WO2018006063A1 - Compositions and methods for the treatment of bacterial infections - Google Patents

Compositions and methods for the treatment of bacterial infections Download PDF

Info

Publication number
WO2018006063A1
WO2018006063A1 PCT/US2017/040467 US2017040467W WO2018006063A1 WO 2018006063 A1 WO2018006063 A1 WO 2018006063A1 US 2017040467 W US2017040467 W US 2017040467W WO 2018006063 A1 WO2018006063 A1 WO 2018006063A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
compound
pharmaceutically acceptable
acceptable salt
acid
Prior art date
Application number
PCT/US2017/040467
Other languages
French (fr)
Inventor
James M. Balkovec
Daniel C. BENSEN
Timothy Blizzard
Allen Borchardt
Thomas P. Brady
Zhi-yong CHEN
Quyen-Quyen Thuy Do
Wanlong Jiang
Thanh Lam
Jeffrey B. LOCKE
Alain Noncovich
Leslie W. TARI
Original Assignee
Cidara Therapeutics, 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
Application filed by Cidara Therapeutics, Inc. filed Critical Cidara Therapeutics, Inc.
Publication of WO2018006063A1 publication Critical patent/WO2018006063A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/50Cyclic peptides containing at least one abnormal peptide link
    • C07K7/54Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring
    • C07K7/60Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring the cyclisation occurring through the 4-amino group of 2,4-diamino-butanoic acid
    • C07K7/62Polymyxins; Related peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K9/00Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof
    • C07K9/006Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof the peptide sequence being part of a ring structure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the disclosure relates to compounds, compositions, and methods for inhibiting bacterial growth (e.g., Gram-negative bacterial growth) and for the treatment of bacterial infections (e.g., Gram-negative bacterial infections).
  • such compounds contain dimers of cyclic heptapeptides conjugated to one or more monosaccharide or oligosaccharide moieties, which interact, directly or indirectly, with an immune cell.
  • compounds containing dimers of cyclic heptapeptides may be conjugated to at least 2 (e.g., 2-1 0, 2-9, 2-8, 2-7, 2-6, 2-5, 2-4, or 2-3) monosaccharide or oligosaccharide moieties (e.g., rhamnose).
  • compounds containing dimers of cyclic heptapeptides conjugated to at least 2 (e.g., 2-1 0, 2-9, 2-8, 2-7, 2-6, 2-5, 2-4, or 2-3) monosaccharide or oligosacchari
  • heptapeptides may be conjugated to at least 3 (e.g., 3-10, 3-9, 3-8, 3-7, 3-6, 3-5, or 3-4) monosaccharide or oligosaccharide moieties (e.g., rhamnose).
  • compounds containing dimers of cyclic heptapeptides may be conjugated to at least 4 (e.g., 4-10, 4-9, 4-8, 4-7, 4-6, or 4-5)
  • LPS lipopolysaccharides
  • the disclosure features a compound described by formula (I):
  • M1 includes a first cyclic heptapeptide including a linking nitrogen and M2 includes a second cyclic heptapeptide including a linking nitrogen; each E is, independently, a monosaccharide or oligosaccharide moiety; L' is a linker covalently attached to E and to the linking nitrogen in each of M1 and M2; and n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
  • L' in formula (I) is described by formula (L):
  • L is a remainder of L';
  • A1 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M1 or is absent; and
  • A2 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M2 or is absent.
  • the compound is described by formula (II):
  • L is a remainder of L'; n is 1 , 2, 3, or 4 (e.g., 1 or 2) ; each of R 1 , R 12 , R' 1 , and R' 12 is, independently, a lipophilic moiety, a polar moiety, or H; each of R 1 1 , R 13 , R 14 , R' 1 1 , R' 13 , and R' 14 is, independently, optionally substituted C1 -C5 alkamino, a polar moiety, a positively charged moiety, or H; each of R 15 and R' 15 is, independently, a lipophilic moiety or a polar moiety; each of R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R' 2 , R' 3 , R' 4 , R' 5 , R' 6 , R' 7 , R' 8 , R' 9 , and R' 10 is, independently, a lipophil
  • heterocycloalkenyl optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or two R groups on the same or adjacent atoms join to form an optionally substituted 5-8 membered ring; each of a', b', c', a, b, and c is, independently, 0 or 1 ; each of N 1 , N 2 , N 3 , N 4 , N' 1 , N' 2 , N' 3 , and N' 4 is a nitrogen atom ; each of C 1 , C 2 , C 3 , C' 1 , C' 2 , and C' 3 is a carbon atom, or a pharmaceutically acceptable salt thereof.
  • the compound includes at least one optionally substituted 5-8 membered ring formed by joining (i) R 2 , R 3 , and C 1 ; (ii) R 3 , R 4 , N 1 , and C 1 ; (iii) R 5 , R 6 , and C 2 ; (iv) R 6 , R 7 , N 2 , and C 2 ; (v) R 8 , R 9 , and C 3 ; (vi) R 9 , R 10 , N 3 , and C 3 ; (vii) R' 2 , R' 3 , and C' 1 ; (viii) R' 3 , R' 4 , N' 1 , and C' 1 ; (ix) R' 5 , R' 6 , and C' 2 ; (x) R' 6 , R' 7 , N' 2 , and C' 2 ; (xi) R' 8 , R' 9 , and C' 3 ; or (xii) R' 9 ,
  • each of R 1 , R 12 , R' 1 , and R' 12 is, independently, a lipophilic moiety; each of R 1 1 , R 13 , R 14 , R' 1 1 , R' 13 , and R' 14 is, independently, optionally substituted C1 -C5 alkamino, a polar moiety, or a positively charged moiety; and/or each of R 15 and R' 15 is, independently, a polar moiety.
  • each of R 1 and R 12 is a lipophilic moiety. In some embodiments, each of R' 1 and R' 12 is a lipophilic moiety. In some embodiments, each lipophilic moiety is, independently, optionally substituted C1 -C20 alkyi, optionally substituted C5-C15 aryl, optionally substituted C6-C35 alkaryl, or optionally C5-C1 0 substituted heteroaryl.
  • each lipophilic moiety is, independently, C1 -C8 alkyi, methyl substituted C2-C4 alkyi, (C1 -C10)alkylene(C6)aryl, phenyl substituted (C1 -C10)alkylene(C6)aryl, or alkyi substituted C4-C9 heteroaryl.
  • each lipophilic moiety is, independently, benzyl, isobutyl, sec-butyl, isopropyl, n-propyl, methyl, biphenylmethyl, n-octyl, or methyl substituted indolyl.
  • each of R 1 1 , R 13 , R 14 , R' 1 1 , R' 13 , and R' 14 is independently optionally substituted C1 -C5 alkamino. In some embodiments, each of R 1 1 , R 13 , R 14 , R' 1 1 , R' 13 , and R' 14 is
  • each of R 15 and R' 15 is a polar moiety.
  • each polar moiety includes a hydroxyl group, a carboxylic acid group, an ester group, or an amide group.
  • each polar moiety is hydroxyl substituted C1 -C4 alkyi.
  • each polar moiety is CH(CH 3 )OH.
  • the compound is described by formula (IV):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl, or a
  • the compound is described by formula (IV-1 ):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl, or a
  • the compound is described by formula (IV-2):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl, or a
  • the compound is described by formula (V):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R 2 , R 6 , R' 2 , and R' 6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally
  • the compound is described by formula (V-1 ):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (V-2):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (V-3):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (V-4):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (V-5):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R 2 , R 6 , R 8 , R' 2 , R' 6 , and R' 8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalky
  • the compound is described by formula (VI-1 ):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (VI-2):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (VI-3):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (VI-4):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (VII):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R 2 , R 6 , R 8 , R' 2 , and R' 6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 - C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally
  • the compound is described by formula (VIII):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R 2 , R 6 , R 8 , and R' 2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8
  • the compound is described by formula (IX):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R 2 , R 6 , and R' 2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 hetero
  • the compound is described by formula (IX-1 ):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (IX-2):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (X):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R 2 and R' 2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkyny
  • the compound is described by formula (XI):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R 2 and R 6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl
  • the compound is described by formula (XI-1 ):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (XI-2):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (XII):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R 2 and R' 2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkyny
  • the compound is described by formula (XII-1 ):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • A1 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M1 ;
  • A2 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M2;
  • E 1 is a first monosaccharide or oligosaccharide moiety;
  • E' 1 is a second monosaccharide or oligosaccharide moiety;
  • L, L 1 , and L' 1 are remainders of L'; or a pharmaceutically acceptable salt thereof.
  • the compound is described by formula (XIII-1 ) :
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g CH2CH(CH3)2), or cyclohexylmethyl; each of R 6 and R' 6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl,
  • the compound is described by formula (XIII-2):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • the compound is described by formula (XIII-3):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
  • A1 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M1 ;
  • A2 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M2, or is absent;
  • L and L 1 are remainders of L'; or a pharmaceutically acceptable salt thereof.
  • R 2 is optionally substituted C1 -C5 alkamino. In some embodiments of the compounds described herein, R' 2 is optionally substituted C1 -C5 alkamino. In some embodiments, the optionally substituted C1 -C5 alkamino is CH2NH2 or CH2CH2NH2.
  • R 2 is a polar moiety. In some embodiments of the compounds described herein, R' 2 is a polar moiety. In some embodiments of the compounds described herein, R 6 is a polar moiety. In some embodiments of the compounds described herein, R' 6 is a polar moiety. In some embodiments, the polar moiety includes a hydroxyl group, a carboxylic acid group, an ester group, or an amide group. In some embodiments, the polar moiety is hydroxyl substituted C1 -C4 alkyl. In some embodiments, the polar moiety is CH(CH3)OH or CH2OH.
  • R 8 is optionally substituted C1 -C5 alkamino. In some embodiments of the compounds described herein, R' 8 is optionally substituted C1 -C5 alkamino. In some embodiments, the optionally substituted C1 -C5 alkamino is CH2NH2 or CH2CH2NH2. In some embodiments of the compounds described herein, R 8 is optionally substituted C5-C1 5 aryl. In some embodiments of the compounds described herein, R' 8 is optionally substituted C5-C15 aryl. In some embodiments, the optionally substituted C5-C15 aryl is naphthyl.
  • R 6 , R 7 , N 2 , and C 2 together form a 5- or 6-membered ring including C4-C5 heterocycloalkyl including an N heteroatom and additional 0 or 1 heteroatom independently selected from N, O, and S; and wherein R' 6 , R' 7 , N' 2 , and C' 2 together form a 5- or 6-membered ring including C4-C5 heterocycloalkyl including an N heteroatom and additional 0 or 1 heteroatom independently selected from N, O, and S.
  • each of R 2 and R' 2 is C1 - C4 heteroalkylene.
  • each of R 2 and R' 2 is -(CH2)2NH- or -CH2NH-.
  • the compound is described by formula (XV):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl, or a
  • L', L, L 1 , or L' 1 includes one or more optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C1 5 arylene, optionally substituted C2-C15 heteroarylene, O, S, NR', P, carbonyl,
  • the backbone of L', L, L 1 , or L' 1 consists of one or more optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20
  • heteroalkenylene optionally substituted C2-C20 alkynylene, optionally substituted C2-C20
  • heteroalkynylene optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20
  • heterocycloalkylene optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15
  • R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyi , optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl , optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl , optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl , optionally substituted C8-C20 heterocycloalkynyl , optionally substituted C5-C15
  • L', L, L 1 , or L' 1 is oxo substituted.
  • the backbone of L', L, L 1 , or L' 1 includes no more than 250 atoms.
  • L', L, L 1 , or L' 1 is capable of forming an amide, a carbamate, a sulfonyl, or a urea linkage.
  • L, L 1 , or L' 1 is a bond.
  • L is described by formula (L-l) :
  • L A is described by formula G A1 -(Z A1 ) g i-(Y A1 )hi-(Z A2 )ii-(Y A2 )ji -(Z A3 )ki -(Y A3 )ii -(Z A4 ) m i -(Y A4 )ni-(Z A5 )oi - G A2 ;
  • L B is described by formula G B1 -(Z B1 )g 2 -(Y B1
  • L c is described by G A1 is a bond attached to Q in formula (L-l) ; G A2 is a bond attached to A1 or M1 if A1 is absent; G B1 is a bond attached to Q in formula (L-l) ; G B2 is a bond attached to A2 or M2 if A2 is absent; G C1 is a bond attached to Q in formula (L-l) ; G C2 is a bond attached to E; each of Z M , Z A2 , Z A3 , Z M , Z A5 , Z B1 , Z B2 , Z B3 , Z B4 , Z B5 , Z C1 , Z C2 , Z C3 , Z C4 , and Z C5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 - C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20
  • heteroalkenylene optionally substituted C2-C20 alkynylene, optionally substituted C2-C20
  • heteroalkynylene optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20
  • heterocycloalkylene optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of Y A1 , Y A2 , Y A3 , Y A4 , Y B1 , Y B2 , Y B3 , Y B4 , Y C1 , Y C2 , Y C3 , and Y C4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl , phosphate, phosphoryl, or imino; R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalky
  • L is
  • R * is a bond or includes one or more of optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2- C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15 heteroarylene, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl,
  • L is described by formula (L-lla) or formula (L-llb):
  • L A is described by formula G A1 -(Z A1 ) g i-(Y A1 )hi-(Z A2 )ii-(Y A2 )ji -(Z A3 )ki -(Y A3 )ii -(Z A4 ) m i -
  • L B is described by formula G B1 -(Z B1 ) g 2-(Y B1 )h2-(Z B2 )i2-(Y B2 )j2-(Z B3 ) k 2-(Y B3 )i2-(Z B4 )m2-
  • L c is described by formula G c1 -(Z c1 ) g 3-(Y c1 )h3-(Z C2 )i 3 -(Y C2 )j3-(Z C3 )k3-(Y C3 )i3-(Z C4 )m3- (Y C4 )n3-(Z C5 )o3-G C2 ;
  • L D is described by formula G D1 -(Z D1 ) g 4-(Y D1 )h4-(Z D2 )i 4 -(Y D2 )j4-(Z D3 )k4-(Y D3 )i4-(Z D4 )m4-
  • G A1 is a bond attached to N A in formula (L-lla) or N A in formula (L-llb);
  • G A2 is a bond attached to A1 or M1 if A1 is absent;
  • G B1 is a bond attached to N A in formula (L-lla) or N A in formula (L- llb);
  • G B2 is a bond attached to A2 or M2 if A2 is absent;
  • G C1 is a bond attached to N B in formula (L-lla) or C in formula (L-llb);
  • G C2 is a bond attached to a first monosaccharide or oligosaccharide moiety, E 1 ;
  • G D1 is a bond attached to N B in formula (L-lla) or C in formula (L-llb);
  • G D2 is a bond attached to a second monosaccharide or oligosaccharide moiety, E' 1 ;
  • L is
  • L is described by formula (L-lll):
  • H A1 is a bond attached to A2; H A2 is a bond attached to A1 ; each of W A1 , W A2 , W A3 , W M , and W A5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5
  • L 1 is described by formula (L-IV):
  • H B1 is a bond attached to A1 ;
  • H B2 is a bond attached to a first monosaccharide or oligosaccharide moiety, E 1 ;
  • each of W B1 , W B2 , W B3 , W B4 , and W B5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4- C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8
  • cycloalkynyl optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; and each of g2, h2, i2, j2, k2, I2, m2, n2, and o2 is,
  • L 1 is
  • L' 1 is described by formula (L-V):
  • H C1 is a bond attached to A2; H C2 is a bond attached to a second monosaccharide or oligosaccharide moiety, E' 1 ; each of W C1 , W C2 , W C3 , W C4 , and W C5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 al
  • E, E 1 , or E' 1 is
  • E, E 1 , or E' 1 is
  • E, E 1 , or E' 1 is any one of the moieties in Tables 2A and 2B.
  • E, E 1 , or E' 1 directly or indirectly activates an immune cell.
  • E, E 1 , or E' 1 is a ligand to an innate immune receptor.
  • the innate immune receptor is AICL, BDCA2, CLEC2, Complement receptor 3,
  • Complement receptor 4 DCIR, dectin-1 , dectin-2, DC-SIGN, a C-Type lectin receptor, MMR, langerin, TLR2, Mincle, MBL, or KCR.
  • E, E 1 , or E' 1 binds to an antibody.
  • the antibody is a natural antibody.
  • the natural antibody is an antibody of the immunoglobulin M (IgM) isotype.
  • the antibody binds to a moiety in Tables 2A and 2B.
  • the antibody is anti-aGal antibody or anti-aRha antibody.
  • the monosaccharide moiety has one optionally substituted C6-C9 monosaccharide residue.
  • the oligosaccharide moiety has 2-18 optionally substituted C6-C9 monosaccharide residues. In some embodiments, the oligosaccharide moiety has 2-12 optionally substituted C6-C9 monosaccharide residues.
  • each of the optionally substituted C6-C9 monosaccharide residues is, independently, glucose (Glc), galactose (Gal), mannose (Man), allose (All), altrose (alt), gulose (Gul), idose (ido), talose (Tal), fucose (Fuc), rhamnose (Rha or L-Rha), thia-rhamnose (thia-Rha or thia-L-Rha), quinovose (Qui), 2-deoxyglucose (2-dGlc), glucosamine (GlcN), galactosamine (GaIN), mannosamine (ManN), fucosamine (FucN), quinovosamine (QuiN), N-Acetyl- glucosamine (GlcNAc), N-Acetyl-galactosamine (GalNAc), N-Acetyl-mannosamine (ManNAc), N-acetyl
  • each of the optionally substituted C6-C9 monosaccharide residues is, independently, an optionally substituted C6 monosaccharide residue.
  • the optionally substituted C6 monosaccharide residue is Glc, Gal, Man, All, Alt, Gul, Ido, or Tal. In some embodiments, the optionally substituted C6 monosaccharide residue is Fuc, Rha or L-Rha, thia-Rha or thia-L-Rha, Qui, or 2-dGlc. In some embodiments, the optionally substituted C6 monosaccharide residue is GlcN, GaIN, ManN, FucN, or QuiN.
  • the optionally substituted C6 monosaccharide residue is N- GlcNAc, GalNAc, ManNAc, FucNAc, QuiNAc, GIcA, GalA, ManA, or IdoA. In some embodiments, the optionally substituted C6 monosaccharide residue is Glc-ol, Gal-ol, or Man-ol. In some embodiments, the optionally substituted C6 monosaccharide residue is Fru, Sor, Tag. In some embodiments, the optionally substituted C6 monosaccharide residue is The, Aco, Dig, Cym, Abe, Col, Tyv, Asc, Par, or MurNAc.
  • the optionally substituted C6 monosaccharide residue is Rha, Gal, Glc, GlcA, GlcNAc, GalNAc, GlcN(Gc) (N-Glycolyl-Glucosamine), Neu5Ac, Neu5Gc, Fuc, Man, -hkPCbMan (mannose phosphate), 6-H2PO3GIC (glucose phosphate), Mur (muramoyl), Mur-L-Ala-D-i-Gln-Lys, (Mur)-3-0-GlcNAc, sulfate-galactose (Su-Gal), disulfate-galactose (Su 2 -Gal), sulfate-glucose (Su-Glc), sulfate-GlcNAc (Su-GlcNAc), or sulfate-GalNAc (Su-GalNAc).
  • the optionally substituted C6 monosaccharide residue is optionally substituted Rha. In some embodiments, the optionally substituted C6 monosaccharide residue is Rha.
  • the optionally substituted C6 monosaccharide residue is L-Rha.
  • the optionally substituted C6 monosaccharide residue is optionally substituted Gal or optionally substituted Glc.
  • the optionally substituted Gal is optionally substituted a1 -3Gal.
  • the optionally substituted Glc is an optionally substituted ⁇ -glucan having 1 -6 Glc moieties.
  • the optionally substituted ⁇ -glucan is
  • the optionally substituted ⁇ -glucan is laminarin.
  • each of the optionally substituted C6-C9 monosaccharide residues is, independently, an optionally substituted C9 monosaccharide residue, wherein the optionally substituted C9 monosaccharide residue is Sia, Neu, Neu5Ac, or Neu5Gc.
  • At least one optionally substituted C6-C9 monosaccharide residue is substituted with 1 -3 substituents independently selected from sulfate, phosphate, methyl, acetyl, natural amino acids, and non-natural amino acids. In some embodiments, at least one optionally substituted C6- C9 monosaccharide residue is substituted with 1 -3 substituents independently selected from sulfate, phosphate, methyl, and acetyl. In some embodiments, at least one optionally substituted C6-C9 monosaccharide moiety is substituted with 1 -3 substituents independently selected from natural and non- natural amino acids. In some embodiments, the natural amino acid is alanine, lysine, serine, glutamine or asparagine.
  • the disclosure features a compound of formula (XVI):
  • each A is an independently selected amino acid
  • L is a linker that, when m is 2, 3, 4, or 5, is bound to any of A
  • each E is independently selected from a monosaccharide or an oligosaccharide
  • m is 0, 1 , 2, 3, 4, or 5
  • n is 1 , 2, 3, 4, or 5
  • Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof.
  • Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof.
  • At least one of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
  • at least two of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
  • at least three of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
  • At least four of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least five of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
  • each Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the disclosure features a compound of formula (XVII):
  • each A 1 and A 2 is an independently selected amino acid
  • L is a linker that, when m is 1 , 2, 3, 4, or 5, is bound to a nitrogen atom in any A 1 and a nitrogen atom in any A 2
  • each E is independently selected from a monosaccharide or an oligosaccharide
  • each m is independently 0, 1 , 2, 3, 4, or 5
  • n is 1 , 2, 3, 4, or 5
  • Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof.
  • Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each
  • At least one (e.g., at least two, three, four, or five) of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
  • each Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyl, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-
  • each m is independently 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
  • n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-
  • the disclosure features a compound of formula (XVIII):
  • each A 1 and A 2 is an independently selected amino acid
  • X is absent or is -CH2CH2C(0)NH-
  • each E is independently selected from a monosaccharide or an oligosaccharide
  • each m is independently 0, 1 , 2, 3, 4, or 5
  • n is 1 , 2, 3, 4, or 5
  • Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid
  • d is an integer from 0 to 10
  • e is an integer from 0 to 10; or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalan
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2 or 3; d is an integer from 1 to 10; e is an integer from 1 to 10; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
  • m is 2. In some embodiments, m is 3.
  • m is 2; d is 1 ; e is 1 ; and X is - CH2CH2C(0)NH-; or a pharmaceutically acceptable salt thereof.
  • E is , or a
  • the disclosure features a compound of formula (XIX):
  • each A 1 and A 2 is an independently selected amino acid
  • each E is independently selected from a monosaccharide or an oligosaccharide
  • each m is independently 0, 1 , 2, 3, 4, or 5
  • n is 1 , 2, 3, 4, or 5
  • Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid
  • d is an integer from 0 to 10
  • e is an integer from 0 to 10
  • f is an integer from 0 to 10
  • g is an integer from 0 to 10; or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • Q 6 is selected from one of
  • a compound of formula (XIX) the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2; d is 1 ; e is 1 ; f is 3; g is 1 ; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-a
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2; d is 1 ; e is 1 ; f is 1 ; g is 1 ; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
  • E is
  • E is or a
  • the disclosure features a compound of formula
  • each A 1 and A 2 is an independently selected amino acid
  • each E is independently selected from a monosaccharide or an oligosaccharide
  • each m is independently 0, 1 , 2, 3, 4, or 5
  • each n is independently 1 , 2, 3, 4, or 5
  • Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid
  • d is an integer from 0 to 1 0
  • e is an integer from 0 to 10
  • f is an integer from 0 to 10
  • g is an integer from 0 to 25; or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine,
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2; d is 1 ; e is 1 ; f is 1 ; g is 8 to 25; E is a
  • n 1 ; or a pharmaceutically acceptable salt thereof.
  • E is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • each A 1 and A 2 is an independently selected amino acid
  • each E is independently selected from a monosaccharide or an oligosaccharide
  • each m is independently 0, 1 , 2, 3, 4, or 5
  • each n is independently 1 , 2, 3, 4, or 5
  • Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid
  • d is an integer from 0 to 15; or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine,
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid and piperazine-2-carboxylic acid; m is 2; d is 10; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
  • E is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • the disclosure features a compound of formula (XXIII):
  • Y is , -C(0)CH2CH2-, -CH2-, or is absent;
  • X is -C(0)CH2CH2- or is absent;
  • each m is independently 0, 1 , 2, 3, 4, or 5;
  • each n is independently 1 , 2, 3, 4, or 5;
  • Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid;
  • d is an integer from 0 to 15;
  • e is an integer from 1 to 10;
  • f is an integer from 1 to 5; and
  • g is an integer from 1 to 5; or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalan
  • neopentylglycine m is 2, 3 or 4; d is 1 ; when Y is , e is 4; E is a monosaccharide; f is 1 or 2; g is 1 or 2; and n is 1 ; or a pharmaceutically acceptable salt thereof.
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 3-aminoalanine, 2-piperazinecarboxylic acid, 2-aminohexanoic
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalan
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid and threonine; and m is 3; or a pharmaceutically acceptable salt thereof.
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalan
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 2,4- diaminobutyric acid, 3-aminoalanine, 2-aminohexanoic acid, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine, or a pharmaceutically acceptable salt thereof.
  • m is 4, d is 1 , f is 1 , and g is 1 , or a pharmaceutically acceptable salt thereof. In some embodiments, m is 3, f is 2, and g is 1 , or a pharmaceutically acceptable salt thereof.
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2- aminooctanoic acid and threonine; f is 1 ; and g is 1 ; or a pharmaceutically acceptable salt thereof.
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2- aminohexanoic acid, 2-aminooctanoic acid and threonine, or a pharmaceutically acceptable salt thereof; m is 4; f is 1 ; and g is 1 ; or a pharmaceutically acceptable salt thereof.
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2-aminohexanoic acid, 2-aminooctanoic acid and threonine, or a pharmaceutically acceptable salt thereof; m is 3; f is 1 ; and g is 1 ; or a pharmaceutically acceptable salt thereof.
  • E is ; or a
  • E is , or a
  • each A 1 and A 2 is an independently selected amino acid
  • X is -C(0)CH2CH2CH2-Y- or - C(0)CH2CH2C(0)NH-;
  • Y is heteroaryl;
  • each E is independently selected from a monosaccharide or an oligosaccharide;
  • each m is independently 0, 1 , 2, 3, 4, or 5;
  • each n is independently 1 , 2, 3, 4, or 5;
  • Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; and
  • d is an integer from 0 to 15; or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalan
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 3-(2-naphthyl)alanine, and threonine, or a pharmaceutically acceptable salt thereof.
  • X is -C(0)CH2CH2CH2-Y-; m is 3; d is 3; and Y is 1 ,4-triazololyl; or a pharmaceutically acceptable salt thereof.
  • X is -C(0)CH2CH2C(0)-; m is 2; and d is 1 ; or a pharmaceutically acceptable salt thereof.
  • E is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • the disclosure features a compound of formula (XXV):
  • each A 1 and A 2 is an independently selected amino acid
  • each E is independently selected from a monosaccharide or an oligosaccharide
  • each m is independently 0, 1 , 2, 3, 4, or 5
  • each n is independently 1 , 2, 3, 4, or 5
  • Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid
  • d is an integer from 0 to 1 5
  • e is an integer from 0 to 1 5; or a
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-
  • n 1 ; or a pharmaceutically acceptable salt thereof.
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2 or 3; d is an integer from 1 to 3; e is an integer from 1 to 3; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
  • E is or a
  • the disclosure features a compound of formula (XXVI):
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalan
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2; d is 1 ; e is 1 ; f is 1 ; E is a monosaccharide; R is C1 -C1 0 alkyl ; and n is 1 ; or a pharmaceutically acceptable salt thereof.
  • E is or a
  • E is or a
  • the disclosure features a compound of formula (XXVII)
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroa
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; m is 2, 3, or 4; d is 1 ; e is 1 ; f is 1 ; E is a monosaccharide; R is C1 -C10 alkyl; and n is 1 ; or a pharmaceutically acceptable salt thereof.
  • m is 2. In some embodiments, m is 3. In some embodiments, m is 4.
  • E is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • the disclosure features a compound of formula (XXVIII):
  • Y is -C(0)CH2CH2-, -CH2-, or is absent; e is an integer from 1 to 10; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; and d is an integer from 0 to 15; or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • Y is -C(0)CH2CH2-, or a
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroa
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and d is 1 ; or a pharmaceutically acceptable salt thereof.
  • m is 2. In some embodiments of a compound of formula (XXVIII), m is 3. In some embodiments of a compound of formula (XXVIII), m is 4. In some embodiments of a compound of formula (XXVIII), E is
  • E is or a
  • the disclosure features a compound of formula (XXIX):
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroa
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and d is 1 ; or a pharmaceutically acceptable salt thereof.
  • m is 2. In some embodiments of a compound of formula (XXIX), m is 3. In some embodiments of a compound of formula (XXIX), m is 4.
  • E is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • the disclosure features a compound of formula (XXX):
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • Y is -C(0)CH2CH2-, or a
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and d is 1 ; and e is 1 or 2; or a pharmaceutically acceptable salt thereof.
  • m is 2. In some embodiments of a compound of formula (XXX), m is 3. In some embodiments of a compound of formula (XXX), m is 4. pharmaceutically acceptable salt thereof. In some embodiments, E is , or a pharmaceutically acceptable salt thereof.
  • the disclosure features a compound of formula (XXXI) :
  • each Y is independently selected from
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl , 2-butyl , methyl , and 2-aminoethyl , or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • Y is -C(0)CH2CH2-, or a
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroa
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and d is 4 or 5; e is 4 or 5; and f is 1 ; or a pharmaceutically acceptable salt thereof.
  • m is 2. In some embodiments of a compound of formula (XXXI), m is 3. In some embodime m is 4.
  • E is , or a
  • the disclosure features a compound of formula (XXXII):
  • each Y is independently selected from
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • Y is -CH2CH2NHC(0)CH2CH2-, or a pharmaceutically acceptable salt thereof.
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoro
  • n 1 ; or a pharmaceutically acceptable salt thereof.
  • each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and d is an integer from 1 to 4; and e is 1 ; or a pharmaceutically acceptable salt thereof.
  • d is 2. In some embodiments of a compound of formula (XXXII), m is 2. In some embodiments, m is 3. In some embodiments, m is 4.
  • E is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • the disclosure features a compound of formula (XXXIII):
  • d is 2.
  • the compound is of the formula (XXXIII-1 ):
  • the compound is of the formula (XXXI 11-2):
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
  • each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2- methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl. hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
  • a compound of formula (XXXIII) e.g., a compound of formula (XXXIII-1 ) or a compound of formula (XXXIII-2)
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • a compound of formula (XXXIII) e.g., a compound of formula (XXXII 1- ) or a compound of formula (XXXIII-2)
  • the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of
  • each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2- piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-e
  • m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments of a compo (e.g., a compound of formula (XXXII 1-1 ) or a
  • E ically acceptable salt thereof In some embodiments, E ically acceptable salt thereof.
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl; each of R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R' 2 , R' 3 , R' 4 , R' 5 , R' 6 , R' 7 , R' 8 , R' 9 , and R' 10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20
  • each of a', b', c', a, b, and c is, independently, 0 or 1 ;
  • each of N 1 , N 2 , N 3 , N 4 , N' 1 , N' 2 , N' 3 , and N' 4 is a nitrogen atom
  • each of C 1 , C 2 , C 3 , C' 1 , C' 2 , and C' 3 is a carbon atom ;
  • L is a linker comprising at least one optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C3-C15 heteroarylene;
  • each E is, independently, a monosaccharide or oligosaccharide moiety
  • n 1 , 2, 3, or 4,
  • the compound comprises at least one optionally substituted 5-8 membered ring formed by joining (i) R 2 , R 3 , and C 1 ; (ii) R 3 , R 4 , N 1 , and C 1 ; (iii) R 5 , R 6 , and C 2 ; (iv) R 6 , R 7 , N 2 , and C 2 ; (v) R 8 , R 9 , and C 3 ; (vi) R 9 , R 10 , N 3 , and C 3 ; (vii) R' 2 , R' 3 , and C' 1 ; (viii) R' 3 , R' 4 , N' 1 , and C' 1 ; (ix) R' 5 , R' 6 , and C' 2 ; (x) R' 6 , R' 7 , N' 2 , and C' 2 ; (xi) R' 8 , R' 9 , and C' 3 ; or (xii) R' 9 ,
  • L is described by formula (L-VI) :
  • L A is described by formula G A1 -(Z A1 ) g i-(Y A1 )hi-(Z A2 )ii-(Y A2 )ji-(Z A3 )ki-(Y A3 )ii-(Z A4 ) m i-
  • L B is described by formula G B1 -(Z B1 ) g 2-(Y B1 )h2-(Z B2 )i2-(Y B2 )j2-(Z B3 ) k 2-(Y B3 )i2-(Z B4 )m2-(Y B4 )n2-(Z B5 )02-
  • L c is described by formula G c1 -(Z c 1 ) g 3-(Y c1 )h3-(Z C2 )i 3 -(Y C2 )j3-(Z C3 )k3-(Y C3 )i3-(Z C4 )m3-(Y C4 )n3-(Z C5 )03-
  • G A1 is a bond attached to Q in formula (L-VI);
  • G A2 is a bond attached to A1 or M1 if A1 is absent;
  • G B1 is a bond attached to Q in formula (L-VI);
  • G B2 is a bond attached to A2 or M2 if A2 is absent;
  • G C1 is a bond attached to Q in formula (L-VI);
  • G C2 is a bond attached to E; each of Z A1 , Z A2 , Z A3 , Z A4 , Z A5 , Z B1 , Z B2 , Z B3 , Z B4 , Z B5 , Z C1 , Z C2 , Z C3 , Z C4 and Z C5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalky
  • each of Y A1 , Y A2 , Y A3 , Y A4 , Y B1 , Y B2 , Y B3 , Y B4 , Y C1 , Y C2 , Y C3 , and Y C4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;
  • R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; each of g1 , hi , i1 , j1 , k1 , 11 , ml , n
  • Q comprises at least one optionally substituted C3-C20 cycloalkylene, optionally substituted C3- C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C3-C15 heteroarylene.
  • L is described b formula (L-VII):
  • L A is described by formula G A1 -(Z A1 ) g i-(Y A1 )hi-(Z A2 )ii-(Y A2 )ji -(Z A3 )ki -(Y A3 )ii -(Z A4 ) m i -
  • L B is described by formula G B1 -(Z B1 ) g 2-(Y B1 )h2-(Z B2 )i2-(Y B2 )j2-(Z B3 ) k 2-(Y B3 )i2-(Z B4 )m2-(Y B4 )n2-(Z B5 )02-
  • L c is described by formula G c1 -(Z c1 ) g 3-(Y c1 )h3-(Z C2 )i 3 -(Y C2 )j3-(Z C3 )k3-(Y C3 )i3-(Z C4 )m3-(Y C4 )n3-(Z C5 )03-
  • L D is described by formula G D1 -(Z D1 ) g 4-(Y D1 )h4-(Z D2 )i 4 -(Y D2 )j4-(Z D3 )k4-(Y D3 )i4-(Z D4 )m4-(Y D4 )n4-(Z D5 )o4-
  • G A1 is a bond attached to Q in formula (L-VII);
  • G A2 is a bond attached to A1 or M1 if A1 is absent;
  • G B1 is a bond attached to Q in formula (L-VII);
  • G B2 is a bond attached to A2 or M2 if A2 is absent;
  • G C1 is a bond attached to Q in formula (L-VII);
  • G C2 is a bond attached to a first monosaccharide or oligosaccharide moiety, E 1 ;
  • G D1 is a bond attached to N in formula (L-VII);
  • G D2 is a bond attached to a second monosaccharide or oligosaccharide moiety, E 2 ;
  • each of Z A1 Z A2 Z A3 Z A4 Z A5 Z B1 Z B2 Z B3 Z B4 Z B5 Z C1 Z C2 Z C3 Z C4 Z C5 Z D1 Z D2 Z D3 Z D4 and Z D5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20
  • heteroalkylene optionally substituted C2-C20 alkenylene, optionally substituted C2-C20
  • heteroalkenylene optionally substituted C2-C20 alkynylene, optionally substituted C2-C20
  • heteroalkynylene optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene;
  • each of Y A1 Y A2 Y A3 Y A4 Y B1 Y B2 Y B3 Y B4 Y C1 Y C2 Y C3 Y C4 Y D1 Y D2 Y D3 and Y D4 is independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;
  • R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; each of g1 , hi , i1 , j1 , k1 , 11 , ml , n
  • Q comprises at least one optionally substituted C3-C20 cycloalkylene, optionally substituted C3- C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C3-C15 heteroarylene.
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl;
  • each of R 2 , R 6 , R 8 , R' 2 , R' 6 , and R' 8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
  • each E is, independently, a monosaccharide or oligosaccharide moiety
  • n 1 , 2, 3, or 4,
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl;
  • each of R 2 , R 6 , R 8 , R' 2 , and R' 6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
  • each E is, independently, a monosaccharide or oligosaccharide moiety
  • n 1 , 2, 3, or 4,
  • the compound is described by formula (XXXVI):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl;
  • each of R 2 , R 6 , R 8 , and R' 2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
  • each E is, independently, a monosaccharide or oligosaccharide moiety
  • n 1 , 2, 3, or 4,
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl;
  • each of R 2 , R 6 , and R 8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each E is, independently, a monosaccharide or oligosaccharide moiety
  • n 1 , 2, 3, or 4,
  • the compound is described by formula (XXXVIII):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl;
  • each of R 2 , R 6 , R' 2 , and R' 6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
  • each E is, independently, a monosaccharide or oligosaccharide moiety
  • n 1 , 2, 3, or 4,
  • the compound is described by formula (XXXIX):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl;
  • each of R 2 , R 6 , and R' 2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
  • each E is, independently, a monosaccharide or oligosaccharide moiety
  • n 1 , 2, 3, or 4,
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl;
  • each of R 2 and R 6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
  • each E is, independently, a monosaccharide or oligosaccharide moiety
  • n 1 , 2, 3, or 4,
  • the compound is described by formula (XXXXI):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl;
  • each of R 2 and R' 2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
  • each E is, independently, a monosaccharide or oligosaccharide moiety
  • n 1 , 2, 3, or 4,
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl;
  • R 2 is a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
  • each E is, independently, a monosaccharide or oligosaccharide moiety
  • n 1 , 2, 3, or 4,
  • the compound is described by formula (XXXXIII):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl;
  • each E is, independently, a monosaccharide or oligosaccharide moiety
  • n 1 , 2, 3, or 4,
  • L comprises at least one an optionally substituted C3 cycloalkylene or an optionally substituted C3 heterocycloalkylene.
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl; and
  • L comprises an optionally substituted C3 cycloalkylene or an optionally substituted C3 heterocycloalkylene
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl; and
  • L comprises an optionally substituted C3 cycloalkylene or an optionally substituted C3 heterocycloalkylene
  • L is N
  • each q is, independently, an integer from 1 to 1 1 , inclusive.
  • L comprises at least one an optionally substituted C5 cycloalkyl optionally substituted C5 heterocycloalkylene.
  • the compound is described by formula (XXXXVI):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl; and
  • L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene, or a pharmaceutically acceptable salt thereof.
  • the compound is described by formula (XXXXVII):
  • R' 1 and R 1 are, independently, optionally substituted benzyl, optionally substituted
  • C2-C15 heteroaryl optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl; and
  • L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene
  • the compound is described by formula (XXXXVIII):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl; and L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene,
  • the compound is described by formula (XXXXIX):
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl; and
  • L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene
  • each q is, independently, an integer from 1 to 1 1 , inclusive.
  • L comprises at least one an optionally substituted C6 cycloalkylene, at least one an optionally substituted C6 heterocycloalkylene, or at least one an optionally substituted C6 arylene.
  • each of R' 1 and R 1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH 2 CH(CH 3 )2), or cyclohexylmethyl; and L comprises an optionally substituted C6 arylene,
  • L is N
  • each q is, independently, an integer from 1 to 1 1 , inclusive.
  • non-natural amino acids that may be included in a compound described herein (e.g., a compound of any one of formulas (l)-(XXXX)), for example, in the polymyxin core portions (e.g., in the first cyclic heptapeptide and/or the second cyclic heptapeptide) and/or the linker portion of the compound, include, but are not limited to, D-Ser, D-Pro, D-Leu, D-Nle (D-norleucine), D-Thr, D-Val, L-Abu (L-2-aminobutyric acid), 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2- piperazinecarboxylic acid,
  • polymyxin core portions e.g., in the first cyclic heptapeptide and/or the second cyclic heptapeptide
  • polymyxin core portions include D-Ser, D- Pro, D-Leu, D-Nle, D-Thr, D-Val, and/or L-Abu.
  • a concentration of the compound, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to 10,000 nM. In some embodiments, the concentration of the compound, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to equal to 1 ,000 nM. In some embodiments, the concentration of the compound, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to equal to 100 nM.
  • the disclosure features a pharmaceutical composition including a compound described herein (e.g., a compound of any one of formulas (l)-(XXXX)), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition further includes an antibacterial agent.
  • the antibacterial agent is selected from the group consisting of linezolid, tedizolid, posizolid, radezolid, rumblemulin, valnemulin, tiamulin, azamulin, lefamulin, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem, doripenem, imipenem/cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefo
  • a prodrug of tedizolid is tedizolid phosphate.
  • the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, levofloxacin, rifampicin, linezolid, erythromycin, or solithromycin.
  • the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, or levofloxacin.
  • the disclosure features a method of protecting against or treating a bacterial infection in a subject by administering to the subject a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)).
  • a compound described herein e.g., a compound of any one of formulas (l)-(XXXXX)
  • the method further includes administering to the subject an antibacterial agent.
  • the disclosure features a method of protecting against or treating a bacterial infection in a subject by administering to the subject (1 ) a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) and (2) an antibacterial agent.
  • a compound described herein e.g., a compound of any one of formulas (l)-(XXXXX)
  • an antibacterial agent e.g., a compound of any one of formulas (l)-(XXXXX)
  • the bacterial infection is caused by Gram- negative bacteria. In some embodiments, the bacterial infection is caused by a resistant strain of bacteria. In some embodiments, the resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene and/or a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, or the mcr-3 gene. In some embodiments, the resistant strain of bacteria possesses a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of bacteria is a resistant strain of E. coli.
  • the disclosure features a method of protecting against or treating sepsis in a subject by administering to the subject a compound described herein (e.g., a compound of any one of formulas (I)- (XXXXX)).
  • a compound described herein e.g., a compound of any one of formulas (I)- (XXXXX)
  • the disclosure features a method of preventing lipopolysaccharides (LPS) in Gram- negative bacteria from activating an immune system in a subject by administering to the subject a compound described herein (e.g., a compound of any one of formulas (l)-(XXXX)).
  • a compound described herein e.g., a compound of any one of formulas (l)-(XXXX)
  • the method prevents LPS from activating a macrophage.
  • the method prevents LPS-induced nitric oxide (NO) production from a macrophage.
  • the Gram-negative bacteria is a resistant strain of Gram-negative bacteria.
  • the resistant strain of Gram-negative bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene and/or a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of Gram-negative bacteria possesses the mcr- 1 gene, the mcr-2 gene, or the mcr-3 gene. In some embodiments, the resistant strain of Gram- negative bacteria possesses a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of Gram-negative bacteria is a resistant strain of E. coli.
  • the method further includes administering to the subject an antibacterial agent.
  • the compound and the antibacterial agent are administered substantially simultaneously.
  • the compound and the antibacterial agent are administered separately. In some embodiments, the compound is administered first, followed by administering of the antibacterial agent alone. In some embodiments, the antibacterial agent is administered first, followed by administering of the compound alone.
  • the compound and the antibacterial agent are administered substantially simultaneously, followed by administering of the compound or the antibacterial agent alone. In some embodiments, the compound or the antibacterial agent is administered first, followed by administering of the compound and the antibacterial agent substantially simultaneously.
  • administering the compound and the antibacterial agent together lowers the MIC of each of the compound and the antibacterial agent relative to the MIC of each of the compound and the antibacterial agent when each is used alone.
  • the compound and/or the antibacterial agent is administered intramuscularly, intravenously, intradermal ⁇ , intraarterially, intraperitoneally, intralesionally, intracranial ⁇ , intraarticularly, intraprostatically, intrapleural ⁇ , intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctival, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, orally, locally, by inhalation, by injection, or by infusion.
  • the disclosure features a method of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria, by contacting the bacteria or a site susceptible to bacterial growth with a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)).
  • a compound described herein e.g., a compound of any one of formulas (l)-(XXXXX)
  • the method further includes contacting the bacteria or the site susceptible to bacterial growth with an antibacterial agent.
  • the disclosure features a method of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria, including contacting the bacteria or a site susceptible to bacterial growth with (1 ) a compound described herein (e.g., a compound of any one of formulas (l)-(XXXX)) and (2) an antibacterial agent.
  • a compound described herein e.g., a compound of any one of formulas (l)-(XXXXX)
  • an antibacterial agent e.g., a compound of any one of formulas (l)-(XXXXX)
  • the antibacterial agent is selected from the group consisting of linezolid, tedizolid, posizolid, radezolid, rumblemulin, valnemulin, tiamulin, azamulin, lefamulin, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem, doripenem, imipenem/cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cef
  • trimethoprim trimethoprim, prodrugs thereof, and pharmaceutically acceptable salts thereof.
  • a prodrug of tedizolid is tedizolid phosphate.
  • the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, levofloxacin, rifampicin, linezolid, erythromycin, or solithromycin.
  • the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, or levofloxacin.
  • the bacteria are Gram-negative bacteria. In some embodiments, the bacteria are a resistant strain of bacteria. In some embodiments, the resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene and/or a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene. In some embodiments, the resistant strain of bacteria possesses a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of bacteria is a resistant strain of E. coli.
  • the disclosure features any compound described herein (e.g., a compound of any one of formulas (l)-(XXXX) and/or any one of compounds 1 -123, or a pharmaceutically
  • the disclosure provides a compound described by any one Formulas (IV)- (XIII), (XV), or (XXXIV), wherein each of R' 1 and R 1 is, independently, benzyl or CH 2 CH(CH 3 )2.
  • cyclic heptapeptide or “cycloheptapeptide,” as used herein, refers to certain compounds that bind to lipopolysaccharides (LPS) in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics.
  • Cyclic heptapeptides or cycloheptapeptides comprise seven natural or non-natural a-amino acid residues, such as D- or L-amino acid residues, in a closed ring.
  • cyclic heptapeptides are formed by linking the a-carboxyl group of one amino acid to the a-amino group or the ⁇ -amino group of another amino acid and cyclizing.
  • the cyclic heptapeptide comprises a heterocycle comprising carbon and nitrogen ring members, which may be substituted, for example, with amino acid side chains.
  • One nitrogen from an a-amino group in the cyclic heptapeptide is not a ring member and is branched from a ring member of the heterocycle. Thus, this nitrogen is directly attached to a ring member, such as a carbon atom (e.g., an a-carbon atom).
  • This nitrogen atom serves as an attachment point for the cyclic heptapeptide to a linker and/or to a peptide (e.g., a peptide including 1 -5 amino acid residue(s)), and thus is referred to herein as a "linking nitrogen.”
  • the linking nitrogen is directly attached to the ring of the cyclic heptapeptide and is not derived from a side chain, such as an ethylamine side chain.
  • the linking nitrogens in a compound of, e.g., formula (II) or (III), are N 4 and N' 4 .
  • a peptide including one or more (e.g., 1 -5; 1 , 2, 3, 4, or 5) amino acid residues may be covalently attached to a linking nitrogen (e.g., N 4 and/or N' 4 , the nitrogen from an a-amino group) in the cyclic heptapeptide ring.
  • Cyclic heptapeptides may be derived from polymyxins (e.g., naturally existing polymyxins and non-natural polymyxins) and/or octapeptins (e.g., naturally existing octapeptins and non-natural octapeptins).
  • polymyxins examples include, but are not limited to, polymyxin Bi , polymyxin B2, polymyxin B3, polymyxin B4, polymyxin B5, polymyxin ⁇ , polymyxin BHIe, polymyxin B2-lle, polymyxin Ci , polymyxin C2, polymyxin Si , polymyxin Ti , polymyxin T2, polymyxin Ai , polymyxin A2, polymyxin Di , polymyxin D2, polymyxin Ei (colistin A), polymyxin E2 (colistin B), polymyxin E3, polymyxin E 4 , polymyxin E7, polymyxin EHIe, polymyxin Ei-Val, polymyxin Ei-Nva, polymyxin E2-lle, polymyxin E2-Val, polymyxin E2-Nva, polymyxin Es-lle, polymyxin Mi , and polymyxin M2.
  • polymyxin Bi
  • covalently attached refers to two parts of a compound that are linked to each other by a covalent bond formed between two atoms in the two parts of the compound.
  • L is covalently attached to N' 4 , which means that when a' is 0, an atom in L forms a covalent bond with N' 4 in the compound.
  • linker refers to a covalent linkage or connection between two or more components in a compound (e.g., two cyclic heptapeptides and one or more monosaccharide or oligosaccharide moieties in a compound described herein).
  • a compound described herein may contain a linker that has a trivalent structure (e.g., a trivalent linker).
  • a trivalent linker has three arms, in which each arm is conjugated to a component of the compound (e.g., a first arm conjugated to a first cyclic heptapeptide, a second arm conjugated to a second heptapeptide, and a third arm conjugated to one or more monosaccharide or oligosaccharide moieties).
  • a compound described herein may contain two or three linkers (e.g., a compound of formula (XIII) or (XIV)), in which each linker has a divalent structure (e.g., a divalent linker).
  • the first linker may connect the first and second cyclic heptapeptides (e.g., L in the compound of formula (XIII)), the second linker may connect a first monosaccharide or oligosaccharide moiety and a peptide (e.g., a peptide including a 1 -5 amino acid residue(s)) attached to the first cyclic heptapeptide (e.g., L 1 in the compound of formula (XIII)), and a third linker may connect a second monosaccharide or oligosaccharide moiety and a peptide (e.g., a peptide including a 1 -5 amino acid residue(s)) attached to the second cyclic heptapeptide (e.g., L' 1 in the compound of formula (XIII)).
  • a first monosaccharide or oligosaccharide moiety and a peptide e.g., a peptide including a 1 -5 amino
  • Molecules that may be used as linkers include at least two functional groups, which may be the same or different, e.g., two carboxylic acid groups, two amine groups, two sulfonic acid groups, a carboxylic acid group and an amine group, or a carboxy group and a sulfonic acid group.
  • the first functional group may form a covalent linkage with a first component in the compound and the second functional group may form a covalent linkage with the second component in the compound.
  • two arms of a linker may contain two dicarboxylic acids, in which the first carboxylic acid may form a covalent linkage with the first cyclic heptapeptide in the compound and the second carboxylic acid may form a covalent linkage with the second cyclic heptapeptide in the compound, and the third arm of the linker may for a covalent linkage (e.g., a C bond) with one or more monosaccharide or oligosaccharide moieties in the compound.
  • a covalent linkage e.g., a C bond
  • the divalent linker may contain two carboxylic acids, in which the first carboxylic acid may form a covalent linkage with one component (e.g., a first cyclic heptapeptide) in the compound and the second carboxylic acid may form a covalent linkage with another component (e.g., a second cyclic heptapeptide) in the compound.
  • first carboxylic acid may form a covalent linkage with one component (e.g., a first cyclic heptapeptide) in the compound and the second carboxylic acid may form a covalent linkage with another component (e.g., a second cyclic heptapeptide) in the compound.
  • a molecule containing one or more sulfonic acid groups may be used as a linker, in which the sulfonic acid group may form a sulfonamide linkage with a component in the compound.
  • a molecule containing one or more isocyanate groups may be used as a linker, in which the isocyanate group may form a urea linkage with a component in the compound.
  • a molecule containing one or more haloalkyl groups may be used as a linker, in which the haloalkyl group may form a covalent linkage, e.g., C-N and C-0 linkages, with a component in the compound.
  • a linker provides space, rigidity, and/or flexibility between the two or more components.
  • a linker may be a bond, e.g., a covalent bond.
  • the term "bond" refers to a chemical bond, e.g., an amide bond, a disulfide bond, a C-0 bond, a N-N bond, or any kind of bond created from a chemical reaction, e.g., chemical conjugation.
  • a linker includes no more than 250 atoms.
  • a linker includes no more than 250 non- hydrogen atoms.
  • the backbone of a linker includes no more than 250 atoms.
  • the "backbone" of a linker refers to the atoms in the linker that together form the shortest path from one part of a compound to another part of the compound (e.g., the shortest path linking a first cyclic heptapeptide and a second cyclic heptapeptide).
  • the atoms in the backbone of the linker are directly involved in linking one part of a compound to another part of the compound (e.g., linking a first cyclic heptapeptide and a second cyclic heptapeptide).
  • hydrogen atoms attached to carbons in the backbone of the linker are not considered as directly involved in linking one part of the compound to another part of the compound.
  • a linker may comprise a synthetic group derived from, e.g., a synthetic polymer (e.g., a polyethylene glycol (PEG) polymer).
  • a linker may comprise one or more amino acid residues, such as D- or L-amino acid residues.
  • a linker may be a residue of an amino acid sequence (e.g., a 1 -25 amino acid, 1 -10 amino acid, 1 -9 amino acid, 1 -8 amino acid, 1 -7 amino acid, 1 -6 amino acid, 1 -5 amino acid, 1 -4 amino acid, 1 -3 amino acid, 1 -2 amino acid, or 1 amino acid sequence).
  • a linker may comprise one or more, e.g., 1 -100, 1 -50, 1 -25, 1 -10, 1 -5, or 1 -3, optionally substituted alkylene, optionally substituted heteroalkylene (e.g., a PEG unit), optionally substituted alkenylene, optionally substituted heteroalkenylene, optionally substituted alkynylene, optionally substituted heteroalkynylene, optionally substituted cycloalkylene, optionally substituted heterocycloalkylene, optionally substituted cycloalkenylene, optionally substituted heterocycloalkenylene, optionally substituted cycloalkynylene, optionally substituted
  • heterocycloalkynylene optionally substituted arylene, optionally substituted heteroarylene (e.g., pyridine), O, S, NR' (R i is H, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkenyl, optionally substituted heteroalkenyl, optionally substituted alkynyl, optionally substituted heteroalkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted cycloalkenyl, optionally substituted heterocycloalkenyl, optionally substituted cycloalkynyl, optionally substituted heterocycloalkynyl, optionally substituted aryl, or optionally substituted heteroaryl), P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino.
  • R i is H, optionally substituted alkyl, optionally substituted heteroalkyl, optionally
  • a linker may comprise one or more optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene (e.g., a PEG unit), optionally substituted C2-C20 alkenylene (e.g., C2 alkenylene), optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20
  • heteroalkynylene optionally substituted C3-C20 cycloalkylene (e.g., cyclopropylene, cyclobutylene), optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene (e.g., C6 arylene), optionally substituted C2-C15 heteroarylene (e.g., imidazole, pyridine), O, S, NR' (R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2- C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20
  • lipophilic moiety refers to a portion, substituent, or functional group of a compound that is, in general, hydrophobic and non-polar.
  • a moiety is lipophilic if it has a hydrophobicity determined using a cLogP value of greater than 0, such as about 0.25 or greater, about 0.5 or greater, about 1 or greater, about 2 or greater, 0.25-5, 0.5-4 or 2-3.
  • cLogP refers to the calculated partition coefficient of a molecule or portion of a molecule.
  • the partition coefficient is the ratio of concentrations of a compound in a mixture of two immiscible phases at equilibrium (e.g., octanol and water) and measures the hydrophobicity or hydrophilicity of a compound.
  • cLog P can be determined using quantitative structure-property relationship algorithims known in the art (e.g. , using fragment based prediction methods that predict the logP of a compound by determining the sum of its non-overlapping molecular fragments).
  • algorithims for calculating cLog P are known in the art including those used by molecular editing software such as CH EMDRAW® Pro, Version 12.0.2.1092
  • a moiety is considered lipophilic if it has a cLogP value described above in at least one of the above methods.
  • a lipophilic moiety having the stated cLogP value will be considered lipophilic, even though it may have a positive charge or a polar substituent.
  • a lipophilic moiety contains entirely hydrocarbons.
  • a lipophilic moiety may contain one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms independently selected from N, O, and S (e.g. , an indolyl), or one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, halo groups, which, due to the structure of the moiety and/or small differences in electronegativity between the heteroatoms or halo groups and the hydrocarbons, do not induce significant chemical polarity into the lipophilic moiety.
  • a lipophilic moiety having, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms and/or, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, halo atoms may still be considered non-polar.
  • a lipophilic moiety may be optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heteroalkenyl, optionally substituted heteroalkynyl , or optionally substituted heteroaryl, or halo forms thereof, wherein the optional substituents are also lipophilic (such as alkyl, alkenyl , alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl , or heteroaryl) or are not lipophilic but do not change the overall lipophilic character of the moiety, i.e.
  • the moiety has a cLogP value of greater than 0.
  • octanol contains a polar group, OH, but is still a lipophilic moiety.
  • a lipophilic moiety may be benzyl, isobutyl, sec-butyl, isopropyl, n-propyl, methyl, biphenylmethyl, n-octyl , or substituted indolyl (e.g. , alkyl substituted indolyl).
  • a lipophilic moiety may be the side chain of a hydrophobic amino acid residue, e.g ., leucine, isoleucine, alanine, phenylalanine, valine, and proline, or groups such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl , and pyrrolidinyl.
  • lipophilic moieties of the compounds described herein may interact with the hydrophobic portions of lipid A (e.g. , fatty acid side chains of lipid A) when the compounds bind to the membrane of bacterial cells (e.g. , Gram-negative bacterial cells). Due to its position on the cyclic heptapeptide, one or more of R 1 , R 12 , R 15 , R' 1 , R' 12 , and R' 15 may be a lipophilic moiety.
  • positively charged moiety refers to a portion , substituent, or functional group of a compound that contains at least one positive charge.
  • a positively charged moiety contains one or more (e.g., 1 -4, 1 -3, 1 , 2, 3, or 4) heteroatoms independently selected from N, O, and S, for example.
  • a positively charged moiety may possess a pH-dependent positive charge, e.g. , the moiety becomes a positively charged moiety at physiological pH (e.g.
  • pH 7 such as -NH 3 + , -(CH 2 ) 4 NH2, -(CH 2 ) 3 NH2, -(CH 2 ) 2 NH2, -CH2NH2, -(CH 2 )4N(CH 3 )2,
  • a positively charged moiety may be optionally substituted alkamino, optionally substituted heteroalkyl (e.g., optionally substituted heteroalkyl containing 1 -3 nitrogens; -(CH2)4-guanidinium, -(CH2)3-guanidinium, -(CH2)2-guanidinium, -CH2-guanidinium), optionally substituted heterocycloalkyl (e.g., optionally substituted heterocycloalkyl containing 1 -3 nitrogens), or optionally substituted heteroaryl (e.g., optionally substituted heteroaryl containing 1 -3
  • a positively charged moiety may be pH independent such
  • substituents may transform an otherwise lipophilic moiety such as optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heteroalkenyl, optionally substituted heteroalkynyl, or optionally substituted heteroaryl, or halo forms thereof, to a positively charged moiety with the addition of a substituent that imparts a positive charge or a pH dependent positive charge, such as guanidinyl, -NH3 + , -NH2, -NH(CH3), -N(CH3)2, and/or -N(CH3)3 + .
  • a positively charged moiety may be the side chain of an amino acid residue (e.g., a natural or non-natural amino acid residue, such as a D- or L-amino acid residue, that is positively charged at physiological pH (e.g., pH 7), such as the side chain of a basic amino acid residue (e.g., arginine, lysine, histidine, ornithine, diaminobuteric acid, or diaminopropionic acid).
  • positively charged moieties of the compounds described herein interact with the negatively charged portions of lipid A (e.g., phosphates of lipid A) when the compounds bind to the membrane of bacterial cells (e.g., Gram-negative bacterial cells). Due to its position on the cyclic heptapeptide, one or more of R 1 1 , R 13 , R 14 , R' 1 1 , R' 13 , and R' 14 may be a positively charged moiety.
  • polar moiety refers to a portion, substituent, or functional group of a compound that has a chemical polarity induced by atoms with different electronegativity.
  • the polarity of a polar moiety is dependent on the electronegativity between atoms within the moiety and the asymmetry of the structure of the moiety.
  • a polar moiety contains one or more (e.g., 1 -4, 1 -3, 1 , 2, 3, or 4) heteroatoms independently selected from N, O, and S, which may induce chemical polarity in the moiety by having different electronegativity from carbon and hydrogen.
  • a polar moiety interacts with other polar or charged molecules.
  • a polar moiety may be optionally substituted alkamino, optionally substituted heteroalkyl (e.g., N- and/or O-containing
  • heteroalkyl -(CH2)4-carboxylic acid, -(CH2)3-carboxylic acid, -(CH2)2-carboxylic acid, -CH2-carboxylic acid), optionally substituted heterocycloalkyl (e.g., N- and/or O-containing heterocycloalkyl), or optionally substituted heteroaryl (e.g., N- and/or O-containing heteroaryl).
  • a polar moiety may -CH(CH 3 )OH, -CH2OH, -(CH 2 ) 2 CONH2, -CH2CONH2, -CH2COOH, or -(CH 2 ) 2 COOH.
  • substituents may transform an otherwise lipophilic moiety such optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heteroalkenyl, optionally substituted heteroalkynyl, or optionally substituted heteroaryl, or halo forms thereof, to a polar moiety with the addition of a substituent that imparts polarity, such as -OH, -COOH, -COOR, or -CONR2, in which R is H or C1 -C4 alkyl.
  • a polar moiety may be the side chain or a polar or charged amino acid residue (e.g., threonine, serine, glutamine, asparagine, arginine, lysine histidine, aspartic acid, and glutamic acid).
  • a polar moiety is the side chain of threonine.
  • polar moieties of the compounds described herein interact with the negatively charged portions of lipid A (e.g., phosphates of lipid A) when the compounds bind to the membrane of bacterial cells (e.g., Gram-negative bacterial cells). Due to its position on the cyclic heptapeptide, one or more of R 1 , R 12 , R 15 , R' 1 , R' 12 , and R' 15 may be a polar moiety.
  • polymyxin core means a cyclic heptapeptide having the structure:
  • alkyl straight-chain and branched- chain monovalent substituents, as well as combinations of these, containing only C and H when unsubstituted.
  • alkyl group includes at least one carbon-carbon double bond or carbon-carbon triple bond, the alkyl group can be referred to as an "alkenyl” or “alkynyl” group respectively.
  • alkenyl or alkynyl group respectively.
  • the monovalency of an alkyl, alkenyl, or alkynyl group does not include the optional substituents on the alkyl, alkenyl, or alkynyl group.
  • alkyl, alkenyl, or alkynyl group is attached to a compound
  • monovalency of the alkyl, alkenyl, or alkynyl group refers to its attachment to the compound and does not include any additional substituents that may be present on the alkyl, alkenyl, or alkynyl group.
  • the alkyl or heteroalkyl group may contain, e.g., 1 -20.
  • the alkenyl, heteroalkenyl, alkynyl, or heteroalkynyl group may contain, e.g., 2-20, 2-18, 2-16, 2-14, 2-12, 2-10, 2-8, 2-6, or 2-4 carbon atoms (e.g., C2-C20, C2-C18, C2-C16, C2-C14, C2-C12, C2-C10, C2-C8, C2-C6, or C2-C4). Examples include, but are not limited to, methyl, ethyl, isobutyl, sec-butyl, tert-butyl, 2-propenyl, and 3-butynyl.
  • cycloalkyl represents a monovalent saturated or unsaturated non- aromatic cyclic alkyl group.
  • a cycloalkyl may have, e.g., three to twenty carbons (e.g., a C3-C7, C3-C8, C3-C9, C3-C10, C3-C1 1 , C3-C12, C3-C14, C3-C16, C3-C18, or C3-C20 cycloalkyi).
  • Examples of cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl.
  • the cycloalkyi group when the cycloalkyi group includes at least one carbon-carbon double bond, the cycloalkyi group can be referred to as a "cycloalkenyl" group.
  • a cycloalkenyl may have, e.g., four to twenty carbons (e.g., a C4- C7, C4-C8, C4-C9, C4-C10, C4-C1 1 , C4-C12, C4-C14, C4-C16, C4-C18, or C4-C20 cycloalkenyl).
  • Exemplary cycloalkenyl groups include, but are not limited to, cyclopentenyl, cyclohexenyl, and cycloheptenyl.
  • the cycloalkyi group can be referred to as a "cycloalkynyl" group.
  • a cycloalkynyl may have, e.g., eight to twenty carbons (e.g., a C8-C9, C8-C10, C8-C1 1 , C8-C12, C8-C14, C8-C16, C8-C18, or C8-C20 cycloalkynyl).
  • cycloalkyi also includes a cyclic compound having a bridged multicyclic structure in which one or more carbons bridges two non-adjacent members of a monocyclic ring, e.g., bicyclo[2.2.1 .]heptyl and adamantane.
  • cycloalkyi also includes bicyclic, tricyclic, and tetracyclic fused ring structures, e.g., decalin and spiro cyclic compounds.
  • aryl refers to any monocyclic or fused ring bicyclic or tricyclic system which has the characteristics of aromaticity in terms of electron distribution throughout the ring system, e.g., phenyl, naphthyl, or phenanthrene.
  • a ring system contains 5-15 ring member atoms or 5-10 ring member atoms.
  • An aryl group may have, e.g., five to fifteen carbons (e.g., a C5-C6, C5-C7, C5-C8, C5-C9, C5-C10, C5-C1 1 , C5-C12, C5-C13, C5-C14, or C5-C15 aryl).
  • five to fifteen carbons e.g., a C5-C6, C5-C7, C5-C8, C5-C9, C5-C10, C5-C1 1 , C5-C12, C5-C13, C5-C14, or C5-C15 aryl.
  • heteroaryl also refers to such monocyclic or fused bicyclic ring systems containing one or more, e.g., 1 - 4, 1 -3, 1 , 2, 3, or 4, heteroatoms selected from O, S and N.
  • a heteroaryl group may have, e.g., two to fifteen carbons (e.g., a C2-C3, C2-C4, C2-C5, C2-C6, C2-C7, C2-C8, C2-C9.
  • the inclusion of a heteroatom permits inclusion of 5-membered rings to be considered aromatic as well as 6-membered rings.
  • heteroaryl systems include, e.g., pyridyl, pyrimidyl, indolyl, benzimidazolyl, benzotriazolyl, isoquinolyl, quinolyl, benzothiazolyl, benzofuranyl, thienyl, furyl, pyrrolyl, thiazolyl, oxazolyl, isoxazolyl, benzoxazolyl, benzoisoxazolyl, and imidazolyl. Because tautomers are possible, a group such as phthalimido is also considered heteroaryl.
  • the aryl or heteroaryl group is a 5- or 6-membered aromatic rings system optionally containing 1 -2 nitrogen atoms.
  • the aryl or heteroaryl group is an optionally substituted phenyl, pyridyl, indolyl, pyrimidyl, pyridazinyl, benzothiazolyl, benzimidazolyl, pyrazolyl, imidazolyl, isoxazolyl, thiazolyl, or imidazopyridinyl.
  • the aryl group is phenyl.
  • an aryl group may be optionally substituted with a substituent such an aryl substituent, e.g., biphenyl.
  • alkaryl refers to an aryl group that is connected to an alkylene, alkenylene, or alkynylene group. In general, if a compound is attached to an alkaryl group, the alkylene, alkenylene, or alkynylene portion of the alkaryl is attached to the compound.
  • an alkaryl is C6- C35 alkaryl (e.g., C6-C16, C6-C14, C6-C12, C6-C10, C6-C9, C6-C8, C7, or C6 alkaryl), in which the number of carbons indicates the total number of carbons in both the aryl portion and the alkylene, alkenylene, or alkynylene portion of the alkaryl.
  • alkaryls include, but are not limited to, (C1 - C8)alkylene(C6-C12)aryl, (C2-C8)alkenylene(C6-C12)aryl, or (C2-C8)alkynylene(C6-C12)aryl.
  • an alkaryl is benzyl.
  • one or more heteroatoms selected from N, O, and S may be present in the alkylene, alkenylene, or alkynylene portion of the alkaryl group and/or may be present in the aryl portion of the alkaryl group.
  • the substituent may be present on the alkylene, alkenylene, or alkynylene portion of the alkaryl group and/or may be present on the aryl portion of the alkaryl group.
  • amino represents -N(R X )2 or -N + (R X )3, where each R x is,
  • H independently, H, alkyl, alkenyl, alkynyl, aryl, alkaryl, cycloalkyl, or two R x combine to form a
  • the amino group is -NH2.
  • alkamino refers to an amino group, described herein, that is attached to an alkylene (e.g., C1 -C5 alkylene), alkenylene (e.g., C2-C5 alkenylene), or alkynylene group (e.g., C2- C5 alkenylene).
  • alkylene e.g., C1 -C5 alkylene
  • alkenylene e.g., C2-C5 alkenylene
  • alkynylene group e.g., C2- C5 alkenylene
  • the amino portion of an alkamino refers to -N(R X )2 or -N + (R X )3, where each R x is, independently, H, alkyl, alkenyl, alkynyl, aryl, alkaryl, cycloalkyl, or two R x combine to form a heterocycloalkyl.
  • the amino portion of an alkamino is -NH2.
  • An example of an alkamino group is C1 -C5 alkamino, e.g., C2 alkamino (e.g., CH2CH2NH2 or CH2CH2N(CH3)2).
  • heteroalkamino group one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms selected from N, O, and S may be present in the alkylene, alkenylene, or alkynylene portion of the heteroalkamino group.
  • an alkamino group may be optionally substituted.
  • the substituent may be present on the alkylene, alkenylene, or alkynylene portion of the alkamino group and/or may be present on the amino portion of the alkamino group.
  • alkamide refers to an amide group that is attached to an alkylene (e.g., C1 -C5 alkylene), alkenylene (e.g., C2-C5 alkenylene), or alkynylene (e.g., C2-C5 alkenylene) group.
  • alkylene e.g., C1 -C5 alkylene
  • alkenylene e.g., C2-C5 alkenylene
  • alkynylene e.g., C2-C5 alkenylene
  • the amide portion of an alkamide refers to -C(0)-N(R X )2, where each R x is, independently, H, alkyl, alkenyl, alkynyl, aryl, alkaryl, cycloalkyl, or two R x combine to form a heterocycloalkyl.
  • the amide portion of an alkamide is -C(0)NH2.
  • An alkamide group may be -(CH2)2-C(0)NH2 or -CH2-C(0)NH2.
  • heteroalkamide group one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms selected from N, O, and S may be present in the alkylene, alkenylene, or alkynylene portion of the heteroalkamide group.
  • an alkamide group may be optionally substituted.
  • the substituent may be present on the alkylene, alkenylene, or alkynylene portion of the alkamide group and/or may be present on the amide portion of the alkamide group.
  • alkylene refers to divalent groups having a specified size.
  • an alkylene may contain, e.g., 1 -20, 1 -18, 1 -16, 1 -14, 1 - 12, 1 -10, 1 -8, 1 -6, 1 -4, or 1 -2 carbon atoms (e.g., C1 -C20, C1 -C18, C1 -C1 6, C1 -C14, C1 -C12, C1 -C1 0, C1 -C8, C1 -C6, C1 -C4, or C1 -C2).
  • an alkenylene or alkynylene may contain, e.g., 2-20, 2-18, 2-16, 2-14, 2-12, 2-10, 2-8, 2-6, or 2-4 carbon atoms (e.g., C2-C20, C2-C18, C2-C16, C2- C14, C2-C12, C2-C10, C2-C8, C2-C6, or C2-C4).
  • Alkylene, alkenylene, and/or alkynylene includes straight-chain and branched-chain forms, as well as combinations of these. The divalency of an alkylene, alkenylene, or alkynylene group does not include the optional substituents on the alkylene, alkenylene, or alkynylene group.
  • two cyclic heptapeptides may be attached to each other by way of a linker that includes alkylene, alkenylene, and/or alkynylene, or combinations thereof.
  • a linker that includes alkylene, alkenylene, and/or alkynylene, or combinations thereof.
  • Each of the alkylene, alkenylene, and/or alkynylene groups in the linker is considered divalent with respect to the two attachments on either end of alkylene, alkenylene, and/or alkynylene group.
  • a linker includes -(optionally substituted alkylene)-(optionally substituted alkenylene)-(optionally substituted alkylene)-, the alkenylene is considered divalent with respect to its attachments to the two alkylenes at the ends of the linker.
  • the optional substituents on the alkenylene are not included in the divalency of the alkenylene.
  • the divalent nature of an alkylene, alkenylene, or alkynylene group refers to both of the ends of the group and does not include optional substituents that may be present in an alkylene, alkenylene, or alkynylene group. Because they are divalent, they can link together multiple (e.g., two) parts of a compound, e.g., a first cyclic heptapeptide and a second cyclic heptapeptide.
  • Alkylene, alkenylene, and/or alkynylene groups can be substituted by the groups typically suitable as substituents for alkyl, alkenyl and alkynyl groups as set forth herein.
  • -HCR-C ⁇ C- may be considered as an optionally substituted alkynylene and is considered a divalent group even though it has an optional substituent, R.
  • Heteroalkylene, heteroalkenylene, and/or heteroalkynylene groups refer to alkylene, alkenylene, and/or alkynylene groups including one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms, e.g., N, O, and S.
  • a polyethylene glycol (PEG) polymer or a PEG unit -(CH2)2-0- in a PEG polymer is considered a heteroalkylene containing one or more oxygen atoms.
  • cycloalkylene refers to a divalent cyclic group linking together two parts of a compound. For example, one carbon within the cycloalkylene group may be linked to one part of the compound, while another carbon within the cycloalkylene group may be linked to another part of the compound.
  • a cycloalkylene group may include saturated or unsaturated non-aromatic cyclic groups.
  • a cycloalkylene may have, e.g., three to twenty carbons in the cyclic portion of the cycloalkylene (e.g., a C3-C7, C3-C8, C3-C9, C3-C10, C3-C1 1 , C3-C12, C3-C14, C3-C16, C3-C18, or C3-C20 cycloalkylene).
  • the cycloalkylene group includes at least one carbon-carbon double bond
  • the cycloalkylene group can be referred to as a "cycloalkenylene" group.
  • a cycloalkenylene may have, e.g., four to twenty carbons in the cyclic portion of the cycloalkenylene (e.g., a C4-C7, C4-C8, C4-C9. C4-C10, C4-C1 1 , C4- C12, C4-C14, C4-C16, C4-C18, or C4-C20 cycloalkenylene).
  • the cycloalkylene group includes at least one carbon-carbon triple bond
  • the cycloalkylene group can be referred to as a "cycloalkynylene" group.
  • a cycloalkynylene may have, e.g., four to twenty carbons in the cyclic portion of the
  • cycloalkynylene e.g., a C4-C7, C4-C8, C4-C9. C4-C1 0, C4-C1 1 , C4-C12, C4-C14, C4-C16, C4-C18, or C8-C20 cycloalkynylene).
  • a cycloalkylene group can be substituted by the groups typically suitable as substituents for alkyl, alkenyl and alkynyl groups as set forth herein.
  • Heterocycloalkylene refers to a cycloalkylene group including one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms, e.g., N, O, and S. Examples of cycloalkylenes include, but are not limited to, cyclopropylene and cyclobutylene.
  • a tetrahydrofuran may be considered as a heterocycloalkylene.
  • arylene refers to a multivalent (e.g., divalent or trivalent) aryl group linking together multiple (e.g., two or three) parts of a compound. For example, one carbon within the arylene group may be linked to one part of the compound, while another carbon within the arylene group may be linked to another part of the compound.
  • An arylene may have, e.g., five to fifteen carbons in the aryl portion of the arylene (e.g., a C5-C6, C5-C7, C5-C8, C5-C9.
  • arylene group can be substituted by the groups typically suitable as substituents for alkyl, alkenyl and alkynyl groups as set forth herein.
  • Heteroarylene refers to an aromatic group including one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms, e.g., N, O, and S.
  • a heteroarylene group may have, e.g., two to fifteen carbons (e.g., a C2-C3, C2-C4, C2-C5, C2-C6, C2-C7, C2-C8, C2- C9. C2-C10, C2-C1 1 , C2-C12, C2-C13, C2-C14, or C2-C15 heteroarylene).
  • substituents include, but are not limited to, alkyl, alkenyl, alkynyl, aryl, alkaryl, acyl, heteroaryl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroalkaryl, halogen, oxo, cyano, nitro, amino, alkamino, hydroxy, alkoxy, alkanoyl, carbonyl, carbamoyl, guanidinyl, ureido, amidinyl, any of the groups or moieties described above, and hetero versions of any of the groups or moieties described above.
  • Substituents include, but are not limited to, F, CI, methyl, phenyl, benzyl, OR, NR 2 , SR, SOR, S0 2 R, OCOR, NRCOR, NRCONR2, NRCOOR, OCONR2, RCO, COOR, alkyl-OOCR, SO3R, CONR2, SO2NR2, NRSO2NR2, CN, CF 3 , OCF3, SiR 3 , and NO2, wherein each R is, independently, H, alkyl, alkenyl, aryl, heteroalkyl, heteroalkenyl, or heteroaryl, and wherein two of the optional substituents on the same or adjacent atoms can be joined to form a fused, optionally substituted aromatic or nonaromatic, saturated or unsaturated ring which contains 3-8 members, or two of the optional substituents on the same atom can be joined to form an optionally substituted aromatic or nonaromatic, saturated or unsaturated ring which contains 3-8 members
  • an optionally substituted group or moiety refers to a group or moiety (e.g., any one of the groups or moieties described above) in which one of the atoms (e.g., a hydrogen atom) is optionally replaced with another substituent.
  • an optionally substituted alkyl may be an optionally substituted methyl, in which a hydrogen atom of the methyl group is replaced by, e.g., OH.
  • a substituent on a heteroalkyl or its divalent counterpart, heteroalkylene may replace a hydrogen on a carbon or a hydrogen on a heteroatom such as N.
  • group -R-NH-R- may be substituted with an alkamide substituent, e.g., -R-N[(CH 2 C(0)N(CH 3 )2]-R.
  • an optional substituent is a noninterfering substituent.
  • a “noninterfering substituent” refers to a substituent that leaves the ability of the compounds described herein (e.g., compounds of any one of formulas (l)-(XXXX)) to either bind to lipopolysaccharides (LPS) or to kill or inhibit the growth of Gram- negative bacteria qualitatively intact.
  • the substituent may alter the degree of such activity.
  • a noninterfering substituent leaves the ability of a compound described herein (e.g., a compound of any one of formulas (l)-(XXXX)) to kill or inhibit the growth of Gram-negative bacteria qualitatively intact as determined by measuring the minimum inhibitory concentration (MIC) against at least one Gram- negative bacteria as known in the art, wherein the MIC is 32 ⁇ g/mL or less.
  • MIC minimum inhibitory concentration
  • a noninterfering substituent leaves the ability of a compound described herein (e.g., a compound of any one of formulas (l)-(XXXX)) to bind to lipopolysaccharides (LPS) from the cell membrane of Gram-negative bacteria qualitatively intact, as determined by an LPS binding assay (e.g., see Example 103), wherein the compound shows a value of about 1 0% or greater displacement of a fluorogenic substrate at 250 ⁇ of the compound.
  • LPS lipopolysaccharides
  • hetero when used to describe a chemical group or moiety, refers to having at least one heteroatom that is not a carbon or a hydrogen, e.g., N, O, and S. Any one of the groups or moieties described above may be referred to as hetero if it contains at least one heteroatom.
  • a heterocycloalkyi, heterocycloalkenyl, or heterocycloalkynyl group refers to a cycloalkyi, cycloalkenyl, or cycloalkynyl group that has one or more heteroatoms independently selected from, e.g., N, O, and S.
  • An example of a heterocycloalkenyl group is a maleimido.
  • a heteroaryl group refers to an aromatic group that has one or more heteroatoms independently selected from, e.g., N, O, and S.
  • One or more heteroatoms may also be included in a substituent that replaced a hydrogen atom in a group or moiety as described herein.
  • a substituent e.g., methyl
  • the substituent may also contain one or more heteroatoms (e.g., methanol).
  • acyl refers to a group having the structure: , wherein R z is an optionally substituted alkyl, alkenyl, alkynyl, cycloalkyi, cycloalkenyl, cycloalkynyl, aryl, alkaryl, alkamino, heteroalkyl, heteroalkenyl, heteroalkynyl, heterocycloalkyi, heterocycloalkenyl,
  • heterocycloalkynyl heteroaryl, heteroalkaryl, or heteroalkamino.
  • halo refers to any halogen atom, e.g., F, CI, Br, or I. Any one of the groups or moieties described herein may be referred to as a "halo moiety" if it contains at least one halogen atom, such as haloalkyl.
  • hydroxyl represents an -OH group.
  • carbonyl refers to a group having the structure
  • thiocarbonyl refers to a group having the structure: ⁇ ⁇
  • phosphate represents the group having the structure: O "
  • sulfonyl represents the group having the structure V NR
  • amino represents the group having the structure: ⁇ ⁇ , wherein R is an optional substituent.
  • AV-protecting group represents those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used AV-protecting groups are disclosed in Greene, "Protective Groups in Organic Synthesis,” 5th Edition (John Wiley & Sons, New York, 2014), which is incorporated herein by reference.
  • AV-protecting groups include, e.g., acyl, aryloyl, and carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, a-chlorobutyryl, benzoyl, carboxybenzyl (CBz), 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L or D, L-amino acid residues such as alanine, leucine, phenylalanine;
  • sulfonyl-containing groups such as benzenesulfonyl and p-toluenesulfonyl ; carbamate forming groups such as benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p- nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3,4- dimethoxybenzyloxycarbonyl, 3,5-dimethoxybenzyl oxycarbonyl, 2,4-dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4,5-dimethoxybenzyloxycarbonyl,
  • diisopropylmethoxycarbonyl isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2,-trichloroethoxycarbonyl, phenoxycarbonyl, 4-nitrophenoxy carbonyl, fluorenyl-9-methoxycarbonyl (Fmoc), cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl, and phenylthiocarbonyl; alkaryl groups such as benzyl, triphenylmethyl, and benzyloxymethyl ; and silyl groups such as trimethylsilyl.
  • amino acid means naturally occurring amino acids and non-naturally occurring amino acids.
  • naturally occurring amino acids means amino acids including Ala,
  • non-naturally occurring amino acid means an alpha amino acid that is not naturally produced or found in a mammal.
  • non-naturally occurring amino acids include D- amino acids; an amino acid having an acetylaminomethyl group attached to a sulfur atom of a cysteine; a pegylated amino acid; the omega amino acids of the formula NH2(CH2) n COOH where n is 2-6, neutral nonpolar amino acids, such as sarcosine, t-butyl alanine, t-butyl glycine, N-methyl isoleucine, and norleucine; oxymethionine; phenylglycine; citrulline; methionine sulfoxide; cysteic acid; ornithine;
  • diaminobutyric acid 3-aminoalanine; 3-hydroxy-D-proline; 2,4-diaminobutyric acid; 2-aminopentanoic acid; 2-aminooctanoic acid, 2-carboxy piperazine; piperazine-2-carboxylic acid, 2-amino-4-phenylbutanoic acid; 3-(2-naphthyl)alanine, and hydroxyproline.
  • amino acids are a-aminobutyric acid, a-amino-a- methylbutyrate, aminocyclopropane-carboxylate, aminoisobutyric acid, aminonorbornyl-carboxylate, L- cyclohexylalanine, cyclopentylalanine, L-N-methylleucine, L-N-methylmethionine, L-N-methylnorvaline, L- N-methylphenylalanine, L-N-methylproline, L-N-methylserine, L-N-methyltryptophan, D-ornithine, L-N- methylethylglycine, L-norleucine, a-methyl-aminoisobutyrate, a-methylcyclohexylalanine, D-a- methylalanine, D-a-methylarginine, D-a-methylasparagine, D-a-methylaspartate, D-a-methylcysteine
  • amino acid residues may be charged or polar.
  • Charged amino acids include alanine, lysine, aspartic acid, or glutamic acid, or non-naturally occurring analogs thereof.
  • Polar amino acids include glutamine, asparagine, histidine, serine, threonine, tyrosine, methionine, or tryptophan, or non-naturally occurring analogs thereof.
  • a terminal amino group in the amino acid may be an amido group or a carbamate group.
  • antibacterial agent refers to an agent that is used in addition to one or more of the compounds described herein (e.g., compounds of any one of formulas (l)-(XXXX)) in methods of treating a bacterial infection (e.g., Gram-negative bacterial infection) and/or preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria.
  • a bacterial infection e.g., Gram-negative bacterial infection
  • An antibacterial agent may be an agent that prevents the entrance of a bacteria (e.g., a Gram-negative bacteria) into a subject's cells, tissues, or organs, inhibits the growth of a bacteria (e.g., a Gram-negative bacteria) in a subject's cells, tissues, or organs, and/or kills a bacteria (e.g., a Gram-negative bacteria) that is inside a subject's cells, tissues, or organs.
  • a bacteria e.g., a Gram-negative bacteria
  • an antibacterial agent used in addition to a compound described herein is linezolid or tedizolid (e.g. , tedizolid phosphate).
  • bacteria infection refers to the invasion of a subject's cells, tissues, and/or organs by bacteria (e.g., Gram-negative bacteria), thus, causing an infection.
  • bacteria e.g., Gram-negative bacteria
  • the bacteria may grow, multiply, and/or produce toxins in the subject's cells, tissues, and/or organs.
  • a bacterial infection can be any situation in which the presence of a bacterial population(s) is latent within or damaging to a host body.
  • a subject is "suffering" from a bacterial infection when a latent bacterial population is detectable in or on the subject's body, an excessive amount of a bacterial population is present in or on the subject's body, or when the presence of a bacterial population(s) is damaging the cells, tissues, and/or organs of the subject.
  • protecting against refers to preventing a subject from developing a bacterial infection (e.g., a Gram-negative bacterial infection) or decreasing the risk that a subject may develop a bacterial infection (e.g., a Gram-negative bacterial infection).
  • Prophylactic drugs used in methods of protecting against a bacterial infection in a subject are often administered to the subject prior to any detection of the bacterial infection.
  • a subject e.g., a subject at risk of developing a bacterial infection
  • a compound described herein e.g., a compound having any one of formulas (l)-(XXXX)
  • a compound described herein e.g., a compound having any one of formulas (l)-(XXXX)
  • treating refers to a therapeutic treatment of a bacterial infection (e.g., a Gram-negative bacterial infection) in a subject.
  • a therapeutic treatment may slow the progression of the bacterial infection, improve the subject's outcome, and/or eliminate the infection.
  • a therapeutic treatment of a bacterial infection in a subject may alleviate or ameliorate of one or more symptoms or conditions associated with the bacterial infection, diminish the extent of the bacterial infection, stabilize (i.e., not worsening) the state of the bacterial infection, prevent the spread of the bacterial infection, and/or delay or slow the progress of the bacterial infection, as compare the state and/or the condition of the bacterial infection in the absence of the therapeutic treatment.
  • a bacterial infection e.g., a Gram-negative bacterial infection
  • a subject may alleviate or ameliorate of one or more symptoms or conditions associated with the bacterial infection, diminish the extent of the bacterial infection, stabilize (i.e., not worsening) the state of the bacterial infection, prevent the spread of the bacterial infection, and/or delay or slow the progress of the bacterial infection, as compare the state and/or the condition of the bacterial infection in the absence of the therapeutic treatment.
  • LPS-induced nitric oxide (NO) production from a macrophage refers to the ability of the lipopolysaccharides (LPS) in Gram-negative bacteria to activate a macrophage and induce NO production from the macrophage.
  • NO production from a macrophage in response to LPS is a signal of macrophage activation, which may lead to sepsis in a subject, e.g., a Gram-negative bacteria infected subject.
  • the disclosure features compounds (e.g., compounds of any one of formulas (l)-(XXXX)) that are able to bind to LPS in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, thus neutralizing an immune response to LPS (see, e.g., Example 1 1 0).
  • NO production from a macrophage may be measured using available techniques in the art, e.g., a Griess assay, as demonstrated in Example 1 10.
  • resistant strain of bacteria refers to a strain of bacteria (e.g., Gram- negative or Gram-positive bacteria) that is refractory to treatment with an antibiotic, such as an antibiotic described in Section V of Detailed Description.
  • a resistant strain of bacteria contains a mcr- 1 gene, a mcr-2 gene, and/or a mcr-3 gene.
  • a resistant strain of bacteria contains a chromosomal mutation conferring polymyxin resistance. In some embodiments, a resistant strain of bacteria contains a mcr- 1 gene, a mcr-2 gene, and/or a mcr-3 gene in combination with other antibiotic resistance genes. In some embodiments, a resistant strain of bacteria is a resistant strain of E. co// ' (e.g., E. coli BAA-2469).
  • activating an immune cell refers to the ability of a compound to directly or indirectly bind to an immune cell to produce an effective immune response.
  • the ability of a compound to directly or indirectly bind to an immune cell to produce an effective immune response may be quantified by measuring the concentration of the compound at which such immune response is produced.
  • the concentration of a compound that binds to an immune cell receptor such as dectin-1 or binds to an antibody (e.g., anti-aGal or anti-aRha antibody, which then binds to an immune cell) to trigger an effective immune response may be less than or equal to 10,000 nM as measured in accordance with, e.g., an enzyme-linked immunosorbent assay (ELISA), as in Examples 107 and 108.
  • an aGal epitope, that binds to an antibody, such as an anti-aGal antibody may be detected using an ELISA.
  • a compound containing a particular monosaccharide or oligosaccharide moiety may be immobilized on a support or surface using conventional techniques in the art. After the compound is immobilized to the surface, an antibody that is specific for the particular monosaccharide or
  • oligosaccharide moiety in the compound is applied over the surface so it is captured by the compound through binding to the monosaccharide or oligosaccharide moiety in the compound.
  • the antibody is often linked to an enzyme (e.g., horseradish peroxidase) for subsequent signal amplification.
  • the enzyme's substrate e.g., 3,3'-diaminobenzidine
  • a measurable signal e.g., color change.
  • the antibody itself can be detected using a secondary antibody, which is linked to an enzyme.
  • the concentration of a compound that binds to an immune cell receptor such as dectin-1 or binds to an antibody (e.g., anti-aGal or anti-aRha antibody, which then binds to an immune cell) to trigger an effective immune response may be less than or equal to 1000 nM or less than or equal to 100 nM as measured in accordance with an ELISA.
  • innate immune receptor refers to a natural receptor, such as a natural receptor on an immune cell, that binds to a carbohydrate (e.g., a monosaccharide or oligosaccharide moiety) or an optionally substituted carbohydrate and causes a response in the immune system.
  • an innate immune receptor binds to the monosaccharide or oligosaccharide moiety of the compounds described herein (e.g., compounds of any one of formulas (l)-(XXXX)).
  • an innate immune receptor binds to a moiety in Table 2A or 2B.
  • natural antibody refers to a naturally existing antibody in the circulation of a mammal (e.g., a human) that has not been previously exposed to deliberate immunization.
  • a natural antibody is an antibody of the immunoglobulin M (IgM) isotype.
  • a natural antibody binds to the monosaccharide or oligosaccharide moiety of the compounds described herein (e.g., compounds of any one of formulas (l)-(XXXX)).
  • a natural antibody binds to a moiety in Table 2A or 2B.
  • a natural antibody is anti-aGal antibobody or anti-aRha antibody.
  • subject can be a human, non-human primate, or other mammal, such as but not limited to dog, cat, horse, cow, pig, turkey, goat, fish, monkey, chicken, rat, mouse, and sheep.
  • substantially simultaneously refers to two or more events that occur at the same time or within a narrow time frame of each other.
  • a compound described herein e.g., a compound of any one of formulas (l)-(XXXX)
  • an antibacterial agent e.g., linezolid or tedizolid
  • the compound and the antibacterial agent are administered together (e.g., in one pharmaceutical composition) or separately but within a narrow time frame of each other, e.g., within 10 minutes, e.g., 1 0, 9, 8, 7, 6, 5, 4, 3, 2, or 1 minute, or 45, 30, 15, or 1 0 seconds of each other.
  • terapéuticaally effective amount refers to an amount, e.g., pharmaceutical dose, effective in inducing a desired effect in a subject or in treating a subject having a condition or disorder described herein (e.g., a bacterial infection (e.g., a Gram-negative bacterial infection)). It is also to be understood herein that a “therapeutically effective amount” may be interpreted as an amount giving a desired therapeutic and/or preventative effect, taken in one or more doses or in any dosage or route, and/or taken alone or in combination with other therapeutic agents (e.g., an antibacterial agent described herein).
  • an effective amount of a compound is, for example, an amount sufficient to prevent, slow down, or reverse the progression of the bacterial infection as compared to the response obtained without administration of the compound.
  • the term "pharmaceutical composition” refers to a medicinal or pharmaceutical formulation that contains at least one active ingredient (e.g., a compound of any one of formulas (I)- (XXXXX)) as well as one or more excipients and diluents to enable the active ingredient suitable for the method of administration.
  • the pharmaceutical composition of the present disclosure includes pharmaceutically acceptable components that are compatible with a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)).
  • a pharmaceutically acceptable carrier refers to an excipient or diluent in a pharmaceutical composition.
  • a pharmaceutically acceptable carrier may be a vehicle capable of suspending or dissolving the active compound (e.g., a compound of any one of formulas (I)- (XXXXX)).
  • the pharmaceutically acceptable carrier must be compatible with the other ingredients of the formulation and not deleterious to the recipient.
  • the pharmaceutically acceptable carrier must provide adequate pharmaceutical stability to a compound described herein.
  • the nature of the carrier differs with the mode of administration. For example, for oral administration, a solid carrier is preferred; for intravenous administration, an aqueous solution carrier (e.g., WFI, and/or a buffered solution) is generally used.
  • pharmaceutically acceptable salt represents salts of the compounds described herein (e.g., compounds of any one of formulas (l)-(XXXX)) that are, within the scope of sound medical judgment, suitable for use in methods described herein without undue toxicity, irritation, and/or allergic response.
  • Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Pharmaceutical Salts: Properties, Selection, and Use (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting the free base group with a suitable organic acid.
  • FIG. 1 is a schematic illustrating a 96-well checkerboard synergy MIC plate layout.
  • FIG. 2 shows the inhibition of nitric oxide (NO) production by Compound 14.
  • FIGS. 3A-3H show the time-kill analysis for Compound 14 and colistin (COL), respectively, against E. col 7 ATCC 25922, K. pneumoniae ATCC 43816, P. aeruginosa ATCC 27853, and A. baumannii ATCC 17978 at 0, 1 , 2, 4, and 16X CLSI broth microdilution MIC values.
  • FIG. 4A shows the membrane permeabilization induced by Compound 14, Compound 54a, and
  • FIG. 4B shows the bacterial cell death induced by Compound 14, Compound 54a, and Compound 59, as quantified by SYTOX green signal.
  • FIGS. 5A and 5B show that Rha mediates binding of human Rha antibody to E. coli ATCC 25922-bound Compound 14, 54a, and Compound 59 in an antigen-specific manner. Bound antibodies were detected using either an anti-human IgG secondary antibody (FIG. 5A) or an anti-human IgM secondary antibody (FIG. 5B).
  • FIG. 5C shows inhibition of Rha-Ab binding to Compound 14 by L-Rha monosaccharide.
  • FIG. 6 shows Rha-specific binding of purified rabbit Rha antibodies to bacteria.
  • FIG. 7 shows Rha-specific binding of purified human Rha antibodies to bacteria.
  • FIGS. 8A-8F show that the killing of E. coli is enhanced by Compound 14, Compound 54a, and
  • FIGS. 9A-9D show that complement dependent cytotoxicity (CDC) is enhanced by Compound 14, Compund 54a, and Compound 59.
  • FIG. 10 shows that Compound 14 displays rAb-dependent efficacy in a mouse septicemia model.
  • FIG. 1 1 shows that Compound 14 displays rAb-dependent efficacy in a neutropenic mouse E. coli thigh infection model.
  • FIG. 12 shows the titration of rAb in the mouse E. coli septicemia model with a fixed dose of
  • FIG. 13 shows the metabolic stability of Compound 14 in rat, monkey and human hepatocytes.
  • FIG. 14 shows the titration of Compound 14 in functional potassium channel assays.
  • FIG. 15 shows the absorbance values at 450 nm for E. coli incubated with Compound 14 and a control compound (Compound 14 without the monosaccharide portion).
  • the disclosure features compounds, compositions, and methods for the treatment of bacterial infections (e.g., Gram-negative bacterial infections).
  • the compounds disclosed herein include dimers of cyclic heptapeptides (e.g., two polymyxin cores) linked to each other through a linker and further conjugated to one or more monosaccharide or oligosaccharide moieties.
  • the dimers of cyclic heptapeptides are linked to each other through a linker and/or one or two peptides (e.g., a peptide including a 1 -5 amino acid residue(s)).
  • the compounds can be used in the treatment of bacterial infections caused by Gram-negative bacteria.
  • one or more of the compounds described herein are used in combination with an antibacterial agent (e.g., linezolid or tedizolid (e.g., tedizolid phosphate)).
  • an antibacterial agent e.g., linezolid or tedizolid (e.
  • Bacteria cause bacterial infections and diseases such as tuberculosis, pneumonia, and foodborne illnesses. Bacteria may be categorized into two major types: Gram-positive bacteria and Gram-negative bacteria. Gram-positive bacteria possess a thick cell wall containing multiple layers of peptidoglycan and teichoic acids, while Gram-negative bacteria have a relatively thin cell wall containing fewer layers of peptidoglycan that are surrounded by a second lipid membrane containing
  • LPS lipopolysaccharides
  • lipoproteins lipoproteins.
  • LPS also called endotoxins, are composed of
  • Gram-positive bacteria include, but are not limited to, bacteria in the genus Streptococcus (e.g., Streptococcus pyogenes), bacteria in the genus Staphylococcus (e.g., Staphylococcus cohnii), bacteria in the genus Corynebacterium (e.g., Corynebacterium auris), bacteria in the genus Listeria (e.g., Listeria grayi), bacteria in the genus Bacillus (e.g., Bacillus aerius), and bacteria in the genus Clostridium (e.g., Clostridium acetium).
  • Streptococcus e.g., Streptococcus pyogenes
  • Staphylococcus e.g., Staphylococcus cohnii
  • Corynebacterium e.g., Corynebacterium auris
  • Listeria e.g
  • Gram-negative bacteria examples include, but are not limited to, bacteria in the genus Escherichia (e.g., Escherichia coli), bacteria in the genus Klebsiella (e.g., Klebsiella granulomatis, Klebsiella oxytoca, Klebsiella pneumoniae, Klebsiella terrigena, and Klebsiella variicola), bacteria in the genus Acinetobacter (e.g., Acinetobacter baumannii,
  • Pseudomonas e.g., Pseudomonas aeruginosa
  • Neisseria e.g., Neisseria gonorrhoeae
  • Bacteria may evolve to become more or fully resistant to antibiotics. Resistance may arise through natural resistance in certain types of bacteria, spontaneous random genetic mutations, and/or by inter- or intra-species horizontal transfer of resistance genes. Resistant bacteria are increasingly difficult to treat, requiring alternative medications or higher doses, which may be more costly or more toxic. Bacteria resistant to multiple antibiotics are referred to as multidrug resistant (MDR) bacteria.
  • MDR multidrug resistant
  • the mcr- 1 gene encodes a phosphoethanolamine transferase (MCR-1 ) which confers resistance to colistin, a natural polymyxin, through modification of LPS. This is the first known horizontally- transferable resistance determinant for the polymyxin class of antibiotics.
  • the mcr- 1 gene has also been found in bacterial strains which already possess resistance to other classes of antibiotics, such as in carbapenem-resistant Enterobacteriaceae (CRE).
  • An mcr- 1 resistance plasmid refers to a bacterial plasmid that carries mcr- 1 alone or in combination with other antibiotic resistance genes.
  • a mcr- 1 resistance plasmid refers to a bacterial plasmid that carries one or more antibiotic resistance genes.
  • mcr- 1 resistance plasmids include, but are not limited to, pHNSHP45, pMR0516mcr, pESTMCR, pAF48, pAF23, pmcr1 -lncX4, pmcr1 -lncl2, pA31 -12, pVT553, plCBEC72Hmcr, pE15004, pE1 5015, and pE1 5017.
  • the mcr-2 gene also confers resistance to colistin.
  • the mcr-2 gene was identified in porcine and bovine colistin-resistance E.coli that did not contain mcr- 1 (Xavier et al., Euro Si/rve/7/ 21 (27), 2016).
  • the mcr-2 gene is a 1 ,617 bp phspoethanolamine transferase harbored on an lncX4 plasmid.
  • the mcr-2 gene has 76.7% nucleotide identity to mcr- 1.
  • mcr-2 resistance plasmid refers to a bacterial plasmid that carries mcr-2 alone or in combination with other antibiotic resistance genes.
  • a mcr-2 resistance plasmid refers to a bacterial plasmid that carries one or more antibiotic resistance genes.
  • Mcr-2 resistance plasmids include, but are not limited to, pKP37-BE and pmcr2-lncX4.
  • the mcr-3 gene also confers resistance to colistin.
  • the mcr-3 gene is considered to include variants which encode an MCR-3 protein that confers resistance to cholistin, such as mcr-3 which encodes a protein having an amino acid substitution at V413A.
  • the mcr-3 gene was identified in cholistin-resistant E.coli isolated from swine (Clemente et al., 27 th ECCMID, Vienna, Austria, 2017).
  • the mcr3 gene is harbored on an lncX4 plasmid. Analysis of mcr-3 harboring plasmids from E.coli isolates shows that the mobile element harboring mcr-3 is the IS26 element.
