009-02-12 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
EP2088205A1 (en) 2008-02-11 2009-08-12 Institut National De La Sante Et De La Recherche Medicale (Inserm) PSMB10: A diagnosis marker and therapeutic target of chronic rejection.
WO2010036357A1 (en) * 2008-09-29 2010-04-01 Millennium Pharmaceuticals, Inc. Derivatives of 1-amino-2-cyclobutylethylboronic acid
EP2238973A1 (en) * 2009-04-07 2010-10-13 Cephalon France Lyophilized preparations of proteasome inhibitors
WO2011098963A1 (en) * 2010-02-09 2011-08-18 Ranbaxy Laboratories Limited Process for the preparation of bortezomib
WO2010149688A3 (en) * 2009-06-24 2011-08-25 Pharma Mar, S.A. Antitumoral compounds
WO2011133479A2 (en) 2010-04-19 2011-10-27 Niiki Pharma Inc. Combination therapy with a proteasome inhibitor and a gallium complex
JP2012140439A (en) * 2005-07-05 2012-07-26 Trustees Of Tufts College Inhibitor of fibroblast activation protein alpha
CN102844313A (en) * 2010-01-28 2012-12-26 哈佛大学校长及研究员协会 Compositions and methods for enhancing proteasome activity
US8513218B2 (en) 2010-03-31 2013-08-20 Millennium Pharmaceuticals, Inc. Derivatives of 1-amino-2-cyclopropylethylboronic acid
US8530694B2 (en) 2007-08-06 2013-09-10 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
US8541590B2 (en) 2009-12-22 2013-09-24 Cephalon, Inc. Proteasome inhibitors and processes for their preparation, purification and use
EP2730579A1 (en) 2008-06-17 2014-05-14 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
WO2014072985A1 (en) * 2012-11-06 2014-05-15 Natco Pharma Limited Novel boronic acid derivatives as anti cancer agents
EP3021120A1 (en) 2009-02-20 2016-05-18 Michael P. Lisanti Diagnosis, prognosis, therapeutics and methods for treating neoplastic deiseases comprising determining the level of caveolin-1 in a stromal cell sample
WO2016130724A1 (en) 2015-02-11 2016-08-18 Millennium Pharmaceuticals, Inc. Novel crystalline form of a proteasome inhibitor
US9556166B2 (en) 2011-05-12 2017-01-31 Proteostasis Therapeutics, Inc. Proteostasis regulators
US9670150B2 (en) 2012-04-16 2017-06-06 Allergan, Inc. (2-ureidoacetamido)alkyl derivatives as formyl peptide receptor 2 modulators
US9850262B2 (en) 2013-11-12 2017-12-26 Proteostasis Therapeutics, Inc. Proteasome activity enhancing compounds
US9849135B2 (en) 2013-01-25 2017-12-26 President And Fellows Of Harvard College USP14 inhibitors for treating or preventing viral infections
US10022372B2 (en) 2013-04-19 2018-07-17 Thomas Jefferson University Caveolin-1 related methods for treating glioblastoma with temozolomide
WO2018136401A1 (en) * 2017-01-18 2018-07-26 Principia Biopharma Inc. Immunoproteasome inhibitors
US10085987B2 (en) 2012-01-27 2018-10-02 Thomas Jefferson University MCT protein inhibitor-related prognostic and therapeutic methods
WO2019099582A1 (en) * 2017-11-16 2019-05-23 Principia Biopharma Inc. Immunoproteasome inhibitors
WO2019099576A1 (en) * 2017-11-16 2019-05-23 Principia Biopharma Inc. Immunoproteasome inhibitors
EA034601B1 (en) * 2007-08-06 2020-02-25 Милленниум Фармасьютикалз, Инк. Process for producing boronic acids
AU2018233007B2 (en) * 2007-08-06 2020-07-23 Takeda Pharmaceutical Company Limited Proteasome inhibitors
US11241448B2 (en) 2014-05-20 2022-02-08 Millennium Pharmaceuticals, Inc. Methods for cancer therapy
US11560422B2 (en) 2013-12-27 2023-01-24 Zymeworks Inc. Sulfonamide-containing linkage systems for drug conjugates
US11591405B2 (en) 2014-09-17 2023-02-28 Zymeworks Bc Inc. Cytotoxic and anti-mitotic compounds, and methods of using the same
US11617777B2 (en) 2013-03-15 2023-04-04 Zymeworks Bc Inc. Cytotoxic and anti-mitotic compounds, and methods of using the same