  • a mcr-3 resistance plasmid refers to a bacterial plasmid that carries one or more antibiotic resistance genes.
  • resistant strain E. coli BAA-2469 possesses the New Delhi metallo-p-lactamase (NDM-1 ) enzyme, which makes bacteria resistant to a broad range of ⁇ -lactam antibiotics. Additionally, E.
  • coli BAA-2469 is also known to be resistatnt to penicillins (e.g., ticarcillin, ticarcillin/clavulanic acid, piperacillin, ampicillin, and ampicillin/sulbactam), cephalosporins (e.g., cefalotin, cefuroxime, cefuroxime, cefotetan, cefpodoxime, cefotaxime, ceftizoxime, cefazolin, cefoxitin, ceftazidime, ceftriaxone, and cefepime), carbapenems (e.g., doripenem, meropenem, ertapenem, imipenem), quinolones (e.g., nalidixic acid, moxifloxacin, norfloxacin, ciprofloxacin, and levofloxacin), aminoglycosides (e.g., amikacin, gentamicin, and tobra
  • a resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene, and/or a chromosomal mutation conferring polymyxin resistance.
  • a resistant strain of bacteria is a resistant strain of E. coli (e.g., E. coli BAA-2469).
  • the compounds disclosed herein include three components: (i) a first cyclic heptapeptide (e.g., a first polymyxin core), (ii) a second cyclic heptapeptide (e.g., a second polymyxin core), and (iii) one or more monosaccharide or oligosaccharide moieties.
  • the first and second cyclic heptapeptides are linked to each other by way of a linker and/or one or two peptides (e.g., each peptide including a 1 -5 amino acid residue(s)).
  • a cyclic heptapeptide or polymyxin core refers to certain compounds that bind to lipopolysaccharides (LPS) in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics.
  • cyclic heptapeptide refers to certain compounds that kill or inhibit the growth of Gram-negative bacteria as determined by measuring the minimum inhibitory concentration (MIC) against at least one Gram-negative bacteria as known in the art, wherein the MIC is 32 ⁇ g/mL or less.
  • Cyclic heptapeptides are composed of, at least, amino acid residues, each of which may, independently, have a D- or L- configuration, assembled as a cyclic heptapeptide ring.
  • a cyclic heptapeptide includes seven natural or non-natural amino acid residues attached to each other in a closed ring. The ring contains six bonds formed by linking the carbon in the a- carboxyl group of one amino acid residue to the nitrogen in the a-amino group of the adjacent amino acid residue and one bond formed by linking the carbon in the a-carboxyl group of one amino acid residue to the nitrogen in the ⁇ -amino group in the side chain of the adjacent amino acid residue.
  • the nitrogen in the a-amino group of this amino acid residue does not participate directly in forming the ring and serves as the linking nitrogen (thus, referred to as the "linking nitrogen” herein) that links one cyclic heptapeptide or polymyxin core to another cyclic heptapeptide or polymyxin core by way of a linker and/or one or two peptides (e.g., a peptide including a 1 -5 amino acid residue(s)).
  • Compounds described herein contain dimers of cyclic heptapeptides (e.g., two polymyxin cores) that are linked to each other at their linking nitrogens through a linker and/or one or two peptides (e.g., a peptide including a 1 -5 amino acid residue(s)), and one or more monosaccharide or oligosaccharide moieties.
  • cyclic heptapeptides e.g., two polymyxin cores
  • one or two peptides e.g., a peptide including a 1 -5 amino acid residue(s)
  • a peptide including one or more (e.g., 1 -5; 1 , 2, 3, 4, or 5) amino acid residues may be covalently attached to the linking nitrogen of the cyclic heptapeptide or the polymyxin core.
  • Cyclic heptapeptides or polymyxin cores may be derived from polymyxins (e.g., naturally existing polymyxins and non-natural polymyxins) and/or octapeptins (e.g., naturally existing octapeptins and non-natural octapeptins).
  • cyclic heptapeptides may be compounds described in Gallardo-Godoy et al., J. Med. Chem. 59:1068, 2016 (e.g., compounds 1 1 -41 in Table 1 of Gallardo-Godoy et al.), which is incorporated herein by reference in its entirety. Examples of some naturally existing polymyxins and their structures are shown in Table 1 A. Examples of some non-natural polymyxins and their structures are shown in Table 1 B.
  • Table 1 B Non-natural polymyxins and their structures (Dab: diaminobutyric acid; Dap: diaminopropionic acid; Orn: ornithine;
  • compounds described herein bind to the cell membrane of Gram-negative bacteria (e.g., bind to LPS in the cell membrane of Gram-negative bacteria) to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics.
  • the initial association of the compounds with the bacterial cell membrane occurs through electrostatic interactions between the compounds and the anionic LPS in the outer membrane of Gram-negative bacteria, disrupting the arrangement of the cell membrane.
  • compounds described herein may bind to lipid A in the LPS. More specifically, compounds described herein may bind to one or both phosphate groups in lipid A.
  • antibiotic- resistant bacteria e.g., antibiotic-resistant, Gram-negative bacteria
  • compounds described herein may bind to multiple Gram-negative bacterial cells at the same time.
  • the binding of the compounds described herein to the LPS may also displace Mg 2+ and Ca 2+ cations that bridge adjacent LPS molecules, causing, e.g., membrane permeabilization, leakage of cellular molecules, inhibition of cellular respiration, and/or cell death.
  • the monosaccharide or oligosaccharide moieties in the compounds serve as a gradient against which immune cells chemotax to the site of bacterial infection and/or growth.
  • first and second cyclic heptapeptides are the same. In some embodiments, the first and second cyclic heptapeptides are different. Compounds described herein may be synthesized using available chemical synthesis techniques in the art. In some embodiments, available functional groups in the first and second cyclic heptapeptides, the linker, and the one or more monosaccharide or oligosaccharide moieties, e.g., amines, carboxylic acids, and/or hydroxyl groups, may be used in making the compounds described herein.
  • the linking nitrogen in a cyclic heptapeptide may form an amide bond with the carbon in a carboxylic acid group in the linker.
  • a peptide including one or more (e.g., 1 -3; 1 , 2, or 3) amino acid residues may be also be covalently attached to the linking nitrogen of the cyclic heptapeptide through forming an amide bond between the carbon in a carboxylic acid group in the peptide and the linking nitrogen.
  • a molecule may be derivatized using conventional chemical synthesis techniques that are well known in the art.
  • the compounds described herein contain one or more chiral centers.
  • the compounds include each of the isolated stereoisomeric forms as well as mixtures of stereoisomers in varying degrees of chiral purity, including racemic mixtures. It also encompasses the various diastereomers, enantiomers, and tautomers that can be formed.
  • each E is, independently, a monosaccharide or oligosaccharide moiety
  • L' is a linker covalently attached to E and to the linking nitrogen in each of M1 and M2
  • n is 1 , 2, 3, or 4 (e.g., 1 or 2).
  • each E can, independently, be connected to an atom in L'.
  • L' in the compound described by formula (I) is described by formula (L):
  • L is a remainder of L';
  • A1 is a 1 -5 amino acid peptide covalently attached to the linking nitrogen in M1 or is absent; and
  • A2 is a 1 -5 amino acid peptide covalently attached to the linking nitrogen in M2 or is absent.
  • the disclosure provides a compound, or a pharmaceutically acceptable salt thereof, described by formula (II):
  • L is a remainder of L'; n is 1 , 2, 3, or 4 (e.g., 1 or 2); each E is, independently, a monosaccharide or oligosaccharide moiety; each of R 1 , R 12 , R' 1 , and R' 12 is, independently, a lipophilic moiety, a polar moiety, or H; each of R 1 1 , R 13 , R 14 , R' 1 1 , R' 13 , and R' 14 is, independently, optionally substituted C1 -C5 alkamino, a polar moiety, a positively charged moiety, or H; each of R 15 and R' 15 is, independently, a lipophilic moiety or a polar moiety; each of R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R' 2 , R' 3 , R' 4 , R' 5 , R' 6 , R' 7
  • each of R 2 , R 3 , and R 4 is, independently, optionally substituted C1 -C20 alkylene or optionally substituted C1 -C20 heteroalkylene.
  • each of R 5 , R 6 , and R 7 is, independently, optionally substituted C1 -C20 alkylene or optionally substituted C1 -C20 heteroalkylene.
  • each of R 8 , R 9 , and R 10 is, independently, optionally substituted C1 -C20 alkylene or optionally substituted C1 -C20 heteroalkylene.
  • each of R' 2 , R' 3 , and R' 4 is, independently, optionally substituted C1 -C20 alkylene or optionally substituted C1 -C20 heteroalkylene.
  • each of R' 5 , R' 6 , and R' 7 is, independently, optionally substituted C1 -C20 alkylene or optionally substituted C1 -C20 heteroalkylene.
  • each of R' 8 , R' 9 , and R' 10 is, independently, optionally substituted C1 -C20 alkylene or optionally substituted C1 -C20 heteroalkylene.
  • the compound of formula (II) has at least one optionally substituted 5-8 membered ring formed by joining (i) R 2 , R 3 , and C 1 ; (ii) R 3 , R 4 , N 1 , and C 1 ; (iii) R 5 , R 6 , and C 2 ; (iv) R 6 , R 7 , N 2 , and C 2 ; (v) R 8 , R 9 , and C 3 ; (vi) R 9 , R 10 , N 3 , and C 3 ; (vii) R' 2 , R' 3 , and C' 1 ; (viii) R' 3 , R' 4 , N' 1 , and C' 1 ; (ix) R' 5 , R' 6 , and C' 2 ; (x) R' 6 , R' 7 , N' 2 , and C' 2 ; (xi) R' 8 , R' 9 , and C' 3 ; or (xiii
  • each of R 2 , R 3 , and R 4 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
  • R 2 , R 3 , and C 1 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R 4 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R 2 , R 3 , and C 1 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 hetero
  • cycloalkenylene optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.
  • each of R 5 , R 6 , and R 7 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
  • R 5 , R 6 , and C 2 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R 7 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkaryl; or (ii) R 5 , R 6 , and C 2 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O
  • cycloalkenylene optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.
  • each of R 8 , R 9 , and R 10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
  • R 8 , R 9 , and C 3 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R 10 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R 8 , R 9 , and C 3 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 hetero
  • cycloalkenylene optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.
  • each of R' 2 , R' 3 , and R' 4 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
  • R' 2 , R' 3 , and C' 1 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R' 4 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R' 2 , R' 3 , and C' 1 together form
  • cycloalkenylene optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.
  • each of R' 5 , R' 6 , and R' 7 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
  • R' 5 , R' 6 , and C' 2 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R' 7 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R' 5 , R' 6 , and C' 2 together form
  • cycloalkenylene optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.
  • each of R' 8 , R' 9 , and R' 10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
  • R' 8 , R' 9 , and C' 3 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R' 10 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R' 8 , R' 9 , and C' 3 together form
  • cycloalkenylene optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.
  • the disclosure provides a compound, or a pharmaceutically acceptable salt thereof, described by formula (III):

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Compositions and methods for the treatment of bacterial infections include compounds containing dimers of cyclic heptapeptides conjugated to one or more monosaccharide or oligosaccharide moieties. In particular, compounds can be used in the treatment of bacterial infections caused by Gram-negative bacteria.

Description

COMPOSITIONS AND METHODS FOR THE TREATMENT OF BACTERIAL INFECTIONS
Background
The need for novel antibacterial treatments for bacterial infections is significant and especially critical in the medical field. Antibacterial resistance is a serious global healthcare threat. Polymyxins are a class of antibiotics that exhibit potent antibacterial activities against Gram-negative bacteria. However, the use of polymyxins as an antibiotic has been limited due to the associated toxicity and adverse effects (e.g., nephrotoxicity). Moreover, mcr- 1, a plasmid-borne gene conferring bacterial resistance to polymyxins, has a high potential for dissemination and further threatens the efficacy of this class of antibiotics.
Because of the shortcomings of existing antibacterial treatments, combined with the emergence of multidrug-resistant Gram-negative bacteria, there is a need in the art for improved antibacterial therapies having greater efficacy, bioavailability, and reduced toxicity. Summary
The disclosure relates to compounds, compositions, and methods for inhibiting bacterial growth (e.g., Gram-negative bacterial growth) and for the treatment of bacterial infections (e.g., Gram-negative bacterial infections). In particular, such compounds contain dimers of cyclic heptapeptides conjugated to one or more monosaccharide or oligosaccharide moieties, which interact, directly or indirectly, with an immune cell. In certain embodiments, compounds containing dimers of cyclic heptapeptides may be conjugated to at least 2 (e.g., 2-1 0, 2-9, 2-8, 2-7, 2-6, 2-5, 2-4, or 2-3) monosaccharide or oligosaccharide moieties (e.g., rhamnose). In certain embodiments, compounds containing dimers of cyclic
heptapeptides may be conjugated to at least 3 (e.g., 3-10, 3-9, 3-8, 3-7, 3-6, 3-5, or 3-4) monosaccharide or oligosaccharide moieties (e.g., rhamnose). In yet other embodiments, compounds containing dimers of cyclic heptapeptides may be conjugated to at least 4 (e.g., 4-10, 4-9, 4-8, 4-7, 4-6, or 4-5)
monosaccharide or oligosaccharide moieties (e.g., rhamnose). In particular embodiments, compounds containing dimers of cyclic heptapeptides may be conjugated to two monosaccharide or oligosaccharide moieties (e.g., rhamnose). Cyclic heptapeptides bind to lipopolysaccharides (LPS) in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics.
In one aspect, the disclosure features a compound described by formula (I):
(E)n
M2 L' M1
(I)
wherein M1 includes a first cyclic heptapeptide including a linking nitrogen and M2 includes a second cyclic heptapeptide including a linking nitrogen; each E is, independently, a monosaccharide or oligosaccharide moiety; L' is a linker covalently attached to E and to the linking nitrogen in each of M1 and M2; and n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
In some embodiments, L' in formula (I) is described by formula (L):
Figure imgf000003_0001
(L)
wherein L is a remainder of L'; A1 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M1 or is absent; and A2 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M2 or is absent.
In some embodiments, the compound is described by formula (II):
M2 A2 A1 M1
Figure imgf000003_0002
(II)
wherein L is a remainder of L'; n is 1 , 2, 3, or 4 (e.g., 1 or 2) ; each of R1 , R12, R'1 , and R'12 is, independently, a lipophilic moiety, a polar moiety, or H; each of R1 1 , R13, R14, R'1 1 , R'13, and R'14 is, independently, optionally substituted C1 -C5 alkamino, a polar moiety, a positively charged moiety, or H; each of R15 and R'15 is, independently, a lipophilic moiety or a polar moiety; each of R2, R3, R4, R5, R6, R7, R8, R9, R10, R'2, R'3, R'4, R'5, R'6, R'7, R'8, R'9, and R'10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20
heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or two R groups on the same or adjacent atoms join to form an optionally substituted 5-8 membered ring; each of a', b', c', a, b, and c is, independently, 0 or 1 ; each of N1 , N2, N3, N4, N'1 , N'2, N'3, and N'4 is a nitrogen atom ; each of C1 , C2, C3, C'1 , C'2, and C'3 is a carbon atom, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound includes at least one optionally substituted 5-8 membered ring formed by joining (i) R2, R3, and C1 ; (ii) R3, R4, N1 , and C1 ; (iii) R5, R6, and C2; (iv) R6, R7, N2, and C2; (v) R8, R9, and C3; (vi) R9, R10, N3, and C3; (vii) R'2, R'3, and C'1 ; (viii) R'3, R'4, N'1 , and C'1 ; (ix) R'5, R'6, and C'2; (x) R'6, R'7, N'2, and C'2; (xi) R'8, R'9, and C'3; or (xii) R'9, R'10, N'3, and C'3. 1 some embodiments, the compound is described by formula
Figure imgf000004_0001
(III)
or a pharmaceutically acceptable salt thereof.
In some embodiments, each of R1 , R12, R'1 , and R'12 is, independently, a lipophilic moiety; each of R1 1 , R13, R14, R'1 1 , R'13, and R'14 is, independently, optionally substituted C1 -C5 alkamino, a polar moiety, or a positively charged moiety; and/or each of R15 and R'15 is, independently, a polar moiety.
In some embodiments, each of R1 and R12 is a lipophilic moiety. In some embodiments, each of R'1 and R'12 is a lipophilic moiety. In some embodiments, each lipophilic moiety is, independently, optionally substituted C1 -C20 alkyi, optionally substituted C5-C15 aryl, optionally substituted C6-C35 alkaryl, or optionally C5-C1 0 substituted heteroaryl. In some embodiments, each lipophilic moiety is, independently, C1 -C8 alkyi, methyl substituted C2-C4 alkyi, (C1 -C10)alkylene(C6)aryl, phenyl substituted (C1 -C10)alkylene(C6)aryl, or alkyi substituted C4-C9 heteroaryl. In some embodiments, each lipophilic moiety is, independently, benzyl, isobutyl, sec-butyl, isopropyl, n-propyl, methyl, biphenylmethyl, n-octyl, or methyl substituted indolyl.
In some embodiments, each of R1 1 , R13, R14, R'1 1 , R'13, and R'14 is independently optionally substituted C1 -C5 alkamino. In some embodiments, each of R1 1 , R13, R14, R'1 1 , R'13, and R'14 is
In some embodiments, each of R15 and R'15 is a polar moiety. In some embodiments, each polar moiety includes a hydroxyl group, a carboxylic acid group, an ester group, or an amide group. In some embodiments, each polar moiety is hydroxyl substituted C1 -C4 alkyi. In some embodiments, each polar moiety is CH(CH3)OH.
In some embodiments, the compound is described by formula (IV):
Figure imgf000004_0002
(IV) wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl, or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (IV-1 ):
Figure imgf000005_0001
(IV-1 )
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl, or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (IV-2):
Figure imgf000005_0002
(IV-2)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl, or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (V):
Figure imgf000005_0003
(V) wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R6, R'2, and R'6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (V-1 ):
Figure imgf000006_0001
(V-1 )
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (V-2):
Figure imgf000006_0002
(V-2)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (V-3):
Figure imgf000007_0001
(V-3)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (V-4):
Figure imgf000007_0002
(V-4)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (V-5):
Figure imgf000007_0003
(V-5)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof. In some embodiments, the compound is described by formula (VI):
Figure imgf000008_0001
(VI)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R6, R8, R'2, R'6, and R'8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (VI-1 ):
Figure imgf000008_0002
(VI-1 )
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (VI-2):
Figure imgf000009_0001
(VI-2)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (VI-3):
Figure imgf000009_0002
(VI-3)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (VI-4):
Figure imgf000009_0003
(VI-4)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof. In some embodiments, the compound is described by formula (VI-5):
Figure imgf000010_0001
(VI-5)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (VII):
Figure imgf000010_0002
(VII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R6, R8, R'2, and R'6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 - C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (VIII):
Figure imgf000011_0001
(VIII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R6, R8, and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (IX):
Figure imgf000011_0002
(IX)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R6, and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (IX-1 ):
Figure imgf000012_0001
(IX-1 )
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (IX-2):
Figure imgf000012_0002
(IX-2)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (X):
Figure imgf000012_0003
(X)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2 and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XI):
Figure imgf000013_0001
(XI)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2 and R6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XI-1 ):
Figure imgf000013_0002
(XI-1 ) wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XI-2):
Figure imgf000014_0001
(XI-2)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XII):
Figure imgf000014_0002
(XII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2 and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; R6, R7, N2, and C2 together form an optionally substituted 5-8 membered ring including optionally substituted C3-C7 heterocycloalkyl including an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl including an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S; R'6, R'7, N'2, and C'2 together form an optionally substituted 5-8 membered ring including optionally substituted C3-C7 heterocycloalkyi including an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl including an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S; or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XII-1 ):
Figure imgf000015_0001
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound described by formula (XIII):
E' E
L'1 L1
I I
M2— A2— L A1 M1
(XIII)
wherein A1 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M1 ; A2 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M2; E1 is a first monosaccharide or oligosaccharide moiety; E'1 is a second monosaccharide or oligosaccharide moiety; L, L1 , and L'1 are remainders of L'; or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is described by formula (XIII-1 ) :
Figure imgf000016_0001
(XIII-1 )
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g CH2CH(CH3)2), or cyclohexylmethyl; each of R6 and R'6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each of R2 and R'2 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2- C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XIII-2):
Figure imgf000016_0002
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XIII-3):
Figure imgf000017_0001
(XIII-3)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; or a
pharmaceutically acceptable salt thereof.
In some embodiments, the compound described by formula (XIV):
E
L
M2— A2— L A1 M1
(XIV)
wherein A1 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M1 ; A2 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M2, or is absent; L and L1 are remainders of L'; or a pharmaceutically acceptable salt thereof.
In some embodiments of the compounds described herein, R2 is optionally substituted C1 -C5 alkamino. In some embodiments of the compounds described herein, R'2 is optionally substituted C1 -C5 alkamino. In some embodiments, the optionally substituted C1 -C5 alkamino is CH2NH2 or CH2CH2NH2.
In some embodiments of the compounds described herein, R2 is a polar moiety. In some embodiments of the compounds described herein, R'2 is a polar moiety. In some embodiments of the compounds described herein, R6 is a polar moiety. In some embodiments of the compounds described herein, R'6 is a polar moiety. In some embodiments, the polar moiety includes a hydroxyl group, a carboxylic acid group, an ester group, or an amide group. In some embodiments, the polar moiety is hydroxyl substituted C1 -C4 alkyl. In some embodiments, the polar moiety is CH(CH3)OH or CH2OH.
In some embodiments of the compounds described herein, R8 is optionally substituted C1 -C5 alkamino. In some embodiments of the compounds described herein, R'8 is optionally substituted C1 -C5 alkamino. In some embodiments, the optionally substituted C1 -C5 alkamino is CH2NH2 or CH2CH2NH2. In some embodiments of the compounds described herein, R8 is optionally substituted C5-C1 5 aryl. In some embodiments of the compounds described herein, R'8 is optionally substituted C5-C15 aryl. In some embodiments, the optionally substituted C5-C15 aryl is naphthyl.
In some embodiments of the compounds described herein, R6, R7, N2, and C2 together form a 5- or 6-membered ring including C4-C5 heterocycloalkyl including an N heteroatom and additional 0 or 1 heteroatom independently selected from N, O, and S; and wherein R'6, R'7, N'2, and C'2 together form a 5- or 6-membered ring including C4-C5 heterocycloalkyl including an N heteroatom and additional 0 or 1 heteroatom independently selected from N, O, and S. In some embodiments, each of R2 and R'2 is C1 - C4 heteroalkylene. In some embodiments, each of R2 and R'2 is -(CH2)2NH- or -CH2NH-.
In some embodiments, the compound is described by formula (XV):
Figure imgf000018_0001
(XV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl, or a
pharmaceutically acceptable salt thereof.
In some embodiments of the compounds described herein, L', L, L1 , or L'1 includes one or more optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C1 5 arylene, optionally substituted C2-C15 heteroarylene, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino, wherein R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C1 5 heteroaryl.
In some embodiments of the compounds described herein, the backbone of L', L, L1 , or L'1 consists of one or more optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20
heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20
heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20
heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15
heteroarylene, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino, wherein R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyi , optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl , optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl , optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl , optionally substituted C8-C20 heterocycloalkynyl , optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl .
In some embodiments of the compounds described herein, L', L, L1 , or L'1 is oxo substituted. In some embodiments of the compounds described herein, the backbone of L', L, L1 , or L'1 includes no more than 250 atoms.
In some embodiments of the compounds described herein, L', L, L1 , or L'1 is capable of forming an amide, a carbamate, a sulfonyl, or a urea linkage.
In some embodiments of the compounds described herein, L, L1 , or L'1 is a bond.
In some embodiments of the compounds described herein, L is described by formula (L-l) :
Lc
LB-Q-LA
(L-l)
wherein LA is described by formula GA1-(ZA1)gi-(YA1)hi-(ZA2)ii-(YA2)ji -(ZA3)ki -(YA3)ii -(ZA4)mi -(YA4)ni-(ZA5)oi - GA2; LB is described by formula GB1-(ZB1)g2-(YB1
Figure imgf000019_0001
Lc is described by
Figure imgf000019_0002
GA1 is a bond attached to Q in formula (L-l) ; GA2 is a bond attached to A1 or M1 if A1 is absent; GB1 is a bond attached to Q in formula (L-l) ; GB2 is a bond attached to A2 or M2 if A2 is absent; GC1 is a bond attached to Q in formula (L-l) ; GC2 is a bond attached to E; each of ZM , ZA2, ZA3, ZM, ZA5, ZB1 , ZB2, ZB3, ZB4, ZB5, ZC1 , ZC2, ZC3, ZC4, and ZC5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 - C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20
heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20
heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20
heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of YA1 , YA2, YA3, YA4, YB1 , YB2, YB3, YB4, YC1 , YC2, YC3, and YC4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl , phosphate, phosphoryl, or imino; R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl , optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C1 5 heteroaryl; each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , o1 , g2, h2, i2, j2, k2, I2, m2, n2, o2, g3, h3, i3, j3, k3, I3, m3, n3, and o3 is, independently, 0 or 1 ; Q is a nitrogen atom, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C1 5 arylene, or optionally substituted C2-C15 heteroarylene.
In some embodiments of the compounds described herein, L is
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
wherein R* is a bond or includes one or more of optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2- C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15 heteroarylene, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, and imino, and wherein R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C2- C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl.
In some embodiments of the compounds described herein, L is described by formula (L-lla) or formula (L-llb):
Figure imgf000022_0002
(L-lla) (L-llb)
wherein LA is described by formula GA1-(ZA1)gi-(YA1)hi-(ZA2)ii-(YA2)ji -(ZA3)ki -(YA3)ii -(ZA4)mi -
(YA4)m-(ZA5)oi-GA2; LB is described by formula GB1-(ZB1 )g2-(YB1 )h2-(ZB2)i2-(YB2)j2-(ZB3)k2-(YB3)i2-(ZB4)m2-
(YB4)n2-(ZB5)o2-GB2; Lc is described by formula Gc1-(Zc1 )g3-(Yc1 )h3-(ZC2)i3-(YC2)j3-(ZC3)k3-(YC3)i3-(ZC4)m3- (YC4)n3-(ZC5)o3-GC2; LD is described by formula GD1 -(ZD1 )g4-(YD1 )h4-(ZD2)i4-(YD2)j4-(ZD3)k4-(YD3)i4-(ZD4)m4-
(YD4)n4-(ZD5)o4-GD2; GA1 is a bond attached to NA in formula (L-lla) or NA in formula (L-llb); GA2 is a bond attached to A1 or M1 if A1 is absent; GB1 is a bond attached to NA in formula (L-lla) or NA in formula (L- llb); GB2 is a bond attached to A2 or M2 if A2 is absent; GC1 is a bond attached to NB in formula (L-lla) or C in formula (L-llb); GC2 is a bond attached to a first monosaccharide or oligosaccharide moiety, E1 ; GD1 is a bond attached to NB in formula (L-lla) or C in formula (L-llb); GD2 is a bond attached to a second monosaccharide or oligosaccharide moiety, E'1 ; each of ZA1 , ZA2, ZA3, ZA4, ZA5, ZB1 , ZB2, ZB3, ZB4, ZB5, ZC1 , ZC2, ZC3, ZC4, ZC5, ZD1 , ZD2, ZD3, ZD4, and ZD5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of YA1 , YA2, YA3, YA4, YB1 , YB2, YB3, YB4, YC1 , YC2, YC3, YC4, YD1 , YD2, YD3, and YD4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C2- C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; and each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , o1 , g2, h2, i2, j2, k2, I2, m2, n2, o2, g3, h3, i3, j3, k3, I3, m3, n3, o3, g4, h4, i4, j4, k4, I4, m4, n4, and o4 is, independently, 0 or 1 .
In some embodiments of the compounds described herein, L is
Figure imgf000023_0001
In some embodiments of the compounds described herein (e.g., compound of any one of formulas (XIII) and (XIV)), L is described by formula (L-lll):
HA1 -(WA1 )gi -(XA1 )hi -(WA2)ii -(XA2)ji -(WA3)ki -(XA3)ii -(WA )mi -(XA4)m -(WA5)01 -HA2
(L-lll) wherein HA1 is a bond attached to A2; HA2 is a bond attached to A1 ; each of WA1 , WA2, WA3, WM , and WA5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C1 5 arylene, or optionally substituted C2-C15 heteroarylene; each of XA1 , XA2, XA3, XM is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C1 5 heteroaryl; and each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , and o1 is, independently, 0 or 1 . In some embodiments, L is
Figure imgf000024_0001
In some embodiments of the compounds described herein (e.g., compound of any one of formulas (XIII) and (XIV)), L1 is described by formula (L-IV):
HB1-(WB1 )g2-(XB1 )h2-(WB2)i2-(XB2)j2-(WB3)k2-(XB3)l2-(WB4)m2-(XB4)n2-(WB5)o2-HB2
(L-IV)
wherein HB1 is a bond attached to A1 ; HB2 is a bond attached to a first monosaccharide or oligosaccharide moiety, E1 ; each of WB1 , WB2, WB3, WB4, and WB5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4- C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of XB1 , XB2, XB3, XB4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 - C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20
cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; and each of g2, h2, i2, j2, k2, I2, m2, n2, and o2 is,
independently, 0 or 1 . In some embodiments, L1 is
Figure imgf000025_0001
In some embodiments of the compounds described herein (e.g., compound of any one of formulas (XIII) and (XIV)), L'1 is described by formula (L-V):
HC1 -(WC1 )g3-(XC1 )h3-(WC2)i3-(XC2)j3-(WC3)k3-(XC3)l3-(WC4)m3-(XC4)n3-(WC5)o3HC2
(L-V)
wherein HC1 is a bond attached to A2; HC2 is a bond attached to a second monosaccharide or oligosaccharide moiety, E'1 ; each of WC1 , WC2, WC3, WC4, and WC5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of XC1 , XC2, XC3, XC4 is, independently, O, S, NR\ P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C1 5 heteroaryl ; and each of g3, h3, i3, j3, k3, I3, m3, n3, and o3 is, independently, 0 or 1 . In some embodiments, L'1 is
Figure imgf000025_0002
In some embodiments of the compounds described herein, E, E1 , or E'1 is
Figure imgf000025_0003
In some embodiments, E, E1 , or E'1 is
Figure imgf000026_0001
In some embodiments, E, E1 , or E'1 is any one of the moieties in Tables 2A and 2B.
In some embodiments, E, E1 , or E'1 directly or indirectly activates an immune cell.
In some embodiments, E, E1 , or E'1 is a ligand to an innate immune receptor. In some embodiments, the innate immune receptor is AICL, BDCA2, CLEC2, Complement receptor 3,
Complement receptor 4, DCIR, dectin-1 , dectin-2, DC-SIGN, a C-Type lectin receptor, MMR, langerin, TLR2, Mincle, MBL, or KCR.
In some embodiments, E, E1 , or E'1 binds to an antibody. In some embodiments, the antibody is a natural antibody. In some embodiments, the natural antibody is an antibody of the immunoglobulin M (IgM) isotype. In some embodiments, the antibody binds to a moiety in Tables 2A and 2B. In some embodiments, the antibody is anti-aGal antibody or anti-aRha antibody.
In some embodiments, the monosaccharide moiety has one optionally substituted C6-C9 monosaccharide residue.
In some embodiments, the oligosaccharide moiety has 2-18 optionally substituted C6-C9 monosaccharide residues. In some embodiments, the oligosaccharide moiety has 2-12 optionally substituted C6-C9 monosaccharide residues. In some embodiments, each of the optionally substituted C6-C9 monosaccharide residues is, independently, glucose (Glc), galactose (Gal), mannose (Man), allose (All), altrose (alt), gulose (Gul), idose (ido), talose (Tal), fucose (Fuc), rhamnose (Rha or L-Rha), thia-rhamnose (thia-Rha or thia-L-Rha), quinovose (Qui), 2-deoxyglucose (2-dGlc), glucosamine (GlcN), galactosamine (GaIN), mannosamine (ManN), fucosamine (FucN), quinovosamine (QuiN), N-Acetyl- glucosamine (GlcNAc), N-Acetyl-galactosamine (GalNAc), N-Acetyl-mannosamine (ManNAc), N-acetyl- fucosamine (FucNAc), N-acetyl-quinovosamine (QuiNAc), glucuronic acid (GIcA), galacturonic acid (GalA), mannuronic acid (ManA), iduronic acid (IdoA), sialic acid (Sia), neuraminic acid (Neu), N-Acetyl- neuraminic acid (Neu5Ac), N-Glycolyl-neuraminic acid (Neu5Gc), glucitol (Glc-ol), galactitol (Gal-ol), mannitol (Man-ol), fructose (Fru), sorbose (Sor), tagatose (Tag), thevetose (The), acofriose (Aco), digitoxose (Dig), cymarose (Cym), abequose (Abe), colitose (Col), tyvelose (Tyv), ascarylose (Asc), paratose (Par), or N-acetyl-muramic acid (MurNAc).
In some embodiments, each of the optionally substituted C6-C9 monosaccharide residues is, independently, an optionally substituted C6 monosaccharide residue.
In some embodiments, the optionally substituted C6 monosaccharide residue is Glc, Gal, Man, All, Alt, Gul, Ido, or Tal. In some embodiments, the optionally substituted C6 monosaccharide residue is Fuc, Rha or L-Rha, thia-Rha or thia-L-Rha, Qui, or 2-dGlc. In some embodiments, the optionally substituted C6 monosaccharide residue is GlcN, GaIN, ManN, FucN, or QuiN. In some embodiments, the optionally substituted C6 monosaccharide residue is N- GlcNAc, GalNAc, ManNAc, FucNAc, QuiNAc, GIcA, GalA, ManA, or IdoA. In some embodiments, the optionally substituted C6 monosaccharide residue is Glc-ol, Gal-ol, or Man-ol. In some embodiments, the optionally substituted C6 monosaccharide residue is Fru, Sor, Tag. In some embodiments, the optionally substituted C6 monosaccharide residue is The, Aco, Dig, Cym, Abe, Col, Tyv, Asc, Par, or MurNAc. In some embodiments, the optionally substituted C6 monosaccharide residue is Rha, Gal, Glc, GlcA, GlcNAc, GalNAc, GlcN(Gc) (N-Glycolyl-Glucosamine), Neu5Ac, Neu5Gc, Fuc, Man, -hkPCbMan (mannose phosphate), 6-H2PO3GIC (glucose phosphate), Mur (muramoyl), Mur-L-Ala-D-i-Gln-Lys, (Mur)-3-0-GlcNAc, sulfate-galactose (Su-Gal), disulfate-galactose (Su2-Gal), sulfate-glucose (Su-Glc), sulfate-GlcNAc (Su-GlcNAc), or sulfate-GalNAc (Su-GalNAc).
In some embodiments, the optionally substituted C6 monosaccharide residue is optionally substituted Rha. In some embodiments, the optionally substituted C6 monosaccharide residue is Rha.
In some embodiments, the optionally substituted C6 monosaccharide residue is L-Rha.
In some embodiments, the optionally substituted C6 monosaccharide residue is optionally substituted Gal or optionally substituted Glc. In some embodiments, the optionally substituted Gal is optionally substituted a1 -3Gal. In some embodiments, the optionally substituted Glc is an optionally substituted β-glucan having 1 -6 Glc moieties.
In some embodiments, the optionally substituted β-glucan is
Figure imgf000027_0001
pustulan.
In some embodiments, the optionally substituted β-glucan is laminarin.
In some embodiments, each of the optionally substituted C6-C9 monosaccharide residues is, independently, an optionally substituted C9 monosaccharide residue, wherein the optionally substituted C9 monosaccharide residue is Sia, Neu, Neu5Ac, or Neu5Gc.
In some embodiments, at least one optionally substituted C6-C9 monosaccharide residue is substituted with 1 -3 substituents independently selected from sulfate, phosphate, methyl, acetyl, natural amino acids, and non-natural amino acids. In some embodiments, at least one optionally substituted C6- C9 monosaccharide residue is substituted with 1 -3 substituents independently selected from sulfate, phosphate, methyl, and acetyl. In some embodiments, at least one optionally substituted C6-C9 monosaccharide moiety is substituted with 1 -3 substituents independently selected from natural and non- natural amino acids. In some embodiments, the natural amino acid is alanine, lysine, serine, glutamine or asparagine.
In another aspect, the disclosure features a compound of formula (XVI):
Figure imgf000028_0001
(XVI)
wherein each A is an independently selected amino acid; L is a linker that, when m is 2, 3, 4, or 5, is bound to any of A; each E is independently selected from a monosaccharide or an oligosaccharide; m is 0, 1 , 2, 3, 4, or 5; n is 1 , 2, 3, 4, or 5; and Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVI), Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVI), at least one of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least two of Q1 , Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least three of Q1 , Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least four of Q1 , Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least five of Q1 , Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVI), each Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVI), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVI), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000029_0001
Figure imgf000030_0001
or a pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XVII):
Figure imgf000030_0002
(XVII)
wherein each A1 and A2 is an independently selected amino acid; L is a linker that, when m is 1 , 2, 3, 4, or 5, is bound to a nitrogen atom in any A1 and a nitrogen atom in any A2; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; n is 1 , 2, 3, 4, or 5; and Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVII), Q1 , Q2, Q3, Q4, Q5 and Q6 are each
independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVII), at least one (e.g., at least two, three, four, or five) of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVII), each Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVII), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyl, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVII), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof. In some embodiments of a compound of formula (XVII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000031_0001
In some embodiments of a compound of formula (XVII), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5- methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; and each m is independently selected from 1 , 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVII), each m is independently 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVII), n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000032_0003
Figure imgf000032_0001
In some embodiments of a compound of formula (XVII), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5- methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; each m is independently 1 , 2, 3 or 4; E is a monosaccharide; n is 1 , 2, 3, or 4; and the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000032_0004
Figure imgf000032_0002
In another aspect, the disclosure features a compound of formula (XVIII):
Figure imgf000033_0001
(XVIII)
wherein each A1 and A2 is an independently selected amino acid; X is absent or is -CH2CH2C(0)NH-; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; n is 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 10; and e is an integer from 0 to 10; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVIII), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVIII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000033_0002
Figure imgf000034_0001
or a p armaceut ca y accepta e sa t t ereo .
In some embodiments of a compound of formula (XVIII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000034_0003
Figure imgf000034_0002
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVIII), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2- amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2 or 3; d is an integer from 1 to 10; e is an integer from 1 to 10; and E is a monosaccharide; or a pharmaceutically acceptable salt thereof. In some embodiments of a compound of formula (XVIII), each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2 or 3; d is an integer from 1 to 10; e is an integer from 1 to 10; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof. In some embodiments, m is 2. In some embodiments, m is 3.
In some embodiments of a compound of formula (XVIII), d is 1 0; e is 10; and X is absent; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVIII), m is 2; d is 1 ; e is 1 ; and X is - CH2CH2C(0)NH-; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XVIII), E is
pharmaceutically acceptable salt thereof. In some embodiments, E is
Figure imgf000035_0001
, or a
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XIX):
Figure imgf000035_0002
(XIX)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; n is 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 10; e is an integer from 0 to 10; f is an integer from 0 to 10; and g is an integer from 0 to 10; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XIX), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XIX), the combination of Q1 , Q2, Q3, Q4, Q5 and
Q6 is selected from one of
Figure imgf000036_0001
or a p armaceutca y accepta e sat t ereo . In some embodiments of a compound of formula (XIX), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000037_0004
Figure imgf000037_0001
or a p armaceut ca y accepta e sa t t ereo .
In some embodiments of a compound of formula (XIX), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5- methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2; d is 1 ; e is 1 ; f is 3; g is 1 ; and E is a monosaccharide; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XIX), each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2; d is 1 ; e is 1 ; f is 3; g is 1 ; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XIX), E is
pharmaceutically acceptable salt thereof. In some embodiments, E i
Figure imgf000037_0002
s or a
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XX):
Figure imgf000037_0003
(XX) wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 1 0; e is an integer from 0 to 10; f is an integer from 0 to 10; and g is an integer from 0 to 10; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XX), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XX), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000038_0001
Figure imgf000039_0001
or a p armaceut ca y accepta e sa t t ereo .
In some embodiments of a compound of formula (XX), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000039_0003
Figure imgf000039_0002
In some embodiments of a compound of formula (XX), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5- methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2; d is 1 ; e is 1 ; f is 1 ; g is 1 ; and E is a monosaccharide; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XX), each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2; d is 1 ; e is 1 ; f is 1 ; g is 1 ; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof. In some embodiments of a compound of formula (XX), E is
pharmaceutically acceptable salt thereof. In some embodiments, E
Figure imgf000040_0001
is or a
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula
Figure imgf000040_0002
(XXI)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 1 0; e is an integer from 0 to 10; f is an integer from 0 to 10; g is an integer from 0 to 25; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXI), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXI), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000040_0003
Figure imgf000041_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXI), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000041_0003
Figure imgf000041_0002
or a pharmaceutically acceptable salt thereof. In some embodiments of a compound of formula (XXI), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5- methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2; d is 1 ; e is 1 ; f is 1 ; g is 8 to 25; and E is a monosaccharide; or a
pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXI), each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2; d is 1 ; e is 1 ; f is 1 ; g is 8 to 25; E is a
monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXI), E is
pharmaceutically acceptable salt thereof. In some embodiments, E is
Figure imgf000042_0001
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XXII):
Figure imgf000042_0002
(XXII)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and d is an integer from 0 to 15; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXII), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000043_0001
In some embodiments of a compound of formula (XXII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000044_0003
Figure imgf000044_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXII), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5- methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2; d is 10; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXII), each A1 and A2 is independently selected from 2,4-diaminobutyric acid and piperazine-2-carboxylic acid; m is 2; d is 10; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXII), E is
pharmaceutically acceptable salt thereof. In some embodiments, E is
Figure imgf000044_0002
pharmaceutically acceptable salt thereof. In another aspect, the disclosure features a compound of formula (XXIII):
Figure imgf000045_0001
(XXIII)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from
a monosaccharide or an oligosaccharide; Y is
Figure imgf000045_0002
, -C(0)CH2CH2-, -CH2-, or is absent; X is -C(0)CH2CH2- or is absent; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 15; e is an integer from 1 to 10; f is an integer from 1 to 5; and g is an integer from 1 to 5; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIII), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000045_0003
Figure imgf000046_0001
or a p armaceut ca y accepta e sa t t ereo .
In some embodiments of a compound of formula (XXIII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000046_0003
Figure imgf000046_0002
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIII), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2- -5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alph
neopentylglycine; m is 2, 3 or 4; d is 1 ; when Y is
Figure imgf000047_0001
, e is 4; E is a monosaccharide; f is 1 or 2; g is 1 or 2; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIII), each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 3-aminoalanine, 2-piperazinecarboxylic acid, 2-aminohexanoic
acid, 2-aminooctanoic acid, methionine, and threonine; m is 2, 3 or 4; when Y is
Figure imgf000047_0002
, e is 4; d is 1 ; E is a monosaccharide; f is 1 or 2; g is 1 or 2; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIII), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2- amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 3 or 4; d is 1 ; Y is absent; E is a monosaccharide; f is 1 ; g is 1 ; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIII), each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine; and m is 3; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIII), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2- amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2, 3 or 4; d is 1 ; Y is -C(0)CH2CH2-; E is a monosaccharide; f is 1 or 2; g is 1 or 2; and n is 1 ; or a pharmaceutically acceptable salt thereof. In some embodiments, each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 2,4- diaminobutyric acid, 3-aminoalanine, 2-aminohexanoic acid, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine, or a pharmaceutically acceptable salt thereof. In some embodiments, m is 4, d is 1 , f is 1 , and g is 1 , or a pharmaceutically acceptable salt thereof. In some embodiments, m is 3, f is 2, and g is 1 , or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIII), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5- methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2, 3 or 4; d is 1 ; Y is -CH2-; E is a monosaccharide; f is 1 or 2; g is 1 or 2; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments, each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2- aminooctanoic acid and threonine; f is 1 ; and g is 1 ; or a pharmaceutically acceptable salt thereof. In some embodiments, each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2- aminohexanoic acid, 2-aminooctanoic acid and threonine, or a pharmaceutically acceptable salt thereof; m is 4; f is 1 ; and g is 1 ; or a pharmaceutically acceptable salt thereof. In some embodiments, each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminohexanoic acid, 2-aminooctanoic acid and threonine, or a pharmaceutically acceptable salt thereof; m is 3; f is 1 ; and g is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIII), E is ; or a
pharmaceutically acceptable salt thereof. In some embodiments, E is
Figure imgf000048_0001
, or a
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XXIV):
Figure imgf000049_0001
(XXIV)
wherein each A1 and A2 is an independently selected amino acid; X is -C(0)CH2CH2CH2-Y- or - C(0)CH2CH2C(0)NH-; Y is heteroaryl; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and d is an integer from 0 to 15; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIV), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIV), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000049_0002
Figure imgf000050_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIV), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000050_0002
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIV), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2- amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2 or 3; d is an integer from 1 to 3; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIV), each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 3-(2-naphthyl)alanine, and threonine, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIV), X is -C(0)CH2CH2CH2-Y-; m is 3; d is 3; and Y is 1 ,4-triazololyl; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIV), X is -C(0)CH2CH2C(0)-; m is 2; and d is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIV), E is
pharmaceutically acceptable salt thereof. In some embodiments, E is
Figure imgf000051_0001
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XXV):
Figure imgf000051_0002
(XXV)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 1 5; and e is an integer from 0 to 1 5; or a
pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXV), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof. In some embodiments of a compound of formula (XXV), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000052_0001
In some embodiments of a compound of formula (XXV), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000053_0001
or a p armaceut ca y accepta e sa t t ereo .
In some embodiments of a compound of formula (XXV), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5- methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2 or 3; d is an integer from 1 to 3; e is an integer from 1 to 3; E is a
monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXV), each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2 or 3; d is an integer from 1 to 3; e is an integer from 1 to 3; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXV), E is or a
pharmaceutically acceptable salt thereof. In some embodiments, E i
Figure imgf000053_0002
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XXVI):
Figure imgf000053_0003
(XXVI) wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; R is C1 -C20 alkyl; each m is independently 0, 1 , 2, 3, 4, or 5;each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 20; e is an integer from 0 to 20; and f is an integer from 0 to 20; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXVI), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXVI), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000054_0001
Figure imgf000055_0001
or a p armaceut ca y accepta e sa t t ereo .
In some embodiments of a compound of formula (XXVI), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000055_0003
Figure imgf000055_0002
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXVI), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2- amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2 or 3; d is an integer from 1 to 3; e is an integer from 1 to 3; f is an integer from 1 to 3; E is a monosaccharide; R is C1 -C1 0 alkyl ; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXVI), each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2; d is 1 ; e is 1 ; f is 1 ; E is a monosaccharide; R is C1 -C1 0 alkyl ; and n is 1 ; or a pharmaceutically acceptable salt thereof. In some embodiments of a compound of formula (XXVI), E is or a
pharmaceutically acceptable salt thereof. In some embodiments, E is
Figure imgf000056_0001
or a
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XXVII)
Figure imgf000056_0002
(XXVII)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; R is C1 -C20 alkyl; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 1 to 20; e is an integer from 1 to 20; and f is an integer from 1 to 20; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXVII), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXVII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000056_0003
Figure imgf000057_0001
or a p armaceutca y accepta e sat t ereo .
In some embodiments of a compound of formula (XXVII), the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000057_0002
Figure imgf000058_0001
or a p armaceut ca y accepta e sa t t ereo .
In some embodiments of a compound of formula (XXVII), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2- amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2, 3, or 4; d is an integer from 1 to 3; e is an integer from 1 to 3; f is an integer from 1 to 3; E is a monosaccharide; R is C1 -C10 alkyl; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXVII), each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; m is 2, 3, or 4; d is 1 ; e is 1 ; f is 1 ; E is a monosaccharide; R is C1 -C10 alkyl; and n is 1 ; or a pharmaceutically acceptable salt thereof. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, m is 4.
In some embodiments of a compound of formula (XXVII), E is
pharmaceutically acceptable salt thereof. In some embodiments, E is
Figure imgf000058_0002
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XXVIII):
Figure imgf000058_0003
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from
a monosaccharide or an oligosaccharide; Y is
Figure imgf000059_0001
-C(0)CH2CH2-, -CH2-, or is absent; e is an integer from 1 to 10; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and d is an integer from 0 to 15; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXVIII), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXVIII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000059_0002
Figure imgf000060_0001
or a p armaceut ca y accepta e sa t t ereo .
In some embodiments of a compound of formula (XXVIII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000060_0003
Figure imgf000060_0002
or a p armaceut ca y accepta e sa t t ereo .
In some embodiments of a compound of formula (XXVIII), Y is -C(0)CH2CH2-, or a
pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXVIII), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2- amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2, 3, or 4; d is an integer from 1 to 3; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXVIII), each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and d is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXVIII), m is 2. In some embodiments of a compound of formula (XXVIII), m is 3. In some embodiments of a compound of formula (XXVIII), m is 4. In some embodiments of a compound of formula (XXVIII), E is
pharmaceutically acceptable salt thereof. In some embodiments, E is
Figure imgf000061_0001
or a
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XXIX):
Figure imgf000061_0002
(XXIX)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; X is -CH2- or -C(O)-; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and d is an integer from 0 to 15; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIX), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIX), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000061_0003
Figure imgf000062_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIX), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000062_0003
Figure imgf000062_0002
In some embodiments of a compound of formula (XXIX), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2- amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2, 3, or 4; d is an integer from 1 to 3; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof. In some embodiments of a compound of formula (XXIX), each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and d is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXIX), m is 2. In some embodiments of a compound of formula (XXIX), m is 3. In some embodiments of a compound of formula (XXIX), m is 4.
In some embodiments of a compound of formula (XXIX), E is
pharmaceutically acceptable salt thereof. In some embodiments, E is
Figure imgf000063_0001
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XXX):
Figure imgf000063_0002
(XXX)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from
a monosaccharide or an oligosaccharide; Y is
Figure imgf000063_0003
-C(0)CH2CH2-, -CH2-, or is absent; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 15; g is an integer from 0 to 15; and e and e' are each independently an integer from 1 to 3; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXX), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXX), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000064_0001
Figure imgf000065_0001
In some embodiments of a compound of formula (XXX), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000065_0003
Figure imgf000065_0002
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXX), Y is -C(0)CH2CH2-, or a
pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXX), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2- naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4- amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S- methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5- methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2, 3, or 4; d is an integer from 1 to 3; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXX), each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and d is 1 ; and e is 1 or 2; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXX), m is 2. In some embodiments of a compound of formula (XXX), m is 3. In some embodiments of a compound of formula (XXX), m is 4. pharmaceutically acceptable salt thereof. In some embodiments, E is
Figure imgf000066_0001
, or a pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XXXI) :
Figure imgf000066_0002
(XXXI)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from
a monosaccharide or an oligosaccharide; each Y is independently selected from
Figure imgf000066_0003
C(0)CH2CH2-, and -CH2-, or is absent; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 1 to 1 5; each e is independently from 1 to 1 5; each f is independently from 1 to 1 5; and g is from 1 to 1 5; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXI), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl , 2-butyl , methyl , and 2-aminoethyl , or a pharmaceutically acceptable salt thereof. In some embodiments of a compound of formula (XXXI), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000067_0001
In some embodiments of a compound of formula (XXXI), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000068_0001
In some embodiments of a compound of formula (XXXI), Y is -C(0)CH2CH2-, or a
pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXI), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2- amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2, 3, or 4; d is an integer from 1 to 5; e is an integer from 1 to 5; E is a
monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof. In some embodiments, each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and d is 4 or 5; e is 4 or 5; and f is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXI), m is 2. In some embodiments of a compound of formula (XXXI), m is 3. In some embodime m is 4.
In some embodiments of a compound of formula (XXXI),
pharmaceutically acceptable salt thereof. In some embod
Figure imgf000068_0002
iments, E is , or a
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XXXII):
Figure imgf000069_0001
(XXXII)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from
a monosaccharide or an oligosaccharide; each Y is independently selected from
Figure imgf000069_0002
C(0)CH2CH2-, -CH2CH2NHC(0)CH2CH2-, and -CH2-, or is absent; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 1 to 15; each e is independently from 1 to 15; and g is from 1 to 15; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXII), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000069_0003
Figure imgf000070_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXII), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000070_0003
Figure imgf000070_0002
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXII), Y is -CH2CH2NHC(0)CH2CH2-, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXII), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4- phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2- aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2- amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha- neopentylglycine; m is 2, 3, or 4; d is an integer from 1 to 5; e is an integer from 1 to 5; E is a
monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXII), each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and d is an integer from 1 to 4; and e is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXII), d is 2. In some embodiments of a compound of formula (XXXII), m is 2. In some embodiments, m is 3. In some embodiments, m is 4.
In some embodiments of a compound of formula (XXXII), E is
pharmaceutically acceptable salt thereof. In some embodiments, E is
Figure imgf000071_0001
pharmaceutically acceptable salt thereof.
In another aspect, the disclosure features a compound of formula (XXXIII):
Figure imgf000071_0002
(XXXIII)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; each X is independently selected from
Figure imgf000071_0003
, -C(0)CH2CH2C(0)-, -CH2CH2NHC(0)CH2CH2-, -C(O)-, and -CH2-, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 alkenyl, optionally substituted C1 -C20 alkynyl, optionally substituted C1 -C15 aryl, optionally substituted C1 -C15 heteroaryl, or is absent; each Y is independently selected from -CH-, or oxygen; Z is -NH-, -NHC(O)-, oxygen, or sulfur; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 1 to 15; g is an integer from 1 to 15; e and e' are independently from 1 to 10 and or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXIII), d is 2.
In some embodiments of a compound of formula (XXXIII), the compound is of the formula (XXXIII-1 ):
Figure imgf000072_0001
(XXXIII-1 )
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXIII), the compound is of the formula (XXXI 11-2):
Figure imgf000072_0002
(XXXIII-2)
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXIII) (e.g., a compound of formula (XXXIII-1 ) or a compound of formula (XXXIII-2)), each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2- methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl. hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXIII) (e.g., a compound of formula (XXXIII-1 ) or a compound of formula (XXXIII-2)), the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000072_0003
Figure imgf000073_0001
or a p armaceutca y accepta e sat t ereo .
In some embodiments of a compound of formula (XXXIII) (e.g., a compound of formula (XXXII 1- ) or a compound of formula (XXXIII-2)), the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000073_0002
Figure imgf000074_0001
or a p armaceut ca y accepta e sa t t ereo .
In some embodiments of a compound of formula (XXXIII) (e.g., a compound of formula (XXXII 1- ) or a compound of formula (XXXIII-2)), each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2- piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; m is 2, 3, or 4; d is an integer from 1 to 5; e is 1 ; X is -C(0)CH2CH2C(0)-; E is a monosaccharide; and n is 1 ; or a pharmaceutically acceptable salt thereof. In some embodiments, each A1 and A2 is independently selected from 2,4- diaminobutyric acid, 2-aminohexanoic acid, and threonine; and d is 1 ; or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXIII) (e.g., a compound of formula (XXXII 1- ) or a compound of formula (XXXIII-2)), m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments of a compo (e.g., a compound of formula (XXXII 1-1 ) or a
compound of formula (XXXIII-2)), or a pharmaceutically acceptable salt
In some embodiments, E
Figure imgf000074_0002
ically acceptable salt thereof.
In some embodiments, compound is described by formula (XXXIV)
Figure imgf000074_0003
(XXXIV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R3, R4, R5, R6, R7, R8, R9, R10, R'2, R'3, R'4, R'5, R'6, R'7, R'8, R'9, and R'10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or two R groups on the same or adjacent atoms join to form an optionally substituted 5-8 membered ring;
each of a', b', c', a, b, and c is, independently, 0 or 1 ;
each of N1 , N2, N3, N4, N'1 , N'2, N'3, and N'4 is a nitrogen atom;
each of C1 , C2, C3, C'1 , C'2, and C'3 is a carbon atom ;
L is a linker comprising at least one optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C3-C15 heteroarylene;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound comprises at least one optionally substituted 5-8 membered ring formed by joining (i) R2, R3, and C1 ; (ii) R3, R4, N1 , and C1 ; (iii) R5, R6, and C2; (iv) R6, R7, N2, and C2; (v) R8, R9, and C3; (vi) R9, R10, N3, and C3; (vii) R'2, R'3, and C'1 ; (viii) R'3, R'4, N'1 , and C'1 ; (ix) R'5, R'6, and C'2; (x) R'6, R'7, N'2, and C'2; (xi) R'8, R'9, and C'3; or (xii) R'9, R'10, N'3, and C'3.
In some embodiments, L is described by formula (L-VI) :
Lc
LB-Q-LA
(L-VI)
wherein LA is described by formula GA1-(ZA1)gi-(YA1)hi-(ZA2)ii-(YA2)ji-(ZA3)ki-(YA3)ii-(ZA4)mi-
Figure imgf000075_0001
LB is described by formula GB1-(ZB1 )g2-(YB1 )h2-(ZB2)i2-(YB2)j2-(ZB3)k2-(YB3)i2-(ZB4)m2-(YB4)n2-(ZB5)02-
GB2;
Lc is described by formula Gc1-(Zc 1 )g3-(Yc1 )h3-(ZC2)i3-(YC2)j3-(ZC3)k3-(YC3)i3-(ZC4)m3-(YC4)n3-(ZC5)03-
GC2;
GA1 is a bond attached to Q in formula (L-VI);
GA2 is a bond attached to A1 or M1 if A1 is absent;
GB1 is a bond attached to Q in formula (L-VI);
GB2 is a bond attached to A2 or M2 if A2 is absent;
GC1 is a bond attached to Q in formula (L-VI);
GC2 is a bond attached to E; each of ZA1 , ZA2, ZA3, ZA4, ZA5, ZB1 , ZB2, ZB3, ZB4, ZB5, ZC1 , ZC2, ZC3, ZC4 and ZC5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C1 5 arylene, or optionally substituted C2-C15 heteroarylene;
each of YA1 , YA2, YA3, YA4, YB1 , YB2, YB3, YB4, YC1 , YC2, YC3, and YC4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;
R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , o1 , g2, h2, i2, j2, k2, I2, m2, n2, o2, g3, h3, i3, j3, k3, I3, m3, n3, and o3 is, independently, 0 or 1 ;
Q comprises at least one optionally substituted C3-C20 cycloalkylene, optionally substituted C3- C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C3-C15 heteroarylene.
In some embodiments, L is described b formula (L-VII):
Figure imgf000076_0001
-Q- wherein LA is described by formula GA1-(ZA1)gi-(YA1)hi-(ZA2)ii-(YA2)ji -(ZA3)ki -(YA3)ii -(ZA4)mi -
Figure imgf000076_0002
LB is described by formula GB1-(ZB1 )g2-(YB1 )h2-(ZB2)i2-(YB2)j2-(ZB3)k2-(YB3)i2-(ZB4)m2-(YB4)n2-(ZB5)02-
GB2;
Lc is described by formula Gc1 -(Zc1 )g3-(Yc1 )h3-(ZC2)i3-(YC2)j3-(ZC3)k3-(YC3)i3-(ZC4)m3-(YC4)n3-(ZC5)03-
GC2;
LD is described by formula GD1 -(ZD1 )g4-(YD1 )h4-(ZD2)i4-(YD2)j4-(ZD3)k4-(YD3)i4-(ZD4)m4-(YD4)n4-(ZD5)o4-
GD2;
GA1 is a bond attached to Q in formula (L-VII);
GA2 is a bond attached to A1 or M1 if A1 is absent; GB1 is a bond attached to Q in formula (L-VII);
GB2 is a bond attached to A2 or M2 if A2 is absent;
GC1 is a bond attached to Q in formula (L-VII);
GC2 is a bond attached to a first monosaccharide or oligosaccharide moiety, E1 ;
GD1 is a bond attached to N in formula (L-VII);
GD2 is a bond attached to a second monosaccharide or oligosaccharide moiety, E2;
each of ZA1 ZA2 ZA3 ZA4 ZA5 ZB1 ZB2 ZB3 ZB4 ZB5 ZC1 ZC2 ZC3 ZC4 ZC5 ZD1 ZD2 ZD3 ZD4 and ZD5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20
heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20
heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20
heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene;
each of YA1 YA2 YA3 YA4 YB1 YB2 YB3 YB4 YC1 YC2 YC3 YC4 YD1 YD2 YD3 and YD4 is independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;
R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , o1 , g2, h2, i2, j2, k2, I2, m2, n2, o2, g3, h3, i3, j3, k3, I3, m3, n3, o3, g4, h4, i4, j4, k4, I4, m4, n4, and o4 is, independently, 0 or 1 ;
Q comprises at least one optionally substituted C3-C20 cycloalkylene, optionally substituted C3- C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C3-C15 heteroarylene.
In some embodiments the compound is described by formula (XXXV):
Figure imgf000077_0001
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl;
each of R2, R6, R8, R'2, R'6, and R'8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
In some embodiments the compound is described by formula (XXXV):
Figure imgf000078_0001
(XXXV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl;
each of R2, R6, R8, R'2, and R'6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XXXVI):
Figure imgf000079_0001
(XXXVI)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl;
each of R2, R6, R8, and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
In some embodiments the compound is described by formula (XXXVII):
Figure imgf000079_0002
(XXXVII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl;
each of R2, R6, and R8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XXXVIII):
Figure imgf000080_0001
(XXXVIII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl;
each of R2, R6, R'2, and R'6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XXXIX):
Figure imgf000080_0002
(XXXIX)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl;
each of R2, R6, and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
In some embodiments the compound is described by formula (XXXX):
Figure imgf000081_0001
(XXXX)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl;
each of R2 and R6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XXXXI):
Figure imgf000081_0002
(XXXXI) wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl;
each of R2 and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
In some embodiments the compound is described by formula (XXXXII):
Figure imgf000082_0001
(XXXXII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl;
R2 is a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XXXXIII):
Figure imgf000083_0001
(XXXXIII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
In some embodiments, L comprises at least one an optionally substituted C3 cycloalkylene or an optionally substituted C3 heterocycloalkylene.
In some embodiments the compound is described by formula (XXXXIV):
Figure imgf000083_0002
(XXXXIV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and
L comprises an optionally substituted C3 cycloalkylene or an optionally substituted C3 heterocycloalkylene,
or a pharmaceutically acceptable salt thereof.
In some embodiments the compound is described by formula (XXXXV):
Figure imgf000083_0003
(XXXXV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and
L comprises an optionally substituted C3 cycloalkylene or an optionally substituted C3 heterocycloalkylene,
or a pharmaceutically acceptable salt thereof.
In some embodiments, L is
Figure imgf000084_0001
wherein each q is, independently, an integer from 1 to 1 1 , inclusive.
In some embodiments, L comprises at least one an optionally substituted C5 cycloalkyl optionally substituted C5 heterocycloalkylene.
In some embodiments, the compound is described by formula (XXXXVI):
Figure imgf000084_0002
(XXXXVI)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and
L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XXXXVII):
Figure imgf000085_0001
(XXXXVII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted
C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and
L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene,
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XXXXVIII):
Figure imgf000085_0002
(XXXXVIII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene,
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound is described by formula (XXXXIX):
Figure imgf000085_0003
(XXXXIX)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and
L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene,
or a pharmaceutically acceptable salt thereof.
In some embodiments L is
Figure imgf000086_0001
wherein each q is, independently, an integer from 1 to 1 1 , inclusive.
In some embodiments, L comprises at least one an optionally substituted C6 cycloalkylene, at least one an optionally substituted C6 heterocycloalkylene, or at least one an optionally substituted C6 arylene.
In some embodiments the compound is described by formula (XXXXX):
Figure imgf000086_0002
(XXXXX)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and L comprises an optionally substituted C6 arylene,
or a pharmaceutically acceptable salt thereof.
In some embodiments, L is
Figure imgf000087_0001
wherein each q is, independently, an integer from 1 to 1 1 , inclusive.
In some embodiments, non-natural amino acids that may be included in a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)), for example, in the polymyxin core portions (e.g., in the first cyclic heptapeptide and/or the second cyclic heptapeptide) and/or the linker portion of the compound, include, but are not limited to, D-Ser, D-Pro, D-Leu, D-Nle (D-norleucine), D-Thr, D-Val, L-Abu (L-2-aminobutyric acid), 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2- piperazinecarboxylic acid, 2-aminooctanoic acid, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine. In some embodiments, polymyxin core portions (e.g., in the first cyclic heptapeptide and/or the second cyclic heptapeptide) of the compound include D-Ser, D- Pro, D-Leu, D-Nle, D-Thr, D-Val, and/or L-Abu.
In some embodiments of the compounds described herein, a concentration of the compound, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to 10,000 nM. In some embodiments, the concentration of the compound, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to equal to 1 ,000 nM. In some embodiments, the concentration of the compound, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to equal to 100 nM.
In another aspect, the disclosure features a pharmaceutical composition including a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition further includes an antibacterial agent.
In some embodiments, the antibacterial agent is selected from the group consisting of linezolid, tedizolid, posizolid, radezolid, retapamulin, valnemulin, tiamulin, azamulin, lefamulin, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem, doripenem, imipenem/cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefepime, ceftaroline fosamil, ceftobiprole, teicoplanin, vancomycin, telavancin, dalbavancin, oritavancin, clindamycin, lincomycin, daptomycin, solithromycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, spiramycin, aztreonam, furazolidone, nitrofurantoin, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin g, penicillin v, piperacillin, penicillin g, temocillin, ticarcillin, amoxicillin clavulanate, ampicillin/sulbactam,
piperacillin/tazobactam, ticarcillin/clavulanate, bacitracin, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethizole, sulfamethoxazole, sulfanamide, sulfasalazine, sulfisoxazole, trimethoprim-sulfamethoxazole (tmp-smx), sulfonamidochrysoidine, eravacycline, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol(bs), ethionamide, isoniazid, pyrazinamide, rifampicin, rifabutin, rifapentine, streptomycin, arsphenamine, chloramphenicol, fosfomycin, fusidic acid, metronidazole, mupirocin, platensimycin, quinupristin/dalfopristin, thiamphenicol, tigecycline, tinidazole, and trimethoprim, prodrugs thereof, and pharmaceutically acceptable salts thereof.
In some embodiments, a prodrug of tedizolid is tedizolid phosphate. In some embodiments, the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, levofloxacin, rifampicin, linezolid, erythromycin, or solithromycin. In some embodiments, the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, or levofloxacin.
In another aspect, the disclosure features a method of protecting against or treating a bacterial infection in a subject by administering to the subject a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)).
In some embodiments of the method, the method further includes administering to the subject an antibacterial agent.
In another aspect, the disclosure features a method of protecting against or treating a bacterial infection in a subject by administering to the subject (1 ) a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) and (2) an antibacterial agent.
In some embodiments of this aspect of the disclosure, the bacterial infection is caused by Gram- negative bacteria. In some embodiments, the bacterial infection is caused by a resistant strain of bacteria. In some embodiments, the resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene and/or a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, or the mcr-3 gene. In some embodiments, the resistant strain of bacteria possesses a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of bacteria is a resistant strain of E. coli.
In another aspect, the disclosure features a method of protecting against or treating sepsis in a subject by administering to the subject a compound described herein (e.g., a compound of any one of formulas (I)- (XXXXX)).
In another aspect, the disclosure features a method of preventing lipopolysaccharides (LPS) in Gram- negative bacteria from activating an immune system in a subject by administering to the subject a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)). In some embodiments, the method prevents LPS from activating a macrophage. In some embodiments, the method prevents LPS-induced nitric oxide (NO) production from a macrophage. In some embodiments of this aspect of the disclosure, the Gram-negative bacteria is a resistant strain of Gram-negative bacteria. In some embodiments, the resistant strain of Gram-negative bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene and/or a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of Gram-negative bacteria possesses the mcr- 1 gene, the mcr-2 gene, or the mcr-3 gene. In some embodiments, the resistant strain of Gram- negative bacteria possesses a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of Gram-negative bacteria is a resistant strain of E. coli.
In some embodiments of this aspect of the disclosure, the method further includes administering to the subject an antibacterial agent.
In some embodiments, the compound and the antibacterial agent are administered substantially simultaneously.
In some embodiments, the compound and the antibacterial agent are administered separately. In some embodiments, the compound is administered first, followed by administering of the antibacterial agent alone. In some embodiments, the antibacterial agent is administered first, followed by administering of the compound alone.
In some embodiments, the compound and the antibacterial agent are administered substantially simultaneously, followed by administering of the compound or the antibacterial agent alone. In some embodiments, the compound or the antibacterial agent is administered first, followed by administering of the compound and the antibacterial agent substantially simultaneously.
In some embodiments, administering the compound and the antibacterial agent together lowers the MIC of each of the compound and the antibacterial agent relative to the MIC of each of the compound and the antibacterial agent when each is used alone.
In some embodiments of the methods described herein, the compound and/or the antibacterial agent is administered intramuscularly, intravenously, intradermal^, intraarterially, intraperitoneally, intralesionally, intracranial^, intraarticularly, intraprostatically, intrapleural^, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctival, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, orally, locally, by inhalation, by injection, or by infusion.
In another aspect, the disclosure features a method of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria, by contacting the bacteria or a site susceptible to bacterial growth with a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)).
In some embodiments, the method further includes contacting the bacteria or the site susceptible to bacterial growth with an antibacterial agent.
In another aspect, the disclosure features a method of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria, including contacting the bacteria or a site susceptible to bacterial growth with (1 ) a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) and (2) an antibacterial agent.
In some embodiments of the methods described herein, the antibacterial agent is selected from the group consisting of linezolid, tedizolid, posizolid, radezolid, retapamulin, valnemulin, tiamulin, azamulin, lefamulin, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem, doripenem, imipenem/cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefepime, ceftaroline fosamil, ceftobiprole, teicoplanin, vancomycin, telavancin, dalbavancin, oritavancin, clindamycin, lincomycin, daptomycin, solithromycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, spiramycin, aztreonam, furazolidone, nitrofurantoin, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin g, penicillin v, piperacillin, penicillin g, temocillin, ticarcillin, amoxicillin clavulanate,
ampicillin/sulbactam, piperacillin/tazobactam, ticarcillin/clavulanate, bacitracin, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethizole, sulfamethoxazole, sulfanamide, sulfasalazine, sulfisoxazole, trimethoprim-sulfamethoxazole (tmp-smx), sulfonamidochrysoidine, eravacycline, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol(bs), ethionamide, isoniazid, pyrazinamide, rifampicin, rifabutin, rifapentine, streptomycin, arsphenamine, chloramphenicol, fosfomycin, fusidic acid, metronidazole, mupirocin, platensimycin, quinupristin/dalfopristin, thiamphenicol, tigecycline, tinidazole, and
trimethoprim, prodrugs thereof, and pharmaceutically acceptable salts thereof.
In some embodiments, a prodrug of tedizolid is tedizolid phosphate. In some embodiments, the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, levofloxacin, rifampicin, linezolid, erythromycin, or solithromycin. In some embodiments, the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, or levofloxacin.
In some embodiments, the bacteria are Gram-negative bacteria. In some embodiments, the bacteria are a resistant strain of bacteria. In some embodiments, the resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene and/or a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene. In some embodiments, the resistant strain of bacteria possesses a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of bacteria is a resistant strain of E. coli.
In another aspect, the disclosure features any compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX) and/or any one of compounds 1 -123, or a pharmaceutically
acceptablesalt thereof).
In some embodiments, the disclosure provides a compound described by any one Formulas (IV)- (XIII), (XV), or (XXXIV), wherein each of R'1 and R1 is, independently, benzyl or CH2CH(CH3)2.
Definitions
The term "cyclic heptapeptide" or "cycloheptapeptide," as used herein, refers to certain compounds that bind to lipopolysaccharides (LPS) in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics. Cyclic heptapeptides or cycloheptapeptides comprise seven natural or non-natural a-amino acid residues, such as D- or L-amino acid residues, in a closed ring. Generally, cyclic heptapeptides are formed by linking the a-carboxyl group of one amino acid to the a-amino group or the γ-amino group of another amino acid and cyclizing. The cyclic heptapeptide comprises a heterocycle comprising carbon and nitrogen ring members, which may be substituted, for example, with amino acid side chains. One nitrogen from an a-amino group in the cyclic heptapeptide, however, is not a ring member and is branched from a ring member of the heterocycle. Thus, this nitrogen is directly attached to a ring member, such as a carbon atom (e.g., an a-carbon atom). This nitrogen atom serves as an attachment point for the cyclic heptapeptide to a linker and/or to a peptide (e.g., a peptide including 1 -5 amino acid residue(s)), and thus is referred to herein as a "linking nitrogen." The linking nitrogen is directly attached to the ring of the cyclic heptapeptide and is not derived from a side chain, such as an ethylamine side chain. The linking nitrogens in a compound of, e.g., formula (II) or (III), are N4 and N'4.
In some embodiments, a peptide including one or more (e.g., 1 -5; 1 , 2, 3, 4, or 5) amino acid residues (e.g., natural and/or non-natural amino acid residues) may be covalently attached to a linking nitrogen (e.g., N4 and/or N'4, the nitrogen from an a-amino group) in the cyclic heptapeptide ring. Cyclic heptapeptides may be derived from polymyxins (e.g., naturally existing polymyxins and non-natural polymyxins) and/or octapeptins (e.g., naturally existing octapeptins and non-natural octapeptins).
Examples of naturally existing polymyxins include, but are not limited to, polymyxin Bi , polymyxin B2, polymyxin B3, polymyxin B4, polymyxin B5, polymyxin Ββ, polymyxin BHIe, polymyxin B2-lle, polymyxin Ci , polymyxin C2, polymyxin Si , polymyxin Ti , polymyxin T2, polymyxin Ai , polymyxin A2, polymyxin Di , polymyxin D2, polymyxin Ei (colistin A), polymyxin E2 (colistin B), polymyxin E3, polymyxin E4, polymyxin E7, polymyxin EHIe, polymyxin Ei-Val, polymyxin Ei-Nva, polymyxin E2-lle, polymyxin E2-Val, polymyxin E2-Nva, polymyxin Es-lle, polymyxin Mi , and polymyxin M2. In other embodiments, a cyclic heptapeptide may be entirely synthetic and prepared by standard peptide methodology as known in the art.
The term "covalently attached" refers to two parts of a compound that are linked to each other by a covalent bond formed between two atoms in the two parts of the compound. For example, in the compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)), when a' is 0, L is covalently attached to N'4, which means that when a' is 0, an atom in L forms a covalent bond with N'4 in the compound. Similarly, when a' is 1 and b' is 0, L is covalently attached to N'1 ; when b' is 1 , and c' is 0, L is covalently attached to N'2; when c' is 1 , L is covalently attached to N'3; when a is 0, L is covalently attached to N4; when a is 1 and b is 0, L is covalently attached to N1 ; when b is 1 , and c is 0, L is covalently attached to N2; and when c is 1 , L is covalently attached to N3.
The terms "linker," "Ι_'," "L," "L1 ," and "L ," as used herein, refer to a covalent linkage or connection between two or more components in a compound (e.g., two cyclic heptapeptides and one or more monosaccharide or oligosaccharide moieties in a compound described herein). In some embodiments, a compound described herein may contain a linker that has a trivalent structure (e.g., a trivalent linker). A trivalent linker has three arms, in which each arm is conjugated to a component of the compound (e.g., a first arm conjugated to a first cyclic heptapeptide, a second arm conjugated to a second heptapeptide, and a third arm conjugated to one or more monosaccharide or oligosaccharide moieties). In some embodiments, a compound described herein may contain two or three linkers (e.g., a compound of formula (XIII) or (XIV)), in which each linker has a divalent structure (e.g., a divalent linker). For example, the first linker may connect the first and second cyclic heptapeptides (e.g., L in the compound of formula (XIII)), the second linker may connect a first monosaccharide or oligosaccharide moiety and a peptide (e.g., a peptide including a 1 -5 amino acid residue(s)) attached to the first cyclic heptapeptide (e.g., L1 in the compound of formula (XIII)), and a third linker may connect a second monosaccharide or oligosaccharide moiety and a peptide (e.g., a peptide including a 1 -5 amino acid residue(s)) attached to the second cyclic heptapeptide (e.g., L'1 in the compound of formula (XIII)).
Molecules that may be used as linkers include at least two functional groups, which may be the same or different, e.g., two carboxylic acid groups, two amine groups, two sulfonic acid groups, a carboxylic acid group and an amine group, or a carboxy group and a sulfonic acid group. The first functional group may form a covalent linkage with a first component in the compound and the second functional group may form a covalent linkage with the second component in the compound. In some embodiments of a trivalent linker, two arms of a linker may contain two dicarboxylic acids, in which the first carboxylic acid may form a covalent linkage with the first cyclic heptapeptide in the compound and the second carboxylic acid may form a covalent linkage with the second cyclic heptapeptide in the compound, and the third arm of the linker may for a covalent linkage (e.g., a C bond) with one or more monosaccharide or oligosaccharide moieties in the compound. In some embodiments of a divalent linker, the divalent linker may contain two carboxylic acids, in which the first carboxylic acid may form a covalent linkage with one component (e.g., a first cyclic heptapeptide) in the compound and the second carboxylic acid may form a covalent linkage with another component (e.g., a second cyclic heptapeptide) in the compound.
Examples of dicarboxylic acids are described further herein. In some embodiments, a molecule containing one or more sulfonic acid groups may be used as a linker, in which the sulfonic acid group may form a sulfonamide linkage with a component in the compound. In some embodiments, a molecule containing one or more isocyanate groups may be used as a linker, in which the isocyanate group may form a urea linkage with a component in the compound. In some embodiments, a molecule containing one or more haloalkyl groups may be used as a linker, in which the haloalkyl group may form a covalent linkage, e.g., C-N and C-0 linkages, with a component in the compound.
In some embodiments, a linker provides space, rigidity, and/or flexibility between the two or more components. In some embodiments, a linker may be a bond, e.g., a covalent bond. The term "bond" refers to a chemical bond, e.g., an amide bond, a disulfide bond, a C-0 bond, a N-N bond, or any kind of bond created from a chemical reaction, e.g., chemical conjugation. In some embodiments, a linker includes no more than 250 atoms. In some embodiments, a linker includes no more than 250 non- hydrogen atoms. In some embodiments, the backbone of a linker includes no more than 250 atoms. The "backbone" of a linker refers to the atoms in the linker that together form the shortest path from one part of a compound to another part of the compound (e.g., the shortest path linking a first cyclic heptapeptide and a second cyclic heptapeptide). The atoms in the backbone of the linker are directly involved in linking one part of a compound to another part of the compound (e.g., linking a first cyclic heptapeptide and a second cyclic heptapeptide). For examples, hydrogen atoms attached to carbons in the backbone of the linker are not considered as directly involved in linking one part of the compound to another part of the compound. In some embodiments, a linker may comprise a synthetic group derived from, e.g., a synthetic polymer (e.g., a polyethylene glycol (PEG) polymer). In some embodiments, a linker may comprise one or more amino acid residues, such as D- or L-amino acid residues. In some embodiments, a linker may be a residue of an amino acid sequence (e.g., a 1 -25 amino acid, 1 -10 amino acid, 1 -9 amino acid, 1 -8 amino acid, 1 -7 amino acid, 1 -6 amino acid, 1 -5 amino acid, 1 -4 amino acid, 1 -3 amino acid, 1 -2 amino acid, or 1 amino acid sequence). In some embodiments, a linker may comprise one or more, e.g., 1 -100, 1 -50, 1 -25, 1 -10, 1 -5, or 1 -3, optionally substituted alkylene, optionally substituted heteroalkylene (e.g., a PEG unit), optionally substituted alkenylene, optionally substituted heteroalkenylene, optionally substituted alkynylene, optionally substituted heteroalkynylene, optionally substituted cycloalkylene, optionally substituted heterocycloalkylene, optionally substituted cycloalkenylene, optionally substituted heterocycloalkenylene, optionally substituted cycloalkynylene, optionally substituted
heterocycloalkynylene, optionally substituted arylene, optionally substituted heteroarylene (e.g., pyridine), O, S, NR' (Ri is H, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkenyl, optionally substituted heteroalkenyl, optionally substituted alkynyl, optionally substituted heteroalkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted cycloalkenyl, optionally substituted heterocycloalkenyl, optionally substituted cycloalkynyl, optionally substituted heterocycloalkynyl, optionally substituted aryl, or optionally substituted heteroaryl), P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino. For example, a linker may comprise one or more optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene (e.g., a PEG unit), optionally substituted C2-C20 alkenylene (e.g., C2 alkenylene), optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20
heteroalkynylene, optionally substituted C3-C20 cycloalkylene (e.g., cyclopropylene, cyclobutylene), optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene (e.g., C6 arylene), optionally substituted C2-C15 heteroarylene (e.g., imidazole, pyridine), O, S, NR' (R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2- C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl), P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino.
The term "lipophilic moiety," as used herein, refers to a portion, substituent, or functional group of a compound that is, in general, hydrophobic and non-polar. A moiety is lipophilic if it has a hydrophobicity determined using a cLogP value of greater than 0, such as about 0.25 or greater, about 0.5 or greater, about 1 or greater, about 2 or greater, 0.25-5, 0.5-4 or 2-3. As used herein, the term "cLogP" refers to the calculated partition coefficient of a molecule or portion of a molecule. The partition coefficient is the ratio of concentrations of a compound in a mixture of two immiscible phases at equilibrium (e.g., octanol and water) and measures the hydrophobicity or hydrophilicity of a compound. A variety of methods are available in the art for determining cLogP. For example, in some embodiments, cLog P can be determined using quantitative structure-property relationship algorithims known in the art (e.g. , using fragment based prediction methods that predict the logP of a compound by determining the sum of its non-overlapping molecular fragments). Several algorithims for calculating cLog P are known in the art including those used by molecular editing software such as CH EMDRAW® Pro, Version 12.0.2.1092
(Camrbridgesoft, Cambridge, MA) and MARVINSKETCH® (ChemAxon, Budapest, Hungary). A moiety is considered lipophilic if it has a cLogP value described above in at least one of the above methods. A lipophilic moiety having the stated cLogP value will be considered lipophilic, even though it may have a positive charge or a polar substituent.
In some embodiments, a lipophilic moiety contains entirely hydrocarbons. In some embodiments, a lipophilic moiety may contain one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms independently selected from N, O, and S (e.g. , an indolyl), or one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, halo groups, which, due to the structure of the moiety and/or small differences in electronegativity between the heteroatoms or halo groups and the hydrocarbons, do not induce significant chemical polarity into the lipophilic moiety. Thus, in some embodiments, a lipophilic moiety having, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms and/or, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, halo atoms may still be considered non-polar. In some embodiments, a lipophilic moiety may be optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heteroalkenyl, optionally substituted heteroalkynyl , or optionally substituted heteroaryl, or halo forms thereof, wherein the optional substituents are also lipophilic (such as alkyl, alkenyl , alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl , or heteroaryl) or are not lipophilic but do not change the overall lipophilic character of the moiety, i.e. , the moiety has a cLogP value of greater than 0. For example, octanol contains a polar group, OH, but is still a lipophilic moiety. In some embodiments, a lipophilic moiety may be benzyl, isobutyl, sec-butyl, isopropyl, n-propyl, methyl, biphenylmethyl, n-octyl , or substituted indolyl (e.g. , alkyl substituted indolyl). In some embodiments, a lipophilic moiety may be the side chain of a hydrophobic amino acid residue, e.g ., leucine, isoleucine, alanine, phenylalanine, valine, and proline, or groups such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl , and pyrrolidinyl. In some embodiments, lipophilic moieties of the compounds described herein may interact with the hydrophobic portions of lipid A (e.g. , fatty acid side chains of lipid A) when the compounds bind to the membrane of bacterial cells (e.g. , Gram-negative bacterial cells). Due to its position on the cyclic heptapeptide, one or more of R1 , R12, R15, R'1 , R'12, and R'15 may be a lipophilic moiety.
The term "positively charged moiety," as used herein, refers to a portion , substituent, or functional group of a compound that contains at least one positive charge. In some embodiments, a positively charged moiety contains one or more (e.g., 1 -4, 1 -3, 1 , 2, 3, or 4) heteroatoms independently selected from N, O, and S, for example. In some embodiments, a positively charged moiety may possess a pH- dependent positive charge, e.g. , the moiety becomes a positively charged moiety at physiological pH (e.g. , pH 7), such as -NH3 +, -(CH2)4NH2, -(CH2)3NH2, -(CH2)2NH2, -CH2NH2, -(CH2)4N(CH3)2,
-(CH2)3N(CH3)2, -(CH2)2N(CH3)2, -CH2N(CH3)2, -(CH2)4NH(CH3), -(CH2)3NH(CH3),-(CH2)2NH(CH3), and -CH2NH(CH3). In some embodiments, a positively charged moiety may be optionally substituted alkamino, optionally substituted heteroalkyl (e.g., optionally substituted heteroalkyl containing 1 -3 nitrogens; -(CH2)4-guanidinium, -(CH2)3-guanidinium, -(CH2)2-guanidinium, -CH2-guanidinium), optionally substituted heterocycloalkyl (e.g., optionally substituted heterocycloalkyl containing 1 -3 nitrogens), or optionally substituted heteroaryl (e.g., optionally substituted heteroaryl containing 1 -3
nitrogens; -(CH2)4-imidazole, -(CH2)3-imidazole, -(CH2)2-imidazole, -CH2-imidazole). In some
embodiments, a positively charged moiety may be pH independent such
as -CH2N(CH3)3+, -(CH2)2N(CH3)3+, -(CH2)3N(CH3)3+, or -(CH2)4N(CH3)3+. Thus, substituents may transform an otherwise lipophilic moiety such as optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heteroalkenyl, optionally substituted heteroalkynyl, or optionally substituted heteroaryl, or halo forms thereof, to a positively charged moiety with the addition of a substituent that imparts a positive charge or a pH dependent positive charge, such as guanidinyl, -NH3+, -NH2, -NH(CH3), -N(CH3)2, and/or -N(CH3)3+. In some embodiments, a positively charged moiety may be the side chain of an amino acid residue (e.g., a natural or non-natural amino acid residue, such as a D- or L-amino acid residue, that is positively charged at physiological pH (e.g., pH 7), such as the side chain of a basic amino acid residue (e.g., arginine, lysine, histidine, ornithine, diaminobuteric acid, or diaminopropionic acid). In some embodiments, positively charged moieties of the compounds described herein interact with the negatively charged portions of lipid A (e.g., phosphates of lipid A) when the compounds bind to the membrane of bacterial cells (e.g., Gram-negative bacterial cells). Due to its position on the cyclic heptapeptide, one or more of R1 1 , R13, R14, R'1 1 , R'13, and R'14 may be a positively charged moiety.
The term "polar moiety," as used herein, refers to a portion, substituent, or functional group of a compound that has a chemical polarity induced by atoms with different electronegativity. The polarity of a polar moiety is dependent on the electronegativity between atoms within the moiety and the asymmetry of the structure of the moiety. In some embodiments, a polar moiety contains one or more (e.g., 1 -4, 1 -3, 1 , 2, 3, or 4) heteroatoms independently selected from N, O, and S, which may induce chemical polarity in the moiety by having different electronegativity from carbon and hydrogen. In general, a polar moiety interacts with other polar or charged molecules. In some embodiments, a polar moiety may be optionally substituted alkamino, optionally substituted heteroalkyl (e.g., N- and/or O-containing
heteroalkyl; -(CH2)4-carboxylic acid, -(CH2)3-carboxylic acid, -(CH2)2-carboxylic acid, -CH2-carboxylic acid), optionally substituted heterocycloalkyl (e.g., N- and/or O-containing heterocycloalkyl), or optionally substituted heteroaryl (e.g., N- and/or O-containing heteroaryl). In some embodiments, a polar moiety may -CH(CH3)OH, -CH2OH, -(CH2)2CONH2, -CH2CONH2, -CH2COOH, or -(CH2)2COOH. Thus, substituents may transform an otherwise lipophilic moiety such optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heteroalkenyl, optionally substituted heteroalkynyl, or optionally substituted heteroaryl, or halo forms thereof, to a polar moiety with the addition of a substituent that imparts polarity, such as -OH, -COOH, -COOR, or -CONR2, in which R is H or C1 -C4 alkyl. In some embodiments, a polar moiety may be the side chain or a polar or charged amino acid residue (e.g., threonine, serine, glutamine, asparagine, arginine, lysine histidine, aspartic acid, and glutamic acid). In some embodiments, a polar moiety is the side chain of threonine. In some embodiments, polar moieties of the compounds described herein interact with the negatively charged portions of lipid A (e.g., phosphates of lipid A) when the compounds bind to the membrane of bacterial cells (e.g., Gram-negative bacterial cells). Due to its position on the cyclic heptapeptide, one or more of R1 , R12, R15, R'1 , R'12, and R'15 may be a polar moiety.
The term "polymyxin core," as used herein means a cyclic heptapeptide having the structure:
Figure imgf000096_0001
wherein Q1 , Q2 and Q3 are as follows:
Figure imgf000096_0002
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
attachment of the polymyxin core to the remainder of the compounds disclosed herein, including the second polymyxin core, the linker, and E groups of the compounds disclosed herein.
The terms "alkyl," "alkenyl," and "alkynyl," as used herein, include straight-chain and branched- chain monovalent substituents, as well as combinations of these, containing only C and H when unsubstituted. When the alkyl group includes at least one carbon-carbon double bond or carbon-carbon triple bond, the alkyl group can be referred to as an "alkenyl" or "alkynyl" group respectively. The monovalency of an alkyl, alkenyl, or alkynyl group does not include the optional substituents on the alkyl, alkenyl, or alkynyl group. For example, if an alkyl, alkenyl, or alkynyl group is attached to a compound, monovalency of the alkyl, alkenyl, or alkynyl group refers to its attachment to the compound and does not include any additional substituents that may be present on the alkyl, alkenyl, or alkynyl group. In some embodiments, the alkyl or heteroalkyl group may contain, e.g., 1 -20. 1 -18, 1 -16, 1 -14, 1 -12, 1 -10, 1 -8, 1 - 6, 1 -4, or 1 -2 carbon atoms (e.g., C1 -C20, C1 -C18, C1 -C16, C1 -C14, C1 -C12, C1 -C10, C1 -C8, C1 -C6, C1 -C4, or C1 -C2). In some embodiments, the alkenyl, heteroalkenyl, alkynyl, or heteroalkynyl group may contain, e.g., 2-20, 2-18, 2-16, 2-14, 2-12, 2-10, 2-8, 2-6, or 2-4 carbon atoms (e.g., C2-C20, C2-C18, C2-C16, C2-C14, C2-C12, C2-C10, C2-C8, C2-C6, or C2-C4). Examples include, but are not limited to, methyl, ethyl, isobutyl, sec-butyl, tert-butyl, 2-propenyl, and 3-butynyl.
The term "cycloalkyl," as used herein, represents a monovalent saturated or unsaturated non- aromatic cyclic alkyl group. A cycloalkyl may have, e.g., three to twenty carbons (e.g., a C3-C7, C3-C8, C3-C9, C3-C10, C3-C1 1 , C3-C12, C3-C14, C3-C16, C3-C18, or C3-C20 cycloalkyi). Examples of cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl. When the cycloalkyi group includes at least one carbon-carbon double bond, the cycloalkyi group can be referred to as a "cycloalkenyl" group. A cycloalkenyl may have, e.g., four to twenty carbons (e.g., a C4- C7, C4-C8, C4-C9, C4-C10, C4-C1 1 , C4-C12, C4-C14, C4-C16, C4-C18, or C4-C20 cycloalkenyl).
Exemplary cycloalkenyl groups include, but are not limited to, cyclopentenyl, cyclohexenyl, and cycloheptenyl. When the cycloalkyi group includes at least one carbon-carbon triple bond, the cycloalkyi group can be referred to as a "cycloalkynyl" group. A cycloalkynyl may have, e.g., eight to twenty carbons (e.g., a C8-C9, C8-C10, C8-C1 1 , C8-C12, C8-C14, C8-C16, C8-C18, or C8-C20 cycloalkynyl). The term "cycloalkyi" also includes a cyclic compound having a bridged multicyclic structure in which one or more carbons bridges two non-adjacent members of a monocyclic ring, e.g., bicyclo[2.2.1 .]heptyl and adamantane. The term "cycloalkyi" also includes bicyclic, tricyclic, and tetracyclic fused ring structures, e.g., decalin and spiro cyclic compounds.
The term "aryl," as used herein, refers to any monocyclic or fused ring bicyclic or tricyclic system which has the characteristics of aromaticity in terms of electron distribution throughout the ring system, e.g., phenyl, naphthyl, or phenanthrene. In some embodiments, a ring system contains 5-15 ring member atoms or 5-10 ring member atoms. An aryl group may have, e.g., five to fifteen carbons (e.g., a C5-C6, C5-C7, C5-C8, C5-C9, C5-C10, C5-C1 1 , C5-C12, C5-C13, C5-C14, or C5-C15 aryl). The term
"heteroaryl" also refers to such monocyclic or fused bicyclic ring systems containing one or more, e.g., 1 - 4, 1 -3, 1 , 2, 3, or 4, heteroatoms selected from O, S and N. A heteroaryl group may have, e.g., two to fifteen carbons (e.g., a C2-C3, C2-C4, C2-C5, C2-C6, C2-C7, C2-C8, C2-C9. C2-C10, C2-C1 1 , C2-C12, C2-C13, C2-C14, or C2-C15 heteroaryl). The inclusion of a heteroatom permits inclusion of 5-membered rings to be considered aromatic as well as 6-membered rings. Thus, typical heteroaryl systems include, e.g., pyridyl, pyrimidyl, indolyl, benzimidazolyl, benzotriazolyl, isoquinolyl, quinolyl, benzothiazolyl, benzofuranyl, thienyl, furyl, pyrrolyl, thiazolyl, oxazolyl, isoxazolyl, benzoxazolyl, benzoisoxazolyl, and imidazolyl. Because tautomers are possible, a group such as phthalimido is also considered heteroaryl. In some embodiments, the aryl or heteroaryl group is a 5- or 6-membered aromatic rings system optionally containing 1 -2 nitrogen atoms. In some embodiments, the aryl or heteroaryl group is an optionally substituted phenyl, pyridyl, indolyl, pyrimidyl, pyridazinyl, benzothiazolyl, benzimidazolyl, pyrazolyl, imidazolyl, isoxazolyl, thiazolyl, or imidazopyridinyl. In some embodiments, the aryl group is phenyl. In some embodiments, an aryl group may be optionally substituted with a substituent such an aryl substituent, e.g., biphenyl.
The term "alkaryl," refers to an aryl group that is connected to an alkylene, alkenylene, or alkynylene group. In general, if a compound is attached to an alkaryl group, the alkylene, alkenylene, or alkynylene portion of the alkaryl is attached to the compound. In some embodiments, an alkaryl is C6- C35 alkaryl (e.g., C6-C16, C6-C14, C6-C12, C6-C10, C6-C9, C6-C8, C7, or C6 alkaryl), in which the number of carbons indicates the total number of carbons in both the aryl portion and the alkylene, alkenylene, or alkynylene portion of the alkaryl. Examples of alkaryls include, but are not limited to, (C1 - C8)alkylene(C6-C12)aryl, (C2-C8)alkenylene(C6-C12)aryl, or (C2-C8)alkynylene(C6-C12)aryl. In some embodiments, an alkaryl is benzyl. In a heteroalkaryl, one or more heteroatoms selected from N, O, and S may be present in the alkylene, alkenylene, or alkynylene portion of the alkaryl group and/or may be present in the aryl portion of the alkaryl group. In an optionally substituted alkaryl, the substituent may be present on the alkylene, alkenylene, or alkynylene portion of the alkaryl group and/or may be present on the aryl portion of the alkaryl group.
The term "amino," as used herein, represents -N(RX)2 or -N+(RX)3, where each Rx is,
independently, H, alkyl, alkenyl, alkynyl, aryl, alkaryl, cycloalkyl, or two Rx combine to form a
heterocycloalkyl. In some embodiment, the amino group is -NH2.
The term "alkamino," as used herein, refers to an amino group, described herein, that is attached to an alkylene (e.g., C1 -C5 alkylene), alkenylene (e.g., C2-C5 alkenylene), or alkynylene group (e.g., C2- C5 alkenylene). In general, if a compound is attached to an alkamino group, the alkylene, alkenylene, or alkynylene portion of the alkamino is attached to the compound. The amino portion of an alkamino refers to -N(RX)2 or -N+(RX)3, where each Rx is, independently, H, alkyl, alkenyl, alkynyl, aryl, alkaryl, cycloalkyl, or two Rx combine to form a heterocycloalkyl. In some embodiment, the amino portion of an alkamino is -NH2. An example of an alkamino group is C1 -C5 alkamino, e.g., C2 alkamino (e.g., CH2CH2NH2 or CH2CH2N(CH3)2). In a heteroalkamino group, one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms selected from N, O, and S may be present in the alkylene, alkenylene, or alkynylene portion of the heteroalkamino group. In some embodiments, an alkamino group may be optionally substituted. In a substituted alkamino group, the substituent may be present on the alkylene, alkenylene, or alkynylene portion of the alkamino group and/or may be present on the amino portion of the alkamino group.
The term "alkamide," as used herein, refers to an amide group that is attached to an alkylene (e.g., C1 -C5 alkylene), alkenylene (e.g., C2-C5 alkenylene), or alkynylene (e.g., C2-C5 alkenylene) group. In general, if a compound is attached to an alkamide group, the alkylene, alkenylene, or alkynylene portion of the alkamide is attached to the compound. The amide portion of an alkamide refers to -C(0)-N(RX)2, where each Rx is, independently, H, alkyl, alkenyl, alkynyl, aryl, alkaryl, cycloalkyl, or two Rx combine to form a heterocycloalkyl. In some embodiment, the amide portion of an alkamide is -C(0)NH2. An alkamide group may be -(CH2)2-C(0)NH2 or -CH2-C(0)NH2. In a heteroalkamide group, one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms selected from N, O, and S may be present in the alkylene, alkenylene, or alkynylene portion of the heteroalkamide group. In some embodiments, an alkamide group may be optionally substituted. In a substituted alkamide group, the substituent may be present on the alkylene, alkenylene, or alkynylene portion of the alkamide group and/or may be present on the amide portion of the alkamide group.
The terms "alkylene," "alkenylene," and "alkynylene," as used herein, refer to divalent groups having a specified size. In some embodiments, an alkylene may contain, e.g., 1 -20, 1 -18, 1 -16, 1 -14, 1 - 12, 1 -10, 1 -8, 1 -6, 1 -4, or 1 -2 carbon atoms (e.g., C1 -C20, C1 -C18, C1 -C1 6, C1 -C14, C1 -C12, C1 -C1 0, C1 -C8, C1 -C6, C1 -C4, or C1 -C2). In some embodiments, an alkenylene or alkynylene may contain, e.g., 2-20, 2-18, 2-16, 2-14, 2-12, 2-10, 2-8, 2-6, or 2-4 carbon atoms (e.g., C2-C20, C2-C18, C2-C16, C2- C14, C2-C12, C2-C10, C2-C8, C2-C6, or C2-C4). Alkylene, alkenylene, and/or alkynylene includes straight-chain and branched-chain forms, as well as combinations of these. The divalency of an alkylene, alkenylene, or alkynylene group does not include the optional substituents on the alkylene, alkenylene, or alkynylene group. For example, two cyclic heptapeptides may be attached to each other by way of a linker that includes alkylene, alkenylene, and/or alkynylene, or combinations thereof. Each of the alkylene, alkenylene, and/or alkynylene groups in the linker is considered divalent with respect to the two attachments on either end of alkylene, alkenylene, and/or alkynylene group. For example, if a linker includes -(optionally substituted alkylene)-(optionally substituted alkenylene)-(optionally substituted alkylene)-, the alkenylene is considered divalent with respect to its attachments to the two alkylenes at the ends of the linker. The optional substituents on the alkenylene are not included in the divalency of the alkenylene. The divalent nature of an alkylene, alkenylene, or alkynylene group (e.g., an alkylene, alkenylene, or alkynylene group in a linker) refers to both of the ends of the group and does not include optional substituents that may be present in an alkylene, alkenylene, or alkynylene group. Because they are divalent, they can link together multiple (e.g., two) parts of a compound, e.g., a first cyclic heptapeptide and a second cyclic heptapeptide. Alkylene, alkenylene, and/or alkynylene groups can be substituted by the groups typically suitable as substituents for alkyl, alkenyl and alkynyl groups as set forth herein. For example, C=0 is a C1 alkylene that is substituted by an oxo (=0). For
example, -HCR-C≡C- may be considered as an optionally substituted alkynylene and is considered a divalent group even though it has an optional substituent, R. Heteroalkylene, heteroalkenylene, and/or heteroalkynylene groups refer to alkylene, alkenylene, and/or alkynylene groups including one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms, e.g., N, O, and S. For example, a polyethylene glycol (PEG) polymer or a PEG unit -(CH2)2-0- in a PEG polymer is considered a heteroalkylene containing one or more oxygen atoms.
The term "cycloalkylene," as used herein, refers to a divalent cyclic group linking together two parts of a compound. For example, one carbon within the cycloalkylene group may be linked to one part of the compound, while another carbon within the cycloalkylene group may be linked to another part of the compound. A cycloalkylene group may include saturated or unsaturated non-aromatic cyclic groups. A cycloalkylene may have, e.g., three to twenty carbons in the cyclic portion of the cycloalkylene (e.g., a C3-C7, C3-C8, C3-C9, C3-C10, C3-C1 1 , C3-C12, C3-C14, C3-C16, C3-C18, or C3-C20 cycloalkylene). When the cycloalkylene group includes at least one carbon-carbon double bond, the cycloalkylene group can be referred to as a "cycloalkenylene" group. A cycloalkenylene may have, e.g., four to twenty carbons in the cyclic portion of the cycloalkenylene (e.g., a C4-C7, C4-C8, C4-C9. C4-C10, C4-C1 1 , C4- C12, C4-C14, C4-C16, C4-C18, or C4-C20 cycloalkenylene). When the cycloalkylene group includes at least one carbon-carbon triple bond, the cycloalkylene group can be referred to as a "cycloalkynylene" group. A cycloalkynylene may have, e.g., four to twenty carbons in the cyclic portion of the
cycloalkynylene (e.g., a C4-C7, C4-C8, C4-C9. C4-C1 0, C4-C1 1 , C4-C12, C4-C14, C4-C16, C4-C18, or C8-C20 cycloalkynylene). A cycloalkylene group can be substituted by the groups typically suitable as substituents for alkyl, alkenyl and alkynyl groups as set forth herein. Heterocycloalkylene refers to a cycloalkylene group including one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms, e.g., N, O, and S. Examples of cycloalkylenes include, but are not limited to, cyclopropylene and cyclobutylene. A tetrahydrofuran may be considered as a heterocycloalkylene.
The term "arylene," as used herein, refers to a multivalent (e.g., divalent or trivalent) aryl group linking together multiple (e.g., two or three) parts of a compound. For example, one carbon within the arylene group may be linked to one part of the compound, while another carbon within the arylene group may be linked to another part of the compound. An arylene may have, e.g., five to fifteen carbons in the aryl portion of the arylene (e.g., a C5-C6, C5-C7, C5-C8, C5-C9. C5-C10, C5-C1 1 , C5-C12, C5-C13, C5- C14, or C5-C15 arylene). An arylene group can be substituted by the groups typically suitable as substituents for alkyl, alkenyl and alkynyl groups as set forth herein. Heteroarylene refers to an aromatic group including one or more, e.g., 1 -4, 1 -3, 1 , 2, 3, or 4, heteroatoms, e.g., N, O, and S. A heteroarylene group may have, e.g., two to fifteen carbons (e.g., a C2-C3, C2-C4, C2-C5, C2-C6, C2-C7, C2-C8, C2- C9. C2-C10, C2-C1 1 , C2-C12, C2-C13, C2-C14, or C2-C15 heteroarylene).
The term "optionally substituted," as used herein, refers to having 0, 1 , or more substituents, such as 0-25, 0-20, 0-10 or 0-5 substituents. Substituents include, but are not limited to, alkyl, alkenyl, alkynyl, aryl, alkaryl, acyl, heteroaryl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroalkaryl, halogen, oxo, cyano, nitro, amino, alkamino, hydroxy, alkoxy, alkanoyl, carbonyl, carbamoyl, guanidinyl, ureido, amidinyl, any of the groups or moieties described above, and hetero versions of any of the groups or moieties described above. Substituents include, but are not limited to, F, CI, methyl, phenyl, benzyl, OR, NR2, SR, SOR, S02R, OCOR, NRCOR, NRCONR2, NRCOOR, OCONR2, RCO, COOR, alkyl-OOCR, SO3R, CONR2, SO2NR2, NRSO2NR2, CN, CF3, OCF3, SiR3, and NO2, wherein each R is, independently, H, alkyl, alkenyl, aryl, heteroalkyl, heteroalkenyl, or heteroaryl, and wherein two of the optional substituents on the same or adjacent atoms can be joined to form a fused, optionally substituted aromatic or nonaromatic, saturated or unsaturated ring which contains 3-8 members, or two of the optional substituents on the same atom can be joined to form an optionally substituted aromatic or nonaromatic, saturated or unsaturated ring which contains 3-8 members.
An optionally substituted group or moiety refers to a group or moiety (e.g., any one of the groups or moieties described above) in which one of the atoms (e.g., a hydrogen atom) is optionally replaced with another substituent. For example, an optionally substituted alkyl may be an optionally substituted methyl, in which a hydrogen atom of the methyl group is replaced by, e.g., OH. As another example, a substituent on a heteroalkyl or its divalent counterpart, heteroalkylene, may replace a hydrogen on a carbon or a hydrogen on a heteroatom such as N. For example, the hydrogen atom in the
group -R-NH-R- may be substituted with an alkamide substituent, e.g., -R-N[(CH2C(0)N(CH3)2]-R.
Generally, an optional substituent is a noninterfering substituent. A "noninterfering substituent" refers to a substituent that leaves the ability of the compounds described herein (e.g., compounds of any one of formulas (l)-(XXXXX)) to either bind to lipopolysaccharides (LPS) or to kill or inhibit the growth of Gram- negative bacteria qualitatively intact. Thus, in some embodiments, the substituent may alter the degree of such activity. However, as long as the compound retains the ability to bind to LPS and/or to kill or inhibit the growth of Gram-negative bacteria, the substituent will be classified as "noninterfering." In some aspects, a noninterfering substituent leaves the ability of a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) to kill or inhibit the growth of Gram-negative bacteria qualitatively intact as determined by measuring the minimum inhibitory concentration (MIC) against at least one Gram- negative bacteria as known in the art, wherein the MIC is 32 μg/mL or less. In some aspects, a noninterfering substituent leaves the ability of a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) to bind to lipopolysaccharides (LPS) from the cell membrane of Gram-negative bacteria qualitatively intact, as determined by an LPS binding assay (e.g., see Example 103), wherein the compound shows a value of about 1 0% or greater displacement of a fluorogenic substrate at 250 μΜ of the compound.
The term "hetero," when used to describe a chemical group or moiety, refers to having at least one heteroatom that is not a carbon or a hydrogen, e.g., N, O, and S. Any one of the groups or moieties described above may be referred to as hetero if it contains at least one heteroatom. For example, a heterocycloalkyi, heterocycloalkenyl, or heterocycloalkynyl group refers to a cycloalkyi, cycloalkenyl, or cycloalkynyl group that has one or more heteroatoms independently selected from, e.g., N, O, and S. An example of a heterocycloalkenyl group is a maleimido. For example, a heteroaryl group refers to an aromatic group that has one or more heteroatoms independently selected from, e.g., N, O, and S. One or more heteroatoms may also be included in a substituent that replaced a hydrogen atom in a group or moiety as described herein. For example, in an optionally substituted heteroaryl group, if one of the hydrogen atoms in the heteroaryl group is replaced with a substituent (e.g., methyl), the substituent may also contain one or more heteroatoms (e.g., methanol).
The term "acyl," as used herein, refers to a group having the structure:
Figure imgf000104_0001
, wherein Rz is an optionally substituted alkyl, alkenyl, alkynyl, cycloalkyi, cycloalkenyl, cycloalkynyl, aryl, alkaryl, alkamino, heteroalkyl, heteroalkenyl, heteroalkynyl, heterocycloalkyi, heterocycloalkenyl,
heterocycloalkynyl, heteroaryl, heteroalkaryl, or heteroalkamino.
The term "halo" or "halogen," as used herein, refers to any halogen atom, e.g., F, CI, Br, or I. Any one of the groups or moieties described herein may be referred to as a "halo moiety" if it contains at least one halogen atom, such as haloalkyl.
The term "hydroxyl," as used herein, represents an -OH group.
The term "oxo," as used herein, refers to a substituent having the structure =0, where there is a double bond between an atom and an oxygen atom.
O
The term "carbonyl," as used herein, refers to a group having the structure
s
The term "thiocarbonyl," as used herein, refers to a group having the structure: ^ ^
O
i I I l-o-p I -c
The term "phosphate," as used herein, represents the group having the structure: O"
O
¾ I I
|— p-o-
The term "phosphoryl," as used herein, represents the group having the structure: OR
O
4 I I <,
i-o-p-o-
R
The term "sulfonyl," as used herein, represents the group having the structure V NR
The term "imino," as used herein, represents the group having the structure: ^ ^ , wherein R is an optional substituent.
The term "AV-protecting group," as used herein, represents those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used AV-protecting groups are disclosed in Greene, "Protective Groups in Organic Synthesis," 5th Edition (John Wiley & Sons, New York, 2014), which is incorporated herein by reference. AV-protecting groups include, e.g., acyl, aryloyl, and carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, a-chlorobutyryl, benzoyl, carboxybenzyl (CBz), 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L or D, L-amino acid residues such as alanine, leucine, phenylalanine;
sulfonyl-containing groups such as benzenesulfonyl and p-toluenesulfonyl ; carbamate forming groups such as benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p- nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3,4- dimethoxybenzyloxycarbonyl, 3,5-dimethoxybenzyl oxycarbonyl, 2,4-dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4,5-dimethoxybenzyloxycarbonyl,
3,4,5-trimethoxybenzyloxycarbonyl, 1 -(p-biphenylyl)-l -methylethoxycarbonyl, α,α-dimethyl- 3,5-dimethoxybenzyloxycarbonyl, benzhydryloxy carbonyl, t-butyloxycarbonyl (BOC),
diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2,-trichloroethoxycarbonyl, phenoxycarbonyl, 4-nitrophenoxy carbonyl, fluorenyl-9-methoxycarbonyl (Fmoc), cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl, and phenylthiocarbonyl; alkaryl groups such as benzyl, triphenylmethyl, and benzyloxymethyl ; and silyl groups such as trimethylsilyl.
The term "amino acid," as used herein, means naturally occurring amino acids and non-naturally occurring amino acids.
The term "naturally occurring amino acids," as used herein, means amino acids including Ala,
Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, lie, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, and Val.
The term "non-naturally occurring amino acid," as used herein, means an alpha amino acid that is not naturally produced or found in a mammal. Examples of non-naturally occurring amino acids include D- amino acids; an amino acid having an acetylaminomethyl group attached to a sulfur atom of a cysteine; a pegylated amino acid; the omega amino acids of the formula NH2(CH2)nCOOH where n is 2-6, neutral nonpolar amino acids, such as sarcosine, t-butyl alanine, t-butyl glycine, N-methyl isoleucine, and norleucine; oxymethionine; phenylglycine; citrulline; methionine sulfoxide; cysteic acid; ornithine;
diaminobutyric acid; 3-aminoalanine; 3-hydroxy-D-proline; 2,4-diaminobutyric acid; 2-aminopentanoic acid; 2-aminooctanoic acid, 2-carboxy piperazine; piperazine-2-carboxylic acid, 2-amino-4-phenylbutanoic acid; 3-(2-naphthyl)alanine, and hydroxyproline. Other amino acids are a-aminobutyric acid, a-amino-a- methylbutyrate, aminocyclopropane-carboxylate, aminoisobutyric acid, aminonorbornyl-carboxylate, L- cyclohexylalanine, cyclopentylalanine, L-N-methylleucine, L-N-methylmethionine, L-N-methylnorvaline, L- N-methylphenylalanine, L-N-methylproline, L-N-methylserine, L-N-methyltryptophan, D-ornithine, L-N- methylethylglycine, L-norleucine, a-methyl-aminoisobutyrate, a-methylcyclohexylalanine, D-a- methylalanine, D-a-methylarginine, D-a-methylasparagine, D-a-methylaspartate, D-a-methylcysteine, D- a-methylglutamine, D-a-methylhistidine, D-a-methylisoleucine, D-a-methylleucine, D-a-methyllysine, D-a- methylmethionine, D-a-methylornithine, D-a-methylphenylalanine, D-a-methylproline, D-a-methylserine, D-N-methylserine, D-a-methylthreonine, D-a-methyltryptophan, D-a-methyltyrosine, D-a-methylvaline, D- N-methylalanine, D-N-methylarginine, D-N-methylasparagine, D-N-methylaspartate, D-N-methylcysteine, D-N-methylglutamine, D-N-methylglutamate, D-N-methylhistidine, D-N-methylisoleucine, D-N- methylleucine, D-N-methyllysine, N-methylcyclohexylalanine, D-N-methylornithine, N-methylglycine, N- methylaminoisobutyrate, N-(1 -methylpropyl)glycine, N-(2-methylpropyl)glycine, D-N-methyltryptophan, D- N-methyltyrosine, D-N-methylvaline, γ-aminobutyric acid, L-t-butylglycine, L-ethylglycine, L- homophenylalanine, L-a-methylarginine, L-a-methylaspartate, L-a-methylcysteine, L-a-methylglutamine, L-a-methylhistidine, L-a-methylisoleucine, L-a-methylleucine, L-a-methylmethionine, L-a-methylnorvaline, L-a-methylphenylalanine, L-a-methylserine, L-a-methyltryptophan, L-a-methylvaline, N-(N-(2,2- diphenylethyl) carbamylmethylglycine, 1 -carboxy-1 -(2,2-diphenyl-ethylamino) cyclopropane, 4- hydroxyproline, ornithine, 2-aminobenzoyl (anthraniloyl), D-cyclohexylalanine, 4-phenyl-phenylalanine, L- citrulline, a-cyclohexylglycine, L-1 ,2,3,4-tetrahydroisoquinoline-3-carboxylic acid, L-thiazolidine-4- carboxylic acid, L-homotyrosine, L-2-furylalanine, L-histidine (3-methyl), N-(3-guanidinopropyl)glycine, O- methyl-L-tyrosine, O-glycan-serine, meta-tyrosine, nor-tyrosine, L-N,N',N"-trimethyllysine, homolysine, norlysine, N-glycan asparagine, 7-hydroxy-1 ,2,3,4-tetrahydro-4-fluorophenylalanine, 4- methylphenylalanine, bis-(2-picolyl)amine, pentafluorophenylalanine, indoline-2-carboxylic acid, 2- aminobenzoic acid, 3-amino-2-naphthoic acid, asymmetric dimethylarginine, L-tetrahydroisoquinoline-1 - carboxylic acid, D-tetrahydroisoquinoline-1 -carboxylic acid, 1 -amino-cyclohexane acetic acid, D/L- allylglycine, 4-aminobenzoic acid, 1 -amino-cyclobutane carboxylic acid, 2 or 3 or 4-aminocyclohexane carboxylic acid, 1 -amino-1 -cyclopentane carboxylic acid, 1 -aminoindane-1 -carboxylic acid, 4-amino- pyrrolidine-2-carboxylic acid, 2-aminotetraline-2-carboxylic acid, azetidine-3-carboxylic acid, 4-benzyl- pyrrolidine-2-carboxylic acid, tert-butylglycine, b-(benzothiazolyl-2-yl)-alanine, b-cyclopropyl alanine, 5,5- dimethyl-1 ,3-thiazolidine-4-carboxylic acid, (2R,4S)4-hydroxypiperidine-2-carboxylic acid, (2S,4S) and (2S,4R)-4-(2-naphthylmethoxy)-pyrrolidine-2-carboxylic acid, (2S,4S) and (2S,4R)4-phenoxy-pyrrolidine- 2-carboxylic acid, (2R,5S)and(2S,5R)-5-phenyl-pyrrolidine-2-carboxylic acid, (2S,4S)-4-amino-1 -benzoyl- pyrrolidine-2-carboxylic acid, t-butylalanine, (2S,5R)-5-phenyl-pyrrolidine-2-carboxylic acid, 1 - aminomethyl-cyclohexane-acetic acid, 3,5-bis-(2-amino)ethoxy-benzoic acid, 3,5-diamino-benzoic acid, 2- methylamino-benzoic acid, N-methylanthranylic acid, L-N-methylalanine, L-N-methylarginine, L-N- methylasparagine, L-N-methylaspartic acid, L-N-methylcysteine, L-N-methylglutamine, L-N- methylglutamic acid, L-N-methylhistidine, L-N-methylisoleucine, L-N-methyllysine, L-N-methylnorleucine, L-N-methylornithine, L-N-methylthreonine, L-N-methyltyrosine, L-N-methylvaline, L-N-methyl-t- butylglycine, L-norvaline, a-methyl-Y-aminobutyrate, 4,4'-biphenylalanine, a-methylcylcopentylalanine, a- methyl-a-napthylalanine, a-methylpenicillamine, N-(4-aminobutyl)glycine, N-(2-aminoethyl)glycine, N-(3- aminopropyl)glycine, N-amino-a-methylbutyrate, a-napthylalanine, N-benzylglycine, N-(2- carbamylethyl)glycine, N-(carbamylmethyl)glycine, N-(2-carboxyethyl)glycine, N-(carboxymethyl)glycine, N-cyclobutylglycine, N-cyclodecylglycine, N-cycloheptylglycine, N-cyclohexylglycine, N-cyclodecylglycine, N-cylcododecylglycine, N-cyclooctylglycine, N-cyclopropylglycine, N-cycloundecylglycine, N-(2,2- diphenylethyl)glycine, N-(3,3-diphenylpropyl)glycine, N-(3-guanidinopropyl)glycine, N-(1 - hydroxyethyl)glycine, N-(hydroxyethyl))glycine, N-(imidazolylethyl))glycine, N-(3-indolylyethyl)glycine, N- methyl-Y-aminobutyrate, D-N-methylmethionine, N-methylcyclopentylalanine, D-N-methylphenylalanine, D-N-methylproline, D-N-methylthreonine, N-(1 -methylethyl)glycine, N-methyl-napthylalanine, N- methylpenicillamine, N-(p-hydroxyphenyl)glycine, N-(thiomethyl)glycine, penicillamine, L-a-methylalanine, L-a-methylasparagine, L-a-methyl-t-butylglycine, L-methylethylglycine, L-a-methylglutamate, L-a- methylhomophenylalanine, N-(2-methylthioethyl)glycine, L-a-methyllysine, L-a-methylnorleucine, L-a- methylornithine, L-a-methylproline, L-a-methylthreonine, L-a-methyltyrosine, L-N-methyl- homophenylalanine, N-(N-(3,3-diphenylpropyl) carbamylmethylglycine, L-pyroglutamic acid, D- pyroglutamic acid, O-methyl-L-serine, O-methyl-L-homoserine, 5-hydroxylysine, a-carboxyglutamate, phenylglycine, L-pipecolic acid (homoproline), L-homoleucine, L-lysine (dimethyl), L-2-naphthylalanine, L- dimethyldopa or L-dimethoxy-phenylalanine, L-3-pyridylalanine, L-histidine (benzoyloxymethyl), N- cycloheptylglycine, L-diphenylalanine, O-methyl-L-homotyrosine, L-p-homolysine, O-glycan-threoine, Ortho-tyrosine, L-N,N'-dimethyllysine, L-homoarginine, neotryptophan, 3-benzothienylalanine, isoquinoline-3-carboxylic acid, diaminopropionic acid, homocysteine, 3,4-dimethoxyphenylalanine, 4- chlorophenylalanine, L-1 ,2,3,4-tetrahydronorharman-3-carboxylic acid, adamantylalanine, symmetrical dimethylarginine, 3-carboxythiomorpholine, D-1 ,2,3,4-tetrahydronorharman-3-carboxylic acid, 3- aminobenzoic acid, 3-amino-1 -carboxymethyl-pyridin-2-one, 1 -amino-1 -cyclohexane carboxylic acid, 2- aminocyclopentane carboxylic acid, 1 -amino-1 -cyclopropane carboxylic acid, 2-aminoindane-2-carboxylic acid, 4-amino-tetrahydrothiopyran-4-carboxylic acid, azetidine-2-carboxylic acid, b-(benzothiazol-2-yl)- alanine, neopentylglycine, 2-carboxymethyl piperidine, b-cyclobutyl alanine, allylglycine, diaminopropionic acid, homo-cyclohexyl alanine, (2S,4R)- 4-hydroxypiperidine-2-carboxylic acid, octahydroindole-2- carboxylic acid, (2S,4R) and (2S,4R)-4-(2-naphthyl), pyrrolidine-2-carboxylic acid, nipecotic acid, (2S,4R)and (2S,4S)-4-(4-phenylbenzyl) pyrrolidine-2-carboxylic acid, (3S)-1 -pyrrolidine-3-carboxylic acid, (2S,4S)-4-tritylmercapto-pyrrolidine-2-carboxylic acid, (2S,4S)-4-mercaptoproline, t-butylglycine, N,N- bis(3-aminopropyl)glycine, 1 -amino-cyclohexane-1 -carboxylic acid, N-mercaptoethylglycine, and selenocysteine. In some embodiments, amino acid residues may be charged or polar. Charged amino acids include alanine, lysine, aspartic acid, or glutamic acid, or non-naturally occurring analogs thereof. Polar amino acids include glutamine, asparagine, histidine, serine, threonine, tyrosine, methionine, or tryptophan, or non-naturally occurring analogs thereof.
It is specifically contemplated that in some embodiments, a terminal amino group in the amino acid may be an amido group or a carbamate group.
The term "antibacterial agent," as used herein, refers to an agent that is used in addition to one or more of the compounds described herein (e.g., compounds of any one of formulas (l)-(XXXXX)) in methods of treating a bacterial infection (e.g., Gram-negative bacterial infection) and/or preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria. An antibacterial agent may be an agent that prevents the entrance of a bacteria (e.g., a Gram-negative bacteria) into a subject's cells, tissues, or organs, inhibits the growth of a bacteria (e.g., a Gram-negative bacteria) in a subject's cells, tissues, or organs, and/or kills a bacteria (e.g., a Gram-negative bacteria) that is inside a subject's cells, tissues, or organs. Examples of antibacterial agents are described in detail further herein. In some embodiments, an antibacterial agent used in addition to a compound described herein is linezolid or tedizolid (e.g. , tedizolid phosphate).
The term "bacterial infection," as used herein, refers to the invasion of a subject's cells, tissues, and/or organs by bacteria (e.g., Gram-negative bacteria), thus, causing an infection. In some
embodiments, the bacteria may grow, multiply, and/or produce toxins in the subject's cells, tissues, and/or organs. In some embodiments, a bacterial infection can be any situation in which the presence of a bacterial population(s) is latent within or damaging to a host body. Thus, a subject is "suffering" from a bacterial infection when a latent bacterial population is detectable in or on the subject's body, an excessive amount of a bacterial population is present in or on the subject's body, or when the presence of a bacterial population(s) is damaging the cells, tissues, and/or organs of the subject.
The term "protecting against," as used herein, refers to preventing a subject from developing a bacterial infection (e.g., a Gram-negative bacterial infection) or decreasing the risk that a subject may develop a bacterial infection (e.g., a Gram-negative bacterial infection). Prophylactic drugs used in methods of protecting against a bacterial infection in a subject are often administered to the subject prior to any detection of the bacterial infection. In some embodiments of methods of protecting against a bacterial infection, a subject (e.g., a subject at risk of developing a bacterial infection) may be
administered a compound described herein (e.g., a compound having any one of formulas (l)-(XXXXX)) to prevent the bacterial infection development or decrease the risk of the bacterial infection development.
The term "treating" or "to treat," as used herein, refers to a therapeutic treatment of a bacterial infection (e.g., a Gram-negative bacterial infection) in a subject. In some embodiments, a therapeutic treatment may slow the progression of the bacterial infection, improve the subject's outcome, and/or eliminate the infection. In some embodiments, a therapeutic treatment of a bacterial infection (e.g., a Gram-negative bacterial infection) in a subject may alleviate or ameliorate of one or more symptoms or conditions associated with the bacterial infection, diminish the extent of the bacterial infection, stabilize (i.e., not worsening) the state of the bacterial infection, prevent the spread of the bacterial infection, and/or delay or slow the progress of the bacterial infection, as compare the state and/or the condition of the bacterial infection in the absence of the therapeutic treatment.
The phrase "LPS-induced nitric oxide (NO) production from a macrophage," as used herein, refers to the ability of the lipopolysaccharides (LPS) in Gram-negative bacteria to activate a macrophage and induce NO production from the macrophage. NO production from a macrophage in response to LPS is a signal of macrophage activation, which may lead to sepsis in a subject, e.g., a Gram-negative bacteria infected subject. The disclosure features compounds (e.g., compounds of any one of formulas (l)-(XXXXX)) that are able to bind to LPS in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, thus neutralizing an immune response to LPS (see, e.g., Example 1 1 0). NO production from a macrophage may be measured using available techniques in the art, e.g., a Griess assay, as demonstrated in Example 1 10.
The term "resistant strain of bacteria," as used herein, refers to a strain of bacteria (e.g., Gram- negative or Gram-positive bacteria) that is refractory to treatment with an antibiotic, such as an antibiotic described in Section V of Detailed Description. Antibiotics that a strain of bacteria is resistant to do not include the compounds described herein (e.g., compounds of any one of formulas (l)-(XXXXX)). Resistance may arise through natural resistance in certain types of bacteria, spontaneous random genetic mutations, and/or by inter- or intra-species horizontal transfer of resistance genes. In some embodiments, a resistant strain of bacteria contains a mcr- 1 gene, a mcr-2 gene, and/or a mcr-3 gene. In some embodiments, a resistant strain of bacteria contains a chromosomal mutation conferring polymyxin resistance. In some embodiments, a resistant strain of bacteria contains a mcr- 1 gene, a mcr-2 gene, and/or a mcr-3 gene in combination with other antibiotic resistance genes. In some embodiments, a resistant strain of bacteria is a resistant strain of E. co//' (e.g., E. coli BAA-2469).
The term "activating an immune cell," as used herein, refers to the ability of a compound to directly or indirectly bind to an immune cell to produce an effective immune response. The ability of a compound to directly or indirectly bind to an immune cell to produce an effective immune response may be quantified by measuring the concentration of the compound at which such immune response is produced. In some embodiments, the concentration of a compound that binds to an immune cell receptor such as dectin-1 or binds to an antibody (e.g., anti-aGal or anti-aRha antibody, which then binds to an immune cell) to trigger an effective immune response may be less than or equal to 10,000 nM as measured in accordance with, e.g., an enzyme-linked immunosorbent assay (ELISA), as in Examples 107 and 108. For instance, an aGal epitope, that binds to an antibody, such as an anti-aGal antibody, may be detected using an ELISA. In an ELISA, a compound containing a particular monosaccharide or oligosaccharide moiety may be immobilized on a support or surface using conventional techniques in the art. After the compound is immobilized to the surface, an antibody that is specific for the particular monosaccharide or
oligosaccharide moiety in the compound is applied over the surface so it is captured by the compound through binding to the monosaccharide or oligosaccharide moiety in the compound. The antibody is often linked to an enzyme (e.g., horseradish peroxidase) for subsequent signal amplification. During signal amplification, the enzyme's substrate (e.g., 3,3'-diaminobenzidine) is added to produce a measurable signal (e.g., color change). In some embodiments, the antibody itself can be detected using a secondary antibody, which is linked to an enzyme.
In some embodiments, the concentration of a compound that binds to an immune cell receptor such as dectin-1 or binds to an antibody (e.g., anti-aGal or anti-aRha antibody, which then binds to an immune cell) to trigger an effective immune response may be less than or equal to 1000 nM or less than or equal to 100 nM as measured in accordance with an ELISA.
The term "innate immune receptor," as used herein, refers to a natural receptor, such as a natural receptor on an immune cell, that binds to a carbohydrate (e.g., a monosaccharide or oligosaccharide moiety) or an optionally substituted carbohydrate and causes a response in the immune system. In some embodiments, an innate immune receptor binds to the monosaccharide or oligosaccharide moiety of the compounds described herein (e.g., compounds of any one of formulas (l)-(XXXXX)). In some embodiments, an innate immune receptor binds to a moiety in Table 2A or 2B.
The term "natural antibody," as used herein, refers to a naturally existing antibody in the circulation of a mammal (e.g., a human) that has not been previously exposed to deliberate immunization. In some embodiments, a natural antibody is an antibody of the immunoglobulin M (IgM) isotype. In some embodiments, a natural antibody binds to the monosaccharide or oligosaccharide moiety of the compounds described herein (e.g., compounds of any one of formulas (l)-(XXXXX)). In some embodiments, a natural antibody binds to a moiety in Table 2A or 2B. In some embodiments, a natural antibody is anti-aGal antibobody or anti-aRha antibody.
The term "subject," as used herein, can be a human, non-human primate, or other mammal, such as but not limited to dog, cat, horse, cow, pig, turkey, goat, fish, monkey, chicken, rat, mouse, and sheep. The term "substantially simultaneously," as used herein, refers to two or more events that occur at the same time or within a narrow time frame of each other. As disclosed herein, a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) and an antibacterial agent (e.g., linezolid or tedizolid) may be administered substantially simultaneously, which means that the compound and the antibacterial agent are administered together (e.g., in one pharmaceutical composition) or separately but within a narrow time frame of each other, e.g., within 10 minutes, e.g., 1 0, 9, 8, 7, 6, 5, 4, 3, 2, or 1 minute, or 45, 30, 15, or 1 0 seconds of each other.
The term "therapeutically effective amount," as used herein, refers to an amount, e.g., pharmaceutical dose, effective in inducing a desired effect in a subject or in treating a subject having a condition or disorder described herein (e.g., a bacterial infection (e.g., a Gram-negative bacterial infection)). It is also to be understood herein that a "therapeutically effective amount" may be interpreted as an amount giving a desired therapeutic and/or preventative effect, taken in one or more doses or in any dosage or route, and/or taken alone or in combination with other therapeutic agents (e.g., an antibacterial agent described herein). For example, in the context of administering a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) that is used for the treatment of a bacterial infection, an effective amount of a compound is, for example, an amount sufficient to prevent, slow down, or reverse the progression of the bacterial infection as compared to the response obtained without administration of the compound.
As used herein, the term "pharmaceutical composition" refers to a medicinal or pharmaceutical formulation that contains at least one active ingredient (e.g., a compound of any one of formulas (I)- (XXXXX)) as well as one or more excipients and diluents to enable the active ingredient suitable for the method of administration. The pharmaceutical composition of the present disclosure includes pharmaceutically acceptable components that are compatible with a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)).
As used herein, the term "pharmaceutically acceptable carrier" refers to an excipient or diluent in a pharmaceutical composition. For example, a pharmaceutically acceptable carrier may be a vehicle capable of suspending or dissolving the active compound (e.g., a compound of any one of formulas (I)- (XXXXX)). The pharmaceutically acceptable carrier must be compatible with the other ingredients of the formulation and not deleterious to the recipient. In the present disclosure, the pharmaceutically acceptable carrier must provide adequate pharmaceutical stability to a compound described herein. The nature of the carrier differs with the mode of administration. For example, for oral administration, a solid carrier is preferred; for intravenous administration, an aqueous solution carrier (e.g., WFI, and/or a buffered solution) is generally used.
The term "pharmaceutically acceptable salt," as used herein, represents salts of the compounds described herein (e.g., compounds of any one of formulas (l)-(XXXXX)) that are, within the scope of sound medical judgment, suitable for use in methods described herein without undue toxicity, irritation, and/or allergic response. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Pharmaceutical Salts: Properties, Selection, and Use (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting the free base group with a suitable organic acid.
The term "about," as used herein, indicates a deviation of ±5%. For example, about 1 0% refers to from 9.5% to 10.5%.
Definitions of abbreviations used in the disclosure are provided in Table A below:
Table A
Figure imgf000111_0001
Other features and advantages of the compounds described herein will be apparent from the following detailed description and the claims.
Description of the Drawings
FIG. 1 is a schematic illustrating a 96-well checkerboard synergy MIC plate layout.
FIG. 2 shows the inhibition of nitric oxide (NO) production by Compound 14.
FIGS. 3A-3H show the time-kill analysis for Compound 14 and colistin (COL), respectively, against E. col 7 ATCC 25922, K. pneumoniae ATCC 43816, P. aeruginosa ATCC 27853, and A. baumannii ATCC 17978 at 0, 1 , 2, 4, and 16X CLSI broth microdilution MIC values.
FIG. 4A shows the membrane permeabilization induced by Compound 14, Compound 54a, and
Compound 59, as determined by NPN signal.
FIG. 4B shows the bacterial cell death induced by Compound 14, Compound 54a, and Compound 59, as quantified by SYTOX green signal. FIGS. 5A and 5B show that Rha mediates binding of human Rha antibody to E. coli ATCC 25922-bound Compound 14, 54a, and Compound 59 in an antigen-specific manner. Bound antibodies were detected using either an anti-human IgG secondary antibody (FIG. 5A) or an anti-human IgM secondary antibody (FIG. 5B).
FIG. 5C shows inhibition of Rha-Ab binding to Compound 14 by L-Rha monosaccharide.
FIG. 6 shows Rha-specific binding of purified rabbit Rha antibodies to bacteria.
FIG. 7 shows Rha-specific binding of purified human Rha antibodies to bacteria.
FIGS. 8A-8F show that the killing of E. coli is enhanced by Compound 14, Compound 54a, and
Compound 59 in human blood from donor 1 and donor 2.
FIGS. 9A-9D show that complement dependent cytotoxicity (CDC) is enhanced by Compound 14, Compund 54a, and Compound 59.
FIG. 10 shows that Compound 14 displays rAb-dependent efficacy in a mouse septicemia model.
FIG. 1 1 shows that Compound 14 displays rAb-dependent efficacy in a neutropenic mouse E. coli thigh infection model.
FIG. 12 shows the titration of rAb in the mouse E. coli septicemia model with a fixed dose of
Compound 14.
FIG. 13 shows the metabolic stability of Compound 14 in rat, monkey and human hepatocytes. FIG. 14 shows the titration of Compound 14 in functional potassium channel assays.
FIG. 15 shows the absorbance values at 450 nm for E. coli incubated with Compound 14 and a control compound (Compound 14 without the monosaccharide portion).
Detailed Description
The disclosure features compounds, compositions, and methods for the treatment of bacterial infections (e.g., Gram-negative bacterial infections). The compounds disclosed herein include dimers of cyclic heptapeptides (e.g., two polymyxin cores) linked to each other through a linker and further conjugated to one or more monosaccharide or oligosaccharide moieties. The dimers of cyclic heptapeptides are linked to each other through a linker and/or one or two peptides (e.g., a peptide including a 1 -5 amino acid residue(s)). In particular, the compounds can be used in the treatment of bacterial infections caused by Gram-negative bacteria. In some embodiments, one or more of the compounds described herein are used in combination with an antibacterial agent (e.g., linezolid or tedizolid (e.g., tedizolid phosphate)).
I. Bacterial Infections
Pathogenic bacteria cause bacterial infections and diseases such as tuberculosis, pneumonia, and foodborne illnesses. Bacteria may be categorized into two major types: Gram-positive bacteria and Gram-negative bacteria. Gram-positive bacteria possess a thick cell wall containing multiple layers of peptidoglycan and teichoic acids, while Gram-negative bacteria have a relatively thin cell wall containing fewer layers of peptidoglycan that are surrounded by a second lipid membrane containing
lipopolysaccharides (LPS) and lipoproteins. LPS, also called endotoxins, are composed of
polysaccharides and lipid A. These differences in bacterial ceil wall structure can produce differences in antibiotic susceptibility. Examples of Gram-positive bacteria include, but are not limited to, bacteria in the genus Streptococcus (e.g., Streptococcus pyogenes), bacteria in the genus Staphylococcus (e.g., Staphylococcus cohnii), bacteria in the genus Corynebacterium (e.g., Corynebacterium auris), bacteria in the genus Listeria (e.g., Listeria grayi), bacteria in the genus Bacillus (e.g., Bacillus aerius), and bacteria in the genus Clostridium (e.g., Clostridium acetium). Examples of Gram-negative bacteria include, but are not limited to, bacteria in the genus Escherichia (e.g., Escherichia coli), bacteria in the genus Klebsiella (e.g., Klebsiella granulomatis, Klebsiella oxytoca, Klebsiella pneumoniae, Klebsiella terrigena, and Klebsiella variicola), bacteria in the genus Acinetobacter (e.g., Acinetobacter baumannii,
Acinetobacter calcoaceticus, Acinetobacter kookii, and Acinetobacter junii), bacteria in the genus Pseudomonas (e.g., Pseudomonas aeruginosa), bacteria in the genus Neisseria (e.g., Neisseria gonorrhoeae), bacteria in the genus Yersinia (e.g., Yersinia pestis), bacteria in the genus Vibrio (e.g., Vibrio cholerae), bacteria in the genus Campylobacter (e.g., Campylobacter jejuni), and bacteria in the genus Salmonella (e.g., Salmonella enterica).
Bacteria may evolve to become more or fully resistant to antibiotics. Resistance may arise through natural resistance in certain types of bacteria, spontaneous random genetic mutations, and/or by inter- or intra-species horizontal transfer of resistance genes. Resistant bacteria are increasingly difficult to treat, requiring alternative medications or higher doses, which may be more costly or more toxic. Bacteria resistant to multiple antibiotics are referred to as multidrug resistant (MDR) bacteria. For example, the mcr- 1 gene encodes a phosphoethanolamine transferase (MCR-1 ) which confers resistance to colistin, a natural polymyxin, through modification of LPS. This is the first known horizontally- transferable resistance determinant for the polymyxin class of antibiotics. The mcr- 1 gene has also been found in bacterial strains which already possess resistance to other classes of antibiotics, such as in carbapenem-resistant Enterobacteriaceae (CRE). An mcr- 1 resistance plasmid refers to a bacterial plasmid that carries mcr- 1 alone or in combination with other antibiotic resistance genes. A mcr- 1 resistance plasmid refers to a bacterial plasmid that carries one or more antibiotic resistance genes. Examples of mcr- 1 resistance plasmids include, but are not limited to, pHNSHP45, pMR0516mcr, pESTMCR, pAF48, pAF23, pmcr1 -lncX4, pmcr1 -lncl2, pA31 -12, pVT553, plCBEC72Hmcr, pE15004, pE1 5015, and pE1 5017.
In another example, the mcr-2 gene also confers resistance to colistin. The mcr-2 gene was identified in porcine and bovine colistin-resistance E.coli that did not contain mcr- 1 (Xavier et al., Euro Si/rve/7/ 21 (27), 2016). The mcr-2 gene is a 1 ,617 bp phspoethanolamine transferase harbored on an lncX4 plasmid. The mcr-2 gene has 76.7% nucleotide identity to mcr- 1. Analysis of mcr-2 harboring plasmids from E.coli isolates shows that the mobile element harboring mcr-2 is an IS element of the IS1595 superfamily, which are distinguished by the presence of an ISXC -like transposase domain (Xavier et al., supra). The MCR-2 protein was predicted to have two domains, with domain 1 (1 -229 residues) as a transporter and domain 2 (230-538 residues) as a transferase domain. An mcr-2 resistance plasmid refers to a bacterial plasmid that carries mcr-2 alone or in combination with other antibiotic resistance genes. A mcr-2 resistance plasmid refers to a bacterial plasmid that carries one or more antibiotic resistance genes. Mcr-2 resistance plasmids include, but are not limited to, pKP37-BE and pmcr2-lncX4. In a further example, the mcr-3 gene also confers resistance to colistin. The mcr-3 gene is considered to include variants which encode an MCR-3 protein that confers resistance to cholistin, such as mcr-3 which encodes a protein having an amino acid substitution at V413A. The mcr-3 gene was identified in cholistin-resistant E.coli isolated from swine (Clemente et al., 27th ECCMID, Vienna, Austria, 2017). The mcr3 gene is harbored on an lncX4 plasmid. Analysis of mcr-3 harboring plasmids from E.coli isolates shows that the mobile element harboring mcr-3 is the IS26 element. A mcr-3 resistance plasmid refers to a bacterial plasmid that carries one or more antibiotic resistance genes. Furthermore, resistant strain E. coli BAA-2469 possesses the New Delhi metallo-p-lactamase (NDM-1 ) enzyme, which makes bacteria resistant to a broad range of β-lactam antibiotics. Additionally, E. coli BAA-2469 is also known to be resistatnt to penicillins (e.g., ticarcillin, ticarcillin/clavulanic acid, piperacillin, ampicillin, and ampicillin/sulbactam), cephalosporins (e.g., cefalotin, cefuroxime, cefuroxime, cefotetan, cefpodoxime, cefotaxime, ceftizoxime, cefazolin, cefoxitin, ceftazidime, ceftriaxone, and cefepime), carbapenems (e.g., doripenem, meropenem, ertapenem, imipenem), quinolones (e.g., nalidixic acid, moxifloxacin, norfloxacin, ciprofloxacin, and levofloxacin), aminoglycosides (e.g., amikacin, gentamicin, and tobramycin), and other antibiotics (e.g., tetracycline, tigecycline, nitrofurantoin, aztreonam,
trimethoprim/sulfamethoxazole).
In some embodiments, a resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene, and/or a chromosomal mutation conferring polymyxin resistance. In some embodiments, a resistant strain of bacteria is a resistant strain of E. coli (e.g., E. coli BAA-2469).
II. Compounds of the Disclosure
Provided herein are synthetic compounds useful in the treatment of bacterial infections (e.g., Gram-negative bacterial infections). The compounds disclosed herein include three components: (i) a first cyclic heptapeptide (e.g., a first polymyxin core), (ii) a second cyclic heptapeptide (e.g., a second polymyxin core), and (iii) one or more monosaccharide or oligosaccharide moieties. The first and second cyclic heptapeptides are linked to each other by way of a linker and/or one or two peptides (e.g., each peptide including a 1 -5 amino acid residue(s)). In some aspects, a cyclic heptapeptide or polymyxin core, as used herein, refers to certain compounds that bind to lipopolysaccharides (LPS) in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics. In some aspects, cyclic heptapeptide, as used herein, refers to certain compounds that kill or inhibit the growth of Gram-negative bacteria as determined by measuring the minimum inhibitory concentration (MIC) against at least one Gram-negative bacteria as known in the art, wherein the MIC is 32 μg/mL or less. Cyclic heptapeptides are composed of, at least, amino acid residues, each of which may, independently, have a D- or L- configuration, assembled as a cyclic heptapeptide ring. A cyclic heptapeptide includes seven natural or non-natural amino acid residues attached to each other in a closed ring. The ring contains six bonds formed by linking the carbon in the a- carboxyl group of one amino acid residue to the nitrogen in the a-amino group of the adjacent amino acid residue and one bond formed by linking the carbon in the a-carboxyl group of one amino acid residue to the nitrogen in the γ-amino group in the side chain of the adjacent amino acid residue. For the amino acid residue whose nitrogen in the γ-amino group in the side chain participates in forming the ring, the nitrogen in the a-amino group of this amino acid residue does not participate directly in forming the ring and serves as the linking nitrogen (thus, referred to as the "linking nitrogen" herein) that links one cyclic heptapeptide or polymyxin core to another cyclic heptapeptide or polymyxin core by way of a linker and/or one or two peptides (e.g., a peptide including a 1 -5 amino acid residue(s)). Compounds described herein contain dimers of cyclic heptapeptides (e.g., two polymyxin cores) that are linked to each other at their linking nitrogens through a linker and/or one or two peptides (e.g., a peptide including a 1 -5 amino acid residue(s)), and one or more monosaccharide or oligosaccharide moieties.
In some embodiments, a peptide including one or more (e.g., 1 -5; 1 , 2, 3, 4, or 5) amino acid residues (e.g., natural and/or non-natural amino acid residues) may be covalently attached to the linking nitrogen of the cyclic heptapeptide or the polymyxin core. Cyclic heptapeptides or polymyxin cores may be derived from polymyxins (e.g., naturally existing polymyxins and non-natural polymyxins) and/or octapeptins (e.g., naturally existing octapeptins and non-natural octapeptins). In some embodiments, cyclic heptapeptides may be compounds described in Gallardo-Godoy et al., J. Med. Chem. 59:1068, 2016 (e.g., compounds 1 1 -41 in Table 1 of Gallardo-Godoy et al.), which is incorporated herein by reference in its entirety. Examples of some naturally existing polymyxins and their structures are shown in Table 1 A. Examples of some non-natural polymyxins and their structures are shown in Table 1 B.
Table 1 A. Natural polymyxins and their structures
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Table 1 B. Non-natural polymyxins and their structures (Dab: diaminobutyric acid; Dap: diaminopropionic acid; Orn: ornithine;
Abu: 2-aminobutyric acid; NIe: norleucine)
Figure imgf000117_0002
Without being bound by theory, in some aspects, compounds described herein bind to the cell membrane of Gram-negative bacteria (e.g., bind to LPS in the cell membrane of Gram-negative bacteria) to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics. In some embodiments, the initial association of the compounds with the bacterial cell membrane occurs through electrostatic interactions between the compounds and the anionic LPS in the outer membrane of Gram-negative bacteria, disrupting the arrangement of the cell membrane.
Specifically, compounds described herein may bind to lipid A in the LPS. More specifically, compounds described herein may bind to one or both phosphate groups in lipid A. In some embodiments, antibiotic- resistant bacteria (e.g., antibiotic-resistant, Gram-negative bacteria) has one phosphate group in lipid A. In some embodiments, compounds described herein may bind to multiple Gram-negative bacterial cells at the same time. The binding of the compounds described herein to the LPS may also displace Mg2+ and Ca2+ cations that bridge adjacent LPS molecules, causing, e.g., membrane permeabilization, leakage of cellular molecules, inhibition of cellular respiration, and/or cell death. In some embodiments, in addition to disrupting the membrane of bacterial cells, the monosaccharide or oligosaccharide moieties in the compounds serve as a gradient against which immune cells chemotax to the site of bacterial infection and/or growth.
Compounds provided described herein are described by any one of formulas (l)-(XXXXX). In some embodiments, the first and second cyclic heptapeptides are the same. In some embodiments, the first and second cyclic heptapeptides are different. Compounds described herein may be synthesized using available chemical synthesis techniques in the art. In some embodiments, available functional groups in the first and second cyclic heptapeptides, the linker, and the one or more monosaccharide or oligosaccharide moieties, e.g., amines, carboxylic acids, and/or hydroxyl groups, may be used in making the compounds described herein. For example, the linking nitrogen in a cyclic heptapeptide may form an amide bond with the carbon in a carboxylic acid group in the linker. A peptide including one or more (e.g., 1 -3; 1 , 2, or 3) amino acid residues (e.g., natural and/or non-natural amino acid residues) may be also be covalently attached to the linking nitrogen of the cyclic heptapeptide through forming an amide bond between the carbon in a carboxylic acid group in the peptide and the linking nitrogen. In cases where a functional group is not available for conjugation, a molecule may be derivatized using conventional chemical synthesis techniques that are well known in the art. In some embodiments, the compounds described herein contain one or more chiral centers. The compounds include each of the isolated stereoisomeric forms as well as mixtures of stereoisomers in varying degrees of chiral purity, including racemic mixtures. It also encompasses the various diastereomers, enantiomers, and tautomers that can be formed.
The disclosure provides a compound, or a pharmaceutically acceptable salt thereof, described by formula (I):
(E)n
M2 L' M1
(I)
in which wherein M1 includes a first cyclic heptapeptide containing a linking nitrogen and M2 includes a second cyclic heptapeptide containing a linking nitrogen; each E is, independently, a monosaccharide or oligosaccharide moiety; L' is a linker covalently attached to E and to the linking nitrogen in each of M1 and M2; and n is 1 , 2, 3, or 4 (e.g., 1 or 2). In formula (I), each E can, independently, be connected to an atom in L'.
In some embodiments, L' in the compound described by formula (I) is described by formula (L):
Figure imgf000119_0001
(L)
in which L is a remainder of L'; A1 is a 1 -5 amino acid peptide covalently attached to the linking nitrogen in M1 or is absent; and A2 is a 1 -5 amino acid peptide covalently attached to the linking nitrogen in M2 or is absent.
In some embodiments, the disclosure provides a compound, or a pharmaceutically acceptable salt thereof, described by formula (II):
Figure imgf000119_0002
(II)
in which L is a remainder of L'; n is 1 , 2, 3, or 4 (e.g., 1 or 2); each E is, independently, a monosaccharide or oligosaccharide moiety; each of R1 , R12, R'1 , and R'12 is, independently, a lipophilic moiety, a polar moiety, or H; each of R1 1 , R13, R14, R'1 1 , R'13, and R'14 is, independently, optionally substituted C1 -C5 alkamino, a polar moiety, a positively charged moiety, or H; each of R15 and R'15 is, independently, a lipophilic moiety or a polar moiety; each of R2, R3, R4, R5, R6, R7, R8, R9, R10, R'2, R'3, R'4, R'5, R'6, R'7, R'8, R'9, and R'10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or two R groups on the same or adjacent atoms join to form an optionally substituted 5-8 membered ring; each of a', b', c', a, b, and c is, independently, 0 or 1 ; each of N1 , N2, N3, N4, N'1 , N'2, N'3, and N'4 is a nitrogen atom; and each of C1 , C2, C3, C'1 , C'2, and C'3 is a carbon atom.
In some embodiments, when R2, R3, and C1 together form an optionally substituted 5-8 membered ring or when R3, R4, N1 , and C1 together form an optionally substituted 5-8 membered ring, each of R2, R3, and R4 is, independently, optionally substituted C1 -C20 alkylene or optionally substituted C1 -C20 heteroalkylene.
In some embodiments, when R5, R6, and C2 together form an optionally substituted 5-8 membered ring or when R6, R7, N2, and C2 together form an optionally substituted 5-8 membered ring, each of R5, R6, and R7 is, independently, optionally substituted C1 -C20 alkylene or optionally substituted C1 -C20 heteroalkylene.
In some embodiments, when R8, R9, and C3 together form an optionally substituted 5-8 membered ring or when R9, R10, N3, and C3 together form an optionally substituted 5-8 membered ring, each of R8, R9, and R10 is, independently, optionally substituted C1 -C20 alkylene or optionally substituted C1 -C20 heteroalkylene.
In some embodiments, when R'2, R'3, and C'1 together form an optionally substituted 5-8 membered ring or when R'3, R'4, N'1 , and C'1 together form an optionally substituted 5-8 membered ring, each of R'2, R'3, and R'4 is, independently, optionally substituted C1 -C20 alkylene or optionally substituted C1 -C20 heteroalkylene.
In some embodiments, when R'5, R'6, and C'2 together form an optionally substituted 5-8 membered ring or when R'6, R'7, N'2, and C'2 together form an optionally substituted 5-8 membered ring, each of R'5, R'6, and R'7 is, independently, optionally substituted C1 -C20 alkylene or optionally substituted C1 -C20 heteroalkylene.
In some embodiments, when R'8, R'9, and C'3 together form an optionally substituted 5-8 membered ring or when R'9, R'10, N'3, and C'3 together form an optionally substituted 5-8 membered ring, each of R'8, R'9, and R'10 is, independently, optionally substituted C1 -C20 alkylene or optionally substituted C1 -C20 heteroalkylene.
In some embodiments, the compound of formula (II) has at least one optionally substituted 5-8 membered ring formed by joining (i) R2, R3, and C1 ; (ii) R3, R4, N1 , and C1 ; (iii) R5, R6, and C2; (iv) R6, R7, N2, and C2; (v) R8, R9, and C3; (vi) R9, R10, N3, and C3; (vii) R'2, R'3, and C'1 ; (viii) R'3, R'4, N'1 , and C'1 ; (ix) R'5, R'6, and C'2; (x) R'6, R'7, N'2, and C'2; (xi) R'8, R'9, and C'3; or (xii) R'9, R'10, N'3, and C'3.
In some embodiments, when a is 1 in the compound of formula (II), (i) each of R2, R3, and R4 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R2, R3, and C1 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R4 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (iii) R3, R4, N1 , and C1 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted heterocycloalkyi comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, and R2 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.
In some embodiments, when b is 1 in the compound of formula (II), (i) each of R5, R6, and R7 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R5, R6, and C2 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R7 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (iii) R6, R7, N2, and C2 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted heterocycloalkyi comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, and R5 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.
In some embodiments, when c is 1 in the compound of formula (II), (i) each of R8, R9, and R10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R8, R9, and C3 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R10 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (iii) R9, R10, N3, and C3 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted heterocycloalkyi comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, and R8 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.
In some embodiments, when a' is 1 in the compound of formula (II), (i) each of R'2, R'3, and R'4 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R'2, R'3, and C'1 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R'4 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (iii) R'3, R'4, N'1 , and C'1 together form a ring (e.g., an optionally substituted 5- 8 membered ring) comprising optionally substituted heterocycloalkyi comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, and R'2 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.
In some embodiments, when b' is 1 in the compound of formula (II), (i) each of R'5, R'6, and R'7 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R'5, R'6, and C'2 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R'7 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (iii) R'6, R'7, N'2, and C'2 together form a ring (e.g., an optionally substituted 5- 8 membered ring) comprising optionally substituted heterocycloalkyi comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, and R'5 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.
In some embodiments, when c' is 1 in the compound of formula (II), (i) each of R'8, R'9, and R'10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R'8, R'9, and C'3 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyi comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R'10 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyi, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (iii) R'9, R'10, N'3, and C'3 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted heterocycloalkyl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, and R'8 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted
cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.
In some embodiments, the disclosure provides a compound, or a pharmaceutically acceptable salt thereof, described by formula (III):
Figure imgf000124_0001
(III)
in which L is a remainder of L'; n is 1 , 2, 3, or 4; each E is, independently, a monosaccharide or oligosaccharide moiety; each of R1 , R12, R'1 , and R'12 is, independently, a lipophilic moiety; each of R1 1 , R13, R14, R'1 1 , R'13, and R'14 is, independently, optionally substituted C1 -C5 alkamino, a polar moiety, or a positively charged moiety; and/or each of R15 and R'15 is, independently, a polar moiety.
In some embodiments, a lipophilic moiety is optionally substituted C1 -C20 alkyl, optionally substituted C5-C15 aryl, optionally substituted C6-C35 alkaryl, or optionally substituted C2-C1 5 heteroaryl. In some embodiments, a lipophilic moiety is C1 -C8 alkyl, methyl substituted C2-C4 alkyl, (C1 - C10)alkylene(C6)aryl, phenyl substituted (C1 -C10)alkylene(C6)aryl, or alkyl substituted C4-C9 heteroaryl. In some embodiments, in particular, a lipophilic moiety is benzyl, isobutyl, sec-butyl, isopropyl, n-propyl, methyl, biphenylmethyl, n-octyl, or methyl substituted indolyl.
In some embodiments, optionally substituted C1 -C5 alkamino is CH2CH2NH2.
In some embodiments, a polar moiety includes a hydroxyl group, a carboxylic acid group, an ester group, or an amide group. In some embodiments, a polar moiety is hydroxyl substituted C1 -C4 alkyl. In some embodiments, a polar moiety is CH(CH3)OH.
In some embodiments, a compound provided herein is described by any one of formulas (IV)- (XXXIII). (E)n
In the compounds described herein, the portion L' in a compound of formula (I) and the portion
(E)n
L in a compound of any one of formulas (ll)-(XV) or (XXXIV)-(XXXXX) indicate that one or more (e.g., 1 , 2, 3, or 4; 1 -2, 1 -3, or 1 -4) monosaccharide or oligosaccharide moieties may be attached to L' or L at any atom(s) within L' or L. E represents a monosaccharide or oligosaccharide moiety. The squiggly line
(E)n (E)n in the portion L' and L is not to be construed as a single bond between one or more
monosaccharide or oligosaccharide moieties and an atom in L' or L. In some embodiments, when n is 1 , one monosaccharide or oligosaccharide moiety may be attached to an atom in L' or L (see, e.g., Compounds 1 -4, 7-21 , and 23-25). In some embodiments, when n is 2, two monosaccharide or oligosaccharide moieties may be attached to two atoms in L' or L (see, e.g., Compounds 5, 6, and 22). As described further herein, a linker in a compound described herein (e.g., L' or L) may be a branched structure. As described further herein, a linker in a compound described herein (e.g., L' or L) may be a multivalent structure, e.g., divalent, trivalent, tetravalent, pentavalent, hexavalent, or heptavalent structure, containing two, three, four, five, six, or seven arms, respectively. In some embodiments when the linker has three or more (e.g., three, four, five, six, or seven) arms, two of the arms may be attached to the first and second cyclic heptapeptides and the remaining arm(s) (e.g., remaining one, two, three, four, or five arms) may be attached to one or more (e.g., 1 , 2, 3, 4, or 5; 1 -2, 1 -3, 1 -4, or 1 -5)
(E)n (E)n monosaccaride or oligosaccharide moieties. Thus, L' or L in the portion L' or L may have multiple arms to attached to multiple monosaccharide or oligosaccharide moieties. For example, in Compounds 5, 6, and 22, the linker portion L' or L has four arms, in which two arms are attached to the first and second cyclic heptapeptides and two arms are attached to the first and second monosaccharide moieties (L-Rha).
The disclosure also provides a compound of formula (XVI):
Figure imgf000125_0001
(XVI)
wherein each A is an independently selected amino acid; L is a linker that, when m is 2, 3, 4, or 5, is bound to any of A; each E is independently selected from a monosaccharide or an oligosaccharide; m is 0, 1 , 2, 3, 4, or 5; n is 1 , 2, 3, 4, or 5; and Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof.
The disclosure also provides a compound of formula (XVII):
Figure imgf000126_0001
(XVII)
wherein each A1 and A2 is an independently selected amino acid; L is a linker that, when m is 1 , 2, 3, 4, or 5, is bound to a nitrogen atom in any A1 and a nitrogen atom in any A2; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; n is 1 , 2, 3, 4, or 5; and Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof.
The disclosure also provides a compound of formula (XVIII):
Figure imgf000126_0002
(XVIII)
wherein each A1 and A2 is an independently selected amino acid; X is absent or is -CH2CH2C(0)NH-; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; n is 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 10; and e is an integer from 0 to 10; or a
pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XIX):
Figure imgf000127_0001
(XIX)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; n is 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 10; e is an integer from 0 to 10; f is an integer from 0 to 10; and g is an integer from 0 to 10; or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XX):
Figure imgf000127_0002
(XX)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 1 0; e is an integer from 0 to 10; f is an integer from 0 to 10; and g is an integer from 0 to 10; or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXI):
Figure imgf000127_0003
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 1 0; e is an integer from 0 to 10; f is an integer from 0 to 10; g is an integer from 0 to 25; or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXII):
Figure imgf000128_0001
(XXII)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and d is an integer from 0 to 15; or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXIII):
Figure imgf000128_0002
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from
a monosaccharide or an oligosaccharide; Y is
Figure imgf000128_0003
-C(0)CH2CH2-, -CH2-, or is absent; X is -C(0)CH2CH2- or is absent; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 15; e is an integer from 1 to 10; f is an integer from 1 to 5; and g is an integer from 1 to 5; or a pharmaceutically acceptable salt thereof. The disclosure also features a compound of formula (XXIV):
Figure imgf000129_0001
(XXIV)
wherein each A1 and A2 is an independently selected amino acid; X is -C(0)CH2CH2CH2-Y- or - C(0)CH2CH2C(0)NH-; Y is heteroaryl; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and d is an integer from 0 to 15; or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXV):
Figure imgf000129_0002
(XXV)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 1 5; and e is an integer from 0 to 1 5; or a
pharmaceutically acceptable salt thereof. The disclosure also features a compound of formula (XXVI):
Figure imgf000130_0001
(XXVI)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; R is C1 -C20 alkyl; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 20; e is an integer from 0 to 20; and f is an integer from 0 to 20; or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXVII):
Figure imgf000130_0002
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; R is C1 -C20 alkyl; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 1 to 20; e is an integer from 1 to 20; and f is an integer from 1 to 20; or a pharmaceutically acceptable salt thereof. The disclosure also features a compound of formula (XXVIII):
Figure imgf000131_0001
(XXVIII)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from
a monosaccharide or an oligosaccharide; Y is
Figure imgf000131_0002
-C(0)CH2CH2-, -CH2-, or is absent; e is an integer from 1 to 10; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and d is an integer from 0 to 15; or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXIX):
Figure imgf000131_0003
(XXIX)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; X is -CH2- or -C(O)-; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and d is an integer from 0 to 15; or a pharmaceutically acceptable salt thereof. The disclosure also features a compound of formula (XXX):
Figure imgf000132_0001
(XXX)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from
a monosaccharide or an oligosaccharide; Y is
Figure imgf000132_0002
, -C(0)CH2CH2-, -CH2-, or is absent; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 15; g is an integer from 0 to 15; and e and e' are each independently an integer from 1 to 3; or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXXI):
Figure imgf000132_0003
(XXXI)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from
a monosaccharide or an oligosaccharide; each Y is independently selected from
Figure imgf000132_0004
C(0)CH2CH2-, and -CH2-, or is absent; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 1 to 15; each e is independently from 1 to 15; each f is independently from 1 to 15; and g is from 1 to 15; or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXXII):
Figure imgf000133_0001
(XXXII)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from
a monosaccharide or an oligosaccharide; each Y is independently selected from
Figure imgf000133_0002
C(0)CH2CH2-, -CH2CH2NHC(0)CH2CH2-, and -CH2-, or is absent; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 1 to 15; each e is independently from 1 to 15; and g is from 1 to 15; or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXXIII):
Figure imgf000133_0003
(XXXIII)
wherein each A1 and A2 is an independently selected amino acid; each E is independently selected from a monosaccharide or an oligosaccharide; each X is independently selected from
Figure imgf000134_0001
, -C(0)CH2CH2-, -CH2CH2NHC(0)CH2CH2-, -C(O)-, and -CH2-, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 alkenyl, optionally substituted C1 -C20 alkynyl, optionally substituted C1 -C15 aryl, optionally substituted C1 -C15 heteroaryl, or is absent; each Y is independently selected from -CH-, or oxygen; Z is -NH-, -NHC(O)-, oxygen, or sulfur; each m is independently 0, 1 , 2, 3, 4, or 5; each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 1 to 15; g is an integer from 1 to 15; e and e' are independently from 1 to 10 and or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXIII), the compound has the formula (XXXII 1- ):
Figure imgf000134_0002
(XXXIII-1 )
or a pharmaceutically acceptable salt thereof.
In some embodiments of a compound of formula (XXXIII), the compound has the formula (XXXIII-
2):
Figure imgf000134_0003
(XXXIII-2)
or a pharmaceutically acceptable salt thereof. The disclosure also features a compound of formula (XXXIV):
Figure imgf000135_0001
(XXXIV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R3, R4, R5, R6, R7, R8, R9, R10, R'2, R'3, R'4, R'5, R'6, R'7, R'8, R'9, and R'10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4- C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C1 5 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or two R groups on the same or adjacent atoms join to form an optionally substituted 5-8 membered ring; each of a', b', c', a, b, and c is, independently, 0 or 1 ; each of N1 , N2, N3, N4, N'1 , N'2, N'3, and N'4 is a nitrogen atom ; each of C1 , C2, C3, C'1 , C'2, and C'3 is a carbon atom ; L is a linker comprising at least one optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C3-C15 heteroarylene; each E is, independently, a monosaccharide or oligosaccharide moiety; n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXXV):
Figure imgf000135_0002
(XXXV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R6, R8, R'2, R'6, and R'8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each E is, independently, a monosaccharide or oligosaccharide moiety; n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
The disclosure also features a com ound of formula (XXXV):
Figure imgf000136_0001
(XXXV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R6, R8, R'2, and R'6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 - C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each E is, independently, a monosaccharide or oligosaccharide moiety; n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
The disclosure also features a com ound of formula (XXXVI):
Figure imgf000136_0002
(XXXVI)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R6, R8, and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each E is, independently, a monosaccharide or oligosaccharide moiety; n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXXVII):
Figure imgf000137_0001
(XXXVII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R6, and R8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each E is, independently, a monosaccharide or oligosaccharide moiety; n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXXVIII):
Figure imgf000137_0002
(XXXVIII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R6, R'2, and R'6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each E is, independently, a monosaccharide or oligosaccharide moiety; n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXXIX):
Figure imgf000138_0001
(XXXIX)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2, R6, and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each E is, independently, a monosaccharide or oligosaccharide moiety; n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
The disclosure also features a com ound of formula (XXXX):
Figure imgf000138_0002
(XXXX) wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2 and R6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each E is, independently, a monosaccharide or oligosaccharide moiety; n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
The disclosure also features a com ound of formula (XXXXI):
Figure imgf000139_0001
(XXXXI)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each of R2 and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each E is, independently, a monosaccharide or oligosaccharide moiety; n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
The disclosure also features a com ound of formula (XXXXII):
Figure imgf000139_0002
(XXXXII) wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; R2 is a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20
heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each E is, independently, a monosaccharide or oligosaccharide moiety; n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
The disclosure also features a com ound of formula (XXXXIII):
Figure imgf000140_0001
(XXXXIII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; each E is, independently, a monosaccharide or oligosaccharide moiety; n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
The disclosure also features a com ound of formula (XXXXIV):
Figure imgf000140_0002
(XXXXIV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and L comprises an optionally substituted C3 cycloalkylene or an optionally substituted C3 heterocycloalkylene, or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXXXV):
Figure imgf000141_0001
(XXXXV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and L comprises an optionally substituted C3 cycloalkylene or an optionally substituted C3 heterocycloalkylene, or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXXXVI):
Figure imgf000141_0002
(XXXXVI)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene, or a pharmaceutically acceptable salt thereof.
The disclosure also features a com ound of formula (XXXXVII):
Figure imgf000141_0003
(XXXXVII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene, or a pharmaceutically acceptable salt thereof.
The disclosure also features a com ound of formula (XXXXVIII):
Figure imgf000142_0001
(XXXXVIII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene, or a pharmaceutically acceptable salt thereof.
The disclosure also features a com ound of formula (XXXXIX):
Figure imgf000142_0002
(XXXXIX)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene, or a pharmaceutically acceptable salt thereof.
The disclosure also features a compound of formula (XXXXX):
Figure imgf000142_0003
(XXXXX) wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl (e.g., CH2CH(CH3)2), or cyclohexylmethyl; and L comprises an optionally substituted C6 arylene, or a pharmaceutically acceptable salt thereof.
Figure imgf000143_0001
ome embodiments of the compounds described herein, E i V 'ΌΗ larmaceutically acceptable salt thereof.
III. Linkers
A linker refers to a linkage or connection between two or more components in a compound (e.g., two cyclic heptapeptides and one or more monosaccharide or oligosaccharide moieties in a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)). In some embodiments, the linker L' in the compound described by formula (I) is described by formula (L):
Figure imgf000143_0002
(L)
in which L is a remainder of L'; A1 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M1 or is absent; and A2 is a 1 -5 amino acid peptide (e.g., a 1 -4, 1 -3, or 1 -2 amino acid peptide) covalently attached to the linking nitrogen in M2 or is absent. In some embodiments, a compound described herein (e.g., compounds of any one of formulas (l)-(XXXXX)) may contain a linker that has a multivalent structure (e.g., divalent, trivalent, tetravalent, pentavalent, hexavalent, or heptavalent structure). In some embodiments, a compound described herein (e.g., compounds of any one of formulas (l)-(XII), (XIV), (XV), and (XXXIII)-(XXXXX); e.g., Compounds 1 -4, 7- 21 , and 23-25) may contain a linker that has a trivalent structure (e.g., a trivalent linker; a linker of formula (L-l); a linker of formual (L-VI)). A trivalent linker has three arms, in which each arm is conjugated to a component of the compound (e.g., a first arm conjugated to a first cyclic heptapeptide, a second arm conjugated to a second heptapeptide, and a third arm conjugated to a monosaccharide or
oligosaccharide moiety). In some embodiments, a compound described herein (e.g., compounds of formula (XIII); Compounds 5, 6, and 22) may contain a linker that has a tetravalent structure (e.g., a tetravalent linker; a linker of formula (L-lla), (L-l lb) , or (L-VII)). A tetravalent linker has four arms, in which each arm is conjugated to a component of the compound (e.g., a first arm conjugated to a first cyclic heptapeptide, a second arm conjugated to a second heptapeptide, a third arm conjugated to a first monosaccharide or oligosaccharide moiety, and a fourth arm conjugated to a second monosaccharide or oligosaccharide moiety). In some embodiments, the one or more monosaccharide or oligosaccharide moieties in the compounds described herein may each be, independently, connected to an atom in the linker. In some embodiments, a compound described herein may contain two or three linkers (e.g., compounds of formula (XIII) or (XIV)), in which each linker has a divalent structure (e.g., a divalent linker; a linker of any one of formulas (L-III)-(L-V)). For example, the first linker may connect the first and second cyclic heptapeptides, the second linker may connect a first monosaccharide or oligosaccharide moiety and a peptide attached to the first cyclic heptapeptide, and a third linker may connect a second monosaccharide or oligosaccharide moiety and a peptide attached to the second cyclic heptapeptide.
In some embodiments, a linker is described by formula (L-l):
Lc
LB-Q-LA
(L-l)
wherein LA is described by formula GA1-(ZA1 )gi-(YA1 )hi-(ZA2)ii-(YA2)ji -(ZA3)ki -(YA3)ii -(ZA4)mi -(YA4)ni-(ZA5)oi- GA2; LB is described by formula GB1-(ZB1 )g2-(YB1
Figure imgf000144_0001
Lc is described by
Figure imgf000144_0002
GA1 is a bond attached to Q in formula (L-l); GA2 is a bond attached to the first cyclic heptapeptide or a peptide (e.g., a peptide including 1 -5 amino acid residue(s)) attached to the first cyclic heptapeptide, if the peptide is present; GB1 is a bond attached to Q in formula (L-l); GB2 is a bond attached to the second cyclic heptapeptide or a peptide (e.g., a peptide including 1 -5 amino acid residue(s)) attached to the second cyclic heptapeptide, if the peptide is present; GC1 is a bond attached to Q in formula (L-l); GC2 is a bond attached to at least one monosaccharide or oligosaccharide moiety; each of ZA1 , ZA2, ZA3, ZA4, ZA5, ZB1 , ZB2, ZB3, ZB4, ZB5, ZC1 , ZC2, ZC3, ZC4 and ZC5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of YA1 , YA2, YA3, YA4, YB1 , YB2, YB3, YB4, YC1 , YC2, YC3, and YC4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C1 5 heteroaryl; each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , o1 , g2, h2, i2, j2, k2, I2, m2, n2, o2, g3, h3, i3, j3, k3, I3, m3, n3, and o3 is, independently, 0 or 1 ; Q is a nitrogen atom, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C1 5 arylene, or optionally substituted C2-C15 heteroarylene. Linkers of formula (L-l) that may be used in compounds described herein include, but limited to,
Figure imgf000145_0001
Figure imgf000146_0001
wherein R* is a bond or comprises one or more of optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15 heteroarylene, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, and imino, and
wherein R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl.
In some embodiments, a linker is described by formula (L-lla) or (L-llb):
Figure imgf000147_0001
(L-lla) (L-llb)
wherein LA is described by formula GA1-(ZA1 )gi-(YA1 )hi-(ZA2)ii-(YA2)ji -(ZA3)ki -(YA3)ii -(ZA4)mi - (YA4)m-(ZA5)oi-GA2; LB is described by formula GB1-(ZB1 )g2-(YB1 )h2-(ZB2)i2-(YB2)j2-(ZB3)k2-(YB3)i2-(ZB4)m2- (YB4)n2-(ZB5)o2-GB2; Lc is described by formula Gc1-(Zc1 )g3-(Yc1 )h3-(ZC2)i3-(YC2)j3-(ZC3)k3-(YC3)i3-(ZC4)m3- (YC4)n3-(ZC5)o3-GC2; LD is described by formula GD1 -(ZD1 )g4-(YD1 )h4-(ZD2)i4-(YD2)j4-(ZD3)k4-(YD3)i4-(ZD4)m4- (YD4)n4-(ZD5)o4-GD2; GA1 is a bond attached to NA in formula (L-lla) or NA in formula (L-llb); GA2 is a bond attached to the first cyclic heptapeptide or a peptide (e.g., a peptide including 1 -5 amino acid residue(s)) attached to the first cyclic heptapeptide, if the peptide is present; GB1 is a bond attached to NA in L of formula (L-lla) or NA in formula (L-llb); GB2 is a bond attached to the second cyclic heptapeptide or a peptide (e.g., a peptide including 1 -5 amino acid residue(s)) attached to the second cyclic heptapeptide, if the peptide is present; GC1 is a bond attached to NB in formula (L-lla) or C in formula (L-llb); GC2 is a bond attached to a first monosaccharide or oligosaccharide moiety; GD1 is a bond attached to NB in formula (L- lla) or C in formula (L-llb); GD2 is a bond attached to a second monosaccharide or oligosaccharide moiety; each of ZA1 ZA2 ZA3 ZA4 ZA5 ZB1 ZB2 ZB3 ZB4 ZB5 ZC1 ZC2 ZC3 ZC4 ZC5 ZD1 ZD2 ZD3 ZD4 and ZD5 is independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C1 5 arylene, or optionally substituted C2-C15 heteroarylene; each of YA1 , YA2, YA3, YA4, YB1 , YB2, YB3, YB4, YC1 , YC2, YC3, YC4 YD1 , YD2, YD3, and YD4 is, independently, O, S, NR\ P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 - C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20
cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; and each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , o1 , g2, h2, i2, j2, k2, I2, m2, n2, o2, g3, h3, i3, j3, k3, I3, m3, n3, o3, g4, h4, i4, j4, k4, I4, m4, n4, and o4, is, independently, 0 or 1 . Linkers of formulas (L-lla) and (L-llb) that may be used in compounds described herein include, but are not limited to,
Figure imgf000148_0001
In some embodiments, a linker is described b formula (L-VI):
Figure imgf000148_0002
(L-VI)
wherein LA is described by formula GA1-(ZA1 )gi-(YA1 )hi-(ZA2)ii-(YA2)ji -(ZA3)ki -(YA3)ii -(ZA4)mi -(YA4)ni-(ZA5)oi- GA2; LB is described by formula GB1-(ZB1 )g2
Lc is described by formula Gc1 -(Zc1 )g3-(Yc1
Figure imgf000148_0003
is a bond attached to Q in formula (L-VI); GA2 is a bond attached to A1 or M1 if A1 is absent; GB1 is a bond attached to Q in formula (L-VI); GB2 is a bond attached to A2 or M2 if A2 is absent; GC1 is a bond attached to Q in formula (L-VI); GC2 is a bond attached to E; each of ZA1 , ZA2, ZA3, ZM, ZA5, ZB1 , ZB2, ZB3, ZB4, ZB5, ZC1 , ZC2, ZC3, ZC4, and ZC5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2- C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20
heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of YA1 , YA2, YA3, YA4, YB1 , YB2, YB3, YB4, YC1 , YC2, YC3, and YC4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C1 5 heteroaryl; each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , o1 , g2, h2, i2, j2, k2, I2, m2, n2, o2, g3, h3, i3, j3, k3, I3, m3, n3, and o3 is, independently, 0 or 1 ; Q comprises at least one optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C3-C15 heteroarylene.
In some embodiments, a linker is described b formula (L-VII):
Figure imgf000149_0001
(L-VII)
wherein LA is described by formula GA1-(ZA1 )gi-(YA1 )hi-(ZA2)ii-(YA2)ji -(ZA3)ki -(YA3)ii -(ZA4)mi -(YA4)ni-(ZA5)oi - GA2; LB is described by formula GB1-(ZB1 )g2-(YB1 )h2-(ZB2)i2-(YB2)j2-(ZB3)k2-(YB3)i2-(ZB4)m2-(YB4)n2-(ZB5)02-GB2; Lc is described by formula Gc1 -(Zc1 )g3-(Yc1 )h3-(ZC2)i3-(YC2)j3-(ZC3)k3-(YC3)i3-(ZC4)m3-(YC4)n3-(ZC5)03-GC2^ LD is described by formula GD1-(ZD1 )g4-(YD 1 )h4-(ZD2)i4-(YD2)j4-(ZD3)k4-(YD3)i4-(ZD4)m4-(YD4)n4-(ZD5)o4-GD2; GA1 is a bond attached to Q in formula (L-VII); GA2 is a bond attached to A1 or M1 if A1 is absent; GB1 is a bond attached to Q in formula (L-VII); GB2 is a bond attached to A2 or M2 if A2 is absent; GC1 is a bond attached to Q in formula (L-VII); GC2 is a bond attached to a first monosaccharide or oligosaccharide moiety, E1 ; GD1 is a bond attached to N in formula (L-VII); GD2 is a bond attached to a second
monosaccharide or oligosaccharide moiety, E2; each of ZA1 , ZA2, ZA3, ZM, ZA5, ZB1 , ZB2, ZB3, ZB4, ZB5, ZC1 , ZC2, ZC3, ZC4, ZC5, ZD1 , ZD2, ZD3, ZD4, and ZD5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of YA1 , YA2, YA3, YA4, YB1 , YB2, YB3, YB4, YC1 , YC2, YC3, YC4, YD1 , YD2, YD3, and YD4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2- C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , o1 , g2, h2, i2, j2, k2, I2, m2, n2, o2, g3, h3, i3, j3, k3, I3, m3, n3, o3, g4, h4, i4, j4, k4, I4, m4, n4, and o4 is, independently, 0 or 1 ; Q comprises at least one optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C3-C15 heteroarylene.
In some embodiments, a linker provides space, rigidity, and/or flexibility between the two cyclic heptapeptides. In some embodiments, a linker may be a bond, e.g., a covalent bond, e.g., an amide bond, a disulfide bond, a C-N bond, a N-N bond, or any kind of bond created from a chemical reaction, e.g., chemical conjugation. In some embodiments, a linker (Ι_', L, L1, or L'1 as shown in any one of formulas (l)-(XVII) and (XXXIV)-(XXXXX)) includes no more than 250 atoms (e.g., 1 -2, 1 -4, 1 -6, 1 -8, 1 -10, 1-12, 1-14, 1-16, 1-18, 1-20, 1-25, 1-30, 1-35, 1-40, 1-45, 1-50, 1-55, 1-60, 1-65, 1-70, 1-75, 1-80, 1-85, 1-90, 1-95, 1-100, 1-110, 1-120, 1-130, 1-140, 1-150, 1-160, 1-170, 1-180, 1-190, 1-200, 1-210, 1-220, 1- 230, 1-240, or 1-250 atom(s); 250, 240, 230, 220, 210, 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40, 35, 30, 28, 26, 24, 22, 20, 18, 16, 14, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 atom(s)). In some embodiments, a linker (Ι_', L, L1, or L'1 as shown in any one of formulas (l)-(XVII) and (XXXIV)-(XXXXX)) includes no more than 250 non-hydrogen atoms (e.g., 1-2, 1-4, 1-6, 1-8, 1-10, 1-12, 1-14, 1-16, 1-18, 1-20, 1-25, 1-30, 1-35, 1-40, 1-45, 1-50, 1-55, 1-60, 1-65, 1-70, 1-75, 1-80, 1-85, 1-90, 1-95, 1-100, 1-110, 1-120, 1-130, 1-140, 1-150, 1-160, 1-170, 1-180, 1-190, 1-200, 1-210, 1- 220, 1 -230, 1 -240, or 1 -250 non-hydrogen atom(s); 250, 240, 230, 220, 210, 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40, 35, 30, 28, 26, 24, 22, 20, 18, 16, 14, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 non-hydrogen atom(s)). In some embodiments, the backbone of a linker (L\ L, L1, or L'1 as shown in any one of formulas (l)-(XVII) and (XXXIV)-(XXXXX)) includes no more than 250 atoms (e.g., 1 -2, 1 -4, 1 -6, 1 -8, 1 -10, 1 -12, 1 -14, 1 -16, 1 -18, 1 -20, 1 -25, 1 -30, 1 -35, 1 -40, 1 - 45, 1-50, 1-55, 1-60, 1-65, 1-70, 1-75, 1-80, 1-85, 1-90, 1-95, 1-100, 1-110, 1-120, 1-130, 1-140, 1-150, 1 -160, 1 -170, 1 -180, 1 -190, 1 -200, 1 -210, 1 -220, 1 -230, 1 -240, or 1 -250 atom(s); 250, 240, 230, 220,
210, 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45,40, 35, 30, 28, 26, 24, 22, 20, 18, 16, 14, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 atom(s)). The "backbone" of a linker refers to the atoms in the linker that together form the shortest path from one part of the compound to another part of the compound. The atoms in the backbone of the linker are directly involved in linking one part of the compound to another part of the compound. For examples, hydrogen atoms attached to carbons in the backbone of the linker are not considered as directly involved in linking one part of the compound to another part of the compound.
Molecules that may be used to make linkers (Ι_', L, L1, or L'1 as shown in any one of formulas (I)- (XVII) and (XXXIV)-(XXXXX)) include at least two functional groups, e.g., two carboxylic acid groups. In some embodiments of a trivalent linker, two arms of a linker may contain two dicarboxylic acids, in which the first carboxylic acid may form a covalent linkage with the first cyclic heptapeptide in the compound and the second carboxylic acid may form a covalent linkage with the second cyclic heptapeptide in the compound, and the third arm of the linker may for a covalent linkage (e.g., a C-0 bond) with one or more monosaccharide or oligosaccharide moieties in the compound. In some embodiments of a divalent linker, the divalent linker may contain two carboxylic acids, in which the first carboxylic acid may form a covalent linkage with one component (e.g., a first cyclic heptapeptide) in the compound and the second carboxylic acid may form a covalent linkage with another component (e.g., a second cyclic heptapeptide) in the compound. In some embodiments, when the first and/or second cyclic heptapeptide is attached to a peptide (e.g., a peptide including a 1 -5 amino acid residue(s)) at the linking nitrogen, the linker may form a covalent linkage with the peptide. In other embodiments of a divalent linker, one end of the linker may form a covalent linkage with the first or second cyclic heptapeptide in the compound and the other end of the linker may form a covalent linkage (e.g., a C-0 bond) with one or more monosaccharide or oligosaccharide moieties.
In some embodiments, dicarboxylic acid molecules may be used as linkers (e.g., a dicarboxylic acid linker). For example, the first carboxylic acid in a dicarboxylic acid molecule may form a covalent linkage with the linking nitrogen of the first cyclic heptapeptide and the second carboxylic acid may form a covalent linkage with the linking nitrogen of the second cyclic heptapeptide. In some embodiments, when the first and/or second cyclic heptapeptide is attached to a peptide (e.g., a peptide including a 1 -5 amino acid residue(s)) at the linking nitrogen, the first carboxylic acid in a dicarboxylic acid linker may form a covalent linkage with the terminal amine group at the end of the peptide that is attached to the first cyclic heptapeptide and the second carboxylic acid in the dicarboxylic acid linker may form a covalent linkage with the terminal amine group at the end of the peptide that is attached to the second cyclic heptapeptide.
mited to,
Figure imgf000151_0001
Figure imgf000152_0001
In some embodiments, dicarboxylic acid molecules, such as the ones described herein, may be further functionalized to contain one or more additional functional groups, which may be used to conjugate to one or more monosaccharide or oligosaccharide moieties.
In some embodiments, a molecule containing one or more sulfonic acid groups may be used to form a linker, in which the sulfonic acid group may form a sulfonamide linkage with the linking nitrogen in a cyclic heptapeptide. In some embodiments, a molecule containing one or more isocyanate groups may be used to form a linker, in which the isocyanate group may form a urea linkage with the linking nitrogen in a cyclic heptapeptide. In some embodiments, a molecule containing one or more haloalkyl groups may be used to form a linker, in which the haloalkyl group may form a covalent linkage, e.g., C-N and C-0 linkages, with a cyclic heptapeptide.
In some embodiments, a linker (Ι_', L, L1 , or L'1 as shown in any one of formulas (l)-(XVII) and (XXXIV)-(XXXXX)) may comprise a synthetic group derived from, e.g., a synthetic polymer (e.g., a polyethylene glycol (PEG) polymer). In some embodiments, a linker may comprise one or more amino acid residues. In some embodiments, a linker may be an amino acid sequence (e.g., a 1 -25 amino acid, 1 -10 amino acid, 1 -9 amino acid, 1 -8 amino acid, 1 -7 amino acid, 1 -6 amino acid, 1 -5 amino acid, 1 -4 amino acid, 1 -3 amino acid, 1 -2 amino acid, or 1 amino acid sequence). In some embodiments, a linker (L\ L, L1 , or L'1 as shown in any one of formulas (l)-(XVII) and (XXXIV)-(XXXXX)) may include one or more optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene (e.g., a PEG unit), optionally substituted C2-C20 alkenylene (e.g., C2 alkenylene), optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20
heteroalkynylene, optionally substituted C3-C20 cycloalkylene (e.g., cyclopropylene, cyclobutylene), optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene (e.g., C6 arylene), optionally substituted C2-C15 heteroarylene (e.g., imidazole, pyridine), O, S, NR' (R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2- C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl), P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino.
In some embodiments, L in a compound of any one of formulas (ll)-(XII) and (XVII) may have a formula of (L-l), (L-lla), or (L-llb). In some embodiments, L in a compound of formula (XIII) may have formula (L-lll), L1 in a compound of formula (XIII) may have formula (L-IV), and L'1 in a compound of formula (XIII) may have formula (L-V). In some embodiments, L in a compound of any one of formulas (XXXIV)-(XXXXX) may have a formula of (L-VI) or (L-VII).
Covalent conjugation of two or more components in a compound using a linker may be accomplished using well-known organic chemical synthesis techniques and methods. Complementary functional groups on two components may react with each other to form a covalent bond. Examples of complementary reactive functional groups include, but are not limited to, e.g., amine and activated carboxylic acid, thiol and maleimide, activated sulfonic acid and amine, isocyanate and amine, azide and alkyne, and alkene and tetrazine.
Other examples of functional groups capable of reacting with amino groups include, e.g., alkylating and acylating agents. Representative alkylating agents include: (i) an a-haloacetyl group, e.g., XCH2CO- (where X=Br, CI, or I); (ii) a N-maleimide group, which may react with amino groups either through a Michael type reaction or through acylation by addition to the ring carbonyl group; (iii) an aryl halide, e.g., a nitrohaloaromatic group; (iv) an alkyl halide; (v) an aldehyde or ketone capable of Schiff's base formation with amino groups; (vi) an epoxide, e.g., an epichlorohydrin and a bisoxirane, which may react with amino, sulfhydryl, or phenolic hydroxyl groups; (vii) a chlorine-containing of s-triazine, which is reactive towards nucleophiles such as amino, sufhydryl, and hydroxyl groups; (viii) an aziridine, which is reactive towards nucleophiles such as amino groups by ring opening; (ix) a squaric acid diethyl ester; and (x) an a-haloalkyl ether. Examples of amino-reactive acylating groups include, e.g., (i) an isocyanate and an isothiocyanate; (ii) a sulfonyl chloride; (iii) an acid halide; (iv) an active ester, e.g., a nitrophenylester or N- hydroxysuccinimidyl ester; (v) an acid anhydride, e.g., a mixed, symmetrical, or N-carboxyanhydride; (vi) an acylazide; and (vii) an imidoester. Aldehydes and ketones may be reacted with amines to form Schiff's bases, which may be stabilized through reductive amination.
It will be appreciated that certain functional groups may be converted to other functional groups prior to reaction, for example, to confer additional reactivity or selectivity. Examples of methods useful for this purpose include conversion of amines to carboxyls using reagents such as dicarboxylic anhydrides; conversion of amines to thiols using reagents such as N-acetylhomocysteine thiolactone, S- acetylmercaptosuccinic anhydride, 2-iminothiolane, or thiol-containing succinimidyl derivatives;
conversion of thiols to carboxyls using reagents such as a -haloacetates; conversion of thiols to amines using reagents such as ethylenimine or 2-bromoethylamine; conversion of carboxyls to amines using reagents such as carbodiimides followed by diamines; and conversion of alcohols to thiols using reagents such as tosyl chloride followed by transesterification with thioacetate and hydrolysis to the thiol with sodium acetate.
IV. Monosaccharide or Oligosaccharide Moieties
A monosaccharide moiety is a molecular moiety that has one optionally substituted C6-C9 (exclusive of the substituents) monosaccharide residue. An oligosaccharide moiety is a molecular moiety that includes at least two, e.g., 2-1 50 (e.g., 2-149, 2-140, 2-130, 2-120, 2-1 1 0, 2-100, 2-90, 2-80, 2-70, 2- 60, 2-50, 2-40, 2-30, 2-20, 2-18, 2-16, 2-14, 2-12, 2-10, 2-9, 2-8, 2-7, 2-6, 2-5, 2-4, 2-3, or 2)) optionally substituted C6-C9 (exclusive of the substituents) monosaccharide residues. In some embodiments, an oligosaccharide moiety includes 2-1 8 optionally substituted C6-C9 (exclusive of the substituents) monosaccharide residues. In some embodiments, an oligosaccharide moiety includes 2-12 optionally substituted C6-C9 (exclusive of the substituents) monosaccharide residues. In some embodiments, an oligosaccharide moiety has a molecule weight that does not exceed 30 kDa, 25 kDa, 24 kDa, 23 kDa, 22 kDa, 21 kDa, 20 kDa, 19 kDa, 1 8 kDa, 17 kDa, 16 kDa, 15 kDa, 14 kDa, 13 kDa, 12 kDa, 1 1 kDa, 10 kDa, 9 kDa, 8 kDa, 7 kDa, 6 kDa, 5 kDa or 3 kDa.
In some embodiments, a monosaccharide moiety has an optionally substituted C6-C9 monosaccharide residue selected from glucose (Glc), galactose (Gal), mannose (Man), allose (All), altrose (alt), gulose (Gul), idose (ido), talose (Tal), fucose (Fuc), rhamnose (Rha; also called L-rhamnose or L-Rha), thia-rhamnose (thia-Rha; also called thia-L-rhamnose or thia-L-Rha), quinovose (Qui), 2- deoxyglucose (2-dGlc), glucosamine (GlcN), galactosamine (GaIN), mannosamine (ManN), fucosamine (FucN), quinovosamine (QuiN), N-Acetyl-glucosamine (GlcNAc); N-Acetyl-galactosamine (GalNAc), N- Acetyl-mannosamine (ManNAc), N-acetyl-fucosamine (FucNAc), N-acetyl-quinovosamine (QuiNAc), glucuronic acid (GlcA), galacturonic acid (GalA), mannuronic acid (ManA), iduronic acid (IdoA), sialic acid (Sia), neuraminic acid (Neu), N-Acetyl-neuraminic acid (Neu5Ac), N-Glycolyl-neuraminic acid (Neu5Gc), glucitol (Glc-ol), galactitol (Gal-ol), mannitol (Man-ol), fructose (Fru), sorbose (Sor), tagatose (Tag), thevetose (The), acofriose (Aco), digitoxose (Dig), cymarose (Cym), abequose (Abe), colitose (Col), tyvelose (Tyv), ascarylose (Asc), paratose (Par), or N-acetyl-muramic acid (MurNAc). In some embodiments, a monosaccharide moiety has an optionally substituted C6-C9 monosaccharide residue selected from Rha, Gal, Glc, GIcA (Glucuronic acid), GlcNAc, GalNAc, GlcN(Gc) (N-Glycolyl-Glucosamine), Neu5Ac, Neu5Gc (N-Glycolyl-neuraminic acid), Fuc, Man, -htePCbMan (mannose phosphate), 6-H2PO3GIC (glucose phosphate), Mur (muramoyl), Mur-L-Ala-D-i-Gln-Lys, (Mur)- 3-O-GlcNAc, sulfate-galactose (Su-Gal), disulfate-galactose (Su2-Gal), sulfate-glucose (Su-Glc), sulfate- GlcNAc (Su-GlcNAc), or sulfate-GalNAc (Su-GalNAc).
Rhamnose (Rha) occurs in nature in its L-form, thus, rhamnose is also referred to as L-rhamnose or L-Rha. Rhamnose may be in a form (also called a-Rha or a-L-Rha) or β form (also called β-Rha or β- L-Rha). Thia-rhamnose has -SH attached at the anomeric carbon instead of -OH. Thia-rhamnose is also referred to as thia-Rha or thia-L-Rha. Thia-rhamnose may be in a form (also called thia-a-Rha or thia-a-L-Rha) or in β form (also called thia^-Rha or thia^-L-Rha). The structures of Rha, a-Rha, and β-Rha are shown below. In some embodiments, the monosaccharide moiety is an optionally substituted a-Rha or an optionally substituted thia-a-Rha. In some embodiments, a compounds described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) may contain a-Rha.
Figure imgf000155_0001
In some embodiments, in an oligosaccharide moiety, each of the optionally substituted C6-C9 monosaccharide residues may, independently, joined to an adjacent monosaccharide residue through an O-glycosidic, S-glycosidic, N-glycosidic linkage, or C-glycosidic. The binding at an O-glycosidic, S- glycosidic, N-glycosidic, or C-glycosidic linkage may be an a- or β-configuration, for example, through 1 ,2- , 1 ,3-, 1 ,4-, 1 ,6-, 2,3-, 2,6-, or 2,8-linkage, or a linkage such as 3-0, for example, a1 -2, a1 -3, a1 -4, a1 -6, a2-3, a2-6, a2-8, β1 -2, β1 -3, β1 -4, or β1 -6. In some embodiments, each of the optionally substituted C6- C9 monosaccharide residues is, independently, glucose (Glc), galactose (Gal), mannose (Man), allose (All), altrose (alt), gulose (Gul), idose (ido), talose (Tal), fucose (Fuc), rhamnose (Rha), quinovose (Qui), 2-deoxyglucose (2-dGlc), glucosamine (GlcN), galactosamine (GaIN), mannosamine (ManN), fucosamine (FucN), quinovosamine (QuiN), N-Acetyl-glucosamine (GlcNAc); N-Acetyl-galactosamine (GalNAc), N- Acetyl-mannosamine (ManNAc), N-acetyl-fucosamine (FucNAc), N-acetyl-quinovosamine (QuiNAc), glucuronic acid (GIcA), galacturonic acid (GalA), mannuronic acid (ManA), iduronic acid (IdoA), sialic acid (Sia), neuraminic acid (Neu), N-Acetyl-neuraminic acid (Neu5Ac), N-Glycolyl-neuraminic acid (Neu5Gc), glucitol (Glc-ol), galactitol (Gal-ol), mannitol (Man-ol), fructose (Fru), sorbose (Sor), tagatose (Tag), thevetose (The), acofriose (Aco), digitoxose (Dig), cymarose (Cym), abequose (Abe), colitose (Col), tyvelose (Tyv), ascarylose (Asc), paratose (Par), or N-acetyl-muramic acid (MurNAc). In some embodiments, each of the optionally substituted C6-C9 monosaccharide residues is, independently, Rha, Gal, Glc, GIcA (Glucuronic acid), GlcNAc, GalNAc, GlcN(Gc) (N-Glycolyl-Glucosamine), Neu5Ac, Neu5Gc (N-Glycolyl-neuraminic acid), Fuc, Man, -H2P03Man (mannose phosphate), 6-H2PO3GIC
(glucose phosphate), Mur (muramoyl), Mur-L-Ala-D-i-Gln-Lys, (Mur)-3-0-GlcNAc, sulfate-galactose (Su- Gal), disulfate-galactose (Su2-Gal), sulfate-glucose (Su-Glc), sulfate-GlcNAc (Su-GlcNAc), or sulfate- GalNAc (Su-GalNAc). In some embodiments, an oligosaccharide moiety may be straight or branched. In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety is substituted with one or more, such as 1 -3, substituents independently selected from sulfate, phosphate, methyl, acetyl, naturally amino acids, and non-naturally occurring amino acids.
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety is substituted with one or more, such as 1 -3, substituents independently selected from sulfate, phosphate, methyl, acetyl.
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety is substituted with one or more, such as 1 -3, substituents independently selected from naturally occurring amino acids and non- naturally occurring amino acids. If the monosaccharide residue is linked to an amino acid, such as serine, the linkage may be, for example, a a1 -O linkage. If the linkage is through a sulfate, the linkage may be through a hydroxyl group, for example, a 2-0, 3-0 , 4-0, or 6-0 linkage, such as 3-0-Su-Gal, (6- 0-Su)Gal, (6-0-Su)Glc, 6-0-Su-GlcNAc, (6-0-Su)GalNAc, 3,6-0-Su2-Gal, 3,4-0-Su2-Gal, or 4-0-Su-Gal. Naturally occurring amino acids include Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, He, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, and Val. A non-naturally occurring amino acid" is an amino acid that is not naturally produced or found in a mammal. Examples of non-naturally occurring amino acids include D- amino acids; an amino acid having an acetylaminomethyl group attached to a sulfur atom of a cysteine; a pegylated amino acid; the omega amino acids of the formula NH2(CH2)nCOOH where n is 2-6, neutral nonpolar amino acids, such as sarcosine, t-butyl alanine, t-butyl glycine, N-methyl isoleucine, and norleucine; oxymethionine; phenylglycine; citrulline; methionine sulfoxide; cysteic acid; ornithine;
diaminobutyric acid; and hydroxyproline. Other amino acids are a-aminobutyric acid, a-amino-a- methylbutyrate, aminocyclopropane-carboxylate, aminoisobutyric acid, aminonorbornyl-carboxylate, L- cyclohexylalanine, cyclopentylalanine, L-N-methylleucine, L-N-methylmethionine, L-N-methylnorvaline, L- N-methylphenylalanine, L-N-methylproline, L-N-methylserine, L-N-methyltryptophan, D-ornithine, L-N- methylethylglycine, L-norleucine, a-methyl-aminoisobutyrate, a-methylcyclohexylalanine, D-a- methylalanine, D-a-methylarginine, D-a-methylasparagine, D-a-methylaspartate, D-a-methylcysteine, D- a-methylglutamine, D-a-methylhistidine, D-a-methylisoleucine, D-a-methylleucine, D-a-methyllysine, D-a- methylmethionine, D-a-methylornithine, D-a-methylphenylalanine, D-a-methylproline, D-a-methylserine, D-N-methylserine, D-a-methylthreonine, D-a-methyltryptophan, D-a-methyltyrosine, D-a-methylvaline, D- N-methylalanine, D-N-methylarginine, D-N-methylasparagine, D-N-methylaspartate, D-N-methylcysteine, D-N-methylglutamine, D-N-methylglutamate, D-N-methylhistidine, D-N-methylisoleucine, D-N- methylleucine, D-N-methyllysine, N-methylcyclohexylalanine, D-N-methylornithine, N-methylglycine, N- methylaminoisobutyrate, N-(1 -methylpropyl)glycine, N-(2-methylpropyl)glycine, D-N-methyltryptophan, D- N-methyltyrosine, D-N-methylvaline, γ-aminobutyric acid, L-t-butylglycine, L-ethylglycine, L- homophenylalanine, L-a-methylarginine, L-a-methylaspartate, L-a-methylcysteine, L-a-methylglutamine, L-a-methylhistidine, L-a-methylisoleucine, L-a-methylleucine, L-a-methylmethionine, L-a-methylnorvaline, L-a-methylphenylalanine, L-a-methylserine, L-a-methyltryptophan, L-a-methylvaline, N-(N-(2,2- diphenylethyl) carbamylmethylglycine, 1 -carboxy-1 -(2,2-diphenyl-ethylamino) cyclopropane, 4- hydroxyproline, ornithine, 2-aminobenzoyl (anthraniloyl), D-cyclohexylalanine, 4-phenyl-phenylalanine, L- citrulline, a-cyclohexylglycine, L-1 ,2,3,4-tetrahydroisoquinoline-3-carboxylic acid, L-thiazolidine-4- carboxylic acid, L-homotyrosine, L-2-furylalanine, L-histidine (3-methyl), N-(3-guanidinopropyl)glycine, O- methyl-L-tyrosine, O-glycan-serine, meta-tyrosine, nor-tyrosine, L-N,N',N"-trimethyllysine, homolysine, norlysine, N-glycan asparagine, 7-hydroxy-1 ,2,3,4-tetrahydro-4-fluorophenylalanine, 4- methylphenylalanine, bis-(2-picolyl)amine, pentafluorophenylalanine, indoline-2-carboxylic acid, 2- aminobenzoic acid, 3-amino-2-naphthoic acid, asymmetric dimethylarginine, L-tetrahydroisoquinoline-1 - carboxylic acid, D-tetrahydroisoquinoline-1 -carboxylic acid, 1 -amino-cyclohexane acetic acid, D/L- allylglycine, 4-aminobenzoic acid, 1 -amino-cyclobutane carboxylic acid, 2 or 3 or 4-aminocyclohexane carboxylic acid, 1 -amino-1 -cyclopentane carboxylic acid, 1 -aminoindane-1 -carboxylic acid, 4-amino- pyrrolidine-2-carboxylic acid, 2-aminotetraline-2-carboxylic acid, azetidine-3-carboxylic acid, 4-benzyl- pyrrolidine-2-carboxylic acid, tert-butylglycine, b-(benzothiazolyl-2-yl)-alanine, b-cyclopropyl alanine, 5,5- dimethyl-1 ,3-thiazolidine-4-carboxylic acid, (2R,4S)4-hydroxypiperidine-2-carboxylic acid, (2S,4S) and (2S,4R)-4-(2-naphthylmethoxy)-pyrrolidine-2-carboxylic acid, (2S,4S) and (2S,4R)4-phenoxy-pyrrolidine- 2-carboxylic acid, (2R,5S)and(2S,5R)-5-phenyl-pyrrolidine-2-carboxylic acid, (2S,4S)-4-amino-1 -benzoyl- pyrrolidine-2-carboxylic acid, t-butylalanine, (2S,5R)-5-phenyl-pyrrolidine-2-carboxylic acid, 1 - aminomethyl-cyclohexane-acetic acid, 3,5-bis-(2-amino)ethoxy-benzoic acid, 3,5-diamino-benzoic acid, 2- methylamino-benzoic acid, N-methylanthranylic acid, L-N-methylalanine, L-N-methylarginine, L-N- methylasparagine, L-N-methylaspartic acid, L-N-methylcysteine, L-N-methylglutamine, L-N- methylglutamic acid, L-N-methylhistidine, L-N-methylisoleucine, L-N-methyllysine, L-N-methylnorleucine, L-N-methylornithine, L-N-methylthreonine, L-N-methyltyrosine, L-N-methylvaline, L-N-methyl-t- butylglycine, L-norvaline, a-methyl-Y-aminobutyrate, 4,4'-biphenylalanine, a-methylcylcopentylalanine, a- methyl-a-napthylalanine, a-methylpenicillamine, N-(4-aminobutyl)glycine, N-(2-aminoethyl)glycine, N-(3- aminopropyl)glycine, N-amino-a-methylbutyrate, a-napthylalanine, N-benzylglycine, N-(2- carbamylethyl)glycine, N-(carbamylmethyl)glycine, N-(2-carboxyethyl)glycine, N-(carboxymethyl)glycine, N-cyclobutylglycine, N-cyclodecylglycine, N-cycloheptylglycine, N-cyclohexylglycine, N-cyclodecylglycine, N-cylcododecylglycine, N-cyclooctylglycine, N-cyclopropylglycine, N-cycloundecylglycine, N-(2,2- diphenylethyl)glycine, N-(3,3-diphenylpropyl)glycine, N-(3-guanidinopropyl)glycine, N-(1 - hydroxyethyl)glycine, N-(hydroxyethyl))glycine, N-(imidazolylethyl))glycine, N-(3-indolylyethyl)glycine, N- methyl-Y-aminobutyrate, D-N-methylmethionine, N-methylcyclopentylalanine, D-N-methylphenylalanine, D-N-methylproline, D-N-methylthreonine, N-(1 -methylethyl)glycine, N-methyl-napthylalanine, N- methylpenicillamine, N-(p-hydroxyphenyl)glycine, N-(thiomethyl)glycine, penicillamine, L-a-methylalanine, L-a-methylasparagine, L-a-methyl-t-butylglycine, L-methylethylglycine, L-a-methylglutamate, L-a- methylhomophenylalanine, N-(2-methylthioethyl)glycine, L-a-methyllysine, L-a-methylnorleucine, L-a- methylornithine, L-a-methylproline, L-a-methylthreonine, L-a-methyltyrosine, L-N-methyl- homophenylalanine, N-(N-(3,3-diphenylpropyl) carbamylmethylglycine, L-pyroglutamic acid, D- pyroglutamic acid, O-methyl-L-serine, O-methyl-L-homoserine, 5-hydroxylysine, a-carboxyglutamate, phenylglycine, L-pipecolic acid (homoproline), L-homoleucine, L-lysine (dimethyl), L-2-naphthylalanine, L- dimethyldopa or L-dimethoxy-phenylalanine, L-3-pyridylalanine, L-histidine (benzoyloxymethyl), N- cycloheptylglycine, L-diphenylalanine, O-methyl-L-homotyrosine, L-p-homolysine, O-glycan-threoine, Ortho-tyrosine, L-N.N'-dimethyllysine, L-homoarginine, neotryptophan, 3-benzothienylalanine, isoquinoline-3-carboxylic acid, diaminopropionic acid, homocysteine, 3,4-dimethoxyphenylalanine, 4- chlorophenylalanine, L-1 ,2,3,4-tetrahydronorharman-3-carboxylic acid, adamantylalanine, symmetrical dimethylarginine, 3-carboxythiomorpholine, D-1 ,2,3,4-tetrahydronorharman-3-carboxylic acid, 3- aminobenzoic acid, 3-amino-1 -carboxymethyl-pyridin-2-one, 1 -amino-1 -cyclohexane carboxylic acid, 2- aminocyclopentane carboxylic acid, 1 -amino-1 -cyclopropane carboxylic acid, 2-aminoindane-2-carboxylic acid, 4-amino-tetrahydrothiopyran-4-carboxylic acid, azetidine-2-carboxylic acid, b-(benzothiazol-2-yl)- alanine, neopentylglycine, 2-carboxymethyl piperidine, b-cyclobutyl alanine, allylglycine, diaminopropionic acid, homo-cyclohexyl alanine, (2S,4R)- 4-hydroxypiperidine-2-carboxylic acid, octahydroindole-2- carboxylic acid, (2S,4R) and (2S,4R)-4-(2-naphthyl), pyrrolidine-2-carboxylic acid, nipecotic acid, (2S,4R)and (2S,4S)-4-(4-phenylbenzyl) pyrrolidine-2-carboxylic acid, (3S)-1 -pyrrolidine-3-carboxylic acid, (2S,4S)-4-tritylmercapto-pyrrolidine-2-carboxylic acid, (2S,4S)-4-mercaptoproline, t-butylglycine, N,N- bis(3-aminopropyl)glycine, 1 -amino-cyclohexane-1 -carboxylic acid, N-mercaptoethylglycine, and selenocysteine. In some embodiments, amino acid residues may be charged or polar. Charged amino acids include alanine, lysine, aspartic acid, or glutamic acid, or non-naturally occurring analogs thereof. Polar amino acids include glutamine, asparagine, histidine, serine, threonine, tyrosine, methionine, or tryptophan, or non-naturally occurring analogs thereof.