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7223745B2 (en) * 2003-08-14 2007-05-29 Cephalon, Inc. Proteasome inhibitors and methods of using the same
DE102005005397B4 (en) * 2005-02-05 2008-08-21 Lts Lohmann Therapie-Systeme Ag Isolation of N-butylbenzenesulfonamide, synthesis of benzenesulfonamide derivatives, and use of N-butylbenzenesulfonamide and benzenesulfonamide derivatives for the treatment of benign prostatic hyperplasia and / or prostate carcinoma
WO2009026579A1 (en) * 2007-08-23 2009-02-26 Cornell Research Foundation, Inc. Proteasome inhibitors and their use in treating pathogen infection and cancer
US7838673B2 (en) * 2007-10-16 2010-11-23 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
CA2763471A1 (en) * 2009-05-27 2010-12-02 Cephalon, Inc. Combination therapy for the treatment of multiple myeloma
ES2768990T3 (en) 2009-07-20 2020-06-24 Bristol Myers Squibb Co Combination of anti-CTLA4 antibodies with gemcitabine for the synergistic treatment of proliferative diseases
JP2013508397A (en) * 2009-10-22 2013-03-07 ポリメディックス・インコーポレーテッド Method for preparing polymer compound
JP2011184344A (en) * 2010-03-08 2011-09-22 Kao Corp p21 EXPRESSION PROMOTER
TW201309303A (en) 2011-03-03 2013-03-01 Cephalon Inc Proteasome inhibitor for the treatment of lupus
CA2833775A1 (en) 2011-06-22 2012-12-27 Cephalon, Inc. Proteasome inhibitors and processes for their preparation, purification and use
ES2820714T3 (en) 2011-10-26 2021-04-22 Allergan Inc Substituted N-urea amino acid amide derivatives as modulators of formylated peptide receptor 1 (FPRL-1) receptor
CN104126017A (en) 2011-11-11 2014-10-29 米伦纽姆医药公司 Biomarkers of response to proteasome inhibitors
EP2776043B1 (en) 2011-11-11 2018-02-21 Millennium Pharmaceuticals, Inc. Biomarkers of response to proteasome inhibitors
EP2810066B1 (en) 2012-01-24 2019-07-31 Millennium Pharmaceuticals, Inc. Methods of treatment of cancer
JP2013006855A (en) * 2012-09-03 2013-01-10 Millennium Pharmaceuticals Inc Proteasome inhibitor
SG11201507113RA (en) 2013-03-06 2015-10-29 Allergan Inc Use of agonists of formyl peptide receptor 2 for treating ocular inflammatory diseases
CA2899804A1 (en) 2013-03-06 2014-09-12 Allergan, Inc. Use of agonists of formyl peptide receptor 2 for treating dermatological diseases
SG11201605260VA (en) 2013-12-27 2016-07-28 Zymeworks Inc Var2csa-drug conjugates
US9493439B1 (en) 2014-04-07 2016-11-15 University Of Kentucky Research Foundation Proteasome inhibitors
MX2017003999A (en) * 2014-10-01 2018-02-09 Merck Patent Gmbh Boronic acid derivatives.
CA2992981C (en) 2015-07-23 2023-10-17 Taipei Medical University Aminonaphthoquinone compounds and pharmaceutical composition for blocking ubiquitination-proteasome system in diseases
BR112018001424B1 (en) 2015-07-30 2024-02-06 Calgent Biotechnology Co., Ltd COMPOUNDS, PHARMACEUTICAL COMPOSITION AND USES OF A COMPOUND
CN106588965A (en) * 2015-10-15 2017-04-26 北京大学 Urea peptidomimetic boric acid compound as well as pharmaceutical composition, preparation method and application thereof
CN105732683B (en) * 2016-03-25 2018-10-16 南京林业大学 The dipeptide boronic acid and its ester type compound, preparation method and its usage of a kind of carboxylic acid and alpha amino acid composition
CN106008572B (en) * 2016-05-23 2018-08-17 成都千禧莱医药科技有限公司 A kind of dipeptide boronic acid compound and preparation method and purposes
JP6223508B2 (en) * 2016-06-27 2017-11-01 ミレニアム ファーマシューティカルズ, インコーポレイテッドMillennium Pharmaceuticals, Inc. Proteasome inhibitor
CN108440583B (en) * 2017-01-23 2020-12-04 成都奥璟生物科技有限公司 Boric acid derivative and its medicinal composition
KR20190127681A (en) * 2017-02-17 2019-11-13 프레세니어스 카비 온콜로지 리미티드 Improved Preparation of Boric Acid Ester
BR112020005079A2 (en) 2017-09-14 2020-09-15 Glaxosmithkline Intellectual Property Development Limited combination treatment for cancer
JP2018024694A (en) * 2017-10-03 2018-02-15 ミレニアム ファーマシューティカルズ, インコーポレイテッドMillennium Pharmaceuticals, Inc. Proteasome inhibitor
US10537585B2 (en) 2017-12-18 2020-01-21 Dexcel Pharma Technologies Ltd. Compositions comprising dexamethasone
CN113105486B (en) * 2021-02-24 2023-08-15 南京师范大学 Boric acid ester compound and pharmaceutically acceptable salt thereof, and preparation method and application thereof
WO2023220641A2 (en) 2022-05-11 2023-11-16 Juno Therapeutics, Inc. Methods and uses related to t cell therapy and production of same
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996014857A1 (en) * 1994-11-14 1996-05-23 Cephalon, Inc. Multicatalytic protease inhibitors
WO1999030707A1 (en) * 1997-12-16 1999-06-24 Cephalon Inc. Multicatalytic protease inhibitors for use as anti-tumor agents
WO2003033507A1 (en) * 2001-10-12 2003-04-24 Kyorin Pharmaceutical Co., Ltd. Benzylmalonic acid derivatives and proteasome inhibitors contaiing the same
WO2005016859A2 (en) * 2003-08-14 2005-02-24 Cephalon, Inc. Proteasome inhibitors and methods of using the same

Family Cites Families (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3301354A1 (en) 1983-01-18 1984-07-19 Matth. Hohner Ag, 7218 Trossingen ELECTRONIC MUSIC INSTRUMENT
US4537773A (en) 1983-12-05 1985-08-27 E. I. Du Pont De Nemours And Company α-Aminoboronic acid derivatives
US4499082A (en) * 1983-12-05 1985-02-12 E. I. Du Pont De Nemours And Company α-Aminoboronic acid peptides
JPS61233689A (en) * 1985-03-22 1986-10-17 メルク・パテント・ゲゼルシヤフト・ミツト・ベシユレンクテル・ハフツング Heterocyclic boron compound
US5242904A (en) 1987-06-05 1993-09-07 The Dupont Merck Pharmaceutical Company Peptide boronic acid inhibitors of trypsin-like proteases
US5187157A (en) 1987-06-05 1993-02-16 Du Pont Merck Pharmaceutical Company Peptide boronic acid inhibitors of trypsin-like proteases
US5250720A (en) 1987-06-05 1993-10-05 The Dupont Merck Pharmaceutical Company Intermediates for preparing peptide boronic acid inhibitors of trypsin-like proteases
EP0315574A3 (en) 1987-11-05 1990-08-22 Hoechst Aktiengesellschaft Renin inhibitors
AU3418389A (en) 1988-03-28 1989-10-16 Regents Of The University Of California, The Nerve growth factor peptides
US5023236A (en) 1988-04-07 1991-06-11 Corvas, Inc. Factor VII/VIIA active site inhibitors
US4963655A (en) 1988-05-27 1990-10-16 Mayo Foundation For Medical Education And Research Boron analogs of amino acid/peptide protease inhibitors
US5159060A (en) 1988-05-27 1992-10-27 Mayo Foundation For Medical Education And Research Cytotoxic boronic acid peptide analogs
US5106948A (en) 1988-05-27 1992-04-21 Mao Foundation For Medical Education And Research Cytotoxic boronic acid peptide analogs
DE3827340A1 (en) 1988-08-12 1990-02-15 Hoechst Ag USE OF (ALPHA) -AMINOBORONIC ACID DERIVATIVES FOR THE PROPHYLAXIS AND TREATMENT OF VIRUS DISEASES
HUT62312A (en) 1990-03-05 1993-04-28 Cephalon Inc Process for producing chymotrypsin-like proteases and their inhibitors
EP0583536B1 (en) 1992-08-14 1997-03-05 The Procter & Gamble Company Liquid detergents containing an alpha-amino boronic acid
WO1994009653A1 (en) 1992-11-02 1994-05-11 Quest International B.V. Tobacco material containing micro-organism cells
CA2155554A1 (en) 1993-02-10 1994-08-18 Alfred L. Goldberg Role of atp-ubiquitin-dependent proteolysis in mhc-1 restricted antigen presentation and inhibitors thereof
IL109319A0 (en) 1993-04-27 1994-07-31 Du Pont Merck Pharma Amidino and guanidino substituted boronic acid compounds
US5658885A (en) 1993-04-27 1997-08-19 The Dupont Merck Pharmaceutical Company Amidino and guanidino substituted boronic acid inhibitors of trypsin-like enzymes
EP0699074B1 (en) 1993-04-30 2002-11-13 Merck & Co. Inc. Thrombin inhibitors
US5672582A (en) 1993-04-30 1997-09-30 Merck & Co., Inc. Thrombin inhibitors
FR2707085B1 (en) 1993-06-30 1995-08-18 Adir New alpha amino acid derivatives, process for their preparation and pharmaceutical compositions containing them.
ES2170104T3 (en) 1993-10-01 2002-08-01 Merrell Pharma Inc INHIBITORS OF THE PRODUCTION OF BETA-AMILOID PROTEINS.
IL111175A0 (en) 1993-10-07 1994-12-29 Du Pont Merck Pharma Electrophilic peptide analogs as inhibitors of trypsin-like serine proteases and pharmaceutical compositions containing them
GB9401483D0 (en) 1994-01-26 1994-03-23 Sandoz Ltd Organic compounds
US5693617A (en) 1994-03-15 1997-12-02 Proscript, Inc. Inhibitors of the 26s proteolytic complex and the 20s proteasome contained therein
US6660268B1 (en) 1994-03-18 2003-12-09 The President And Fellows Of Harvard College Proteasome regulation of NF-KB activity
US6083903A (en) 1994-10-28 2000-07-04 Leukosite, Inc. Boronic ester and acid compounds, synthesis and uses
US5550262A (en) 1994-11-14 1996-08-27 Cephalon, Inc. Multicatalytic protease inhibitors
US5834487A (en) 1996-09-24 1998-11-10 Cv Therapeutics Inhibition of 26S and 20S proteasome by indanones
ZA979327B (en) 1996-10-18 1998-05-11 Vertex Pharma Inhibitors of serine proteases, particularly hepatitis C virus NS3 protease.