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety may be in a closed ring form
(i.e., where the aldehyde/ketone carbonyl carbon (C=0) and hydroxyl group (-OH) in the open-chain monosaccharide residue reacts to form a hemiacetal with a new C-O-C bridge) or an open ring form.
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety is an optionally substituted C6 monosaccharide residue.
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety is an optionally substituted C9 monosaccharide residue, e.g., sialic acid (Sia), Neuraminic acid (Neu), N-Acetyl-neuraminic acid
(Neu5Ac), or N-Glycolyl-neuraminic acid (Neu5Gc).
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety may include a hexose residue, e.g., glucose (Glc), galactose (Gal), mannose (Man), allose (All), altrose (alt), gulose (Gul), idose
(ido), or talose (Tal).
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety may include a deoxyhexose residue, e.g., a hexose residue without the hydroxyl group at the 6-position or the 2-position, e.g., fucose (Fuc), rhamnose (Rha), quinovose (Qui), or 2-deoxyglucose (2-dGlc).
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety may also include an aminohexose residue, e.g., a hexose residue with an amino group or an N-acetylated amino group at the 2 -position, e.g., glucosamine (GlcN), galactosamine (GaIN), mannosamine (ManN), fucosamine (FucN), quinovosamine (QuiN), N-Acetyl-glucosamine (GlcNAc), N-Acetyl-galactosamine (GalNAc), N-Acetyl- mannosamine (ManNAc), N-acetyl-fucosamine (FucNAc), or N-acetyl-quinovosamine (QuiNAc).
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety may include a uronic acid residue, e.g., a hexose residue with a negatively charged carboxylate at the 6-position, e.g., glucuronic acid (GlcA), galacturonic acid (GalA), mannuronic acid (ManA), or iduronic acid (IdoA).
In addition to hexose residues, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety may include a sialic acid residue, e.g., a residue of a nine-carbon acidic sugar, e.g., a residue of. Sialic acid (Sia), Neuraminic acid (Neu), N-Acetyl-neuraminic acid (Neu5Ac), or N-Glycolyl-neuraminic acid (Neu5Gc).
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety may include a sugar alcohol, e.g., glucitol (Glc-ol), galactitol (Gal-ol), or mannitol (Man-ol).
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety may include other compounds, e.g., thevetose (The), acofriose (Aco), digitoxose (Dig), cymarose (Cym), abequose (Abe), colitose (Col), tyvelose (Tyv), ascarylose (Asc), paratose (Par), and N-acetyl-muramic acid (MurNAc).
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety is Gal, such as aGal (for example, a1 -3Gal). An aGal epitope, i.e., an oligosaccharide moiety that exhibits specific binding to an anti-aGal antibody, may also include a moiety comprising an a-D-galactopyranoside moiety, Gal, Galal-3Gal, Galal-4Gal, Galal-6Gal, Galal-3Gala1 -3GlcNAc, Galal-3Gala1 -4Gal, Galal-3Gala1 -4GlcNAc, Galal-3Gala1 -4Glc, Galal-3Gala1 -4[3-deoxy]GlcNAc, Galal-3Gala1 -4[6-deoxy]GlcNAc, Galal-3Gala1 - 4Gala1 -3Gal, Galal-3Gala1 -4GlcNAca1 -3Gala1 -4Glc, and any multimers and combinations thereof. Galal-3Gal 1 -4GlcNAc-R (aGal epitope) glyconjugates have been reported in Macher, et al. Biochim Biophys Acta 1780: 75-88, 2008.
Exemplary aGal epitopes that may be included in a compound described here (e.g., a compound of any one of formulas (l)-(XXXXX)) are shown below:
Figure imgf000159_0001
If present, optional substituents may include 1 -3 substituents independently selected from sulfate, phosphate, methyl, acetyl, naturally occurring amino acid residues, and non-naturally occurring amino acid residues on each monosaccharide residue. These may be linked through an O, S, or N, such as a sulfamate linkage through an S. The amino acid residues may be charged or polar and includes isomers thereof. Charged amino acids include alanine, lysine, aspartic acid, or glutamic acid. Polar amino acids include glutamine, asparagine, histidine, serine, threonine, tyrosine, methionine or tryptophan. For example, the amino acid substituent, if present, may be alanine, lysine, serine, glutamine, or i-glutamine, or asparagine.
In some embodiments, the optionally substituted monosaccharide residue(s) (i.e., at least one monosaccharide residue) in the monosaccharide or oligosaccharide moiety may contain at least one (such as 1 -12) of the following optionally substituted monosaccharide residues: Rha, Gal, Glc, GlcA (Glucuronic acid), GlcNAc, GalNAc, GlcN(Gc) (N-Glycolyl-Glucosamine), Neu5Ac, Neu5Gc (N-Glycolyl- neuraminic acid), Fuc, Man, -htePCbMan (mannose phosphate), 6-H2PO3GIC (glucose phosphate), Mur (muramoyl), Mur-L-Ala-D-i-Gln-Lys, (Mur)-3-0-GlcNAc, sulfate-galactose (Su-Gal), disulfate-galactose (Su2-Gal), sulfate-glucose (Su-Glc), sulfate-GlcNAc (Su-GlcNAc), or sulfate-GalNAc (Su-GalNAc).
In some embodiments, the 1 -12 optionally substituted C6-C9 monosaccharide residues in the monosaccharide or oligosaccharide moiety may be β-glucan residues. In some embodiments, the β- Glucan residue includes 2-12 optionally substituted glucopyranosyl monosaccharide residues each independently joined to an adjacent glucopyranosyl monosaccharide residue via an O-glycosidic, S- glycosidic, N-glycosidic, or C-glycosidic, linkage to form β-linked chains, which retain the ability to bind dectin-1 . Optional substituents may include, e.g., 1 -3 substituents, such as with acetyl, sulfate, phosphate, or a natural or non-natural amino acid. Some examples of β-glucan residues include thia-β- glucan residues such as the structures shown below:
Figure imgf000160_0001
In some embodiments, an oligosaccharide moiety may be a β-glucan having more than 10 monosaccharides and/or a molecular weight of at least 3 kDa (e.g., 3-30 kDa; e.g., 3-29, 3-25, 3-20, 3-15, 3-10, 3-8, 3-6, 3-5, or 3-4 kDa). For example, an oligosaccharide moiety may be a β(1→3, 1→6)-glucan, e.g., laminarin, or a β(1→6)-glucan, e.g., pustulan.
In some embodiments, a monosaccharide or oligosaccharide moiety directly or indirectly activates an immune cell. In some embodiments, the monosaccharide or oligosaccharide moiety directly binds an immune cell. For example, β-glucans bind dectin-1 receptors. When bound to dectin-1 , which internalizes the β-glucan, β-glucan mediates the production of reactive oxygen species (ROS), activation of NF- B, and subsequent secretion of proinflammatory cytokines. The β-giucan receptor, dectin-1 , is predominantly expressed on the surface of cells of monocytes/macrophages and neutrophils. In some embodiments, the monosaccharide or oligosaccharide moiety indirectly binds an immune cell. For example, without being bound by theory, the monosaccharide or oligosaccharide moiety may bind to an antibody. The antibody in turn may bind to Fc receptors found on the surface of certain on immune cells including, among others, B lymphocytes, follicular dendritic cells, natural killer cells, macrophages, neutrophils, eosinophils, basophils, and mast cells. For example, aGal or aRha will bind to anti-aGal or anti-aRha antibody, respectively, thereby indirectly activating immune cells.
In some embodiments, a monosaccharide or oligosaccharide moiety is a ligand to an innate immune receptor. In some embodiments, the innate immune receptor is AICL, BDCA2, CLEC2, Complement receptor 3, Complement receptor 4, DCIR, dectin-1 , dectin-2, DC-SIGN, a C-Type lectin receptor, MMR, langerin, TLR2, Mincle, MBL, or KCR. In some embodiments, the monosaccharide or oligosaccharide moiety binds to an antibody. In some embodiments, the antibody is a natural antibody. In some embodiments, the natural antibody is an antibody of the immunoglobulin M (IgM) isotype.
Glycans bound by antibodies contained in intravenous immunoglobulin (IVIG) are studied by Gunten et al., J. Allergy Clin Immunol. 123:1268, 2009, e.g., see Tables I and E1 of Gunten et al., which is incorporated herein by reference in its entirety. In some embodiments, a monosaccharide or oligosaccharide moiety in a compound described herein (e.g., a compound of any one of formulas (I)- (XXXXX)) may be any one of the glycans listed in Tables I and E1 of Gunten et al. Examples of glycans studied by Gunten et al. are shown in Table 2A. Anti-carbohydrate antibodies found in normal sera have been studied by Huflejt et al., Molecular Immunology 46:3037-3049, 2009, using a library of glycans shown in Table 2B.
Table 2A
Structure of Glycan Structure of Glycan
Neu5Aca2-3Galp1 -3GlcNAcp1 -3Galp1 -
1 GlcNAcpl -2Gcrtp1 -3GalNAca 10
3GlcNAcp
NeuAca2-6Galp1 -4GlcNAcp1 -3Galp1 -
2 GlcNAcal -3Galp1 -4GlcNAcp 1 1
3GlcNAcp
GlcNAcpl -3(GlcNAcp1 -4)(GlcNAcp1 -
3 12 a-L-Rha
6)GlcNAc
4 GlcNAcpl -4MDPLys 13 [su]3Galp1 -3GlcNAcp
5 GlcNAcpl -4(GlcNAcp1 -6)GalNAca 14 HOOC(CH3)CH-3-0-GlcNAcp1 -4GlcNAcp
6 GlcNAcpl -4Galp1 -4GlcNAcp Galpl -3(Fuca1 -4)GlcNAcp1 -2Mana1 - 3[Galp
7 Gala1 -3Galp1 -3GlcNAcp 15
1 -3(Fuca1 -4)GlcNAcp1 -2Mana1 -6]Man p1 -4GlcNAcp1 -4GlcNAcp
8 NeuAc(9Ac)a2-3Galp1 -3GlcNAcp
Neu5Aca2-3Galp1 -4GlcNAcp1 -3Galp1 -
9
3GlcNAcp
Table 2B
(Gc: glyc ;olyl; GIcA: glucuronic acid; Mur: muramoyi; O S: oligo saccharide; P: phosphate; Ser. Serine; Sia: Neu5Ac Su. Sulfate.)
oiruciure oi uiycan oiruciure οτ iycan
1 Neu5Aca2-6Galp1 -4GlcNAcp- 80 GlcNAcpl -3Galp1 -3GalNAca- Structure of Glycan Structure of Glycan
Galp1-4GlcNAcp1-3
Galp1-4GlcNAcp- 81 GalNAcal -3(Fuca1 -2)Galp1 -4GlcNAcp- Galp1-4GlcNAcp1-6
Neu5Gca2-6Galp1 -4GlcNAcp- 82 GalNAca1-0-Ser
Fuca1-3
GlcNAcp- 83 GlcNAcpl -6Galp1 -4GlcNAcp- Neu5Aca2-3Galp1-4
Mana1-3
Manal -6(Mana1 -3)Manp- 84 GlcNAcpl -6(Galp1 -3)GalNAca- Mana1-6
Manal -3
Manpl -4GlcNAcp1 -4GlcNAcp- 85 GlcNAcp1-3Galp1-4Glcp- Mana1-6
Neu5Aca2-8Neu5Aca2- 86 Galpl -4GlcNAcp1 -3GalNAca-
Galpl -4(Fuca1 -3)GlcNAcp- 87 GlcAp1-3Galp-
3-0-Su-Galp1 -4(Fuca1 -3)GlcNAcp- 88 Neu5Aca2-8Neu5Acp-OCH2C6H4-p-
Fuca1-2Galp1-4GlcNAcp- 89 Neu5Aca2-3Galp1 -4Glcp-
Fuca1-3
6-O-Su-GlcNAcp- 90 Fuca1-2Galp- Neu5Aca2-3Galp1-4
Neu5Aca2-3Galp1 -3(Fuca1 -4)GlcNAcp- 91 3,4-0-Su2-Galp1 -4GlcNAcp-
GlcNAcp1-6
Galp1-4GlcNAc- 92 Neu5Aca2-6Galp- Galp1-4GlcNAcp1-3
(Neu5Aca2-6Galp1 -4GlcNAcp1 -2Mana1 )2 -
93 Gala1-3Galp- 3,6-Manp1 -4GlcNAcp1 -4GlcNAcp-
(Galpl -4GlcNAcp1 -2Mana1 )2-3,6-Manp1 -
94 Galp1-4Glcp- 4GlcNAcp1-4GlcNAcp-
(GlcNAcpl -2Mana1 )2-3,6-Manp1 -
95 GalNAcp1-3Galp- 4GlcNAcp1-4GlcNAcp-
Fucal -2Galp1 -4(Fuca1 -3)GlcNAcp- 96 Gala-
Galp1-3(Fuca1-4) GlcNAcp- 97 GlcNAcp1-3Manp-
Neu5Aca- 98 Galp1-3GalNAca-
Neu5Aca2-3Galp1-3-(6-0-Su)GalNAca- 99 Gala1-3GalNAca-
(Galp1-4GlcNAc-p1-3)3p- 100 Neu5Gca2-6GalNAca-
Mana1-6Manp- 101 Gala1-4Galp1-4GlcNAcp-
Neu5Aca2-6Galp1-4-(6-0-Su)GlcNAcp- 102 GalNAcal -3(Fuca1 -2)Gal-
Neu5Aca2-3Galp1-4-(6-0-Su)GlcNAcp- 103 Glcp- Structure of Glycan Structure of Glycan
Fuca1-3
GlcNAcp- 104 Glcp1-4Glcp- Neu5Aca2-3(6-0-Su)Galp1 -4
Neu5Aca2-8Neu5Aca-OCH2C6H4-p- 105 GlcAp1-6Galp-
3-0-Su-Galp1 -3(Fuca1 -4)GlcNAcp- 106 GalNAcp-
Glca- 107 GalNAcp1-4GlcNAcp-
3-O-Su-Galp- 108 Glca1-4Glcp-
GlcN(Gc)p- 109 Gala1-3GalNAcp-
Galp1-4GlcNAcp- 110 Neu5Aca2-6GalNAca-
Neu5Aca2-3Galp1 -4GlcNAcp- 111 Galp1-4(6-0-Su)Glcp-
Neu5Aca2-3Galp1 -3GalNAca- 112 Galp1-3Galp-
GlcNAcp1-3
Galp1-4GlcNAcp- 113 GalNAca- Galp1-4GlcNAcp1-6
Neu5Aca2-3Galp- 114 Gala1-6Glcp-
Manal -3(Mana1 -6)Manp- 115 Manp1-4GlcNAcp-
Neu5Aca2-3Galp1 -4Glcp- 116 Glcp1-6Glcp-
Galpl -4GlcNAcp1 -3Galp1 -4GlcNAcp- 117 GlcNAcpl -4GlcNAcp-Asn
3-0-Su-Galp1 -4GlcNAcp1 -3Galp1 -
118 Gala1-3Galp1-4GlcNAcp- 4GlcNAc-
Mana- 119 Fuca1-2Galp1-4Glcp-
Fucal -4GlcNAcp1 -3Galp1 -4Glcp-Fuca1 -
3-0-Su-Galp1-4Glcp- 120
2Galp1-3
Neu5Gca- 121 Galp1-2Galp-
(Glca1-4)3p- 122 Gala1-4Galp1-4Glcp-
GlcNAcpl -3(GlcNAcp1 -6)Galp1 -4GlcNAcp- 123 Galp-
3-0-Su-Galp1-4GlcNAcp- 124 Galp1-3Galp1-4GlcNAcp-
GlcNAcpl -3Galp1 -4GlcNAcp- 125 Gala1-2Galp-
(Glca1-6)4p- 126 6-O-Su-GlcNAcp-
GlcAp- 127 GlcNAcp1-4GlcNAcp-
Neu5Aca2-6Galp1 -4Glcp- 128 Galpl -4GlcNAcp1 -6GalNAca-
(Neu5Aca2-8)3p- 129 GlcAp1-3GlcNAcp-
Fucal -2Galp1 -3(Fuca1 -4)GlcNAcp- 130 Galpl -4GlcNAcp1 -3Galp1 -4Glcp- Structure of Glycan Structure of Glycan
Fucal -3GlcNAcp1 -3Galp-Neu5Aca2- Fucal -3GlcNAcp1 -3Galp1 -4Glcp-Fuca1 -
131
3Galp1 -4 2Galp1 -4
6-H2P03Mana- 132 Galp1 -3GalNAcp-
3-0-Su-Galp1 -4(6-0-Su)GlcNAcp- 133 GlcNAcp1 -3GalNAca-
Galpl -4GlcNAcp1 -6Galp1 -4GlcNAcp - 134 Gala1 -3(Fuca1 -2)Galp-
3,6-0-Su2-Galp1 -4GlcNAcp- 135 GalNAcp1 -4Galp1 -4Glcp-
Glca1 -6Glca1 -6Glcp- 136 GlcNAcpl -GalNAca-
Galpl -4GlcNAcp1 -6(Galp1 -3)GalNAca- 137 Galpl -3GlcNAcp1 -3Galp1 -4Glcp-
Neu5Aca-OCH2C6H4-p- 138 GalNAcal -3(Fuca1 -2)Galp-
6-0-Su-Galp1 -4GlcNAcp- 139 GlcNAcpl -4-(Mur)-3-0-GlcNAcp-
Neu5Aca2-3Galp1 -3(Fuca1 -4)GlcNAcpl- 140 GlcNAcp-
GlcAa- 141 Galp1 -3GlcNAcp-
Galp1 -4(6-0-Su)GlcNAcp- 142 GalNAca1 -3Galp-
Neu5Aca2-8Neu5Aca2-3Galp1 -4Glcp- 143 GlcNAcpl -3(GlcNAcp1 -6)GalNAca-
Fuca- 144 Galal -3(Fuca1 -2)Galp1 -4GlcNAcp-
3-0-Su-Galp1 -3GalNAca- 145 Galpl -3GalNAcp1 -4Galp1 -4Glcp-
6-0-Su-Galp1 -4(6-0-Su)GlcNAcp- 146 Gala1 -4GlcNAcp-
3,6-0-Su2-Galp1 -4(6-0-Su)GlcNAcp- 147 Fuca1 -4GlcNAcp-
3-0-Su-Galp1 -4(6-0-Su)Glcp- 148 Fuca1 -3GlcNAcp-
6-0-Su-Galp1 -4(6-0-Su)Glcp- 149 Rhaa-
Neu5Aca2-6(Galp1 -3)GalNAca- 150 Gala1 -3(Fuca1 -2)Galp-
(Glca1 -4)4p- 151 GlcNAcpl -4GlcNAcp1 -4GlcNAcp-
Neu5Aca2-6(Galp1 -3)GlcNAcp1 -3Galp1 -
152 4-0-Su-Galp1 -4GlcNAcp- 4Glcp-
Galpl -4GlcNAcp1 -3GalNAca-Galp1 -
153 GalNAca1 -3GalNAcp- 4GlcNAcp1 -6
Fucal -2Galp1 -3GlcNAcp- 154 (GlcNAcp1 -4)6p-
Fucal -2Galp1 -3GalNAca- 155 Neu5Aca2-3Galp1 -3GlcNAcp-
6-H2PO3GICP- 156 (GlcNAcp1 -4)5p-
6-0-Su-Galp1 -4Glcp- 157 GlcNAcpl -4Mur-L-Ala-D-i-Gln-Lys
Galal -3Galp1 -4(Fuca1 -3)GlcNAcp- 158 3-0-Su-Galp1 -3GlcNAcp- Parameters that describe properties of immunoprofiles of carbohydrate-binding antibodies are median and median absolute deviation. A low median indicates that most donors show low intensities of antibodies bound to the given glycan, while a large median suggests that there is a significant number of donors with large antibody binding intensities to the given glycan. Any of the glycan moieties in Tables 2A and 2B may be used as the monosaccharide or oligosaccharide moiety herein.
In some embodiments, a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) may contain one or more monosaccharide or oligosaccharide moieties having the structures
Figure imgf000165_0001
V. Antibacterial Agents
In some embodiments, one or more antibacterial agents may be administered in combination (e.g., administered substantially simultaneously (e.g., in the same pharmaceutical composition or in separate pharmaceutical compositions) or administered separately at different times) with a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)).
Antibacterial agents may be grouped into several classes, e.g., quinolones, carbapenems, macrolides, DHFR inhibitors, aminoglycosides, ansamycins (e.g., geldanamycin, herimycin, and rifaximin), carbacephem (e.g., loracarbef), cephalosporins (e.g., cefadroxil, cefaolin, cefalotin, cefalothin, cephalexin, e.g., cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefdinir, cefditoren,
cefoperazone, cefotaxime, cefpodoxime, ceftazidime, and ceftobiprole), glycopeptides (e.g., teicoplanin, vancomycin, telavancin, dalbavancin, and oritavancin), lincosamides (e.g., clindamycin and lincomycin), lipopeptides (e.g., daptomycin), monobactams (e.g., aztreonam), nitrofurans (e.g., furazolidone and nitrofurantoin), oxazolidinones, pleuromutilins, penicillins , sulfonamides, and tetracyclines (e.g., eravacycline, demeclocycline, doxycycline, minocycline, oxytetracycline, and tetracycline). Quinolones include, but are not limited to, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, and temafloxacin. Carbapenems include, but are not limited to, ertapenem, doripenem,
imipenem/cilastatin, and meropenem. Macrolides include, but are not limited to, solithromycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, and spiramycin. In some embodiments, a macrolide is solithromycin. DHFR inhibitors include, but are not limited to, diaminoquinazoline, diaminopyrroloquinazoline, diaminopyrimidine, diaminopteridine, and diaminotriazines. Aminoglycosides include, but are not limited to, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, and spectinomycin. Oxazolidinones include, but are not limited to, linezolid, tedizolid, posizolid, radezolid, and furazolidone. Pleuromutilins include, but are not limited to, retapamulin, valnemulin, tiamulin, azamulin, and lefamulin. Penicillins include, but are not limited to, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin G, penicillin V, piperacillin, penicillin G, temocillin, and ticarcillin. Sulfonamides include, but are not limited to, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethizole, sulfamethoxazole, sulfanamide, sulfasalazine, sulfisoxazole, trimethoprim-sulfamethoxazole (Co-trimoxazole) (TMP-SMX), and sulfonamidochrysoidine.
In some embodiments, the antibacterial agent used in combination with a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) is selected from the group consisting of Iinezolid, tedizolid, posizolid, radezolid, retapamulin, valnemulin, tiamulin, azamulin, lefamulin, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem, doripenem, imipenem/cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefepime, ceftaroline fosamil, ceftobiprole, teicoplanin, vancomycin, telavancin, dalbavancin, oritavancin, clindamycin, lincomycin, daptomycin, solithromycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, spiramycin, aztreonam, furazolidone, nitrofurantoin, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin g, penicillin v, piperacillin, penicillin g, temocillin, ticarcillin, amoxicillin clavulanate, ampicillin/sulbactam,
piperacillin/tazobactam, ticarcillin/clavulanate, bacitracin, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethizole, sulfamethoxazole, sulfanamide, sulfasalazine, sulfisoxazole, trimethoprim-sulfamethoxazole (tmp-smx), sulfonamidochrysoidine, eravacycline, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol(bs), ethionamide, isoniazid, pyrazinamide, rifampicin, rifabutin, rifapentine, streptomycin, arsphenamine, chloramphenicol, fosfomycin, fusidic acid, metronidazole, mupirocin, platensimycin, quinupristin/dalfopristin, thiamphenicol, tigecycline, tinidazole, and trimethoprim, prodrugs thereof, and pharmaceutically acceptable salts thereof.
In some embodiments, the antibacterial agent used in combination with a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) is tedizolid, azithromycin, meropenem, amikacin, levofloxacin, rifampicin, Iinezolid, erythromycin, or solithromycin. In some embodiments, the antibacterial agent used in combination with a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) is tedizolid, azithromycin, meropenem, amikacin, or levofloxacin. In some embodiments, the antibacterial agent used in combination with a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) is tiamulin. In some embodiments, the antibacterial agent used in combination with a compound described herein (e.g., a compound of any one of formulas (I)- (XXXXX)) is solithromycin. VI. Methods
Methods described herein include, e.g., methods of protecting against or treating a bacterial infection (e.g., a Gram-negative bacterial infection) in a subject and methods of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria (e.g., Gram-negative bacteria). A method of treating a bacterial infection (e.g., a Gram-negative bacterial infection) in a subject includes administering to the subject a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) or a pharmaceutical composition thereof. In some embodiments, the bacterial infection is caused by Gram- negative bacteria. In some embodiments, the bacterial infection is caused by a resistant strain of bacteria. In some embodiments, the resistant strain of bacteria is a resistant strain of E. coli. A method of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria (e.g., Gram-negative bacteria) includes contacting the bacteria (e.g., Gram-negative bacteria) or a site susceptible to bacterial growth (e.g., Gram-negative bacterial growth) with a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) or a pharmaceutical composition thereof. In some embodiments, the bacterial infection is caused by Gram-negative bacteria. In some embodiments, the bacterial infection is caused by a resistant strain of bacteria. In some embodiments, the resistant strain of bacteria is a resistant strain of E. coli. In some embodiments, a compound used in any methods described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) may bind to LPS in the cell membrane of Gram- negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics. Additionally, the monosaccharide or oligosaccharide moieties in the compounds described herein also serve as a gradient against which immune cells chemotax to the site of bacterial infection and/or growth.
Moreover, methods described herein also include methods of protecting against or treating sepsis in a subject by administering to the subject a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)). In some embodiments, the method further includes administering to the subject an antibacterial agent. Methods described herein also include methods of preventing lipopolysaccharides (LPS) in Gram-negative bacteria (e.g., a resistant strain of Gram-negative bacteria or a resistant strain of E. coli (e.g., E. coli BAA-2469)) from activating a immune system in a subject by administering to the subject a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)). In some embodiments of the method, the method prevents LPS from activating a macrophage. In some embodiments, the method further includes administering to the subject an antibacterial agent. In some embodiments, a compound used in any methods described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) may bind to LPS in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics.
In some embodiments, the methods described herein may further include administering to the subject an antibacterial agent in addition to a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)). Methods described herein also include methods of protecting against or treating a bacterial infection in a subject by administering to said subject (1 ) a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) and (2) an antibacterial agent. Methods described herein also include methods of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria, by contacting the bacteria or a site susceptible to bacterial growth with (1 ) a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) and (2) an antibacterial agent.
In some embodiments, the compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) is administered first, followed by administering of the antibacterial agent alone. In some embodiments, the antibacterial agent is administered first, followed by administering of the compound described herein alone. In some embodiments, the compound described herein and the antibacterial agent are administered substantially simultaneously (e.g., in the same pharmaceutical composition or in separate pharmaceutical compositions). In some embodiments, the compound described herein or the antibacterial agent is administered first, followed by administering of the compound described herein and the antibacterial agent substantially simultaneously (e.g., in the same pharmaceutical composition or in separate pharmaceutical compositions). In some embodiments, the compound described herein and the antibacterial agent are administered first substantially simultaneously (e.g., in the same pharmaceutical composition or in separate pharmaceutical compositions), followed by administering of the compound described herein or the antibacterial agent alone. In some embodiments, when a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) and an antibacterial agent are administered together (e.g., substantially simultaneously in the same or separate pharmaceutical compositions, or separately in the same treatment regimen), the MIC of each of the compound and the antibacterial agent may be lower than the MIC of each of the compound and the antibacterial agent when each is used alone in a treatment regimen.
VII. Pharmaceutical Compositions and Preparations
A compound described herein may be formulated in a pharmaceutical composition for use in the methods described herein. In some embodiments, a compound described herein may be formulated in a pharmaceutical composition alone. In some embodiments, a compound described herein may be formulated in combination with an antibacterial agent in a pharmaceutical composition. In some embodiments, the pharmaceutical composition includes a compound described herein (e.g., a compound described by any one of formulas (l)-(XXXXX)) and pharmaceutically acceptable carriers and excipients.
Acceptable carriers and excipients in the pharmaceutical compositions are nontoxic to recipients at the dosages and concentrations employed. Acceptable carriers and excipients may include buffers such as phosphate, citrate, HEPES, and TAE, antioxidants such as ascorbic acid and methionine, preservatives such as hexamethonium chloride, octadecyldimethylbenzyl ammonium chloride, resorcinol, and benzalkonium chloride, proteins such as human serum albumin, gelatin, dextran, and
immunoglobulins, hydrophilic polymers such as polyvinylpyrrolidone, amino acid residues such as glycine, glutamine, histidine, and lysine, and carbohydrates such as glucose, mannose, sucrose, and sorbitol.
Examples of other excipients include, but are not limited to, antiadherents, binders, coatings, compression aids, disintegrants, dyes, emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, sorbents, suspensing or dispersing agents, or sweeteners. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine,
methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.
The compounds herein may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts. These salts may be acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the compounds herein be prepared from inorganic or organic bases. Frequently, the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases are well-known in the art, such as hydrochloric, sulphuric, hydrobromic, acetic, lactic, citric, or tartaric acids for forming acid addition salts, and potassium hydroxide, sodium hydroxide, ammonium hydroxide, caffeine, various amines, and the like for forming basic salts. Methods for preparation of the appropriate salts are well-established in the art.
Representative acid addition salts include, but are not limited to, acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate,
camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, and valerate salts. Representative alkali or alkaline earth metal salts include, but are not limited to, sodium, lithium, potassium, calcium, and magnesium, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, and ethylamine.
Depending on the route of administration and the dosage, a compound herein or a
pharmaceutical composition thereof used in the methods described herein will be formulated into suitable pharmaceutical compositions to permit facile delivery. A compound (e.g., a compound of any one of formulas (l)-(XXXXX)) or a pharmaceutical composition thereof may be formulated to be administered intramuscularly, intravenously (e.g., as a sterile solution and in a solvent system suitable for intravenous use), intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctival, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, orally (e.g., a tablet, capsule, caplet, gelcap, or syrup), topically (e.g., as a cream, gel, lotion, or ointment), locally, by inhalation, by injection, or by infusion (e.g., continuous infusion, localized perfusion bathing target cells directly, catheter, lavage, in cremes, or lipid compositions). Depending on the route of administration, a compound herein or a pharmaceutical composition thereof may be in the form of, e.g., tablets, capsules, pills, powders, granulates, suspensions, emulsions, solutions, gels including hydrogels, pastes, ointments, creams, plasters, drenches, osmotic delivery devices, suppositories, enemas, injectables, implants, sprays, preparations suitable for iontophoretic delivery, or aerosols. The compositions may be formulated according to conventional pharmaceutical practice.
A compound described herein may be formulated in a variety of ways that are known in the art.
For use as treatment of human and animal subjects, a compound described herein can be formulated as pharmaceutical or veterinary compositions. Depending on the subject (e.g., a human) to be treated, the mode of administration, and the type of treatment desired, e.g., prophylaxis or therapy, a compound described herein is formulated in ways consonant with these parameters. A summary of such techniques is found in Remington: The Science and Practice of Pharmacy, 22nd Edition, Lippincott Williams & Wilkins (2012); and Encyclopedia of Pharmaceutical Technology, 4th Edition, J. Swarbrick and J. C. Boylan, Marcel Dekker, New York (2013), each of which is incorporated herein by reference.
Formulations may be prepared in a manner suitable for systemic administration or topical or local administration. Systemic formulations include those designed for injection (e.g., intramuscular, intravenous or subcutaneous injection) or may be prepared for transdermal, transmucosal, or oral administration. The formulation will generally include a diluent as well as, in some cases, adjuvants, buffers, and preservatives. The compounds can be administered also in liposomal compositions or as microemulsions. Systemic administration may also include relatively noninvasive methods such as the use of suppositories, transdermal patches, transmucosal delivery and intranasal administration. Oral administration is also suitable for compounds herein. Suitable forms include syrups, capsules, and tablets, as is understood in the art.
The pharmaceutical compositions can be administered parenterally in the form of an injectable formulation. Pharmaceutical compositions for injection can be formulated using a sterile solution or any pharmaceutically acceptable liquid as a vehicle. Formulations may be prepared as solid forms suitable for solution or suspension in liquid prior to injection or as emulsions. Pharmaceutically acceptable vehicles include, but are not limited to, sterile water, physiological saline, and cell culture media (e.g., Dulbecco's Modified Eagle Medium (DMEM), a-Modified Eagles Medium (a-MEM), F-12 medium). Such injectable compositions may also contain amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, such as sodium acetate and sorbitan monolaurate. Formulation methods are known in the art, see e.g., Pharmaceutical Preformulation and Formulation, 2nd Edition, M. Gibson, Taylor & Francis Group, CRC Press (2009).
The pharmaceutical compositions can be prepared in the form of an oral formulation.
Formulations for oral use include tablets containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients. These excipients may be, for example, inert diluents or fillers (e.g., sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, starches including potato starch, calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate, or sodium phosphate); granulating and disintegrating agents (e.g., cellulose derivatives including microcrystalline cellulose, starches including potato starch, croscarmellose sodium, alginates, or alginic acid); binding agents (e.g., sucrose, glucose, sorbitol, acacia, alginic acid, sodium alginate, gelatin, starch, pregelatinized starch, microcrystalline cellulose, magnesium aluminum silicate, carboxymethylcellulose sodium, methylcellulose, hydroxypropyl methylcellulose, ethylcellulose, polyvinylpyrrolidone, or polyethylene glycol); and lubricating agents, glidants, and antiadhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc). Formulations for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., potato starch, lactose, microcrystalline cellulose, calcium carbonate, calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil. Powders, granulates, and pellets may be prepared using the ingredients mentioned above under tablets and capsules in a conventional manner using, e.g., a mixer, a fluid bed apparatus or a spray drying equipment.
Other pharmaceutically acceptable excipients for oral formulations include, but are not limited to, colorants, flavoring agents, plasticizers, humectants, and buffering agents. Formulations for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., potato starch, lactose, microcrystalline cellulose, calcium carbonate, calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil. Powders, granulates, and pellets may be prepared using the ingredients mentioned above under tablets and capsules in a conventional manner using, e.g., a mixer, a fluid bed apparatus or a spray drying equipment.
Dissolution or diffusion controlled release of a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) or a pharmaceutical composition thereof can be achieved by appropriate coating of a tablet, capsule, pellet, or granulate formulation of the compound, or by incorporating the compound into an appropriate matrix. A controlled release coating may include one or more of the coating substances mentioned above and/or, e.g., shellac, beeswax, glycowax, castor wax, carnauba wax, stearyl alcohol, glyceryl monostearate, glyceryl distearate, glycerol palmitostearate, ethylcellulose, acrylic resins, dl-polylactic acid, cellulose acetate butyrate, polyvinyl chloride, polyvinyl acetate, vinyl pyrrolidone, polyethylene, polymethacrylate, methylmethacrylate, 2-hydroxymethacrylate, methacrylate hydrogels, 1 ,3 butylene glycol, ethylene glycol methacrylate, and/or polyethylene glycols. In a controlled release matrix formulation, the matrix material may also include, e.g., hydrated
methylcellulose, carnauba wax and stearyl alcohol, carbopol 934, silicone, glyceryl tristearate, methyl acrylate-methyl methacrylate, polyvinyl chloride, polyethylene, and/or halogenated fluorocarbon.
The pharmaceutical composition may be formed in a unit dose form as needed. The amount of active component, e.g., a compound described herein (e.g., a compound of any one of formulas (I)- (XXXXX)), included in the pharmaceutical compositions are such that a suitable dose within the designated range is provided (e.g., a dose within the range of 0.01 -100 mg/kg of body weight). VIII. Routes of Administration and Dosages
In any of the methods described herein, compounds herein may be administered by any appropriate route for treating or protecting against a bacterial infection (e.g., a Gram-negative bacterial infection), or for preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria (e.g., Gram- negative bacteria). Compounds described herein may be administered to humans, domestic pets, livestock, or other animals with a pharmaceutically acceptable diluent, carrier, or excipient. In some embodiments, administering comprises administration of any of the compounds described herein (e.g., compounds of any one of formulas (l)-(XXXXX)) or compositions intramuscularly, intravenously (e.g., as a sterile solution and in a solvent system suitable for intravenous use), intradermal^, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally,
intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctival, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, orally (e.g., a tablet, capsule, caplet, gelcap, or syrup), topically (e.g., as a cream, gel, lotion, or ointment), locally, by inhalation, by injection, or by infusion (e.g., continuous infusion, localized perfusion bathing target cells directly, catheter, lavage, in cremes, or lipid compositions). In some embodiments, if an antibacterial agent is also administered in addition to a compound described herein, the antibacterial agent or a pharmaceutical composition thereof may also be administered in any of the routes of administration described herein.
The dosage of a compound described herein (e.g., a compound of any one of formulas (I)- (XXXXX)) or a pharmaceutical compositions thereof depends on factors including the route of administration, the disease to be treated (e.g., the extent and/or condition of the bacterial infection), and physical characteristics, e.g., age, weight, general health, of the subject. Typically, the amount of the compound or the pharmaceutical composition thereof contained within a single dose may be an amount that effectively prevents, delays, or treats the bacterial infection without inducing significant toxicity. A pharmaceutical composition may include a dosage of a compound described herein ranging from 0.01 to 500 mg/kg (e.g., 0.01 , 0.1 , 0.2, 0.3, 0.4, 0.5, 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, 150, 200, 250, 300, 350, 400, 450, or 500 mg/kg) and, in a more specific embodiment, about 0.1 to about 30 mg/kg and, in a more specific embodiment, about 1 to about 30 mg/kg. In some embodiments, when a compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) and an antibacterial agent are administered together (e.g., substantially simultaneously in the same or separate
pharmaceutical compositions, or separately in the same treatment regimen), the dosage needed of the compound described herein may be lower than the dosage needed of the compound if the compound was used alone in a treatment regimen.
A compound described herein (e.g., a compound of any one of formulas (l)-(XXXXX)) or a pharmaceutical composition thereof may be administered to a subject in need thereof, for example, one or more times (e.g., 1 -10 times or more; 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 1 0 times) daily, weekly, monthly, biannually, annually, or as medically necessary. Dosages may be provided in either a single or multiple dosage regimens. The timing between administrations may decrease as the medical condition improves or increase as the health of the patient declines. The dosage and frequency of administration may be adapted by the physician in accordance with conventional factors such as the extent of the infection and different parameters of the subject.
EXAMPLES
General Schemes
The compounds of the disclosure and their precursors may be synthesized by a number of methods known to those skilled in the art. The following Scheme 1 and Scheme 2 illustrate several methods and strategies that may be employed but are not meant to be limiting. Cyclic heptapeptides that comprise residues M1 and M2 may be prepared by synthesis using solid phase peptide methods or by solution phase methods. Alternately, the cyclic heptapeptides that comprise residues M1 and M2 may be derived from certain compounds known as polymyxins or from octapeptins which may be derived from fermentation sources or from synthetic sources. The protected M1 and M2 residues may be obtained by first protection of the polymyxin or octapeptin followed by enzymatic hydrolysis (e.g. with savinase or other enzymes known to those skilled in the art). Synthetic, non-natural polymyxins or their
corresponding cyclic heptapeptides, may also be used in the preparation of M1 and M2 and may be prepared by methods known to those who are skilled in the art.
The cyclic heptapeptides may be optionally derivatized with 1 , 2 or 3 amino acids which may be the same or different. Using a suitable linker group L, the two halves which may be the same or different, may be coupled. Appending a suitable E group, which may or may not contain part of the linker L, and subsequent deprotection affords the compounds of the present disclosure.
Alternatively, the linker group L' may be prepared from the individual amino acid groups and L by methods known to those in the art. L' may be built up sequentially or by a convergent synthesis and then coupled to the cyclic heptapeptides that comprise the residues M1 and M2. The L' group may be assembled with one or more E groups attached or the E group(s) may be appended after complete construction of L'. After deprotection, the compounds of the immediate disclosure may be obtained. Alternatively, when a, b, a', and b' are 1 , A1 -M1 or A2-M2 may be derived from a polymyxin or octapeptin precursor by first protection followed by enzymatic hydrolysis (e.g. with papain). Subsequent coupling with L, E or L'-E groups and deprotection, if required, provides the compounds of the present disclosure. Alternatively, when a, b, c, a', b' and c' are 0, the cyclic heptapeptides that comprise the residues M1 and M2 may be coupled directly with L, E or L'-E groups followed by deprotection, if required, to give the compounds of the immediate disclosure.
Other methods known by those skilled in the art may be employed to prepare symmetric compounds (i.e. those where A1 and A2 are the same and M1 and M2 are the same), or asymmetric compounds where A1 -M1 is different from A2-M2 and where L may or may not be symmetric.
The E group(s) may be appended at the time L' is synthesized or it may be appended after the L' is completely assembled or it may be appended after M1 , M2 and L' are coupled. Using orthogonal protection schemes known to those skilled in the art, allows different E groups (e.g. a-L-Rha, a-Gal epitope, laminarin) to be attached in the same molecule. The E groups themselves may possess part of the final linker group L' and as such, L' would be made up of the linker used to assemble M2-L'-M1 and
(E)n
E-L' to give the compounds of the present disclosure ^ . Scheme 1. Synthesis of compound
Figure imgf000174_0001
E
M2— L'— M
Figure imgf000174_0002
Scheme 2. Synthesis of compounds
Figure imgf000175_0001
Figure imgf000175_0002
Figure imgf000175_0003
General Methods
Preparative HPLC was performed using the following: Teledyne Isco HP C1 8, 50g column. Eluent: CH3CN/H2O/ 0.1 % formic acid or 0.1 % trifluoroacetic acid; various linear gradients as necessary at 40 mL /min on an Isco Combiflash Rf LC unit, or Isco EZ prep HPLC, with UV Detection at 220 and 254 nm on a Luna 5 micron C1 8, 100 A, AXIA 1 00 x 30 mm. Eluent: CH3CN/H2O/ 0.1 % formic acid or 0.1 % trifluoroacetic acid; various linear gradients as necessary at 25 mL /min on the Gilson System ; 215 Liquid Handler, Gilson UV-VIS 155, Gilson 305 pump and Detector. UV detection at 220 and 254 nm.
Analytical LC/MS: High resolution liquid chromatography mass spectrometry (HRES-LC/MS) was performed using a Waters Q-TOF Premier mass spectrometer with an electrospray probe coupled with a Waters Acquity H-class UPLC system with a diode array detector set to collect from 210 nm to 400 nm. A gradient of 0.1 % formic acid in water (A) and 0.1 % formic acid in acetonitrile (B) was run from 15%B to 95% over 10 min using a 100 x 2.1 mm 1 .7 μ Phenomenex Kinetex C18 column at 40 °C.
Liquid chromatography mass spectrometry was performed using an Agilent 6120 mass spectrometer an electrospray probe coupled with an Agilent 1260 HPLC system with a variable wavelength detector set to either 220 nm or 254 nm. A gradient of 0.1 % formic acid in water (A) and 0.1 % formic acid in acetonitrile (B) was run from 15%B to 99% over 3.5 min using a 50x3.0 mm 2.6μ Phenomenex Gemini-NX column at 30°C.
1 H-NMR (1 H NMR) spectra were acquired on a Bruker 300 MHz system using a 5 mm QNP probe and chemical shifts are reported as ppm (δ) downfield from tetramethylsilane.
Compounds herein may be made using synthetic methods known in the art, including procedures analogous to those disclosed below. Exemplary protocols, techniques, reagents, and solvents for synthetic methods known in the art are provided, for example, in Caren, S., Practical Synthetic Organic Chemistry: Reactions, Principles, and Techniques. Wiley, 201 1 , and Benoiton, N.L., Chemistry of Peptide Synthesis. CRC Press, 2005, each of which is incorporated by reference in their entirety.
Example 1. Synthesis of L-Rhamnose-PEG1-NH2 (INT-1 )
Figure imgf000176_0001
Step a. Synthesis of benzyl (2-(2-hydroxyethoxy)ethyl)carbamate
To a solution of 2-(2-aminoethoxy)ethanol (10.5 g, 0.1 mol) in 200 mL THF was added Hunig's base (28 mL, 0.2mol) under the ice-water bath, then CBz-CI (18g, 0.1 mol) in 50 mL THF was added dropwise to the above cooled solution. The resultant solution was stirred for overnight and then concentrated to remove solvent, the residue was partitioned by ethyl acetate (300mL) and water 150 mL, the organic layer was washed by 5% sodium bicarbonate (100 mL), brine (1 00 mL), and 1 N HCI (1 00 mL) and then brine, respectively. The organic layer was dried and concentrated for next step. Yield of products 18 g, 75%. Ή NMR (300 MHz, DMSO-afe) δ 7.34 (d, J = 3.2 Hz, 5H), 5.01 (d, J = 2.5 Hz, 2H), 3.40 (m, 8H).
Step b. Synthesis of (2R,3R,4R,5S,6S)-2-(2-(2-(((benzyloxy)carbonyl)amino)ethoxy)ethoxy)-6- methyltetrahydro-2H-pyran-3,4,5-triyl triacetate
To a solution of benzyl (2-(2-hydroxyethoxy)ethyl)carbamate (14g, 56 mmol) and per-acetyl-L- rhamnose (20 g, 56 mmol) in 200 mL dry DCM under the ice-water bath, then BF3-Et20 (15 mL, 1 10 mmol) was added dropwise to the above cooled solution. After stirring overnight, the reaction was quenched with saturated NaHCCb (aq) and diluted with EtOAc. The organic phase was washed sat. NaHC03 (aq) and brine. The combined organic layers were dried over anhydrous Na2S04. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 1 00% acetonitrile and water, using 0.1 %
trifluoroacetic acid as the modifier. The product was isolated as an oil, 18 g (63%). LC/MS [M]+1 512.2.
Step c. Synthesis of benzyl (2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H- pyran-2-yl)oxy)ethoxy)ethyl)carbamate
A solution of (2R,3R,4R,5S,6S)-2-(2-(2-(((benzyloxy)carbonyl)amino)ethoxy)ethoxy)-6- methyltetrahydro-2H-pyran-3,4,5-triyl triacetate (26 g, 50 mmol) and hydrazine (25 mL) in anhydrous methanol was stirred overnight, The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil, 18.5 g (95%). LC/MS [M+H]+ 386.2. The material was subjected to high vacuum to remove excess hydrazine.
Step d. Synthesis of (2R,3R,4R,5R,6S)-2-(2-(2-aminoethoxy)ethoxy)-6-methyltetrahydro-2H-pyran- 3,4,5-triol (L-Rhamnose-PEG1-NH2, INT-1)
Benzyl (2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2- yl)oxy)ethoxy)ethyl)carbamate (14.6 mg, 38 mmol) was dissolved into 30 mL MeOH and 30 mL ethyl acetate, then 5 mg of 5% Pd on charcoal was added above solution, and the mixture was stirred at room temperature under a hydrogen atmosphere overnight. The palladium on charcoal was removed by filtration after completion of the reaction by LCMS. The filtrate was concentrated and used in the next step without any purification. 1 H NMR (300 MHz, methanol-dt) δ 4.75(s, 1 H) 3.84 (ddt, J = 8.5, 3.3, 1 .6 Hz,
2H), 3.76 - 3.52 (m, 7H), 3.47 - 3.28 (m, 4H), 3.15 (t, J = 5.1 Hz, 2H), 1 .28 (d, J = 6.2 Hz, 3H). 13C NMR (75 MHz, methanol-ck) δ 100.36, 72.52, 70.93, 70.72, 70.1 1 , 68.45, 66.60, 66.27, 39.29, 16.61 . LCMS 252.2 [M+H]+ Example 2. Synthesis of 4-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4- oxobutanoic acid (INT-2)
Procedu e A
Succinic anhydride (156 mg, 1 .57 mmol) was added to a stirring mixture of INT-1 (375 mg, 1 .49 mmol) and DIEA (193 mg, 1 .49 mmol) in MeOH (7 mL) and the reaction was stirred at RT for 3 hours at room temperature. The solvent was reduced by 80% on the rotary evaporator and the mixture was applied to reversed phase HPLC (5 to 50% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were pooled and lyophilized to afford 4-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoic acid (INT-2) as a clear vicous oil. Yield: 75%. LC/MS [M-H-]- 350.2. Procedu e B
Figure imgf000178_0001
Figure imgf000178_0002
Step a. Synthesis of benzyl (2-(2-hydroxyethoxy)ethyl)carbamate
To the solution of 2-(2-aminoethoxy)ethanol (301 .0 g, 2.863mol, 1 .Oeq) in 1500ml_ DCM was added Et3N (343. Og, 3.390mol, 1 .2eq) under the ice-water bath (control the temperature below 20°C), then CBz-CI (488. Og, 2.863mol, 1 .Oeq) was added dropwise to the above cooled solution (control inside temperature below 20°C). The resultant solution was stirred for overnight (>16 hrs) and then washed with water 1500 mL, sat. NaHC03 (1500 mL), 1 N HCI (1500 mL) and brine (1500 mL), respectively. The organic layer was dried (200 g of Na2S04) and concentrated for next step directly. Yield of product 639 g, 92%. Colorless oil.
Step b. Per-acetylation of L-rhamnose
To a solution of L-rhamnose (69.0 g, 0.3799 mol, 1 .0 eq) and DMAP (1 .40 g, 0.02x) in pyridine (280 mL, 4V/M) was added acetic anhydride (232.0 g, 2.273 mol, 6.0 equiv) dropwise under the ice-water bath (to control the temperature of the reaction below 20°C), The mixture was stirred at ambient temperature (>12 hrs), and volatiles were removed under reduced pressure (toluene azeotrope). The crude product was dissolved in EtOAc (800 mL), washed with hydrochloric acid (1 N, 800 mL*2), sat. NaHCO3 (800 mL), brine (800 mL), and dried with sodium sulfate (100 g). Volatiles were removed under reduced pressure, and crude per-acetyl-L-rhamnose (131 .0 g, yield: 100%) was used directly for the next step. Step c. Synthesis of (2R,3R,4R,5S,6S)-2-(2-(2-(((benzyloxy)carbonyl)amino)ethoxy)ethoxy)-6- methyltetrahydro-2H-pyran-3,4,5-triyl triacetate
Figure imgf000179_0001
To the solution of benzyl (2-(2-hydroxyethoxy)ethyl)carbamate (48.8 g, 0.204 mol, 1 .20 eq) and per-acetyl-L-rhamnose (56.6 g, 0.170 mol, 1 .00 eq) in 500 mL dry DCM under the ice-water bath, then BF3-Et20 (52.5 mL, 0.416 mol, 2.5 eq) was added dropwise to the above cooled solution (control the temperature below 20°C). After stirring overnight, the reaction was quenched by 700 mL of sat. NaHCCb (aq), concentrated and purified by chromatography on silica gel (90g/500g), eluent:PE (1 L),
PE/EA=5/1 (10.8L), PE/EA=3/1 (20L). Yield: 50.0 g, 57%, of a colorless oil.
Step d. Synthesis of benzyl (2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H- pyran-2-yl)oxy)ethoxy)ethyl)carbamate
Figure imgf000179_0002
To a solution of (2R,3R,4R,5S,6S)-2-(2-(2-(((benzyloxy)carbonyl)amino)ethoxy)ethoxy)-6- methyltetrahydro-2H-pyran-3,4,5-triyl triacetate (33.7 g, 0.066 mol, 1 .0 eq) in methanol (200 mL) was added NaOMe (1 .14 g in 50 mL of MeOH, 0.050 mol, 0.75 eq) and stirred overnight. The resultant solution was concentrated and dissolved in 800 mL of THF, then filtered through a pad of silica gel (20 g), and washed with THF (400 mL*3) and concentrated to give a colorless oil (23.4 g, 92%). Step e. Synthesis of (2R,3R,4R,5R,6S)-2-(2-(2-aminoethoxy)ethoxy)-6-methyltetrahydro-2H-pyran- 3,4,5-triol (L-Rhamnose-PEG1-NH2) (INT-1)
Figure imgf000179_0003
Benzyl (2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2- yl)oxy)ethoxy)ethyl)carbamate (21 .2 g, 0.066 mol, 1 .0 eq) was dissolved into 300 mL MeOH, then 2.2 g of 5% palladium on charcoal (0.1 x) was added to the above solution and the mixture was stirred at room temperature under the hydrogen atmosphere for overnight. The palladium on charcoal was removed by filtration after the reaction was complete as judged by TLC. The filtrate was concentrated to give INT-1 as a colorless oil (13.4 g, 97%). LCMS 252.2[M+H]+ Step f. Synthesis of 4-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4- oxobutanoi -2)
Figure imgf000180_0001
A solution of INT-1 (10.93 g, 0.043 mol, 1 .0 eq) and succinic anhydride (4.35 g, 0.043 mol, 1 .0 eq) in anhydrous MeOH (1 00 mL) was stirred at RT for overnight (>12 hrs). Then the solvent was removed and the residue was purified by flash silica gel column chromatography (18 g/100 g), eluent: DCM/MeOH=10/1 (1 .8L), DCM/MeOH=6/1 (1 .6L) to give INT-2 (12.1 g, 82 %) as a colorless oil. 1 HNMR (400 MHz, methanol-ck): 4.72 (s, 1 H); 3.82-3.75 (m, 2H); 3.67-3.53 (m, 7H); 3.39-3.35 (m, 2H); 3.31 (m, 1 H); 2.61 -2.47 (m, 4H); 1 .26 (d, 6.4 Hz, 3H).
Example 3. Synthesis of 2-aminoethyl 2,3,4,6-tetra-0-acetyl-beta-D-glucopyranosyl-(1->3)-2,4,6-tri-
0-acetyl-3-thio-beta-D-glucopyranosyl-(1->3)-2,4,6-tri-0-acetyl-1 ,3-dithio-beta-D-glucopyranoside
(INT-3)
Figure imgf000180_0002
Figure imgf000181_0001
Step a. Synthesis of (3aR,5R,6R,6aR)-5-((R)-2,2-dimethyl-1 ,3-dioxolan-4-yl)-2,2- dimethyltetrahydro-2H-furo[2,3-d][1 ,3]dioxol-6-yl trifluoromethanesulfonate
Triflic anhydride (7.8 mL, 46.39 mmol) was added over 30 minutes to a cold (0°C) solution of the diacetone allofuranose (10.50 g, 40.34 mmol) in pyridine (100 mL). After stirring in the ice-bath for 1 hour, the reaction was diluted with ethyl acetate (250 mL) and the organic layer was washed with a mixture of saturated sodium bicarbonate and brine (50 mL + 50 mL), dried (Na2S04) and concentrated under reduced pressure to provide the crude triflate. The crude residue was purified using a normal phase flash column (0-10% EA/Hex) to give the title compound (14.15 g, 89%) as a colorless oil. 1 HNMR (300 MHz, Chloroform-d): 5.85 (d, 1 H); 4.93 (dd, 1 H); 4.79 (dd, 1 H); 4.21 (dd, 1 H); 4.1 1 -4.17 (dt, 1 H); 3.90-3.95 (dt, 1 H); 1 .61 (s, 3H); 1 .47 (s, 3H); 1 .41 (s, 3H); 0.3 (s, 3H).
Step b. Synthesis of Thio-linked Disaccharide: ((2R,3R,4S,5R,6S)-2-[(acetyloxy)methyl]-6- {[(3aR,5R,6S,6aS)-5-(2,2-dimethyl-1 ,3-dioxolan-4-yl)-2,2-dimethyltetrahydro-2H-furo[2,3- d][1 ,3]dioxol-6-yl]sulfanyl}oxane-3,4,5-triyl triacetate)
Sodium hydride was added to a solution of 2,3,4,6-tetra-0-acetyl-l-thio-p-D-glucopyranose (14.00 g, 38.42 mmol) in dry THF (450 mL in a 2000-mL RB Flask) at 0 °C. The suspension was stirred under nitrogen until hydrogen formation had ceased. To this solution, 1 ,7,10-trioxa-4,13-diazacyclopentadecane (Kryptofix 21 , 4,10-diaza-16-crown-5-ether, 1 .53 g, 9.92 mmol) and a solution of (3aR,5R,6R,6aR)-5-((R)- 2,2-dimethyl-1 ,3-dioxolan-4-yl)-2,2-dimethyltetrahydro-2H-furo[2,3-d][1 ,3]dioxol-6-yl
trifluoromethanesulfonate (13.09 g, 46.39 mmol) in THF (200 mL) was added via cannula transfer and the mixture was stirred for 2 h at 0 °C under nitrogen, then at room temperature for overnight. The reaction mixture was concentrated under reduced pressure. The residue was dissolved in DCM (400 mL), washed with water (200 mL), dried (Na2S04), and concentrated. Added 40 mL of EtOH to the residue to dissolve the foam followed by swirling the flask. The foam slowly turn to white solid. After standing still for 5 minutes, breaking down the solid cake and filtering and washing with 20% aq. EtOH (100 mL). After drying, 10.28 g of the title compound was obtained (44%). Check TLC of the mother liquor and some of the product remained in the mother liquor which was purified with normal phase flash column. 1 HNMR (300 MHz, Chloroform-d): 5.86 (d, H), 5.25 (dd, 1 H), 5.10 (m, 2H), 4.86 (d, H), 4.76 (d, H), 3.95-4.42 (m, 6H), 3.64-3.78 (m, H), 3.58 (d, H), 2.1 1 (s, 3H), 2.09 (s,3H), 2.06 (s, 3H), 2.03 (s, 3H), 1 .54 (s, 3H), 1 .45 (s, 3H), 1 .35 (s, 3H), 1 .35 (s, 3H) Step c. Synthesis of 1 ,2,4,6-tetra-0-acetyl-3-S-(2,3,4,6-tetra-0-acetyl-beta-D-glucopyranosyl)-3- thio-D-glucopyranose
A solution of ((2R,3R,4S,5R,6S)-2-[(acetyloxy)methyl]-6-{[(3aR,5R,6S,6aS)-5-(2,2-dimethyl-1 ,3- dioxolan-4-yl)-2,2-dimethyltetrahydro-2H-furo[2,3-d][1 ,3]dioxol-6-yl]sulfanyl}oxane-3,4,5-triyl triacetate) (10.28 g) in 60% aq. AcOH/TFA (20:1 , 1 05 mL) was heated for 5 hours at 70 °C, then concentrated under reduced pressure and the residue dissolved in water and freeze-dried. Conventional acetylation of the resulting solid (1 :1 Ac20/pyridine, 200 mL) yielded the product. Rotovaped to remove most of Ac20/Py, diluted with DCM (400 mL) and washed with 0.5 N HCI 200 mL. 200 mL of DCM extracted the aqueous layer once. Combined DCM extract and dried with Na2S04. DCM was removed under reduced pressure. The residue was precipitated with 30% EA/Hex to give a white solid. Filtration and 30%EA/Hex wash and dry. Check TLC of mother liquor, the impurities and the desired product have almost equal amount.
Purification was not performed due to the small amount in the mother liquor. The precipitation gave peracetylated (1 -3>)-thiodisaccharide (1 ,2,4,6-tetra-0-acetyl-3-S-(2,3,4,6-tetra-0-acetyl-beta-D- glucopyranosyl)-3-thio-D-glucopyranose) 9.770 g (83.0%) and a column purification gave 1 .384 g (1 1 .8%). 1 HNMR showed alpha/beta ratio 1 :1 (from ~H NMR integration of H-1 signals).
Step d. Synthesis of (1->3)-Thiodisaccharide Bromide: 2,4,6-tri-0-acetyl-3-S-(2,3,4,6-tetra-0-acetyl- beta-D-g lucopyranosyl)-3-th io-alpha-D-g lucopyranosyl bromide
To a solution of 1 ,2,4,6-tetra-0-acetyl-3-S-(2,3,4,6-tetra-0-acetyl-beta-D-glucopyranosyl)-3-thio- D-glucopyranose (1 1 .15 g, 16.06 mmol) in dry DCM (1 60 mL) at 0 °C was added dropwise commercial 33% HBr in AcOH (34.9 mL, 12 equiv.). After the mixture was stirred for 3 hours in an ice-bath, TLC (1 :1 EtOAc-hexane) showed complete conversion of the SM into a single product. DCM was added (240 mL), and the solution was washed with cold water (2 x 100 mL) and dried with Na2S04. DCM was removed under reduced pressure. The residue was precipitated with 30% EA/Hex to give a white solid. Filtration and 30% EA/Hex wash and dry to give the title compound (10.50 g, 91 .4%) as white a powder. 1 HNMR spectrum was identical with literature data for the compound [B. Sylla, et al.; J. Med.
Chem., 2014, 57 (20), pp 8280-8292].
Step e. Synthesis of (1->3)-Thiodisaccharide Thiol: 2,4,6-tri-0-acetyl-3-S-(2,3,4,6-tetra-0-acetyl- beta-D-glucopyranosyl)-1 ,3-dithio-beta-D-glucopyranose
Thiourea (5.56 g, 73.4 mmol, 5 equiv.) was added into a solution of the 2,4,6-tri-0-acetyl-3-S- (2,3,4,6-tetra-0-acetyl-beta-D-glucopyranosyl)-3-thio-alpha-D-glucopyranosyl bromide (10.5 g, 14.6 mmol) from Step d. in dry acetone (1 00 mL). The reaction mixture was refluxed with TLC monitoring. After 10 minutes, the reaction mixture became a clear solution. After 2hrs, another 5 equiv. of thiourea was added and the mixture was refluxed for another 2 hours. After completion of conversion of starting material (checked by TLC), the reaction mixture was cooled to room temperature. Acetone was removed by reduced pressure. To the residue solid was added Chloroform and water (1 :1 , 300 mL) and stirred at 85 °C for 1 hour with condenser. The mixture was cooled to room temperature and extracted with DCM (3 x 200 mL). Dried and remove solvent. Crystallization in EtOH gave the title compound as a white solid (9.01 g, 92%). H NMR (300 MHz, Chloroform-d): 5.17 (dd, 1 H), 4.95-5.12 (m, 2H), 4.85-4.98 (m, 2H), 4.67 (d, 1 H), 4.43 (dd, 1 H), 4.27 (ddd, 1 H), 4.18 (m, 1 H), 4.06-4.16 (2H), 3.64-3.76 (m, 2H), 2.96 (dd, 1 H), 2.35 (d, 1 H, SH), 2.16 (s, 3H), 2.09 (s, 3H), 2.08 (s, 3H), 2.06 (s, 3H), 2.02 (s, 3H), 2.01 (s, 3H), 1 .00 (s, 3H). 13C NMR (75 MHz, Chloroform-d): 170.6 (CO), 170.5 (CO), 170.2 (CO), 169.4 (CO), 169.3 (CO), 169.1 (2CO), 84.1 (CH), 80.5 (CH), 78.5 (CH), 75.7 (CH), 75.4 (CH), 73.6 (CH), 70.1 (CH), 68.2 (CH), 66.5 (CH), 52.5 (CH), 61 .9 (CH), 52.0 (CH), 21 .0 (CH3), 20.8 (CH3), 20.7 (CH3), 20.5 (3 CH3), 20.4 (CH3).
Step f. Synthesis of Thio-linked Trisaccharide: 2,4,6-tri-0-acetyl-3-S-(2,3,4,6-tetra-0-acetyl-beta-D- glucopyranosyl)-1 ,3-dithio-beta-D-glucopyranose
Sodium hydride (0.808 g, 29.2 mmol) was added to a solution of 2,4,6-tri-0-acetyl-3-S-(2,3,4,6- tetra-0-acetyl-beta-D-glucopyranosyl)-1 ,3-dithio-beta-D-glucopyranose (9.01 g, 13.47 mmol) in dry THF (450 mL in a 2000-mL RB Flask) at 0 °C. The suspension was stirred under nitrogen until hydrogen formation had ceased. To this solution, 1 ,7,10-trioxa-4,13-diazacyclopentadecane (Kryptofix 21 , 4,10- diaza-16-crown-5-ether, 0.535 g, 2.43 mmol) and a solution of (3aR,5R,6R,6aR)-5-((R)-2,2-dimethyl-1 ,3- dioxolan-4-yl)-2,2-dimethyltetrahydro-2H-furo[2,3-d][1 ,3]dioxol-6-yl trifluoromethanesulfonate (5.551 g, 14.1 5 mmol) in THF (200 mL) was added via cannula transfer and the mixture was stirred for 2 hours at 0 °C under Nitrogen, then at room temperature for overnight. The reaction mixture was concentrated under reduced pressure. A solution of the residue was dissolved in DCM (400 mL), washed with water (200 mL), dried (Na2S04), and concentrated. Added 40 mL of EtOH to the crude product to dissolve the foam followed by swirling the flask. The white precipitate was formed. Filtration and wash with 30% EtOAc/Hex and drying gave the title compound 6.55 g (53.4%). Check TLC of the mother liquor and some of the product remained in the mother liquor which was purified with normal phase flash column. 1 H NMR (300 MHz, Chloroform-d): δ 5.83 (d, J = 3.5 Hz, 1 H), 5.18 (dd, J = 9.3 Hz, 1 H), 5.07 (dd, J = 10.2 Hz, 1 H), 5.06 (dd, J = 9.6 Hz, 1 H), 4.88 (ddd, J = 9.7, 9.7, 9.9 Hz, 1 H), 4.83 (d, J = 3.6 Hz, 1 H), 4.63 (dd, J = 14.2, 10.0 Hz, 2H), 4.37 - 4.21 (m, 3H), 4.21 - 4.05 (m, 5H), 4.04 - 3.96 (m, 1 H), 3.76 - 3.61 (m, 2H), 3.54 (d, J = 3.7 Hz, 1 H), 3.00 (dd, J = 10.8 Hz, 1 H), 2.15 (s, 3H), 2.10 (s, 3H), 2.08 (s, 3H), 2.08 (s, 3H), 2.02 (s, 3H), 2.01 (s, 3H), 1 .99 (s, 3H), 1 .51 (s, 3H), 1 .42 (s, 3H), 1 .34 (s, 3H), 1 .32 (s, 3H). 13C NMR (75 MHz, Chloroform-d): δ 170.53 (e), 170.48(e), 170.16(e), 169.42(e), 1 69.26(e), 169.09(e), 168.47(e), 1 1 1 .92(e), 109.45(e), 104.82(o), 86.04(o), 84.52(o), 84.31 (o), 80.1 1 (o), 78.43(o), 75.59(o), 73.72(o), 73.62(o), 71 .74(0), 70.02(0), 68.15(o), 67.31 (e), 66.62(o), 62.51 (e), 61 .92(e), 52.18(o), 50.07(o), 26.87(o, CH3), 26.59(0, CH3), 26.32(0, CH3), 25.33(o, CH3), 20.83(o, CH3), 20.73(o, CH3), 20.68(o, CH3), 20.53(o, 3CH3), 20.39(0, CH3).
Step g. Synthesis of Peracetylated (1->3)-Thiotrisaccharide: 2,3,4,6-tetra-O-acetyl-beta-D- glucopyranosyl-(1->3)-2,4,6-tri-0-acetyl-3-thio-beta-D-glucopyranosyl-(1->3)-1 ,2,4,6-tetra-0-acetyl- 3-thio-D-glucopyranose
A solution of 2,4,6-tri-0-acetyl-3-S-(2,3,4,6-tetra-0-acetyl-beta-D-glucopyranosyl)-1 ,3-dithio-beta- D-glucopyranose 6.55 g, 7.19 mmol) in 60% aq. AcOH/TFA (10:0.5, 105 mL) was heated for 5 hours at 70 °C, then concentrated under reduced pressure and the residue dissolved in water and freeze-dried. Conventional acetylation of the resulting solid (1 :1 Ac20/pyridine, 1 00 mL) yielded the desired product. Rotovaped to remove most of Ac20/Py, diluted with DCM (400 mL) and washed with 0.5N HCI 200 mL. 200 mL of DCM extracted the aqueous layer once. Combined DCM extract and dried with Na2S04. DCM was removed by reduced pressure. Normal phase silica column purification (0-70% EtOAc/Hex) gave the title compound (6.51 g, 90.6%) as a white solid. Step h. Synthesis of (1->3)-Thiotrisaccharide Bromide: 2,3,4,6-tetra-O-acetyl-beta-D- glucopyranosyl-(1->3)-2,4,6-tri-0-acetyl-3-thio-beta-D-glucopyranosyl-(1->3)-1 ,2,4,6-tetra-0-acetyl- 3-thio-D-glucopyranose
To a solution of the 2,3,4,6-tetra-0-acetyl-beta-D-glucopyranosyl-(1 ->3)-2,4,6-tri-0-acetyl-3-thio- beta-D-glucopyranosyl-(1 ->3)-1 ,2,4,6-tetra-0-acetyl-3-thio-D-glucopyranose (6.51 g, 6.52 mmol) in dry CH2CI2 (70 mL) at 0 °C was added dropwise commercial 33% HBr in AcOH (14.2 mL, 12 equiv.). After the mixture was stirred for 3 hours at ice-bath, TLC (1 :1 EtOAc-hexane) showed complete conversion of the starting material into a single product. DCM was added (200mL), and the solution was washed with cold water (100 mL). DCM was used to extract on the aqueous layer. Combined DCM layers were washed with water (1 00 mL). Dried and removed solvent. Ethanol (Hot, 50 mL) dissolved the residue to precipitate the title compound (4.43 g, 67%). 1 H NMR (300 MHz, chloroform-d): 6.65 (d, J = 3.63, 1 H), δ 5.18 (dd, J = 9.3 Hz, 1 H), 5.02 - 5.13 (m, 2H), 4.80 - 5.01 (m, 4H), 4.72 (d, J = 9.7, 1 H), 4.65 (d, J = 1 0.2, 1 H), 4.07 - 4.35 (m, 7H), 3.67 - 3.78 (m, 2H), δ 3.32 (dd, J = 1 1 .1 Hz, 1 H), 3.00 (dd, J = 10.5 Hz, 1 H), 2.16 (s, 3H), 2.14 (s, 3H), 2.12 (s, 6H), 2.1 1 (s, 3H), 2.10 (s, 3H), 2.08 (s, 3H), 2.04 (s, 3H), 2.02 (s, 3H), 2.00 (s, 3H). 13C NMR (75 MHz, Chloroform-d): δ 170.55 (e), 170.53 (e), 170.50 (e), 1 70.21 (e), 169.43 (e), 169.40(e), 169.38 (e), 169.30 (e), 169.10 (e), 168.53 (e), 89.00 (0), 84.23 (0), 77.82 (0), 75.58 (0), 73.61 (0), 73.61 (0), 73.00 (0), 71 .83 (0, 2C), 71 .48 (0), 69.96 (0) , 68.01 (0), 66.41 (0), 64.65 (0), 62.32 (e), 61 .86 (e), 61 .51 (e), 52.17 (0), 46.83 (0), 20.78 (0), 20.74 (0, 2C), 20.67 (0), 20.64 (0), 20.60 (0, 3C), 20.54 (0), 20.44 (0). Step i. Synthesis of (1->3)-Thiotrisaccharide Thiol: 2,3,4,6-tetra-0-acetyl-beta-D-glucopyranosyl-(1- >3)-2,4,6-tri-0-acetyl-3-thio-beta-D-glucopyranosyl-(1->3)-2,4,6-tri-0-acetyl-1 ,3-dithio-beta-D- glucopyranose
A similar procedure was used as that in Example 3, Step e. Crystallization from EtOH gave 3.50 g of the title compound as a white solid. Column purification of mother liquor gave a white solid (0.55 g). The yield was 95.9%. Ή NMR (300 MHz, Chloroform-d): δ 5.16 (dd, J = 9.3 Hz, 1 H), 4.99 -5.12 (m, 2H), 4.79 - 4.80 (m, 4H), 4.64 (d, J = 9.9, 1 H), 4.61 (d, J = 4.0, 1 H), 4.41 (t, J = 4.5 Hz, 1 H), 4.30 - 4.04 (m, 6H), 3.60 - 3.75 (m, 3H), 2.85 - 3.00 (m, 2H), 2.38 (d, J = 9.9 Hz, 1 H), 2.14 (s, 3H), 2.10 (s, 3H), 2.08 (s, 9H), 2.07 (s, 6H), 2.01 (s, 3H), 1 .99 (s, 3H), 1 .98 (s, 3H). 13C NMR (75 MHz, Chloroform-d): δ 170.68 (e), 170.64 (e), 170.55 (e), 170.17 (e), 169.50 (e), 169.39 (e), 169.29 (e), 169.1 0 (e), 1 68.95 (e), 1 68.56 (e), 84.87 (0), 84.10 (0), 80.73 (0), 78.45 (0), 77.84 (0), 75.55 (0), 74.99 (0), 73.60 (0), 71 .84 (0), 69.96 (0), 68.04 (0), 66.65 (0), 66.14 (0), 62.49 (e, 2C), 61 .84 (e), 52.12 (0), 51 .84 (0), 20.98 (0), 20.88 (0), 20.77 (0), 20.74 (0), 20.65 (0), 20.59 (0), 20.55 (0, 3C), 20.41 (0). Step j. Synthesis of Peracetylated (1->3)-Thiotrisaccharide ethyleneamine: 2-aminoethyl 2,3,4,6- tetra-0-acetyl-beta-D-glucopyranosyl-(1->3)-2,4,6-tri-0-acetyl-3-thio-beta-D-glucopyranosyl-(1->3)- 2,4,6-tri-0-acetyl-1 ,3-dithio-beta-D-glucopyranoside (INT-3)
Part 1 (Mitsunobu Reaction). Trimethylphosphine (TMP, 1 .0M in THF, 2.1 ml_, 2.06 mmol) was added to a solution of 1 ,1 '-(azodicarbonyl)dipiperidine (ADDP, 0.519 g, 2.06 mmol) in 25 mL of THF at 0°C and stirred for 30 min. N-Cbz-ethanolamine (0.201 g, 1 .03 mmol) and 2,3,4,6-tetra-O-acetyl-beta-D- glucopyranosyl-(1 ->3)-2,4,6-tri-0-acetyl-3-thio-beta-D-glucopyranosyl-(1 ->3)-2,4,6-tri-0-acetyl-1 ,3-dithio- beta-D-glucopyranose (0.54 mmol in 5 mL of THF) were added sequentially to the solution, with further stirring at room temperature for 2 hours. Any precipitate was then filtered off and the solution evaporated to dryness. Normal phase silica gel column purification (0-70% EtOAc/Hex) gave the desired product as a white foam (0.946 g, 80%). 13C NMR (75 MHz, Chloroform-d): δ 170.70 (e), 1 70.59 (e), 1 70.57 (e), 170.20 (e), 169.46 (e), 169.36 (e), 169.30 (e), 169.12 (e), 168.66 (e), 168.57 (e), 1 56.27 (e), 136.44 (e), 128.54 (o, 2C), 128.19 (o, 3C), 86.05 (o), 85.16 (o), 84.10 (o), 78.1 1 (o), 77.75 (o), 75.56 (o), 73.60 (o), 71 .73 (o), 71 .23 (o), 69.95 (o), 68.04 (o), 66.70 (e), 66.59 (o), 66.29 (o), 62.46 (e), 61 .85 (e), 52.18 (o), 51 .81 (o), 41 .13 (e), 31 .35 (e), 20.94 (o), 20.85 (o), 20.77 (o), 20.67 (o, 2C), 20.61 (o), 20.58 (o, 2C), 20.55 (o), 20.44 (o).
Part 2 (Hydrogenolysis). The compound from Example 3, Step j, Part 1 (0.500 g, 0.435 mmol) and 30% Pd/C (0.463 g, 1 .30 mmol) were in the flask with 20 mL of EtOH and 5 mL of CHCI3. Under H2, the reaction mixture was stirred overnight. Check TLC and mass. Celite filtration and wash with EtOH gave the title compound 0.44 g (100%). Mass showed a strong detectable positive charge signal at tr=3 min with 8 min (5~95%)/highMW method (found M+H+: 101 6.2).
Figure imgf000185_0001
INT-3 (1 eq) is dissolved in methanol and excess NaOMe (10 eq) is added. The reaction is stirred until LCMS shows substantial formation of the desired product (INT-4) with an exact mass of 595.14.
Example 5. Synthesis of (1->3)-Thiotrisaccharide ethyleneamine: 4-[(2-{[beta-D-glucopyranosyl-(1 >3)-3-thio-beta-D-glucopyranosyl-(1->3)-3-thio-beta-D-glucopyranosyl]thio}ethyl)amino]-4- oxobutanoic acid (INT-5)
Figure imgf000185_0002
Step a. Succinic anhydride (0.0879 g, 0.869 mmol) was added to INT-3 (0.4417 g, 0.435 mmol) in DCM/Py (20 mL). The reaction mixture was stirred at room temperature overnight. LC/MS 8min Positive/negative showed the desired peak mass at 4.3 min on negative ionization. The solvent was removed and the residue was loaded onto an Isco Gold 24g column and eluted with 4-6%MeOH/DCM to give 0.27 g of the desired product. 13C NMR (75 MHz, Chloroform-d) δ 172.68, 1 70.94, 170.88, 170.65, 170.23, 169.62, 169.44, 169.35, 169.16, 168.99, 168.64, 85.52, 85.30, 84.08, 78.1 0, 77.57, 75.52, 73.60, 71 .75, 71 .32, 69.95, 68.03, 66.60, 66.41 , 62.56, 61 .86, 52.1 6, 51 .87, 50.68, 39.40, 30.89, 30.22, 29.68, 20.98, 20.86, 20.78, 20.66, 20.63, 20.58, 20.43. Step b. NaOMe in MeOH (25%, 1 .42 mL, 25 equiv.) was added into a 50-mL RB Flask which contained the product of Step a (0.270 g, 0.242 mmol) in 5 mL of MeOH. The mixture was stirred at room temperature for 60 hours. It was then acidified with 1 N aq. HCI and purified with reversed phase C18 column (0-3% CAN/water) to give 0.168 g (99.8%) of the title compound (INT-5). LC/MS 8 min pos/neg method showed negative mass at the very beginning (M-H, 694.2). 13C NMR (75 MHz, water-d): δ 181 .01 (e), 175.85 (e), 86.73 (o), 85.96 (o), 84.51 (o), 81 .72 (o), 81 .58 (o), 79.67 (o), 77.08 (o), 72.57 (o), 72.24 (o), 71 .81 (o), 69.27 (o), 66.88 (o, 2C), 61 .02 (e), 60.92 (e), 60.59 (e), 56.01 (o), 55.76 (o), 39.36 (e), 32.92 (e), 32.25 (e), 29.10 (e).
Figure imgf000186_0001
Step a. Synthesis of 1 ,2,3,4-tetra-0-acetyl-6-deoxy-6-iodo-beta-D-glucopyranose
Iodine (9.42 g, 37.1 mmol) was added at room temperature to a mixture of 1 ,2,3,4-tetra-O-acetyl- beta-D-glucose (9.93 g, 28.55 mmol) and chlorodiphenylphosphine (CIDPP, 6.82 mL, 37.1 mmol) and imidazole (4.28 g, 62.8 mmol) in toluene (-100 mL). After 5 minutes of stirring the reaction mixture started to produce heat. The temperature rose to about 50 °C. After 30 minutes the reaction mixture was heated at 50 °C for 6 hours when TLC indicated that the reaction was complete. Workup: The reaction mixture was cooled to room temperature and poured into an equal volume of saturated Na2C03 in a separatory funnel. The funnel was shaken for 5 minutes while iodine was added portion wise until the toluene phase remained iodine-colored. The aqueous layer was separated and the organic layer was washed with saturated aqueous Na2S203 to remove the excess iodine and then washed with water and brine. Dried and removed the solvent. Purified with 0-40% EA/Hex to give 8.02 g of iodide 2 as a white solid (61 %). 1 H NMR (300 MHz, Chloroform-d) δ 5.77 (d, J = 8.2 Hz, 1 H), 5.28 (dd, J = 9.4 Hz, 1 H), 5.15 (dd, J = 9.4, 8.2 Hz, 1 H), 5.01 (dd, J = 9.4 Hz, 1 H), 3.59 (ddd, J = 9.54 6.4, 3.0 Hz, 1 H), 3.35 (dd, J = 1 1 .3, 3.0 Hz, 1 H), 3.1 8 (dd, J = 1 1 .3, 6.4 Hz, 1 H), 2.15 (s, 3H), 2.08 (s, 3H), 2.05 (s, 3H), 2.04 (s, 3H).
Step b. Synthesis of (1->6)-Thiodisaccharide: 1 ,2,3,4-tetra-0-acetyl-6-S-(2,3,4,6-tetra-0-acetyl- beta-D-g lucopyranosyl)-6-th io-beta-D-g lucopyranose
Sodium hydride (0.576 g, 14.4 mmol) was added to a solution of 2,3,4,6-tetra-0-acetyl-l-thio-p-D- glucopyranose (5.01 g, 13.75 mmol) in dry THF (130 mL) at 0 °C. The suspension was stirred under nitrogen until hydrogen formation had ceased. The resulting solution was then concentrated under reduced pressure, and the residue was dissolved in 60 mL of DMF. To this solution, 1 ,2,3,4-tetra-O- acetyl-6-deoxy-6-iodo-beta-D-glucopyranose (1 eq in 30 mL of DMF) was added. The mixture was stirred for 3 hours at room temperature under nitrogen, then concentrated under reduced pressure. A solution of the residue in DCM (250 mL) was washed with water (50 mL x 2), dried (Na2S04), and concentrated. Crystallization of the crude product was accomplished from EtOH. After drying, 7.23 g of the title compound as a white powder was obtained (79%). 1 HNMR showed that there was no alpha isomer. Mother liquor was purified by silica column. After removal of solvents, the residue was subjected to EtOH precipitation and 0.37 g of a white crystalline solid was obtained (4.1 %). Both materials showed the correct 1 HNMR and 13CNMR, identical to the reference.
Step c. Synthesis of (1->6)-Thiodisaccharide Bromide: 2,3,4-tri-0-acetyl-6-S-(2,3,4,6-tetra-0- acetyl-beta-D-glucopyranosyl)-6-thio-alpha-D-glucopyranosyl bromide
To a solution of 1 ,2,3,4-tetra-0-acetyl-6-S-(2,3,4,6-tetra-0-acetyl-beta-D-glucopyranosyl)-6-thio- beta-D-glucopyranose (7.600 g, 10.94 mmol) in dry DCM (120 mL) at 0 °C was added dropwise commercial 33% HBr in AcOH (23.8 mL, 12 equiv.). After the mixture was stirred for 3 hours in an ice- bath, TLC (1 :1 EtOAc-hexane) showed complete conversion of the starting material into a single product. DCM was added (240 mL), and the solution was washed with cold water (2 x 100 mL). After removal of the solvents, the residue was subjected to EtOH precipitation and 6.87 g of the title compound (as a white crystalline solid) was obtained (87.8%). Both showed the correct 1 HNMR and 13CNMR, identical to the reference. Step d. Synthesis of (1->6)-Thiodisaccharide Thiol: 2,3,4-tri-0-acetyl-6-S-(2,3,4,6-tetra-0-acetyl- beta-D-glucopyranosyl)-1 ,6-dithio-beta-D-glucopyranose
A similar procedure was used as that in Example 3, Step e. Crystallization from EtOH gave 3.74 g of the title compound (white solid) from 4.27 g of 2,3,4-tri-0-acetyl-6-S-(2,3,4,6-tetra-0-acetyl-beta-D- glucopyranosyl)-6-thio-alpha-D-glucopyranosyl bromide (94% yield). 1 HNMR and 13CNMR were identical to the reference (Carbohydrate Research, 281 (1996) 99-1 18).
Step e. Synthesis of (1->6)-Thiotrisaccharide: 2,3,4,6-tetra-0-acetyl-beta-D-glucopyranosyl-(1->6)- 2,3,4-tri-0-acetyl-6-thio-beta-D-glucopyranosyl-(1->6)-1 ,2,3,4-tetra-0-acetyl-6-thio-beta-D- glucopyranose
Reaction and workup procedure are analogous to those described above in Step b. Purification: normal phase silica gel column 0-90% purify the crude residue. The collection was clean by TLC but contained NMP. After removing EA/Hex, added water and lyophilized twice to remove NMP. 1 HNMR is identical to the reference (Carbohydrate Research, 281 (1996) 99-1 18). 4.62 g of the title compound was obtained (73% based on bromide).
Step f. Synthesis of (1->6)-Thiotrisaccharide Bromide: 2,3,4,6-tetra-O-acetyl-beta-D- glucopyranosyl-(1->6)-2,3,4-tri-0-acetyl-6-thio-beta-D-glucopyranosyl-(1->6)-2,3,4-tri-0-acetyl-6- thio-alpha-D-glucopyranosyl bromide
An analogous procedure to that used in Step c of this Example was used. 4.26 g of the title compound was obtained as a white solid (82%). 13C NMR (75 MHz, CDC ): δ 170.69 (e), 170.12 (e, 2C), 169.83 (e), 169.76 (e), 169.70 (e), 169.64 (e), 169.49 (e), 169.44 (e), 169.37 (e), 86.17 (o), 83.48 (o), 83.1 8 (o), 78.26 (o), 76.00 (o), 74.33 (o), 73.72 (o), 73.56 (o), 71 .52 (o), 70.67 (o), 70.43 (o), 70.09 (o), 70.00 (o), 69.87 (o), 68.20 (o), 62.07 (e), 31 .07 (e), 30.17 (e), 20.84 (o, CH3), 20.76 (o, 3 CH3), 20.70 (o, CH3), 20.66 (o, CH3), 20.65 (o, CH3), 20.61 (o, 3 CH3).
Step g. Synthesis of (1->6)-Thiotrisaccharide Thiol: 2,3,4,6-tetra-O-acetyl-beta-D-glucopyranosyl-
(1->6)-2,3,4-tri-0-acetyl-6-thio-beta-D-glucopyranosyl-(1->6)-2,3,4-tri-0-acetyl-1 ,6-dithio-beta-D- glucopyranose
An analogous procedure as that used in Example 3, Step e was used. 3.20 g of the title compound was obtained as a white solid (75%). 1 H NMR (300 MHz, Chloroform-d): δ 5.10 -5.29 (m, 3H), 4.91 - 5.07 (m, 5H), 4.74 (d, J = 10.1 , 1 H), 4.68 (d, J = 1 0.4, 1 H), 4.62 (dd, J = 9.8 Hz, 1 H), 4.30 (dd, J = 4.9, 12.4, 1 H), 4.18 (dd, J = 1 .6, 1 1 .0, 1 H), 3.74 - 3.84 (m, 2H), 3.65 - 3.73 (m, 2H), 2.79 - 2.90 (m, 2H), 2.42 (d, J = 9.8 Hz, 1 H), 2.15 (s, 3H), 2.1 1 (s, 3H), 2.1 0 (s, 3H), 2.09 (s, 6H), 2.08 (s, 3H), 2.05 (s, 3H), 2.03 (s, 6H), 2.02 (s, 3H). 13C NMR (75 MHz, CDCI3) δ 170.69 (e), 170.17 (e), 170.13 (e), 170.1 1 (e), 169.65 (e), 169.62 (e, 2C), 169.42 (e), 169.40 (e), 169.36 (e), 83.44 (o), 83.12 (o), 78.92 (o), 78.54 (o), 77.93 (o), 76.10 (o), 73.73 (o), 73.65 (o, 2C), 73.40 (o), 71 .52 (o), 71 .40 (o), 70.44 (o), 69.74 (o), 68.1 7 (o), 62.04 (e), 31 .23 (e), 31 .05 (e), 20.85 (o), 20.79 (o, 2C), 20.76 (o, 2C), 20.71 (o), 20.63 (o, 4C). Step h. Synthesis of Peracetylated (1->6)-Trisaccharide ethyleneamine: 2-aminoethyl 2,3,4,6-tetra- 0-acetyl-beta-D-glucopyranosyl-(1->6)-2,3,4-tri-0-acetyl-6-thio-beta-D-glucopyranosyl-(1->6)-2,3,4- tri-0-acetyl-1 ,6-dithio-beta-D-glucopyranoside
Part 1 (Mitsunobu Reaction). The reaction was carried out in a similar manner as Example 3, Step j, Part 1 except the purification method was different. Reversed phase C18 column (10-80%
ACN/water with 0.1 %TFA) was used. 1 .02 g of the desired (1 ->6) product was obtained (87%) from 2,3,4,6-tetra-0-acetyl-beta-D-glucopyranosyl-(1 ->6)-2,3,4-tri-0-acetyl-6-thio-beta-D-glucopyranosyl-(1 - >6)-2,3,4-tri-0-acetyl-1 ,6-dithio-beta-D-glucopyranose (1 .00 g 1 .03 mmol). 13C NMR (75 MHz, CDC ) δ 170.66 (e), 170.18 (e), 170.13(e), 1 70.06 (e), 1 69.67(e), 169.61 (e), 169.44 (e, 2C), 169.40 (e), 169.34 (e), 156.33 (e), 136.57 (e), 136.40 (e), 128.51 (o), 128.10 (o, 2C), 83.76 (o, 2C), 83.53 (o), 77.57 (o), 76.20 (o), 73.65 (o), 73.49 (o, 2C), 73.45 (o), 71 .49 (o), 71 .45 (o), 70.1 6 (o), 70.13 (o), 69.61 (o), 68.1 5 (o), 66.81 (e), 66.62 (e), 62.17 (e), 62.1 0 (e), 43.47 (e), 41 .05 (e), 20.83 (o, CH3), 20.78 (o, CH3), 20.73 (o, CH3), 20.69 (o, 2 CH3), 20.60 (o, 5 CH3).
Part 2 (Hydrogenolysis). The reaction was carried out in a similar manner as Example 3, Step j, Part 2 to obtain INT-6. Ion found by LCMS (M+H+: 101 6.1 ).
Exampl -7
Figure imgf000189_0001
In a manner similar to that described for the synthesis of INT-4 but using INT-6 as the starting material, INT-7 is obtained having an exact mass of 595.14.
Example 8. Synthesis of (1->6)-Thiotrisaccharide ethyleneamine: 4-[(2-{[beta-D-glucopyranosyl- (1->6)-6-thio-beta-D-glucopyranosyl-(1->6)-6-thio-beta-D-glucopyranosyl]thio}ethyl)amino]-4- oxobutanoic acid (INT-8)
Figure imgf000189_0002
Step a. An analogous procedure was used as that for the preparation of the compound from Example 5, Step a. The desired compound (0.480 g) was obtained in 99% yield. 13C NMR (75 MHz, CDCI3): δ 172.65 (e), 170.75 (e), 170.22 (e), 1 70.1 7 (e), 170.09 (e), 169.78 (e), 169.75 (e, 3C), 169.57 (e), 169.49 (e), 169.46 (e), 83.85 (o), 83.63 (o), 83.55 (o), 77.49 (o), 77.43 (o), 76.24 (o), 73.65 (o), 73.43 (o), 73.38 (o), 71 .58 (o), 71 .50 (o), 70.25 (o, 2C), 69.59 (o), 68.16 (o), 62.14 (e), 39.69 (e), 32.29 (e), 31 .29 (e), 30.78 (e), 30.38 (e, 2C), 20.86 (o,CH3), 20.79 (o,CH3), 20.75 (o, CH3), 20.72 (o, 3 CH3), 20.62 (o, 4 CH3). Step b. An analogous procedure was used as that for the preparation of the compound from Example 5, Step b. INT-8 (0.275 g, 99% yield) was obtained. Negative ion was found (M-H-: 694.0). 13C NMR (75 MHz, D20): δ 180.94 (e), 175.76 (e), 87.07 (0) , 86.04 (0) , 85.34 (0) , 79.74 (0) , 79.23 (0) , 79.20 (0) , 77.14 (0) , 76.87 (0, 2C), 72.70 (0) , 72.46 (0) , 72.41 (0, 2C), 72.33 (0) , 69.37 (0) , 60.77 (e), 39.43 (e), 33.07 (e), 32.94 (e), 32.30 (e), 32.01 (e), 29.51 (e).
Example 9. Synthesis of INT-9
Figure imgf000190_0001
Figure imgf000190_0002
reflux 85°C
Figure imgf000190_0003
Step a. Synthesis of benzyl [2-(2-hydroxyethoxy)ethyl]carbamate
Triethylamine (1 .31 mL, 9.32 mmol) was added to a cold (0 °C) solution of 2-(2- aminoethoxy)ethan-1 -ol (CAS# 929-06-6, 1 .00 g, 9.32 mmol) in anhydrous THF (100 mL). To the mixture was then added slowly Cbz chloride (1 .50 mL, 10.3 mmol). After stirring at room temperature for 1 hour, the reaction was diluted with ethyl acetate (250 mL) and the organic layer was washed with a mixture of saturated sodium bicarbonate and brine (50 mL + 50 mL), dried (Na2S04) and concentrated under reduced pressure to provide the crude product. The crude residue was purified by normal phase silica flash column (0-10% EA/Hex) to give the desired product (14.1 5 g, 89%) as a colorless oil. 1 HNMR (300MHz, Chloroform-d): δ 7.45 - 7.29 (m, 5H), 5.1 8 (bs, 1 H, NH), 5.13 (m, 2H), 3.75 (td, J = 5.6, 3.9 Hz, 2H), 3.59 (m, 4H), 3.43 (td, J = 5.6, 3.9 Hz, 2H), 2.06 (t, J = 6.0 Hz, 1 H, OH). Step b. Synthesis of Per-acetyl Rhamnose Bromide: 2,3,4-tri-0-acetyl-6-deoxy-beta-L- mannopyranosyl bromide
To a solution of L-rhamnose (69.0 g, 0.3799 mol, 1 .0 eq) and DMAP (1 .40 g, 0.02x) in pyridine (280mL, 4V/M) was added acetic anhydride (232.0 g, 2.273 mol, 6.0 equiv) dropwise with cooling by an ice-water bath (control the temperature below 20°C). The mixture was stirred at ambient temperature (12 hrs), and volatiles were removed under reduced pressure (toluene azeotrope). The crude product was dissolved in EtOAc (800 mL), washed with hydrochloric acid (1 N, 800 mL*2), sat. NaHCO3 (800 mL), brine (800 mL), and dried with sodium sulfate (100g). Volatiles were removed under reduced pressure, and crude acetate (131 .0g, yield: 100%) was used directly for the next step. To a solution of the crude peracetylated rhamnose (6.34 g, 15.3 mmol) in dry DCM (150 mL) at 0 °C was added dropwise commercial 33% HBr in AcOH (33.2 mL, 12 equiv.). After the mixture was stirred for 3 hours at ice-bath, TLC (1 :1 EtOAc-hexane) showed complete conversion of the mixed SM into a single product. DCM was added (240mL), and the solution was washed with cold water (2 x 100 mL) and dried with Na2S04. DCM was removed by reduced pressure. The residue was purified using silica column (0-50% EA/Hex) to give a white solid (Rf = 0.5 at 30%EA/Hex) in 84% yield.