GB9623908D0 (en) 1996-11-18 1997-01-08 Hoffmann La Roche Amino acid derivatives
US6096778A (en) 1997-10-07 2000-08-01 Cephalon, Inc. α-ketoamide multicatalytic protease inhibitors
US6075150A (en) 1998-01-26 2000-06-13 Cv Therapeutics, Inc. α-ketoamide inhibitors of 20S proteasome
GB9806815D0 (en) 1998-03-30 1998-05-27 Hoffmann La Roche Amino acid derivatives
US6462019B1 (en) 1998-07-10 2002-10-08 Osteoscreen, Inc. Inhibitors of proteasomal activity and production for stimulating bone growth
EP0995757A3 (en) 1998-08-26 2002-05-22 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Bivalent inhibitors of the proteasome
CA2347275C (en) 1998-10-20 2010-03-09 Millennium Pharmaceuticals, Inc. Method for monitoring proteasome inhibitor drug action
CA2359561A1 (en) 1999-01-20 2000-07-27 Shiro Akinaga Proteasome inhibitors
EP1053750A1 (en) 1999-04-22 2000-11-22 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Use of proteasome-inhibitor for the induction of programmed cell death (apoptosis)
ES2223512T3 (en) 1999-04-27 2005-03-01 Novartis Ag USE OF 2,4-DIAMINO-3-HYDROXICARBOXYLIC ACID DERIVATIVES AS INHIBITORS OF PROTEASOMA.
SE9901573D0 (en) 1999-05-03 1999-05-03 Astra Ab New compounds
WO2001002424A2 (en) 1999-07-07 2001-01-11 Du Pont Pharmaceuticals Company Peptide boronic acid inhibitors of hepatitis c virus protease
US7122627B2 (en) 1999-07-26 2006-10-17 Bristol-Myers Squibb Company Lactam inhibitors of Hepatitis C virus NS3 protease
AU7832000A (en) 1999-09-23 2001-04-24 Washington University Compounds directed against pilus biogenesis and activity in pathogenic bacteria;methods and compositions for synthesis thereof
WO2001028579A2 (en) 1999-10-20 2001-04-26 Osteoscreen, Inc. Inhibitors of proteasomal activity for stimulating bone and hair growth
AU8063701A (en) 2000-07-21 2002-02-05 Schering Corp Novel peptides as NS3-serine protease inhibitors of hepatitis C virus
AU2002218133A1 (en) 2000-10-12 2002-04-22 Viromics Gmbh Agents for the treatment of viral infections
EP1360189A1 (en) 2001-01-25 2003-11-12 Millennium Pharmaceuticals, Inc. Formulation of boronic acid compounds
CN1531653A (en) 2001-04-09 2004-09-22 ����������˹�ٰ���Ф Method for identification of proteins from intracellular bacteria
US7214769B2 (en) 2001-05-23 2007-05-08 The Curators Of The University Of Missouri Method for inverse solid phase synthesis of peptides
ATE317850T1 (en) 2001-05-30 2006-03-15 Novartis Pharma Gmbh 2-((N-(2-AMINO-3-(HETEROARYL- OR - ARYL)PROPIONYL)AMINOACYL)AMINO)- ALKYLBORONIC ACID DERIVATIVES
AU2002324734A1 (en) 2001-08-16 2003-03-03 Levente Fabry-Asztalos Borinic acid protease inhibitors
JPWO2003033506A1 (en) * 2001-10-12 2005-02-03 杏林製薬株式会社 Aminoboranoic acid derivatives and proteasome inhibitors containing the same
JP4412586B2 (en) * 2002-01-08 2010-02-10 エーザイ・アール・アンド・ディー・マネジメント株式会社 Eponemycin and epoxomycin analogues and their uses
US7442830B1 (en) * 2007-08-06 2008-10-28 Millenium Pharmaceuticals, Inc. Proteasome inhibitors
NZ582693A (en) 2007-08-06 2012-01-12 Millennium Pharm Inc Boron-containing proteasome inhibitors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996014857A1 (en) * 1994-11-14 1996-05-23 Cephalon, Inc. Multicatalytic protease inhibitors
WO1999030707A1 (en) * 1997-12-16 1999-06-24 Cephalon Inc. Multicatalytic protease inhibitors for use as anti-tumor agents
WO2003033507A1 (en) * 2001-10-12 2003-04-24 Kyorin Pharmaceutical Co., Ltd. Benzylmalonic acid derivatives and proteasome inhibitors contaiing the same
WO2005016859A2 (en) * 2003-08-14 2005-02-24 Cephalon, Inc. Proteasome inhibitors and methods of using the same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE CA [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; SIMOV, BILJANA PERIC ET AL: "Chiral carbanions. Part 4: Borylation of (trimethylsilyl)methyl N,N-dialkylcarbamates - Diastereoselectivity and structural studies" XP002321002 retrieved from STN Database accession no. 142:93882 & SYNTHESIS , (16), 2704-2710 CODEN: SYNTBF; ISSN: 0039-7881, 2004, *

Cited By (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2006213814B2 (en) * 2005-02-11 2011-10-13 Cephalon, Inc. Proteasome inhibitors and methods of using the same
US7468383B2 (en) 2005-02-11 2008-12-23 Cephalon, Inc. Proteasome inhibitors and methods of using the same
US8283367B2 (en) 2005-02-11 2012-10-09 Cephalon, Inc. Proteasome inhibitors and methods of using the same
WO2006086600A1 (en) * 2005-02-11 2006-08-17 Cephalon, Inc. Proteasome inhibitors and methods of using the same
JP2012140439A (en) * 2005-07-05 2012-07-26 Trustees Of Tufts College Inhibitor of fibroblast activation protein alpha
US8871745B2 (en) 2007-08-06 2014-10-28 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
AU2018233007B2 (en) * 2007-08-06 2020-07-23 Takeda Pharmaceutical Company Limited Proteasome inhibitors
EA028622B1 (en) * 2007-08-06 2017-12-29 Милленниум Фармасьютикалз, Инк. Proteasome inhibitors
EA034601B1 (en) * 2007-08-06 2020-02-25 Милленниум Фармасьютикалз, Инк. Process for producing boronic acids
US8772536B2 (en) 2007-08-06 2014-07-08 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
KR101831674B1 (en) 2007-08-06 2018-02-23 밀레니엄 파머슈티컬스 인코퍼레이티드 Proteasome inhibitors
WO2009020448A1 (en) * 2007-08-06 2009-02-12 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
EP2527347A1 (en) 2007-08-06 2012-11-28 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
EP3210987A1 (en) 2007-08-06 2017-08-30 Millennium Pharmaceuticals, Inc. Process for the synthesis of proteasome inhibitors
TWI511972B (en) * 2007-08-06 2015-12-11 Millennium Pharm Inc Proteasome inhibitors
US8530694B2 (en) 2007-08-06 2013-09-10 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
AU2007357338B2 (en) * 2007-08-06 2014-03-20 Takeda Pharmaceutical Company Limited Proteasome inhibitors
EP2088205A1 (en) 2008-02-11 2009-08-12 Institut National De La Sante Et De La Recherche Medicale (Inserm) PSMB10: A diagnosis marker and therapeutic target of chronic rejection.
EP3536693A1 (en) 2008-06-17 2019-09-11 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
US9175018B2 (en) 2008-06-17 2015-11-03 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
US10526351B2 (en) 2008-06-17 2020-01-07 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
EP2730579A1 (en) 2008-06-17 2014-05-14 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
EP2730581A1 (en) 2008-06-17 2014-05-14 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
EP2730580A1 (en) 2008-06-17 2014-05-14 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
US10604538B2 (en) 2008-06-17 2020-03-31 Millenium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
EP2733147A1 (en) 2008-06-17 2014-05-21 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
US11485746B2 (en) 2008-06-17 2022-11-01 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
US9175017B2 (en) 2008-06-17 2015-11-03 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