Step c. Synthesis of Peracetylated Rhamnosyl Thiol: 2,3,4-tri-0-acetyl-6-deoxy-1-thio-alpha-L- mannopyranose
Thiourea (2.82 g, 37.0 mmol, 5 equiv.) was added into a solution of the above 2,3,4-tri-O-acetyl-
6-deoxy-beta-L-mannopyranosyl bromide in dry acetone (40mL). The reaction mixture was refluxed with TLC monitoring. After 10 minutes, the reaction mixture became a clear solution. After 2 hrs, another 5 equiv. of thiourea was added and the mixture was refluxed for another 2 hours. After completion of conversion of starting material (checked by TLC), the reaction mixture was cooled to room temperature. Acetone was removed under reduced pressure. To the solid residue was added chloroform and water (1 :1 , 300 mL) and stirred at 85 °C for 1 hour with condenser. The mixture was cooled to room
temperature and extracted with DCM (3 x 200 mL), the DCM layer dried, filtered and the solvent removed. The residue was purified by silica gel chromatography (0-50% EA/Hex) to give a white solid (Rf = 0.5 at 30%EA/Hex) in over 80% yield. 1 H NMR (300 MHz, Chloroform-d): δ 5.49 (dd, J = 6.9, 1 .3 Hz, 1 H), 5.33 (s, 1 H), 5.38 - 5.26 (m, 1 H), 5.1 0 (t, J = 9.4 Hz, 1 H), 4.30 - 4.06 (m, 1 H), 2.26 (d, J = 7.0 Hz, 1 H), 2.1 7 (s, 3H), 2.08 (s, 3H), 2.01 (s, 3H), 1 .52 (d, J = 6.2 Hz, 3H). 13C NMR (75 MHz, Chloroform-d): 170.0 (CO), 170.0 (CO), 169.3 (CO), 84.1 (CH), 76.7 (CH), 72.3 (CH), 71 .0 (CH), 69.5 (CH), 67.6 (CH), 20.9 (CH3), 20.8 (CH3), 20.7 (CH3), 17.3 (CH3). Step d. Synthesis of Rhamnose-PEG1-NHCbz: benzyl (2-{2-[(2,3,4-tri-0-acetyl-6-deoxy-alpha-L- mannopyranosyl)thio]ethoxy}ethyl)carbamate
Trimethylphosphine (TMP, 1 .0M in THF, 1 .3 mL, 1 .3 mmol) was added to a solution of 1 ,1 '- (azodicarbonyl)dipiperidine (ADDP, 0.329 g, 1 .3 mmol) in 2 mL of THF at 0 °C and stirred for 30 min. The Cbz-2-(2-hydroxyethoxy)ethyl alcohol (0.164 g, 0.686 mmol) from Step a and the 2,3,4-tri-0-acetyl-6- deoxy-1 -thio-alpha-L-mannopyranose from Step b (in 2 mL of THF) were added sequentially to the solution, with further stirring at room temperature for 2 hours. Any precipitate was then filtered off and the solution evaporated to dryness. Normal phase silica gel column purification (0-70% EtOAc/Hex) gave the desired product as a white foam (0.946 g, 80%). 1 H NMR (300 MHz, Chloroform-d): δ 7.42 - 7.28 (m, 5H), 5.40 - 5.25 (m, 2H, NH, CH), 5.28 (d, J = 1 .5 Hz, 1 H), 5.22 (dd, J = 10.0, 3.3 Hz, 1 H), 5.17 - 5.05 (m, 3H), 4.20 (m, 1 H), 3.64 (m, 2H), 3.53 (m, 2H), 3.38 (m, 2H), 2.84 (m, 1 H), 2.72 (m, 1 H), 2.13 (s, 3H), 2.05 (s, 3H), 1 .99 (s, 3H), 1 .23 (d, J = 6.2 Hz, 3H). 13C NMR (75 MHz, CDCI3) δ 170.19 (e), 169.99 (e), 169.88 (e), 156.47 (e), 136.57 (e), 128.50 (o, 2C), 128.07 (o, 3C), 82.64 (o), 77.52 (CDCI3), 77.09 (CDCI3), 76.67 (CDCI3), 71 .52 (o), 71 .1 5 (o), 70.55 (e), 69.94 (e), 69.24 (o), 67.1 9 (o), 66.64 (e), 40.87 (e), 30.72 (e), 20.95 (o), 20.82 (o), 20.70 (o), 1 7.37 (o). Step e. Synthesis of Rhamnose-PEG1-NH Succinic Anhydride Adduct: 4-oxo-4-[(2-{2-[(2,3,4-tri-0- acetyl-6-deoxy-alpha-L-mannopyranosyl)thio]ethoxy}ethyl)amino]butanoic acid
Part 1 (Hydrogenolysis): Benzyl (2-{2-[(2,3,4-tri-0-acetyl-6-deoxy-alpha-L- mannopyranosyl)thio]ethoxy}ethyl)carbamate (0.200 g, 0.379 mmol) and 30% Pd/C (0.134 g, 0.379 mmol) were loaded in the flask with 8 mL of EtOAc and 2 ml_ of CHCI3. Under H2, the reaction mixture was stirred overnight. Check TLC and mass spectrum. The material was filtered through celite and washed with EtOH to give the title compound 0.14 g. Mass spectrum showed a strong detectable positive charge signal (5-95%) (found M+H+: 394.2 and M-Boc+H+: 293.2). Part 2 (Succinic anhydride adduct): To the material from Part 1 in DCM/Py (10 mL) was added succinic anhydride (0.540 g, 0.534 mmol, 1 .5 equiv.). The reaction mixture was stirred at room temperature overnight. LC/MS 8min Positive/negative showed the desired peak mass at 3.72 min on both positive (M+H+: 494.2) and negative ionization (492.2). The solvent was removed and the residue was loaded onto a silica gel column and eluted with 4-6% MeOH/DCM to give the title compound. 13C NMR (75 MHz, CDCI3) δ 175.94 (CO), 172.53 (CO), 170.42 (CO), 170.02 (2CO), 82.53 (CH), 71 .63 (CH), 71 .08 (CH), 70.51 (CH2), 69.60 (CH2), 69.25 (CH), 67.20 (CH), 39.38 (CH2), 30.82 (CH2), 30.71 (CH2), 28.9 (CH2), 21 .00 (CH3), 20.80 (CH3), 20.70 (CH3), 17.36 (CH3).
Step f. Synthesis of Rhamnosyl-PEG1-Succinic acid linker: 4-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)thio]ethoxy}ethyl)amino]-4-oxobutanoic acid (INT-9)
NaOMe in MeOH (0.33 mL, 1 .42 mmol) was added into the 20-mL vial which contained the product of Step e (0.140 g, 0.284 mmol) in 3 mL of MeOH. Stirred at room temperature for overnight. LC/MS 5 min pos/neg method showed desired mass at 2.01 7 min (M+H+: 368.2 and M-H-: 366.2).
Acidified to pH~3 with AcOH and the solvent was evaporated under reduced pressure. The residue as purified by C1 8 reverse phase chromatography to give the title compound INT-9.
Example 10. Synthesis of INT-10 (Gala1-3Galp1-4GlcNAcp-OCH2CH2NH2)
Figure imgf000192_0001
Step b. Synthesis of galactose coupling partner
Figure imgf000193_0001
Step c. Synthesis of protected 2-glucosamine-1-azidoethylether
Figure imgf000193_0002
Step d. Synthesis of INT-10 (Gala1-3Galp1-4GlcNAcp-OCH2CH2NH2)
Figure imgf000193_0003
INT-10 (Galal -3Galp1 -4GICNACP-OCH2CH2NH2) was prepared in a manner similar to that described in WO 2014/151423 A1 (Example 1 , Scheme 5) with the modifications noted in the preceding scheme detailing the preparation. 1 H NMR (400 MHz, D2O) : δ 5.03 (d, 3.6 Hz, 1 H), 4.41 (d, 7.6 Hz, 2H), 4.10-4.04 (m, 2H), 4.06-3.49 (m, 15H). 2.73-2.71 (m, 2H), 1 .93 (s, 3H). LCMS: m/z calcd for
C22H40N2O16: 588.24; found: 589.2 [M+H]+ Example 11. Degradation of polymyxin B to tri-Boc polymyxin B cycloheptapeptide (INT-11 )
Figure imgf000194_0001
polymyxin B
INT-11
Polymyxin B (100 g, 72.2 mmol) was dissolved in acetonitrile (1 000 mL, 10V) and water (500 mL, 5V) and stirred at room temperature for 10 mins. TEA (58.5 g, 8.0 eq) was added and the mixture stirred for a further 10 mins. B0C2O (94.6 g, 6.0 eq) was subsequently added in one portion and the mixture stirred for 6 hrs at 20 °C. Savinase (300 mL) was then added. The pH of the resulting mixture was adjusted to 9.0 with aq 4M sodium hydroxide solution (10 mL) and the reaction mixture stirred at 25°C. Additional savinase (100 mL, 1 V) was added after 17 hrs, and another quantity of savinase (100 mL x2, 1 VX2) was added after 26 hrs. After an overall reaction time of 80 hrs, the mixture was diluted with EA (2000 mL, 20V). After separation of the layers, the organic phase was washed with 0.1 M NaOH solution (1000 mL x2, 10V x2), then water (1000 mL, 10V). The organic layer was dried over anhydrous Na2S04, filtered and the solvent evaporated at reduced pressure. The residue was purified by silica gel chromatography eluting with 80% (EtOAc : MeOH : Η2ΟΝΗ3 = 40:1 0:1 ) in ethyl acetate to give the title compound INT-1 1 (49.8 g, 65.0%). LCMS: m/z (M + H)+ calcd for CsoHeaNnO :! 061 .61 ; found:1062.5
Example 12. Degradation of polymyxin E to tri-Boc polymyxin E cycloheptapeptide (INT-12)
Figure imgf000194_0002
INT-12
Procedure A
Colistin sulfate (5.0 g, 3.95 mmol) was dissolved in acetonitrile (50 mL) and water (25 mL) and stirred at room temperature for 10 minutes. Triethylamine (3.2 mL, 23.0 mmol) was added and the mixture stirred for a further 10 minutes. Di-tert-butyl dicarbonate (5.0 g, 23.0 mmol) was subsequently added in one portion and the mixture stirred for 16 hours. Savinase (Novozymes) (15 mL) was then added, followed by 4 M sodium hydroxide solution (0.5 mL) and the reaction mixture stirred at room temperature for 5 days. The mixture was diluted with ethyl acetate and water. After separation of the layers, the organic phase was washed with 0.1 M sodium hydroxide solution (x2), then water. The organic layer was dried over magnesium sulfate, filtered and the solvent evaporated at reduced pressure. The residue was purified by reversed phase chromatography (1 0-95% acetonitrile/di water containing 0.1 % formic acid: 25 minute gradient). The pure fractions were pooled and lyophilized to afford title compound INT-12 as a formate salt, white powder. (2.28 g, 56%). m/z 1028, [M+H]+
Procedu e B
Colistin sulfate (50.0 g, 39.5 mmol) was dissolved in acetonitrile (500 mL, 10V) and water (250 mL, 5V) and stirred at room temperature for 10 mins. TEA (24.2 g, 6.0eq) was added and the mixture stirred for a further 10 mins. B0C2O (52.2 g, 6.0 eq) was subsequently added in one portion and the mixture stirred for 29 hrs. LCMS showed material no being detected. Savinase (1 50 mL) was then added. The pH of the resulting mixture was adjusted to 9.0 with aq 4M sodium hydroxide solution (5 mL) and the reaction mixture stirred at 25°C. Additional savinase (50 mL, 1 V) was added after 79 hrs, and another quantity of savinase (50 mL, 1 V) was added after 103 hrs. After an overall reaction time of 162 hrs, the mixture was diluted with EA (1000 mL, 20V). After separation of the layers, the organic phase was washed with 0.1 M NaOH solution (500 mL x2, 10V x2), then water (500 mL, 10V). The organic layer was dried over anhydrous Na2S04, filtered and the solvent evaporated at reduced pressure. The residue was purified by silica gel chromatography eluting with 80% (EtOAc:MeOH:NH4OH=40:10:1 ) in ethyl acetate to give the title compound INT-12 (20.2 g, 49.3%). LCMS: m/z (M + H)+ calcd for 047Η85ΝιιΟΐ4:1027.63; found:1028.5.
Example 13. Preparation of tetra-Boc Dab-polymyxin E cycloheptapeptide (INT-13)
Step a. Synthesis of Cbz-L-Dab-IN -12
Figure imgf000195_0001
INT-12 (0.20 g, 0.195 mmol) , Z-L-Dab(Boc)-OH-DCHA (0.124 g, 0.233 mmol), and HOBT (0.029 g, 0.214 mmol) were dissolved in DMF (2 mL) and EDC (0.041 g, 0.214 mmol) was added and the reaction was stirred for 2 hours. The mixture was applied directly to reversed phase HPLC (25-95% acetonitrile in Dl water containing 0.1 % formic acid:20 minute gradient). The pure fractions were lyophilized to afford 0.197 g of the title compound as a white solid. Yield: 74%. LC/MS (M+2H)/2 = 682.0. Step b. Synthesis of INT-13
Figure imgf000196_0001
The Cbz-L-Dab-INT-12 (0.197 g, 0.194 mmol) was stirred in methanol (15 mL) in the presence of 5% Pd/C (50 mg) under 1 atm of hydrogen gas for 45 minutes. The mixture was filtered, concentrated and applied to reversed phase HPLC (25-95% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were lyophilized to afford 0.140 g of the title compound as a white solid. Yield: 72%. LC/MS (M+2H)/2= 565.0 (loss of 1 Boc in LC/MS).
Example 14. Preparation of tetra-Boc Dab-polymyxin B cycloheptapeptide (INT-14)
Step a. Synthesis of Cbz-L-Dab-I -11
Figure imgf000196_0002
INT-1 1 (2 g, 1 .88 mmol), Z-L-Dab(Boc)-OH-DCHA (1 .20 g, 2.33 mmol), and N-methylmorpholine (0.62 mL, 5.65 mmol) were dissolved in DMF (6 mL) and HATU (0.858 g, 2.25 mmol) was added and the reaction was stirred for 45 minutes. The mixture was applied directly to reversed phase HPLC (25-95% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were lyophilized to afford 2.1 g of the title compound as a white solid. Yield: 79%. LC/MS (M+2H)/2 = 599.0 (Cbz protected intermediate, loss of 2 Boc groups on LC/MS).
Step b. Preparation of INT-14
Figure imgf000196_0003
The Cbz-L-Dab-INT-12 (2.1 g, 1 .50 mmol) was stirred in methanol (25 mL) in the presence of 5% Pd/C (125 mg) under 1 atm of hydrogen gas for 45 minutes. The mixture was filtered, concentrated and applied to reversed phase HPLC (25-95% acetonitrile in Dl water: 20 minute gradient). The pure fractions were lyophilized to afford 1 .92 g of INT-14 as a white solid. Yield: 96%. LC/MS (M+2H)/2=532.0 (loss of 2 Boc groups on LC/MS). Example 15. Synthesis of INT-15 (tetra-Boc polymyxin E nonapeptide)
Figure imgf000197_0001
Procedu e A
INT-15 was prepared from INT-13 and Z-Thr-OH in a manner similar to that described for INT-16. Yield 73%. LC/MS (M+2H)/2 = 565.8 (loss of 2 Boc groups on LC/MS).
Procedure B
Step a. Enzymatic hydrolysis of colistin to polymyxin E nonapeptide (PMEN)
Colistin sulfate was dissolved in phosphate buffer (1 liter, pH 7, 25 mM), and KCI (1 .5g), EDTA (500 mg), Cysteine-HCI (2g) and Immobilised papain (10 mL, Thermofisher, 16-40 BAEE/mg before immobilisation) was added then the pH was brought back to 7 with potassium phosphate, dibasic. The reaction was shaken at 37 SC for 72 hours. The solids were removed by filtration and the aqueous reaction mixture was lyophilized. The white solids were dissolved in 1 N HCI and purified (in 3 aliquots) by reversed phase HPLC (5-50% acetonitrile in Dl water containing 0.1 % TFA: 20 minute gradient). Pure fractions were pooled and lyophilized to afford the product as a white solid. LC/MS 929.5 and 465.0 ((M+2H)/2).
Step b. Selective protection of polymyxin E nonapeptide to tetra-Boc polymyxin E nonapeptide (INT-15)
The title compound may be prepared in a manner analogous to the following literature procedure: O'Dowd, et al. Tetrahedron Letters, Volume 48, Issue 1 1 , 12 March 2007, Pages 2003-2005.
Example 16. Synthesis of INT-16 (tetra-Boc polymyxin B nonapeptide)
Figure imgf000197_0002
Procedu e A
INT-14 (0.22 g, 0.174 mmol), Z-Thr-OH (0.053 g, 0.209 mmol), and N-methylmorpholine (0.057 mL, 5.65 mmol) were dissolved in DMF (6 mL) and HATU (0.070 g, 0.209 mmol) was added and the reaction was stirred for 45 minutes. The mixture was applied directly to reversed phase HPLC (25-95% acetonitrile in Dl water containing 0.1 % formic acid:20 minute gradient). The pure fractions were lyophilized to afford 2.1 g of the title compound as a white solid. LC/MS (M+2H)/2 = 649.4 (Cbz protected intermediate, loss of 2 boc groups on LC/MS). The Cbz protected intermediate was taken up in methanol (10 mL) and stirred in the presence of 5% Pd/C (40 mg) under 1 atmosphere of hydrogen gas for 45 minutes. The mixture was filtered and applied to reversed phase HPLC (20-95% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were lyophilized to afford 0.187 g of the title compound as a white solid. Yield: 78%, 2 steps. LC/MS (M+2H)/2 = 582.4 (loss of 2 Boc groups on LC/MS).
Procedure B
Tetra-Boc polymyxin B nonapeptide is prepared from polymyxin B via polymyxin B nonapeptide (PMBN) in a manner similar to that described in Example 15, Procedure B.
Example 17. Synthesis of INT-17 (penta-Boc polymyxin E decapeptide)
Figure imgf000198_0001
Penta-Boc polymyxin E decapeptide was prepared analogously to penta-Boc polymyxin B decapeptide (see Example 18), except that polymyxin E was substituted for polymyxin B as the starting material.
Exam -18 (penta-Boc polymyxin B decapeptide)
Figure imgf000199_0001
Step a. Synthesis of Cbz-penta-Boc polymyxin B decapeptide
A solution of INT-1 1 (2.25 g, 2.18 mmol), (2S)-2-[(N-{(2S)-2-{[(benzyloxy)carbonyl]amino}-4-[(tert- butoxycarbonyl)amino]butanoyl}-L-threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoic acid (1 .36 g, 2.08 mmol), and DIEA (1 .20 mL, 6.87 mmol), in DMF (5 mL ), were treated with HATU (0.831 g, 2.18 mmol), while stirring at room temperature. After 30 minutes, the product was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 20% to 1 00% methanol and water, using no modifier. Yield 2.50 g, 72% yield, lon(s) found by LCMS: [(M-1 Boc)+2H]/2 = 799.5, [(M-2Boc)+2H]/2 = 749.5
Step b. Synthesis of penta-Boc polymyxin B decapeptide
A solution of Cbz-penta-Boc polymyxin B decapeptide (2.5 g, 1 .47 mmol) dissolved in methanol (10 mL), was treated with 5% Pd/C (0.5 g), flushed with hydrogen and stirred for 12 hr. The reaction was filtered through Celite, concentrated, and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 1 .98 g, 86% yield, lon(s) found by LCMS: [(M-1 Boc)+2H]/2 = 732.4
Example 19. Synthesis of INT-19 (penta-Boc polymyxin E undecapeptide)
Penta-Boc polymyxin E undecapeptide was prepared analogously to penta-Boc polymyxin B undecapeptide (see Example 20), except that penta-Boc polymyxin E decapeptide was substituted for penta-Boc polymyxin B decapeptide as the starting material. E
Figure imgf000200_0001
Step a. Synthesis of Cbz penta-Boc polymyxin B undecapeptide
A solution of penta-Boc polymyxin B decapeptide (prepared as described in Example 18) (1 .31 g, 0.838 mmol), (2S)-2-{[(benzyloxy)carbonyl]amino}octanoic acid (0.25 g, 0.880 mmol), and DIEA (0.46 mL, 2.64 mmol), in DMF (5 mL ), were treated with HATU (0.334 g, 0.880 mmol), while stirring at room temperature. After 30 minutes, the product was used in the next step without purification, lon(s) found by LCMS: [(M-1 Boc)+2H]/2 = 870.0, [(M-2Boc)+2H]/2 = 820.0. Step b. Synthesis of penta-Boc polymyxin B undecapeptide
The solution from Step a was treated with 5% Pd/C (0.5g), flushed with hydrogen and stirred for 2 hr. The reaction was filtered through Celite, concentrated, and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.83 g, 58% yield, lon(s) found by LCMS: [(M-1 Boc)+2H]/2 = 803.0, [(M-2Boc)+2H]/2 = 753.0
Example 21. Synthesis of INT-21 (H-Thr-YBoc-Dab-OMe)
Proce
Figure imgf000200_0002
Step a. Preparation of Cbz-Thr-yBoc-Dab-OMe
NH2-Dab(Boc)-OMe (HCI salt) (5.000g, 1 eq.), Z-NH-L-Thr-OH (5.049g, 1 .05eq.), EDCI (5.350g, 1 .5eq.), HOBt (3.733g, 1 .5eq.) and NaHC03 (3.095g, 2eq.) were weighed into a 100- mL round bottom flask. 24 mL of DCM/DMF (4:1 ) was added into the flask. The mixture was stirred at room temperature for about 3 hrs. (TLC or LC/MS monitoring). After completion, EtOAc (200 mL) was added to dilute the reaction mixture and washed with 1 N aq. HCI, saturated NaHC03 and brine. Dried with Na2S04 and condensed. The residue was purified with normal phase silica (2-7% MeOH/DCM) to give 8.24 g pure desired product (>95%).
Figure imgf000201_0001
Step b. Removal of the Cbz Group
Cbz-Thr-yBoc-Dab-OMe (8.24 g) was dissolved in 1 00 mL of MeOH/EtOAc. And 5%Pd/C (3.75 g, 0.1 eq) was added. Under H2 balloon, the reaction mixture was stirred for 2 h. Check TLC and LC/MS to determine completion of the reaction. The reaction was subjected to Celite filtration and with MeOH washing of the solids. Dried to give 5.87 g of free amine (>99%). The material may be used in the next step without purification.
Procedu e B
Figure imgf000201_0002
Z-Thr-OH (2.0 g, 7.9 mmol), methyl (2S)-2-amino-4-[(tert-butoxymethyl)amino]butanoate hydrochloride (2.2 g, 8.7 mmol), HOBt (1 .2 g, 8.7 mmol), EDC (1 .7 g, 8.7 mmol) and DIEA (2.0 g, 1 6 mmol) were stirred in DMF (15 mL) at ambient temperature for 2 hours. The mixture was diluted with ethyl acetate (30 mL) and aqueous 1 N HCI (50 mL). The aqueous phase was extracted with ethyl acetate (3x, 30 mL) and the combined organic extracts were dried over sodium sulfate, filtered and concentrated. The crude dipeptide was stirred in a methanol (50 mL) under 1 atmosphere of hydrogen gas in the presence of 5% Pd/C (200 mg) for 45 minutes. The mixture was filtered, concentrated and purified by reversed phase HPLC (5 to 75% acetonitrile in Dl water containing 0.1 % TFA: 20 minute gradient). The pure fractions were pooled and lyophilized to afford H-Thr-yBoc-Dab-OMe as a white solid. Yield: 82%. LC/MS [M+H+]+ 334.8.
Example 22. Synthesis of INT-22
Step a. Synthesis of methyl (2S)-2-[(N-{(2S)-2-{[(2S)-2-
{[(benzyloxy)carbonyl]amino}octanoyl]amino}-4-[(tert-butoxycarbonyl)amino]butanoyl}-L threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate
Figure imgf000201_0003
A solution of methyl (2S)-2-[(N-{(2S)-2-amino-4-[(tert-butoxycarbonyl)amino]butanoyl}-L- threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate (0.600 g, 1 .124 mmol),(2S)-2- {[(benzyloxy)carbonyl]amino}octanoic acid (0.330 g, 1 .124 mmol), and DIEA (0.59 mL, 3.37 mmol) in DMF (4 mL), was treated with HATU (0.470g, 1 .24 mmol), and stirred for 30 min. The reaction was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid
chromatograph eluted with 10% to 100% acetonitrile and water, using formic acid (0.1 %) as the modifier. Yield 0.726 g, (79%). lon(s) found by LCMS: (M+H)+ = 809.5
Step b. Synthesis of methyl (2S)-2-[(N-{(2S)-2-{[(2S)-2-aminooctanoyl]amino}-4-[(tert- butoxycarbonyl)amino]butanoyl}-L-threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate
Figure imgf000202_0001
A mixture of methyl (2S)-2-[(N-{(2S)-2-{[(2S)-2-{[(benzyloxy)carbonyl]amino}octanoyl]amino}-4- [(tert-butoxycarbonyl)amino]butanoyl}-L-threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate (0.300 g, 0.371 mmol), 5% Pd/C (0.150 g), and methanol (4 mL), were charged with hydrogen from a balloon. After 2 hr, the product was filtered through Celite, concentrated, and used in the next step without further purification. Mass recovery 0.250 g, quantitative, lon(s) found by LCMS: (M+H)+ = 675.4
Figure imgf000202_0002
Step a. Synthesis of benzyl diethyl 2,2',2"-nitrilotriacetate
Benzyl bromoacetate (4.6 g, 20 mmol) was added into the solution of diethyl 2,2'- azanediyldiacetate (3.8 g, 20 mmol) and DIPEA (3.6 mL, 30 mmol) in 60 mL DMF, the resultant solution was stirred at room temperature for overnight. The reaction solution was concentrated and purified by flash chromatography to provide products. 1 H NMR (300 MHz, Chloroform-d) δ 7.37 (m, 5H), 5.17 (s, 2H), 4.18 (q, J = 6.9 Hz, 4H), 3.78 (s, 2H), 3.68 (s, 4H), 1 .27 (t, J = 8.2Hz, 6H). LC/MS, 338.2 [M+H]+.
Step b. Synthesis of [bis(2-ethoxy-2-oxoethyl)amino]acetic acid
Benzyl diethyl 2,2',2"-nitrilotriacetate (4.6 g, 13.6 mmol) was dissolved into 30 mL MeOH and 30 mL ethyl acetate, then 1 g of 5% palladium on charcoal was added above solution and the mixture was stirred at room temperature under an hydrogen atmosphere overnight. The solids were removed by filtration after completion of the reaction as judged by LCMS. The filtrate was concentrated and used in the next step without any purification. 1 H NMR (300 MHz, Chloroform-d) δ 4.24 (q, J = 7.1 Hz, 4H), 3.61 (s, 4H), 3.53 (d, J = 10.1 Hz, 2H), 1 .31 (t, J = 7.2 Hz, 6H). LC/MS, 247.2 [M+H]+ .
Step c. Synthesis of diethyl 2,2'-({2-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}azanediyl)diacetate
To the solution of [bis(2-ethoxy-2-oxoethyl)amino]acetic acid (4.4 g, 18 mmol), 2-(2- aminoethoxy)ethyl 6-deoxy-alpha-L-mannopyranoside (4.51 g, 18 mmol) in DMF (50 mL) was added EDC (4 g, 20 mmol), HOBT (2.7g, 20 mmol), Hunig's base (4.2 mL, 30 mmol) at room temperature. The solution was stirred for overnight. The resultant solution was removed DMF and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil, 7.0 g, 81 % yield. 1 H NMR (300 MHz, methanol-ck) δ 4.19 (qd, J = 7.2, 3.0 Hz, 2H), 3.88 - 3.55 (m, 4H), 3.54 - 3.33 (m, 3H), 2.94 (s, 1 H), 2.83 (s, 1 H), 2.36 (t, J = 8.1 Hz, OH), 2.03 (p, J = 7.7 Hz, OH), 1 .43 - 1 .21 (m, 4H). LC/MS 481 .2 [M+H]+
Step d. Synthesis of 2,2'-({2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2- oxoethyl}azanediyl)diacetic acid
Lithium hydroxide (20 mmol) in 10 mL H2O was added to the solution of diethyl 2,2'-({2-
[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}azanediyl)diacetate (4.80g, 10 mmol) in mix solvent of 10 mL H2O, 20 mL MeOH and 20 mL THF. The resultant solution was stirred for 1 hours at room temperature. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. 5.7g, yield of 92%. 1 H NMR (300 MHz, methanol-ck) δ 4.73 (s, 1 H), 3.80 (dd, J = 15.5, 5.5 Hz, 2H), 3.73 - 3.61 (m, 7H), 3.60 (d, J = 10.6 Hz, 1 H), 3.59 (s, 6H), 3.40 (d, J = 23.5 Hz, 7H), 1 .27 (d, J = 6.3 Hz, 3H). LC/MS 425.2 [M+H]+ Step e. Synthesis of dibenzyl 14-{2-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}-4,12,16,24-tetraoxo-8,20-dioxa- 5,11 ,14,17,23-pentaazaheptacosane-1 ,27-dioate
To the solution 2,2'-({2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2- oxoethyl}azanediyl)diacetic acid (Example 23, Step d) (1 .3 g, 3 mmol) and 4-(benzyloxy)-4-oxobutanoic acid (1 .8 g, 6.1 mmol) in DMF (30 ml_) was added EDC (1 .4g, 7 mmol), HOBT (1 .0g, 7 mmol), Hunig's base (1 .4 ml_, 10 mmol) at room temperature. The solution was stirred for overnight. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 1 00% acetonitrile and water, using 0.1 %
trifluoroacetic acid as the modifier. The product was obtained as an oil, 2.2 g, 75% yield. 1 H NMR (300 MHz, methanol-dt) δ 7.36 (s, 10H), 5.13 (s, 4H), 4.72 (s, 1 H), 3.81 (s, 2H), 3.70 - 3.46 (m, 16H), 3.46 - 3.24 (m, 85H), 2.70 (q, J = 6.9, 5.4 Hz, 5H), 2.55 (d, J = 7.0 Hz, 3H), 1 .27 (d, J = 6.2 Hz, 3H). LC/MS 977.5 [M+H]+ Step f. Synthesis of 14-{2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2- oxoethyl}-4,12,16,24-tetraoxo-8,20-dioxa-5,11 ,14,17,23-pentaazaheptacosane-1 ,27-dioic acid
Dibenzyl 14-{2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}- 4,12,16,24-tetraoxo-8,20-dioxa-5,1 1 ,14,17,23-pentaazaheptacosane-1 ,27-dioate (816mg, 0.836 mmol) was dissolved into 5 mL MeOH and 5 mL ethyl acetate, then 0.5 g of 5% Palladium on charcoal was added above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere for overnight. The palladium charcoal was removed by filtration after completed the reaction by LCMS. The filtrate was concentrated and used next step without any purification. 1 H NMR (300 MHz, methanol- dt) 5 4.73(s, 1 H) 3.74 (s, 7H), 3.70 - 3.49 (m, 15H), 3.43 (dd, J = 14.3, 7.8 Hz, 8H), 3.36 (s, 48H), 2.69 - 2.43 (m, 8H), 1 .27 (d, J = 6.2 Hz, 3H). LC/MS, 797.4 [M+H]+ .
Step g. Synthesis of per-Boc-(Compound 1)
To the solution of 14-{2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2- oxoethyl}-4,12,16,24-tetraoxo-8,20-dioxa-5,1 1 ,14,17,23-pentaazaheptacosane-1 ,27-dioic acid (80 mg, 0.10 mmol), triethylamine (0.07 mL, 0.5 mmol) and INT-16 (270 mg, 0.2 mmol) in 5 mL DMF was added HATU (78 mg, 0.2 mmol). The reaction was stirred for 1 hr and the resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. Yield of product, tri-tert-butyl {[(2S,5R,8S,1 1 S,14S,17S,22S)-5-benzyl-22-{[(8S,1 1 S,41 S,44S)-8- {[(3S,6S,9S,12S,15R,18S,21 S)-15-benzyl-6,9,18-tris{2-[(tert-butoxycarbonyl)amino]ethyl}-3-[(1 R)-1 - hydroxyethyl]-12-(2-methylpropyl)-2,5,8,1 1 ,14,17,20-heptaoxo-1 ,4,7,10,13,16,19-heptaazacyclotricosan- 21 -yl]carbamoyl}-44-{2-[(tert-butoxycarbonyl)amino]ethyl}-1 1 ,41 -bis[(1 R)-1 -hydroxyethyl]-2,2-dimethyl- 4,10,13,16,24,28,36,39,42,45-decaoxo-26-(2-oxo-2-{[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyloxan-2-yl]oxy}ethoxy)ethyl]amino}ethyl)-3,20,32-trioxa-5,9,12,17,23,26,29,35,40,43- decaazapentatetracontan-45-yl]amino}-17-[(1 R)-1 -hydroxyethyl]-8-(2-methylpropyl)-3,6,9,12,1 5,18,23- heptaoxo-1 ,4,7,10,13,16,19-heptaazacyclotricosane-2,1 1 ,14-triyl]tri(ethane-2,1 -diyl)}triscarbamate. 220 mg, 65% yield.
Step h. Synthesis of Compound 1
The per-Boc-(Compound 1 ) from Step g was treated in 2 ml_ DCM and 2 ml_ TFA at room temperature, the solution was stirred 10 min, then concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield of (2S,5S,35S,38S)-2,38-bis(2-aminoethyl)- 5,35-bis[(1 R)-1 -hydroxyethyl]-4,7,10,18,22,30,33,36-octaoxo-20-(2-oxo-2-{[2-(2-{[(2R,3R,4R,5R,6S)- 3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}ethoxy)ethyl]amino}ethyl)-N~1 ~,N~39~- bis(3S,6S,9S,12S,15R,18S,21 S)[6,9,18-tris(2-aminoethyl)-1 5-benzyl-3-[(1 R)-1 -hydroxyethyl]-12-(2- methylpropyl)-2,5,8,1 1 ,14,17,20-heptaoxo-1 ,4,7,1 0,13,16,1 9-heptaazacyclotricosan-21 -yl]-14,26-dioxa- 3,6,1 1 ,17,20,23,29,34,37-nonaazanonatriacontane-1 ,39-diamide was 0.120 g, 67% yield, lon(s) found by LCMS: (M+2H)/2 = 1343.7, (M+3H)/3 = 896.2, (M+4H)/4 =672.4, (M+5H)/5=538.1 .
Example 2
Figure imgf000205_0001
The title compound was prepared analogously to Compound 1 except INT-15 was substituted for INT-16 in the sequence, lon(s) found by LCMS (M+2H)/2 = 1309.8, (M+3H)/3 =873.5, (M+4H)/4 =655.4. (M+5H)/5 =524.5.
Exampl
Figure imgf000205_0002
The title compound was prepared analogously to Compound 10. lon(s) found by LCMS: (M+2H)/2 = 1480.8. (M+3H)/3 = 987.5, (M+4H)/4 =740.9, (M+5H)/5 =592.9. (M+6H)/6 =494.3. Exam
Figure imgf000206_0001
The title compound was prepared analogously to Compound 2. lon(s) found by LCMS: (M+2H)/2 = 1445.8, (M+3H)/3 = 964.9. (M+4H)/4 =724.0, (M+5H)/5 =579.3.
Example 27. Preparation of Compound 5
Step a. Preparation of benzyl {21-[(6-deoxy-alpha-L-mannopyranosyl)oxy]-15-oxo-3,6,9,12,19- pentaoxa-16-azahenicosan-1-yl}carbamate
Figure imgf000206_0002
2-(2-aminoethoxy)ethyl 6-deoxy-alpha-L-mannopyranoside (500 mg, 1 .25 mmol), 3-oxo-1 -phenyl- 2,7,10,13,16-pentaoxa-4-azanonadecan-19-oic acid (325 mg, 1 .29 mmol), EDC (287 mg, 1 .50 mmol) and DIEA (193 mg, 1 .50 mmol) were stirred together in DMF (1 .5 mL) for 3 hours. The mixture was applied directly to reversed phase HPLC (20-95% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were pooled and lyophilized to afford benzyl {21 -[(6-deoxy-alpha-L- mannopyranosyl)oxy]-1 5-oxo-3,6,9,12,19-pentaoxa-16-azahenicosan-1 -yl}carbamate as a clear oil. Yield: 91 %. LC/MS [m+H]+: 633.6.
Step b. Synthesis of 1-[(6-deoxy-alpha-L-mannopyranosyl)oxy]-7,23-dioxo-3,10,13,16,19- pentaoxa-6,22-diazahexacosan-26-oic acid
Figure imgf000206_0003
Benzyl {21 -[(6-deoxy-alpha-L-mannopyranosyl)oxy]-15-oxo-3,6,9,12,19-pentaoxa-16- azahenicosan-1 -yl}carbamate (250 mg, 0.395 mmol) was stirred under 1 atmosphere of hydrogen gas i the presence of 5% Pd/C (70 mg) at room temperature for 2 hours. The mixture was filtered,
concentrated, and taken up in methanol (4 mL) and succinic anhydride (40 mg, 0.395 mmol) and trimethylamine (40 mg, 0.395 mmol) were added. The reaction was stirred at room temperature for 2 hours and then concentrated, taken up in Dl water (2 mL) and applied to reversed phase HPLC (5-95% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were pooled and lyophilized to afford 1 -[(6-deoxy-alpha-L-mannopyranosyl)oxy]-7,23-dioxo-3,1 0,13,16,19-pentaoxa- 6,22-diazahexacosan-26-oic acid as a clear, viscous oil. Yield: 85%. LC/MS [M-H-] = 597.4.
Step c. Sy
Figure imgf000207_0001
HATU (1 12 mg, 0.250 mmol) was added to stirring mixture of tri-tert-butyl
{[(2S,5R,8S,1 1 S,14S,17S,22S)-22-({(2S,5S,24S,27S)-29-amino-2-(2-aminoethyl)-27-
{[(3S,6S,9S,12S,15R,18S,21 S)-15-benzyl-6,9,18-tris{2-[(tert-butoxycarbonyl)amino]ethyl}-3-[(1 R)-1 - hydroxyethyl]-12-(2-methylpropyl)-2,5,8,1 1 ,14,17,20-heptaoxo-1 ,4,7,10,13,16,19-heptaazacyclotricosan- 21 -yl]carbamoyl}-5,24-bis[(1 R)-1 -hydroxyethyl]-4,7,22,25-tetraoxo-10,13,1 6,1 9-tetraoxa-3, 6,23,26- tetraazanonacosanan-1 -oyl}amino)-5-benzyl-17-[(1 R)-1 -hydroxyethyl]-8-(2-methylpropyl)- 3,6,9,12,15,18,23-heptaoxo-1 ,4,7,10,13,16,19-heptaazacyclotricosane-2,1 1 ,14-triyl]tri(ethane-2,1 - diyl)}triscarbamate (279 mg, 0.1 00 mmol), 1 -[(6-deoxy-alpha-L-mannopyranosyl)oxy]-7,23-dioxo- 3,10,13,16,19-pentaoxa-6,22-diazahexacosan-26-oic acid (150 mg, 0.250 mmol), and DIEA (77 mg, 0.601 mmol) in DMF (4 mL). The reaction was stirred at room temperature for 4 hours then applied directly to reversed phase HPLC (25-95% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were pooled and concentrated. The Boc-protected intermediate was stirred in a 1 /1 mixture of TFA/DCM containing 0.1 % thioanisole (5 mL) at room temperature for 30 minutes. The solvent was removed by rotary evaporation and the crude product was purified by to reversed phase HPLC (0-35% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were pooled and lyophilized to afford (23S,26S,45S,48S)-26,45-bis[(1 R)-1 -hydroxyethyl]- 16,1 9,25,28,43,46, 52, 55-octaoxo-N~1 ~,N~70~-bis[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyloxan-2-yl]oxy}ethoxy)ethyl]-N~23~,N~48~-bis(3S,6S,9S,12S,15R,18S,21 S)[6,9,18-tris(2- aminoethyl)-15-benzyl-3-[(1 R)-1 -hydroxyethyl]-12-(2-methylpropyl)-2,5,8,1 1 ,14,17,20-heptaoxo- 1 , 4,7,10,13, 16,1 9-heptaazacyclotricosan-21 -yl]-3, 6, 9, 12,31 , 34,37,40,59,62,65, 68-dodecaoxa- 15,20,24,27,44,47,51 ,56-octaazaheptacontane-1 ,23,48,70-tetracarboxamide (Compound 5) as a white solid. Yield: 10%, 2 steps. LC/MS (M+4H)/4 = 837.0.
Example 28. Synthesis of Compound 6
Figure imgf000208_0001
Step a. Synthesis of benzyl bis(2-oxo-2-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyltetrahydro-2H-pyran-2-yl)oxy)ethoxy)ethyl)amino)ethyl)carbamate
To the solution rhamnose(2R,3R,4R,5R,6S)-2-(2-(2-aminoethoxy)ethoxy)-6-methyltetrahydro-2H- pyran-3,4,5-triol (INT-1 ) (1 .0 mmol) and 2,2'-(((benzyloxy)carbonyl)azanediyl)diacetic acid (0.50 g, 2 mmol) in DMF (1 0 mL) was added EDC (1 .0 g, 5 mmol), HOBT (750 mg, 5 mmol) and Hunig's base (1 .4 mL, 10 mmol) at room temperature. The solution was stirred overnight. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil 620 mg, 48% yield. LC/MS 734.3 [M+H]+
Step b. Synthesis of ethyl 6-(2-ethoxy-2-oxoethyl)-4,8,11-trioxo-9-(2-oxo-2-((2-(2- (((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2- yl)oxy)ethoxy)ethyl)amino)ethyl)-17-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H- pyran-2-yl)oxy)-15-oxa-3,6,9,12-tetraazaheptadecan-1 -oate
Benzyl bis(2-oxo-2-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2- yl)oxy)ethoxy)ethyl)amino)ethyl)carbamate (400 mg, 0.05 mmol) was dissolved into 5 mL MeOH and 5 ml_ ethyl acetate, then 100 mg of 5% palladium on charcoal was added to the above solution and the mixture was stirred at room temperature under a hydrogen atmosphere for overnight. The palladium on charcoal was removed by filtration after completion of the reaction by LCMS. The filtrate was
concentrated and used for next step without any purification. LC/MS [M]+1 600.3.
Step c. Synthesis of ethyl 12-(2-ethoxy-2-oxoethyl)-7,10-dioxo-9-(2-oxo-2-{[2-(2-
{[(2/?,3/?,4/?,5/?,6¾-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}ethoxy)ethyl]amino}ethyl)-1-
{[(2/?,3/?,4/?,5/?,6¾-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}-3-oxa-6,9,12-triazatetradecan-14-oate
The amine product from the previous step was dissolved into 5 mL DMF, then 2-(bis(2-ethoxy-2- oxoethyl)amino)acetic acid (240 mg, 1 mmol), EDC (1200 mg, 10 mmol), HOBT (150 mg, 5 mmol),
Hunig's base (0.28 mL, 2 mmol) were added to the solution at room temperature. The solution was stirred overnight. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil 320 mg, 75% yield. LC/MS [M+H]+ 829.4
Step d. Synthesis of 12-(carboxymethyl)-7,10-dioxo-9-(2-oxo-2-{[2-(2-{[(2/?,3/?,4/?,5/?,6S)-3,4,5- trihydroxy-6-methyloxan-2-yl]oxy}ethoxy)ethyl]amino}ethyl)-1-{[(2/?,3/?,4/?,5/?,6¾-3,4,5-trihydroxy- 6-methyloxan-2-yl]oxy}-3-oxa-6,9,12-triazatetradecan-14-oic acid
Lithium hydroxide (24 mg, 1 mmol) in 3 mL H2O was added to the solution of ethyl 6-(2-ethoxy-2- oxoethyl)-4,8,1 1 -trioxo-9-(2-oxo-2-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H- pyran-2-yl)oxy)ethoxy)ethyl)amino)ethyl)-17-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H- pyran-2-yl)oxy)-15-oxa-3,6,9,12-tetraazaheptadecan-1 -oate (200 mg, 0.25 mmol) in a mixed solvent of 3 mL MeOH and 3 mL THF. The resultant solution was stirred for 1 hour at room temperature then concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil, 180 mg 90% yield. LC/MS [M+H]+ 773.2.
Step e. Preparation of dibenzyl 14-(2-(bis(2-oxo-2-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyltetrahydro-2H-pyran-2-yl)oxy)ethoxy)ethyl)amino)ethyl)amino)-2-oxoethyl)-4,12,16,24- tetraoxo-8,20-dioxa-5,11 ,14,17,23-pentaazaheptacosane-1 ,27-dioate
12-(carboxymethyl)-7,10-dioxo-9-(2-oxo-2-{[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-ethyloxan -2-yl]oxy}ethoxy)ethyl]amino}ethyl)-1 -{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}-3-oxa- 6,9,12-triazatetradecan-14-oic acid (150 mg, 0.2 mmol), benzyl 4-{[2-(2-aminoethoxy)ethyl]amino}-4- oxobutanoate (100 mg, 0.34 mmol), EDC (160 mg, 0.8 mmol), HOBt (120 mg, 0.8 mmol) and TEA(0.14 mL, 1 mmol) were dissolved into 5 mL DMF and the solution was stirred for overnight. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil 120 mg, 62% yield. LC/MS [M+H]+ 1325.6. Step f. Synthesis of 14-(2-(bis(2-oxo-2-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyltetrahydro-2H-pyran-2-yl)oxy)ethoxy)ethyl)amino)ethyl)amino)-2-oxoethyl)-4,12,16,24- tetraoxo-8,20-dioxa-5, 11 ,14,17,23-pentaazaheptacosane-1 ,27-dioic acid
Dibenzyl 14-(2-(bis(2-oxo-2-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H- pyran-2-yl)oxy)ethoxy)ethyl)amino)ethyl)amino)-2-oxoethyl)-4,12,16,24-tetraoxo-8,20-dioxa-
5,1 1 ,14,17, 23-pentaazaheptacosane-1 ,27-dioate (100 mg, 0.08 mmol) was dissolved into 3 mL MeOH and 3 mL ethyl acetate, then 30 mg of 5% palladium on charcoal was added above solution and the mixture was stirred at room temperature under a hydrogen atmosphere overnight. The solids were removed by filtration after completion of the reaction as evidenced by LCMS. The filtrate was
concentrated and used for next step without any purification. LC/MS [M+H]+ 1 145.5.
Step
Figure imgf000210_0001
The title compound was prepared analogously to the procedure described for Compound 1 (Steps g and h) except 14-(2-(bis(2-oxo-2-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro- 2H-pyran-2-yl)oxy)ethoxy)ethyl)amino)ethyl)amino)-2-oxoethyl)-4,12,1 6,24-tetraoxo-8,20-dioxa- 5,1 1 ,14,17,23-pentaazaheptacosane-1 ,27-dioic acid (Example 28, Step f) was used as the linker, lon(s) found by LCMS (M+3H)/3 =1012.2, (M+4H)/4 =759.4. (M+5H)/5 =607.7. (M+6H)/6 =506.6.
Example 2
Figure imgf000210_0002
The title compound was prepared analogously to Compound 10. lon(s) found by LCMS (M+3H)/3 = 1 037.2, (M+4H)/4 =778.2, (M+5H)/5 =622.7. (M+6H)/6 =518.1 . Example 30.
Figure imgf000211_0001
Step a. Synthesis of di-(4-formylphenyl)methyl amino-PEG10-acid
Terephthalaldehyde (536.4 mg, 4.0 mmol) and amino-PEG1 0-acid (529.6 mg, 1 mmol) were dissolved in anhydrous EtOH (8 mL) by gently heating with a heat gun. After cooling to room temperature, sodium triacetoxyborohydride (635.8 mg, 3 mmol) was added in two portions over 30 minutes to the solution. The mixture was stirred for 2 hours and directly purified by RPLC (0 to 100% acetonitrile in water). Yield 289 mg, 37.7 %. lon(s) found by LCMS: [M + H]+ = 766.
Step b. Synthesis of N-(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl) di-(4- formylphenyl) Compound 8 precursor
To a solution of a mixture of di-(4-formylphenyl)methyl amino-PEG10-acid (120 mg, 0.157 mmol) and HATU (71 .5 mg, 0.188 mmol) in anhydrous DMF (0.5 mL) was added DIPEA (40 mg, 0.316 mmol). After the solution was stirred for 10 minutes, it was added with 2-(2-aminoethoxy)ethyl 6-deoxy-alpha-L- altropyranoside TFA salt (59.2 mg, 0.236 mmol). The resulting mixture was stirred for 30 minutes, then directly purified through RPLC (0 to 40% acetonitrile and water). Yield 1 10 mg, 70.1 %. lon(s) found by LCMS calculated: (M+2H)/2 = 500. Step c. Synthesis of 4-Z-1-Boc-piperazine-INT-14
A mixture of (R)-4-(benzyloxy)carbonyl)-1 -(tert-butoxycarbonyl)piperazine-2-carboxylic acid (1 75 mg, 0.48 mmol) and HATU (190 mg, 0.5 mmol) was dissolved in anhydrous DMF (1 mL), and the solution was treated with DIPEA (130 mg, 1 mmol). After the mixture was stirred for 10 minutes, it was added with INT-14 (505 mg, 0.4 mmol). The resulting mixture was stirred for 1 hour, then purified through RPLC (25 to 100 % acetonitrile and water, using 0.1 % formic acid as modifier). Yield 572.4 mg, 94.9 %. lon(s) found by LCMS: [(M - 2Boc)/2]+1 = 705. Step d. Synthesis of 1-Boc-piperazine-INT-14
4-Z-1 -Boc-piperazine-INT-14 (522.9 mg, 0.325 mmol) was dissolved in MeOH (20 mL) and treated with Pd/C. The suspension was stirred under hydrogen for 1 .5 hours. Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation to afford the title product as a tan solid. Yield 465 mg, 97%. lon(s) found by LCMS calculated: [(M-Boc)/2]+1 = 688.
Step e. Synthesis of Compound 8
A mixture of 1 -Boc-piperazine-INT-14 (98 mg, 0.0644 mmol) and N-(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl) di-(4-formylphenyl) Compound 8 precursor (32 mg, 0.032 mmol) from Step b was dissolved in anhydrous EtOH (-2 mL). NaBH(OAc)3 (41 mg) was added, and the resulting mixture was stirred overnight. Four additional portions of NaBH(OAc)3 (41 mg x 4) were added every 1 hour. Stirring of the reaction was continued for another 2 hours, then purified through HPLC (25 to 50 % acetonitrile and water, using 0.1 % formic acid as modifier). The collected fractions were lyophilized to a white solid (1 9.7 mg). The materials were treated with TFA (-0.5 mL), and the solution was stirred for 20 minutes. The mixture was purified by HPLC (0 to 8.5 % acetonitrile and water, using 0.1 % formic acid). lon(s) found by LCMS: (M+5H)/5 = 583.
Figure imgf000212_0001
Step a. Synthesis of (S)-benzyl 3-((tert-butoxycarbonyl)amino)-4-(octylamino)-4-oxobutanoate
To a solution of n-octylamine (650 mg, 5 mmol) and (S)-4-(benzyloxy)-2-((tert- butoxycarbonyl)amino)-4-oxobutanoic acid (1 .65 g, 5 mmol) in DMF (30 mL) was added EDC (1 .2 g, 6 mmol), HOBT (900 mg, 6 mmol), Hunig's base (1 .4 mL, 10 mmol) at room temperature. The solution was stirred overnight. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil 2.1 g, 85% yield. LC/MS 435.3 [M+H]+ Step b. Synthesis of (S)-diethyl 2,2'-((2-((4-(benzyloxy)-1-(octylamino)-1 ,4-dioxobutan-2-yl)amino)- 2-oxoethyl)azanediyl)diacetate
(S)-benzyl 3-((tert-butoxycarbonyl)amino)-4-(octylamino)-4-oxobutanoate (440 mg, 1 mmol) was treated with TFA and stirred for half hour and then concentrated and dried under high vacuum used for next step without purification.
The deprotected product from the previous step was mixed with [bis(2-ethoxy-2- oxoethyl)amino]acetic acid (250 mg, 1 mmol) in DMF (10 mL) was added EDC (250 mg, 1 .25 mmol), HOBT (200 mg, 1 .25 mmol), Hunig's base (0.3 mL, 2 mmol) at room temperature. The solution was stirred overnight. The solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil 450 mg, 85% yield. LC/MS 564.3 [M+H]+
Step c. Synthesis of (S)-ethyl 3-(2-ethoxy-2-oxoethyl)-7-(octylcarbamoyl)-5,9-dioxo-15- (((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2-yl)oxy)-13-oxa-3,6,10- triazapentadecan-1-oate
(S)-diethyl 2,2'-((2-((4-(benzyloxy)-1 -(octylamino)-l ,4-dioxobutan-2-yl)amino)-2- oxoethyl)azanediyl)diacetate (300 mg, 0.5 mmol) was dissolved into 3 mL MeOH and 3 mL ethyl acetate, then 0.1 g of 5% palladium on charcoal was added above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere overnight. The palladium on charcoal was removed by filtration after completion of the reaction by LCMS. The filtrate was concentrated and used in the next step without any purification. LCMS [M+H]+ calculated 473.3.
To the above debenzylated products and 2-(2-aminoethoxy) ethyl 6-deoxy-alpha-L-mannopyranoside (INT-1 ) (150 mg, 0.6 mmol) in DMF (5 mL) was added EDC (120 mg, 0.6 mmol), HOBT (100 mg, 0.6 mmol) and Hunig's base (0.14 mL, 1 mmol) at room temperature. The solution was stirred overnight. The mixture was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 1 00% acetonitrile and water, using 0.1 %
trifluoroacetic acid as the modifier. The product was obtained as an oil, LC/MS 707.4 [M+H]+ Step d. Synthesis of (S)-3-(carboxymethyl)-7-(octylcarbamoyl)-5,9-dioxo-15-(((2R,3R,4R,5R,6S)- 3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2-yl)oxy)-13-oxa-3,6,10-triazapentadecan-1-oic acid
Lithium hydroxide (24 mg, 1 mmol) in 3 mL H2O was added to a solution of (S)-ethyl 3-(2-ethoxy- 2-oxoethyl)-7-(octylcarbamoyl)-5,9-dioxo-15-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H- pyran-2-yl)oxy)-13-oxa-3,6,10-triazapentadecan-1 -oate (210 mg, 0.3 mmol) in a mixed solvent of 3 mL MeOH and 3 mL THF. The resultant solution was stirred for 1 hour at room temperature. The reaction mixture was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 1 00% acetonitrile and water, using 0.1 %
trifluoroacetic acid as the modifier. The product was obtained as an oil, 180 mg 85% yield. LC/MS [M+H] 651 .3.
Step e. Synthesis of dibenzyl 14-(2-(((S)-1-(octylamino)-1 ,4-dioxo-4-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5- trihydroxy-6-methyltetrahydro-2H-pyran-2-yl)oxy)ethoxy)ethyl)amino)butan-2-yl)amino)-2- oxoethyl)-4,12,16,24-tetraoxo-8,20-dioxa-5,11 ,14,17,23-pentaazaheptacosane-1 ,27-dioate
The above (S)-3-(carboxymethyl)-7-(octylcarbamoyl)-5,9-dioxo-1 5-(((2R,3R,4R,5R,6S)-3,4,5- trihydroxy-6-methyltetrahydro-2H-pyran-2-yl)oxy)-13-oxa-3,6,10-triazapentadecan-1 -oic acid (130 mg, 0.2 mmol) and benzyl 4-((2-(2-aminoethoxy)ethyl)amino)-4-oxobutanoate (150 mg, 0.6 mmol) in DMF (5 mL) was added EDC (120 mg, 0.6 mmol), HOBT (100 mg, 0.6 mmol), Hunig's base (0.14 mL, 1 mmol) at room temperature. The solution was stirred overnight. The mixture was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluting with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil, LC/MS 602.3 [M+2H]/2
Step f. Synthesis of 14-(2-(((S)-1-(octylamino)-1 ,4-dioxo-4-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5- trihydroxy-6-methyltetrahydro-2H-pyran-2-yl)oxy)ethoxy)ethyl)amino)butan-2-yl)amino)-2- oxoethyl)-4,12,16,24-tetraoxo-8,20-dioxa-5,11 ,14,17,23-pentaazaheptacosane-1 ,27-dioic acid
Dibenzyl 14-(2-(((S)-1 -(octylamino)-l ,4-dioxo-4-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyltetrahydro-2H-pyran-2-yl)oxy)ethoxy)ethyl)amino)butan-2-yl)amino)-2-oxoethyl)-4,12,16,24- tetraoxo-8,20-dioxa-5,1 1 ,14,17,23-pentaazaheptacosane-1 ,27-dioate..(120 mg, 0.1 mmol) was dissolved into 3 mL MeOH and 3 mL ethyl acetate. Then 0.1 g of 5% palladium on charcoal was added to the solution, and the mixture was stirred at room temperature under the hydrogen atmosphere for overnight. The palladium on charcoal was removed by filtration after completion of the reaction as determined by LCMS. The filtrate was concentrated and used in the next step without any purification. LCMS: [M+2H]/2 512.3.
Step g. Preparation of Compound 9
The title compound was prepared analogously to Compound 1 utilizing 14-(2-(((S)-1 -(octylamino)- 1 ,4-dioxo-4-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2- yl)oxy)ethoxy)ethyl)amino)butan-2-yl)amino)-2-oxoethyl)-4,12,16,24-tetraoxo-8,20-dioxa-5,1 1 ,14,17,23- pentaazaheptacosane-1 ,27-dioic acid (Example 31 , Step f) as the linker, lon(s) found by LCMS: (M+2H)/2 = 1456.8, (M+3H)/3 = 970.6, (M+4H)/4 =728.90, (M+5H)/5 =583.3.
Example 32. Synthesis of Compound 10
Figure imgf000215_0001
Step a. Synthesis of 6-(2-ethoxy-2-oxoethyl)-4,8-dioxo-
3,12,15,18,21 ,24,27,30,33,36,39,42,45,48,51 ,54,57,60,63,66,69,72,75,78,81 -pentacosaoxa-6,9- diazatetraoctacontan-84-oic acid
To a solution of [bis(2-ethoxy-2-oxoethyl)amino]acetic acid (62 mg, 25 mmol and amino-PEG24-t- butyl ester (600 mg, 0.5 mmol ) in DMF (5 mL) was added EDC (120 mg, 0.6 mmol), HOBT (100 mg, 6 mmol) and Hunig's base (0.3 mL, 2 mmol) at room temperature. The solution was stirred overnight. The DMF was removed under reduced pressure and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier.
The above product was treated with TFA and stirred for 30 min, concentrated and dried under high vacuum and used directly for the next step without purification. LC/MS [M/2-t-Butyl]+1 688.4.
Step b. Synthesis of ethyl 3-(2-ethoxy-2-oxoethyl)-5,81-dioxo-87-(((2R,3R,4R,5R,6S)-3,4,5- trihydroxy-6-methyltetrahydro-2H-pyran-2-yl)oxy)- 9,12,15,18,21 ,24,27,30,33,36,39,42,45,48,51 ,54,57,60,63,66,69,72,75,78,85-pentacosaoxa-3,6,82- triazaheptaoctacontan-1 -oate
To a solution of 6-(2-ethoxy-2-oxoethyl)-4,8-dioxo- 3,12,15,18,21 ,24,27,30,33,36,39,42,45,48,51 ,54,57,60,63,66,69,72,75,78,81 -pentacosaoxa-6,9- diazatetraoctacontan-84-oic acid (270 mg, 0.2 mmol) and 2-(2-aminoethoxy)ethyl 6-deoxy-alpha-L- mannopyranoside (75 mg, 0.3 mmol) in DMF (5 mL) was added EDC (70 mg, 0.35 mmol), HOBT (60 mg, 0.4 mmol) and Hunig's base (0.3 mL, 2 mmol) at room temperature. The solution was stirred overnight. The reaction mixture was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil, 150 mg 75% yield. LC/MS (M+2H)/2 805.
Step c. Synthesis of 3-(carboxymethyl)-5,81-dioxo-87-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyltetrahydro-2H-pyran-2-yl)oxy)-
9,12,15,18,21 ,24,27,30,33,36,39,42,45,48,51 ,54,57,60,63,66,69,72,75,78,85-pentacosaoxa-3,6,82- triazaheptaoctacontan-1-oic acid
Lithium hydroxide (12 mg, 0.5 mmol) in 3 mL H2O was added to the solution of 3-(2-ethoxy-2- oxoethyl)-5,81 -dioxo-87-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2-yl)oxy)- 9, 12, 15, 18,21 ,24,27,30,33,36,39,42,45,48, 51 ,54,57,60,63, 66, 69, 72, 75,78, 85-pentacosaoxa-3, 6,82- triazaheptaoctacontan-1 -oate (150 mg, 0.1 mmol) in mix solvent of 3 mL MeOH and 3 mL THF. The resultant solution was stirred for 1 hour at room temperature. The reaction mixture was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid
chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil, 120 mg 75% yield. LC/MS (M+2H)/2 776.9. Step d. Synthesis of dibenzyl 14-(2,78-dioxo-84-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyltetrahydro-2H-pyran-2-yl)oxy)-
6,9,12,15,18,21 , 24,27,30,33,36,39,42,45,48,51 , 54,57,60,63,66,69,72,75,82-pentacosaoxa-3,79- diazatetraoctacontyl)-4,12,16,24-tetraoxo-8,20-dioxa-5,11 ,14,17,23-pentaazaheptacosane-1 ,27- dioate
To a solution 3-(carboxymethyl)-5,81 -dioxo-87-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyltetrahydro-2H-pyran-2-yl)oxy)-
9, 12, 15, 18,21 ,24,27,30,33,36,39,42,45,48, 51 ,54,57,60,63, 66, 69, 72, 75,78, 85-pentacosaoxa-3, 6,82- triazaheptaoctacontan-1 -oic acid (150 mg, 0.1 mmol) and benzyl 4-((2-(2-aminoethoxy)ethyl)amino)-4- oxobutanoate (1 00 mg, 0.3 mmol) in DMF (33 mL) was added EDC (60 mg, 0.3 mmol), HOBT (50 mg, 0.3 mmol) and Hunig's base (0.14 mL, 1 mmol) at room temperature. The solution was stirred overnight. The reaction mixture was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil, LC/MS 1053.1 [M/2]+H+
Step e. Synthesis of 14-(2,78-dioxo-84-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H- pyran-2-yl)oxy)-6,9,12,15,18,21 , 24,27,30,33,36,39,42,45,48,51 , 54,57,60,63,66,69,72,75,82- pentacosaoxa-3,79-diazatetraoctacontyl)-4,12,16,24-tetraoxo-8,20-dioxa-5,11 ,14,17,23- pentaazaheptacosane-1 ,27-dioic acid
Dibenzyl 14-(2,78-dioxo-84-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2- yl)oxy)-6,9,12,1 5,1 8,21 ,24,27,30,33,36,39,42,45,48,51 ,54,57,60,63,66, 69, 72,75, 82-pentacosaoxa-3, 79- diazatetraoctacontyl)-4,12,16,24-tetraoxo-8,20-dioxa-5,1 1 ,14,17,23-pentaazaheptacosane-1 ,27-dioate (1 10 mg, 0.05 mmol) was dissolved into 3 mL MeOH and 3 mL ethyl acetate, then 0.1 g of 5% palladium on charcoal was added above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere for overnight. The palladium on charcoal was removed by filtration after the reaction was determined to reach completion by LCMS. The filtrate was concentrated and used next step without any purification. LCMS: [M+2H]/2 963.0
Step f. Preparation of Compound 10
The title compound was prepared analogously using the procedure described in Example 23
(Steps g and h) but utilized 14-(2,78-dioxo-84-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro- 2H-pyran-2-yl)oxy)-6, 9, 12, 15, 18,21 ,24,27,30,33,36,39,42,45,48, 51 ,54,57,60,63,66, 69,72,75,82- pentacosaoxa-3,79-diazatetraoctacontyl)-4,12,16,24-tetraoxo-8,20-dioxa-5,1 1 ,14,17,23- pentaazaheptacosane-1 ,27-dioic acid (Example 32, Step e) as the linker, lon(s) found by LCMS:
(M+3H)/3 = 1272.1 , (M+4H)/4 =954.3, (M+5H)/5 =763.6. (M+6H)/6 =636.5. Example 33. Synthesis of Compound 11
Figure imgf000218_0001
Step a. Synthesis of dimethyl (2S,5S,8S,16S,19S,22S)-12-[(benzyloxy)carbonyl]-2,8,16,22- tetrakis{2-[(tert-butoxycarbonyl)amino]ethyl}-5,19-bis[(1 R)-1-hydroxyethyl]-4,7,10,14,17,20- hexaoxo-3,6,9,12,15,18,21 -heptaazatricosane-1 ,23-dioate
A stirring solution of methyl (2S)-2-[(N-{(2S)-2-amino-4-[(tert-butoxycarbonyl)amino]butanoyl}-L- threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate (0.399 g, 0.748 mmol), 2,2'- {[(benzyloxy)carbonyl]azanediyl}diacetic acid (0.100 g, 0.374 mmol), and DIEA(0.261 mL, 1 .50 mmol), in DMF (1 mL), were treated with a solution of HATU (0.285 g, 0.748 mmol) added dropwise over 30 minutes, at room temperature. The desired product was isolated by reversed phase liquid
chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.200 g, 41 %. lon(s) found by LCMS: (M+H)+ = 1298.7 Step b. Synthesis of (2S,5S,8S,16S,19S,22S)-12-[(benzyloxy)carbonyl]-2,8,16,22-tetrakis{2-[(tert- butoxycarbonyl)amino]ethyl}-5,19-bis[(1 R)-1-hydroxyethyl]-4,7,10,14,17,20-hexaoxo- 3,6,9,12,15,18,21 -heptaazatricosane-1 ,23-dioic acid
A solution of dimethyl (2S,5S,8S,1 6S,19S,22S)-12-[(benzyloxy)carbonyl]-2,8,16,22-tetrakis{2- [(tert-butoxycarbonyl)amino]ethyl}-5,19-bis[(1 R)-1 -hydroxyethyl]-4,7,10,14,17,20-hexaoxo- 3,6, 9, 12, 15, 18,21 -heptaazatricosane-1 ,23-dioate (0.380 g, 0.242 mmol), in methanol (1 mL) was treated with a solution of lithium hydroxide (0.028 g, 1 .1 71 mmol), in water (1 mL), then stirred at room temperature for 30 minutes. The reaction was made slightly acidic (pH=5) with concentrated HCI (a few drops). The desired product was isolated directly by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.1 64 g, 44%. lon(s) found by LCMS: [(M-2Boc)+2H]/2 = 535.8, [(M-3Boc)+2H]/2 =485.8 Step c. Synthesis of Cbz-deca-Boc Intermediate
A solution of (2S,5S,8S,16S,19S,22S)-12-[(benzyloxy)carbonyl]-2,8,16,22-tetrakis{2-[(tert- butoxycarbonyl)amino]ethyl}-5,19-bis[(1 R)-1 -hydroxyethyl]-4,7,10,14,17,20-hexaoxo-3,6,9,12,1 5,1 8,21 - heptaazatricosane-1 ,23-dioic acid (0.164 g, 0.129 mmol), INT-1 1 (0.329 g, 0.310 mmol), DIEA (0.146 mL, 0.839 mmol), and DMF (1 mL), was treated with a solution of HATU (0.175 g, 0.460 mmol) in DMF (1 mL), added dropwise over 30 minutes. The crude reaction mixture was taken on to the next step without purification, lon(s) found by LCMS: [(M-2Boc)+3H]/3 = 1 053.3, [(M-3Boc)+3H]/3 = 1 019.9, [(M- 4Boc)+3H]/3 = 986.6. Step d. Synthesis of deca-Boc Intermediate
Crude Cbz-deca-Boc Intermediate from Step c (DMF solution) was diluted with methanol (10 mL), charged with 5%Pd/C (0.1 50 g), and hydrogen from a balloon. The reaction was monitored by LCMS. After 2 hr the mixture was filtered through Celite, concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.252 g, 61 % (two steps), lon(s) found by LCMS: (M+3H)/3 = 1 075.3
Step e. Synthesis of deca-Boc-(Compound 11)
A solution of deca-Boc-intermediate from Step d (0.100 g, 0.031 mmol), 4-[(2-{2-[(6-deoxy-alpha- L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoic acid (INT-2) (0.012 g, 0.0341 mmol, and DIEA (0.016 mL, 0.093 mmol), in DMF (1 mL), were treated with HATU (0.014 g, 0.037 mmol), while stirring at room temperature. After 30 minutes, the product was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 1 00% methanol and water, using no modifier. Yield 0.095 g, 86% yield, lon(s) found by LCMS: [(M-2Boc)+3H]/3 = 1 1 19.7, [(M- 3Boc)+3H]/3 = 1086.3
Step f. Synthesis of Compound 11
A solution of deca-Boc-(Compound 1 1 ) (0.095 g, 0.027 mmol), dissolved in DCM (1 mL), was treated with TFA (1 mL), while stirring at room temperature. After 5 minutes, the product was
concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield 0.061 g, 75% yield, lon(s) found by LCMS: (M+3H)/3 = 852.8, (M+4H)/4 = 639.8 Example 34.
Figure imgf000220_0001
Compound 12 was prepared analogously to Compound 1 1 , where 4-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoic acid was substituted with 1 -[(6-deoxy-alpha-L- mannopyranosyl)oxy]-7,10-dioxo-3,14,17,20,23-pentaoxa-6,1 1 -diazahexacosan-26-oic acid in the first step of the sequence, lon(s) found by LCMS: (M+4H)/4 = 701 .6, (M+5H)/5 = 561 .5
Example 35. Synthesis of Compo
Figure imgf000220_0002
Step a. Synthesis of 5-(2-{[(benzyloxy)carbonyl]amino}ethoxy)benzene-1 ,3-dicarboxylic acid
Dimethyl-5-hydroxyisophthalate (1 .0 g, 4.76 mmol), benzyl (2-bromoethyl)carbamate (1 .5 g, 6.19 mmol) and cesium carbonate (1 .6 g, 4.76 mmol) were stirred together in DMF (3 mL) at 50 °C for 12 hours. The mixture was cooled, filtered and applied directly to reversed phase HPLC (20-95% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were concentrated and stirred in a 1 /1 /2 mixture of methanol/THF/DI water (15 mL) containing lithium hydroxide (0.68 g, 28.6 mmol) at room temperature for 2 hours. The mixture was neutralized with 1 N HCI aqueous, extracted into ethyl acetate, concentrated and applied to reversed phase HPLC (10-95% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were pooled and lyophilized to afford 5-(2-{[(benzyloxy)carbonyl]amino}ethoxy)benzene-1 ,3-dicarboxylic acid as a white solid. Yield: 79%, 2 steps. LC/MS [M-H-] - 358.4. Step b. Synthesis of (2S,2'S)-2,2'-([5-(2-{[(benzyloxy)carbonyl]amino}ethoxy)-1 ,3- phenylene]bis{carbonylazanediyl[(2S,3R)-3-hydroxy-1-oxobutane-2,1-diyl]azanediyl})bis{4-[(tert- butoxycarbonyl)amino]butanoic acid}
HATU (550 mg, 1 .22 mmol) was added to a stirring mixture of 5-(2- {[(benzyloxy)carbonyl]amino}ethoxy)benzene-1 ,3-dicarboxylic acid (200 mg, 0.56 mmol), INT-21 (408 mg, 0.122 mmol), DIEA (431 mg, 3.34 mmol) in DMF (4 mL). The mixture was stirred for 30 minutes and then applied directly to reversed phase HPLC (20-95% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were concentrated and stirred in a 1 /1 /2 mixture of methanol/THF/DI water (15 mL) containing lithium hydroxide (80 mg, 3.3 mmol) at room temperature for 2 hours. The mixture was neutralized with 1 N HCL aqueous and extracted into ethyl acetate, concentrated and applied to reversed phase HPLC (1 0-95% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were pooled and lyophilized to afford (2S,2'S)-2,2'-([5-(2- {[(benzyloxy)carbonyl]amino}ethoxy)-1 ,3-phenylene]bis{carbonylazanediyl[(2S,3R)-3-hydroxy-1 - oxobutane-2,1 -diyl]azanediyl})bis{4-[(tert-butoxycarbonyl)amino]butanoic acid} as a white solid. Yield: 88 %, 2 steps. LC/MS (M+2H)/2 = 481 .2.
Step c. Synthesis of hexa-tert-butyl ([5-(2-aminoethoxy)-1 ,3- phenylene]bis{carbonylazanediyl[(2S,3R)-3-hydroxy-1-oxobutane-2,1-diyl]azanediyl{(2S)-4-[(tert- butoxycarbonyl)amino]-1-oxobutane-2,1-diyl}azanediyl[(2S,5R,8S,11S,14S,17S,22S)-5-benzyl-17- [(1 R)-1-hydroxyethyl]-8-(2-methylpropyl)-3,6,9,12,15,18,23-heptaoxo-1 ,4,7,10,13,16,19- heptaazacyclotricosane-22,2,11 ,14-tetrayl]tri(ethane-2,1-diyl)})hexakiscarbamate
HATU (174 mg, 0.457 mmol) was added to a stirring mixture of (2S,2'S)-2,2'-([5-(2- {[(benzyloxy)carbonyl]amino}ethoxy)-1 ,3-phenylene]bis{carbonylazanediyl[(2S,3R)-3-hydroxy-1 - oxobutane-2,1 -diyl]azanediyl})bis{4-[(tert-butoxycarbonyl)amino]butanoic acid} (200 mg, 0.208 mmol), INT-1 1 (485 mg, 457 mmol) and DIEA (161 mg, 1 .25 mmol) in DMF (4 mL). The mixture was stirred for 30 minutes and then applied directly to reversed phase HPLC (30-95% methanol in Dl water containing 0.1 % formic acid: 25 minute gradient). The pure fractions were pooled and concentrated. The CBZ-protected intermediate was taken up in methanol (15 m) and stirred for 1 hour in the presence of 5% Pd/C (75 mg) under 1 atmosphere of hydrogen. The mixture was filtered, concentrated and applied to reversed phase HPLC (20-95% methanol in Dl water containing 0.1 % formic acid: 25 minute gradient). The pure fractions were pooled and concentrated to afford hexa-tert-butyl ([5-(2-aminoethoxy)-1 ,3- phenylene]bis{carbonylazanediyl[(2S,3R)-3-hydroxy-1 -oxobutane-2,1 -diyl]azanediyl{(2S)-4-[(tert- butoxycarbonyl)amino]-1 -oxobutane-2,1 -diyl}azanediyl[(2S,5R,8S,1 1 S,14S,17S,22S)-5-benzyl-17-[(1 R)- 1 -hydroxyethyl]-8-(2-methylpropyl)-3,6,9,12,15,18,23-heptaoxo-1 ,4,7,1 0,13,16,19- heptaazacyclotricosane-22,2,1 1 ,14-tetrayl]tri(ethane-2,1 -diyl)})hexakiscarbamate as a white solid. Yield: 66 %, 2 steps. LC/MS [m-boc/3+H+] = 872.5.
Step d. Synthesis of Compound 13
HATU was added to a stirring mixture of 4-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoic acid (1 8 mg, 0.051 mmol) , hexa-tert-butyl ([5-(2- aminoethoxy)-1 ,3-phenylene]bis{carbonylazanediyl[(2S,3R)-3-hydroxy-1 -oxobutane-2,1 - diyl]azanediyl{(2S)-4-[(tert-butoxycarbonyl)amino]-1 -oxobutane-2,1 - diyl}azanediyl[(2S,5R,8S ,1 1 S ,14S ,17S,22S)-5-benzyl-17-[(1 R)-1 -hydroxyethyl]-8-(2-methylpropyl)- 3,6,9,12,15,18,23-heptaoxo-1 ,4,7,10,13,16,19-heptaazacyclotricosane-22,2,1 1 ,14-tetrayl]tri(ethane-2,1 - diyl)})hexakiscarbamate (100 mg, 0.034 mmoL) and DIEA (13 mg, 0.103 mmol) in DMF (1 .5 mL). The mixture was stirred for 30 minutes and then applied directly to reversed phase HPLC (30-95% acetonitrile in Dl water containing 0.1 % formic acid: 25 minute gradient). The pure fractions were pooled and concentrated and the resulting white solid was stirred in a 1 /1 mixture of TFA/DCM containing 0.1 % thioanisole (4 mL) at room temperature for 30 minutes. The solvent was removed by rotovap, azeotroped twice with toluene. The residue was dissolved in Dl water (1 mL) and applied to reversed phase HPLC (0- 35% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were pooled and lyophilized to afford the title compound (Compound 13) as a white solid. Yield: 23%, 2 steps. LC/MS (M+3H)/3 = 816.8. Example 36. Synthesis of Compound 14
Figure imgf000222_0001
Step a. Synthesis of methyl (2S)-2-[(N-{(2S)-2-{[(2S)-2-
{[(benzyloxy)carbonyl]amino}octanoyl]amino}-4-[(tert-butoxycarbonyl)amino]butanoyl}-L- threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate
A stirring solution of methyl (2S)-2-[(N-{(2S)-2-amino-4-[(tert-butoxycarbonyl)amino]butanoyl}-L- threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate (0.300 g, 0.562 mmol), (2S)-2- {[(benzyloxy)carbonyl]amino}octanoic acid (0.165 g, 0.562 mmol), and DIEA (0.294 mL, 1 .69 mmol), in DMF (1 mL), were treated with HATU (0.235 g, 0.618 mmol) at room temperature. The desired product was isolated by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using 0.1 % TFA as the modifier. Yield 0.296g, 65%. lon(s) found by LCMS: (M+H)+ = 808.5 Step b. Synthesis of methyl (2S)-2-[(N-{(2S)-2-{[(2S)-2-aminooctanoyl]amino}-4-[(tert- butoxycarbonyl)amino]butanoyl}-L-threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate
A solution of methyl (2S)-2-[(N-{(2S)-2-{[(2S)-2-{[(benzyloxy)carbonyl]amino}octanoyl]amino}-4- [(tert-butoxycarbonyl)amino]butanoyl}-L-threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate (1 .160 g, 1 .433 mmol), in methanol (5 mL), was charged with 5%Pd/C (0.400 g), and hydrogen from a balloon. When the reaction was complete by LCMS (1 hr), the mixture was filtered through Celite, concentrated and taken-on to the next step without purification. Yield 0.630 g, 65%. lon(s) found by LCMS: (M+H)+ = 675.4
Step c. Synthesis of dimethyl (2S,5S,8S,11S,19S,22S,25S,28S)-15-[(benzyloxy)carbonyl]-2,8,22,28- tetrakis{2-[(tert-butoxycarbonyl)amino]ethyl}-11 ,19-dihexyl-5,25-bis[(1 R)-1-hydroxyethyl]- 4,7,10,13,17,20,23,26-octaoxo-3,6,9,12,15,18,21 , 24,27-nonaazanonacosane-1 ,29-dioate
A solution of methyl (2S)-2-[(N-{(2S)-2-{[(2S)-2-aminooctanoyl]amino}-4-[(tert- butoxycarbonyl)amino]butanoyl}-L-threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate (0.626 g, 0.928 mmol), 2,2'-{[(benzyloxy)carbonyl]azanediyl}diacetic acid (0.124 g, 0.464 mmol), and DIEA(0.364 mL, 2.088 mmol) in DMF (3 mL), were treated with a solution of HATU (0.362 g, 0.951 mmol) in DMF (1 mL), dropwise over 15 minutes. The desired product was isolated by reversed phase liquid
chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.383 g, 52%. lon(s) found by LCMS: [(M-2Boc)+2H]/2 =690.9, [(M-4Boc)+2H]/2 = 590.9
Step d. Synthesis of (2S,5S,8S,11S,19S,22S,25S,28S)-15-[(benzyloxy)carbonyl]-2,8,22,28-tetrakis{2-
[(tert-butoxycarbonyl)amino]ethyl}-11 ,19-dihexyl-5,25-bis[(1 R)-1-hydroxyethyl]-
4,7,10,13,17,20,23,26-octaoxo-3,6,9,12,15,18,21 , 24,27-nonaazanonacosane-1 ,29-dioic acid
A solution of dimethyl (2S,5S,8S,1 1 S,19S,22S,25S,28S)-15-[(benzyloxy)carbonyl]-2, 8,22,28- tetrakis{2-[(tert-butoxycarbonyl)amino]ethyl}-1 1 ,19-dihexyl-5,25-bis[(1 R)-1 -hydroxyethyl]-
4,7,10,13,17,20,23,26-octaoxo-3,6,9,12,15,18,21 ,24,27-nonaazanonacosane-1 ,29-dioate (0.383 g, 0.242 mmol), in hot methanol (10 mL) was treated with a solution of lithium hydroxide (0.0232 g, 0.969 mmol), in water (2 mL), while stirring at room temperature. The desired product was isolated by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 1 00% methanol and water, using no modifier. Yield 0.340 g, 90%. lon(s) found by LCMS: [(M-1 Boc)+2H]/2 = 726.9, [(M-2Boc)+2H]/2 = 676.9
Step e. Synthesis of Cbz-deca-Boc-(Compound 14) Precursor
A solution of (2S,5S,8S,1 1 S,19S,22S,25S,28S)-15-[(benzyloxy)carbonyl]-2,8,22,28-tetrakis{2- [(tert-butoxycarbonyl)amino]ethyl}-1 1 ,19-dihexyl-5,25-bis[(1 R)-1 -hydroxyethyl]-4,7,10,13,17,20,23,26- octaoxo-3,6,9,12,1 5,1 8,21 ,24,27-nonaazanonacosane-1 ,29-dioic acid (0.357 g, 0.230 mmol), INT-1 1 (0.488 g, 0.460 mmol), DIEA (0.191 mL, 1 .095 mmol), and DMF (5 mL), was treated with HATU (0.175 g, 0.460 mmol). The crude reaction mixture was taken on to the next step without purification, lon(s) found by LCMS: [(M-3Boc)+3H]/3 = 1 1 14.0
Step f. Synthesis of deca-Boc-(Compound 14) Precursor
Crude Cbz-deca-Boc-(Compound 14) Precursor was diluted with methanol (1 0 mL), charged with 5%Pd/C (0.250 g), and hydrogen from a balloon. The reaction was monitored by LCMS. After 2 hr the mixture was filtered through Celite, concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 1 00% methanol and water, using no modifier. Yield 0.139 g, 18% (two steps), lon(s) found by LCMS: [(M-2Boc)+3H]/3 = 1 102.6
Step g. Synthesis of deca-Boc-(Compound 14)
Deca-Boc-(Compound 14) Precursor (0.070 g, 0.0200 mmol), INT-2 (0.042 g, 0.120 mmol), DIEA (0.063 mL, 0.359 mmol), and DMF (1 mL) were treated with HATU (0.046 g, 0.120 mmol), while stirring at room temperature. The reaction was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.070 g, 91 % yield, lon(s) found by LCMS: [(M-2Boc)+3H]/3 = 1213.7, [(M-3Boc)+3H]/3 = 1 1 80.4, [(M-4Boc)+3H]/3 = 1 147.0
Step h. Synthesis of Compound 14
Deca-Boc-(Compound 14) (0.070 g, 0.01 82 mmol) was dissolved in DCM (2 mL) and treated with TFA (1 mL ), while stirring at room temperature. After 30minutes, the reaction was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid
chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid(0.1 %) as the modifier. Yield of Compound 14 was 0.013g, 25% yield, lon(s) found by LCMS: (M+4H)/4 = 710.4, (M+5H)/5 = 568.5, (M+6H)/6 = 473.9
Figure imgf000225_0001
Step a. Synthesis of Cbz-deca-Boc-(Compound 14) Precursor
A solution of INT-20 (2.23 g, 1 .308 mmol), 2,2'-{[(benzyloxy)carbonyl]azanediyl}diacetic acid
(0.175 g, 0.654 mmol), DIEA (0.718 mL, 4.12 mmol), and DMF (10 mL), was treated with a solution HATU (0.522 g, 1 .373 mmol) in DMF (3 mL) via syringe pump over 1 hr, while stirring at room temperature. The crude reaction mixture was taken on to the next step without purification, lon(s) found by LCMS: [(M- 3Boc)+3H]/3 = 1 1 14.0
Step b. Synthesis of deca-Boc-(Compound 14) Precursor
Crude Cbz-deca-Boc-(Compound 14) Precursor in DMF, was charged with 5% Pd/C (1 .0 g), and hydrogen gas from a balloon. The reaction was monitored by LCMS. After 2 hr the mixture was filtered through Celite, concentrated and purified by C18 reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 20% to 100% methanol and water, using no modifier. Yield 1 .46 g, 64% (two steps), lon(s) found by LCMS: [(M-2Boc)+3H]/3 = 1 1 02.6
Step c. Synthesis of deca-Boc-(Compound 14)
A solution of deca-Boc-(Compound 14) Precursor (1 .46 g, 0.41 6 mmol), 4-[(2-{2-[(6-deoxy-alpha- L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoic acid (INT-2) (0.161 g, 0.458 mmol), DIEA
(0.239 mL, 1 .374 mmol), and DMF (5 mL) were treated with a solution of HATU (0.174 g, 0.457 mmol) in DMF (3 mL), dropwise over 30 minutes, while stirring at room temperature. The reaction was purified by C18 reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 1 .56 g, 97% yield, lon(s) found by LCMS: [(M-2Boc)+3H]/3 = 1213.7, [(M-3Boc)+3H]/3 = 1 180.4, [(M-4Boc)+3H]/3 = 1 147.0 Step d. Synthesis of Compound 14
Deca-Boc-(Compound 14) (1 .56 g, 0.406 mmol) was dissolved in DCM (10 mL) and treated with TFA (10 mL), while stirring at room temperature for 5 minutes. The reaction was concentrated at 0°C via rotary evaporation, stored under high vacuum for 1 hr to remove residual TFA, dissolved in methanol and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid
chromatograph eluted with 0% to 30% acetonitrile and water over 20 minutes, using formic acid (0.1 %) as the modifier. After analysis by HPLC the desired fractions are pooled and concentrated to colorless foam. Yield of Compound 14 was 1 .18 g, 96% yield, lon(s) found by LCMS: (M+4H)/4 = 710.4, (M+5H)/5 = 568.5, (M+6H)/6 = 473.9
E
Figure imgf000226_0001
Figure imgf000226_0002
Step a. Synthesis of Cbz-deca-Boc-(Compound 15) Precursor
A stirring solution of INT-17 (0.200 g, 0.131 mmol), Cbz-L-aspartic acid (0.017 g, 0.065 mmol), and DIEA (0.068 mL, 0.392 mmol), dissolved in DMF (1 mL), was treated with a solution of HATU (0.050 g, 0.0131 mmol) in DMF (1 .0 mL), dropwise over 30 minutes. After 1 hour the product was taken on to the next step without further purification, lon(s) found by LCMS: [(M-3Boc)+3H]/3 = 997.3, [(M- 4Boc)+3H]/3 = 964.0, [(M-5Boc)+3H]/3 = 930.6 Step b. Synthesis of deca-Boc-(Compound 15) Precursor
A solution of Cbz-deca-Boc-(Compound 15) Precursor (431 mg, 0.131 mmol) dissolved in methanol (5 mL), was charged with 5% Pd/C (0.050 g), flushed with hydrogen from a balloon, and stirred under a hydrogen atmosphere until Cbz removal was complete by LCMS (~0.5hr). The crude product was isolated by filtration through Celite, then concentrated to an oil, which was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 1 00% methanol and water, using no modifier. Yield for two steps 0.147 g, 71 % yield, lon(s) found by LCMS: [(M-3Boc)+3H]/3 = 952.6, [(M-4Boc)+3H]/3 = 91 9.3 Step c. Synthesis of deca-Boc-Compound 15
A solution of deca-Boc-(Compound 15) Precursor (0.1 06 g, 0.0335 mmol), INT-2 (0.013 g, 0.0370 mmol), and DIEA (0.019 mL, 0.1 1 1 mmol), in DMF (1 mL), were treated with HATU (0.046 g, 0.120 mmol), while stirring at room temperature. After 30 minutes, the product was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 1 00% methanol and water, using no modifier. Yield 0.1 15 g, 98% yield, lon(s) found by LCMS: [(M- 2Boc)+3H]/3 = 1097.0, [(M-3Boc)+3H]/3 = 1063.7
Step d. Synthesis of Compound 15
A solution of deca-Boc-(Compound 15) (0.109 g, 0.031 mmol) in DCM (1 mL) was treated TFA (1 mL), while stirring at room temperature. After 5 minutes the solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 1 00% acetonitrile and water, using formic acid as the modifier. Yield of Compound 15 was 0.033 g, 40% yield, lon(s) found by LCMS: (M+3H)/3 = 830.2, (M+4H)/4 =622.9, (M+5H)/5 =498.5. Example 39. Sy
Figure imgf000227_0001
A stirring solution of INT-17 (0.150 g, 0.098 mmol), fumaric acid (0.0057 g, 0.049 mmol), and DIEA (0.051 mL, 0.294 mmol) in DMF (1 mL), was treated with a solution of HATU (0.037 g, 0.098 mmol) in DMF (0.5 mL), dropwise over 15 minutes. After 1 hour the product was isolated by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 1 00% methanol and water, using no modifier. Yield 0.108 g, 70% yield, lon(s) found by LCMS: [(M- 3Boc)+3H]/3 = 947.0, [(M-4Boc)+3H]/3 = 913.6, [(M-5Boc)+3H]/3 = 880.3
Figure imgf000228_0001
Step a. Synthesis of Benzyl-deca-Boc-(Compound 16) Precursor
Benzyl-deca-Boc-(Compound 1 6) Precursor was prepared analogously as in Example 39, where 1 -benzyl-3,3-azetidinedicarboxylic acid, was substituted for fumaric acid, lon(s) found by LCMS: [(M- 2Boc)+3H]/3 = 1020.0, [(M-3Boc)+3H]/3 = 986.6
Step b. Synthesis of deca-Boc-(Compound 16) Precursor
A solution of benzyl-deca-Boc-(Compound 16) Precursor (0.175 g, 0.537 mmol) dissolved in methanol (5 mL), was charged with 5% Pd/C (0.050 g), flushed with hydrogen gas from a balloon, and stirred under a hydrogen atmosphere until debenzylation was complete by LCMS (~2hr). The crude product was isolated by filtration through Celite, then concentrated to an oil, and used in the next step without further purification. Step c. Synthesis of deca-Boc-(Compound 16)
A solution of deca-Boc-(Compound 16) Precursor (0.175 g, 0.0552 mmo), 4-[(2-{2-[(6-deoxy- alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoic acid (INT-2) (0.0291 g, 0.0828 mmol) and DIEA (0.029 mL, 0.166 mmol), in DMF(1 mL ), was treated with a solution of HATU (0.0220 g, 0.0580 mmol) in DMF (0.5 mL ), dropwise over 30 minutes. After stirring for 1 hr at room temperature, the product was isolated by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.089 g, 46%. lon(s) found by LCMS: [(M-1 Boc)+3H]/3 = 1 134.3, [(M-2Boc)+3H]/3 = 1 1 01 .0 Step d. Synthesis of Compound 16
A solution of deca-Boc-(Compound 16) (0.025 g, 0.00767 mmol), dissolved in methylene chloride (1 mL) was treated with TFA (1 mL), while stirring at room temperature. After 5 minutes, the product was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield 0.01 Og , 58% yield, lon(s) found by LCMS: (M+2H)/2 = 1250.7, (M+3H)/3 = 834.2
Example 41. Synthesis of Compound 17
Figure imgf000229_0001
Step a. Synthesis of the dimethyl ester linker
To a solution of a mixture of 14-{2-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}-4,12,16,24-tetraoxo-8,20-dioxa-5,1 1 ,14,17,23- pentaazaheptacosane-1 ,27-dioic acid (from Example 23, Step f) (24.9 mg, 0.0312 mmol), methyl (2S)-2- [(N-{(2S)-2-amino-4-[(tert-butoxycarbonyl)amino]butanoyl}-L-threonyl)amino]-4-[(tert- butoxycarbonyl)amino]butanoate (36.8 mg, 0.07 mmol), and EteN (20.2 mg, 0.2 mmol) in anhydrous DMF (0.5 mL) was added dropwise a solution of HATU (26.2 mg, 0.069 mmol) in anhydrous DMF (0.5 mL) over 30 minutes. The reaction was stirred for 1 hour, then directly purified through RPLC (15 to 55 % acetonitrile and water). Yield 48.7 mg, 85.4%. lon(s) found by LCMS: [(M - Boc)/2]+1 = 864.5.
Step b. Synthesis of the linker
The dimethyl ester linker (Step a) was dissolved in MeOH (1 mL). After the solution was cooled in an ice-water bath, a solution of LiOH (10 mg, 0.25 mmol) was added. The reaction was stirred for 2 hours, then acidified by formic acid (0.02 mL) and purified through RPLC (5 to 78 % MeOH and water). Yield 46.4 mg, 96.9%. lon(s) found by LCMS: [(M - Boc)/2]+1 = 850.
Step c. Synthesis of Compound 17
To a solution of a mixture of the linker (Step b) (46.4 mg, 0.026 mmol), INT-1 1 (60.6 mg, 0.057 mmol), and EteN (1 0.1 mg, 0.1 mmol) in anhydrous DMF (1 mL) was added dropwise a solution of HATU (21 mg, 0.055 mmol) in anhydrous DMF (0.5 mL) over 30 minutes. The reaction was stirred for 1 hour, then directly purified through RPLC (25 to 100 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid (79.5 mg, LCMS: [(M - Boc)/3]+1 = 872). The material was dissolved in TFA (~ 0.5 mL). After the solution was stirred for 15 minutes, it was purified by HPLC (3 to 12 % acetonitrile and water). Yield 10.8 mg, 10.3%. lon(s) found by LCMS: (M+4H)/4 = 722.
Example 42. Synth
Figure imgf000230_0001
The title compound was prepared analogously to Compound 1 but utilized 2,2'-({2-[(2-{2-[(6- deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}azanediyl)diacetic acid (Example 23, Step d) as the linker, lon(s) found by LCMS (M+2H)/2 = 1 157.6, (M+3H)/3 =772.1 , (M+4H)/4 =579.3. (M+5H)/5 =463.7. Example 43. Synthesis of Compound 19
Step a. Synthesis of dimethyl 5-(2-aminoethoxy)benzene-1 ,3-dicarboxylate
Dimethyl-5-hydroxyisophthalate (2 g, 9.5 mmol), benzyl (2-bromoethyl)carbamate (1 .6 g, 12.4 mmol) and cesium carbonate (3 g, 9.5 mmol) were stirred together in DMF (6 mL) at 50 °C for 12 hours. The mixture was cooled, filtered and applied directly to reversed phase HPLC (20-95% acetonitrile in Dl water containing 0.1 % formic acid: 20 minute gradient). The pure fractions were concentrated and stirred in a mixture of methanol (25 mL) and 5% Pd/C (200 mg) under 1 atmosphere of hydrogen gas for 12 hours. The mixture was filtered, concentrated and purified by reversed phase HPLC (5-95% acetonitrile in Dl water with no modifier: 20 minute gradient) to afford dimethyl 5-(2-aminoethoxy)benzene-1 ,3- dicarboxylate as a clear oil. Yield: 84%, 2 steps. LC/MS [M+H+]+ 254.2. Step b. Synthesis of 5-{2-[4-(1-{2-[2-(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethoxy)ethoxy]ethyl}-1 H-1 ,2,3-triazol-4- yl)butanamido]ethoxy}benzene-1 ,3-dicarboxylic acid
HATU (419 mg, 1 .1 mmol, in 1 mL DMF) was added to a stirring mixture of 4-(1 -{2-[2-(2-{2-[(6- deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethoxy)ethoxy]ethyl}-1 H-1 ,2,3-triazol-4-yl)butanoic acid (526 mg, 1 .1 mmol), dimethyl 5-(2-aminoethoxy)benzene-1 ,3-dicarboxylate (254 mg, 1 .0 mmol) and diisopropylethylamine (259 mg, 2.0 mmol) in DMF (3 mL). The mixture was stirred for 45 minutes then applied directly to reversed phase HPLC (5-95% acetonitrile in Dl water containing 0.1 % TFA: 20 minute gradient). The pure fractions were concentrated and stirred in 1 /1 /2 mixture of THF/methanol/DI water (15 mL) containing lithium hydroxide (144 mg, 6.0 mmol) for 20 minutes. The mixture was acidified with TFA (~1 mL) and the volume was concentrated by half on the rotary evaporator. The mixture was applied to reversed phase HPLC (0-40% acetonitrile in Dl water containing 0.1 % TFA:20 minute gradient). The pure fractions were pooled and lyophilized to afford 5-{2-[4-(1 -{2-[2-(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethoxy)ethoxy]ethyl}-1 H-1 ,2,3-triazol-4-yl)butanamido]ethoxy}benzene-1 ,3- dicarboxylic acid as a light greenish colored oil. Yield: 29%, 2 steps. LC/MS [m/2-H]- 683.4.
Step c. Synthesis of (2R)-2-[3-{[(1S)-1-carboxy-2-(naphthalen-2-yl)ethyl]carbamoyl}-5-(2-{4-[1-(2- {2-[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}ethoxy)ethoxy]ethoxy}ethyl)- 1 H-1 ,2,3-triazol-4-yl]butanamido}ethoxy)benzamido]-3-(naphthalen-2-yl)propanoic acid
HATU (347 mg, 0.92 mmol, in 1 mL DMF) was added to a stirring mixture of 5-{2-[4-(1 -{2-[2-(2-{2-
[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethoxy)ethoxy]ethyl}-1 H-1 ,2,3-triazol-4- yl)butanamido]ethoxy}benzene-1 ,3-dicarboxylic acid (285 mg, 0.42 mmol), methyl (2R)-2-amino-3- (naphthalen-2-yl)propanoate hydrochloride (243 mg, 0.92 mmol) and triethylamine (252 mg, 2.5 mmol) in DMF (2 mL). The mixture was stirred for 45 minutes then applied directly to reversed phase HPLC (5-95% acetonitrile in Dl water containing 0.1 % TFA: 20 minute gradient). The pure fractions were concentrated and stirred in 1 /1 /2 mixture of THF/methanol/DI water (10 mL) containing lithium hydroxide (60 mg, 2.5 mmol) for 20 minutes. The mixture was acidified with TFA (~1 mL) and the volume was concentrated by half on the rotary evaporator. The mixture was applied to reversed phase HPLC (0-40% acetonitrile in Dl water containing 0.1 % TFA: 20 minute gradient). The pure fractions were pooled and lyophilized to afford (2R)-2-[3-{[(1 S)-1 -carboxy-2-(naphthalen-2-yl)ethyl]carbamoyl}-5-(2-{4-[1 -(2-{2-[2-(2-{[(2R,3R,4R,5R,6S)- 3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}ethoxy)ethoxy]ethoxy}ethyl)-1 H-1 ,2,3-triazol-4- yl]butanamido}ethoxy)benzamido]-3-(naphthalen-2-yl)propanoic acid as a light greenish colored oil. Yield: 43%, 2 steps. LC/MS [M+H+]+ 1079.4. Step d. Synthesis of N~1 ~,N~3~-bis[(2S)-1-{[(2S,3R)-1-{[(2S)-4-amino-1-oxo-1-
{[(3S,6S,9S,12S,15R,18S,21S)-6,9,18-tris(2-aminoethyl)-15-benzyl-3-[(1 R)-1-hydroxyethyl]-12-(2- methylpropyl)-2,5,8,11 ,14,17,20-heptaoxo-1 , 4,7,10,13, 16,19-heptaazacyclotricosan-21 - yl]amino}butan-2-yl]amino}-3-hydroxy-1-oxobutan-2-yl]amino}-3-(naphthalen-2-yl)-1-oxopropan-2- yl]-5-(2-{4-[1-(2-{2-[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2- yl]oxy}ethoxy)ethoxy]ethoxy}ethyl)-1 H-1 ,2,3-triazol-4-yl]butanamido}ethoxy)benzene-1 ,3- dicarboxamide (Compou
Figure imgf000232_0001
HATU (50 mg, 0.13 mmol, in 0.5 mL DMF) was added dropwise over 30 minutes to a stirring mixture of INT-16 (181 mg, 0.13 mmol), (2R)-2-[3-{[(1 S)-1 -carboxy-2-(naphthalen-2-yl)ethyl]carbamoyl}-5- (2-{4-[1 -(2-{2-[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2- yl]oxy}ethoxy)ethoxy]ethoxy}ethyl)-1 H-1 ,2,3-triazol-4-yl]butanamido}ethoxy)benzamido]-3-(naphthalen-2- yl)propanoic acid (65 mg, 0.060 mmol), and triethylamine (36 mg, 0.36 mmol) in DMF (1 .5 mL) and the reaction was stirred for an additional 30 minutes at room temperature. The mixture was applied directly to reversed phase HPLC (20-95% methanol in Dl water containing no modifier: 20 minute gradient). The pure fractions containing the desired intermediate (LC/MS (M+3H)/3+1 156.8:loss of 2 Bocs) were pooled, concentrated and dried under high vacuum. The Boc-protected intermediate was stirred in a 1 /1 mixture of TFA/DCM containing thioanisole (60 mg, 0.482 mmol) for 20 minutes at ambient temperature. The solvent was removed on the rotary evaporator and purified by to reversed phase HPLC (0-35% acetonitrile in Dl water containing 0.1 % TFA: 20 minute gradient). The pure fractions were pooled and lyophilized to afford N~1 ~,N~3~-bis[(2S)-1 -{[(2S,3R)-1 -{[(2S)-4-amino-1 -oxo-1 - {[(3S,6S,9S,12S,15R,18S.21 S)-6,9,18-tris(2-aminoethyl)-15-benzyl-3-[(1 R)-1 -hydroxyethyl]-12-(2- methylpropyl)-2,5,8,1 1 ,14,17,20-heptaoxo-1 ,4,7,1 0,13,16,1 9-heptaazacyclotricosan-21 -yl]amino}butan-2- yl]amino}-3-hydroxy-1 -oxobutan-2-yl]amino}-3-(naphthalen-2-yl)-1 -oxopropan-2-yl]-5-(2-{4-[1 -(2-{2-[2-(2- {[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}ethoxy)ethoxy]ethoxy}ethyl)-1 H-1 ,2,3-triazol- 4-yl]butanamido}ethoxy)benzene-1 ,3-dicarboxamide (Compound 19) as a white solid. Yield: 41 %, 2 steps. LC/MS (M+3H)/3+ 990.3.
Figure imgf000233_0001
Step a. Synthesis of di-tert-butyl 37-(2-(benzyloxy)-2-oxoethyl)-35,39-dioxo- 4,7,10,13,16,19,22,25,28,31 , 43,46,49,52,55,58,61 ,64,67,70-icosaoxa-34,37,40-triazatriheptacontane- 1 ,73-dioate
To the solution of 2,2'-((2-(benzyloxy)-2-oxoethyl)azanediyl)diacetic acid (70 mg, 0.25 mmol) and amino-PEG10-tert-butyl ester (290 mg, 0.5 mmol) in DMF (5 ml_) was added EDC (120 mg, 0.6 mmol), HOBT (90 mg, 0.6 mmol) and Hunig's base (1 .4 ml_, 10 mmol) at room temperature. The solution was stirred overnight. The reaction mixture was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil, 21 0 mg 75% yield. LC/MS (M+2H)/2 709.0.
Step b. Synthesis of 3-(38,38-dimethyl-2,36-dioxo-6,9,12,15,18,21 , 24,27,30,33,37-undecaoxa-3- azanonatriacontyl)-41 ,41-dimethyl-5,39-dioxo-9,12,15,18,21 ,24,27,30,33,36,40-undecaoxa-3,6- diazadotetracontan-1-oic acid
Di-tert-butyl 37-(2-(benzyloxy)-2-oxoethyl)-35,39-dioxo- 4,7,10,13,16,19,22,25,28,31 ,43,46,49,52,55,58,61 ,64,67,70-icosaoxa-34,37,40-triazatriheptacontane- 1 ,73-dioate (210 mg, 0.15 mmol) was dissolved into a mixture of 5 mL MeOH and 5 mL ethyl acetate, then 0.1 g of 5% palladium on charcoal was added above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere overnight. The palladium on charcoal was removed by filtration after completion of the reaction as determined by LCMS. The filtrate was concentrated and used in the next step without any purification. LC/MS [M/2-t-Butyl]+1 663.9. Step c. Synthesis of 35,39-dioxo-37-(2-oxo-2-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyltetrahydro-2H-pyran-2-yl)oxy)ethoxy)ethyl)amino)ethyl)-
4,7,10,13,16,19,22,25,28,31 , 43,46,49,52,55,58,61 ,64,67,70-icosaoxa-34,37,40-triazatriheptacontane- 1 ,73-dioic acid
To a solution of 3-(38,38-dimethyl-2,36-dioxo-6,9,12,15,18,21 ,24,27,30,33,37-undecaoxa-3- azanonatriacontyl)-41 ,41 -dimethyl-5,39-dioxo-9,12,1 5,18,21 ,24,27,30,33,36,40-undecaoxa-3,6- diazadotetracontan-1 -oic acid (0.1 5 mmol) and INT-1 (50 mg, 0.2 mmol) in DMF (5 mL) was added EDC (20 mg, 0.1 mmol), HOBT (15mg, 0.1 mmol) and Hunig's base (0.3 mL, 0.2 mmol) at room temperature. The solution was stirred overnight. The reaction mixture was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil, 150 mg 75% yield. LC/MS [M/2-2X t-Butyl]+1 724.4.