US8859504B2 (en) 2008-06-17 2014-10-14 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
EP2730581B1 (en) 2008-06-17 2016-04-27 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
EP2318419B1 (en) 2008-06-17 2015-04-08 Millennium Pharmaceuticals, Inc. Boronate ester compounds and pharmaceutical compositions thereof
US8664200B2 (en) 2008-09-29 2014-03-04 Millennium Pharmaceuticals, Inc. Derivatives of 1-amino-2-cyclobutylethylboronic acid
KR101739470B1 (en) 2008-09-29 2017-05-24 밀레니엄 파머슈티컬스 인코퍼레이티드 Derivatives of 1-amino-2-cyclobutylethylboronic acid
US9771381B2 (en) 2008-09-29 2017-09-26 Millennium Pharmaceuticals, Inc. Derivatives of 1-amino-2-cyclobutylethylboronic acid
US10035811B2 (en) 2008-09-29 2018-07-31 Millennium Pharmaceuticals, Inc. Derivatives of 1-amino-2-cyclobutylethylboronic acid
EA030539B1 (en) * 2008-09-29 2018-08-31 Милленниум Фармасьютикалз, Инк. Derivatives of 1-amino-2-cyclobutylethylboronic acid
AU2009297113B2 (en) * 2008-09-29 2016-11-10 Millennium Pharmaceuticals, Inc. Derivatives of 1-amino-2-cyclobutylethylboronic acid
WO2010036357A1 (en) * 2008-09-29 2010-04-01 Millennium Pharmaceuticals, Inc. Derivatives of 1-amino-2-cyclobutylethylboronic acid
EP3021120A1 (en) 2009-02-20 2016-05-18 Michael P. Lisanti Diagnosis, prognosis, therapeutics and methods for treating neoplastic deiseases comprising determining the level of caveolin-1 in a stromal cell sample
WO2010114982A3 (en) * 2009-04-03 2011-09-01 Cephalon, Inc. Lyophilization cakes of proteasome inhibitors
US8785418B2 (en) 2009-04-03 2014-07-22 Cephalon, Inc. Lyophilization cakes of proteasome inhibitors
AU2010232632C1 (en) * 2009-04-03 2016-09-22 Cephalon, Inc. Lyophilization cakes of proteasome inhibitors
AU2010232632B2 (en) * 2009-04-03 2016-04-21 Cephalon, Inc. Lyophilization cakes of proteasome inhibitors
TWI484981B (en) * 2009-04-03 2015-05-21 Cephalon Inc Lyophilized preparations of proteasome inhibitors
EP2238973A1 (en) * 2009-04-07 2010-10-13 Cephalon France Lyophilized preparations of proteasome inhibitors
WO2010149688A3 (en) * 2009-06-24 2011-08-25 Pharma Mar, S.A. Antitumoral compounds
US8541590B2 (en) 2009-12-22 2013-09-24 Cephalon, Inc. Proteasome inhibitors and processes for their preparation, purification and use
CN102844313A (en) * 2010-01-28 2012-12-26 哈佛大学校长及研究员协会 Compositions and methods for enhancing proteasome activity
US10351568B2 (en) 2010-01-28 2019-07-16 President And Fellows Of Harvard College Compositions and methods for enhancing proteasome activity
WO2011098963A1 (en) * 2010-02-09 2011-08-18 Ranbaxy Laboratories Limited Process for the preparation of bortezomib
US9023832B2 (en) 2010-03-31 2015-05-05 Millennium Pharmaceuticals, Inc. Derivatives of 1-amino-2-cyclopropylethylboronic acid
US8513218B2 (en) 2010-03-31 2013-08-20 Millennium Pharmaceuticals, Inc. Derivatives of 1-amino-2-cyclopropylethylboronic acid