The above product was treated with TFA and stirred for 30 minutes, concentrated and dried under high vacuum and used in the next step without purification.
Step d. Prepa
Figure imgf000234_0001
To a solution of 14-{2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2- oxoethyl}-4,12,16,24-tetraoxo-8,20-dioxa-5,1 1 ,14,17,23-pentaazaheptacosane-1 ,27-dioic acid (150 mg, 0.1 mmol), triethylamine (0.07 mL, 0.5 mmol) and INT-16 (270 mg, 0.2 mmol) in 5 mL DMF was added HATU (78 mg, 0.2 mmol). The reaction was stirred for 1 hr and then the reaction mixture was
concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. Yield of product, 1 00 mg, 35% yield.
Per-Boc-(Compound 20) from the previous step was treated in 2 mL DCM and 2 mL TFA at room temperature, the solution was stirred 10 min, then concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield 0.015 g, 34% yield, lon(s) found by LCMS: (M+3H)/3 = 1 1 13.0, (M+4H)/4 = 835, (M+5H)/5 =668.2 (M+6H)/6=557. Example 45. Synth
Figure imgf000235_0001
To the solution of 2,2'-({2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2- oxoethyl}azanediyl)diacetic acid (Example 23, Step d) (10 mg, 0.023 mmol), triethylamine (0.07 mL, 0.5 mmol) and INT-18 (74 mg, 0.047 mmol) in 5 mL DMF was added HATU (19 mg, 0.02 mmol) in 1 mL DMF by syringe pump over 1 hour. The reaction mixture was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. Yield of product, 30 mg, 37% yield.
The per-Boc-(Compound 21 ) from the previous step was treated in 2 mL DCM and 2 mL TFA at room temperature, the solution was stirred 10 min, then concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield of Compound 21 was 0.01 5 g, 34% yield. lon(s) found by LCMS: (M+2H)/2 = 1257.7, (M+3H)/3 = 838.8, (M+4H)/4 =629.4, (M+5H)/5=503.7.
Example 46. Synthesis of (2S)-4-amino-2-((S)-6-(2-(((2S,3R)-1-(((S)-4-amino-1-oxo-1-
(((3S,6S,9S,12S,15R,18S,21S)-6,9,18-tris(2-aminoethyl)-15-benzyl-3-((R)-1-hydroxyethyl)-12- isobutyl-2,5,8,11 ,14,17,20-heptaoxo-1 , 4,7,10,13, 16,19-heptaazacyclotricosan-21 -yl)amino)butan-2- yl)amino)-3-hydroxy-1-oxobutan-2-yl)amino)-2-oxoethyl)-2-((R)-1-hydroxyethyl)-4,8,11-trioxo-9-(2- oxo-2-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2- yl)oxy)ethoxy)ethyl)amino)ethyl)-17-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H- pyran-2-yl)oxy)-15-oxa-3,6,9,12-tetraazaheptadecanamido)-N-((3S,6S,9S,15R,18S,21 S)-6,9,18-tris(2-
aminoethyl)-12-benzyl-3-((R)-1-hydroxyethyl)-15-isobutyl-2,5,8,11 ,14,17,20-heptaoxo- 1 ,4,7,10,13,16,19
Figure imgf000236_0001
The title compound was prepared analogously to Compound 1 utilizing 6-(carboxymethyl)-4,8,1 1 - trioxo-9-(2-oxo-2-((2-(2-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2- yl)oxy)ethoxy)ethyl)amino)ethyl)-17-(((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2- yl)oxy)-15-oxa-3,6,9,12-tetraazaheptadecan-1 -oic acid (Example 28, Step d) as the linker, lon(s) found by LCMS: (M+2H)/2 = 1331 .7, (M+3H)/3 = 888.1 , (M+4H)/4 =666.4, (M+5H)/5 =533.3.
Figure imgf000236_0002
Step a. Synthesis of Cbz-D-Thr-Dab(Boc)-OH: (2S)-2-({N-[(benzyloxy)carbonyl]-D- threonyl}amino)-4-[(tert-butoxycarbonyl)amino]butanoic acid
Cbz-D-Thr-OH (commercial) (1 .92 g, 7.37 mmol), Dab(Boc)-OMe (commercial) (2.00 g, 7.37 mmol), EDCI (4.263 g, 1 1 .05 mmol), HOBt (1 .493 g, 1 1 .05 mmol) and NaHCOs (1 .238 g, 14.74 mmol) were weighed into a 100 mL round bottom flask. 50 mL of DCM/DMF (4:1 ) was added into the flask. The mixture was stirred at room temperature overnight (TLC or LC/MS monitoring). After completion, EtOAc (200 mL) was added to dilute the reaction mixture and washed with 1 N aq. HCI, saturated NaHCCb and brine. The organic layer was dried with Na2S04 and concentrated. The residue was purified with normal phase silica (2-7% MeOH/DCM) to give 3.00 g pure desired product as a white solid (87%). Ions were found as: positive charge (M-Boc+H+: 368.2), negative charge (M-H-: 467.2). The solid was dissolved in 10 mL of THF/water (1 :1 ) and treated with LiOH (0.95 equiv.) for 30 minutes at room temperature. The reaction mixture was acidified to pH~2 with 1 N aqueous HCI. THF was removed under reduced pressure. The residue was extracted with EtOAc. After drying and concentration, Cbz-D-Thr-Dab(Boc)-OH was obtained in quantitative yield. Ions were found as: positive charge (M-Boc+H+: 354.2), negative charge (M-H-: 452.2). Step b. Synthesis of D-Thr-Dab(Boc)-OMe: Methyl (2S)-2-({N-[(benzyloxy)carbonyl]-D- threonyl}amino)-4-[(tert-butoxycarbonyl)amino]butanoate~methyl (2S)-4-[(tert- butoxycarbonyl)amino]-2-(D-threonylamino)butanoate
(D-Thr-Dab(Boc)-OMe ) was obtained by hydrogenolysis of the compound from Step a. in quantitative yield. Ion was found as (M+H+: 334.2).
Step c. Synthesis of Cbz-Dab(Boc)-D-Thr-Dab(Boc)-OH : (2S)-2-[(N-{(2S)-2-
{[(benzyloxy)carbonyl]amino}-4-[(tert-butoxycarbonyl)amino]butanoyl}-D-threonyl)amino]-4-[(tert- butoxycarbonyl)amino]butanoic acid
D-Thr-Dab(Boc)-OMe (0.713 g, 2.14 mmol), Z-NH-Dab(Boc)-OH (DCHA salt) (1 .199 g, 2.25 mmol), EDCI (0.615 g, 3.21 mmol), HOBt (0.433 g, 3.21 mmol) and NaHCOs (0.359 g, 4.28 mmol) were weighed into a 100- mL round bottom flask. 20 mL of DCM/DMF (4:1 ) was added into the flask. The mixture was stirred at room temperature overnight. After completion, EtOAc (100 mL) was added to dilute the reaction mixture and washed with 1 N aq. HCI, saturated NaHCOe and brine. Dried with Na2S04 and condensed. The residue was purified with normal phase silica (2-7% MeOH/DCM) to give 1 .186 g pure desired product Cbz-Dab(Boc)-D-Thr-Dab(Boc)-OMe as a white solid (83%). Ion was found as (M-
Boc+H+: 568.2). This white solid was dissolved in 10 mL of THF/water (1 :1 ) and treated with LiOH (1 .05 equiv.) for half hour at room temperature. The reaction mixture was acidified to pH~2 with 1 N aqueous HCI. THF was removed under reduced pressure. The residue was extracted with EtOAc. After drying and condensation Cbz-Dab(Boc)-D-Thr-Dab(Boc)-OH was obtained in quantitative yield. Ions were found as: positive charge (M-Boc+H+: 554.2), negative charge (M-H~: 653.2).
Step d. Synthesis of Dab(Boc)-D-Thr-Dab(Boc)-INT-11 : (2S)-2-[(N-{(2S)-2-
{[(benzyloxy)carbonyl]amino}-4-[(tert-butoxycarbonyl)amino]butanoyl}-D-threonyl)amino]-4-[(tert- butoxycarbonyl)amino]butanoic acid--tri-tert-butyl {[(2S,5R,8S,11 S,14S,17S,22S)-22- ({(8S,11 R,14S)-8-amino-14-{2-[(tert-butoxycarbonyl)amino]ethyl}-11-[(1S)-1-hydroxyethyl]-2,2- dimethyl-4,9,12,15-tetraoxo-3-oxa-5,10,13-triazapentadecan-15-yl}amino)-5-benzyl-17-[(1 R)-1- hydroxyethyl]-8-(2-methylpropyl)-3,6,9,12,15,18,23-heptaoxo-1 ,4,7,10,13,16,19- heptaazacyclotricosane-2,11 ,14-triyl]tri(ethane-2,1-diyl)}triscarbamate
INT-1 1 (0.709 g, 0.668 mmol) and Cbz-Dab(Boc)-D-Thr-Dab(Boc)-OH (0.502 g, 1 .577 mmol) were dissolved in 10 mL of DMF at room temperature followed by adding DIPEA (0.23 mL,1 .34 mmol). To this mixture was slowly added HATU (0.305 g, 0.801 mmol) in 1 .2 ml_ of DMF via syringe pump. After overnight of stirring at room temperature, the reaction mixture was condensed under reduced pressure. The residue was purified with reversed phase C18 with acetonitrile/water (0.1 %TFA) to give 1 .039 g of Cbz-protected Dab(Boc)-D-Thr-Dab(Boc)-INT-1 1 . Ion was found as ((M-2Boc+2H+)/2: 749.6). The Cbz protecting group was removed with 5% Pd/C in MeOH/EtOAc (1 :1 , 1 0 mL) under hydrogen balloon pressure at room temperature for 2 hours. Celite filtration gave Dab(Boc)-D-Thr-Dab(Boc)-INT-1 1 in quantitative yield. Ion was found as ((M+2H+)/2: 782.4).
Step e. Synthesis of Compound 23
The synthesis of Compound 23 was done in an analogous manner to Compound 21 by coupling
Dab(Boc)-D-Thr-Dab(Boc)-INT-1 1 with 2,2'-({2-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}azanediyl)diacetic acid (Example 23, Step d) followed by Boc deprotection with TFA. HPLC with ACN/water (0.1 % TFA) provided Compound 23 as a white powder after lyophilization. Ions were found as ((M+3H+)/3: 837.8).
Example 48. Synthesi
Figure imgf000238_0001
The title compound was prepared analogously to Compound 9 except the diacid of Example 31 , Step d was used as the linker, lon(s) found by LCMS: (M+2H)/2 = 1270.7, (M+3H)/3 = 847.5, (M+4H)/4 =635.9, (M+5H)/5 =508.9
Example 49. Prepara
Figure imgf000239_0001
The title compound was prepared analogously to Compound 24 but utilized INT-15 in place of INT-16 as the starting material, lon(s) found by LCMS: (M+3H)/3 = 891 .5, (M+4H)/4 =668.9, (M+5H)/5 =535.3
Example 50. Synthesis of INT-23
Figure imgf000239_0002
Figure imgf000240_0001
Figure imgf000240_0002
Step a. Synthesis of MeO-L-Dap-L-Thr-Cbz
MeO-L-Dap(p-Boc)-NH2 (HCI salt) (5.000g, 1 eq.), Z-NH-Thr-OH (5.049g, 1 .05eq.), EDCI (5.350g, 1 .5eq.), HOBt (3.733g, 1 .5eq.) and NaHC03 (3.095g, 2eq.) were weighed into a 100-mL round bottom flask. 24 mL of DCM/DMF (4:1 ) was added into the flask. The mixture was stirred at room temperature for less than 3 hrs. (TLC or LC/MS monitoring). After the completion, EtOAc (200 mL) was added to dilute the reaction mixture and washed with 1 N aq. HCI, saturated NaHC03 and brine. Dried with Na2S04 and condensed. The residue was purified with normal phase silica (2-7% MeOH/DCM) to give 8.24g pure desired product (>95%). Mass showed a strong detectable positive charge signal at t=4.1 07 min with 8 min (5-95%) (found M-Boc+H+: 368.21 ).
Step b. Synthesis of MeO-L-Dap-L-Thr-NH2
A solution of MeO-L-Dap-L-Thr-Cbz (1 .00g, 2.205 mmol) in methanol (1 OmL) was charged with 5%Pd/C (0.500 g), and hydrogen from a balloon. The reaction was monitored by LCMS. After 1 hr the mixture was filtered through Celite, concentrated and used immediately in the next step without purification.
Step c. Synthesis of MeO-L-Dap-L-Thr-L-Dab-Cbz
A solution of MeO-L-Dap-L-Thr-NH2(0.704g, 2.205mmol), (Ny-Boc)-L-Dab-Cbz (1 .295 g, 2.426 mmol), DIEA (1 .268 mL, 7.28 mmol), in DMF (8mL), was treated with a solution of HATU (0.922g,
2.426mmol) in DMF(4mL) dropwise via syringe pump over 1 hr. After 1 .5 hr the mixture was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 1 00% methanol and water, using no modifier. Yield 1 .37 g, 95%. Step d. Synthesis of HO-L-Dap-L-Thr-L-Dab-Cbz
A solution of MeO-L-Dap-L-Thr-L-Dab-Cbz (1 .37g, 2.096mmol), in THF (5 mL) and water (5mL) was treated with LiOH (0.0552g, 2.310mmol) in one portion, at room temperature. LCMS after 15minutes show no starting material. The reaction was made slightly acidic with cone. HCI (aq), then extracted into ethyl acetate, and dried over sodium sulfate. The product was >90% pure by LCMS and was used without further purification, lon(s) found by LCMS: (M-H)- 638.2
Step e. Synthesis of ( Boc)- PM B H- L- Dap- L-Th - L- Dab-C bz
A solution of HO-L-Dap-L-Thr-L-Dab-Cbz (1 .190g, 1 .860mmol), Boc-PMB heptapeptide (Int 1 1 ,
1 .976 g, 1 .8603 mmol), DIEA (1 .07 mL, 6.14 mmol), in DMF (8 mL), was treated with a solution of HATU(0.778g, 2.046mmol) in DMF(2mL) dropwise via syringe pump over 1 hr. After 1 .5 hr the mixture was taken on to the next step without purification, lon(s) found by LCMS: [(M-1 Boc)+2H]/2 = 793.0, [(M- 2Boc)+2H]/2 = 743.0
Step f. Synthesis of (Boc)-PMBH-L-Dap-L-Thr-L-Dab-NH2
A solution of crude (Boc)-PMBH-L-Dap-L-Thr-L-Dab-Cbz (1 .86 mmol) in methanol (10mL) was charged with 5%Pd/C (1 .00 g), and hydrogen from a balloon. The reaction was monitored by LCMS. After 2 hr the mixture was filtered through Celite, concentrated and. purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 1 .43g. 49% . lon(s) found by LCMS: [(M-1 Boc)+2H]/2 = 725.9
Step g. Synthesis of (Boc)-PMBH-L-Dap-L-Thr-L-Dab-(L-amino-octanoyl-Cbz)
A solution of (Boc)-PMBH-L-Dap-L-Thr-L-Dab-NH2 (0.7g, 0.452mmol), Cbz-L-amino-octanoic acid(0.139g, 0.474mmol), and DIEA(0.248mL, 1 .423mmol) in DMF(5mL) was treated with a solution of HATU(0.180g, 0.474mmol) in DMF(1 mL) dropwise over 1 h. After 1 .5h the mixture was taken on to the next step without purification, lon(s) found by LCMS: (M+2H)/2 = 913.6, [(M-1 Boc)+2H]/2 = 863.6, [(M- 2Boc)+2H]/2 = 813.6
Step h. Synthesis of (Boc)-PMBH-L-Dap-L-Thr-L-Dab-(L-amino-octanoyl-NH2) (INT-23)
A solution of crude (Boc)-PMBH-L-Dap-L-Thr-L-Dab-(L-amino-octanoyl-Cbz) (0.452 mmol) was charged with 5%Pd/C (0.40 g), and hydrogen from a balloon. The reaction was monitored by LCMS. After 2 hr the mixture was filtered through Celite, concentrated and. purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.616g. 80% . lon(s) found by LCMS: [(M-1 Boc)+2H]/2 = 796.5, [(M-2Boc)+2H]/2 = 746.5, [(M-3Boc)+2H]/2 = 696.5 Example 51. Synthesis of INT-24
Figure imgf000242_0001
INT-24 was prepared analogously to INT-23 where MeO-L-Dap(p-Boc)-NH2 (HCI salt) was replaced with MeO-D-Dap(p-Boc)-NH2 (HCI salt) in the first step of that sequence, lon(s) found by LCMS: [(M-1 Boc)+2H]/2 = 796.5, [(M-2Boc)+2H]/2 = 746.5, [(M-3Boc)+2H]/2 = 696.5
Example 52. Synthesis of INT-25
Figure imgf000242_0002
INT-25 was prepared analogously to INT-23 where MeO-L-Dap(p-Boc)-NH2 (HCI salt) was replaced with MeO-L-Dab(p-Boc)-NH2 (HCI salt) in the first step of that sequence, lon(s) found by LCMS: [(M-2Boc)+2H]/2 = 753.0
Example 53. Synthesis of 3-{[(tert-butoxycarbonyl)(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]methyl}cyclopropane-trans-1 ,2-dicarboxylic acid (pair of diastereomers) (INT-26)
Figure imgf000242_0003
The aldehyde (dimethyl 3-formylcyclopropane-trans-1 ,2-dicarboxylate, racemic, prepared based on Reference Chem. Eur. J., 2013, 19, 6766-6773 , 0.785 g, 4.22 mmol) was dissolved in 20 mL of DCM. To this solution at room temperature, were added Rha-PEG1 -NH2 (INT-1 ) (1 .0596 g, 4.22 mmol, dissolved in 10 mL of 4:1 DCM/MeOH) and TFA (0.65 mL, 8.43 mmol). The mixture was stirred for 20 minutes followed by addition of NaBH(OAc)3 (1 .787 g, 8.43 mmol). The reaction mixture was stirred overnight to give the desired product (dimethyl 3-{[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]methyl}cyclopropane-trans-1 ,2-dicarboxylate, a pair of diastereomers) . Mass showed a strong detectable positive charge signal at tr=0.50 min with 8 min LC- MS run (found M+H+; 422.2). To this reaction mixture, were added DIPEA (2.20mL, 12.6 mmol) and Boc anhydride (2.03 mL, 8.40 mmol). After 3 hrs of stirring, the solvents were removed and the residue was purified with normal phase silica (2-7% MeOH/DCM) to give the desired product (dimethyl 3-{[(tert- butoxycarbonyl)(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)^
trans-1 ,2-dicarboxylate as a pair of diastereomers). Mass spectrum showed a strong detectable positive/negative charge signal at tr=3.60 min with 5 min polar run (2-95%) method (found Positive M- Boc+H+: 394.20; Negative M-H+; 492.00).
Example 54. Synthesis of INT-27
Figure imgf000243_0001
HATU (1 .56 g, 4.1 1 mmol) in DMF (1 .5 mL) was added, dropwise, to a solution of 2,2'- {[(benzyloxy)carbonyl]azanediyl}diacetic acid (0.5g, 1 .87 mmol), norleu-OMe hydrochloride salt (0.71 g, 3.93 mmol), and triethylamine (1 .13 g, 1 1 .23 mmol) in DMF (5 mL) over a period of 30 minutes. The mixture was stirred for an additional 20 minutes then applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the CBZ-protected di-ester as a clear oil, LC/MS [m+H]+ = 522.6. The di-ester intermediate was stirred in a 1 /1 /2 mixture (10 mL) of THF/methanol/water containing LiOH (0.1 8 g, 7.48 mmol) for 20 minutes. The mixture was applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford (2S,2'S)-2,2'-({[(benzyloxy)carbonyl]azanediyl}bis[(1 - oxoethane-2,1 -diyl)azanediyl])dihexanoic acid as a white solid. Yield: 43%, 2 steps. LC/MS [m-H]- =
492.3.
Figure imgf000243_0002
lnt-1 1 (500 mg, 0.47 mmol), HOBt (68 mg, 0.50 mmol), EDC (96 mg, 0.50 mmol), DIEA (130 mg, 1 mmol) and CBZ-(D)-Ser-OH (0.50 mmol) were stirred in DMF (3 mL) at room temperature for 2 hours. The mixture was applied directly to reversed phase HPLC (25-95% acetonitrile in Dl water containing 0.1 % TFA: 20 minute gradient). The pure fractions were pooled, concentrated and directly taken up in methanol (10 mL) and stirred in the presence of 5% Pd/C (75 mg) under 1 atmosphere of hydrogen gas for 1 hour. The mixture was filtered, concentrated and purified by reversed phase HPLC (10-95% acetonitrile in Dl water, no modifier: 20 minute gradient). The pure fractions were pooled and lyophilized to afford tri-tert-butyl {[(2S,5R,8S,1 1 S,14S,17S,22S)-5-benzyl-17-[(1 R)-1 -hydroxyethyl]-8-(2- methylpropyl)-3,6,9,12,15,18,23-heptaoxo-22-(D-serylamino)-1 ,4,7,10,13,16,19-heptaazacyclotricosane- 2,1 1 ,14-triyl]tri(ethane-2,1 -diyl)}triscarbamate (D-ser-(lnt-1 1 )) as a white solid. Yield: 83%. LC/MS [M- Boc+2H]/2 = 525.6.
Step b. Synthesis of L-thr-D-ser-(lnt-11)
L-threonyl-N-[(3S,6S,9S,12S,15R,18S.21 S)-15-benzyl-6,9,18-tris{2-[(tert- butoxycarbonyl)amino]ethyl}-3-[(1 R)-1 -hydroxyethyl]-12-(2-methylpropyl)-2,5,8,1 1 ,14,1 7,20-heptaoxo- 1 ,4,7,10,13,16,1 9-heptaazacyclotricosan-21 -yl]-D-serinamide (L-thr-D-ser-(lnM )) was prepared from tri- tert-butyl {[(2S,5R,8S,1 1 S,14S,17S,22S)-5-benzyl-17-[(1 R)-1 -hydroxyethyl]-8-(2-methylpropyl)- 3,6,9,12,15,18,23-heptaoxo-22-(D-serylamino)-1 ,4,7,10,13,16,19-heptaazacyclotricosane-2,1 1 ,14- triyl]tri(ethane-2,1 -diyl)}triscarbamate (D-ser-(lnt-1 1 )) and Z-Thr-OH using an analogous procedure to that described in Step a. Yield: 81 %. LC/MS [M-2 Boc+2H]/2 = 525.8.
Step c. Synthesis of Z-(Y-Boc)-Dab-L-thr-D-ser-(lnt-11)
INT-28 was prepared from the product of Step b and Z-(Y-Boc)-Dab-OH in an analogous manner as described in Step a. Yield: 78%. LC/MS [M+2H]/2 = 676.2 (loss of 1 Boc group).
Step d. Synthsis of INT-28
HATU (0.1 1 g, 0.30 mmol) in DMF (0.5 ml) was added, dropwise, to a stirring mixture of the product of Step c (0.44 g, 0.30 mmol), and INT-27 (0.70 g, 0.14 mmol) in DMF (1 .5 mL) over a period of 20 minutes. The mixture was applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 30% to 95% methanol and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the CBZ-protected intermediate, LC/MS [M+3H]/3 1020.0 (loss of 2 boc groups). The CBZ-intermediate was stirred in methanol (10 mL) in the presence of 5% Pd/C (100 mg) under 1 atmosphere of hydrogen for 2 hours. The mixture was filtered and concentrated then purified by to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 30% to 95% acetonitrile and water using no modifier. The pure fractions were pooled and lyophilized to afford the title compound as a white solid. Yield: 51 %, 2 steps. LC/MS [M+2H]/2 = 1562.6 (loss of 2 Boc groups). Example 56. Synthesis of INT-29
Figure imgf000245_0001
HATU (0.70 g, 1 .87 mmol) in DMF (1 ml_) was added, dropwise, to a solution of 1 -tert-butyl 4- methyl 4-aminopiperidine-1 ,4-dicarboxylate (0.48 g, 1 .87 mmol), CBZ-norleu-OH (0.45 g, 1 .70 mmol), and triethylamine (0.51 g g, 5.09 mmol) in DMF (2 ml_) over a period of 20 minutes. The mixture was stirred for an additional 20 minutes then applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the CBZ-protected intermediate as white solid. The intermediate was stirred under 1 atmosphere of hydrogen in the presence of 5% Pd/C (100 mg) for 2 hours. The mixture was filtered, concentrated, and applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford 1 -tert-butyl 4-methyl 4-(L-norleucylamino)piperidine-1 ,4-dicarboxylate as a white solid. Yield: 96%, 2 steps. LC/MS [m+H]+ = 372.3.
Example 57. Synthesis of INT-30
Figure imgf000245_0002
HATU (156 mg, 0.41 mmol) in DMF (1 mL) was added, dropwise, to a solution of INT-39 (144 mg, 0.41 mmol), INT-29 (265 mg, 0.32 mmol), and triethylamine (80 mg, 0.79 mmol) in DMF (5 mL) over a period of 30 minutes. The mixture was stirred for an additional 20 minutes then applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the di-ester as a clear oil, LC/MS [M+2H]/2 = 487.3 (loss of 2 boc groups). The di- ester intermediate was stirred in a 1 /1 /2 mixture (8 mL) of THF/methanol/water containing LiOH (30 mg, 1 .26 mmol) for 20 minutes. The mixture was applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 1 0% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford INT-30 as a white solid. Yield: 81 %, 2 steps. LC/MS [M+2H]/2 = 473.5 (loss of 2 boc groups).
Example 58. Synthesis of INT-31
Figure imgf000246_0001
HATU (156 mg, 0.41 mmol) in DMF (0.7 mL) was added, dropwise, to a solution of INT-30 (0.25 g, 0.22 mmol), H-Thr-(Y-Boc)Dab-OMe (0.16 g, 0.48 mmol), and triethylamine (1 10 mg, 1 .09 mmol) in DMF (1 .5 mL) over a period of 30 minutes. The mixture was stirred for an additional 20 minutes then applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 25% to 95% methanol and water using no modifier. The pure fractions were pooled and lyophilized to afford the di-ester as a clear oil, LC/MS [M+2H]/2 = 788.5 (loss of 2 boc groups). The di-ester intermediate was stirred in a 1 /1 /2 mixture (8 mL) of THF/methanol/water containing LiOH (31 mg, 1 .31 mmol) for 20 minutes. The mixture was applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 20% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford INT-31 as a white solid. Yield: 77%, 2 steps. LC/MS [M+2H]/2 = 774.6 (loss of 2 boc groups).
Example 59. Synthesis of INT-32
Figure imgf000246_0002
HATU (0.55 g, 1 .46 mmol) in DMF (1 mL) was added, dropwise, to a solution of INT-1 1 (1 .41 g, 1 .33 mmol), CBZ-Fluoro-Alanine-OH (0.35 g, 1 .46mmol), and triethylamine (0.40 g, 3.98 mmol) in DMF (2 mL) over a period of 20 minutes. The mixture was stirred for an additional 20 minutes then applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the CBZ-protected intermediate as white solid. The intermediate was stirred under 1 atmosphere of hydrogen in the presence of 5% Pd/C (100 mg) for 2 hours. The mixture was filtered, concentrated, and applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using no modifier. The pure fractions were pooled and lyophilized to afford the title compound as a white solid. Yield: 21 %, 2 steps. LC/MS [M+2H]/2 = 476.6 (loss of 2 Boc groups).
Example 60. Synthesis of INT-33
Figure imgf000247_0001
INT-33 was prepared from INT-32 and CBZ-Thr-OH by an analogous method as that for INT-32. Yield: 76%. LC/MS [M+2H]/2 = 527.0 (loss of 2 boc groups).
Example 61. Synthesis of INT-34
Figure imgf000247_0002
INT-34 was prepared from H-(Yboc)-Dab-OMe and CBZ-NorLeu-OH by a similar method as that for INT-32. Yield: 68%. LC/MS [m+H]+ = 246.4 (artifactual loss of 1 boc group).
Example 62. Synthesis of INT-35
Figure imgf000247_0003
HATU (6.3 g, 16.6 mmol) in DMF (2 mL) was added, dropwise, to a solution of 2,2'- {[(benzyloxy)carbonyl]azanediyl}diacetic acid (2.1 g, 7.9 mmol), INT-34 (5.7 g, 16.6 mmol), and triethylamine (4.0 g, 39.5 mmol) in DMF (4 mL) over a period of 20 minutes. The mixture was stirred for an additional 20 minutes then applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the CBZ-protected intermediate as white solid. The intermediate was stirred under 1 atmosphere of hydrogen in the presence of 5% Pd/C (500 mg) for 2 hours. The mixture was filtered, concentrated, and applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using no modifier. The pure fractions were pooled and lyophilized to afford the title compound as a white solid. Yield: 51 %, 2 steps. LC/MS [M+2H]/2 = 588.0 (loss of 2 boc groups).
Example 63. Synthesis of INT-36
Figure imgf000248_0001
INT-36 was prepared from INT-35 and INT-2 by a procedure analogous to the one for INT-30. Yield: 81 %, 2 steps. LC/MS [M-2H]/2 = 445.4 (loss of 2 Boc groups).
Example 64. Synthesis of INT-37a and INT-37b
Figure imgf000248_0002
HATU (3.1 g, 8.1 mmol) in DMF (5 mL) was added, dropwise, to a solution of racemic-transl - (ferf-butoxycarbonyl)pyrrolidine-3,4-dicarboxylic acid (1 g, 3.9 mmol), H-Norleu-OMe-hydrochloride (1 .5 g, 8.1 mmol), and triethylamine (4.0 g, 39.5 mmol) in DMF (10 mL) over a period of 20 minutes. The mixture was stirred for an additional 20 minutes then applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 1 0% to 95% acetonitrile and water using 0.1 % TFA modifier. The two diasteomers were separated and pooled and lyophilized separately into the more polar diasteomer (a) and the less polar diastereomer (b): LC/MS [m+H]+ = 414.2 (loss of 1 boc group) for both boc-protected intermediates. Each diastereomer was stirred in a 1 /1 mixture of DCM/TFA (8 mL) for 20 minutes then concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 0% to 75% acetonitrile and water using no modifier. Yield: 88% (combined yield of both), 2 steps. LC/MS [M+H]+ = 414.2.
Note: The diastereomers have been arbitrarily assigned based on HPLC retention time. Example 65. Synthesis of INT-38a and INT-38b
Figure imgf000249_0001
INT-38a and INT-38b were prepared from INT-37a and INT-37b and 4-[(2-{2-[(6-deoxy-alpha-L mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoic acid by a procedure analogous to the one for INT-30. Yield: 56%, 2 steps. LC/MS [m-H]- = 717.4.
Example 66. Synthesis of 2,2'-({4-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoyl}azanediyl)diacetic acid (INT-39)
Figure imgf000249_0002
Step a. Synthesis of diethyl 2,2'-({4-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoyl}azanediyl)diacetate
A mixture of 4-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoic acid (INT-2) (530 mg, 1 .51 mmol) and diethyl iminodiacetate (343 mg, 1 .82 mmol) was dissolved in anhydrous DMF (3 ml) and DIPEA (390 mg, 3 mmol). The solution was cooled in an ice-water bath and added with a solution of HATU (631 mg, 1 .66 mmol) in DMF (3 ml) via syringe pump at a rate of 1 .5 ml/hr. After the addition, the reaction was stirred for one more hour and then purified though RPLC (130 g, 5 to 15 % acetonitrile and water, using 0.1 % TFA as modifier). The collected fractions were concentrated by rotary evaporation and further dried under high vacuum to afford the title product as clear oil (680 mg, 86.2 %). Ion found by LCMS: [M]+ = 523.
Step b. Hydrolysis of diethyl 2,2'-({4-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoyl}azanediyl)diacetate
The material from Step a was dissolved in a 1 :1 mixture of MeOH and THF (6 ml). The solution was cooled in an ice-water bath and added with a solution of LiOH (1 00 mg, 4 mmol) in water (3 ml). The resulting mixture was stirred at 0 °C to room temperature overnight. It was cooled back to an ice-water bath, and the pH was adjusted to ~ 7 by 4N HCI solution in dioxane. The organic solution was partially removed by rotary evaporation. The residue was purified through RPLC (150 g, 0 to 20 % MeOH and water, using 0.1 % TFA as modifier). Yield 564 mg, 93 % (INT-39). Ion found by LCMS: [M]+ = 467.
Example 67. Synthesis of INT-40
Figure imgf000250_0001
Step a. Synthesis of methyl N-{(2S)-2-{[(benzyloxy)carbonyl]amino}-4-[(tert- butoxycarbonyl)amino]butanoyl}-L-threoninate
A reaction flask was charged with a strong stirrer, L-threonine methyl ester HCI (2.4 g, 14 mmol) and anhydrous DMF (1 0 ml). The material was gently heated to dissolve. After cooled to room temperature, the solution was added with NaZ-Nv-Boc-L-2,4-diaminobutyric acid dicyclohexylammonium (6.4 g, 12 mmol), DIPEA (3.64 g, 28 mmol), DMF (10 ml), and DCM (20 ml). The resulting suspension was added with a solution of HATU (4.8 g, 12.6 mmol) in DMF (12 ml) via syringe pump at a rate of 5 ml/hr. After the addition, the mixture was extracted with water (100 ml) and DCM (80 mL x 2). The combined organic layers were concentrated by rotary evaporation. The residue was purified through RPLC (150 g, 0 to 42 % acetonitrile and water, using 0.1 % TFA as modifier). Yield (5.25 g, 93.7 %). Ion found by LCMS: [M-Boc]+ = 368.
Step b. Synthesis of methyl N-{(2S)-2-amino-4-[(tert-butoxycarbonyl)amino]butanoyl}-L- threoninate
The step-a product (2.05 g, 4.28 mmol) was dissolved in MeOH (~ 20 ml). The solution was treated with Pd/C (1 g), and the resulting mixture was stirred under hydrogen for 2 hours. Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation then further dried under high vacuum to a white solid. Yield (1 .41 g, 98.8 %). Ion found by LCMS: [M]+ = 334.
Step c. Synthesis of methyl (2S)-2-aminooctanoate
A suspension of (S)-2-aminooctanoic acid (650 mg, 4.08 mmol) in MeOH (8 ml) was cooled in an ice-water bath and added dropwise with thionyl chloride (0.5 ml). The mixture was warmed to room temperature and heated at 60 °C under nitrogen for 3 hours. It was then partially concentrated by rotary evaporation, and the residue was purified through RPLC (100 g, 0 to 25 % acetonitrile and water). The collected fractions were concentrated by rotary evaporation and further dried under high vacuum to afford the product as a clear oil (693 mg, 98 %). Ion found by LCMS: [M]+ = 174.
Step d. Synthesis of methyl (15S)-1-[(6-deoxy-alpha-L-mannopyranosyl)oxy]-15-hexyl-11-(2-{[(2S)-
1-methoxy-1-oxooctan-2-yl]amino}-2-oxoethyl)-7,10,13-trioxo-3-oxa-6,11 ,14-triazahexadecan-16- oate
A mixture of methyl (2S)-2-aminooctanoate (490 mg, 2.8 mmol) and 2,2'-({4-[(2-{2-[(6-deoxy- alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoyl}azanediyl)diacetic acid (600 mg, 1 .29 mmol) was dissolved in anhydrous DMF (2 ml) and DIPEA (702 mg, 5.4 mmol). The solution was cooled in an ice-water bath and drop-wise added with a solution of HATU (1 .06 g, 2.8 mmol) in DMF (3 ml) via syringe pump at a rate of 1 .5 ml/hr. After the addition, the reaction was directly purified through RPLC (100 g, 5 to 100 % acetonitrile and water). Yield 528.7 mg, 52.8 %. Ion found by LCMS: [M]+ = 777.
Step e. Synthesis of (15S)-11-(2-{[(1S)-1-carboxyheptyl]amino}-2-oxoethyl)-1-[(6-deoxy-alpha-L- mannopyranosyl)oxy]-15-hexyl-7,10,13-trioxo-3-oxa-6,11 ,14-triazahexadecan-16-oic acid
A solution of step-d product (528.7 mg, 0.681 mmol) in a 1 :1 mixture of MeoH:THF (6 ml) was cooled in an ice-water bath. It was added with a solution of LiOH (40 mg, 1 .67 mmol) in water (3 ml). The mixture was stirred for 3 hours, then neutralized by 4N HCI solution in dioxane. After partially concentrated, the reaction was directly purified through RPLC (100 g, 0 to 80 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 487.4 mg, 95.6 %. Ion found by LCMS: [M]+ = 749. Step f. Synthesis of INT-40-dimethyl ester
A mixture of the step-b product (481 mg, 1 .44mmol) and the step-e product (487.4 mg, 0.65 mmol) was dissolved in anhydrous DMF (2 ml) and DIPEA (338 mg, 2.6 mmol). The solution was cooled in an ice-water bath and added with a solution of HATU (532 mg, 1 .4 mmol) in DMF (2 ml) via syringe pump at a rate of 1 ml/hr. After the addition, the mixture was stirred for 30 minutes, then directly purified through RPLC (100 g, 1 0 to 100 % acetonitrile and water). Yield 735 mg, 81 .9 %. Ion found by LCMS: [(M-2Boc)/2]+ = 590.
Step g. Hydrolysis of INT-40-dimethyl ester
A solution of step-f product (735 mg, 0.533 mmol) in a 1 :1 mixture of MeOH:THF (10 ml) was cooled in an ice-water bath and treated with a solution of LiOH (40 mg, 1 mmol). The mixture was stirred for 3 hours, then neutralized by 4N HCI solution in dioxane. After partially concentrated, the mixture was purified through RPLC (150 g, 5 to 90 % MeOH and water, using 0.1 % TFA as modifier). Yield 712 mg, 98.8 %, white solid. Ion found by LCMS: [(M-2Boc)/2]+ = 576.
Example 68. Preparation of INT-41
Figure imgf000252_0001
Step a. Synthesis of 2,2'-(2-(benzyloxy)-2-oxoethylazanediyl)diacetic acid
Benzyl Bromo-acetate (4.6 g, 20 mmol) was added into the solution of 2,2'-azanediyldiacetic acid (2.7g, 20mmol) and DIPEA (5.6 g, 40 mmol) in 60 mL DMF, the resulted solution was stirred at room temperature for overnight. The reaction solution was concentrated and purified by flash chromatography to provide products. LC/MS, 282.1 [M+H]+.
Step b. Synthesis of benzyl 2-((2-oxo-2-(2-(2-((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyltetrahydro-2H-pyran-2-yloxy)ethoxy)ethylamino)ethyl)(2-oxo-2-(2-(2-((2R,3R,4R,5S)-3,4,5- trihydroxytetrahydro-2H-pyran-2-yloxy)ethoxy)ethylamino)ethyl)amino)acetate
To the solution of 2,2'-(2-(benzyloxy)-2-oxoethylazanediyl)diacetic acid (280mg, 1 mmol) in 5ml_
DMF were added (2R,3R,4R,5S)-2-(2-(2-aminoethoxy)ethoxy)tetrahydro-2H-pyran-3,4,5-triol (INT-1 ) (520mg, 2mmol), EDC (600mg, 3mmol), HOBT (420mg, 3mmol), Hunigs base (1 .4ml_, 10mmol) at room temperature. The solution was stirred for overnight. The resulted solution was removed DMF and purified by reverse phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoacetic acid as the modifier. Yield of oil product, 520mg, 72% yield. LC/MS 734.3 [M+H]
Step c. Synthesis of 2-((2-oxo-2-(2-(2-((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H- pyran-2-yloxy)ethoxy)ethylamino)ethyl)(2-oxo-2-(2-(2-((2R,3R,4R,5S)-3,4,5-trihydroxytetrahydro- 2H-pyran-2-yloxy)ethoxy)ethylamino)ethyl)amino)acetic acid
Benzyl 2-((2-oxo-2-(2-(2-((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2- yloxy)ethoxy)ethylamino)ethyl)(2-oxo-2-(2-(2-((2R,3R,4R,5S)-3,4,5-trihydroxytetrahydro-2H-pyran-2- yloxy)ethoxy)ethylamino)ethyl)amino)acetate (500mg, 0.7mmol) was dissolved into 5mL MeOH and 3 mL ethyl acetate, then 200mg of 5% Palladium on charcoal was added above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere for overnight. The palladium charcoal was removed by filtration after completed the reaction by LCMS. The filtrate was concentrated and used next step without any purification. LC/MS, 644.3 [M+H]+ . Step d. Synthesis of (S)-benzyl 17-(2-(bis(2-oxo-2-(2-(2-((2S,3S,4S,5S,6R)-3,4,5-trihydroxy-6- methyltetrahydro-2H-pyran-2-yloxy)ethoxy)ethylamino)ethyl)amino)acetamido)-7,11-dioxo-9- oxo-2-(2-(2-((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2- yloxy)ethoxy)ethylamino)ethyl)-1-((2R,3R,4R,5R,6S)-3,4 trihydroxy-6-methyltetrahydro-2H-pyran- 2-yloxy)-3-oxa-6,9,12-triazaoctadecan-18-oate
To the solution of 2-((2-oxo-2-(2-(2-((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H- pyran-2-yloxy)ethoxy)ethylamino)ethyl)(2-oxo-2-(2-(2-((2R,3R,4R,5S)-3,4,5-trihydroxytetrahydro-2H- pyran-2-yloxy)ethoxy)ethylamino)ethyl)amino)acetic acid (400mg, 0.6mmol) in 5ml_ DMF were added (S)-benzyl 2,6-diaminohexanoate (70mg, 0.3mmol), EDC (200mg, 1 mmol), HOBT (150mg, 1 mmol), Hunigs base (0.28mL, 2mmol) at room temperature. The solution was stirred for overnight. The resulted solution was removed DMF and purified by reverse phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 1 00% acetonitrile and water, using 0.1 % trifluoacetic acid as the modifier. Yield of oil product, 360mg, 78% yield. LC/MS 758.4 [(M+2H)/2].
Step e. Synthesis of (S)-17-(2-(bis(2-oxo-2-(2-(2-((2S,3S,4S,5S,6R)-3,4,5-trihydroxy-6- methyltetrahydro-2H-pyran-2-yloxy)ethoxy)ethylamino)ethyl)amino)acetamido)-7,11-dioxo-9-(2- oxo-2-(2-(2-((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2- yloxy)ethoxy)ethylamino)ethyl)-1-((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran- 2-yloxy)-3-oxa-6,9,12-triazaoctadecan-18-oic acid (INT-41)
(S)-benzyl 17-(2-(bis(2-oxo-2-(2-(2-((2S,3S,4S,5S,6R)-3,4,5-trihydroxy-6-methyltetrahydro-2H- pyran-2-yloxy)ethoxy)ethylamino)ethyl)amino)acetamido)-7,1 1 -dioxo-9-(2-oxo-2-(2-(2-((2R,3R,4R,5R,6S)- 3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2-yloxy)ethoxy) ethylamino)ethyl)-1 -((2R,3R,4R,5R,6S)- 3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2-yloxy)-3-oxa-6,9,12-triazaoctadecan-18-oate (300 mg, 0.2 mmol) was dissolved into 2ml_ MeOH and 2 ml_ ethyl acetate, then 100mg of 5% Palladium on charcoal was added above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere for overnight. The palladium charcoal was removed by filtration after completed the reaction by LCMS. The filtrate was concentrated and used next step without any purification. LC/MS, 713.7
[(M+2H)/2] . Exam le 69. Synthesis of Compound 26
Figure imgf000254_0001
Step a. Synthesis of methyl (2S)-2-[(2-{[(benzyloxy)carbonyl]amino}ethyl)amino]-4-[(fert- butoxycarbonyl)amino]butanoate
To a solution of (S)-methyl-2-amino-4-((tert-butoxycarbonyl)amino)butanoate HCI (537.5 mg, 2 mmol) in DMF/water (2/0.5 ml) was added benzyl 2-bromoethylcarbamate (567.8 mg, 2.2 mmol) and sodium bicarbonate (252 mg, 3 mmol). After the resulting mixture was stirred for 3 hours, it was added with an additional amount of benzyl 2-bromoethylcarbamate (258 mg, 1 mmol). The reaction was continued overnight and purified through RPLC (1 50 g, 5 to 100 % acetonitrile and water). The collected fractions were concentrated by rotary evaporation to afford the title compound as a clear oil (750 mg, 91 .6%). Ion found by LCMS: [M]+ = 410.
Step b. Synthesis of (8S,11 £,15S)-9,14-bis(2-{[(benzyloxy)carbonyl]amino}ethyl)-15-{2-[(fert- butoxycarbonyl)amino]ethyl}-8-carboxy-2,2-dimethyl-4,10,13-trioxo-3-oxa-5,9,14-triazahexadec-11- en-16-oic acid
A flame-dried reaction flask was filled with nitrogen and charged with step-a product (328 mg, 0.8 mmol), chloroform (2 ml) and DIPEA (41 6 mg, 3.2 mmol). After the solution was cooled in an ice-water bath, it was syringe-pumped added with a solution of fumaryl chloride (61 mg, 0.4 mmol) in chloroform (0.6 ml) at a rate of 0.2 ml/hr. An additional amount of DIPEA (208 mg, 1 .6 mmol) was added for every hour. After the completion of the fumaryl chloride addition, the reaction was stirred for one more hour and then concentrated by rotary evaporation. The residue was purified through RPLC (50 g, 10 to 70 % acetonitrile and water, using 0.1 % TFA as modifier). The collected fractions were concentrated by rotary evaporation to a purple solid (LCMS: [M]+ = 899). The material was re-dissolved in 1 :1 MeOH:THF (3 ml). After cooled in an ice-water bath, the solution was added with LiOH solution (20 mg, 0.8 mmol) in water (1 .5 ml). The mixture was stirred for 3 hours and then acidified by formic acid. It was purified through RPLC (50 g, 20 to 78 % MeOH and water). The collected fractions were concentrated by rotary evaporation and further dried under high vacuum to afford the title product as a white solid (156 mg, 22.4 %). Ion found by LCMS: [M-Boc]+1 = 771 .
Step c. Synthesis of A , ,Af-bis(2-aminoethyl)butanediamide-INT-16
To a mixture of step-b product (56 mg, 0.0643 mmol), INT-16 (192.9 mg, 0.141 mmol) and DIPEA
(78 mg, 0.6 mmol) in anhydrous DMF (1 ml) was added with a solution of HATU (26.9 mg, 0.07 mmol) in DMF (1 ml) via syringe pump at 0.5 ml/hr rate. After the addition, the mixture was stirred for one more hour and then added with Pd/C and EtOAc. The resulting mixture was stirred under hydrogen for 4 hours. Pd/C was filtered off, and the filtrate was concentrate. The residue was purified through RPLC (50 g, 20 to 100 % MeOH and water, using 0.1 % formic acid as modifier). The collected fractions were
concentrated by rotary evaporation to a white solid (34.5 mg, 16.3%). Ions found by LCMS: [(M-Boc)/3]+1 = 1 065, [(M-3Boc)+3H]/3 = 999.
Step d. Synthesis of deca-Boc-Compound 26 precursor
To a solution of a mixture of 4-[(2-{2-[(6-deoxy-a -L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4- oxobutanoic acid (INT-2) (10 mg, 0.0285 mmol) and HOBT hydrate (5.7 mg, 0.037 mmol) in anhydrous DMF (0.5 ml) was added EDC-HCI (6.1 mg, 0.032 mmol). After stirred for 10 minutes, it was added with a solution of step-c product (34.5 mg, 0.0105 mmol) and DIPEA (50 mg, 0.385 mmol) in DMF (1 ml). The reaction was stirred for 2 hours, then directly purified through RPLC (50g, 20 to 100 % MeOH and water). The collected fractions was concentrated by rotary evaporation to a white solid (24.9 mg, LCMS [(M- Boc)/3]+1 = 1287, [(M-2Boc)+3H]/3 = 1254).
Step e. Removal of the Boc group
The step-d product was re-dissolved in TFA/DCM (1 :1 , - 1 ml). After the solution was stirred for 15 minutes, it was purified through HPLC (5 to 22 % acetonitrile and water, using 0.1 % TFA as modifier). Yield (5.8 mg, 13.5 %). Ions found by LCMS: (M+3H)/3 = 987, (M+4H)/4 = 740, (M+5H)/5 = 592. Example 70. Synthesis of Compound 27
Figure imgf000256_0001
HATU (40 mg, 0.106 mmol) in DMF (0.5 ml) was added, dropwise, to a stirring mixture of INT-28 (0.23 g, 0.071 mmol), and 4-[(2-{2-[(6-deoxy-alpha-L mannopyranosyl)oxy]ethoxy}ethyl)amino]-4- oxobutanoic acid (INT-2) (38 mg, 0.107 mmol) in DMF (1 .5 mL) over a period of 20 minutes. The mixture was applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 30% to 95% methanol and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the boc-protected intermediate; LC/MS [M+3H]/3 1086.6 (loss of 2 boc groups). The boc-intermediate was stirred in a 1 /1 mixture of DCM/TFA (5 mL) containing thioanisole (71 mg, 0.071 mmol) for 20 minutes. The mixture was concentrated on the rotovap then applied to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 0% to 55% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the title compound as a white solid. Yield: 41 %, 2 steps. LC/MS [M+3H]/3 = 919.8.
Figure imgf000256_0002
Step a. Coupling of INT-20
To the solution of 2,2'-({2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2- oxoethyl}azanediyl)diacetic acid (1 0 mg, 0.023 mmol), triethyl amine (0.07 mL, 0.5 mmol) and INT-20 (85 mg, 0.05 mmol) in 5ml_ DMF was added HATU (40 mg, 0.1 mmol) in 1 mL DMF by syringe pump over 1 hour. Then the resulted solution was concentrated and purified by reverse phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. Yield of product, 60mg, 70% yield.
Step b. Removal of the Boc groups
The material from Step a was treated in 2 mL DCM and 2mL TFA at room temperature, the solution was stirred 10 min, then concentrated and purified by reverse phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield 35 mg, 60% yield (Compound 28). lon(s) found by LCMS:
(M+3H)/3 = 932.9, (M+4H)/4 =699.9, (M+5H)/5=560.1 .
Figure imgf000257_0001
Step a. Coupling to INT-20
To the solution of (S)-3-(carboxymethyl)-7-(octylcarbamoyl)-5,9-dioxo-1 5-(((2R,3R,4R,5R,6S)- 3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2-yl)oxy)-13-oxa-3,6,10-triazapentadecan-1 -oic acid (15 mg, 0.023 mmol), triethyl amine (0.07mL, 0.5mmol) and INT-20 (85mg, 0.05mml) in 5mL DMF was added HATU (19 mg, 0.5mmol) in 1 mL DMF by syringe pump over 1 hour. Then the resulted solution was concentrated and purified by reverse phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoacetic acid as the modifier. Yield of product, 45mg, 51 % yield.
Step b. Removal of the Boc groups
The per-Boc product from Step a was treated in 2 mL DCM and 2mL TFA at room temperature, the solution was stirred 10 min, then concentrated and purified by reverse phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield 15mg, 48% yield (Compound 29). lon(s) found by LCMS:
(M+3H)/3 = 1008.3, (M+4H)/4 =756.5, (M+5H)/5=605.4, (M+6H)/6=504.6.
Figure imgf000258_0001
HATU (78 mg, 0.20 mmol) in DMF (0.5 mL) was added, dropwise, to a solution of INT-31 (0.17 g, 0.097 mmol), trisBoc-PMBH (INT-1 1 ) (227 mg, 0.21 mmol), and triethylamine (59 mg, 0.58 mmol) in DMF (1 .5 mL) over a period of 30 minutes. The mixture was stirred for an additional 20 minutes then applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 25% to 95% methanol and water using no modifier. The boc-protected intermediate was stirred in a 1 /1 mixture of DCM/TFA (4 mL) containing thioanisole (121 mg, 0.97 mmol) for 20 minutes. The solvent was removed by rotovap and the residue was dried under high vacuum and then purified by reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 0% to 45% acetonitrile and water using no modifier. The pure fractions were pooled and lyophilized to afford the title compound as a white solid. Yield: 59%, 2 steps. LC/MS [M+3H]/3 = 945.9.
Figure imgf000259_0001
Step a. Synthesis of deca-Boc Compound 31 benzyl ester
Deca-Boc Compound 31 benzyl ester was prepared analogously to Example 37 (alternate procedure), where racemic (1 R,2R)-3-[(benzyloxy)carbonyl]cyclopropane-1 ,2-dicarboxylic acid, was substituted for 2,2'-{[(benzyloxy)carbonyl]azanediyl}diacetic acid in the first step of the sequence.
Step b. Synthesis of deca-Boc Compound 31 carboxylic acid
Crude deca-Boc benzyl ester Intermediate (0.373g, 0.103 mmol) from Step a (DMF solution) was charged with 5%Pd/C (0.200 g), and hydrogen from a balloon. The reaction was monitored by LCMS. After 12 hr the mixture was filtered through Celite, concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.295 g, 81 % (two steps), lon(s) found by LCMS: [(M- 3Boc)+3H]/3 = 1083.0, [(M-4Boc)+3H]/3 = 1049.7, [(M-5Boc)+3H]/3 = 1016.3
Step c. Synthesis of deca-Boc Compound 31
A solution of deca-Boc Compound 31 carboxylic acid (0.295g, 0.0831 mmol), L-Rhamnose-PEG1 - NH2 (INT-1 ) (0.01 15g, 0.0457mmol), DIEA(0.0239DL, 0.137mmol), in DMF(3mL), was treated with a solution of HATU(0.0174g, 0.0457mmol) in DMF(0.5mL) dropwise via syringe pump over 1 hr. After 1 .5 hr the mixture was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.057 g, 36%. lon(s) found by LCMS: (M+3H)/3 = 1260.7, [(M-1 Boc)+3H]/3 = 1227.4, [(M-2Boc)+3H]/3 = 1 1 94.0 Step d. Synthesis of Compound 31
A solution of deca-Boc-Compound 31 (0.234 g, 0.0659 mmol), dissolved in DCM (1 ml_), was treated with TFA (1 ml_), while stirring at room temperature. After 5 minutes, the product was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield 0.027 g, 55% yield (mixture of diastereomers). lon(s) found by LCMS: (M+3H)/3 = 927.2, (M+4H)/4 = 695.7, (M+5H)/5 = 556.7, (M+6H)/6 = 464.1 , (M+7H)/7 = 398.0
Figure imgf000260_0001
Compound 32 was prepared analogously to example 37 (alternate procedure) where intermediate INT-20 was replaced with intermediate INT-23 in the first step of that sequence, lon(s) found by LCMS: (M+3H)/3 = 938.1 , (M+4H)/4 = 703.8, (M+5H)/5 = 563.3
Example 76. Synthesis of Compound 33
Figure imgf000261_0001
Compound 33 was prepared analogously to Example 75 where INT-23 was replaced with INT-24 in the first step of that sequence, lon(s) found by LCMS: (M+4H)/4 = 703.8, (M+5H)/5 = 563.3, (M+6H)/6 = 469.6, (M+7H)/7 = 402.6
Figure imgf000261_0002
Figure imgf000262_0001
Step a. Synthesis of ethyl 32-(2-ethoxy-2-oxoethyl)-3,31 -dioxo-1 -phenyl-2,7,10,13,16,19,22,25,28- nonaoxa-4,32-diazatetratriacontan-34-oate
To the solution of 3-oxo-1 -phenyl-2,7,10,13,16,19,22,25,28-nonaoxa-4-azahentriacontan-31 -oic acid (1 g, 2.5 mmol) in DMF (20 mL) were added diethyl 2,2'-azanediyldiacetate (0.71 g, 3.7 mmol), EDC (600mg, 3mmol), HOBT (0.45g, 3.0mmol), Hunigs base (0.7 mL, 5mmol) at room temperature. The solution was stirred overnight. The resultant solution was removed DMF and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. Yield of oil product, 1 .2 g, 65% yield. LC/MS 747.4 [M+H]
Step b. Synthesis of 32-(carboxymethyl)-3,31-dioxo-1 -phenyl-2,7,10,13,16,19,22,25,28-nonaoxa- 4,32-diazatetratriacontan-34-oic acid
Lithium hydroxide (50 mg, 2 mmol) in 2 mL H2O was added to the solution of ethyl 32-(2-ethoxy- 2-oxoethyl)-3,31 -dioxo-1 -phenyl-2,7,10,13,16,19,22,25,28-nonaoxa-4,32-diazatetratriacontan-34-oate (800mg, 1 mmol) in mix solvent of 2 mL H2O, 2mL MeOH and 2mL THF. The resulting solution was stirred for 1 hour at room temperature. The resulted solution was concentrated and purified by reverse phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoro-acetic acid as the modifier. LC/MS 691 .3 [M+H]+
Step c. Synthesis of Bis (penta-Boc-polymyxin B decapeptide)-32-(carboxymethyl)-3,31 -dioxo-1 - phenyl-2,7,10,13,16,19,22,25,28-nonaoxa-4,32-diazatetratriacontan-34-amide
To a solution of 32-(carboxymethyl)-3,31 -dioxo-1 -phenyl-2,7,10,13,16,19,22,25,28-nonaoxa-4,32- diazatetratriacontan-34-oic acid (70 mg, 0.1 mmol), triethyl amine (0.14 mL, 1 mmol) and INT-18 (300 mg, 0.2 mmol) in 5 mL DMF was added HATU (80 mg, 0.2 mmol) in 1 mL DMF by syringe pump over 1 hour. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. Yield of product, 230 mg, 70% yield.
Step d. Synthesis of deca-Boc-lntermediate
The Cbz-protected product from Step c (120 mg, 0.03 mmol) was dissolved into a mixture of 2 mLs of MeOH and 2 mLs of ethyl acetate, then 100 mg of 5% palladium on charcoal was added to the above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere overnight. The palladium charcoal was removed by filtration after completion of the reaction as indiciated by HPLC. The filtrate was concentrated and used next step without any purification.
Step e. Coupling to INT-41
To the solution of (S)-17-(2-(bis(2-oxo-2-(2-(2-((2S,3S,4S,5S,6R)-3,4,5-trihydroxy-6- methyltetrahydro-2H-pyran-2-yloxy)ethoxy)ethylamino)ethyl)amino)acetamido)-7,1 1 -dioxo-9-(2-oxo-2-(2- (2-((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2-yloxy) ethoxy)ethylamino)ethyl)-1 - ((2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyltetrahydro-2H-pyran-2-yloxy)-3-oxa-6,9,12-triazaoctadecan- 18-oic acid (27mg, 0.02 mmol), triethyl amine (0.07mL, 0.5mmol) and INT-41 (70 mg, 0.02 mmol) in 5 mL DMF was added HATU (19 mg, 0.05 mmol) in 1 mL DMF by syringe pump over 1 hour. Then the resulted solution was concentrated and purified by reverse phase liquid chromatography (RPLC) using an Isco
CombiFlash liquid chromatograph eluted with 5% to 1 00% acetonitrile and water, using 0.1 % trifluoacetic acid as the modifier. Yield of product, 60mg, 55% yield.
Step f. Removal of the Boc groups
The product from Step e was treated in 2 mL DCM and 2 mL TFA at room temperature, the solution was stirred 10 min, then concentrated and purified by reverse phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield 16 mg, 38% yield (Compound 34). lon(s) found by LCMS:
(M+4H)/4 = 1014.1 , (M+5H)/5 =81 1 .4, (M+6H)/6=676.4, (M+6H)/6=579.9.
Example 78. Synthesis of Compound 35
Figure imgf000264_0001
Step a. Synthesis of Met-INT-18
A reaction flask was filled with nitrogen and charged with a mixture of Fmoc-L-Met-OH (44.6 mg,
0.12 mmol) and HOBT hydrate (26 mg, 0.17 mmol) and anhydrous DMF ((1 ml). EDC-HCI (25.2 mg, 0.132 mmol) was added, and the resulting mixture was stirred for 15 minutes, then added with INT-18 (168 mg, 0.1 mmol) and DIPEA (91 .5 mg, 0.7 mmol). After stirred for 1 hour, the reaction mixture was quenched with water (0.1 ml) and stirred for 10 minutes. Piperidine (3 drops) was then added, and the resulting mixture was stirred for 2 hours. It was then directly purified through RPLC (50 g, 20 to 82 % MeOH and water). Yield 139.6 mg, 82.4 %. Ions found by LCMS: [(M-Boc)/2]+1 = 798.
Step b. Synthesis of decaBoc-Compound 35 precursor
A reaction flask was filled with nitrogen and charged with a mixture of 2,2'-({4-[(2-{2-[(6-deoxy- alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoyl}azanediyl)diacetic acid (1 1 mg, 0.0236 mmol), step-a product (84.8 mg, 0.05 mmol), anhydrous DMF (0.5 ml), and DIPEA (39 mg, 0.3 mmol). The mixture was cooled in an ice-water bath and added with a solution of HATU (19 mg, 0.05 mmol) in DMF (0.5 ml) via syringe pump at a rate of 0.5 ml/hr. After the addition, the reaction was stirred for one more hour. It was then purified through RPLC (50 g, 20 to 100 % MeOH and water). Yield 53.7 mg, 40.4 %. Ions found by LCMS: [(M-Boc)/3]+1 = 1240, [(M-2Boc)/3]+= 1207, [(M-3Boc)/3]+= 1 173.
Step c. Removal of the Boc groups
The step-b product was dissolved in TFA (-0.5 ml). The solution was stirred at room temperature for 15 minutes, then directly purified through HPLC (5 to 20 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 20.4 mg, %. Ions found by LCMS: [M/3]+ = 940, [M/4]+ = 705, [M/5]+ = 564, [M/6]+ = 470.
Example 79. Synthesis of Compound 36
Figure imgf000265_0001
The title compound was prepared analogously to Compound 35. Ions found by LCMS: [M/3]+ 950, [M/4]+ = 713, [M/5]+ = 570, [M/6]+ = 475.
Example 80. Synthesis of Compound 37
Figure imgf000265_0002
The title compound was prepared analogously to Compound 35. Ions found by LCMS: [M/3]+ 961 , [M/4]+ = 721 , [M/5]+ = 577, [M/6]+ = 481 .
Example 81. Synthesis of Compound 38
Figure imgf000266_0001
The title compound was prepared analogously to Compound 35. Ions found by LCMS: [M/4]+ 698, [M/5]+ = 558, [M/6]+ = 466, (M+7H)/7= 399.
Example 82. Synthesis of Compound 39
Figure imgf000266_0002
The title compound was prepared analogously to Compound 35 Ions found by LCMS: [M/3]+ 940, [M/4]+ = 705, [M/5]+ = 564, [M/6]+ = 470, [M/7]+ = 403.
Example 83. Synthesis of Compound 40
Figure imgf000266_0003
Compound 40 was prepared analogously as the compound of Example 37 (alternate procedure) where intermediate INT-20 was replaced with INT-25 in the first step of that sequence, lon(s) found by LCMS: (M+3H)/3 = 946.9, (M+4H)/4 = 710.4, (M+5H)/5 = 568.5, (M+6H)/6 = 473.9, (M+7H)/7 = 406.4 Example 84. Synthesis of Compound 41
Figure imgf000267_0001
Step a. Synthesis of Ai-{[(9W-fluoren-9-yl)methoxy]carbonyl}-S-propyl-L-homocysteine
A solution of NaOH (600 mg, 15 mmol) in 1 :1 mixture of water/EtOH (10 ml) was bubbled with nitrogen and heated at 40 °C. It was added with L-homocysteine (675.5 mg, 5 mmol), followed by propyl p-toluenesulfonate (1 .09 g, 5.05 mmol). The mixture was stirred under nitrogen at 40 °C for 1 .5 hours, then to room temperature overnight. EtOH was partially removed by rotary evaporation, and the residue was extracted with DCM (~ 30 ml). THF (10 ml) was added to the aqueous solution, followed by Fmoc- Osu (1 .7 g, 5 mmol). After the mixture was stirred for 1 hour, it was added with an additional amount of Fmoc-Osu (337 mg, 1 mmol). The reaction was continued for one more hour. THF was partially removed by rotary evaporation. The white suspension residue was directly purified through RPLC (150 g, 5 to 58 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 1 .05 g, 50.3 %. Ion found by LCMS: [M+1 ]+ = 400.
Step b. Synthesis of (propyl)-homocys-INT-18
Figure imgf000267_0002
The title compound was prepared analogously to Met- INT- 18. Ion found by LCMS: [(M-Boc)/2]+
= 812. Step c. Synthesis of Compound 41
Figure imgf000268_0001
The title compound was prepared analogously to Compound 35 Ions found by LCMS: [M/3]+ 959, [M/4]+ = 719, [M/5]+ = 575, [M/6]+ = 480.
Example 85. Synthesis of Compound 42
Figure imgf000268_0002
Step a. Synthesis of (cyclopropyl)-ala-INT-18
Figure imgf000268_0003
A mixture of Fmoc-L-cyclopropylalanine (42.2 mg, 0.12 mmol) and HOBT hydrate (25.7 mg, 0.168 mmol) was dissolved in an anhydrous DMF (0.5 ml). EDC-HCI (25.2 mg, 0.132 mmol) was added, and the resulting mixture was stirred for 15 minutes. It was added with INT-18 (168 mg, 0.1 mmol), followed by DIPEA (130 mg, 1 mmol). After stirred for 1 hour, the reaction mixture was quenched with water (0.1 ml) and stirred for 10 more minutes. Piperidine (3 drops) was then added, and the resulting mixture was stirred for 2 hours. It was then directly purified through RPLC (50 g, 20 to 100 % MeOH and water). Yield 145.7 mg, 87 %. Ion found by LCMS: [(M-Boc)/2]+1 = 788. Step b. Synthesis of decaBoc-Compound 42 precursor
A mixture of 2,2'-({4-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4- oxobutanoyl}azanediyl)diacetic acid (15.7 mg, 0.0336 mmol) and the step-b product (1 15 mg, 0.0687 mmol) was dissolved in anhydrous DMF (0.5 ml) and DIPEA (39 mg, 0.3 mmol). The solution was cooled in an ice-water bath and added with a solution of HATU (28 mg, 0.074 mmol) in DMF (0.5 ml) via syringe pump at a rate of 0.5 ml/hr. After the addition, the reaction was stirred for one more hour and directly purified through RPLC (50 g, 20 to 100 % MeOH and water). Yield 73.2 mg, 57.6 %. Ions found by LCMS: [(M-Boc)/3]+ = 1227, [(M-2Boc)+3H]/3 = 1 193, [(M-3Boc)/3]+= 1 160, [(M-5Boc)/3]+= 1093.
Step c. Removal of the Boc groups
DecaBoc-Compound 42 precursor was dissolved in TFA (-0.5 ml). The solution was stirred at room temperature for 15 minutes, then directly purified through HPLC (5 to 20 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 20.4 mg, 20.4 %. Ions found by LCMS: [M/3]+ = 926, [M/4]+ = 695, [M/5]+ = 556, [M/6]+ = 463.
Example 86. Synthesis of Compound 43
Figure imgf000269_0002
Step a. Synthesis of L-Thre-IN -18
Figure imgf000270_0001
To a mixture of Z-thr-OH (30.4 mg, 0.12 mmol) and HOBT hydrate (25.7 mg, 0.168 mmol) was dissolved in anhydrous DMF (0.5 ml) was added EDC-HCI (25.2 mg, 0.132 mmol). The resulting mixture was stirred for 15 minutes, then added with INT-18 (168 mg, 0.1 mmol), followed by DIPEA (130 mg, 1 mmol). After stirring for 1 hour, the reaction mixture was purified through RPLC (50 g, 20 to 1 00 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid (LCMS: [(M- 2B0c)/2]+ = 800). The material was re-dissolved in MeOH (~ 20 ml) and added with Pd/C. The resulting mixture was stirred under hydrogen for 2 hours. Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation and further dried under high vacuum. Yield 120 mg, 72 %. Ion found by LCMS: [(M- Boc)/2]+1 = 783.
Step b. Synthesis of decaBoc-Compound 43 precursor
Figure imgf000270_0002
The title compound was prepared analogously to decaBoc-Compound 42 precursor. Ions found by LCMS: [(M-Boc)/3]+ = 1220, [(M-2Boc)+3H]/3 = 1 1 87, [(M-3Boc)/3]+= 1 153.
Step c. Removal of the Boc group
The reaction was performed analogously to the removal of the Boc group of deca-Boc Compound 42 precursor. Ions found by LCMS: [M/5]+ = 552, [M/6]+ = 460, [M/7]+ = 395. Example 87.
Figure imgf000271_0001
Compound 44 was prepared from INT-33 and INT-36 as described in the procedure for the synthesis of Compound 30. Yield: 15%, 2 steps. LC/MS [M+3H]/3 = 921 .0.
Example 88. Synthesis of Compound 45
Figure imgf000271_0002
Step a. Coupling of INT-18
To the solution of 2,2'-({2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2- oxoethyl}azanediyl)diacetic acid (1 0mg, 0.023 mmol), triethyl amine (0.07ml_, 0.5mmol) and INT-18 (74mg, 0.047mml) in 5ml_ DMF was added HATU(40mg, O.o2mmol) in 1 mL DMF by syringe pump over 1 hour. Then the resulted solution was concentrated and purified by reverse phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoacetic acid as the modifier. Yield of product, 30mg, 37% yield.
Step b. Removal of Boc groups
The product from Step a was treated in 2 ml_ DCM and 2ml_ TFA at room temperature, the solution was stirred 10 min, then concentrated and purified by reverse phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield 15mg, 70% yield, lon(s) found by LCMS: (M+2H)/2 = 1298.8, (M+3H)/3 = 866.2, (M+4H)/4 =649.9, (M+5H)/5=519.1 .
Example 89. Synthesis of Compound 46
Figure imgf000272_0001
Step a. Synthesis of (2¾-2-({[(9W-fluoren-9-yl)methoxy]carbonyl}amino)-5-methylhex-4-enoic acid
c
Figure imgf000272_0002
(S)-2-amino-5-methylhex-4-enoic acid (715.9 mg, 5 mmol) was dissolved in water (3 ml) by gently heating with a heat-gun. After cooled to room temperature, the solution was added with THF (6 ml) and a solution of NaOH (370 mg, 9.25 mmol) in water (3 ml). Fmoc-Osu (1 .7 g, 5.05 mmol) was added, and the resulting mixture was stirred for 1 .5 hours. It was then directly purified through RPLC (150 g, 5 to 60 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 598.6 mg, 32.7%. Ion found by LCMS: [M+1 ]+ = 366. Step b. Synthesis of 5-methyl
Figure imgf000273_0001
The title compound was prepared analogously to (cyclopropyl)-ala-INT-l 8. Ion found by LCMS: [(M-Boc)/2]+ = 795.
Step c. Synthesis of Deca-Boc Compound 46 precursor
Figure imgf000273_0002
A mixture of 2,2'-({4-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4- oxobutanoyl}azanediyl)diacetic acid (18 mg, 0.0385 mmol), 5-methylhex-INT-18 (143.5 mg, 0.085 mmol), DIPEA (42 mg, 0.4 mmol) and anhydrous DMF (1 ml). It was cooled in an ice-water bath and added with a solution of HATU (32.2 mg, 0.0847 mmol) in DMF (0.5 ml) via syringe pump at a rate of 0.5 ml/hr. After the addition, the reaction was stirred for one more hour. It was then purified through RPLC (50 g, 20 to 100 % MeOH and water). Yield 1 03.4 mg, 70.5 %. Ions found by LCMS: [(M-Boc)/3]+1 = 1236, [(M- 2Boc)/3]+= 1203, [(M-3Boc)/3]+= 1 170.
Step d. Removal of the Boc groups
Deca-Boc Compound 46 precursor product (50 mg, 0.013 mmol)) was dissolved in a 1 :1 mixture of TFA:DCM (-0.5 ml). The solution was stirred at room temperature for 15 minutes, then directly purified through HPLC (5 to 20 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 10.4 mg,
20.3%. Ions found by LCMS: [M/3]+ = 935 , [M/4]+ = 702, [M/5]+ = 562, [M/6]+ = 468. Example 90. Synthesis of Compound 47
Figure imgf000274_0001
Deca-Boc Compound 46 precursor (53 mg, 0.138 mmol) was dissolved in MeOH (~ 5 ml). The solution was added with 1 0% Pd/C, and the mixture was stirred under hydrogen overnight. Pd/C was filtered off, and the filtrated was concentrated by rotary evaporation to a white solid (44 mg, 0.01 15 mmol, 83 %). The material was dissolved to a a 1 :1 mixture of TFA:DCM which was precooled in an ice-water bath. The solution was stirred at 0°C to room temperature for 1 hour. It was then directly purified through HPLC. Yield 23.2 mg, 51 .3%. Ions found by LCMS: [M/3]+ = 937 , [M/4]+ = 703, [M/5]+ = 562, [M/6]+ = 469.
Example 91. Synt
Figure imgf000274_0002
Compound 48 was prepared analogously to example 37 (alternate procedure) where 2,2'- {[(benzyloxy)carbonyl]azanediyl}diacetic acid was replaced with N-[(benzyloxy)carbonyl]-D-aspartic acid in the first step of that sequence, lon(s) found by LCMS: (M+3H)/3 = 852.8, (M+4H)/4 = 639.9, (M+5H)/5 = 512.1 , (M+6H)/6 = 427.0 Example 92. Synt
Figure imgf000275_0001
Compound 49 was prepared analogously to example 37 (alternate procedure) where 2,2'- {[(benzyloxy)carbonyl]azanediyl}diacetic acid was replaced with N-[(benzyloxy)carbonyl]-L-aspartic acid in the first step of that sequence, lon(s) found by LCMS: (M+3H)/3 = 852.8, (M+4H)/4 = 639.9, (M+5H)/5 = 512.1 , (M+6H)/6 = 427.0
Example 93. Synthesis of Compound 50
Figure imgf000275_0002
Step a. Synthesis of Gly-IN
Figure imgf000275_0003
To a mixture of Z-glycine (92 mg, 0.44 mmol) and HOBT hydrate (94.2 mg, 0.616 mmol) in anhydrous DMF (1 ml) was added EDC-HCI (92.4 mg, 0.484 mmol). The resulting mixture was stirred for 15 minutes, then added with INT-18 (671 .2 mg, 0.4 mmol), followed by DIPEA (390 mg, 3 mmol). After stirred for 3 hours, the reaction mixture was purified through RPLC (50 g, 20 to 90 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid (LCMS: [M-2Boc)/2]+ = 778). The material was re-dissolved in MeOH (10 ml) and added with Pd/C. The mixture was stirred under hydrogen overnight. Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation and further dried under high vacuum. Yield 586.4 mg, 90.4 %. Ion found by LCMS: [(M-Boc)/2]+1 = 760. Step 2. Removal of the Boc group
The reaction was preformed analogously to the removal of the Boc group of deca-Boc Compound 42 precursor. Ions found by LCMS: [M/3]+ = 891 , [M/4]+ = 668, [M/5]+ = 535 , [M/6]+ = 446.
Example 94. Synthesis of Compound 51
Figure imgf000276_0001
The title compound was prepared analogously to Compound 50. Ions found by LCMS: [M/3]+ = 928, [M/4]+ = 696, [M/5]+ = 557 , [M/6]+ = 464, [M/7]+ = 398.
Example 95. Synthesis of Compound 52
Figure imgf000276_0002
The title compound was prepared analogously to Compound 50. Ions found by LCMS: [M/3]+ = 937, [M/4]+ = 703, [M/5]+ = 563, [M/6]+ = 469, [M/7]+ = 402. Example 96. Synthesis of Compound 53
Figure imgf000277_0001
Step a. Synthesis of undeca-Boc-Compound 53
A solution of intermediate INT-20 (0.148 g, 0.0868 mmol), intermediate INT-26 (0.021 Og, 0.0423), and DIEA(0.045ml_, 0.260mmol), in DMF(3ml_), was treated with a solution of HATU(0.0330g,
0.0868mmol) in DMF(1 mL) dropwise via syringe pump over 1 hr. After 1 .5 hr the mixture was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 1 00% methanol and water, using no modifier. Yield 0.127 g, 77%. lon(s) found by LCMS: did not ionize
Step b. Synthesis of Compound 53
A solution of undeca-Boc-Compound 53 (0.127 g, 0.0327 mmol), dissolved in DCM (1 mL), was treated with TFA (1 mL), while stirring at room temperature. After 5 minutes, the product was
concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using 0.1 % formic acid as the modifier. Yield 0.090 g, 85% yield, lon(s) found by LCMS: (M+4H)/4 = 692.2, (M+5H)/5 = 553.9, (M+6H)/6 = 461 .8, (M+7H)/7 = 395.9
Example 97. Synthesis of Compound 54a and Compound 54b (Structures assigned arbitrarily)
Figure imgf000278_0001
Procedure A
The title compounds were prepared from INT-38a and INT-38b and INT-18 in a manner similar to that described in Example 73. Yield: 5% for Compound 54a and 51 % for Compound 54b. LC/MS
[M+3H]/3 = 937.4 (for both isomers).
Procedure B (Compond 54a)
Figure imgf000279_0001
Step a. Synthesis of diastereomer a and diastereomer b
HATU (3.1 g, 8.1 mmol) in DMF (5 mL) was added, dropwise, to a solution of racemic-transl -
(ferf-butoxycarbonyl)pyrrolidine-3,4-dicarboxylic acid (1 g, 3.9 mmol), H-Norleu-OMe-hydrochloride (1 .5 g, 8.1 mmol), and triethylamine (4.0 g, 39.5 mmol) in DMF (10 mL) over a period of 20 minutes. The mixture was stirred for an additional 20 minutes then applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 1 0% to 95% acetonitrile and water using 0.1 % TFA modifier. The two diastereomers were separated and pooled and lyophilized separately to yield a more polar compound (diastereomer a) and a less polar compound (diastereomer b) by reversed phase HPLC: LC/MS [m+H]+ = 414.2 (loss of 1 Boc group) for either Boc-protected
intermediates. Each diastereomer was stirred in a 1 /1 mixture of DCM/TFA (8 mL) for 20 minutes then concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 0% to 75% acetonitrile and water using no modifier. Yield: 88%, 2 steps. LC/MS [m+H]+ = 414.2.
Note: The configuration of the more polar isomer (diastereomer a) has been arbitrarily assigned the (S,R,R,S) stereochemistry and the configuration of the less polar isomer (diastereomer b) has been arbitrarily assigned the (S,S,S,S) stereochemistry.
Step b. Conjugation of diastereomer a with INT-2
EDC (0.22 g, 1 .16 mmol) was added to a stirring mixture of deprotected diastereomer a (from Step a) (0.40g, 0.97 mmol), 4-[(2-{2-[(6-deoxy-alpha-L mannopyranosyl)oxy]ethoxy}ethyl)amino]-4- oxobutanoic acid (INT-2) (0.41 g, 1 .1 6 mmol), HOBT (0.18g, 1 .1 6 mmol) and triethylamine (0.12 g, 1 .16 mmol) in DMF (4 mL). The mixture was stirred for 2 hours and then applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford 660 mg of the di-ester intermediate; LC/MS [m+H]+ = 747.6. The di-ester was stirred in a 1 /1 /2 mixture of methanol/THF/DI water (6 mL) containing LiOH (46 mg, 1 .93 mmol) at ambient temperature for 20 minutes at which point LC/MS analysis confirmed complete hydrolysis to the di-acid product. The mixture was acidified to pH~5 with glacial acetic acid and the volatile solvents were removed by rotovap. The mixture was applied to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford Rhamnose-706-linker A as a white solid. 71 %, 2 steps. LC/MS [m-H]- = 717.2. LC/MS [m+H]+ = 719.2.
Step c. Synthesis of Compound 54a
The title compound was prepared using the product of Step b and INT-18 using a procedure similar to that described in Example 73 for Compound 30. Yield: 53% for Compound 54a LC/MS
[m/3+H]+ = 937.4.
Procedure C (Compound 54b)
Figure imgf000280_0001
Compound 54b was prepared by an analogous procedure as described for Compound 54a in Procedure B, Steps b and c, using diastereomer b and INT-18. Yield 54%. LC/MS [m/3+H]+ = 937.4. Example 98. Synthesis of Compound 55
Figure imgf000281_0001
Step a. Synthesis of L-Arg-INT-18
Figure imgf000281_0002
A mixture of Z-Arg-OH (68 mg, 0.22 mmol) and INT-1 8 (212 mg, 0.2 mmol) was dissolved in anhydrous DMF (1 ml) and DIPEA (91 mg, 0.7 mmol). The solution was cooled in an ice-water bath and added with a solution of HATU (83.6 mg, 0.22 mmol) in DMF (0.5 ml) via syringe pump at a rate of 0.5 ml/hr. After the addition, the mixture was stirred for 30 more minutes, then directly purified through RPLC (50 g, 10 to 90 % acetonitrile and water). The collected fractions were concentrated to a white solid (LCMS: [(M-2Boc)/2]+ = 626). The material was re-dissolved in MeOH (~ 10 ml), and the solution was added with Pd/C. The resulting mixture was stirred under hydrogen for 4 hours. Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation and further dried in high vacuum to afford the title compound as a white solid (236.8 mg, 97.2 %). Ion found by LCMS: [M/2]+ = 610.
Step b. Synthesis of Octa-Boc Compound 55 precursor
Figure imgf000282_0001
A mixture of INT-40 (40 mg, 0.0347 mmol), L-Arg-INT-18 (90 mg, 0.07 mmol) was dissolved in in anhydrous DMF (0.5 ml) and DIPEA (52 mg, 0.4 mmol). The solution was cooled in an ice-water bath and added with a solution of HATU (26.6 mg, 0.07 mmol) in DMF (0.5 ml) via syringe pump at a rate of 0.5 ml/hr. After the addition, the reaction was stirred for 30 more minutes. It was then purified through RPLC (50 g, 20 to 100 % MeOH and water). Yield 72 mg, 53.7 %. Ions found by LCMS: (M+3H)/3 = 1251 , [(M- Boc)/3]+= 1218, [(M-5Boc)/3]+= 1084.
Step c. Removal of the Boc groups
Compound 55 Boc intermediate (72 mg, 0.0186 mmol) was dissolved in TFA (~ 0.5 ml). The solution was stirred at room temperature for 15 minutes, then directly purified through RPLC (50 g, 5 to 38 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 27 mg, 35.5 %. Ions found by LCMS: [M/3]+ = 984, [M/4]+ = 738, [M/5]+ = 591 , [M/6]+ = 492, [M/7]+ = 422.
Example 99. Synthesis of Compound 56
Figure imgf000282_0002
The title compound was prepared analogously to Compound 55. Ions found by LCMS: [M/3]+ = 956, [M/4]+ = 717, [M/5]+ = 574, [M/6]+ = 478, [M/7]+ = 410. Example 100. Synthesis of Compound 57
Figure imgf000283_0001
The title compound was prepared analogously to Compound 55. Ions found by LCMS: [M/3]+ 965, [M/4]+ = 724, [M/5]+ = 580, [M/6]+ = 483.
Example 101. Synthesis of Compound 58
Figure imgf000283_0002
The title compound was prepared analogously to Compound 55. Ions found by LCMS: [M/3]+ 972, [M/4]+ = 730, [M/5]+ = 584, [M/6]+ = 487.
Example 102. Synthesis of Compound 59
Figure imgf000284_0001
Step a. Preparation of di-tert-butyl N,N-bis[2-(benzyloxy)-2-oxoethyl]-L-aspartate
Benzyl Bromo-acetate (4.6 g, 20 mmol) was added into the solution di-tert-butyl L-aspartate (2.45 g, 10 mmol) and DIPEA (3.6 mL, 30 mmol) in 60 mL DMF, the resultant solution was stirred at room temperature overnight. The reaction solution was concentrated and purified by flash chromatography to provide products. Yield of oil product, LC/MS, 542.3 [M+H]+.
Step b. Preparation of N,N-bis[2-(benzyloxy)-2-oxoethyl]-L-aspartic acid
The above product di-tert-butyl N,N-bis[2-(benzyloxy)-2-oxoethyl]-L-aspartate (1 g, 1 .84 mmol) was treated with 5 mL trifluoroacetic acid and 5 mL DCM, the solution was stirred for a half hour and then concentrated and dried under high vacuum used for next step without purification. Yield of oil product, LC/MS, 430.1 [M+H]+.
Step c. Preparation of dibenzyl 2,2'-{[(2S)-1 ,4-dioxo-1 ,4-bis{[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5- trihydroxy-6-methyloxan-2-yl]oxy}ethoxy)ethyl]amino}butan-2-yl]azanediyl}diacetate
The above crude product N,N-bis[2-(benzyloxy)-2-oxoethyl]-L-aspartic acid (1 .84 mmol) was redissolved in 20 mL DMF, then INT-1 . (1 .25 g, 5 mmol), EDC (1 .0 g, 5 mmol), HOBT (0.75 g, 5 mmol), Hunig's base (1 .4 mL, 10 mmol) were added the solution at room temperature. The resultant solution was stirred overnight. The resultant solution was removed DMF and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil. LC/MS 896.4 [M+H].
Step d. Preparation of 2,2'-{[(2S)-1 ,4-dioxo-1 ,4-bis{[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyloxan-2-yl]oxy}ethoxy)ethyl]amino}butan-2-yl]azanediyl}diacetic acid Dibenzyl 2,2'-{[(2S)-1 ,4-dioxo-1 ,4-bis{[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2- yl]oxy}ethoxy)ethyl]amino}butan-2-yl]azanediyl}diacetate (500 mg, 0.56 mmol) was dissolved into 5 mL MeOH and 5 mL ethyl acetate, then 200 mg of 5% palladium on charcoal was added above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere overnight. The palladium charcoal was removed by filtration after completed the reaction by LCMS. The filtrate was concentrated and used next step without any purification. LC/MS, 71 6.3 [M+H]+ .
Step e. Preparation of per-Boc-Compound 59 (Standard Procedure A)
To the solution of 2,2'-{[(2S)-1 ,4-dioxo-1 ,4-bis{[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyloxan-2-yl]oxy}ethoxy)ethyl]amino}butan-2-yl]azanediyl}diacetic acid (72 mg, 0.1 mmol), triethyl amine (0.07 mL, 0.5 mmol) and INT-20 (340 mg, 0.2 mmol) in 5 mL DMF was added HATU (78 mg, 0.2 mmol). The reaction was stirred for 1 hr and then the resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. Yield of product (per-Boc-Compound 59), 220 mg, 65% yield.
Step f. Preparation of Compound 59 (Standard Procedure B)
Per-Boc-Compound 59 from previous step was treated in 2 mL DCM and 2 mL trifluoroacetic acid at room temperature, the solution was stirred 10 min, then concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using trifluoroacetic acid as the modifier. Yield 0.120 g, 67% yield, lon(s) found by LCMS: [M/3]+1 = 1029.9, [M/4]+1 =772.7, [M/5]+1 =61 8.4, [M/6]+1 =515.5.
Example 103. Binding of Selected Compounds from Examples 23-49, 69-102, and 163-165 to Lipopolysaccharide (LPS)
Binding of compounds to LPS derived from Pseudomonas aeruginosa (ATCC 27316) was determined according to the following procedure. LPS from other strains of bacteria may be substituted to determine binding of compounds with any particular LPS sample.
Figure imgf000286_0001
Step a. Synthesis of methyl (2S)-2-[(N-{(2S)-2-{[(2S)-2-
{[(benzyloxy)carbonyl]amino}octanoyl]amino}-4-[(tert-butoxycarbonyl)amino]butanoyl}-L- threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate
A solution of methyl (2S)-2-[(N-{(2S)-2-amino-4-[(tert-butoxycarbonyl)amino]butanoyl}-L- threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate (0.600 g, 1 .124 mmol),(2S)-2- {[(benzyloxy)carbonyl]amino}octanoic acid (0.330 g, 1 .124 mmol), and DIEA (0.59 mL, 3.37 mmol) in DMF(4 mL), was treated with HATU (0.470 g, 1 .24 mmol), and stirred for 30min. The reaction was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid
chromatograph eluted with 10% to 100% acetonitrile and water, using formic acid (0.1 %) as the modifier. Yield 0.726 g, 79% yield, lon(s) found by LCMS: (M+H)+ = 809.5
Step b. Synthesis of methyl (2S)-2-[(N-{(2S)-2-{[(2S)-2-aminooctanoyl]amino}-4-[(tert- butoxycarbonyl)amino]butanoyl}-L-threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate
A mixture of methyl (2S)-2-[(N-{(2S)-2-{[(2S)-2-{[(benzyloxy)carbonyl]amino}octanoyl]amino}-4- [(tert-butoxycarbonyl)amino]butanoyl}-L-threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate (0.300 g, 0.371 mmol), 5%Pd/C(0.150g), and methanol(4 mL), were charged with hydrogen from a balloon. After 2hr the product was filtered through Celite, concentrated, and used in the next step without further purification. Mass recovery 0.250 g, quantitative, lon(s) found by LCMS: (M+H)+ = 675.4
Step c. Synthesis of methyl (2S)-4-[(tert-butoxycarbonyl)amino]-2-({N-[(2S)-4-[(tert- butoxycarbonyl)amino]-2-{[(2S)-2-{[5-(dimethylamino)naphthalene-1- sulfonyl]amino}octanoyl]amino}butanoyl]-L-threonyl}amino)butanoate
A solution of methyl (2S)-2-[(N-{(2S)-2-{[(2S)-2-aminooctanoyl]amino}-4-[(tert- butoxycarbonyl)amino]butanoyl}-L-threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate (0.60 g), and dansyl chloride (0.1 10 g, 0.408 mmol), in NMP (1 .5 mL) was treated with N-methylmorpholine (0.081 mL, 0.741 mmol). After 30 minutes the product was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 1 00% acetonitrile and water, using formic acid (0.1 %) as the modifier. Yield 0.336 g, 85% yield, lon(s) found by LCMS: (M+H)+ = 908.5
Step d. Synthesis of (2S)-4-[(tert-butoxycarbonyl)amino]-2-({N-[(2S)-4-[(tert- butoxycarbonyl)amino]-2-{[(2S)-2-{[5-(dimethylamino)naphthalene-1- sulfonyl]amino}octanoyl]amino}butanoyl]-L-threonyl}amino)butanoic acid
A solution of methyl (2S)-4-[(tert-butoxycarbonyl)amino]-2-({N-[(2S)-4-[(tert- butoxycarbonyl)amino]-2-{[(2S)-2-{[5-(dimethylamino)naphthalene-1 - sulfonyl]amino}octanoyl]amino}butanoyl]-L-threonyl}amino)butanoate (0.285 g, 0.314 mmol), in methanol (10 mL), was treated with lithium hydroxide (0.050 g, 2.1 mmol) in water(1 mL), while stirring at room temperature. After 30 minutes the mixture was titrated with concentrated HCI(aq.) until slightly acidic by pH paper, concentrated, and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using no modifier. Yield 0.267 g, 95% yield, lon(s) found by LCMS: LCMS: (M+H)+ = 894.5
Step e. Synthesis of penta-Boc-(Compound 60)
A solution of (2S)-4-[(tert-butoxycarbonyl)amino]-2-({N-[(2S)-4-[(tert-butoxycarbonyl)amino]-2-
{[(2S)-2-{[5-(dimethylamino)naphthalene-1 -sulfonyl]amino}octanoyl]amino}butanoyl]-L- threonyl}amino)butanoic acid(0.267 g, 0.299 mmol), INT-1 1 (0.349 g, 0.328 mmol), and DIEA (0.172 mL, 0.985 mmol) in DMF (1 mL), was treated with HATU (0.129 g, 0.328 mmol), while stirring at room temperature. The product was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.267 g, 46% yield, lon(s) found by LCMS: (M+2H)/2 = 969.5, [(M-1 Boc)+2H]/2 = 919.5, [(M-2Boc)+2H]/2 = 869.5
Figure imgf000287_0001
Penta-Boc-(Compound 60) (0.267 mg, 0.138 mmol) was treated with TFA (3 mL) for 15 minutes, then concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco
CombiFlash liquid chromatograph eluted with 0% to 1 00% acetonitrile and water, using formic acid as the modifier. The appropriate fractions were collected, pooled and concentrated to give after drying and lyophilization, Compound 60. Yield 0.053 g, 27% yield, lon(s) found by LCMS: [M+2H]/2 = 719.4 Part 2. Binding assay
The LPS binding of compounds was measured by fluorescence of a compound bound to LPS using a Tecan Infinite Pro fluorescence spectrophotometer set at an excitation wavelength of 340 nm and emission wavelength of 485 nm. Displacement assays were performed by recording the fluorescence after the addition of inhibitors at 250 μΜ final concentration. Assays were performed in 96-well microtiter plates using 4 μg of P. aeruginosa LPS (ATCC 27316) and 6 μΜ Compound 60 in 10 mM HEPES buffer (pH 7.5). Percent displacement is expressed relative to a no inhibitor control and corrected for intrinsic inhibitor fluorescence.
Part 3. Results
Table 3. % Displacement of Fluorescent Probe Compound 60 from P. aeruginosa LPS
Figure imgf000288_0001
Compound % Displacement Compound % Displacement
Compound 29 97.3 Compound 58 49.2
Compound 30 79.7 Compound 59 94.2
Compound 31 96 Compound 75 90.1
Compound 32 97 Compound 76 93.1
Compound 33 98.8 Compound 77 87.5
Compound 42 92.2 Compound 78 86.9
Compound 43 87.9 Compound 79 95.7
Compound 44 48.5 Compound 80 97.6
Compound 45 58.6
Example 104. Antibacterial Activity of Selected Compounds from Examples 23-49, 69-101 , and 151-209
Part 1. Methods
Generation of E. co/ BW25113 + pUC18-mcr-1.
An mcr- 1 expression plasmid was constructed (GenScript; Piscataway, NJ) using the pUC18 high copy number plasmid (Yanisch-Perron, et al., 1985). A 1649-bp fragment was synthesized containing, in order from 5' to 3': EcoR1 restriction site (5'-GAATTC-3'), a ribosomal binding site (5'-AGGAGG-3'), a 5- bp native start codon upstream sequence (5'-TTCTC-3') from the mcr- /-bearing plasmid pHNSHP45
(Liu, YY, et al., 2015; GenBank Accession # KP347127), the 1626-bp mcr- 1 open reading frame with stop codon ("orf00073" of GenBank Accession # KP347127), and finally an Xba\ restriction site sequence (5'- TCTAGA-3'). Restriction digestion and subsequent ligation into the multiple cloning site of the pUC18 backbone resulted in directional integration of the mcr- 1 gene. The pUC1 8-mcr-1 plasmid was transformed into chemically-competent E. coli BW251 13 (Coli Genetic Stock Center #7636) as described previously (Chung, et al., 1989), recovered for 1 h shaking at 37°C in Super Optimal broth with Catabolite repression (SOC) media and then aliquots were plated on Luria-Bertani (LB) media containing 100 μg/mL of carbenicillin. Putative transformant colonies were then verified in MIC assays. Strains and culture conditions.
Bacterial strains were stored as glycerol stocks at -80 °C prior to culturing on cation-adjusted
Mueller-Hinton agar (MHA; BD cat. no. 21 1438) or in cation-adjusted Mueller-Hinton broth (MHB; BD cat. no.212322) cultured at 37°C. Antibacterial activity was assessed against a strain panel consisting of
Escherichia coli BW251 13 wild-type (Coli Genetic Stock Center #7636), BW251 13 + pUC1 8-mcr-1 , BW251 13 COLr 4X-12 mutant (an uncharacterized, spontaneous mutant selected by plating BW251 13 on
MHA containing 1 μg/mL COL, or 4X the MIC value), and ATCC BAA-2469 (ATCC - American Type
Culture Collection), Acinetobacter baumannii ATCC A5075 (ATCC), Pseudomonas aeruginosa PA01
(ATCC BAA-47, ATCC), and Klebsiella pneumoniae ATCC 10031 (ATCC). MIC (Minimum Inhibitory Concentration) assays.
MIC assays were performed according to CLSI broth microdilution guidelines (M07-A9, M100- S23) with the exception of using a 1 00 μΙ_ assay volume and preparing stock compounds at 50X final concentration. Briefly, stock solutions of all antibacterial agents were prepared fresh in appropriate solvents (i.e. DMSO, Dl water, ethanol, etc.). Stock concentrations were made at 50X the highest final assay concentration and serially diluted 2-fold, 12 times in a 96-well PCR plate (VWR cat. no. 83007- 374). Bacterial cell suspensions generated from MHA plate cultures were prepared in 0.85% saline and adjusted to -0.1 Οϋβοο nm. Next, cell suspensions were diluted 1 :200 in MHB (BD cat. no. 212322) to a concentration of ~5 x 105 CFU (Colony Forming Units)/ml_. Ninety-eight microliters of each cell suspension in MHB were added to test wells in a 96-well assay plate (Costar cat. no. 3370). A Beckman Multimek 96 liquid handling robot was used to dispense 2 μΙ_ of each 50X stock compound into the plate containing 98 μΙ_ of each strain in MHB (2% final solvent concentration). Plates were shaken then incubated at 37°C overnight (16-20 h). MIC values were read visually at 100% growth inhibition for all compounds.
Part 2. Results
Table 4. MICs ^g/mL)
Figure imgf000290_0001
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
* ND: not determined
Example 105. Synergistic Activity of Selected Compounds from Examples 23-35, 38-49, 69-101 , and 151-172
Fixed concentration synergy MIC assays. The synergistic potential of test compounds was investigated through assessment of MIC values in the presence of a fixed concentration of erythromycin (ERY; Sigma cat. no. E5389). The test compound was run as a dilution series as performed under the CLSI-based conditions described in Example 51 for single agents using E. coli BW251 13 (or E. coli BW251 13 + pUC18-mcr-1 ), however each well was stamped a subsequent time with Beckman Multimek 96 liquid handling robot to aliquot a 2 μΙ_ volume of 0.05 mg/mL ERY (solubilized in 50% ethanol) across the 12-point, 2-fold dilution series to give a resulting concentration of ~1 μg/mL ERY. MIC values for the test compounds alone were compared with those containing the fixed concentration of ERY to determine if the polymyxin-based compound increased the susceptibility of the strain to ERY. Table 5. MICs in combination/absence of 1 μg/mL erythromycin and resultant fold-synergy
Figure imgf000296_0001
E. coli + 1 £. co/ + 1
Fold Fold
Compound E. coli Mg/mL E. coli Mg/mL
Synergy Synergy erythromycin erythromycin
BW25113
BW25113 +
BW25113 BW25113 + pUC18- pUC18-mcr-1 mcr-1
Compound 22 16 1 16 256 4 64
Compound 23 128 2 64 256 128 2
Compound 24 2 1 2 8 2 4
Compound 25 2 2 1 4 4 1
Compound 26 4 2 2 16 16 1
Compound 27 1 1 1 1 1 1
Compound 28 2 2 1 2 2 1
Compound 29 4 4 1 4 4 1
Compound 30 1 1 1 128 2 64
Compound 31 4 2 2 4 2 2
Compound 32 2 2 1 2 2 1
Compound 33 2 1 2 2 2 1
Compound 34 2 2 1 256 256 1
Compound 35 1 1 1 4 2 2
Compound 36 0.5 0.5 1 32 16 2
Compound 37 1 1 1 32 32 1
Compound 38 1 1 1 16 16 1
Compound 39 2 2 1 2 2 1
Compound 40 4 4 1 8 4 2
Compound 41 4 2 2 4 2 2
Compound 42 2 2 1 4 2 2
Compound 43 1 2 0.5 64 64 1
Compound 44 2 2 1 4 4 1
Compound 45 4 2 2 8 4 2
Compound 46 2 2 1 2 2 1
Compound 47 2 2 1 2 2 1 £. coli + 1 E. coli + 1
Fold Fold
Compound E. coli Mg/mL £. co/ Mg/mL
Synergy Synergy erythromycin erythromycin
BW25113
BW25113 +
BW25113 BW25113 + pUC18- pUC18-mcr-1 mcr-1
Compound 48 1 1 1 16 64 0.25
Compound 49 2 1 2 16 16 1
Compound 50 2 1 2 64 64 1
Compound 51 2 1 2 8 2 4
Compound 52 2 2 2 2
Compound 53 2 2 1 2 2 1
Compound 54a 1 1 1 1 2 0.5
Compound 54b 2 1 2 2 2 1
Compound 55 2 2 1 4 4 1
Compound 56 2 2 1 1 2 0.5
Compound 57 2 2 1 2 2 1
Compound 58 4 4 1 8 8 1
Compound 59 2 2 1 2 2 1
Compound 61 1 1 1 64 64 1
Compound 62 1 1 1 32 32 1
Compound 63 2 1 2 128 64 2
Compound 64 4 2 2 4 4 1
Compound 65 2 4 0.5 2 4 0.5
Compound 66 2 1 2 2 2 1
Compound 67 2 2 1 4 2 2
Compound 68 2 2 1 2 2 1
Compound 69 1 1 1 2 2 1
Compound 70 2 2 1 2 2 1
Compound 71 1 1 1 64 64 1
Compound 72 2 2 1 2 2 1
Compound 73 1 1 1 2 2 1
Compound 74 2 2 1 2 4 0.5
Compound 75 4 2 2 16 4 4
Compound 76 4 4 1 8 4 2 E. coli + 1 £. co// + 1
Fold Fold
Compound E. coli Mg/mL £. co/ Mg/mL
Synergy Synergy erythromycin erythromycin
BW25113
BW25113 +
BW25113 BW25113 + pUC18- pUC18-mcr-1
mcr-1
Compound 77 4 4 1 32 4 8
Compound 78 4 4 1 8 4 2
Compound 79 4 2 2 2 2 1
Compound 80 4 4 1 4 4 1
Compound 81 2 2 1 2 2 1
Compound 82 2 2 1 2 2 1
Compound 83 4 4 1 4 4 1
Compound 84 1 1 1 1 1 1
Compound 85 2 2 1 2 2 1
Compound 86 2 1 2 2 1 2
Compound 87 1 1 1 1 1 1
Example 106. Synergistic Activity of Compound 1
Checkerboard synergy assays.
Checkerboard synergy assays were performed as previously described (Eliopoulos, G.M. and R.C. Moellering, 1996; Moody, J., 2004). In master stock plates the stocks of two compounds
("Compound A" - test compound, and "Compound B" - a known antibacterial agent) for synergistic evaluation were prepared. A liquid stock of Compound A was prepared fresh in Dl water to a concentration 50X the desired final synergy assay concentration. Into wells A1 through H1 (vertical, Y- axis column on left edge of plate) of a 96-well PCR plate (VWR cat. no. 83007-374) Compound A was aliquoted in 100 μΙ_ volumes. To all other wells on the plate, with the exception of A12 through H12, 50 μΙ_ of Dl water was added. A multichannel pipette was then used to transfer 50 μΙ_ from column A1 -H1 to column A2-H2. Following mixing, tips were discarded and the process was repeated across the plate to create a series of 1 1 , serial two-fold dilutions spanning columns 1 through 1 1 .
A stock of Compound B was prepared in a similar fashion in the appropriate solvent (DMSO, Dl water, ethanol, etc.) at 50X the desired final synergy assay concentration. In a separate 96-well PCR plate, 100 μΙ_ aliquots of Compound B were added to wells in the top row of the plate spanning A1 through A12. In all other wells, excluding the bottom row (H1 -H12), 50 μΙ_ of the corresponding Compound B solvent were added. In a similar fashion to Compound A, Compound B was serially diluted, two-fold down the plate using a multichannel pipette, resulting in 7 rows comprised of 12 wells with 50 μΙ_ diluted Compound B. Checkerboard synergy assays were performed using the same assay setup as the standard MIC assays previously described previously herein (i.e. inoculum, media, volume, 96-well assay plates, incubation time, incubation temperature, 100% inhibition assay endpoint, etc.). However, rather than a single addition of 2 μΙ_ of 50X stock compound solution to the assay plate containing MHB pre-inoculated with bacterial cells using the Multimek 96 liquid handling robot, two sequential additions were made (one from the Compound A 50X stock plate and one from the Compound B 50X stock plate). The resulting checkerboard plates contained a 7 x 1 1 -well grid (A1 -G1 x A1 -A1 1 ) of synergy wells containing varying proportions of Compounds A and B, an 1 1 -point dilution series of Compound A alone (wells H1 -H1 1 , assay concentration range "I X to 1 /1024X), a 7-point dilution series of Compound B alone (wells A12- G12, assay concentration range "I X to 1 /64X) and a positive growth control well containing no drug (well H12) (FIG. 1 ).
Following overnight incubation, checkerboard synergy plates were read visually at 100% growth inhibition. MIC values for Compound A and B were recorded from their respective single-drug dilution series wells and an optimal synergy well (or wells) was identified as that for which growth inhibition was achieved using a combination of the lowest possible concentrations of both drugs. The fraction inhibitory concentration (FIC) was calculated for each compound by dividing the combination MIC value in the optimal synergy well by the MIC value for the compound alone. The summation of the FIC values for Compounds A and B was then used to calculate the fractional inhibitory concentration index (FIG) and synergy was defined as an FICI value of <0.50.
FIC Compound A = MIC of Compound A in combination/MIC of Compound A alone
FIC Compound B = MIC of Compound B in combination/MIC of compound B alone
Figure imgf000300_0001
Table 6. MICs of Compound 1 (CMP 1 ) and various antibiotics against Gram-negative bacterial strains and FICI values of Compound 1 in combination with those antibiotics
Figure imgf000300_0002
Example 107. Inoculation of mice or rabbits with OVA-Rha-linked vaccine, isolation of serum and purification of anti-Rha antibodies
Part 1. Preparation of OVA-Rha-linked vaccine (OVA-Rha)
Step a. Synthesis of N-(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)-4-[(2,5- dioxopyrrolidin-1-yl)oxy]-4-oxobutanamide
(0-(N-Succinimidyl)-1 ,1 ,3,3-tetramethyl uronium tetrafluoroborate-' STU") (206 mg, 0.68 mmol) was added to a stirring mixture of 4-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4- oxobutanoic acid (INT-2) (200 mg, 0.57 mmol) and triethylamine (690 mg, 0.68 mmol) in DMF (4 mL) and the reaction was stirred for 4 hours at room temperature. The solvent was removed on a rotary evaporator then dried under high vacuum for 12 hours. The crude product mixture was used for protein conjugation without further treatment. (~2 mg of the crude product was treated with excess benzylamine and analyzed by LC/MS to confirm formation of the activated ester). Step b. Conjugation to ovalbumin
A solution of N-(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)-4-[(2,5-dioxopyrrolidin- 1 -yl)oxy]-4-oxobutanamide (30 mg/mL in 2x PBS pH 7.4) was added to the same volume of ovalbumin (10 mg/mL in 2x PBS pH 7.4) and was gently stirred at 25°C for 1 h, followed by 5 h incubation at 4°C. The solution was concentrated and buffer exchanged into 1 x PBS pH 7.4 using Amicon Centrifugal Filter Devices (10,000 MWCO). Extensive buffer washes removed unconjugated linker from the protein solution. Solution purity and a molecular weight shift were confirmed by SDS-PAGE and size exclusion chromatography (Superdex 75). The solution was determined to be endotoxin free using the Endosafe Assay (Charles River). The glycoprotein solution was quantitated using a NanoDrop 2000
Spectrophotometer and stored at 4°C for immunizations.
Part 2. Vaccination of animals for production of anti-Rha serum
Mice were vaccinated subcutaneously in a similar manner to that described in ACS Chem Biol. 201 1 Feb 18;6(2):1 85-91 . Female ICR mice (17-20 g each) were immunized q2wksx4 with 30 μg of OVA-Rha as follows: Day 0: each mouse received a subcutaneous injection of 150 μΙ_ of OVA-Rha + Complete Freund's Adjuvant (CFA, Sigma F5881 ); Day 14: 1 50 μΙ_ of OVA-Rha + Incomplete Freund's Adjuvant (IFA Sigma F5506); Day 28: 150 μΙ_ OVA-Rha + saline. An additional immunization on Day 42 using 150 μΙ_ OVA-Rha + saline can be used to further increase titers of anti-Rha antibody. Serum was isolated subsequently from mouse blood according to standard protocol. Serum production may be accomplished at least 7 to 21 days after the last vaccination. Part 3. Vaccination of rabbits
Sixty day old New Zealand rabbits were vaccinated subcutaneously with OVA-Rha ± CFA or IFA. Rabbits were held at indicated time points and serum wasw prepared using a standard procedure.
Week 0: 1 st immunization with 0.25 mg of OVA-Rha in Complete Freund's Adjuvant
Week 2: 2nd immunization with 0.2 mg of OVA-Rha in Incomplete Freund's Adjuvant (IFA)
Week 4: 3rd immunization with 0.2 mg of OVA-Rha in IFA
Week 5: 1 st production bleed - -20-25 mL of serum
Week 6: 4th immunization with 0.2 mg of OVA-Rha in IFA
Week 7: 2nd Production bleed - -20-25 mL of serum
Week 8: 3rd Production bleed - -20-25 mL of serum
Part 4. Purification of Rha-specific antibodies from serum
Step a. Preparation of the anti-rhamnose antibody affinity column
6-Aminohexyl-Agarose (5 mL, 4% crosslinked beaded agarose, -5 μιηοΙ per mL) suspended in saline was centrifuged (3500 rpm for 5 min), then decanted to remove water. The resulting solid was suspended in DMF (4 x 15 mL), centrifuged (3500 rpm for 5 min) and decanted. The agarose was resuspended in DMF (1 mL), and treated with INT-2 (0.140 g, 0.40 mmol), followed by DIEA (0.21 mL, 1 .20 mmol), and HATU (0.152 g, 0.40 mmol). The mixture was rotated for 3 hr at room temperature, then transferred to a plastic column and washed with phosphate buffered saline (5 x 15 mL, pH 7.4). The solid supported a-L-rhamnose was stored in phosphate buffered saline (pH 7.4) with 20% ethanol, at room temperature.
Step b. Purification of anti-rhamnose antibody from rabbit serum
Serum from immunized rabbits was diluted 2-fold in 2X PBS pH 7.4. The solution was passaged over the solid supported a-L-rhamnose column (from Step a) by gravity flow followed by a wash step using 10 CV of 1 X PBS pH 7.4. Protein was eluted using 20 mM Glycine Buffer pH 2.0 and immediately adjusted to a neutral pH by the addition of 1 M Tris pH 9.0. The eluate was buffer exchanged with 1 X PBS pH 7.4 and concentrated in Amicon centrifugal concentrators (10K MWCO). The presence of IgG was determined by reducing and non-reducing SDS-PAGE and quantitated using a NanoDrop 2000 Spectrophotometer. The resultant purified antibodies can be used in place of serum adjusting for relative antibody titers.
Example 108. Rha binding assay - Plate ELISA
Part 1. Binding of anti-rhamnose (anti-Rha) antibodies to selected compounds from Examples 23- 49 and 69-101
Test compound (2 μg) was immobilized to 96 well microtiter plates using the TaKaDa peptide coating kit (Clontech). Binding was detected with 3-fold dilutions of polyclonal rabbit anti-rhamnose serum (from Example 54) or na'ive rabbit serum (as a control), starting with a 1 :450 dilution, and ending with a 1 :984,150 dilution. Bound rabbit antibody was detected using a 1 :5,000 dilution of anti-rabbit IgG HRP antibody and TMB development solution. After stopping the reaction with 1 M H2SO4, the absorbance at 450 nm (A450 nm) was read using a Magellan plate reader and plotted against serum dilution factor using GraphPad Prism and Microsoft Excel for data analysis. A BCso was calculated and is the reciprocal of the serum dilution at 50% maximum absorbance at 450 nm. Part 2. Results
Table 7. BCso values for selected Compounds from Examples 23-49 and 69-101
Figure imgf000303_0001
* Reciprocal of serum dilution giving 50% signal
Example 109. Rha binding assay - Whole cell ELISA
Part 1. Binding of anti-rhamnose antibodies to selected E. co//'-bound compounds 23-49 and 69- 101.
96 Well microtiter plates coated with E. coli were incubated with a 5-fold dilution series of test compound, starting with 4,000 ng and ending with 0.0512 ng. Binding was detected with a fixed
1 :100,000 dilution of polyclonal rabbit anti-rhamnose serum (from Example 54) or naive rabbit serum (as a control). Bound rabbit antibody was detected using a 1 :5,000 dilution of anti-rabbit IgG HRP antibody and TMB development solution. After stopping the reaction with 1 M H2SO4, the absorbance at 450 nm (A450 nm) was read using a Magellan plate reader and plotted against the amount of compound in ng using GraphPad Prism and Microsoft Excel for data analysis. Part 2. Results
Table 8 below shows the ECso values for selected Compounds from Examples 23-49 and 69-101 . FIG. 15 shows the absorbance values at 450 nm for E. coli incubated with Compound 14 and a control compound (Compound 14 without the monosaccharide portion).
Table 8
Figure imgf000304_0001
* Concentration of compound that gave 50% of total signal (ng)
Example 110. LPS Neutralization by Selected Compounds from Examples 23-49 and 69-101 Part a. Method
The murine macrophage cell line RAW 264.7 (ATCC, TIB-71 ) was grown to about 70 % confluency in DMEM + 10 % FBS in T75 cm2 flasks at 37 °C / 5 % CO2 for 2-3 days. Cells were washed once with 10 mL DPBS and scraped into 5 mL of media. After centrifugation at 800 rpm for 5 min, the cells were counted with a hemocytometer after 1 :10 dilution in media with 0.04 % trypan blue. Cells were adjusted to 2E6/ mL in DMEM + 1 0 % FBS.
RAW 264.7 cells were added to a 96-well plate at 2 x 105 cells per well in 100 μΙ_ in DMEM + 10 % FBS and incubated for 24 h at 37°C 1 5% CO2. Medium was aspirated and cells washed once with 200 μΙ_ DPBS before addition of test reagents. Test reagents and media only control were prepared in RPMI without phenol red + 5% FBS.
Table 9. Test article preparation
Test Compounds Polymyxin B LPS (positive control)
5 \iqlmL· 5 pQlmL 10 \iqlmL·
7.5 μΙ_ 15 μΙ_ Polymyxin B 1 μΙ_ LPS
(2 mg/mL ) (1 mg/mL ) (1 mg/mL )
1492.5 μΐ media 1485 μΐ media 999 μΐ media
1 .5 mL 1 .5 mL 1 mL
100 it of reagents were added to wells at 0.5 and 5 μς/ mL final concentrations. The cells were incubated with reagents for 1 h at 37°C + 5% CO2. After 15 min, 100 μί medium containing LPS at 0.001 , 0.01 , 0.1 , 1 and 10 μς/ mL was added to appropriate wells and the plates were incubated for 24 h at 37°C + 5% CO2. Controls received medium only. After 24h, 150 μΙ of each supernatant was transferred to a new 96 well FLAT bottom plate. 50 μί of the transferred supernatant was used for a Griess assay (measurement of nitric oxide (NO) production). The remainder was stored at 4 °C for ELISA. A Griess assay was performed according to manufacturer's (Promega) instructions. Absorbance at 540 nm was read and NO production in μΜ was calculated by linear regression analysis of the NO standard curve using Graphpad Prism 6 software.
Part b. Results: Inhibition of Nitric Oxide (NO) production by Selected Compounds from
Examples 23-49 and 69-101 in RAW cells stimulated with LPS
NO production in response to LPS (endotoxin) is a signal of macrophage activation which may lead to sepsis in an infected host. Inhibition of NO production through LPS binding and neutralization is demonstrated by Compound 14 of the present disclosure (FIG. 2) at clinically relevant concentrations of LPS that have been correlated with sepsis in humans which is less than 1 ng/mL in the plasma (see, e.g., Opal et al., J. Infect Dis. 180:1584-9, 1999).
Example 111. In vivo activity in a mouse model of bacterial sepsis for selected compounds from Examples 23-49 and 69-101
Mouse strain
Experiments utilized female C57BI/6 mice (18 - 22 grams) from Charles River Laboratories
(Wilmington, Massachusetts) and were allowed to acclimate for 5 days prior to start of study. Animals were housed 6 per cage with access to food and water ad libitum.
Inoculum preparation and infection
E. coli ATCC 25922 was obtained from the American Type Culture Collection (Manassas, Va) and was prepared for infection from an overnight plate culture (Trypicase Soy agar media). A portion of the plate was re-suspended in sterile saline and adjusted to an OD of 0.1 at 625 nm. The adjusted bacterial suspension was further diluted in 5% hog gastric mucin to target an infecting inoculum of 1 .0x103 CFU/mouse. Plate counts of the inoculum were performed to confirm inoculum concentration. To initiate the infection, mice were injected with 0.5 mL of the suspension intraperitoneally (IP).
Treatment/Efficacy
When indicated, groups of mice received an IV injection of rabbit anti-serum or purified antibody (+Ab) 24 hours prior to infection at a dose of 1 :4000. Mice that received no serum or antibody are indicated by (-Ab). Beginning at one hour post infection, mice were dosed with either vehicle or test article. Vehicle/diluent was sterile 0.9% saline for all test articles. All animals were dosed IP at 10 mL /kg. Mice were euthanized at 16 hours post infection. Kidneys were aseptically removed, weighed, homogenized, serially diluted and plated. The CFUs were enumerated after overnight incubation. The average and standard deviations for each group were determined via accepted methodology. Results
Table 10. Survival and CFU reduction in kidneys of mice (n=6 per group) infected with E. co//' and treated with colistin or Compound
Figure imgf000306_0001
Table 1 1 . Survival and CFU reduction in kidneys of mice (n=6 or 10 per group) infected with E. co//' and treated with meropenem or Compound
Figure imgf000306_0002
Table 12. Survival and CFU reduction in kidneys of mice (n=6 per group) infected with E. co//' and treated with meropenem or Compound
Figure imgf000306_0003
Change in logi0
Dose (mg/kg) Number of logioCFU/
Group ID St. Dev. CFU/g kidney versus
(IP, QD) survivors g kidney
16 hour controls
5.0 (-Ab) 6 3.30 0.69 -3.78
Compound 1 1
5.0 (+Ab) 6 2.1 1 0.00 -4.97
Additional efficacy was seen in the presence of serum with Compound 1 , Compound 1 1 , and Compound 14 indicating enhanced efficacy due to conjugation of antigen and recognition by anti-Rha antibody. Example 112. Antibacterial Activity of Compounds 14 and 54a
Part 1. Methods
Generation of E. co/ BW25113 + pUC18-mcr-2.
An mcr-2 expression plasmid was constructed (GenScript; Piscataway, NJ) using the pUC18 high copy number plasmid (Yanisch-Perron, et al., 1985). A 1640-bp fragment was synthesized containing, in order from 5' to 3': EcoR1 restriction site (5'-GAATTC-3'), a ribosomal binding site (5'-AGGAGG-3'), a 5- bp native start codon upstream sequence (5'-TTTCT-3') from the mcr-2-bearing plasmid pKP37-BE (Xavier et al., Euro Surveill A (27), 201 6; GenBank Accession # LT598652), the 1 617-bp mcr-2 open reading frame with stop codon (locus tag # A7J1 1_03753), and finally an Xba\ restriction site sequence (5'-TCTAGA-3'). Restriction digestion and subsequent ligation into the multiple cloning site of the pUC18 backbone resulted in directional integration of the mcr-2 gene. The pUC1 8-mcr-2 plasmid was transformed into chemically-competent E. coli BW251 13 (Coli Genetic Stock Center #7636) as described previously (Chung, et al., 1989), recovered for 1 h shaking at 37°C in Super Optimal broth with Catabolite repression (SOC) media and then aliquots were plated on Luria-Bertani (LB) media containing 100 μg/mL of carbenicillin. Putative transformant colonies were then verified in MIC assays. Susceptibility of Compounds 14 and 54a, COL (colistin), and PMB (polymyxin B) were then assessed vs. WT E. coli BW251 13 and the pUC18-mcr-2 transformant strain.
Strains and culture conditions.
Bacterial strains were stored as glycerol stocks at -80 °C prior to culturing at 37°C on cation- adjusted Mueller-Hinton agar (MHA; BD cat. no. 21 1438) or in cation-adjusted Mueller-Hinton broth
(MHB; BD cat. no. 212322). Antibacterial activity was assessed against Escherichia coli BW251 13 wild- type (Coli Genetic Stock Center #7636) and BW251 13 + pUC18-mcr-2. Additionally MIC90 assays were run on panels of COL-S (colistin-susceptible) clinical isolates collected from US sites between 2012 and 2016 for Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa and Acinetobacter baumannii (n=20/species) as well as a panel of COL-R (colistin-resistant) isolates (n=25, composed of a mixture of K. pneumoniae, P. aeruginosa and A. baumannii). Finally, E. coli ATCC 25922 WT and a panel of spontaneous mutants selected with Compound 14 and COL were assessed for changes in susceptibility to selecting drugs and a panel of compounds to assess cross-resistance.
MIC (Minimum Inhibitory Concentration) assays.
MIC assays were performed according to CLSI broth microdilution guidelines (M07-A9, M100-
S23) with the exception of using a 1 00 μί assay volume and preparing stock compounds at 50X final concentration. Briefly, stock solutions of all antibacterial agents were prepared fresh in appropriate solvents (i.e. DMSO, Dl water, ethanol, etc.). Stock concentrations were made at 50X the highest final assay concentration and serially diluted 2-fold, 12 times in a 96-well PCR plate (VWR cat. no. 83007- 374). Bacterial cell suspensions generated from MHA plate cultures were prepared in 0.85% saline and adjusted to -0.1 Οϋβοο nm. Next, cell suspensions were diluted 1 :200 in MHB (BD cat. no. 212322) to a concentration of ~5 x 105 CFU (Colony-Forming Units)/mL. Ninety-eight microliters of each cell suspension in MHB were added to test wells in a 96-well assay plate (Costar cat. no. 3370). A Beckman Multimek 96 liquid handling robot was used to dispense 2 μί of each 50X stock compound into the plate containing 98 μΙ_ of each strain in MHB (2% final solvent concentration). Plates were shaken then incubated at 37°C overnight (16-20 h). MIC values were read visually at 100% growth inhibition for all compounds.
Part 2. Results
Table 13. Broth microdilution MICs ^g/mL) for Compounds 14 and 54a, PMB, and COL against Mcr-2- expressing E. coli
Figure imgf000308_0001
Tables 14A-14D. MICso and MIC90 analysis of Compounds 14 (CMP 14) and 54a (CMP 54a), PMB, COL, TIG (tigecycline), and MERO (meropenem) against COL-S E. coli (Table 14A), K. pneumoniae (Table 14B), P. aeruginosa (Table 14C), and A. baumannii (Table 14D) clinical isolate panels
Table 14A (COL-S E. coli)
Figure imgf000308_0002
MIC (Mg/mL)
# Strain CMP 14 CMP 54a PMB COL TIG MERO
3 MMX 8415 2 1 0.5 0.25 0.25 0.015
4 MMX 8416 2 2 0.5 0.5 0.25 0.015
5 MMX 8419 2 2 0.5 0.25 0.5 0.03
6 MMX 8421 2 1 0.5 0.5 0.25 0.015
7 MMX 8433 2 2 0.5 0.5 0.5 0.03
8 MMX 8880 2 1 0.5 0.25 0.25 0.015
9 MMX 8883 2 1 0.5 0.25 0.5 0.015 0 MMX 8885 2 1 0.5 0.5 0.25 0.015 1 MMX 8889 2 2 0.5 0.5 0.125 0.015 2 MMX 8890 2 1 0.5 0.25 0.25 0.015 3 MMX 8891 2 1 0.5 0.25 0.25 0.015 4 MMX 8915 2 1 0.5 0.25 0.125 0.015 5 MMX 8917 2 1 1 0.25 0.25 0.015 6 MMX 8920 2 1 0.5 0.5 0.25 0.015 7 MMX 8921 2 1 0.5 0.25 0.25 0.015 8 MMX 8923 2 1 0.25 0.5 0.25 0.015 9 MMX 8926 2 1 0.5 0.25 0.25 0.015 0 MMX 9013 2 1 0.5 0.5 0.25 0.015
MICso 2 1 0.5 0.25 0.25 0.015
MIC90 2 2 0.5 0.5 0.5 0.03 range 2-2 1-2 0.25-1 0.25-0.5 0.125-0.5 0.015-0.03
Table 14B (COL-S K. pneumoniae)
Figure imgf000309_0001
MIC (Mg/mL)
# Strain CMP 14 CMP 54a PMB COL TIG MERO 2 MMX 6379 4 2 1 0.25 1 0.03 3 MMX 6380 2 2 0.5 0.25 1 0.03 4 MMX 6381 2 1 0.5 0.25 1 0.03 5 MMX 6384 2 2 0.5 0.25 2 0.03 6 MMX 6386 2 1 0.5 0.5 1 8 7 MMX 8708 4 2 0.5 0.25 2 0.03 8 MMX 9030 2 1 0.5 0.25 1 0.06 9 MMX 61 17 4 2 0.5 0.25 1 0.06 0 MMX 6383 2 1 0.5 0.25 1 0.06
MICso 2 2 0.5 0.25 1 0.03
MIC90 4 2 0.5 0.5 2 0.5 range 2-8 1-2 0.5-1 0.25-0.5 1-8 0.03-8
Table 14C (COL-S P. aeruginosa)
Figure imgf000310_0001
MIC (Mg/mL)
# Strain CMP 14 CMP 54a PMB COL TIG MERO 0 MMX 8848 2 1 1 4 >8 1
MICso 2 1 1 1 >8 0.5
MIC90 4 2 1 2 >8 >8 range 2-8 1-2 0.5-1 0.5-4 >8->8 0.125->8
Table 14D (COL-S A. baumannii)
Figure imgf000311_0001
Table 14E. MICso and MIC90 analysis of Compounds 14 (CMP 14) and 54a (CMP 54a), PMB, COL, TIG, MERO, CAZ (ceftazidime), AMIK (amikacin), and LEVO (levofloxacin) against COL-R mixed-species clinical isolate panel
Figure imgf000312_0001
MIC (Mg/mL)
CMP CMP MER AMI
Species Strain PMB COL TIG CAZ LEVO
14 54a 0 K
K.
MMX
pneumoni 16 16 64 128 1 32 >256 16 32
6269
ae
K.
MMX
pneumoni 4 2 1 16 0.5 0.03 0.25 1 0.06
6382
ae
K.
MMX
pneumoni 4 4 64 256 2 64 >256 64 64
881 1
ae
P.
MMX
aeruginos 2 1 16 16 >8 1 4 4 0.5
7874
a
P.
MMX
aeruginos 8 8 256 1024 >8 8 64 32 >256
6977
a
A. MMX
2 1 16 256 4 128 >256 16 32 baumannii 8537
A. MMX
2 1 2 8 4 64 256 4 16 baumannii 8644
A. MMX
32 >32 64 256 4 128 256 8 16 baumannii 8990
A. MMX
2 1 8 128 4 64 >256 2 128 baumannii 6971
A. MMX >102 >25
4 2 256 2 16 32 8 baumannii 6973 4 6
A. MMX >25
2 1 8 32 4 32 128 8 baumannii 6975 6
A. MMX >25
4 2 32 512 4 64 >256 8 baumannii 8381 6
A. MMX >102 >25
8 8 256 4 8 128 16 baumannii 8383 4 6 MIC (Mg/mL)
CMP CMP MER AMI
# Species Strain PMB COL TIG CAZ LEVO
14 54a 0 K
A MMX >102 >25
23 4 2 256 2 16 64 8 baumannii 8384 4 6
A. MMX >25
24 4 1 8 64 2 32 128 8 baumannii 8386 6
A. MMX
25 2 2 2 8 2 32 128 4 16 baumannii 6338
A. MMX
26 2 ND* ND 128 ND 128 ND ND ND baumannii 8537
MICso 4 2 32 128 2 32 >256 32 32
>102 >25
MIC90 16 16 256 8 128 >256 256
4 6
1- 8- 1-
0.5- 0.03- 0.25- 0.06- range 2-32 1->32 >102 >102 >25
>8 256 >256 >256 4 4 6
* ND: not determined
Table 15. Summary of MIC90 analysis of Compounds 14 (CMP 14) and 54a (CMP 54a), PMB, COL, TIG, MERO, CAZ (ceftazidime), AMIK (amikacin), and LEVO (levofloxacin) against COL-S species and COL-R mixed-species clinical isolate panels
Figure imgf000314_0001
MIC90 (range) ( g/mL)
CMP CMP
Species PMB COL TIG MERO CAZ AM IK LEVO
14 54a
>1024 128 >256 >256 256
COL-R 16 (2- 16 (1 - 256 (1 - 8 (0.5-
(8- (0.03- (0.25- (n=25) 32) >32) >1024) >8) (1 - (0.06- >1024) 256) >256) >256) >256)
Table 16. Summary of MIC90 analysis of Compound 84, Compound 90, Compound 91 , Compound 1 1 0, and COL, TIG and MERO against COL-S species and COL-R mixed-species clinical isolate panels
Figure imgf000315_0001
Example 113. Time-kill analysis of Compound 14 and colistin (COL) against E. coli ATCC 25922
Time-kill assays were performed according to CLSI guidelines (Methods for Determining Bactericidal Activity of Antimicrobial Agents; Approved Guideline. CLSI document M26-A (ISBN 1 -56238- 384-1 )). An overnight Mueller-Hinton agar (MHA) culture of E. coli ATCC 25922 was resuspended in 0.85% NaCI to -0.1 ODeoo and added to 10 mL Mueller-Hinton broth (MHB) medium to achieve a mid-105 CFU/mL starting inoculum. Compound 14 and COL were then added to the inoculated MHB to produce concentrations equivalent to 0, 1 , 2, 4, and 16X the corresponding broth microdilution MIC value for each strain (Compound 14 - 2 μg/mL and COL - 0.5 μg/mL). Time-kills were performed in baffled, ventilated 125 mL Erlenmeyer flasks (TriForest, cat. no. FBC0125S) incubated 8ί 37 in a shaking incubator. Aliquots were removed from each flask at 0, 1 , 3, 6, 9, and 24 hours and serially diluted 1 :10 in 0.85% NaCI. Fifty microliters of each serial dilution were spread onto MHA plates and incubated for 24 hours at 37°C (assay limit of detection: 20 CFU/mL). Colonies were counted and CFU/mL values were calculated and plotted using GraphPad Prism 6 (GraphPad Software, Inc.) (FIGS. 3A and 3B). Fungistatic activity was defined by CFU reductions <3-logs from the starting inoculum and fungicidal activity was defined by CFU reductions≥3-logs (denoted by dashed lines in FIGS. 3A and 3B).
Similar assays were also performed using K. pneumoniae ATCC 43816, P. aeruginosa ATCC
27853, and A. baumannii ATCC 17978 and the results are shown in FIGS. 3C-3H.
Example 114. Spontaneous mutation frequency determination and cross-resistance analysis.
The spontaneous mutation frequency of Compound 14 and COL were determined for E. coli ATCC 25922. Three independent colonies of E. coli ATCC 25922 were picked from an overnight MHA plate and restreaked on fresh MHA plates and grown overnight. The following day a loop was used to scrape cells to generate an Ο βοο -0.80 suspension in 0.85% NaCI. One-hundred microliters (-6-8x108 CFU) of each culture was then spread with sterile glass beads on plates (90 mm x 90 mm) containing 40 ml_ of MHA prepared with either 1 X or 2X the corresponding concentration of Compound 14 or COL needed to completely inhibit growth. Plates were incubated at 37°C for 48 hours and then putative mutant colonies were picked with a loop and streaked onto MHA plates containing the same
concentration of Compound 14 or COL that they were isolated from. Spontaneous mutation frequencies were then calculated by dividing the number of colonies that successfully regrew after 24 hours by the starting inoculum plated on each plate (Table 1 7). None of the 14 Compound 14-selected mutants and 2 of the 47 COL-selected spontaneous E. coli ATCC 25922 spontaneous mutants demonstrated broth microdilution MIC shifts 2-fold for the respective selecting compound (data not shown). The remaining 45 COL-selected mutants were evaluated in an MIC assay to determine the fold-shift changes vs. COL as well as cross-resistance to PMB, Compound 14, and MERO (Table 18). Table 17. Selection of Compound 14 and COL spontaneous mutants in E. coli ATCC 25922 on MHA plates containing 1 X or 2X the respective agar inhibition concentrations
Figure imgf000316_0001
All Compound 14 colonies selected grew upon restreaking on 1 X MIC agar media, however, when tested in broth microdilution MIC, failed to demonstrate≥2-fold reductions in susceptibility. 2AII but 2 of the COL-selected colonies at "I X or 2X MIC demonstrated≥2-fold reductions in susceptibility upon broth microdilution MIC testing.
Table 18. E. coli ATCC 25922 COL-R spontaneous mutants cross-resistance MIC panel (n=45 mutant strains)
Figure imgf000317_0001
Figure imgf000318_0001
Example 115. Membrane Permeability
The outer membrane of Gram-negative bacteria is composed of negatively charged LPS which constitutes a potent permeability barrier against the entry of hydrophobic compounds into cells. The ability of Compound 14 to damage the outer membrane was investigated using 1 -N-phenylnaphthylamine (NPN), an uncharged lipophilic dye. NPN fluoresces weakly in aqueous environments, but its
fluorescence greatly increases in hydrophobic environments such as the cell membrane. Therefore, the analysis of changes in fluorescence intensity of NPN in compound treated bacteria constitutes a sensitive and convenient method to evaluate changes in outer membrane permeability. Cell death was evaluated in parallel by use of SYTOX® Green, a green-fluorescent nuclear and chromosome counterstain that is impermeable to live cells.
Log-phase bacteria of two colistin-susceptible (E. coli ATCC25922 and K. pneumoniae 43816) and three colistin-resistant strains (K. pneumoniae MMX6949, P. aeruginosa MMX6977 and A. baumannii MMX8537) were used in the studies. Bacteria were treated with Compound 14, Compound 54a, Compound 59, or colistin at concentrations ranging from 0.1 to 5 μΜ in RPMI for 90 min. Compound 14, Compound 54a, and Compound 59 more potently induced membrane permeability for all bacteria when compared to COL (FIG. 4A). Bacterial cell death against COLs susceptible strains was similar for Compound 14 and COL, whereas Compound 54a and Compound 59 killed all strains tested at lower concentrations (FIG. 4B). Treatment of COLR strains with Compound 14 and Compound 54a resulted in significant cell death. These data demonstrate, Compound 14 and its analogs potently disrupt the outer membrane resulting in cell death in both COLs and COLR strains.
Damaging the outer membrane of Gram-negative bacteria has several important consequences:
(1 ) it can lead to direct killing of bacteria as shown by MIC assays and in the experiments described here,
(2) it sensitizes bacteria to killing by natural antimicrobials or in combination with other antibiotics, and (3) it results in increased complement fixation (data not shown) and CDC (complement dependent cytotoxicity).
Example 116. Antibacterial Activity of Compounds 14 against MDR CRE clinical isolates
Compound 14 was further evaluated against a panel of seven characterized multidrug resistant (MDR) carbapenem-resistant Enterobacteriaceae (CRE) clinical isolates, some of which possessed the mcr- 1 gene (Table 1 9). Similarly to the MDR isolates in the COLR MIC90 panel, Compound 14 had consistent activity against these MDR CRE strains, including those possessing mcr- 1. Compound 14 had MICs of 2-4 μg/mL, except against a mucoid-producing strain (K. pneumoniae 30660 (Kp 30660)) where the MIC was 8 μg/mL. These data show the promising activity of Compound 14 against even the most problematic clinical isolates (such as the ceftazidime-avibactam resistant Klebsiella strain Kp 47772) independent of the potential added benefits from immune system engagement. Table 19
MOR fCs E co// 47554 Kf>1 4876S Kp 47768 K{ 47772 Kp 47640 Kp 30660* Kp 30684
ae
» .ν· ;; ίϊ -Ϊ 'ίϊ, <¾¾»■,,,, Sfa
i.i1. i;f."'. i!,
■Λ¾. *iss Μκ<«.
<wf83. iSuft s .AS.
! W.
AO!SpKIS, iCfrttrtOS, vVT 0¾pC iCrsoi 3(> W OmsK¾6 ν'ί'Γ Offif-KSe
MiC (Mg m!)
4 4
Meropenem 64 84 32 4 64 s 8
Cettaziiiitns-avibeetam >£4 1 4 266 >64 0.5 ·;
CeitaskSitnfc Hi4 >64 58 812 >S4 54 S4
Cefepitae >W >16 H28 s 16 18 S
2 >32 »258 8 32 32 32
64 NT-1 NT 64 S4 6
Coiisiin 2 1 <2 <2 ■- 8 <C.5
Ttgeeye!me >4 1 NT NT 1 i
' Kp - KMmz!ls prsimo m; 5 mucoid producer; · OMP - Outer Mem&rane Protein; 4 Not Tes ed
Example 117. Compound Recognition by Rha Abs and Rha-Coordinated Binding to Bacteria
Rha-specific Abs (from vaccinated rabbits or natural Abs purified from human serum) were affinity purified and binding of Compound 14 to purified anti-Rha antibodies was measured using Surface Plasmon Resonance (BiaCore 3000). Binding was measured by flowing Compound 14 (analyte) over a CM5 chip coated with immobilized antibody (ligand). Equilibrium dissociation constants (KDs) of 87.3 nM and 66.8 nM were measured for complexes of Compound 14 with rabbit and human antibody, respectively. For Compound 59, which possesses two Rha molecules, KDs were 2.4 nM and 6.0 nM, respectively (data not shown).
The ability of Rha Ab to engage compound bound to bacteria was determined. ELISA plates were coated with 50 μΙ_ of a log-phase culture of E. coli ATCC 25922 (Ο βοο = 0.5)/well and dried.
Coated plates were then incubated with Compound 14, Compound 54a, and Compound 59 or compounds that do no have Rha molecules: Compound X and Compound Y (structrures shown further below) at indicated concentrations. Binding was then probed with purified human anti-Rha Ab at a fixed concentration. Bound antibodies were then detected using either an anti-human IgG secondary antibody (FIG. 5A) or an anti-human IgM secondary antibody (FIG. 5B). All three Rha-containing compounds (Compound 14, Compound 54a, and Compound 59) mediated binding of purified human Rha-Ab to E. coli. Binding was dose-dependent and both IgG (FIG. 5A) and IgM (FIG. 5B) isotypes were detected. To further demonstrate Rha-specific binding, competition assays using ELISA were performed with soluble L-rhamnose. As before, E. coli coated plates were incubated with various concentrations of Compound 14 and human anti-Rha Ab was added in the absence or presence (0.01 -1 mM) of L-rhamnose. Binding of Rha ab was detected using an anti-human IgG secondary antibody. L-rhamnose spike-in resulted in dose-dependent inhibition of anti-Rha Ab binding, confirming specificity for this carbohydrate (FIG. 5C). Furthermore, binding of purified rabbit or human anti-Rha Abs to six bacterial strains immobilized on ELISA plates was evaluated using Compound 14, Compound 54a, and Compound 59, colistin, and compounds that do no have Rha molecules: Compound X and Compound Y, as described above. ELISA plates were coated with 50 μί of a log-phase culture of the indicated bacterial strains (Ο βοο = 0.5)/ well and dried. Compounds were added at the indicated concentrations and probed with purified rabbit anti- Rha antibodies or human anti-Rha antibodies at a fixed concentration. Bound antibodies were detected using an anti-human IgG secondary antibody. Only Rha-containing compounds (Compound 14, Compound 54a, and Compound 59) mediated binding of purified rabbit (FIG. 6) or human anti-Rha abs (FIG. 7) to bacteria. Binding intensity was strain and dose-dependent. Binding of Rha antibodies to live bacteria was confirmed by flow cytometry (data not shown).
Collectively, these data demonstrate that L-rhamnose is effective at recruiting rabbit or human Rha antibodies to a wide variety of bacterial strains.
Figure imgf000321_0001
Compound X
Figure imgf000321_0002
Compound Y
Example 118. Rha-lnduced Enhancement of Bacterial Cytotoxicity in Whole Blood
In addition to possessing intrinsic antibiotic activity, the compounds may opsonize bacteria and recruit immune effectors to the site of infection. Using whole blood killing assays, immune-mediated enhancement of bactericidal activity has been demonstrated. 4x 105 CFU/mL of E. coli 25922 were incubated in the presence or absence of Compound 14, Compound X, or colistin (COL) at sub-inhibitory concentrations (0.02 μΜ or about 0.03 μg/mL) in RPMI medium (FIGS. 8A and 8D) or human blood (FIGS. 8B and 8E). After 90 minutes of incubation, bacteria were harvested and colony forming units
(CFUs) were counted. The drug concentrations used were too low to inhibit bacterial growth in the absence of human blood (FIG. 8A, drug vs. RPMI alone). To correct for the intrinsic activity of human blood, the antibacterial activity of blood from donor 1 was measured - it reduced the CFUs/mL from 520k to 72k (86% reduction). Compound X or COL also improved antibacterial activity of blood. Compound 14 however, enhanced bacterial killing by more than tenfold (p<0.0001 , ANOVA with Tukey's adjustment), clearly demonstrating the Rha-mediated engagement of immune components present in blood (FIG. 8B, whole blood). This reduced the percentage of surviving bacteria from approximately 13% when treated with whole blood alone (or whole blood plus COL) to less than 1 % (FIG. 8C). The percentage survival was calculated relative to RPMI control for each compound to account for differences in non-Rha-induced antimicrobial activity. Results shown represent average with dots for individual data points and was analyzed by one-way ANOVA (****P<0.0001 ). Donor 1 had anti-Rha Ab titers of 12,800 for IgG and 200 for IgM.
Using blood from another independent donor, killing enhancement with Compound 14 was retested, and compared to enhancement with Compound 54a and Compound 59. Killing enhancement observed with Compound 14 (donor 2, FIGS. 8D-8F) was similar to levels observed with donor 1 .
Compound 59 and Compound 54a were superior, enhancing bacterial killing by 4- and 1 0-fold, respectively, compared with Compound 14. It is important to note that anti-Rha IgG titers were lower in this particular donor (lgG=800, lgM=200) demonstrating that immune mediated killing enhancement is retained across a wide range of titers. Compound 54a and Compound 59 have consistently shown improved immune mediated activity as compared to Compound 14, demonstrating that the Rha-mediated activity of small molecule compounds can be optimized.
Of note, when mouse blood (spiked with rAb) was tested for whole blood killing using identical assay conditions, antibacterial activity was not observed in the presence or absence of compound (data not shown) highlighting key differences between mouse and human blood. One potentially important difference between human and mouse blood is white blood cell composition - human white blood cells contain -60% neutrophils, while much lower percentages are found in mouse blood. This is important when interpreting in vivo efficacy studies performed in mice as the immune-mediated component of bacteria killing may be underestimated in this species.
The human whole blood data presented above clearly demonstrate that the compounds benefit from immune-mediated enhancement of bactericidal activity. Importantly, multiple effector mechanisms potentially contribute to killing, including CDC, ADCC, and potentiation of the activity of antimicrobial peptides. The relative importance of respective mechanisms may vary in patients depending on individual predispositions. However, in cases where the pathogen displays resistance to antibiotic treatment, clinical outcomes may strongly depend on the successful interaction of these host immune mechanisms with the bacterium.
Example 119. Compound-Induced Enhancement of Complement-Dependent Cytotoxicity (CDC)
The complement system is one of the first lines of defense against invading pathogens, and plays an essential role in both innate and adaptive immunity. The complement cascade can be activated through distinct pathways; the classical pathway (CP), the lectin pathway (LP) or the alternative pathway (AP). The classical pathway is initiated by antibodies binding to the surface of the invading pathogen, while the alternative pathway acts through spontaneous hydrolysis of the protein C3. Whether initiated by either pathway, direct killing of pathogens by complement operates through the formation of the membrane attack complex (MAC). This activity can be measured in normal human serum (NHS) but is abolished in heat inactivated serum (HIS).
To demonstrate compound-mediated enhancement of complement activity, CDC assays were developed. Log phase E. Coli ATCC 25922 were generated by overnight culture and regrowth until OD6oo=0.4 (1 e8 CFU/ml). 1 x 106 CFU/mL of E. coli 25922 were incubated with sub-inhibitory
concentrations of 0.02 μΜ (-0.03 μg/mL) of Compond 14, Compound 54a or Compound 59, Compound X, or COL in RMPI alone (data not shown) or RPMI supplemented with 10% normal human serum (NHS) (FIGS. 9A and 9B) or 10% heat-inactivated (HIS) (FIGS. 9C and 9D). Plates were incubated at 37 °C for 90 minutes and surviving bacteria were determined by CFU enumeration (Rha titer lgG=6400 lgM=800). The low drug concentrations used did not inhibit bacterial growth in the absence of serum (RPMI alone, data not shown). Addition of NHS alone (FIGS. 9A and 9B) had little impact on bacterial growth compared to bacteria grown in RPMI (1 x 1 06 to 4x 106). Notably, while Compound X or COL displayed no antibacterial activity in NHS, Compound 14 enhanced bacterial killing by 22%, Compound 54a by 82%, and Compound 59 by 57% (FIG. 9B). This activity was not seen when the experiment was run with heat inactivated serum (HIS) (FIGS. 9Cand 9D), demonstrating that the effect was mediated by complement. These data derived from ex vivo killing assays suggest that the compounds with Rha attached recruit and activate both complement and cell-mediated immune effector arms of the immune system. Initial investigation of cell-mediated effects demonstrated the compounds increased neutrophil activity (data not shown).
Example 120. Mouse Models of Infection
The in vivo activity of Compound 14 has been evaluated in two key efficacy models, mouse septicemia (sepsis, bacteremia) and a more stringent neutropenic thigh infection model. Plans are to further evaluate Compound 14 in these models with other G- pathogens (K. pneumoniae, P. aeruginosa, A. baumanii, and ColR pathogens) and in a lung infection model. The septicemia model is useful for compound screening and can provide two readouts, survival or kidney CFUs. Animals are infected IP and treated one hour later by the IP route. The thigh infection model, requires penetration of drug into muscle. The readout is thigh muscle CFUs. Animals are inoculated directly into the thigh and drug is dosed IP. It generally takes a higher dose of drug to demonstrate efficacy in the thigh versus the septicemia model. The success of Compound 14 in the immunocompromised thigh infection model supports its use in a non- immune-competent population.
Mice lack natural Rha Abs but they can be acquired by immunization or by passive transfer. Rha linked to ovalbumin provided a vaccine (OVA-R2) to immunize mice to raise anti-Rha antibodies.
However, Ab titers of mice immunized in this way were quite variable. To overcome this and obtain consistent titers, passive transfer of the antibodies was achieved by IV injection of affinity-purified antibodies isolated from the serum of OVA-R2 vaccinated New Zealand white rabbits (rAbs). Importantly, rAbs have been shown to bind murine Fc receptors as well as murine complement and, therefore, are expected to mediate an effect although the response may be less robust compared to a homogeneous system (i.e., a clinical setting).
Example 121. Immune-Competent Mouse Septicemia Model
Rha-mediated activity could be evaluated in vivo using an established, acute disseminated E. coli mouse septicemia model. Both intrinsic antibacterial activity of Compound 14 and additional immune- dependent efficacy relying on the presence of rAbs were investigated. Mice (n=12) infected with E. coli 25922 were given a single dose IP of Comppound 14 + rAb (FIG. 10,■), Compound 14 only (FIG. 10, o), or COL only.
FIG. 10 shows a dose ranging study with Compound 14 (MIC 2 μg/mL against the challenge strain) in a mouse E. coli septicemia model where kidney CFUs at 16 hours were enumerated. Improved efficacy was seen at 3 and 5 mpk in the presence of rAb with an additional ~1 -log reduction in burdens (P<0.05) versus Compound 14 in the absence of antibody or colistin (MIC 0.5 μg/mL) at 1 mpk. Plasma exposure of Compound 14 in this study was estimated at 2-3 μghr/mL. Importantly, the bacterial burden in most mice in the "+rAb" groups was at the limit of detection (LOD) suggesting the actual effect may be significantly greater than it appears in the figure. In a similar study (data not shown), Compound X (no Rha attached) did not show rAb-enhanced efficacy while Compound 14 showed a dose-dependent reduction in CFUs which was improved in the presence of rAb. Example 122. Immune-Suppressed Mouse Thigh Infection Model
FIG. 1 1 shows a dose ranging study with Compound 14 (MIC 2 μg/mL) in a neutropenic mouse E. coli thigh infection model where bacterial burden in thigh muscle tissue at 24 hours was determined. Mice (n=6) infected with E. coli AT CC 25922 were administered Compound 14 either once (qd), twice (bid) or three times (tid) in one day either by IV (10 or 15 mg/kg/day), IP (20, 30, 60 or 90 mg/kg/day), or SC (20, 30, 60 or 90 mg/kg/day) routes. COL was administered IV qd (1 mg/kg/day) or IP qd (3 mg/kg/day) and gentamicin was administered SC bid (20 mg/kg/day). Only mice receiving Compound 14 were administered 1 00 μΙ of rabbit rAb 24h prior to E. coli challenge. Efficacy was demonstrated in all Compound 14 treated groups (+rAb), as well as in COL (MIC 0.5 μg/mL) and gentamicin (positive control) treated mice. E. coli thigh CFUs were significantly lower (p <0.01 ) in Compound 14, COL, and gentamicin treated mice. Compound 14 administered either IV, IP or SC bid or tid showed the greatest reduction in CFUs/thigh versus qd dosing. Higher variability seen in the Compound 14 SC treated mice was perhaps due to variable distribution to thigh tissues after SC dosing especially during the short duration of the study (24h endpoint). Compound 14 demonstrated robust efficacy at estimated plasma exposures (13.4 μghr/mL) that are well below the NOAEL.
In a similar study activity of Compound 14 against K. pneumoniae in an immune-competent thigh model was also examined. In that study, an approximate 1 .5 log reduction in thigh burden was achieved when the animals were given a single IV dose of Compound 14 at 5 mg/kg (data not shown). Example 123. Antibody Titration Study
To investigate the minimum concentration of rAb necessary to engage the Rha of Compound 14 and produce an effect the concentration of rAb was varied in our standard E. coli Septicemia Model. Concentrations of rAb from (1000) to (32,000) in 4-fold increments (upper titrations not graphed for clarity; FIG. 12) were tested with Compound 14 fixed at 3 mg/kg (IP, single dose) and kidney burden elaborated at 16 hours. Mice (n=6) survived in all but the vehicle control group with 4 deaths.
As highlighted in FIG. 12, even at the lowest rAb concentration tested the Rha is fully engaged in this experiment and the maximum effect is achieved. However, it is worth noting that the majority of animals in the -i-rAb group are at the studies limit of detection, therefore it is possible that some differences in efficacy could be teased out if Compound 14 was tested at a lower concentration. Additional studies will be conducted to further clarify the low end of rAb concentration required for an effect in a murine system. It is encouraging that in our current test system potent efficacy is seen at modest Rha Ab titers.
Example 124. Drug Metabolism and Pharmacokinetics (DMPK) and Drug Stability
The plasma concentration-time profile and resulting pharmacokinetics of Compound 14 was determined upon dosing in mice (IV and IP; efficacy route), rats (IV and subcutaneous (SC); toxicology route), and cyno monkeys (IV and SC) (Table 20). It should be noted that the pharmacokinetics of Compound 14 (IP 10 mg/kg ± rAbs) was comparable with or without co-administered rAbs. The plasma half-life was approximately 1 .2 h, 0.5 and 0.6 h, in the mouse, rat and monkey, respectively. Plasma clearance in the respective animal species tested (mouse, rat, and monkey) appears to be low (<10% of corresponding liver blood flow). As plasma protein binding of Compound 14 was high (>99% bound) in all species including human, this could likely account for the overall lower clearance observed.
Table 20
AUCV* AUCinf CL v„ V, F
Dose
Species Route (hr) (jig/inL)
(mg/kg) (μο-hr mL) (iiS-hi mi l (hr) (mL/miii/kg) (iuL/kg) (mL/kg) < )
Mouse IV bolus 3 NA 27.2 15.5 15.5 1.19 3.56 466 388 NA
IP 3 0.50 1.S0 2.09 2.30 NA NA NA NA 15%
IP 10 0.50 4.75 4.99 5.49 NA NA \ NA 1 1%
IP (+ rAbs) 10 1.00 2.74 3.78 6.72 NA NA NA NA 13%
SC 3 0.33 0.965 I 50 1.73 NA N A . NA 11%
Rat IV bolus 10 "MA 34.3 20.3 20.4 0.52 8.17 268 368 NA
SC 10 2.67 1.84 9.65 15.3 NA N A NA NA 75%
Monkey FV bohis 1 1.00 4.06 2.54 2.83 0.61 6.12 258 308
Figure imgf000325_0001
IV 1-hr inf 3 1.00 27.0 37.5 37.7 0.57 1.34 83.8 66.1 A
TV I-Iir inf 10 1.00 49.0 126 127 1.2 1.32 166 136
SC 10 1.17 25.1 77.7 88.8 MA NA. NA NA 70%
Bioavailability from SC dosing appeared to be high at about 70-75% in the species tested although IP dosing only generated a bioavailability of about 13% (repeated to confirm result). In the rat, parent drug was not detected in the urine. More recently, the plasma concentration-time profile of Compound 14 was characterized following a toxicokinetics study of IV 1 -hr infusion from a monkey tolerability study at doses of 3, 10, and 20 mg/kg. The resulting exposures suggest dose proportionality from 3 to 10 mg/kg. An earlier 1 mg/kg IV bolus administration was also studied.
Moreover, in vitro metabolic stability experiments carried out with un-labeled Compound 14 incubations in whole blood and liver microsomes/hepatocytes indicate that the compound is stable. FIG. 13 shows the metabolic stability of Compound 14 in rat, monkey and human hepatocytes.
Furthermore, from in vivo studies, we have further profiled for metabolites employing
LC-QTof-MS mass defect filtering in select plasma/urine samples from the rat and have not detected any Ph1 or oxidative biotransformation products. We had also specifically looked for precursors of the parent compound that are available as synthetic standards, including those that involve cleavage of the linker, but have not found any to date. Therefore, the bifunctional compound appears to be stable in vivo. Thus far, there does not appear to be significant metabolism and it may be that we will need to characterize its metabolism using radio-labeled compound. We are also characterizing the concentrations of Compound 14 in various tissues. Example 125. Off-Target Activity
Compound 14 was evaluated in the CEREP SafetyScreen44 battery of in vitro
enzyme/receptor/ion channel assays at 10 μΜ. Six CNS targets were inhibited at 55-85% (a2A adrenergic, κ-opioid, 5-HTI B, 5-HT2a receptors and dopamine and norepinephrine transporters) and of lesser concern since Compound 14 is unlikely to cross the blood brain barrier due to its multiple charge state and size. Screening against a broader panel is planned. Binding affinity (ICso) to the rat potassium A-type channel (KA), a neuronal target, was 330 nM; COL had a ICso of 9610 nM. The binding assay results were followed up in functional human voltage-gated potassium channels Kv1 .1 , Kv1 .2 and Kv1 .6 using an electrophysiological platform (lonWorks Quattro). Results from this functional assay suggest that antagonism of the channels occurred at much higher concentrations than observed in the binding assay with ICso values estimated to be >100 μΜ (FIG. 14). Minimal CYP450 inhibition (1 A2, 2B6, 2C8, 2C9, 2C19, 2D6, 3A4) was observed when tested at a range of Compound 14 concentrations with ICso values determined to be >30 μΜ.
Example 126. Toxicology
Compound 14 in 5-Day Rat Toxicology Study. COL induces renal toxicity in humans at clinically relevant doses. Compound 14 was evaluated for nephrotoxicity using a rat model predictive of COL-induced renal injury in humans. Compound 14 was administered SC for 5 days to rats at 25 mg/kg/day BID, which generates plasma exposures (-24 μghr/mL), 12-fold higher than exposures required for efficacy in the mouse septicemia model. COL was similarly administered at 25 mg/kg/day, a dose that generates therapeutic plasma levels in humans. Urine was collected daily and monitored for albumin levels, a biomarker for renal proximal tubule injury. In contrast to COL, Compound 14 did not elevate urinary albumin. Lack of renal toxicity with Compound 14 was confirmed microscopically in kidneys collected on Day 6 (data not shown). Compound 14-treated rats demonstrated no adverse clinical pathology, hematology, or histopathology (major organs). Acute histamine-like effects were seen (similar to COL) but were rat specific; there were no adverse observations in singly-dosed cynos.
Histamine-like effects are not expected in humans, as none are seen with COL. Since these effects are likely Cmax-related, slow infusion of drug may minimize any potential histamine-related responses. Example 127. Compound 14 Safety Margins
Based on the more stringent thigh infection model that utilized an E. coli strain with an MIC of 2 μg/mL, results from the 5-day rat toxicology and single dose cyno tolerability studies, as well as exposures from mouse, rat, and cyno PK studies, the following margins have been calculated (Table 21 ). Based on projected efficacious plasma AUCs (10 mg/kg, bid, IP) and exposures as outlined below, there is a≥2.9-fold margin established regarding nephrotoxicity (likely to be wider) and a >19-fold margin regarding neuromuscular effects. Now that the effect in cynos has been correlated with Cmax, various dosing protocols will be explored (tid, 2h infusion or continuous infusion) to allow better toxicological assessment using higher exposures.
Table 21
Figure imgf000327_0001
Example 128. Synergistic Activity of of Compounds 14 and 54a
Part 1. Methods
Checkerboard synergy assays.
The synergistic activity of Compound 14 in combination with Gram-positive agents (AZITHRO
(azithromycin) and RIF (rifampicin)) and Gram-negative agents ((TIG (tigecycline), MERO (meropenem), LEVO (levofloxacin), AMIK (amikacin), and CAZ/AVB (ceftazidime/avibactam)) was assessed for the seven COL-R clinical isolates possessing the highest MIC values for Compound 14 (e.g.,≥8 μς/ιηί). Checkerboard synergy assays were performed as previously described (see, e.g., Eliopoulos and Moellering, "Antibiotics in Laboratory Medicine," 4th Edition (The Williams & Wilkins Co., Baltimore, Maryland, 1996), and Moody, "Clincal Microbiology Procedures Handbook," 2nd Edition, Volume 2 (Washington: ASM Press, 2004)). In master stock plates, the stocks of two compounds (Compound A, which is Compound 14 in this Example; and Compound B, which is a known antibacterial agent) for synergistic evaluation were prepared. A liquid stock of Compound A was prepared fresh in Dl water to a concentration 50X the desired final synergy assay concentration. Into wells A1 through H1 (vertical, Y- axis column on left edge of plate) of a 96-well PCR plate (VWR cat. no. 83007-374) Compound A was aliquoted in 100 μΙ_ volumes. To all other wells on the plate, with the exception of A12 through H12, 50 μΙ_ of Dl water was added. A multichannel pipette was then used to transfer 50 μΙ_ from column A1 -H1 to column A2-H2. Following mixing, tips were discarded and the process was repeated across the plate to create a series of 1 1 , serial two-fold dilutions spanning columns 1 through 1 1 .
A stock of Compound B was prepared in a similar fashion in the appropriate solvent (DMSO, Dl water, ethanol, etc.) at 50X the desired final synergy assay concentration. In a separate 96-well PCR plate, 100 μΙ_ aliquots of Compound B were added to wells in the top row of the plate spanning A1 through A12. In all other wells, excluding the bottom row (H1 -H12), 50 μΙ_ of the corresponding
Compound B solvent were added. In a similar fashion to Compound A, Compound B was serially diluted, two-fold down the plate using a multichannel pipette, resulting in 7 rows comprised of 12 wells with 50 μΙ_ diluted Compound B. In some cases a second dilution plate, setup in the same fashion as the first, was needed to capture synergy present at drug concentrations less than the range on the first plate.
Checkerboard synergy assays were performed using the same assay setup as the standard MIC assays previously described previously herein (i.e. inoculum, media, volume, 96-well assay plates, incubation time, incubation temperature, 100% inhibition assay endpoint, etc.). However, rather than a single addition of 2 μΙ_ of 50X stock compound solution to the assay plate containing MHB pre-inoculated with bacterial cells using the Multimek 96 liquid handling robot, two sequential additions were made (one from the Compound A 50X stock plate and one from the Compound B 50X stock plate). The resulting checkerboard plates contained a 7 x 1 1 -well grid (A1 -G1 x A1 -A1 1 ) of synergy wells containing varying proportions of Compounds A and B, an 1 1 -point dilution series of Compound A alone (wells H1 -H1 1 , assay concentration range 1 X to 1 /1024X), a 7-point dilution series of Compound B alone (wells A12- G12, assay concentration range 1 X to 1 /64X) and a positive growth control well containing no drug (well H12) (FIG. 1 ).
Following overnight incubation, checkerboard synergy plates were read visually at 100% growth inhibition. MIC values for Compound A and B were recorded from their respective single-drug dilution series wells and an optimal synergy well (or wells) was identified as that for which growth inhibition was achieved using a combination of the lowest possible concentrations of both drugs. The fraction inhibitory concentration (FIC) was calculated for each compound by dividing the combination MIC value in the optimal synergy well by the MIC value for the compound alone. The summation of the FIC values for Compounds A and B was then used to calculate the fractional inhibitory concentration index (FIG) and synergy was defined as an FICI value of <0.50.
FIC Compound A = MIC of Compound A in combination/MIC of Compound A alone
FIC Compound B = MIC of Compound B in combination/MIC of compound B alone
Figure imgf000328_0001
Part 2. Results
Table 22. Summary of checkerboard synergy analysis for Compound 14 in combination with Gram-positive and Gram-negative standard of care agents for COL-R isolates possessing the highest
MIC values for Compound 14
MIC (Mg/mL)
Ki pneumonae.
6951
Figure imgf000329_0001
Ki pneumonae.
Compound 14 alone 16 1 6 MMX 6956 16 8 8 4 32
Compound 14 combe ) 4 ί 4 4 4 4 2
AZITHRO alone 256 25 56 Ki pneumonae.128 512 128 1024 1024
AZITHRO combo 32 3 2 8 MMX 6961 32 64 1024 1
FICI 0.38 0.. 38 0.31 0.56 1 2 0.06
Compound 14 alone 32 3 2 8 Ki pneumonae. 8 4 8 64
Compound 14 combe ) 4 > 2 2 MMX 6269 4 2 2
RIF alone 16 6 4 32 32 32 2 2
Pi aerugnosa.
RIF combo 0.125 i 4 8 32 0.125 0.125
6977
FICI 0.13 0. 13 0.38 0.5 2 0.31 0.09
Compound 14 alone 32 1 6 16 8 8 A biiaumann. 4 64
Compound 14 combe ) 8 i 8 8 4 2 MMX 8383 4
TIG alone 8 ί > 2 1 16 4 4
A biiaumann.
TIG combo 2 1 0.5 1 2 2 0.125
8990
FICI 0.5 0." 75 0.75 2 0.63 1 0.09
Compound 14 alone 16 1 6 8 8 4 4 64
Compound 14 combe ) 8 1 6 4 4 4 2 2
MERO alone 32 1∑ Ϊ8 32 32 8 8 64
MERO combo 8 Y >8 4 4 4 1 1
FICI 0.75 : » 0.63 0.63 1 0.63 0.05
Compound 14 alone 16 1 6 16 8 8 4 64
Compound 14 combe ) 4 ί 4 0.125 4 2 2
LEVO alone 256 25 56 64 32 1024 8 16
LEVO combo 32 6 4 32 16 256 2 0.25
FICI 0.38 0 5 0.75 0.52 0.75 0.75 0.05
Compound 14 alone 16 1 6 8 8 8 8 64 MIC (Mg/mL)
Figure imgf000330_0001
Compound 14 comb o Ki pneumonae 0..031 4 0.031 4 0.5 4 16
AMIK alone 64 MMX 6951 32 32 16 64 1024 4
AMIK combo 32 16 16 8 32 0.016 1
Ki pneumonae.
FICI 0.5 0.75 0.5 1 0.56 0.5 0.5
6956
Compound 14 alons 3 16 16 8 16 8 8 64
Compound 14 comb o 8 8 1 4 4 2 8
Ki pneumonae.
CAZ/AVB alone 8 8 16 8 8 64 64
6961
CAZ/AVB combo 2 1 8 4 1 4 16
FICI 0.75 0.63 0.63 0.75 0.63 0.31 0.38
Ki pneumonae.
6269
Example 129. Synthesis of INT-42 (fe -butyl (2S)-2-aminooctanoate)
Pi aerugnosa.
6977
Figure imgf000330_0002
A suspension of (S)-2-aminooctano c acid (3.2 g, 20 mmol) in tert-butyl acetate A biiaumann. (30 mL) was cooled in an ice-water bath and added with perchloric acid 70 % (4.4 g). After the mixture w MMX 8383as stirred at 0 °C to room temperature for 2.5 days, it was concentrated by rotary evaporation. The remaining was diluted with MeOH (5 mL) and poured into a beaker containing ice (50 g) and NaOH pellet (2.3 g) A bii.aumann A. fter all the NaOH was consumed, the pH was adjusted to -7.5 with solid Na2C03. The suspension was MMX 8990 extracted by DCM (1 00 mL x 5). The combined organic layers were dried over Na2S04, concentrated by rotary evaporation and further dried under high vacuum. Yield 1 .93 g, 44.8 %. Ion found by LCMS: [M- tBu + H]+ = 160.
Example 130. Synthesis of INT-43
Figure imgf000331_0001
Step a. Synthesis of methyl (15S)-1-[(6-deoxy-alpha-L-mannopyranosyl)oxy]-15-hexyl-11-(2-{[(2S)- 1-methoxy-1-oxooctan-2-yl]amino}-2-oxoethyl)-7,10,13-trioxo-3-oxa-6,11 ,14-triazahexadecan-16- oate
A mixture of methyl (2S)-2-aminooctanoate (490 mg, 2.8 mmol) and INT-39 (600 mg, 1 .29 mmol) was dissolved in anhydrous DMF (2 mL) and DIPEA (702 mg, 5.4 mmol). The solution was cooled in an ice-water bath followed by dropwise addition of a solution of HATU (1 .06 g, 2.8 mmol) in DMF (3 mL) via syringe pump at a rate of 1 .5 ml/hr. After the addition, the reaction was directly purified using RPLC (100 g, 5 to 100 % acetonitrile and water). Yield 528.7 mg, 52.8 %. Ion found by LCMS: [M + H]+ = 777.
Step b. Synthesis of (15S)-11-(2-{[(1S)-1-carboxyheptyl]amino}-2-oxoethyl)-1-[(6-deoxy-alpha-L- mannopyranosyl)oxy]-15-hexyl-7,10,13-trioxo-3-oxa-6,11 ,14-triazahexadecan-16-oic acid
A solution of step-a product (528.7 mg, 0.681 mmol) in a 1 :1 mixture of MeOH:THF (6 mL) was cooled in an ice-water bath. It was added with a solution of LiOH (40 mg, 1 .67 mmol) in water (3 mL). The mixture was stirred for 3 hours, then neutralized by 4N HCI solution in dioxane. After partial concentration, the reaction was directly purified by RPLC (100 g, 0 to 80 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 487.4 mg, 95.6 %. Ion found by LCMS: [M + H]+ = 749.
Figure imgf000331_0002
706-Linker A (more polar, by HPLC RT) and 707-Linker B (less polar, by HPLC RT)
Step a. Coupling of L-nor-Leu Methyl Ester and Removal of Boc Group
HATU (3.1 g, 8.1 mmol) in DMF (5 mL) was added, dropwise, to a solution of racemic-transl - (ferf-butoxycarbonyl)pyrrolidine-3,4-dicarboxylic acid (1 g, 3.9 mmol), L-Norleu-OMe-hydrochloride (1 .5 g, 8.1 mmol), and triethylamine (4.0 g, 39.5 mmol) in DMF (10 mL) over a period of 20 minutes. The mixture was stirred for an additional 20 minutes then applied directly to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 1 0% to 95% acetonitrile and water using 0.1 % TFA modifier. The two diastereomers were separated and pooled and lyophilized separately into the more polar diastereomer (a) and the less polar diastereomer (b): LC/MS [M-Boc+H]+ = 414.2 for both Boc-protected intermediates. Each diastereomer was stirred in a 1 /1 mixture of DCM/TFA (8 mL) for 20 minutes then concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 0% to 75% acetonitrile and water using no modifier to afford the TFA salt after lyophilization as a hydroscopic white solid. Yield: 88%, 2 steps. LC/MS [M+H]+ = 414.2.
Note: The stereochemistry of the trans-pyrrolidine dicarboxylate diastereomers was arbitrarily assigned based on HPLC retention time.
Step b. Cbz Protection and Hydrolysis of Methyl Esters
Cbz-NHS ester (227 mg, 0.91 mmol) was added to a stirring mixture of the product from Step a
(more polar diastereomer) (400 mg, 0.76 mmol) and triethylamine in DMF (10 mL). The mixture was stirred for 4 hours and then applied to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 5% to 95% acetonitrile and using 0.1 % THFA modifier. The solvent was removed by rotovap and the di-ester was hydrolyzed stirring in a 1 /1 /2 mixture of methanol/THF/DI water containing LiOH (0.72 g, 30 mmol) for 30 minutes. The mixture was adjusted to pH 5 with acetic acid then concentrated and applied to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were lyophilized to afford INT-44a as a white solid. Yield: 78%, 2 steps. LC/MS [M+H]+ = 520.2.
Example 132. Synthesis of INT-44
Figure imgf000332_0001
HATU (644mg, 1 .69 mmol), in DMF (2 mL) was added, dropwise over 10 minutes, to a stirring mixture of INT-18 (2.65 g, 1 .69 mmol), INT-44a (400 mg, 0.77 mmol), and triethylamine (466 mg, 4.62 mmol) in DMF (6 mL) and the reaction was stirred for an additional 30 minutes. Acetic acid ( 2 mL) was added, followed by 5% Pd/C (400 mg) and the mixture was stirred under an atmosphere of hydrogen for 12 hours. The mixture was filtered through celite and applied to reversed phase liquid chromatography (RPLC) using an Isco Combiflash liquid chromatograph eluted with 35 % to 95% acetonitrile and water using 0.1 % TFA modifier. Yield: 35%. LC/MS [M+3H/3]+ =1 092.8.
Example 133. Preparation of INT-45
Figure imgf000333_0001
Tri-tert-butyl {[(2S,5R,8S,1 1 S,14S,17S,22S)-22-({(8S,1 1 S,14R)-8-amino-14-{2-[(tert- butoxycarbonyl)amino]ethyl}-1 1 -[(1 R)-1 -hydroxyethyl]-2,2-dimethyl-4,9,12,15-tetraoxo-3-oxa-5,10,13- triazapentadecan-15-yl}amino)-5-benzyl-17-[(1 R)-1 -hydroxyethyl]-8-(2-methylpropyl)-3, 6,9,12,15,18,23- heptaoxo-1 ,4,7,10,13,16,19-heptaazacyclotricosane-2,1 1 ,14-triyl]tri(ethane-2,1 -diyl)}triscarbamate was prepared by an analogous procedure to the preparation of INT-18 except D-Dab was utilized instead of L- Dab. Positive mass ion was found as (M - Boc + 2H+)/2: 732.5), tr = 1 .89 minutes with 5 min gradient (40-95%) method.
Example 134. Preparation of INT-46. Trans-3-({4-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanamido}methyl)cyclopropane-1 ,2- dicarboxylic acid (pair of diastereomers)
Figure imgf000333_0002
(pair of diastereomers) Step a: Synthesis of dimethyl trans-3-(hydroxymethyl)cyclopropane-1 ,2-dicarboxylate
To a solution of aldehyde (see Preparation of INT-26, Step c, 4.496 g, 24.15 mmol) in THF (177 ml_) in a 1000 ml_ round bottom flask was added glacial AcOH (30 ml_, THF:AcOH = 177:30) and NaBh CN subsequently at 0 °C. The reaction was warmed up to room temperature for 15 min. Water (177 ml_) was added and pH was adjusted to 7 with saturated NaHCCb solution. The aqueous layer was extracted with ether (3 x 100 mL) and the combined organic layers were dried over Na2S04. Solvents were removed. Silica gel column chromatography (0.5-6% MeOH/DCM, UV@214nm) to give the alcohol 2.85 g (63%). Ή NMR (300 MHz, Chloroform-d) δ 3.99 (dd, J = 1 1 .9, 5.4 Hz, 1 H), 3.84 (dd, J = 12.5, 7.4 Hz, 1 H), 3.75 (s, 3H), 3.73 (s, 3H), 2.38 (d, J = 7.4 Hz, 2H), 2.14 (qd, J = 7.4, 5.4 Hz, 1 H).
Step b: Synthesis of dimethyl trans-3-(bromomethyl)cyclopropane-1 ,2-dicarboxylate
To a stirred solution of alcohol (2.85 g, 15.1 mmol) in CH2CI2 (130 mL) at room temperature were added PPh3 (6.02 g, 22.7 mmol) and CBr4 (7.70 g, 22.8 mmol). After being stirred for 1 hr, the mixture was concentrated under reduced pressure and the residue was purified by column chromatography (10-15% Et20/Hexanes, UV@214nm) on silica gel to give bromide as a colorless oil (3.58 g, 94%). 13C NMR (100 MHz, Chloroform-d) δ 170.8, 1 69.7, 52.5, 52.4, 30.4, 29.8, 28.5, 28.3
Step c: Synthesis of dimethyl trans-3-(azidomethyl)cyclopropane-1 ,2-dicarboxylate
To a stirred solution of bromide (3.58 g, 14.3 mmol) in DMF (0.1 5 M) at room temperature were added TBAI (1 1 .7 g, 31 .1 mmol) and NaN3 (4.68 g, 71 .3 mmol). After stirring at 80 °C for 2 hrs., the mixture was cooled to room temperature and then treated with H2O (two volumes). The mixture was extracted with hexane (3x). Combined organic layer was dried over Na2S04 and concentrated under reduced pressure to give the azide as a colorless oil (2.96 g, 97%), which was pure enough and was used for the next reaction without purification: Rf = 0.2 (Et2O/hexane=10:90); 1 H NMR (300 MHz, Chloroform-d) δ 3.76 (s, 3H), 3.74 (s, 3H), 3.68-3.48 (m, 2H),2.43 (dd, J = 9.2, 4.8 Hz, 1 H), 2.30 (t, J = 5.4 Hz, 1 H), 2.21 -2.08 (m, 1 H).
Step d: Synthesis of dimethyl trans-3-{[(tert-butoxycarbonyl)amino]methyl}cyclopropane-1 ,2- dicarboxylate
To a stirred solution of the azide (2.96 g, 13.9 mmol) in THF (20 mL) and MeOH (40 mL) at room temperature were added B0C2O (15.3 g, 69.4 mmol) and Pd(OH)2 (2.43 g, 3.47 mmol). After stirring under a hydrogen balloon for 3 hrs, the mixture was filtered through a pad of Celite and the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (ether/hexane = 20:80 - 60:40) to give N-Boc amine as a colorless oil (3.31 g, 83%): Rf = 0.33
(ether/hexane = 30:70); Ή NMR (300 MHz, Chloroform-d) δ 4.71 (bs, 1 H), 3.74 (s, 3H), 3.72 (s, 3H), 3.60 - 3.56 (m, 1 H), 3.39 - 3.27 (m, 1 H), 2.34 (dd, J = 9.2, 4.8 Hz, 1 H), 2.25 (dd, J = 5.4 Hz, 1 H), 2.13 (d, J = 7.5 Hz, 1 H), 1 .46 (s, 9H). Step e: Synthesis of dimethyl ester of INT-46. Dimethyl trans-3-({4-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanamido}methyl)cyclopropane-1 ,2- dicarboxylate (a pair of diastereomers)
The Boc amine (0.180 g, 0.627 mmol) was treated with 4NHCI in dioxane (4 mL) for 1 hour followed by removing excess HCI and dioxane. The amine HCI salt and INT-2 (0.330 g, 0.940 mmol) were dissolved in DMF 10 mL followed by adding DIPEA (0.16 mL, 0.94 mmol). HATU (0.357 g, 0.940 mmol) was added into the reaction mixture at once. After 1 hr, DMF was removed and the residue was loaded into reversed phase prep-HPLC (0-1 5%ACN/Water with 0.1 %TFA) to give 0.238 g (73%). Positive mass ion was found as (M + H+): 521 .2, tr = 2.53 minutes with 8 min (5-95%) method.
Step f : Synthesis of INT-46. Trans-3-({4-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanamido}methyl)cyclopropane-1 ,2- dicarboxylic acid (a pair of diastereomers)
Dimethyl ester of INT-46 (0.238 g, 0.457 mmol) was mixed with LiOH (0.0237 g, 0.960 mmol) in 6 mL of THF/Water (1 :1 ). Stirred for less than 1 hrs. 1 N HCI to acidify. Removed THF and extracted with EtOAC to give INT-46 (0.220 g, 97%). Positive/negative mass ions were found as (M + H+): 493.2, (M - H+): 491 .2, tr = 0.52 minutes with 5 min (5-40%) method.
Figure imgf000335_0001
INT-47 was prepared using a similar procedure as that for INT-16 except D-Dab is used in place of L-Dab. Positive mass ions were found as (M - Boc + 2H+)/2: 632.4 (M + H+): 1364.1 , tr = 1 .41 minutes with 5 min (40-95%) method. Example 136. Preparation of INT-48
Figure imgf000336_0001
INT-47 (0.904 g, 0.663 mmol), Z-L-2-amino-octanoic acid (0.214 g, 0.729 mmol) and DIPEA (0.17 ml_, 0.99 mmol) were dissolved in 10 mL of DMF. HATU (0.378 g, 0.995 mmol) was added as DMF solution (5 mL) via syringe pump. Stirred till the reaction completed. Added 5% Pd/C (0.423 g, 0.1 99 mmol) and stirred under hydrogen balloon for 3 hours. Removed DMF and purified with reversed phase prep-HPLC to give INT-48 (0.806 g, 81 %). Positive mass ions were found as (M - Boc + 2H+)/2: 703.0, tr = 4.72 minutes with 8 min (5-95%) method.
Example 137. Preparation of INT-49. Trans-3-{(1 E)-3-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-3-oxoprop-1-en-1-yl}cyclopropane-1 ,2-dicarboxylic acid and trans-3-{3-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-3- oxopropyl}cyclopropane-1 ,2-dicarboxylic acid (pair of trans-cyclopropyl diastereomers)
Figure imgf000336_0002
Step a: Synthesis of di-tert-butyl trans-3-[(1 E)-3-methoxy-3-oxoprop-1-en-1-yl]cyclopropane-1 ,2- dicarboxylate
The sulfonium salt (see INT-26 step a for procedure, 6.000 g, 24.88 mmol)) was added into a flask with 50 mL of THF at 0 °C. t-BuOK (1 M THF, 24.9 mL, 24.9 mmol) was added and stirred for 20 min, then at -78 °C, di-tert-butyl fumarate (4.982 g, 20.73 mmol) and 20 mL of THF were added respectively. The reaction mixture was stirred at -78 °C for 5 hrs and then let warm-up to room temperature. After the reaction was completed the reaction was quenched with brine (60 mL). The organic layer was separated and the aq. layer was extracted with ether twice. Dried and 0-15% Hex/EA silica column purification to give the desired product 0.983 g (15%). 13C NMR (75 MHz, Chloroform-d) δ 169.49, 167.78, 166.09, 143.1 1 , 123.53, 82.03, 81 .82, 77.47, 77.04, 76.62, 51 .45, 30.29, 29.91 , 29.57, 28.02, 27.99.
Step b: Synthesis of (2E)-3-[trans-2,3-bis(tert-butoxycarbonyl)cyclopropyl]prop-2-enoic acid
The product of Step a (0.212 g, 0.650 mmol) was mixed with LiOH (0.0400 g, 1 .63 mmol) in 6 mL of THF/Water (1 :1 ). The reaction mixture was stirred for less than three days. 1 N HCI was added to acidify the reaction mixture. Removed THF and extracted with EtOAC. Purified by reversed phase prep- HPLC to give the desired acid 0.170 g (83%). HPLC showed tr = 7.2 min for the product. Step c: Synthesis of di-tert-butyl ester of INT-49 and INT-49a. Di-tert-butyl trans-3-{(1 E)-3-[(2-{2- [(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-3-oxoprop-1-en-1-yl}cyclopropane- 1 ,2-dicarboxylate (mixture of diastereomers)
The acid (0.140 g, 0.448 mmol) and INT-1 (0.135 g, 0.538 mmol) were dissolved in 10 mL of DMF followed by adding DIPEA (0.26 mL, 0.67 mmol). HATU (0.256 g, 0.672 mmol) was added into the reaction solution at once. After 1 hr, DMF was removed and the residue was loaded into reversed phase prep-HPLC (0~15%ACN/Water with 0.1 %TFA) to give the di-tert-butyl ester of INT-49, 0.207 g (85%). Positive mass ion was found as (M + H+): 546.2, tr = 4.30 minutes with 8 min (5-95%) method.
Step d: Synthesis of INT-49. Trans-3-{(1 E)-3-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-3-oxoprop-1-en-1-yl}cyclopropane-1 ,2-dicarboxylic acid
The di-tert-butyl ester (0.200 g, 0.367 mmol) was mixed with 6 mL of TFA and stirred for less than 1 hrs. Removed TFA and purified reversed phase prep-HPLC to give INT-49 (mixture) (0.130 g, 83%). Positive/negative mass ions were found as (M + H+): 434.2, (M - H+): 432.2, tr = 2.1 1 minutes with 5 min (5-40%) method. The product was obtained as a mixture of trans-cyclopropyl dicarboxylate
diastereomers. Example 138. Preparation of INT-50
Figure imgf000338_0001
The synthesis of INT-50 was analogous to that for INT-20 except D-Dab was used in place of L- Dab in the synthesis. Positive mass ion was found as (M - 2Boc + 2H+)/2: 803.3, tr = 2.44 minutes with 5 min (40-95%) method.
Example 139. Preparation of INT-51 and INT-52
Figure imgf000338_0002
Step a: Hydrogenation of di-tert-butyl trans-3-[(1 E)-3-methoxy-3-oxoprop-1-en-1-yl]cyclopropane- 1 ,2-dicarboxylate
The olefin (see INT-49, Step a for a procedure, 2.00 g, 6.13 mmol) in 40 mL of THF was treated with 5% Pd/C. The mixture was degassed by bubbling argon through the solution and then it was stirred under a H2 atmosphere (balloon pressure), the reaction mixture was stirred for 2h. Celite filtration and solvent removal gave the clean desired product in 1 00% yield. 1 H NMR (300 MHz, Chloroform-d) δ 3.69 (s, 3H), 2.30 - 1 .95 (m, 2H), 2.15 (ddd, J = 9.5, 4.7, 0.7 Hz, 1 H), 2.09 - 1 .95 (m, 2H), 1 .94 - 1 .78 (m, 1 H), 1 .77 - 1 .63 (m, 1 H), 1 .48 (s, 9H), 1 .46 (s, 9H).
Step b: Synthesis of 3-[trans-2,3-bis(tert-butoxycarbonyl)cyclopropyl]propanoic acid Mixed the di-tert-butyl ester (2.012 g, 6.13 mmol) with LiOH (0.182 g, 7.35 mmol) in 32 mL of THF/Water (1 :1 mL). The mixture was stirred for about three days. 1 N HCI was added to acidify the reaction mixture. Removed THF and extracted with EtOAC to give the desired acid 1 .926 g (100%). 1 H NMR (300 MHz, Chloroform-d) δ 2.43 (t, J = 7.4 Hz, 2H), 2.17 (dd, J = 9.4, 4.7 Hz, 1 H), 2.14 - 1 .95 (m, 2H), 1 .96 - 1 .84 (m, 1 H), 1 .83 - 1 .68 (m, 1 H), 1 .48 (s, 9H), 1 .46 (s, 9H).
Step c: Synthesis of di-tert-butyl trans-3-{3-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-3-oxopropyl}cyclopropane-1 ,2-dicarboxylate (a pair of diastereomers)
The acid (1 .926 g, 6.13 mmol) and INT-1 (1 .693 g, 6.74 mmol) were dissolved in 10 mL of DMF followed by adding DIPEA (1 .60 mL, 9.1 9 mmol). HATU (3.494 g, 9.19 mmol) was added into the reaction solution at once. After 1 hr, DMF was removed and the residue was loaded into reversed phase prep- HPLC (0~15%ACN/Water with 0.1 %TFA) to give 2.002 g (60%). Positive mass ions were found as (M + H+): 548.2 and (M - CeHnOs + H+): 400.2, tr = 4.46 minutes with 8 min (5-95%) method.
Step d: Synthesis of trans-3-{3-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]- 3-oxopropyl}cyclopropane-1 ,2-dicarboxylic acid (a pair of diastereomers)
Mixed the di-tert-butyl ester (2.000 g, 3.65 mmol) with TFA (30 mL). Stirred for less than 1 hrs. Removed TFA followed by adding water and lyophilization to give the diacid (100%). Negative mass ion was found as (M - H+): 434.2, tr = 2.20 minutes with 5 min (5-40%) method.
Step e: Synthesis of INT-51 and INT-52
The diacid (1 .590 g, 3.652 mmol) and methyl (2S)-2-aminooctanoate HCI salt (1 .608 g, 7.668 mmol) were dissolved in 10 mL of DMF/DCM (1 :4) followed by adding NaHCC (1 .227 g, 14.61 mmol), HOAt (1 .242 g, 9.129 mmol) and EDCI (1 .750 g, 9.129 mmol). After 3hrs, the reaction mixture was diluted with EtOAc (100 mL) and washed with 1 N HCI, saturated NaHCCb and brine. Solvents were removed and the residue was loaded into reversed phase prep-HPLC (0-1 5%ACN/Water with 0.1 %TFA) to give INT- 51 as Peak-1 (polar) and INT-52 as Peak-2 (less polar). Positive/negative mass ions were found as (M + H+): 747.2 and (M - CeHnOs + H+): 600.4, (M - H+): 745.0, tr = 4.84 minutes with 8 min (5-95%) method for INT-51 and (M + H+): 747.2 and (M - CeHnOs + H+): 600.4, (M - H+): 745.0, tr = 5.37 minutes with 8 min (5-95%) method for INT-52. The stereochemistry of the trans-cyclopropane was arbitrarily assigned.
Example 140. Preparation of INT-53 N~1 ~,N~31 ~-bis(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)-4,7,10,13,19,22,25,28-octaoxa-16-azahentriacontane-1 ,31- diamide and INT-54 1-[(6-deoxy-alpha-L-mannopyranosyl)oxy]-22-{21-[(6-deoxy-alpha-L- mannopyranosyl)oxy]-15-oxo-3,6,9, 12,19-pentaoxa-16-azahenicosan-1 -yl}-7,23-dioxo- 3,10,13,16,19-pentaoxa-6,22-diazahexacosan-26-oic acid
Figure imgf000340_0001
Step a: Synthesis of 16-[(benzyloxy)carbonyl]-4,7,10,13,19,22,25,28-octaoxa-16-azahentriacontane- 1 ,31-dioic acid
NH-(PEG4-COOH (1 .441 g, 2.806 mmol) was dissolved in 1 5 mL of THF and 10%aq. Na2C03 (15 mL) and treated by CbzCI (0.62 mL, 4.2 mmol) by dropwise addition with ice-water bath cooling. After stirring overnight, the mixture was acidified with 1 N HCI and THF/water was removed followed by adding minimal amount of NMP/water. Prep-HPLC purification gave the desired Cbz protected diacid 1 .53 g, 84%. Positive/negative mass ions were found as (M + H+): 648.4, (M - H+): 646.2, tr = 3.92 minutes with 8 min (5-95%) method.
Step b: Synthesis of INT-53. N~1 ~,N~31 ~-bis(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)-4,7,10,13,19,22,25,28-octaoxa-16-azahentriacontane-1 ,31- diamide
The Cbz protected diacid (1 .53 g, 2.36 mmol), INT-1 (1 .247 g, 4.961 mmol), and DIPEA (1 .03 mL, 5.91 mmol) were mixed in DMF (20mL). HATU (2.245 g, 5.905 mmol) in DMF (13 mL) was pumped into the mixture in 4hrs. The reaction was stirred for another hour after the completion of HATU addition. DMF was removed and the residue was purified by prep-HPLC to give the Cbz protected INT-53, 2.142 g, 81 %. Positive mass ion was found as (M - 2C6H11 O5 - Bn + H+): 698.4, tr = 4.37 minutes with 8 min (5-95%) method. The Cbz protected INT-53 was dissolved in ethanol and treated with 5%Pd/C and hydrogen balloon for 2hrs. Celite filtration and solvent removal gave INT-53 1 .88 g, 100%. Positive mass ions were found as (M - 2C6Hn 04 + 2H+)/2: 344.6, (M - CeH C + 2H+)/2: 417.6.
Step c. Synthesis of INT-54
INT-53 (1 .156 g, 1 .18 mmol), succinic acid monobenzyl ester (0.2937 g, 1 .41 1 mmol), and DIPEA (0.61 mL, 3.5 mmol) were mixed in DMF (1 0 mL). HATU (0.671 g, 1 .76 mmol) in DMF (6 mL) was pumped into the mixture in 4hrs. The reaction was stirred for another 1 hr after the completion of HATU addition. DMF was removed and the residue was purified by prep-HPLC to give 1 .05 g, 76%. The benzyl ester was dissolved in ethanol and treated with 5% Pd/C and hydrogen balloon for 2hrs. Celite filtration and solvent removal gave INT-54 which was used for next step reaction without purification. Positive mass ions were found as (M - 2C6Hi i04 + 2H+)/2: 394.8, (M - C6Hii04 + 2H+)/2: 467.8, (M + 2H+)/2: 540.8. Example 141. Preparation of INT-55 and INT-56
Figure imgf000341_0001
Step a. Synthesis of INT-55. Methyl (4S,12S)-4,12-dihexyl-3,6,10-trioxo-2-oxa-5,8,11- triazatridecan-13-oate
2,2'-{[(benzyloxy)carbonyl]azanediyl}diacetic acid (0.9994 g, 3.740 mmol), methyl (2S)-2- aminooctanoate HCI salt (1 .647 g, 7.854 mmol), EDCI (1 .792 g, 9.350 mmol), HOAt (2.273 g, 9.350 mmol) and NaHCCb (1 .257 g, 14.96 mmol) were mixed in 20 mL of DCM/DMF (4:1 ). The reaction mixture was stirred for 3hrs. EtOAc was added into the reaction mixture to dilute followed by washing with 1 NHCI, saturated NaHC03 and brine wash. After removal of solvents the residue was loaded into normal phase prep-HPLC, 0.5~7%MeOH/DCM to purify. 2.008 g of the desired product (Cbz protected INT-55) was obtained, 93%. Rf = 0.6 7% MeOH/DCM. 1 H NMR (300 MHz, Chloroform-d) δ 8.25 (d, J = 7.6 Hz, 1 H), 7.42 (d, J = 7.9 Hz, 1 H), 7.39 - 7.22 (m, 5H), 5.08 - 5.20 (m, 2H), 4.65 - 4.52 (m, 1 H), 4.51 - 4.39 (m, 1 H), 4.00 (d, J = 5.5 Hz, 4H), 3.72 (s, 3H), 3.67 (s, 3H), 1 .58 - 1 .90 (m, 4H), 1 .40 - 1 .15 (m, 16H), 0.87 (t, J = 19.7 Hz, 6H, 2CH3). 13C NMR (75 MHz, CDC ) δ 172.99, 172.90, 169.35, 169.28, 155.85, 135.97, 128.44, 128.05, 127.66, 67.91 , 53.32, 53.20, 52.57, 52.55, 52.53, 52.27, 52.13, 32.10, 31 .68, 31 .54, 28.81 , 25.47, 25.25, 22.53, 14.02. The Cbz protected INT-55 was dissolved in ethanol and treated with 5%Pd/C and hydrogen balloon for 2hrs. Celite filtration and solvent removal gave INT-55 used for next step reaction without purification. Positive/negative mass ions were found as (M + H+): 444.4, (M - H+): 442.2, tr = 3.95 minutes with 8 min (5-95%) method.
Step b. Synthesis of INT-56
INT-54 (0.26 g, 0.228 mmol), INT-55 (0.127 g, 0.228 mmol), and DIPEA (0.079 mL, 0.46 mmol) were mixed in 5 mL of DMF. HATU was added via syringe pump (2 mL, 1 mL/hr). The reaction solution was stirred for another hour. DMF was removed and the residue was purified by prep-HPLC to give 0.224 g, 65%. Positive mass ions were found as (M - 2 CeH C + 2H+)/2: 607.6, (M - CeH C + 2H+)/2: 680.4, (M + 2H+)/2: 753.6, tr = 2.1 0 minutes with 5 min (40-95%) method. Example 142. Preparation of INT-57 (15S)-11-(2-{[(1S)-1-carboxyheptyl]amino}-2-oxoethyl)-1-[(6- deoxy-alpha-L-mannopyranosyl)thio]-15-hexyl-7,10,13-trioxo-3-oxa-6,11 ,14-triazahexadecan-16-oic acid
Figure imgf000342_0001
Per-acetated INT-9 (0.0730 g, 1 .48 mmol), INT-55 (0.0656 g, 0.148 mmol) and DIPEA (0.052 mL, 0.30 mmol) were mixed in 4 mL of DMF. HATU (0.0844 g, 0.222 mmol) was added via syringe pump (1 mL, 1 mL/hr). The reaction solution was stirred for another hour after completion of addition. DMF was removed and the residue was purified by prep-HPLC to give Per-acetated dimethyl ester of INT-57, 0.083 g, 61 %. Positive mass ion was found as (M - CeH C + H+): 647.0, tr = 3.46 minutes with 5 min (40-95%) method. Per-acetated dimethyl ester of INT-57 (0.083 g, 0.090 mmol) was dissolved in 6 mL of THF/water (1 :1 ) and treated with LiOH (0.01 1 g, 0.46 mmol). After acidification and ethyl acetate extraction, clean INT-57 was obtained in 100%. Positive/negative mass ions were found as (M + H+): 765.4, (M - H+): 763.4, tr = 4.10 minutes with 8 min (5-95%) method.
Example 143. Preparation of INT-58 and INT-59
Figure imgf000342_0002
Step a. Synthesis of INT-58
The Cbz protected diacid (see INT-53, Step a for procedure, 0.200 g, 0.309 mmol), INT-53 (0.6355 g, 0.6484 mmol), and DIPEA (0.22 ml_, 1 .2 mmol) were mixed in DMF (1 OmL). HATU (0.294 g, 0.772 mmol) in DMF (3 mL) was pumped into the mixture in 4hrs. The reaction was stirred for another hour after the completion of HATU addition. DMF was removed and the residue was purified by prep-
HPLC to give the Cbz protected INT-58, 0.660 g, 83%. Positive mass ions were found as (M - 4CeHn04 + 3H+)/3: 662.9, (M - 3C6Hii04 + 3H+)/3: 71 1 .4, (M - 2C6Hii04 + 3H+)/3: 760.3, (M - CeHnC + 3H+)/3: 809.0, tr = 3.28 minutes with 8 min (5-95%) method. The Cbz protected INT-58 was dissolved in ethanol and treated with 5%Pd/C and hydrogen balloon for 2hrs. Celite filtration and solvent removal gave INT-58 1 .88 g, 1 00%. Positive mass ions were found as (M - CeH C + 2H+)/2: 667.0, (M - 2C6Hn04 + 3H+)/3: 716.0, (M - C6Hii04 + 3H+)/3: 764.6, (M + 3H+)/3: 813.4, (M - CeHnC + 2H+)/2: 1 146.4, (M + 2H+)/2: 1219.2.
Step b. Synthesis of 4-(bis{2-[(1-methoxy-1-oxooctan-2-yl)amino]-2-oxoethyl}amino)-4- oxobutanoic acid
Succinic acid monobenzyl ester (0.568 g, 2.73 mmol), INT-55 (1 .10 g, 2.48 mmol) and DIPEA (1 .30 mL, 7.44 mmol) were mixed in 20 mL of DMF. HATU (1 .414 g, 3.72 mmol) was added via syringe pump (10 mL, 2mL/hr). The reaction solution was stirred for another hour after the completion of addition. DMF was removed and the residue was purified by prep-HPLC to give dimethyl benzyl triester, 1 .245 g, 79%. Positive mass ion was found as (M + H+): 634.4, tr = 2.85 minutes with 7 min (60-95%) method.
Dimethyl benzyl triester (1 .245 g, 1 .964 mmol) was dissolved in ethanol and treated with 5% Pd/C and H2 balloon for 2 hrs. Celite filtration gave the desired acid in 1 00%. Positive mass ion was found as (M + H+): 544.0, tr = 3.01 minutes with 5 min (40-95%) method. Step c. Synthesis of INT-59
The acid from Step b (0.0950 g, 0.175 mmol), INT-58 (0.387 g, 0.159 mmol) and DIPEA (0.083 mL, 0.47 mmol) were mixed in 4 mL of DMF. HATU (0.0901 g, 0.238 mmol) was added via syringe pump (2 mL, 1 mL/hr). The reaction solution was stirred for another hour after the completion of addition. DMF was removed and the residue was purified by prep-HPLC to give dimethyl ester of INT-59, 0.31 8 g, 66%. Positive mass ions were found as (M - 4C6Hn04 + 3H+)/3: 793.4, (M - 3CeHn04 + 3H+)/3: 842.3, (M -
2C6Hii04 + 3H+)/3: 890.3, (M - CeHnC + 3H+)/3: 939.6, (M + 3H+)/3: 987.9, tr = 4.18 minutes with 8 min (5-95%) method. Dimethyl ester of INT-59 (0.318 g, 0.107 mmol) was dissolved in 8 mL of THF/water (1 :1 ) and treated with LiOH (0.0054 g, 0.23 mmol). After acidification THF was removed and 3mL of NMP was added. Then most of Water was removed and the residue was load to pep-HPLC to purify. 0.289 g of INT-59 was obtained (92%). Positive mass ions were found as (M - 3C6Hn04 + 4H+)/4: 624.8, (M
4C6Hii04 + 3H+)/3: 784.0, (M - 3C6Hi i04 + 3H+)/3: 832.8, (M - 2C6Hii04 + 3H+)/3: 881 .8, (M - CeHnC + 3H+)/3: 929.8, (M + 3H+)/3: 979.8, tr = 3.80 minutes with 8 min (5-95%) method. Example 144. Preparation of INT-60. 2,2'-{[(2-{3-[(3,4,5-Trihydroxy-6-methyloxan-2- yl)oxy]propanamido}acetamido)acetyl]azanediyl}diacetic acid
Figure imgf000344_0001
Step a. Synthesis of 3-[(2,3,4-tri-0-acetyl-6-deoxyhexopyranosyl)oxy]propanoic acid
To a solution of the per-acetated bromo-L-rhamnopyranose (1 .600 g, 4.531 mmol) and benzyl 3- hydroxypropanoate (0.6280 g, 3.485 mmol) in 50 mL of dry CH2CI2 at room temperature were added silver triflate (1 .075 g, 4.182mmol) and 4A molecular sieves (12 g). After the mixture was stirred for 2 hrs. at room temperature. Two Grams of Celite was added into the reaction mixture followed by filtration. The filtrate was condensed and purified with 1 -5% MeOH/DCM to give the desired product 0.853 g, 54%. 1 H NMR (300 MHz, CDCI3) δ 7.36, 7.35, 7.34, 7.32, 7.31 , 7.31 , 7.29, 7.29, 7.28, 5.25, 5.24, 5.22, 5.21 , 5.20, 5.20, 5.1 9, 5.14, 5.08, 5.06, 5.04, 5.03, 5.00, 4.73, 4.73, 4.02, 4.00, 3.99, 3.98, 3.97, 3.95, 3.92, 3.90, 3.89, 3.88, 3.87, 3.86, 3.85, 3.83, 3.73, 3.71 , 3.70, 3.69, 3.67, 3.66, 3.44, 2.67, 2.65, 2.63, 2.13, 2.12, 2.04, 2.02, 1 .98, 1 .96, 1 .30, 1 .20, 1 .18. 13C NMR (75 MHz, CDC ) δ 170.89, 170.01 , 169.95, 169.87, 135.68, 128.56, 128.24, 128.21 , 97.60, 77.57, 77.1 5, 76.72, 71 .00, 69.70, 69.06, 66.51 , 66.44, 63.37, 34.59, 20.84, 20.76, 20.67, 17.34. The benzyl ester was hydrogenated (5% Pd/C, H2 balloon) to the free acid in quantitative yield.
Step b. Synthesis of N-{3-[(2,3,4-tri-0-acetyl-6-deoxyhexopyranosyl)oxy]propanoyl}glycylglycine The propanoic acid (from Step a, 0.6830 g, 1 .885 mmol), NH2-Gly-Gly-OBn HCI salt (0.4877 g,
1 .885 mmol), EDCI (0.5420 g, 2.828 mmol), HOAt (0.3849 g, 2.828 mmol) and NaHCOs (0.6334 g, 7.540 mmol) were mixed in 1 0 mL of (DCM/DMF (4:1 ). The reaction solution was stirred overnight. The reaction mixture was diluted with EtOAc and washed with 1 NHCI, saturated NaHCCb and brine. Reversed phase prep-HPLC purification gave the desired product 1 .020 g, 95%. Positive mass ion was found as (M + H+): 567.0, tr = 4.23 minutes with 8 min (5-95%) method. The benzyl ester was hydrogenated (5% Pd/C, H2 balloon) to the free acid in quantitative yield.
Step c. Synthesis of INT-60
The acid (from Step b, 0.8410 g, 1 .782 mmol), dimethyl 2,2'-azanediyldiacetate (0.3837 g, 1 .942 mmol) and DIPEA (0.46 mL, 2.6 mmol) were mixed in 10 mL of DMF. To the solution, was added HATU (1 .007 g, 2.648 mmol) in DMF (5 mL) with syringe pump at 1 mL/min. The reaction solution was stirred for another hour after completion of addition. Reverse phase prep-HPLC purification gave the desired product, dimethyl ester of INT-60, 1 .016 g, 93%. Positive mass ion was found (M + H+): 61 9.8, tr = 3.36 minutes with 8 min (5-95%) method. Dimethyl ester of INT-60 (1 .016 g, 1 .640 mmol) was dissolved in 20 ml_ of THF/water (1 :1 ) and treated with LiOH (0.3927 g, 16.40 mmol). After acidification THF was removed and 3ml_ of NMP was added. Then most of Water was removed and the residue was load to pep-HPLC to purify. 0.567 g of INT-60 was obtained (74%). Negative mass ions were found as (M - C6Hi i 04 - H+): 302.0, (M - H+): 464.0, tr = 0.80 minutes with 5 min (5-60%) method.