US8703743B2 (en) 2010-03-31 2014-04-22 Millennium Pharmaceuticals, Inc. Derivatives of 1-amino-2-cyclopropylethylboronic acid
WO2011133479A2 (en) 2010-04-19 2011-10-27 Niiki Pharma Inc. Combination therapy with a proteasome inhibitor and a gallium complex
US10532996B2 (en) 2011-05-12 2020-01-14 Proteostasis Therapeutics, Inc. Proteostasis regulators
US9556166B2 (en) 2011-05-12 2017-01-31 Proteostasis Therapeutics, Inc. Proteostasis regulators
US10085987B2 (en) 2012-01-27 2018-10-02 Thomas Jefferson University MCT protein inhibitor-related prognostic and therapeutic methods
US9670150B2 (en) 2012-04-16 2017-06-06 Allergan, Inc. (2-ureidoacetamido)alkyl derivatives as formyl peptide receptor 2 modulators
WO2014072985A1 (en) * 2012-11-06 2014-05-15 Natco Pharma Limited Novel boronic acid derivatives as anti cancer agents
US9849135B2 (en) 2013-01-25 2017-12-26 President And Fellows Of Harvard College USP14 inhibitors for treating or preventing viral infections
US11617777B2 (en) 2013-03-15 2023-04-04 Zymeworks Bc Inc. Cytotoxic and anti-mitotic compounds, and methods of using the same
US10022372B2 (en) 2013-04-19 2018-07-17 Thomas Jefferson University Caveolin-1 related methods for treating glioblastoma with temozolomide
US11242361B2 (en) 2013-11-12 2022-02-08 Proteostasis Therapeutics, Inc. Proteasome activity enhancing compounds
US9850262B2 (en) 2013-11-12 2017-12-26 Proteostasis Therapeutics, Inc. Proteasome activity enhancing compounds
US11958873B2 (en) 2013-11-12 2024-04-16 Kineta, Inc. Proteasome activity enhancing compounds
US11560422B2 (en) 2013-12-27 2023-01-24 Zymeworks Inc. Sulfonamide-containing linkage systems for drug conjugates
US11241448B2 (en) 2014-05-20 2022-02-08 Millennium Pharmaceuticals, Inc. Methods for cancer therapy
US11591405B2 (en) 2014-09-17 2023-02-28 Zymeworks Bc Inc. Cytotoxic and anti-mitotic compounds, and methods of using the same
WO2016130724A1 (en) 2015-02-11 2016-08-18 Millennium Pharmaceuticals, Inc. Novel crystalline form of a proteasome inhibitor
US11154563B2 (en) 2017-01-18 2021-10-26 Principia Biopharma Inc. Immunoproteasome inhibitors
WO2018136401A1 (en) * 2017-01-18 2018-07-26 Principia Biopharma Inc. Immunoproteasome inhibitors
AU2018210839B2 (en) * 2017-01-18 2022-01-20 Principia Biopharma Inc. Immunoproteasome inhibitors
US11225493B2 (en) 2017-11-16 2022-01-18 Principia Biopharma Inc. Immunoproteasome inhibitors
KR20200090810A (en) * 2017-11-16 2020-07-29 프린시피아 바이오파마, 인코퍼레이티드 Immunoproteasome inhibitors
WO2019099582A1 (en) * 2017-11-16 2019-05-23 Principia Biopharma Inc. Immunoproteasome inhibitors
WO2019099576A1 (en) * 2017-11-16 2019-05-23 Principia Biopharma Inc. Immunoproteasome inhibitors
AU2018369787B2 (en) * 2017-11-16 2023-04-20 Principia Biopharma Inc. Immunoproteasome inhibitors
US11827656B2 (en) 2017-11-16 2023-11-28 Principia Biopharma Inc. Immunoproteasome inhibitors
KR102647050B1 (en) 2017-11-16 2024-03-14 프린시피아 바이오파마, 인코퍼레이티드 Immunoproteasome inhibitors

Also Published As

Publication number Publication date
EA010804B1 (en) 2008-12-30
DK1660507T4 (en) 2017-03-27
NO20061197L (en) 2006-05-11
PL1660507T3 (en) 2010-01-29
EP1660507B1 (en) 2009-08-05
DE602004022424D1 (en) 2009-09-17
ES2330008T3 (en) 2009-12-03
HRP20090590T1 (en) 2009-12-31
IS8343A (en) 2006-03-08
JP4917431B2 (en) 2012-04-18
HK1091839A1 (en) 2007-01-26
EA200600414A1 (en) 2006-08-25
CN102603781A (en) 2012-07-25
JP2007502304A (en) 2007-02-08
US20090291918A1 (en) 2009-11-26
US20050107307A1 (en) 2005-05-19
US8058262B2 (en) 2011-11-15
IL173722A (en) 2012-06-28
AU2004268946B2 (en) 2012-01-19
CA2535686C (en) 2016-03-22
US7915236B2 (en) 2011-03-29
US7576206B2 (en) 2009-08-18
NO20150303L (en) 2006-05-11
CY1109551T1 (en) 2014-08-13
PL1660507T5 (en) 2017-12-29
HRP20090590T4 (en) 2017-06-16
EP1660507B9 (en) 2010-03-24
US20120041196A1 (en) 2012-02-16
AU2004268946A1 (en) 2005-03-10
MXPA06001687A (en) 2007-04-17
BRPI0413582B8 (en) 2021-05-25
IL173722A0 (en) 2006-07-05
WO2005021558A3 (en) 2005-05-12
ES2330008T5 (en) 2017-06-28
PT1660507E (en) 2009-10-15
KR20060120585A (en) 2006-11-27
US9233115B2 (en) 2016-01-12
NZ545775A (en) 2009-11-27
EP1660507B2 (en) 2017-01-04
US8546608B2 (en) 2013-10-01
DK1660507T3 (en) 2009-11-02
ATE438650T2 (en) 2009-08-15
IL217623A (en) 2015-06-30
KR101093880B1 (en) 2011-12-13
CA2535686A1 (en) 2005-03-10
US20110144033A1 (en) 2011-06-16
US20140088042A1 (en) 2014-03-27
SI1660507T1 (en) 2010-01-29
EP1660507A2 (en) 2006-05-31
IS3013B (en) 2019-05-15
BRPI0413582B1 (en) 2020-01-07
BRPI0413582A (en) 2006-10-17

Similar Documents

Publication Publication Date Title
EP1660507B1 (en) Proteasome inhibitors and methods of using the same
EP1846424B1 (en) Proteasome inhibitors and methods of using the same
EP1658255B1 (en) Proteasome inhibitors and methods of using the same
AU2011265405B2 (en) Proteasome inhibitors and methods of using the same

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200480030152.0

Country of ref document: CN

AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2535686

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: PA/a/2006/001687

Country of ref document: MX

Ref document number: 200601293

Country of ref document: ZA

Ref document number: 12006500325

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 173722

Country of ref document: IL

Ref document number: 1020067003068

Country of ref document: KR

Ref document number: 2006523419

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 545775

Country of ref document: NZ

Ref document number: 2004786507

Country of ref document: EP

Ref document number: 2004268946

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 565/KOLNP/2006

Country of ref document: IN

Ref document number: 00565/KOLNP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 200600414

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 2004268946

Country of ref document: AU

Date of ref document: 20040813

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004268946

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2004786507

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0413582

Country of ref document: BR

WWP Wipo information: published in national office

Ref document number: 1020067003068

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 217623

Country of ref document: IL