Example 145. Preparation of INT-61. 2,2'-{[(2-{(R)-3-[(3,4,5-Trihydroxy-6-methyloxan-2- yl)oxy]butanamido}acetamido)acetyl]azanediyl}diacetic acid
Figure imgf000345_0001
Step a: Synthesis of (R)-3-[(2,3,4-tri-0-acetyl-6-deoxyhexopyranosyl)oxy]butanoic acid
To a solution of the per-acetated bromo-L-rhamnopyranose (1 .900 g, 5.380 mmol) and benzyl (R)-3-hydroxybutanoate (0.8708 g, 4.483 mmol) in 80 mL of dry CH2CI2 at room temperature were added silver trifiate (1 .256 g, 4.887 mmol) and 4A molecular sieves (20 g). After the mixture was stirred for 2 hrs. at room temperature. 2 gram of Celite was added into the reaction mixture followed by filtration. The filtrate was condensed and purified with 1 -5% MeOH/DCM to give the desired product 1 .018 g, 49%.1 H NMR (300 MHz, CDCI3) 7.33 (s, 2H), 7.38 - 7.23 (m, 2H), 5.35 - 5.08 (m, 4H), 5.02 (t, J = 9.9 Hz, 1 H), 4.84 (d, J = 1 .8 Hz, 1 H), 4.34 - 4.17 (m, 1 H), 3.98 - 3.82 (m, 1 H), 2.65 (dd, J = 15.5, 8.0 Hz, 1 H), 2.47
(dd, J = 15.5, 4.9 Hz, 1 H), 2.14 - 1 .92 (m, 7H), 1 .17 (dd, J = 10.1 , 6.2 Hz, 5H). 13C NMR (75 MHz, CDCI3) δ 170.83, 170.12, 1 69.96, 1 69.86, 135.67, 128.53, 128.19, 94.73, 77.61 , 77.18, 76.76, 71 .03, 70.30, 69.48, 69.10, 66.62, 66.39, 42.00, 20.85, 20.66, 18.84, 17.26.The benzyl ester was hydrogenated (5% Pd/C, H2 balloon) to the free acid in quantitative yield.
Step b. Synthesis of N-{(R)-3-[(2,3,4-tri-0-acetyl-6-deoxyhexopyranosyl)oxy]butanoyl}glycylglycine
The (R)-3-butanioc acid (from Step a, 0.8120 g, 2.1 58 mmol), NH2-Gly-Gly-OBn HCI salt (0.5582 g, 2.158 mmol), EDCI (0.6204 g, 3.236 mmol), HOAt (0.4405 g, 3.236 mmol) and NaHCOs (0.7250 g, 8.630 mmol) were mixed in 10 mL of (DCM/DMF (4:1 ). The reaction solution was stirred overnight. The reaction mixture was diluted with EtOAc and washed with 1 NHCI, saturated NaHCCb and brine. Reverse phase prep-HPLC purification gave the desired product 1 .224 g, 98%. Positive mass ion was found as (M + H+): 581 .2, tr = 4.38 minutes with 8 min (5-95%) method. The benzyl ester was hydrogenated (5% Pd/C, H2 balloon) to the free acid 1 .01 1 g, 98%. Positive/negative mass ions were found as (M + H+): 491 .2, (M - H+): 489.2, tr = 2.45 minutes with 5 min (5-95%) method. Step c. Synthesis of INT-61
The acid (from Step b, 1 .01 1 g, 2.061 mmol), dimethyl 2,2'-azanediyldiacetate (0.4481 g, 2.268 mmol) and DIPEA (0.54 mL, 3.1 mmol) were mixed in 10 mL of DMF. To the solution, was added HATU (1 .176 g, 3.092 mmol) in DMF (5 mL) with syringe pump at 1 mL/min. The reaction solution was stirred for another hour after completion of addition. Reverse phase Prep-HPLC purification gave the desired product, dimethyl ester of INT-61 , 1 .173 g, 90%. Positive mass ion was found (M + H+): 633.8, tr = 3.45 minutes with 8 min (5-95%) method. Dimethyl ester of INT-61 (1 .173 g, 1 .851 mmol) was dissolved in 20 mL of THF/water (1 :1 ) and treated with LiOH (0.4434 g, 18.51 mmol). After acidification THF was removed and 3mL of NMP was added. Then most of Water was removed and the residue was load to pep-HPLC to purify. 0.773 g of INT-61 was obtained (87%). Negative mass ions were found as (M - H+): 478.0, tr = 0.60 minutes with 5 min (5-60%) method.
Example 146. Preparation of INT-62
Figure imgf000346_0001
INT-62
INT-62 was prepared from INT-1 1 and Z-D-Ser-OH in an analogous manner as described in Example 14. Yield: 81 %. LC/MS ((M+2H)/2) = 525.6 (loss of 1 Boc group).
Step a. Synthesis of the Protected Octapeptide
A solution of HATU (1 .97 g, 5.18 mmol) in DMF (9 mL) was added via a syringe pump at a rate of 2.5 mL/hr into a solution of INT-1 1 (5.0 g, 4.71 mmol), Z-D-Ser-OH (1 .24 g mg, 5.18 mmol),
diisopropylethylamine (1 .82 g, 14.12 mmol) in DMF (35.5 mL). After the addition, the reaction mixture was stirred for an additional hour then purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5 to 35 to100% methanol and water to yield the desired compound (6.05 g, 1 00%). Ion found by LC/MS [(M-2Boc+2H)/2]+ = 542.4. Step b. Synthesis of INT-62
To a solution of the product from Step a. in methanol (31 mL) was charged with 5% Pd/C (94.2 mg) and H2 gas balloon. The reaction was stirred at room temperature overnight. Next day, the reaction mixture was filtered through a pad of Celite ® and washed with methanol then concentrated. The residue was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5 to 78% methanol and water to yield the title compound as a free base (2.61 g, 48%). Ion found by LC/MS [(M-2Boc+2Na)/2] = 497.6.
Example 147. Preparation of INT-63
Figure imgf000347_0001
INT-63 was prepared from INT-62 and Z-Thr-OH in an analogous manner as described i e 16 (Procedure A). Yield: 66%. LC/MS ((M+2HV2) = 525.8 (loss of 2 Boc groups).
Example 148. Preparation of INT-64
Figure imgf000347_0002
Step a. Synthesis of the Protected Dipeptide
The synthesis of the protected dipeptide was accomplished by a similar procedure using L- threonine methyl ester hydrochloride (1 .0 equiv.), Z-Dab(Boc)-OH dcha (1 .05 equiv.), HOBt (1 .5 equiv.), EDCI (1 .5 equiv.), and NaHCCb (2.0 equiv.) to yield the methyl ester. Hydrolysis of the methyl ester with LiOH (3.0 equiv.) in MeOH:THF:H20 (1 :1 :2) to yielded the desired compound (51 %). Ion found by LC/MS [M-H]+ = 452.2. Step b. Coupling of the Dipeptide to INT-62
A solution of HATU (218.34 mg, 0.57 mmol) in DMF (1 mL) was added via a syringe pump at a rate of 2.5 mL/hr into a solution of 836-INT-B (0.6 g, 0.52 mmol), 836-INT-E (260.41 g mg, 0.57 mmol), diisopropylethylamine (202.25 mg, 1 .57 mmol) in DMF (6 mL). After the addition, the reaction mixture was stirred for an additional hour then purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5 to 35 to 100% methanol and water, using 0.1 % TFA as modifier to yield the title compound (577.0 mg, 66%). Ion found by LC/MS [(M-2Boc+2H)/2]+ = 694.5.
Step c. Synthesis of INT-64
A solution of the Step-b Product (51 1 .0 mg, 303.09 mmol) in MeOH (6 mL) was charged with 5% Pd/C (96.76 mg, 0.045 mmol) and H2 gas balloon. The reaction mixture was stirred at room temperature overnight. After the reaction was complete, it was filtered through a pad of Celite®, washed with methanol then concentrated. The residue was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5 to 30 to 100% methanol and water to yield INT-64 as the free base (191 .0 mg, 43%). Ion found by LC/MS [(M-3Boc+3H)/3]+ = 385.4.
Example 149. Preparation of INT-65
Figure imgf000348_0001
INT-65 was prepared from INT-64 and Z-L-Norleu-OH in an analogous manner as described for INT-62. Yield: 67%. LC/MS [M+2H/2] 682.6 (loss of 2 Boc groups).
xample 150. Preparation of INT-66
Figure imgf000349_0001
derived from more polar intermediate
Note: The absolute configuration of the trans-pyrrolidne dicarboxamide is arbitrarily assigned but the product is derived from the more polar trans-cyclopentane-(bis-octanoate methyl ester)
Step a. Synthesis of dimethyl (3R,4R)-1-benzylpyrrolidine-3,4-dicarboxylate
To a solution of dimethyl fumarate (1 .44 g, 10 mmol) in acetonitrile (40 mL) was added N- (methyoxymethyl)-N-(trimethylsilylmethyl)benzylamine (2.37 g, 10 mmol) followed by lithium fluoride (410 mg, 15.8 mmol). After the mixture was stirred for 1 .5 days, it was concentrated by rotary evaporation. The residue was purified by RPLC (150 g, 0 to 35 % acetonitrile and water, using 0.1 % TFA as modifier). The collected was concentrated by rotary evaporation and further dried in high vacuum to afford the product as a yellow oil TFA salt. Yield 3.0 g, 76.7 %. Ion found by LCMS: [M + H]+ = 278.
Step b. Synthesis of (3R,4R)-1-benzylpyrrolidine-3,4-dicarboxylic acid
The step-a product (3 g, 7.67 mmol) was dissolved in 1 :1 mixture of MeOH/THF (16 mL). The solution was cooled in an ice-water bath and added with a solution of LiOH (384 mg, 15.3 mmol) in water (10 mL). After the reaction mixture was stirred overnight, it was added with an additional amount of LiOH (300 mg) in water (3 mL) and continued at ~ 5-10 °C for 6 hours. 4N HCI solution in dioxane (3 mL) was slowly added to adjust pH to ~ 6. The organic solvents was removed by rotary evaporation, and the aqueous solution was purified by RPLC (150 g, 0 to 50 % acetonitrile and water). Yield 1 .57 g, 82 %. Ion found by LCMS: [M + H]+ = 250. Step c. Synthesis of dimethyl (2S,2'S)-2,2'-{[(3R,4R)-1-benzylpyrrolidine-3,4- diyl]bis(carbonylazanediyl)}dioctanoate
A mixture of step-b product (498.6 mg, 2 mmol) and methyl (2S)-2-aminooctanoate HCI (922.7 mg, 4.4 mmol) was dissolved in anhydrous DMF (4 mL) and DIPEA (1 .69 g, 13 mmol). The resulting mixture was added with a solution of HATU (1 .67 g, 4.4 mmol) in DMF (5 mL) via syringe pump at a rate of 5 ml/hr. After the addition, the reaction was directly purified by RPLC (1 50 g, 10 to 70 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 540 mg of the desired product as more polar isomer, 48.2 %. Ion found by LCMS: [M + H]+ = 560.
Step d. Removal of the benzyl protection
The step-c more polar isomer (540 mg, 0.96 mmol) was dissolved in MeOH. The solution was added with Pd(OH)2 and stirred under hydrogen for 20 hours. Pd(OH)2 was filtered off, and the filtrate was concentrated to dryness. Yield 450 mg, 99.8 %. Ion found by LCMS: [M + H]+ = 470.
Step e. Synthesis of dimethyl (2S,2'S)-2,2'-{[(3/?,4/¾-1-{4-[(2-{2-[(6-deoxy-a-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoyl}pyrrolidine-3,4- diyl]bis(carbonylazanediyl)}dioctanoate
A mixture of the step-d product (450 mg, 0.958 mmol) and INT-2 (405.5 mg, 1 .15 mmol) was dissolved in anhydrous DMF (2 mL) and DIPEA (390 mg, 3 mmol). After the solution was cooled in an ice- water bath, it was drop-wise added with a solution of HATU (437 mg, 1 .15 mmol) in DMF (1 mL) at a rate of 2 ml/hr. After the addition, the reaction was stirred for 30 more minutes and purified by RPLC (150 g, 10 to 70 % acetonitrile and water). Yield 250 mg, 32.4 %. Ion found by LCMS: [M + H]+ = 803. Step f. Synthesis of INT-66
The step-e product (250 mg, 0.31 mmol) was dissolved in a 1 :1 mixture of MeOH:THF (4 mL). After the solution was cooled in an ice-water bath, it was added with a solution of LiOH (32 mg, 1 .34 mmol)) in water (2 mL) by three portions over 1 .5 hours and stirred for one more hour. It was then acidified by 4N HCI solution in dioxane (0.3 mL) and purified by RPLC (50 g, 5 to 55 % acetonitrile and water, using 0.1 % TFA as modifier). The yield of INT-66 was 171 mg, 68.1 %. Ion found LCMS: [M + H]+ = 775.
Figure imgf000351_0001
The dicarboxylic acid INT-26 (0.042 g, 0. 85 mmol) and INT-45 (0.266 g, 0.1 70 mmol) were dissolved in 5 mL of DMF followed by addition of DI PEA (0.037 ml_, 0.21 mmol). To this mixture was added HATU (0.0808 g, 0.213 mmol) in 1 .5 mL of DMF via syringe pump over 2 hours at room temperature. After completion of HATU addition, the reaction mixture was stirred for another hour at room temperature. Then the reaction mixture was concentrated under reduced pressure. The residue was purified by reversed phase prep-HPLC with ACN/water (0.1 %TFA) to give the per-Boc-Compound 61 as Peak-1 (polar) and per-Boc-Compound 62 as Peak-2 (less polar). The mass spectrum showed strong detectable positive charge signals at tr = 2.19 and 2.43 min with 7 min (60-95%) method [found positive ion (M - 5Boc - t-Bu -C6H11 O5 + 4H+)/4: 738.0]. The Boc groups of Peak-1 and Peak-2 were removed (each compound individually) by treating with TFA for about an 1 hour. Most of the TFA was removed and the residue was dissolved in a minimum amount of NMP and loaded onto a Preparative HPLC with ACN/water (0.1 % TFA). The desired material (either Compound 61 or Compound 62) was collected and lyophilized to give the product as a white powder of TFA salt. Positive mass ion was found as [(M - C6H11 O5 + 5H+)/5: 465.1 ] for both Compound 61 and Compound 62. The stereochemistry of the trans- cyclopropyl dicarboxylate was arbitrarily assigned.
Figure imgf000352_0001
Step a. Preparation of dibenzyl 2,2'-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]
ethoxy}ethyl)azanediyl]diacetate
Benzyl bromo-acetate (4.0g, 17.4 mmol) was added into the solution INT-1 . L-Rhamnose-PEG1 - NH2 (1 .75 g, 7 mmol) and DIPEA (3.6 mL, 30 mmol) in 60 mL DMF, the resulting solution was stirred at room temperature overnight. The reaction solution was concentrated and purified by flash
chromatography to provide the desired product. Yield of oil product, LC/MS, 548.2 [M+H]+.
Step b. Preparation of 2,2'-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]-ethoxy}ethyl)- azanediyl]diacetic acid
Dibenzyl 2,2'-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy] ethoxy}ethyl)azanediyl] diacetate (1 .0g, 2 mmol) was dissolved into 10 mL MeOH and 10 mL ethyl acetate, then 200 mg of 5% palladium on charcoal was added above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere overnight. The palladium charcoal was removed by filtration after completed the reaction by LCMS. The filtrate was concentrated and used next step without any purification. Yield of oil product, LC/MS, 368.2 [M+H]+ .
Step c. Preparation of per-Boc-Compound 63
Standard procedure A: To the solution of 2,2'-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]- ethoxy}ethyl)-azanediyl]diacetic acid (40 mg, 0.1 mmol), triethyl amine (0.07 mL, 0.5 mmol) and INT-18 (320 mg, 0.2 mmol) in 5 mL DMF was added HATU (78 mg, 0.2 mmol). The reaction was stirred for 1 hr and then the resulting solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. Yield of product (per-Boc-Compound 63), 220 mg, 65% yield.
Step d. Preparation of Compound 63
Standard procedure B: Per-Boc-Compound 63 from the previous step was treated in 2 mL
DCM and 2 ml_ trifluoroacetic acid at room temperature, the solution was stirred 1 0 min, then concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using trifluoroacetic acid as the modifier. Yield 0.120 g, 67% yield, lon(s) found by LCMS: [M+2H]/2 = 1229.2, [M+3H]/3 = 819.8,
[M+4H]/4 = 615.1 , [M+5H]/5 =492.3.
Example 153: Synthesis of Compound 64
Figure imgf000353_0001
Step a. O-Alkylation of S-Hydroxy-I .S-Benzenedicarboxyiic Acid Dimethyl Ester and Removal of the Cbz Group
Benzyl (2-bromoethyl)carbamate ( .5 g, 6.0 mmol), 5-hydroxy-l ,3-benzenedicarboxylic acid dimethyl ester (1 .0 g, 4.8 mmol), and cesium carbonate (2.0 g, 6 mmol) were stirred in DMF (10 mL) at 60°C for 4 hours. The mixture was diluted with D! water (200 mL) and extracted into ethyl acetate (75 mL, 3x). The combined organic extracts were dried over sodium sulfate and concentrated. The residue was purified by Silica gel chromatography (hexanes/ethyi acetate) to afford 1 .4 g of the Cbz protected intermediate as a white solid. The Cbz-protected intermediate was stirred in methanol (100 mL) in the presence of 5% Pd/C (200 mg) under 1 atmosphere of hydrogen for 2 hours. The mixture was filtered through ceiiie and concentrated to afford 1 .1 g of the product as a white solid. Yield: 83 %. LC/MS [ +H] = 254.4. Step b. Synthesis of the Hemi-succinate
Succinic anhydride (0.6 g, 6 mmol) and the Step-a product (1 .1 g, 4.3 mmol) were stirred in methanol (20 mL) for 2 hours then concentrated. The mixture was acidified with glacial acetic acid and concentrated. The residue was purified by RPLC (50 g, 5 % to 95% acetonitrile and water using 0.1 % TFA modifier). The pure fractions were pooled and lyophilized to afford 1 g of the product as a white solid. Yield: 67%. LC/MS [M+H]+ = 354.0.
Step c. Coupling to INT-1
HATU (0.75 g, 2 mmol) in DMF (2 mL) was added, dropwise over a period of 20 minutes, to a stirring mixture of the Step-b product (0.63 g, 1 .8 mmol), INT-1 (0.45g, 1 .8 mmol), and triethylamine (0.36 g, 3.6 mmol) in DMF (4 mL) and then stirred for an additional 20 minutes. The mixture was applied to RPLC (150 g, 5 % to 95% acetonitrile and water using 0.1 % TFA modifier). The pure fractions were pooled and lyophilized and then stirred for 30 minutes in a 1 /1 /2 mixture of THF/methanol/DI water containing LiOH (1 50 mg, 6 mmol). The mixture was acidified with glacial acetic acid and concentrated. The residue was purified by RPLC (50 g, 1 0 % to 95% acetonitrile and water using 0.1 % TFA modifier). The pure fractions were pooled and lyophilized to afford 625 mg of the product. Yield: 61 %. LC/MS
[M+H]+ = 559.6.
Step d. Synthesis of Compound 64
HATU (42 mg, 0.1 1 mmol), in DMF (1 mL) was added, dropwise over 20 minutes, to a stirring mixture of INT-18 (192, mg, 0.1 1 mmol), the Step-c product (30 mg, 0.054 mmol), and triethylamine (32 mg, 0.31 mmol) in DMF (2 mL). The reaction was stirred for 30 additional minutes and applied to RPLC (50 g, 30 % to 95% methanol and water using 0.1 % TFA modifier). The pure fractions were concentrated and then stirred in a 1 /1 mixture of TFA/DCM (5 mL) at ambient temperature for 20 minutes. The solvent was removed by rotovap and the residue was purified by RPLC (50 g, 0 % to 75% acetonitrile and water using 0.1 % TFA modifier). The pure fraction were pooled and lyophilized to afford the product as a white solid. Yield: 67%, 2 steps. LC/MS [M+3H/3] = 977.9. Example 154: Synthesis of Compound 65
Figure imgf000355_0001
Step a. Synthesis of the Dipeptide
HATU (2.89 g, 7.50mmol), in DMF (4 mL) was added, dropwise over 30 minutes, to a stirring mixture of Cbz-S-1 -amino-octanoic acid (2.00 g, 6.82 mmol), L-DAB-methyl ester hydrochloride (1 .75 g, 7.50 mmol), and triethylamine (2.07 mg, 20.5 mmol) in DMF (6 mL) and the reaction was stirred for an additional 30 minutes. The mixture was applied directly to RPLC (150 g, 15 % to 95% acetonitrile and water using 0.1 % TFA modifier). The pure fractions were pooled and lyophilized and then taken up in methanol (150 mL), 5% Pd/C (400 mg) was added and the mixture was stirred under an atmosphere of hydrogen for 12 hours. The mixture was filtered through Celite and concentrated to afford 2.2 g of the product as a white solid. Yield: 86%. LC/MS [M+H]+ = 374.6.
Step b. Coupling to 2,2'-{[(benzyloxy)carbonyl]azanediyl}diacetic Acid
The Step-b product was prepared from 2,2'-{[(benzyloxy)carbonyl]azanediyl}diacetic acid and the Step-a product in a similar procedure as described in Step a. Yield: 86%. LC/MS [M+H]+ = 844.6 .
Step c. Coupling to INT-2 and Methyl Ester Hydrolysis
HATU (406 mg, 1 .07 mmol), in DMF (2 mL) was added, dropwise over 30 minutes, to a stirring mixture of the Step-b product (820 mg, 0.97 mmol), INT-2 (375 mg, 1 .07 mmol), and triethylamine (294 mg, 2.91 mmol) in DMF (3 mL) and the reaction was stirred for an additional 30 minutes. The mixture was applied directly to RPLC (150 g, 15 % to 95% acetonitrile and water using 0.1 % TFA modifier). The pure fractions were pooled and lyophilized and then stirred for 30 minutes in a 1 /1 /2 mixture of
THF/methanol/DI water containing LiOH (1 16 mg, 4.86 mmol). The mixture was acidified with glacial acetic acid and concentrated. The residue was purified by RPLC (150 g, 10 % to 95% acetonitrile and water using 0.1 % TFA modifier). The pure fractions were pooled and lyophilized to afford 350 mg of the product. Yield: 86%. LC/MS [M-2boc+2H/2]+ = 475.4.
Step d. Preparation of Compound 65
The title compound was prepared from INT-63 and the Step-c product in a similar manner as described in the procedure for the preparation of Compound 64. Yield: 39%. LC/MS [M+3H/3]+ = 938.5.
Figure imgf000356_0001
The title compound was prepared analogously using standard procedure A and B of Example 152. lon(s) found by LCMS [M+3H/3] = 913.9, [M+4H/4] =685.6, [M+5H/5] =548.7. [M+6H/6] =457.4.
Example 156: Synthesis of Compound 67
Figure imgf000356_0002
The title compound was prepared analogously to Example 37, where INT-20 was prepared with D-aminooctanoic acid in place of L-aminooctanoic acid in that sequence of reactions, lon(s) found by LCMS: (M+3H)/3 = 946.9, (M+4H)/4 = 710.4, (M+5H)/5 = 568.5, (M+6H)/6 = 473.9
Example 157: Synthesis of Compound 68
Figure imgf000357_0001
Step a. Preparation of diethyl 2,2'-{[2-({2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethyl}amino)-2- oxoethyl]azanediyl}diacetate
To the solution of [bis(2-ethoxy-2-oxoethyl)amino]acetic acid (0.41 g, 2 mmol), 2-aminoethyl 6- deoxy-alpha-L-mannopyranoside (0.5 g, 2 mmol) in DMF (20 mL) was added EDC (400 mg, 2 mmol), HOBT (300 mg, 2 mmol), Hunig's base (0.42 mL, 3 mmol) at room temperature. The solution was stirred overnight. The resultant solution was removed DMF and purified by reversed phase liquid
chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil. LC/MS 437.2 [M+H]
Step b. Preparation of 2,2'-({2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]- 2-oxoethyl}azanediyl)diacetic acid
Lithium hydroxide (120 mg, 5mmol) in 5 mL H2O was added to the solution of diethyl 2,2'-({2-[(2- {2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}azanediyl)-diacetate (0.41 g, 0.94 mmol) in a mixture of 5 mL H2O, 5 mL MeOH and 5 mL THF. The resultant solution was stirred for 1 hour at room temperature. The solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoro-acetic acid as the modifier. The product was obtained as an oil. LC/MS 381 .1 [M+H]+
Step c. Preparation of Compound 68
The title compound was prepared analogously using standard procedure A and B of Example 152. lon(s) found by LCMS [M+3H/3] = 91 8.2, [M+4H/4] =688.9, [M+5H/5] =551 .3. [M+6H/6] =459.6.
Example 158: Synthesis of Compound 69 and Compound 70
Figure imgf000358_0001
The title compounds were prepared analogously to Example 157, where the 2,2'-({2-[(2-{2-[(6- deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}azanediyl)diacetic acid was replaced with (1 R, 2R)-3-{(1 E)-3-[(2-{2-[(6-deoxy-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-3-oxoprop-1 -en-1 - yl}cyclopropane-1 ,2-dicarboxylic acid (INT-49) in the first step of that sequence. The product was obtained as a separable mixture of diastereomers in the final step (RPLC), one diastereomer with a shorter retention time (more polar diastereomer) and one diastereomer with a longer retention time (less polar diastereomer). The relative stereochemistry of the diastereomers was arbitrarily assigned, lon(s) found by LCMS (for both diastereomers): (M+2H)/2 = 1403.3, (M+3H)/3 = 935.9, (M+4H)/4 = 702.2.
Figure imgf000359_0001
The title compound was prepared analogously to the preparation of Compound 68. lon(s) found by LCMS
[M+3H/3] = 823.1 , [M+4H/4] =618.3, [M+5H/5] =494.9. [M+6H/6] =412.6.
Figure imgf000359_0002
Figure imgf000360_0001
Step a. Synthesis of D-Asn-INT-11
A solution of Z-D-Asn-OH (87.9 mg, 0.33 mmol) and HOBT hydrate (70.7 mg, 0.463 mmol) in anhydrous DMF (1 mL) was cooled in an ice-water bath. EDC-HCI (76.8 mg, 0.4 mmol) was added, and the resulting mixture was stirred for 15 minutes. It was then added with a solution of INT-1 1 (319 mg, 0.3 mmol) in anhydrous DMF (1 mL) and DIPEA (130 mg, 1 mmol). After the reaction mixture was stirred for 1 hour, it was directly purified through C18 RPLC (50 g, 10 to 90 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid (LCMS: [(M - 2Boc + 2H)/2]+ = 555). The material was re-dissolved in MeOH (~ 15 mL), and the solution was added with Pd/C. The resulting mixture was stirred under hydrogen for 4 hours. Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation to dryness. Yield 1 85.2 mg, 52.5 %. Ion found by LCMS: [(M - Boc + 2H)/2]+ = 538.
Step b. Synthesis of octa-Boc-Compound 72 precursor
A mixture of INT-40 (59.2 mg, 0.0438 mmol) and D-Asn-INT-1 1 (105.2 mg, 0.0894 mmol) was dissolved in in anhydrous DMF (0.5 mL) and DIPEA (52 mg, 0.4 mmol). After the solution was cooled in an ice-water bath, it was added with a solution of HATU (36.6 mg, 0.0964 mmol) in DMF (0.5 mL) via syringe pump at a rate of 0.5 ml/hr. The reaction was stirred for 30 more minutes. It was then purified by RPLC (50 g, 20 to 100 % MeOH and water). Yield 72 mg, 42.7 %. Ions found by LCMS: [(M + 3H)/3]+ = 1224, [(M - Boc + 3H)/3]+= 1 190, [(M - 2Boc + 3H)/3]+= 1 156, [(M - 3Boc + 3H)/3]+= 1 123, [(M - 6Boc + 3H)/3]+= 1023.
Step c. Removal of the Boc groups
The step-b product (72 mg, 0.01 86 mmol) was dissolved in TFA (~ 0.5 mL) and stirred at room temperature for 15 minutes. It was then directly purified by RPLC (50 g, 5 to 38 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 27 mg, 35.5 %. Ions found by LCMS: [(M + 3H)/3]+ = 956.2, [(M + 4H)/4]+ = 71 7.3, [(M + 5H)/5]+ =574 , [(M + 6H)/6]+ = 478.5. Example 161 : Synthesis of Compound 73
Figure imgf000361_0001
Note: The absolute configuration of the trans-cyclopropane- 1 ,2-dicarboxamide is arbitrarily assigned Step a. Synthesis of 1 ,1-dibenzyl 2,3-di-tert-butyl (2S,3S)-cyclopropane-1 ,1 ,2,3-tetracarboxylate
To a solution of triphenylarsine (306.2 mg, 1 mmol) in EtOH at 0 °C was added di-benzyl malonate (582.8 mg, 2.05 mmol) in EtOH (2 mL). The mixture was added drop-wise a solution of di-tert- butyl acetylene dicarboxylate (452.6 mg, 2 mmol) in acetonitrile (1 .4 mL) via syringe pump at a rate of 1 .4 ml/hr. After the addition, the reaction was stirred at 0 °C to room temperature for 5 days, then concentrated by rotary evaporation. The residue was purified through silica gel column chromatography (80 g, 0 to 25 % EtOAc in hexane). The collected fractions were concentrated by rotary evaporation and further dried in high vacuum to afford the product as a clear oil (674.2 mg, 66.0 %). Ion found by LCMS: [M - 2tBu + H]+ = 399.
Step b. Synthesis of (1S,2S)-3,3-bis[(benzyloxy)carbonyl]cyclopropane-1 ,2-dicarboxylic acid
The step-a product (624.2 mg, 1 .22 mmol) was dissolved in DCM (~ 3 mL), and the solution was treated with a mixture of TFA/anisole (2:0.2 mL). After the mixture was stirred at room temperature overnight, it was concentrated by rotary evaporation. The residue was purified by RPLC (100 g, 1 0 to 65 % acetonitrile and water). Yield 434.5 mg of thick oil, 89.5 %. Ion found by LCMS: [M + H]+ = 399.
Step c. Synthesis of di-carboxylic acid-Compound 73 precursor
A mixture of INT-20 (151 mg, 0.886 mmol) and the step-b product (17.3 mg, 0.0434 mmol) was dissolved in anhydrous DMF (0.5 mL) and DIPEA (65 mg, 0.5 mmol). After the solution was cooled in an ice-water bath, it was drop-wise added with a solution of HATU (37 mg, 0.1 mmol) in anhydrous DMF (0.5 mL) via syringe pump at a rate of 1 ml/hr. After the addition, the reaction was stirred for 30 more minutes and directly purified by RPLC (50 g, 30 to 100 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid. The material was re-dissolved in MeOH (~ 15 mL) and added with Pd/C. The mixture was stirred under hydrogen overnight. Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation and further dried in high vacuum to a white solid. Yield
96.2 mg, 61 .8 % over two steps. Ions found by LCMS: [(M + 3H)/3]+ = 1 1 99, [(M - 4Boc + 3H)/3]+ = 1025.
Step d. Synthesis of Rhamnose Compound 73 precursor
The step-c product (96.2 mg, 0.0268 mmol) was dissolved in anhydrous DMF (0.5 mL) and DIPEA (10.3 mg, 0.1 mmol). After the solution was cooled in an ice-water bath, it was added with HATU (10.2 mg, 0.0268 mmol). The mixture was stirred for 15 minutes, then INT-1 (1 1 mg, 0.044 mmol) was added. The resulting mixture was stirred at 0 °C to room temperature overnight. It was directly purified by RPLC (50 g, 20 to 100 % MeOH and water). Yield 57.6 mg, 56.2 %. Ions found by LCMS: [(M + 3H)/3]+ = 1275, [(M - 3Boc + 3H)/3]+ = 1 176, [M - 4Boc + 3H)/3]+ = 1 142, [(M - 5Boc + 3H)/3]+ = 1 109.
Step e. Removal of the Boc group
The step-d product (57.6 mg, 0.0151 mmol) was dissolved in TFA (~ 1 mL). The solution was stirred for 15 minutes and then directly purified through HPLC (5 to 23 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 13.1 mg, 21 .9 %. Ions found by LCMS: [(M + 4H)/4]+ = 706, [(M + 5H)/5]+ = 565, [(M + 6H)/6]+ = 471 . xample 162: Synthesis of Compound 74
Figure imgf000363_0001
The title compound was prepared analogously to Example 152, where 2,2'-({2-[(2-{2-[(6-deoxy- alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}azanediyl)diacetic acid was replaced with (1 R, 2R)-3-({4-[(2-{2-[(6-deoxy-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4- oxobutanamido}methyl)cyclopropane-1 ,2-dicarboxylic acid (INT-46) in the first step of that sequence. lon(s) found by LCMS: (M+2H)/2 = 1432.8, (M+3H)/3 = 955.6, (M+4H)/4 = 716.9.
Figure imgf000364_0001
The dicarboxylic acid INT-46 (0.052 g, 0. 106 mmol) and INT-48 (0.266 g, 0.170 mmol) were dissolved in 5 mL of DMF followed by addition of DI PEA (0.046 mL, 0.26 mmol). To this mixture was added HATU (0.100 g, 0.264 mmol) in 1 .5 mL of DMF via syringe pump in 2 hours at room temperature. After completion of HATU addition, the reaction mixture was stirred for 1 hr at room temperature. Then the reaction mixture was concentrated under reduced pressure. The residue was purified by reversed phase prep-HPLC with ACN/water (0.1 %TFA) to give the per-Boc-Compound 75 as Peak-1 (polar) and per-Boc-Compound 76 as Peak-2 (less polar). The mass spectrum showed strong detectable positive charge signals at tr = 2.86 and 3.07 min with 7 min (60-95%) method [found positive ion (M - 3Boc +
3H+)/3: 1056.2]. The Boc groups of Peak-1 and Peak-2 were removed by treating with TFA for less than 1 hour. Most of the TFA was removed and the residue was dissolved in a minimum amount of NMP and loaded onto a Preparative HPLC with ACN/water (0.1 % TFA). The desired product was collected and lyophilized to give the product as a white powder of TFA salt. Positive mass ions were found as [(M + 5H+)/5: 534.0, (M + 4H+)/4: 667.0] at tr = 2.21 min with 5 min (5-95%) method for Compound 75 [(M + 5H+)/5: 534.0, (M + 4H+)/4: 667.2] at tr = 2.41 min with 5 min (5-95%) method for Compound 76. The stereochemistry of the trans-cyclopropane dicarboxamide was arbitrarily assigned.
Example 164: Synthesis of Compound 77 and Compound 78
Figure imgf000365_0001
The dicarboxylic acid INT-49 (0.0599 g, 0. 138 mmol) and INT-48 (0.416 g, 0.276 mmol) were dissolved in 5 mL of DMF followed by addition of DI PEA (0.060 mL, 0.35 mmol). To this mixture was added HATU (0.131 g, 0.345 mmol) in 1 .5 mL of DMF via syringe pump in 2 hours at room temperature. After completion of HATU addition, the reaction mixture was stirred for 1 hr at room temperature. Then the reaction mixture was concentrated under reduced pressure. The residue was purified by reversed phase prep-HPLC with ACN/water (0.1 %TFA) to give the per-Boc-Compound 77 as Peak-1 (polar) and per-Boc-Compound 78 as Peak-2 (less polar). The mass spectrum showed strong detectable positive charge signals at tr = 2.96 and 3.18 min with 7 min (60-95%) method [found positive ion (M - CeHnCH -
4Boc + 5H+)/5: 571 .8]. The Boc groups of Peak-1 and Peak-2 were removed by treating with TFA for less than 1 hour. Most of the TFA was removed and the residue was dissolved in a minimum amount of NMP and loaded onto a Preparative HPLC with ACN/water (0.1 % TFA). The desired product was collected and lyophilized to give the product as a white powder of TFA salt. Positive mass ions were found as [(M + 5H+)/5: 522.4, (M + 4H+)/4: 652.4, (M + 3H+)/3: 869.4] at tr = 2.13 min with 5 min (5-95%) method for Compound 77 [(M + 5H+)/5: 522.0, (M + 4H+)/4: 652.2, (M + 3H+)/3: 869.6] at tr = 2.34 min with 5 min (5-95%) method for Compound 78. The stereochemistry of the trans-cyclopropane dicarboxamide was arbitrarily assigned.
Figure imgf000366_0001
The dicarboxylic acid INT-46 (0.0340 g, 0. 0690 mmol) and INT-50 (0.235 g, 0.138 mmol) were dissolved in 5 mL of DMF followed by addition of DI PEA (0.030 mL, 0.17 mmol). To this mixture was added HATU (0.656 g, 0.172 mmol) in 1 .0 mL of DMF via syringe pump in 2 hours at room temperature. After completion of HATU addition, the reaction mixture was stirred for 1 hr at room temperature. Then the reaction mixture was concentrated under reduced pressure. The residue was purified by reversed phase prep-HPLC with ACN/water (0.1 %TFA) to give the per-Boc-Compound 79 as Peak-1 (polar) and per-Boc-Compound 80 as Peak-2 (less polar). The mass spectrum showed strong detectable positive charge signals at tr = 3.07 and 3.26 min with 7 min (60-95%) method [found positive ion (M - ΟβΗηΟδ - 4Boc + 4H+)/4: 852.6, (M - CeHnOs - Boc + 3H+)/3: 1200.8, (M - CeHnC - Boc + 3H+)/3: 1207.0]. The Boc groups of Peak-1 and Peak-2 were removed by treating with TFA for less than 1 hour. Most of the TFA was removed and the residue was dissolved in a minimum amount of NMP and loaded onto a Preparative HPLC with ACN/water (0.1 % TFA). The desired product was collected and lyophilized to give the product as a white powder of TFA salt. Positive mass ions were found as [(M + 6H+)/6: 478.6, (M + 5H+)/5: 574.8] at tr = 2.03 min with 5 min (5-95%) method for Compound 79, [(M + 6H+)/6: 478.4, (M + 5H+)/5: 574.8] at tr = 2.14 min with 5 min (5-95%) method for Compound 80. The stereochemistry of the trans- cyclopropane dicarboxamide was arbitrarily assigned.
Example 166: Synthesis of Compound 81
Part 1. Preparation of the Asparagine Decapeptide
Figure imgf000367_0001
Step a. Synthesis of Z-Thr-Asn-tert-butyl ester
A mixture of Z-Thr-OH (2.79 g, 1 1 mmol) and HOBT (2.14 g, 14 mmol) in anhydrous DMF (8 ml_) was cooled in an ice-water bath and added with EDC HCI (2.3 g, 12 mmol). After stirred for 30 minutes, the reaction was added with a suspension of H-Asn-OtBu (1 .88 g, 1 0 mmol) and DIPEA (2.6 g, 20 mmol) in DMF (10 mL) and DCM (10 mL). The reaction was continued overnight and then extracted with water (50 mL) and DCM (40 mL x 2). The combined organic layers were concentrated by rotary evaporation. The residue was purified by RPLC (100 g, 5 to 50 % acetonitrile and water). Yield 3.5 g, 82.6 %. Ions found by LCMS: [M + H]+ = 424, [M - tBu + H]+ = 368. Step b. Synthesis of Z-Thr-Asn-OH
The step-a product (3.5 g, 8.26 mmol) in DMC (5 mL) was added with TFA (10 mL) and thioanisole (3 mL). After the mixture was stirred at room temperature for 1 day, it was concentrated by rotary evaporation. The residue was purified by RPLC (150 g, 5 to 31 % acetonitrile and water). Yield 448.7 mg, 14.8 %. Ion found by LCMS: [M + H]+ = 368.
Step c. Synthesis of Z-Thr-Asn-INT-11
A mixture of the step-b product (2368 mg, 1 mmol) and HOBT (214.2 mg, 1 .4 mmol) in anhydrous DMF (1 mL) was cooled in an ice-water bath and added with EDC HCI (209 mg, 1 .1 mmol). After stirred for 30 minutes, the reaction was added with INT-1 1 (1 .06 g, 1 mmol) and DIPEA (325 mg, 2.5 mmol) in DMF (1 .5 mL). The reaction was continued for 2 hours and then purified by RPLC (100 g, 15 to 100 % MeOH and water, using 0.1 % TFA as modifier). Yield 1 .3 g, 92 %. Ions found by LCMS: [(M - 2Boc + 2H)/2]+ = 606.5. Step d. Removal of the Cbz protection
Step-c product (1 .3 g, 0.92 mmol) was dissolved in MeOH (30 mL). The solution was added with Pd/C and stirred under hydrogen for 5 hours. Pd/C was removed, and the filtrate was concentrated by rotary evaporation to dryness. Yield 1 .1 g, 93.6 %. Ion found by LCMS: [(M - Boc + 2H)/2]+ = 589. Step e. Synthesis of the Asparagine Decapeptide
A mixture of the step-d product (1 .1 g, 0.861 mmol), Na-Z-NY-Boc-L-2,4-dimaminobutyric acid dicyclohexylammonium (533.7 mg, 1 mmol) and DIPEA (260 mg, 2 mmol) in anhydrous NMP (3 mL) was cooled in an ice-water bath and drop-wise added with a solution of HATU (400 mg, 1 .06 mmol) in NMP (1 .5 mL) via syringe pump at a rate of 1 ml/hr. After the addition, the resulting mixture was stirred for 1 hour and directly purified by RPLC (50 g, 20 to 100 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid. The material was re-dissolved in MeOH (30 mL) and added with Pd/C. The mixture was stirred under hydrogen for 4 hours. Pd/C was removed, and the filtrate was concentrated by rotary evaporation to dryness. Yield 601 mg, 47.2 %. Ion found by LCMS: [(M - Boc +2H)/2]+ = 689.
Figure imgf000369_0001
Step a. Synthesis of Octa-Boc Compound 81 precursor
A mixture of INT-66 (56 mg, 0.0723 mmol) and the asparagine decapeptide (235 mg, 0.159 mmol) was dissolved in anhydrous DMF (1 mL) and DIPEA (65 mg, 0.5 mmol). After cooled in an ice- water bath, the mixture was drop-wise added with a solution of HATU (70.4 mg, 0.185 mmol) in DMF (0.5 mL) at a rate of 1 ml/hr. After the addition, the reaction was stirred for 30 more minutes and then purified by RPLC (150 g, 10 to 100 % MeOH and water, using 0.1 % TFA as modifier). The collected fractions were concentrated by rotary evaporation to a white solid. Yield 68.5 mg, 25.7 %. Ions found by LCMS: [(M - Boc + 3H)/3]+ = 1 1 98, [(M - 2Boc + 3H)/3]+ = 1 165, [(M - 3Boc + 3H)/3]+ = 1 132, [(M - 4Boc + 3H)/3]+ = 1098.
Step b. Removal of the Boc groups to give Compound 81
The step-a product (68.5 mg, 0.186 mmol) was dissolved in TFA (~ 1 mL). The solution was stirred at room temperature for 15 minutes, then directly purified by RPLC (50 g, 0 to 33 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 24.5 mg, 34.7 %. Ions found by LCMS: [(M + 3H)/3]+ = 965, [(M + 4H)/4]+ = 724, [(M + 5H)/5]+ = 579, [(M + 6H)/6]+ = 483.
Figure imgf000370_0001
Step a. Preparation of 4,4'-{[1 ,4-dimethoxy-1 ,4-dioxobutane-2,3-diyl]diazanediyl}bis(4-oxobutanoic acid), meso-
Dimeihyl 2.3-diamino-succinate (meso) (176 mg, 1 mmo!) and succinic anhydride (600 mg, 6 mmo!) were dissolved into 5 mL 10% pyridine in DC , the resulting solution was stirred overnight and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid
chromatograph eluted with 0% to 100% acetonitrile and water, using trifluoroacetic acid as the modifier. The product was obtained as an oil, LCMS: 377.1 , [M+H]+.
Step b. Preparation of dimethyl 2,3-(meso)-bis(4-oxo-4-{[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyloxan-2-yl]oxy}ethoxy)ethyl]amino}butanamido)butanedioate 4,4'-{[1 ,4-Dimethoxy-1 ,4-dioxobutane-2,3-diyl]diazanediyl}bis(4-oxobutanoic acid), meso- (90 mg, 0.23 mmol) was dissolved into 5 mL DMF, then INT-1 (120 mg, 0.5 mmol), EDC (100 mg, 0.5 mmol), HOBT (80 mg, 0.5 mmol), Hunig's base (0.14 mL, 1 mmol) were added the solution at room temperature. The resultant solution was stirred overnight. The resultant solution was removed DMF and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil. LC/MS 843.4 [M+H].
Step c. Synthesis of 2,3-(meso)-bis(4-oxo-4-{[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyloxan-2-yl]oxy}ethoxy)ethyl]amino}butanamido)butanedioic acid
Lithium hydroxide (12 mg, 0.5 mmol) in 2 mL H2O was added to the solution of dimethyl 2,3- (meso)-bis(4-oxo-4-{[2-(2-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}ethoxy) - ethyl]amino}butanamido)butanedioate (30 mg, 0.035 mmol) in 2 mL MeOH and 2mL THF, and the mixture was stirred at room temperature for 1 hours at room temperature. The reaction solution was quenched by drops of acetic acid. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoro-acetic acid as the modifier. The product was obtained as an oil. LC/MS 815.3, [M+H]+.
Step d. Preparation of Compound 82.
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3] = 1062.9, [M+4H/4] =797.5, [M+5H/5]=638.2, [M+6H/6]=531 .9.
Figure imgf000372_0001
Step a. Preparation of diethyl 2,2'-({2-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}azanediyl)diacetate
To the solution of [bis(2-ethoxy-2-oxoethyl)amino]acetic acid (4.4 g, 18 mmol), INT-1 (4.51 g, 18 mmol) in DMF (50 mL) was added EDC (4 g, 20 mmol), HOBT (2.7 g, 20 mmol), Hunig's base (4.2 mL, 30 mmol) at room temperature. The solution was stirred overnight. The resultant solution was removed DMF and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil. LC/MS 481 .2 [M+H]
Step b. Preparation of 2,2'-({2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]- 2-oxoethyl}azanediyl)diacetic acid
Lithium hydroxide (600 mg, 25 mmol) in 10 mL H20 was added to the solution of diethyl 2,2'-({2- [(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}azanediyl)-diacetate (4.80 g, 10mmol) in mix solvent of 10 mL H20, 20 mL MeOH and 20 mL THF. The resultant solution was stirred for 1 hours at room temperature. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil. LC/MS 425.2 [M+H]+
Step c. Preparation of Compound 83
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3] = 928.2, [M+4H/4] =696.4, [M+5H/5]=557.3, [M+6H/6]=464.6.
Figure imgf000373_0001
Step a. Synthesis of N-[(benzyloxy)carbonyl]-3-carbamimidamido-L-alanine
L-2-amino-3-guanidinopropionic acid (1 .83 g, 10 mmol) was dissolved in a solution of NaOH (800 mg, 10 mmol) in water (10 mL). After cooled in ice-water bath, the mixture was added with a solution of N-
(benzyloxycarbonyloxy)succinimide (2.73 g, 1 1 mmol) in THF (10 mL). The resulting mixture was stirred at 0 °C to room temperature overnight. THF was then partially removed by rotary evaporation, and the residue was extracted with a 1 :1 mixture of EtOAc/hexane (40 mL). The aqueous layer was purified by RPLC (150 g, 0 to 40 % acetonitrile and water). Yield 2.05 g, 71 .3 %. Ion found by LCMS: [M + H]+ = 281 . Step b. Synthesis of N-[(benzyloxy)carbonyl)]-3-carbamimidamido-Ala-INT-11
To a solution of a mixture of N-[(benzyloxy)carbonyl]-3-carbamimidamido-L-alanine (420 mg, 1 .5 mmol) and INT-1 1 (1 .06 g, 1 mmol) in anhydrous DMF (2 mL) was added DMAP (24.4 mg, 0.2 mmol) and HATU (570 mg, 1 .5 mmol). After the reaction was stirred overnight, it was added with DIPEA (130 mg, 1 mmol) and continued for 6 more hours. It was then directly purified through RPLC (100 g, 20 to 80 % MeOH and water). Yield 980 mg, 74 %. Ion found by LCMS: [(M - Boc + 2H)/2]+ = 613.
Step c. Synthesis of 3-carbamimidamido-Ala-INT-11
The step-b product (185.4 mg, 0.14 mmol) was re-dissolved in MeOH (20 mL), and the solution was added with Pd/C. The mixture was stirred under hydrogen for 3 hours. Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation to dryness. Yield 165 mg, 98%. Ion found by LCMS: [(M - Boc + 2H)/2]+ = 546.
Step d. Synthesis of octa-Boc-Compound 84 precursor
A mixture of the step-c product (120 mg, 0.1 mmol), INT-40 (66.8 mg, 0.494 mmol), and DMAP (66.8 mg) was dissolved in anhydrous DMF (2 mL). The solution was drop-wise added with a HATU (45.6 mg, 0.12 mmol) solution in DMF (0.5 mL) via syringe pump at a rate of 1 ml/hr. After the addition, the resulting mixture was stirred for one more hour. It was then directly purified by RPLC (50 g, 20 to 95 % MeOH in water, using 0.1 % TFA as modifier). Yield 1 15 mg, 63 %. Ions found by LCMS: [(M - Boc + 3H)/3]+ = 1 1 99, [(M - 2Boc + 3H)/3]+ = 1 166, [(M - 3Boc + 3H)/3]+ = 1 132, [(M - 4Boc + 3H)/3]+ = 1099, [(M - 5Boc + 3H)/3]+ = 1066).
Step e. Removal of the Boc group
The step-d product was dissolved a 1 :1 mixture of TFA:DCM (~ 1 mL). The solution was stirred for 15 minutes, then directly purified through HPLC (0 to 22.4 % acetonitrile in water, using 0.1 % TFA as modifier). Yield 14.8 mg, 12.5 %. Ions found by LCMS: [(M + 3H)/3]+ = 966, [(M + 4H)/4]+ = 724, [(M + 5H)/5]+ = 580, [(M + 6H)/6]+ = 483.
Figure imgf000375_0001
Step a. Synthesis of benzyl (3R,4R)-3,4-bis{[(2S)-1-methoxy-1-oxooctan-2- yl]carbamoyl}pyrrolidine-1-carboxylate
A solution of transl -[(benzyloxy)carbonyl]pyrrolidine-3,4-dicarboxylic acid (3.69g, 12.58 mmol) and methyl (2S)-2-aminooctanoate (4.58g, 26.42 mmol), DIEA(13.8ml_, 79.3mmol) in 20ml_ of DMF was treated with HATU (10. Og, 26.4mmol). After stirring for 30 minutes at room temperature the reaction was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using formic acid (0.1 %) as the modifier. Yield 1 .44 g, 19%. lon(s) found by LCMS: M+H = 604.4
Step b. Synthesis of (2S,2'S)-2,2'-[{(3R,4R)-1-[(benzyloxy)carbonyl]pyrrolidine-3,4- diyl}bis(carbonylazanediyl)]dioctanoic acid
A solution of benzyl (3R, 4R)-3,4-bis{[(2S)-1 -methoxy-1 -oxooctan-2-yl]carbamoyl}pyrrolidine-1 - carboxylate (1 .44g, 2.39mmol) dissolved in THF (1 OmL) was treated with a solution of LiOH(0.143g,
5.96mmol) dissolved in water (10ml_). After stirring for 30 minutes all starting material was consumed by LCMS. Work-up: the reaction was made slightly acidic with concentrated HCI, extracted into ethyl acetate, and dried over sodium sulfate. The resulting crude product was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using formic acid (0.1 %) as the modifier. Yield 1 .14 g, 83%.
Step c. Synthesis of Cbz-pyrrolidine-polymyxin B dimer
A solution of racemic (2S,2'S)-2,2'-[{(3R,4R)-1 -[(benzyloxy)carbonyl]pyrrolidine-3,4- diyl}bis(carbonylazanediyl)]dioctanoic acid (0.401 g, 0.697mmol), INT-18 (2.18g, 1 .39mmol), DIEA(0.76ml_, 4.39mmol) dissolved in DMF(5ml_) was treated with a solution of HATU(0.557g, 1 .46mmol) dissolved in DMF(2mL), dropwise over 60 minutes. The reaction was stirred for an additional hour, then taken-on to the next step without further purification. Step d. Synthesis of Pyrrolidine-polymyxinB dimer
A solution of crude Cbz-pyrrolidine-polymyxinB dimer in DMF was treated with 5%Pd/C (0.75g) and vacuum flushed with hydrogen and stirred under a hydrogen atmosphere. LCMS after 2h shows complete conversion. The reaction was filtered through Celite, concentrated, and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using no modifier. Yield 1 .39 g, 56%. lon(s) found by LCMS: [(M- 2Boc)+3H]/3 = 1 1 1 1 .4, [(M-3Boc)+3H]/3 = 1078.0, [(M-4Boc)+3H]/3 = 1044.7.
Step e. Synthesis of Rhamnose Pyrrolidine-polymyxinB dimer
A solution of pyrrolidine-polymyxinB dimer (0.300g, 0.085mmol), INT-2 (0.0298g, 0.085mmol), and DIEA (0.049mL, 0.280mmol) was treated with a solution of HATU(0.036g, 0.0934mmol) in
DMF(1 .OmL), dropwise over 60 minutes. After an additional hour stirring the solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using 0.1 % TFA as the modifier. Yield 0.41 6 g, 127% (not pure), lon(s) found by LCMS: Did not ionize.
Step f. Deprotection to Compound 85
A solution of rhamnose pyrrolidine-polymyxinB dimer (0.328g, 0.085mmol) dissolved in
DCM(2mL) was treated with TFA(2mL) for 5 minutes. The solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 1 00% acetonitrile and water, using 0.1 % formic acid as the modifier. Yield 0.1 90 g, 67%. lon(s) found by LCMS: (M+3H)/3 = 955.8, (M+4H)/4 = 716.9, (M+5H)/5 = 573.7.
Example 171 : Synthesis of Compound 86
Figure imgf000376_0001
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3] = 1029.9, [M+4H/4] =772.7, [M+5H/5]=61 8.4, [M+6H/6]=515.5.
Figure imgf000377_0001
Step a. Synthesis of diethyl 1-{4-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl) amino]-4-oxobutanoyl}pyrrolidine-2,5-dicarboxylate
To the solution of diethyl pyrrolidine-2,5-dicarboxylate (1 .0 g, 4.6 mmol), INT-2 (4-[(2-{2-[(6- deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoic acid,1 .6 g, 4.6 mmol) in DMF (20 mL) was added EDC (1 g, 5 mmol), HOBT (800 mg, 5 mmol), Hunig's base (1 .4 ml_, 10 mmol) at room temperature. The solution was stirred overnight. The resultant solution was removed DMF and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil. LC/MS 549.3 [M+H]
Step b. Synthesis of 1-{4-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl) amino]-4- oxobutanoyl}pyrrolidine-2,5-dicarboxylic acid
Lithium hydroxide (120 mg, 5 mmol) in 5 mL H20 was added to the solution of diethyl 1 -{4-[(2-{2- [(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl) amino]-4-oxobutanoyl}pyrrolidine-2,5-dicarboxylate (1 .0 g, 1 .8 mmol) in mix solvent of 5 mL MeOH and 5 mL THF. The resultant solution was stirred for 1 hours at room temperature. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoro-acetic acid as the modifier. The product was obtained as an oil. LC/MS 493.2 [M+H]+
Step c. Preparation of Compound 87
The title compound was prepared in an analogous manner as described in standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3] = 955.6, [M+4H/4] =71 6.9, [M+5H/5]=573.7, [M+6H/6]=478.3.
xample 173: Synthesis of Compound 88
Figure imgf000379_0001
Step a. Coupling of L-Nor-Leu Methyl Ester to racemic-trans-1-(tert-butoxycarbonyl)pyrrolidine- 3,4-dicarboxylic acid
To a solution of racemic-trans-1 -(tert-butoxycarbonyl)pyrrolidine-3,4-dicarboxylic acid (257.5 mg, 0.99 mmol), L-Norleu-OMe-hydrochloride (375.29 mg, 2.07 mmol), and diisopropylethylamine (384.79 mg, 2.98 mmol) in DMF (10 ml_) was added a solution of HATU (0.29 g, 0.77 mmol) in DMF (2 ml_) via a syringe pump at a rate of 2.0 mL/hr. After the addition, the reaction mixture was stirred for an additional 1 hour at room temperature. The reaction mixture was purified by reversed phase liquid chromatography
(RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5 to 50% acetonitrile and water, using 0.1 % TFA as modifier to yield two diastereomers: a more polar diastereomer (Step-a more polar isomer) (1 13.1 mg, 22%) and a less polar diastereomer (Step-a less polar isomer) (92.9 mg, 17%). LC/MS [(M- Boc)+H]+ = 414.2 (artefactual loss of 1 boc group) for both boc-protected intermediates (methyl 2- [((3R,4R)-4-{N-[(1 S)-1 -(methoxycarbonyl)pentyl]carbamoyl}-1 [(tert-butyl)oxycarbonyl]pyrrolidin-3- yl)carbonylamino](2S). Note: the isomers were taken forward separately in subsequent syntheses. Step b. Synthesis of INT-37a
A solution of the Step-a more polar isomer (379 mg, 1 .46 mmol) was stirred in TFA:CH2Cl2 (1 :2, 9 mL) at room temperature until completion. The volatiles were removed under reduced pressure then purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid
chromatograph eluted with 0 to 75% acetonitrile and water, using 0.1 % TFA as modifier to yield INT-37a (217.9 mg, 36%). Ion found by LC/MS [M+H]+ = 414.2.
Step c. Conjugation of INT-37a with INT-2
INT-37a (217.3 mg, 0.41 mmol), INT-2, (4-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanoic acid, 159.2 mg, 0.45 mmol), HOBt (61 .22 mg, 0.45 mmol) and triethylamine (129.21 mg, 1 .28 mmol) were stirred in DMF (3 mL). To this solution was added a prepared solution of EDCI (70.34 mg, 0.45 mmol) in DMF (2 mL). The reaction was stirred at room temperature until completion. The reaction mixture was diluted with ethyl acetate and washed with 1 N HCI aqueous solution and saturated NaHCCb aqueous solution. The organic layer was dried over Na2S04 and filtered then concentrated. The residue was purified by reversed phase liquid
chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10 to 50% acetonitrile and water, using 0.1 % TFA as modifier to yield the desired compound (204.2 mg, 66%). Ion found by LC/MS [M+H]+ = 747.3.
Step d. Synthesis of INT-38a
To a solution of Step-c product (204.2 mg, 0.27 mmol) in MeOH:THF:H20 (1 :1 :2, 8 mL) at room temperature was added LiOH (14 mg, 0.58 mmol) until completion monitoring by analytical HPLC. After the reaction was completed, it was quenched with 1 N HCI aqueous solution to pH 3 then concentrated under reduced pressure. The residue was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0 to 50% acetonitrile and water, using 0.1 % TFA as modifier to yield the desired compound (65.2 mg, 33%). Ion found by LC/MS [M-H]+ = 717.3.
Step e. Synthesis of Compound 88
A solution of HATU (45.70 mg, 0.12 mmol) in DMF (0.3 mL) was added via a syringe pump at a rate of 2.5 mL/hr into a solution of INT-38a (40.0 mg, 0.056 mmol), INT-17 (183.89 mg, 0.12 mmol) and diisopropylethylamine (43.12 mg, 0.33 mmol) in DMF (0.9 mL). The reaction mixture was stirred for an additional hour then purified reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5 to 30 to 100% methanol and water, using 0.1 % TFA as modifier to yield the Boc-protected intermediate. The Boc-protected intermediate was stirred in TFA:DCM (1 :1 , 4 mL) at room temperature for 20 minutes. After the reaction was completed, the reaction mixture was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0 to 40% acetonitrile and water, using 0.1 % TFA as modifier to yield the title compound (1 19.5 mg, 55%). lon(s) found by LC/MS [(M+3H)/3]+ = 914.0, [(M+4H)/4]+ = 685.5, [(M+5H)/5]+ = 549.3 and [(M+6H)/6]+ = 457.9.
Example 174: Synthesis of Compound 89
Figure imgf000381_0001
The dimethyl ester INT-51 (0.102 g, 0. 137 mmol) was dissolved in 6 mL of THF/H2O (1 :1 ) followed by treating with LiOH (0.0069 g, 0.29 mmol). After 30 minutes, the reaction was finished and acidified with 1 N HCI and extracted with EtOAc to give the diacid without purification for next step. The diacid and INT-18 (0.4277 g, 0.2735 mmol) were dissolved in 5 mL of DMF followed by addition of DIPEA (0.071 mL, 0.41 mmol). To this mixture was added HATU (0.130 g, 0.342 mmol) in 2.0 mL of DMF via syringe pump in 2 hours at room temperature. After completion of HATU addition, the reaction mixture was stirred for 1 hr at room temperature. Then the reaction mixture was concentrated under reduced pressure. The residue was purified by reversed phase prep-HPLC with ACN/water (0.1 %TFA) to give the per-Boc-Compound 89. The mass spectrum showed strong detectable positive charge signals at tr = 3.77 min with 7 min (60-95%) method [found positive ion (M -CeHnOs - 6Boc + 6H+)/6: 507.4, (M - CeH C - 6Boc + 8H+)/8: 408.2]. The Boc groups of Per-Boc-Compound 89 were removed by treating with TFA for less than 1 hour. Most of the TFA was removed and the residue was dissolved in a minimum amount of NMP and loaded onto a Prep-HPLC with ACN/water (0.1 % TFA). The desired product was collected and lyophilized to give the product as a white powder of TFA salt. Positive mass ions were found as [(M + 6H+)/6: 469.2, (M + 5H+)/5: 562.8] at tr = 2.03 min with 5 min (5-95%) method for Compound 89. Example 175: Synthesis of Compound 90
Figure imgf000382_0001
The dimethyl ester INT-52 (0.133 g, 0. 178 mmol) was dissolved in 8 mL of THF/H2O (1 :1 ) followed by treating with LiOH (0.0090 g, 0.37 mmol). After 30 minutes, the reaction was finished and acidified with 1 N HCI and extracted with EtOAc to give the diacid without purification for next step. The diacid and INT-18 (0.5577 g, 0.3566 mmol) were dissolved in 5 mL of DMF followed by addition of DIPEA (0.093 mL, 0.53 mmol). To this mixture was added HATU (0.1695 g, 0.4577 mmol) in 2.0 mL of DMF via syringe pump in 2 hours at room temperature. After completion of HATU addition, the reaction mixture was stirred for 1 hr at room temperature. Then the reaction mixture was concentrated under reduced pressure. The residue was purified by reversed phase C18 with ACN/water (0.1 %TFA) to give the per- Boc-Compound 90. The mass spectrum showed a strong detectable positive charge signal at tr = 4.26 min with 7 min (60-95%) method [found positive ion (M - 3Boc + 3H+)/3: 1 170.4]. The Boc groups of Per- Boc Compound 90 were removed by treating with TFA for less than 1 hour. Most of the TFA was removed and the residue was dissolved in a minimum amount of NMP and loaded onto a prep-PLC with
ACN/water (0.1 % TFA). The desired product was collected and lyophilized to give the product as a white powder of TFA salt. Positive mass ions were found as [(M + 6H+)/6: 468.8, (M + 5H+)/5: 562.2, (M + 4H+)/4: 702.4] at tr = 2.23 min with 5 min (5-95%) method for Compound 90.
Example 176: Synthesis of Compound 91
Figure imgf000383_0001
INT-56 (0.224 g, 0.149 mmol) was dissolved in 6 mL of THF/water (1 :1 ) and treated with LiOH (0.0075 g, 0.312 mmol). After acidification THF was removed and 3mL of NMP was added. Then most of water was removed and the residue was load to pep-HPLC to purify. 0.219 g of INT-56 diacid was obtained (100%). Positive mass ions were found as (M - 2 CeHnC + 2H+)/2: 593.3, (M - CeHnC + 2H+)/2: 666.2, (M + 2H+)/2: 739.2, tr = 3.93 minutes with 8 min (5-95%) method.
The diacid, INT-18 (0.465 g, 0.297 mmol) and DIPEA (0.078 mL, 0.45 mmol) were mixed in 5 mL of DMF. HATU (0.141 g, 0.371 mmol) was added via syringe pump (2 mL, 1 ml/hr). The reaction solution was stirred for another hour after addition. DMF was removed and the residue was purified by prep-HPLC to give per-Boc- Compound 91 , 0.323 g, 48%. Positive mass ions were found as (M - 2C6H11 O4 + 4H+)/4: 1067.6, (M -4Boc - CeHn Ot + 4H+)/4: 1005.4, tr = 2.55 minutes with 7 min (60-95%) method. The Boc groups of Per-Boc- Compound 91 were removed by treating with TFA for less than 1 hour. Most of the TFA was removed and the residue was dissolved in a minimum amount of NMP and loaded onto prep- HPLC with ACN/water (0.1 % TFA). The desired product was collected and lyophilized to give the product as a white powder of TFA salt. Positive mass ions were found as [(M - 2C6H11 O4 + 5H+)/5: 655.8, (M - C6H11 O4 + 5H+)/5: 684.8, (M + 5H+)/5: 714.2, (M + 4H+)/4: 892.5] at tr = 2.02 min with 5 min (5-95%) method for Compound 91 .
Figure imgf000384_0001
INT-57 (0.069 g, 0.090 mmol), INT-18 (0.282 g, 0.180 mmol) and DIPEA (0.047 ml_, 0.27 mmol) were mixed in 5 mL of DMF. HATU (0.0857 g, 0.226 mmol) was added via syringe pump (2 ml_, 1 ml/hr). The reaction solution was stirred for another hour after addition. DMF was removed and the residue was purified by prep-HPLC to give per-Boc-Compound 92, 0.193 g, 55%. Positive mass ions were found as (M - 3Boc+ 4H+)/4: 889.6, (M - 3Boc+ 3H+)/3: 1 186.8, (M - 3Boc - CeHnC + 4H+)/4: 850.6, tr = 3.08 minutes with 7 min (60-95%) method. The Boc groups of Per-Boc-Compound 92 were removed by treating with TFA for less than 1 hour. Most of the TFA was removed and the residue was dissolved in a minimum amount of NMP and loaded onto prep- HPLC with ACN/water (0.1 % TFA). The desired product was collected and lyophilized to give the product as a white powder of TFA salt. Positive mass ions were found as [(M - C6Hi i04S + 6H+)/6: 443.0, (M - CeH C S + 5H+)/5: 531 .4, (M + 6H+)/6: 476.6, (M + 5H+)/: 571 .4] for Compound 92.
Figure imgf000385_0001
Step a. Preparation of 4-benzyl 1-ferf-butyl W-{[(2S)-1-methoxy-1-oxooctan-2-yl]carbamoyl}-L- aspartate
Methyl (2S)-2-isocyanato-octanoate (200 mg, 1 mmol) was added into the solution of 4-benzyl 1 - tert-butyl L-aspartate (280 mg, 1 .00 mmol) and triethylamine (0.28 mL, 2.0 mmol) in 5 mL DMF at room temperature, the reaction was stirred overnight, then concentrated and the residue was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. Yield 400mg, 75%. lon(s) found by LCMS: M+H = 479.3.
Step b. Preparation of methyl (4S,8S,11 S)-8-[2-(benzyloxy)-2-oxoethyl]-4,11-dihexyl-3,6,9-trioxo-2- oxa-5,7,10-triazadodecan-12-oate
4-benzyl 1 -ferf-butyl A/-{[(2S)-1 -methoxy-1 -oxooctan-2-yl]carbamoyl}-L-aspartate (400 mg, 0.83 mmol) was treated with 5mL trifluoroacetic acid in 10mL DCM and stirred for half hour and then concentrated and dried under high vacuum and used for next step without purification.
The above residue was re-dissolved into 20 mL DMF, followed by addition of methyl (2S)-2-amino- octanoate HCI salt (200 mg, 1 mmol), EDC (200 mg, 1 mmol), HOBT (150 mg, 1 mmol), Hunig's base (0.28 mL, 2 mmol) at room temperature. The solution was stirred overnight. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil. lon(s) found by LCMS: M+H = 578.3.
Step c. Preparation of N-[(2S)-1-methoxy-1-oxooctan-2-yl]-N~2~-{[(2S)-1-methoxy-1-oxooctan-2- yl]carbamoyl}-L-alpha-asparagine
Methyl (4S,8S,1 1 S)-8-[2-(benzyloxy)-2-oxoethyl]-4,1 1 -dihexyl-3,6,9-trioxo-2-oxa-5,7,10- triazadodecan-12-oate (150 mg, 0.26 mmol) was dissolved into 5 mL 5% H20 in MeOH, then 20 mg of 5% Palladium on charcoal was added above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere for 2hours. The palladium charcoal was removed by filtration, and the filtrate was concentrated and used for next step without purification.
Step d. Preparation of methyl (2S,6S)-2-hexyl-6-{[(2S)-1-methoxy-1-oxooctan-2-yl]carbamoyl}-4,8- dioxo-14-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}-12-oxa-3,5,9-triazatetradecan-
1-oate
The residue from Step c. was re-dissolved into 5 mL DMF, followed by addition of INT-1 (65 mg,
0.26 mmol), EDC (1 00 mg, 1 mmol), HOBT (75 mg, 0.5 mmol), Hunig's base (0.14 mL, 1 mmol) at room temperature. The solution was stirred overnight. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil. lon(s) found by LCMS: M+H = 721 .4.
Step e. Preparation of (2S,6S)-6-{[(1S)-1-carboxyheptyl]carbamoyl}-2-hexyl-4,8-dioxo-14- {[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}-12-oxa-3,5,9-triazatetradecan-1-oic acid (non-preferred name)
Methyl (2S,6S)-2-hexyl-6-{[(2S)-1 -methoxy-1 -oxooctan-2-yl]carbamoyl}-4,8-dioxo-14-
{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}-12-oxa-3,5,9-triazatetradecan-1 -oate (0.18 mg, 0.25 mmol), in methanol (1 mL) was treated with a solution of lithium hydroxide (0.024 g, 1 .0 mmol), in water (1 mL) and THF (1 mL), then stirred at room temperature for 30 minutes. The reaction was quenched by drops of acetic acid. The desired product was isolated directly by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as modifier. The product was obtained as an oil. lon(s) found by LCMS: M+H = 693.4.
Step f. Preparation of Compound 93
The title compound was prepared analogously using the standard procedure A and B of Example
102. lon(s) found by LCMS: [M/3]+1 =928.2, [M/4]+1 =696.4, [M/5]+1 =557.3, [M/6]+1 =464.6.
Figure imgf000387_0001
Note: The absolute configuration of the trans-cyclopropane- 1 ,2-dicarboxamide is arbitrarily assigned but the product is derived from the more polar trans-cyclopropane-(bis-octanoate tert-butyl ester)
Step a. Synthesis of 1 -benzyl 2,3-di-ferf-butyl 1 -methyl (2S,3S)-cyclopropane-1 ,1 ,2,3- tetracarboxylate
Triphenylarsine (3.17 g, 1 1 mmol) was dissolved in anhydrous EtOH (50 mL) and mixed with benzyl methyl malonate (4.6 g, 22.09 mmol). After the solution was cooled in an ice-water bath, it was added with di-tert-butyl acetylene dicarboxylate (5 g, 22.09 mmol). The reaction mixture was stirred at 0 °C to room temperature for 10 days. EtOH was removed by rotary evaporation, and the residue was purified through silica gel column chromatography (220 g, 0 to 10 % EtOAc and hexane). Yield 9.2 g, 85.9 %. Ion found by LCMS: [M - 2tBu + H]+ = 323.
Step b. Selective hydrolysis of tert-butyl ester
The step-a product (9 g, 20.7 mmol) was dissolved in a mixture of DCM (30 mL), TFA (30 mL) and thioanisole (7.7g, 62.2 mmol). After the solution was stirred for 1 day, it was concentrated by rotary evaporation. The residue was purified by RPLC (150 g, 5 to 100 % acetonitrile and water). Yield 6.28 g, 94 %. Ion found by LCMS: [M + H]+ = 323. Step c. Synthesis of 1-benzyl 1-methyl (2ff,3/¾-2,3-bis{[(2S)-1-ferf-butoxy-1-oxooctan-2- yl]carbamoyl}cyclopropane-1 ,1-dicarboxylate and its isomer
A mixture of the step-b (646 mg, 2 mmol) and ferf-butyl (2S)-2-aminooctanoate (947 mg, 4.4 mmol) was dissolved in anhydrous DMF (3 m) and DIPEA (1 .14 g, 8.8 mmol). The resulting mixture was drop-wise added with a solution of HATU (1 .67 g, 4.4 mmol) in DMF (5 mL) via syringe pump at a rate of 10 ml/hr. After the addition, an additional amount of DIPEA (600 mg) was added, and the reaction was continued overnight. It was purified by RPLC (1 00 g, 20 to 90 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 374.9 mg of the desired product as more polar isomer (0.523 mmol, 26.1 %). Ions found by LCMS: [M - 2tBu + H]+ = 605 and [M + H]+ = 717. A less polar isomer was also obtained.
Step d. Selective hydrolysis of benzyl ester
The step-c more polar product was dissolved in EtOAC (~ 20 mL). The solution was added with Pd/C and stirred under hydrogen for 5 hours. Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation and further dried under high vacuum. Yield 300.6 mg, 91 .2%. Ion found by LCMS: [M - 2tBu + H]+ = 515.
Step e. Synthesis of methyl (2S,3¾-2,3-bis{[(2¾-1-fert-butoxy-1-oxooctan-2-yl]carbamoyl}-1-[(2-{2- [(6-deoxy-a-L-mannopyranosyl)oxy]ethoxy}ethyl)carbamoyl]cyclopropane-1-carboxylate
To a solution of a mixture of the step-d product (288 mg, 0.56 mmol) and HATU (254.6 mg, 0.67 mmol) in anhydrous DMF (2 mL) was added with DIPEA (130 mg, 1 mmol). After the resulting mixture was stirred for 10 minutes, it was added with INT-1 (185 mg, 0.35 mmol). The reaction was stirred for 1 hour, then purified by RPLC (40 g, 20 to 75 % acetonitrile and water). Yield 287.4 mg, 59.7 %. Ion found by LCMS: [M + H]+ = 860. Step f. Selective hydrolysis of di-tert-butyl ester
The step-e product was dissolved in TFA (~ 2 mL) and thioanisole (0.2 mL). The solution was stirred at room temperature overnight. It was concentrated by rotary evaporator, and the residue was purified by RPLC (40 g, 0 to 40 % acetonitrile and water). Yield 1 1 0.8 mg, 44.4 %. Ion found by LCMS: [M + H]+ = 748
Step g. Synthesis of deca-Boc Compound 94 precursor
A mixture of INT-18 (336 mg, 0.2148 mmol) and the step-f product (78.7 mg, 0.1053 mmol) was dissolved in anhydrous DMF (1 mL) and DIEPA (65 mg, 0.5 mmol). After the mixture was cooled in an ice-water bath, it was drop-wise added with a solution of HATU (87.4 mg, 0.23 mmol) in DMF (0.5 mL) via syringe pump at a rate of 0.5 ml/hr. The reaction was then directly purified by RPLC (40 g, 20 to 96 % MeOH and water, using 0.1 TFA as modifier). Yield 338 mg, 83.6 %. Ions found by LCMS: [(M -3Boc + 3H)/3]+ = 1 1 81 , [(M - 4Boc + 3H)/3]+ = 1 147, [(M - 5Boc + 3H)/3]+ = 1 1 14, [(M - 10Boc + 3H)/3]+ = 947.
Step h. Removal of the Boc groups Step-g product (338 mg, 0.088 mmol) was dissolved in TFA (~ 2 ml_). After the solution was stirred for 15 minutes, it was concentrated by rotary evaporation and purified by RPLC (50 g, 0 to 30 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 281 .3 mg, 80.4 %. Ions found by LCMS: [(M + 4H)/4]+ = 71 0, [(M + 5H)/5]+ = 568, [(M + 6H)/6]+ = 474, [(M +7H)/7]+ = 406.
Step i. Hydrolysis of methyl ester
Step-h product (281 .3 mg, 0.0707 mmol) was dissolved in MeOH (~ 1 mL). After the solution was cooled in ice-water bath, it was added with a solution of LiOH (25 mg, 1 mmol) in water (1 mL). The reaction was stirred for 1 hour and acidified with 4 N HCI solution in dioxane (0.26 mL). It was then directly purified by RPLC (50 g, 0 to 32 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 140 mg, 49.9 %. Ions found by LCMS: [(M + 4H)/4]+ = 706, [(M + 5H)/5]+ = 565, [(M + 6H)/6]+ = 471 , [(M + 7H)/7]+ = 404.
Example 180: Synthesis of Compound 95
Figure imgf000389_0001
The compound was prepared by an analogous procedure as that used to prepare Compound 94 except the less polar isomer from Step c was used instead of the more polar diastereomer. Ions found by LCMS: [(M + 4H)/4]+ = 706, [(M + 5H)/5]+ = 565, [(M + 6H)/6]+ = 471
Example 181 : Synthesis of Compound 96
Figure imgf000390_0001
Figure imgf000391_0001
Step a. Preparation of di-tert-butyl 16-{N~2~,N~2~-bis[2-(benzyloxy)-2-oxoethyl]-N,N-bis(17,17- dimethyl-15-oxo-3,6,9,12,16-pentaoxaoctadecan-1-yl)-L-alpha-asparaginyl}-4,7,10,13,19,22,25,28- octaoxa-16-azahentriacontane-1 ,31 -dioate
N,N-bis[2-(benzyloxy)-2-oxoethyl]-L-aspartic acid (150 mg, 0.28 mmol) and di-tert-butyl
4,7,10,13,19,22,25,28-octaoxa-16-azahentriacontane-1 ,31 -dioate (1 80mg, 0.28mmol) and triethylamine (0.1 mL, 0.7 mmol) were dissolved into 5ml_ DMF, then HATU (120 mg, 0.3mmol) in 1 ml_ DMF was added into the resultant solution by syringe pump over 1 hours, the resultant solution was concentrated and isolated directly by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as modifier. The product was obtained as an oil. lon(s) found by LCMS: [M+2H/2]=823.0.
Step b. Preparation of benzyl (25S)-26-[2-(benzyloxy)-2-oxoethyl]-25-[bis(15-oxo-21- {[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}-3,6,9,12,19-pentaoxa-16-azahenicosan- 1-yl)carbamoyl]-7,23-dioxo-22-(15-oxo-21-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2- yl]oxy}-3,6,9,12,19-pentaoxa-16-azahenicosan-1-yl)-1-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6- methyloxan-2-yl]oxy}-3,10,13,16,19-pentaoxa-6,22,26-triazaoctacosan-28-oate
Di-tert-butyl 1 6-{N~2~,N~2~-bis[2-(benzyloxy)-2-oxoethyl]-N,N-bis(17,17-dimethyl-15-oxo- 3,6,9,12,16-pentaoxaoctadecan-1 -yl)-L-alpha-asparaginyl}-4,7,10,13,19,22,25,28-octaoxa-16- azahentriacontane-1 ,31 -dioate (500 mg, 0.3 mmol) was treated with 5 mL trifluoroacetic acid in 5 mL DCM and stirred for half hour and then concentrated and dried under high vacuum used for next step without purification. The above residue was re-dissolved into 10 mL DMF, followed by addition of by addition of INT-1 (0.3 g, 1 .2 mmol), Hunig's base (0.3 mL, 2 mmol), then HATU (460 mg, 1 .2 mmol) in 5 mL DMF was added into the resultant solution by syringe pump over 2 hours, the resultant solution was concentrated and isolated directly by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as modifier. The product was obtained as an oil. lon(s) found by LCMS: [M+2H/2]=1 177.1 , [M+3H/3]=785.1 , [M+4H/4]=589.1 , [M+5H/5]=471 .4.
Step c. Preparation of (25S)-25-[bis(15-oxo-21-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2- yl]oxy}-3,6,9,12,19-pentaoxa-16-azahenicosan-1-yl)carbamoyl]-26-(carboxymethyl)-7,23-dioxo-22- (15-oxo-21-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}-3,6,9,12,19-pentaoxa-16- azahenicosan-1-yl)-1-{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}-3,10,13,16,19- pentaoxa-6,22,26-triazaoctacosan-28-oic acid
Benzyl (25S)-26-[2-(benzyloxy)-2-oxoethyl]-25-[bis(15-oxo-21 -{[(2R,3R,4R,5R,6S)-3,4,5- trihydroxy-6-methyloxan-2-yl]oxy}-3,6,9,12,1 9-pentaoxa-16-azahenicosan-1 -yl)carbamoyl]-7,23-dioxo-22- (15-0X0-21 -{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}-3, 6,9, 12,19-pentaoxa-1 6- azahenicosan-1 -yl)-1 -{[(2R,3R,4R,5R,6S)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy}-3,10,13,1 6,1 9- pentaoxa-6,22,26-triazaoctacosan-28-oate (120 mg, 0.05 mmol) was dissolved into 5 mL 5% H20 in MeOH, then 20 mg of 5% Palladium on charcoal was added above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere for 2hours. The palladium charcoal was removed by filtration, and the filtrate was concentrated and used for next step without purification.
Step d. Preparation of Compound 96
The title compound was prepared analogously using the standard procedure A and B of Exampli 102. lon(s) found by LCMS: [M+4H/4]=1 136.9, [M+5H/5]=909.7, [M+6H/6]=758.3, [M+7H/7]=650.1 , [M+8H/8]=568.9.
Example 182: Synthesis of Compound 97
Figure imgf000393_0001
Step a. Synthesis of Asn-INT-16
To a solution of INT-16 (464 mg, 0.34 mmol) and Z-L-asparagine (109.2 mg, 0.41 mmol) in anhydrous NMP (2 mL) was added HATU (155.8 mg, 0.41 mmol). The resulting mixture was stirred for 15 minutes, then added with DIPEA (65 mg, 0.5 mmol). After the reaction was stirred for 1 hour, it was diluted with MeOH (~ 15 mL) and treated with Pd/C. The mixture was stirred under hydrogen overnight. Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation. The residue was purified by RPLC (50 g, 15 to 80 % MeOH and water). Yield 296.9 mg, 59.1 %. Ion found by LCMS: [(M - Boc + 2H)/2]+ = 689. Step b. Synthesis of octa-Boc benzyl bis(2-amino-2-oxoethyl)carbamate Compound 97 precursor
To a solution of step-a product (150 mg, 0.1015 mmol) and (2S,2'S)-2,2'- ({[(benzyloxy)carbonyl]azanediyl}bis[(1 -oxoethane-2,1 -diyl)azanediyl])dioctanoic acid (27.5 mg, 0.05 mmol) in anhydrous DMF (1 mL) was added with DIPEA (65 mg, 0.5 mmol) and drop-wise added with a solution of HATU (20.9 mg, 0.55 mmol) in DMF (0.5 mL) via syringe pump at a rate of 1 ml/hr. The reaction was stirred for 15 more minutes and directly purified by RPLC (50 g, 20 to 100 % MeOH and water). Yield 1 16.6 mg, 67.2 %. Ions found by LCMS: [(M - 3Boc + 3H)/3]+ = 1057, [(M - 4Boc + 3H)/3]+ =
1023, [(M - 5Boc +3H)/3]+ = 990.
Step c. Removed of the Cbz protection
The step-b product was dissolved in MeOH (20 mL) and Pd/C was added. The mixture was stirred under hydrogen overnight. Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation to dryness. Yield 104.6 mg, 93.3%. Ions found by LCMS: [(M - 2Boc + 3H)/3]+ = 1 045, [(M - 3Boc + 3H)/3]+ = 1012, [(M - 4Boc + 3H)/3]+ = 978.
Step d. Synthesis of Octa-Boc-Compound 97 precursor
A mixture of step-c product (104.6 mg, 0.0314 mmol) and INT-2 (71 mg, 0.2 mmol) was dissolved in anhydrous DMF (1 mL) and DIPEA (42 mg). After the solution was cooled in an ice-water bath, it was drop-wise added with a solution of HATU (76 mg, 0.2 mmol) in DMF (0.5 mL) via syringe pump at a rate of 1 ml/hr. The reaction was stirred for 15 more minutes and directly purified by RPLC (50 g, 20 to 100 % MeOH and water). Yield 1 15 mg, 96.7 %. Ions found by LCMS: [(M - 3Boc + 3H)/3]+ = 1 123, [(M - 6BOC + 3H)/3]+ = 1023.
Step e. Removal of the Boc protection
Step-d product (1 1 5 mg, 0.031 mmol) was dissolved in TFA:DCM (1 :1 , - 1 mL). The solution was stirred for 15 minutes, then directly purified by RPLC (50 g, 0 to 35 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 45 mg, 38.4 %. Ions found by LCMS: [(M + 3H)/3]+ =956, [(M + 4H)/4]+ = 717, [(M + 5H)/5]+ = 574, [(M + 6H)/6]+ = 478. Example 183: Synthesis of Compound 98
Figure imgf000395_0001
The title compound was prepared analogously to Compound 97. Ions found by LCMS: [(M + 3H)/3]+ = 947, [(M + 4H)/4]+ = 710, [(M + 5H)/5]+ = 568, [(M + 6H)/6]+ = 474.
Example 184: Synthesis of Compound 99
Figure imgf000395_0002
The title compound was prepared from INT-44 and INT-54 in a similar manner as described in Example 37. Yield: 52%. LC/MS [M+4H/4]+ = 884.8. xample 185: Synthesis of Compound 100
Figure imgf000396_0001
The title compound was prepared analogously to Compound 97. Ions found by LCMS: [(M + 3H)/3]+ = 956.7, [(M + 4H)/4]+ = 718, [(M + 5H)/5]+ = 574.
Example 186: Synthesis of Compound 101
Figure imgf000396_0002
INT-59 (0.289 g, 0.0985 mmol), INT-18 (0.323 g, 0.207 mmol) and DIPEA (0.051 mL, 0.30 mmol) were mixed in 5 mL of DMF. HATU (0.0936 g, 0.246 mmol) was added via syringe pump (1 mL, 1 ml/hr). The reaction solution was stirred for another hour after addition. DMF was removed and the residue was purified by prep-HPLC to give per-Boc-Compound 91 , 0.281 g (47%). Positive mass ions were found as (M - 4Boc + 8H+)/8: 703.6, (M - 4C6Hii04 + 5H+)/5: 1087.0, tr = 2.09 minutes with 8 min (40-95%) method. The Boc groups of Per-Boc-Compound 1 01 were removed by treating with TFA for less than 1 hour. Most of the TFA was removed and the residue was dissolved in a minimum amount of NMP and loaded onto a Preparative HPLC with ACN/water (0.1 % TFA). The desired product was collected and lyophilized to give the product as a white powder of TFA salt. Positive mass ions were found as [(M - 4C6Hii04 + 5H+)/5: 888.3, (M -3C6Hii04 + 5H+)/5: 917.7, (M - 2C6Hii04 + 5H+)/5: 947.2, (M - C6Hii04 + 5H+)/5: 976.8, (M + 5H+)/5: 1005.7, (M + 4H+)/4: 1256.7] at tr = 2.26 min with 8 min (5-95%) method for Compoud 101 .
Example 187: Synthesis of Compound 102
Figure imgf000397_0001
The title compound was prepared analogously to compound Compound 97. Ions found by LCMS: [(M + 4H)/4]+ = 733.9, [(M + 5H)/5]+ = 587, [(M + 6H)/6]+ = 489, [(M + 7H)/7]+ = 420.
Figure imgf000398_0001
Step a. Preparation of methyl (2S)-2-({A/6-[(benzyloxy)carbonyl]-A -(fert-butoxycarbonyl)-L- lysyl }am i no)octanoate
Methyl (2S)-2-amino-octanoate (350 mg, 2 mmol), ferf-butyl /^-[(benzyloxyJcarbonylJ-L-lysinate
(770 mg, 2 mmol), EDC (500 mg, 2.5 mmol), HOBT (450 mg, 3 mmol), Hunig's base (0.7 mL, 5 mmol) were mixed in 10 mL DMF at room temperature. The solution was stirred overnight. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 1 00% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product 460 mg (75%) was obtained as an oil. lon(s) found by LCMS: M+H = 536.3. Step b. Preparation of methyl (2¾-2-{[(2S)-6-{[(benzyloxy)carbonyl]amino}-2-({[(2S)-1-methoxy-1- oxooctan-2-yl]carbamoyl}amino)hexanoyl]amino}octanoate
Methyl (2S)-2-({/V6-[(benzyloxy)carbonyl]-/v2-(ferf-butoxycarbonyl)-L-lysyl}amino)octanoate (540mg, 1 .0 mmol) was treated with 2 mL trifluoroacetic acid in 2mL DCM and stirred for half hour and then concentrated and dried under high vacuum used for next step without purification.
The deprotected products from the above step were dissolved into 5 mL DMF and methyl (2S)-2- isocyanatooctanoate (200 mg, 1 mmol) and triethylamine (0.7 mL, 5 mmol) were added at room temperature. The reaction was stirred overnight then concentrated and the residue was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. Yield 460 mg, 75%. lon(s) found by LCMS: M+H = 635.4.
Step c. Preparation of methyl (2S)-2-({[(2S)-6-amino-1-{[(2S)-1-methoxy-1-oxooctan-2-yl]amino}-1- oxohexan-2-yl]carbamoyl}amino)octanoate
Methyl (2S)-2-{[(2S)-6-{[(benzyloxy)carbonyl]amino}-2-({[(2S)-1 -methoxy-1 -oxooctan-2- yl]carbamoyl}amino)hexanoyl]amino}octanoate (150 mg, 0.24 mmol) was dissolved into 5 mL 5% H20 in MeOH, then 50 mg of 5% Palladium on charcoal was added above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere for 2hours. The palladium charcoal was removed by filtration. The residue was used for next step without purification. Step d. Preparation of methyl (2S)-2-{[(2S)-6-{4-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-4-oxobutanamido}-2-({[(2S)-1 -methoxy-1 -oxooctan-2- yl]carbamoyl}amino)hexanoyl]amino}octanoate
The methyl (2S)-2-({[(2S)-6-amino-1 -{[(2S)-1 -methoxy-1 -oxooctan-2-yl]amino}-1 -oxohexan-2- yl]carbamoyl}amino)octanoate from previous step was redissoved into 5mL DMF, followed by addition of INT-2 (83 mg, 0.24 mmol), EDC (100 mg, 0.5 mmol), HOBT (80 mg, 0.5 mmol), Hunig's base (0.14 mL, 1 mmol) at room temperature. The solution was stirred overnight. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil. lon(s) found by LCMS: M+H = 834.5.
Step e. Preparation of (2S)-2-[({(16S,19S)-19-carboxy-1-[(6-deoxy-alpha-L-mannopyranosyl)oxy]- 7,10,17-trioxo-3-oxa-6,11 ,18-triazapentacosan-16-yl}carbamoyl)amino]octanoic acid
Methyl (2S)-2-{[(2S)-6-{4-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-4- oxobutanamido}-2-({[(2S)-1 -methoxy-1 -oxooctan-2-yl]carbamoyl}amino)hexanoyl]-amino}octanoate (0.050 g, 0.06 mmol) in methanol (1 mL) was treated with a solution of lithium hydroxide (0.012 g, 0.5 mmol), in water (1 mL) and THF (1 mL), then stirred at room temperature for 30 minutes. The reaction was quenched by drops of acetic acid. The desired product was isolated directly by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using 0.1 % Trifluoacetic acid as modifier. Yield of oil product 48mg, 100% yields. lon(s) found by LCMS: M+H = 806.5.
Step f. Preparation of Compound 103
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3]=965.9 [M+4H/4]=724.7, [M+5H/5]=579.9, [M+6H/6]=483.5.
Figure imgf000400_0001
Step a. Synthesis of 2,2'-({2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2- oxoethyl}azanediyl)diacetic acid
Lithium hydroxide (480 mg, 20 mmol) in 10mL H2O was added to the solution of diethyl 2,2'-({2- [(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}azanediyl)-diacetate (4.80 g, 10 mmol) in mix solvent of 10 mL H2O, 20 mL MeOH and 20 mL THF. The resultant solution was stirred for 1 hour at room temperature. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoro-acetic acid as the modifier. The product was obtained as an oil. LC/MS 425.2 [M+H]+ Step b. Synthesis of dibenzyl 14-{2-[(2-{2-[(6-deoxy-alpha-L- mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}-4,12,16,24-tetraoxo-8,20-dioxa- 5,11 ,14,17,23-pentaazaheptacosane-1 ,27-dioate
To the solution 2,2'-({2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2- oxoethyl}azanediyl)diacetic acid (1 .3 g, 3 mmol) and 4-(benzyloxy)-4-oxobutanoic acid (1 .8 g, 6.1 mmol) in 30 mL DMF was added EDC (1 .4 g, 7 mmol), HOBT (1 .0 g, 7 mmol), Hunig's base (1 .4 mL, 10 mmol) at room temperature. The solution was stirred overnight. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid
chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil. LC/MS 977.5 [M+H]+
Step c. Synthesis of 14-{2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2- oxoethyl}-4,12,16,24-tetraoxo-8,20-dioxa-5,11 ,14,17,23-pentaazaheptacosane-1 ,27-dioic acid
Dibenzyl 14-{2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}- 4,12, 16,24-tetraoxo-8,20-dioxa-5, 1 1 ,14,17,23-pentaazaheptacosane-1 ,27-dioate (816 mg, 0.836 mmol) was dissolved into 5 mL MeOH and 5 mL ethyl acetate, then 0.5 g of 5% Palladium on charcoal was added above solution, and the mixture was stirred at room temperature under the hydrogen atmosphere overnight. The palladium charcoal was removed by filtration after completed the reaction by LCMS. The filtrate was concentrated and used next step without any purification. LC/MS, 797.4 [M+H]+ .
Step d. Synthesis of Compound 104
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3]=1056.9, [M+4H/4] =792.9, [M+5H/5]=634.6, [M+6H/6]=529.0, [M+7H/7]=453.5.
Example 190: Synthesis of Compound 105
Figure imgf000401_0001
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3]= 1020.2, [M+4H/4] =776.2, [M+5H/5]=613.1 , [M+6H/6]=51 1 .1 .
Figure imgf000402_0001
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3]=924.2, [M+4H/4]=693.4, [M+5H]+1 =554.9, [M+6H/6]=462.6.
Figure imgf000402_0002
The title compound was prepared from INT-38a and INT-65 in a similar manner as described for the synthesis of Compound 64. Yield: 66 %. LC/MS [M+3H/3]+ = 861 .4.
Figure imgf000403_0001
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3]=1023.3, [M+4H/4]=768.4, [M+5H/5]=614.9, [M+6H/6]=512.6.
Figure imgf000404_0001
Step a. Synthesis of D-Ser-INT-11
Z-D-Ser-OH (435.3 mg, 1 .82 mmol) and INT-1 1 (1 .6 g, 1 .51 mmol) were dissolved in anhydrous DMF (2 mL). It was cooled in an ice-water bath and mixed with DIPEA (260 mg) followed by dropwise addition of a solution of HATU (722 mg, 1 .9 mmol) in DMF (2 mL) via syringe pump at a rate of 4 ml/hr. After the addition, the reaction was directly purified by RPLC (100 g, 30 to 85 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid (Ion found by LCMS: [(M - 2Boc + 2H)/2]+ = 542). The material was re-dissolved in MeOH (~ 30 ml_) and added with Pd/C. The mixture was stirred under hydrogen overnight. The Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation and further dried under high vacuum. Yield 1 .23 g, 70.7 %. Ion found by LCMS: [(M -Boc + 2H)/2]+ = 525.
Step b. Synthesis of Thr-(D)-Ser-INT-11
Z-thr-OH (329.3 mg, 1 .3 mmol) and step-a product (1 .23 g, 1 .07 mmol) were dissolved in anhydrous DMF (2 ml_). After the solution was cooled in an ice-water bath, it was mixed with HATU (418 mg, 1 .1 mmol). The mixture was stirred for 10 minutes, then added with DIPEA (195 mg, 1 .5 mmol). The reaction was stirred for 1 hour and directly purified by RPLC (100 g, 30 to 85 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid (Ion found by LCMS: [(M - 2Boc + 2H)/2]+ = 593). The material was re-dissolved in MeOH (~ 30 mL) and added with Pd/C. The mixture was stirred under hydrogen overnight. The Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation and further dried under high vacuum. Yield 1 .03 g, 77 %. Ion found by LCMS: [(M - Boc + 2H)/2]+ = 576.
Step c. Synthesis of Dab-Thr-(D)-Ser-INT-11
Z-thr-OH (123.5 mg, 0.487 mmol) and step-b product (508 mg g, 0.406 mmol) were dissolved in anhydrous DMF (1 mL) and mixed with HATU (190 mg, 0.5 mmol). After the mixture was stirred for 10 minutes, DIPEA (195 mg, 1 .5 mmol) was added. The reaction was stirred for 1 hour and directly purified by RPLC (100 g, 30 to 85 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid (Ion found by LCMS: [(M - 3Boc + 2H)/2]+ = 593). The material was re- dissolved in MeOH (~ 30 mL) and added with Pd/C. The mixture was stirred under hydrogen overnight. The Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation and further dried under high vacuum. Yield 448.7 mg, 81 .8 %. Ion found by LCMS: [(M - 2Boc + 2H)/2]+ = 576.
Step d. Synthesis of hexa-Boc-Compound 109 precursor
To a solution of step-c product (273.2 mg, 0.2 mmol) and INT-43 (46 mg, 0.096 mmol) in anhydrous DMF (1 mL) and DIPEA (65 mg, 0.5 mmol) was drop-wise added a solution of HATU (76 mg, 0.2 mmol) in DMF (0.5 mL) via syringe pump at a rate of 1 ml/hr. The reaction was stirred for 15 more minutes and directly purified by RPLC (50 g, 20 to 100 % MeOH and water). Yield 248 mg, 75.6 %. Ions found by LCMS: [(M - Boc + 3H)/3]+ = 1046.7, [(M - 3Boc + 3H)/3]+ = 1 038, [(M - 5Boc + 3H)/3]+ = 971 .
Step e. Removal of the Boc protection
Step-d product (248 mg, 0.072 mmol) was dissolved in TFA (~1 mL). The solution was stirred for
15 minutes, then directly purified through HPLC (5 to 26 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 71 .4 mg, 29.4 %. Ions found by LCMS: [(M + 4H)/4]+ =704, [(M + 5H)/5]+ = 563.7. Example 195: Synthesis of Compound 110
Figure imgf000406_0001
Step a. Synthesis of Methyl (2S)-2-Aminooctanoate
(2S)-2-Aminooctanoic acid (3.0 g, 18.84 mmol) was stirred and heated in methanol (40 mL) and sulfuric acid (2.0 mL) at 75 °C in a sealed tube overnight. Next day, the reaction mixture was cooled down to room temperature then with vigorously stirring the reaction was quenched with saturated NaHCCb aqueous solution (50 mL). Upon the gas evolution ceased and pH 6, methanol was removed under reduced pressure. The aqueous layer was extracted with ethyl acetate (3 x 30 mL). The combined organic layers were washed with brine, dried over solid Na2S04 and filtered then concentrated. The residue was used in the next step without purification. Ion found by LC/MS [M+H]+ = 174.2.
Step b. Synthesis of pyrrolidine intermediate (a) more polar isomer and pyrrolidine intermediate (b) less polar isomer
To a solution of racemic-trans1 -(tert-butoxycarbonyl)pyrrolidine-3,4-dicarboxylic acid (0.3 g, 1 .16 mmol), methyl (2S)-2-aminooctanoate (0.44 g, 2.55 mmol), and diisopropylethylamine (896.6 mg, 6.94 mmol) in DMF (5.7 mL) was added a solution of HATU (967.95 mg, 2.55 mmol) in DMF (4 mL) via a syringe pump at a rate of 2.5 mL/hr. After the addition, the reaction mixture was stirred for an additional of 1 h at room temperature. The reaction mixture was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5 to 50% acetonitrile/water, using 0.1 % TFA as modifier to yield two diastereomers: the more polar diastereomer (a) (381 .8 mg, 58%) and the less polar diastereomer (b) (344.3 mg, 52%). Ion found by LC/MS [M-Boc+H]+ = 470.4 (loss of 1 boc group) for both Boc-protected compounds, methyl 2-[((3R,4R)-4-{N[(1 S)-1 - (methoxycarbonyl)heptyl]carbamoyl}-1 -[(tert-butyl)oxycarbonyl]pyrrolidine-3- yl)carbonylamino](2S)ocatanoate. Note: the isomers were taken forward separately in subsequent syntheses. Step c. Removal of the Boc Group
The Step-b product (more polar isomer) (381 .8 mg, 0.67 mmol) was stirred in TFAiChteC (1 :1 , 12 ml_) at room temperature then volatiles were removed under reduced pressure. The residue was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid
chromatograph eluted with 5 to 100% methanol and Dl water to yield the title compound (201 .0 mg, 64%). Ion found by LC/MS [M+H]+ = 470.4.
Step d. Acylation with I NT- 2
A solution of HATU (198.21 mg, 0.52 mmol) in DMF (0.8 mL) was added via a syringe pump at a rate of 2.5 mL/hr into a solution of the Step-c Product (204 mg, 0.43 mmol), INT-2 (183.15 mg, 0.52 mmol), diisopropylethylamine (168.29 mg, 1 .30 mmol) in DMF (4 mL). After the addition, the reaction mixture was stirred for an additional hour then purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5 to 80% acetonitrile and water, using 0.1 % TFA as modifier to yield the title compound (221 .5 mg, 64%). Ion found by LC/MS [M+H]+ = 803.4. Step e. Hydrolysis of the Methyl Esters
The Step-d Product (222.0 mg, 0.28 mmol) and LiOH (17.8 mg, 0.74 mmol) were stirred in MeOH:THF:H20 (1 :1 :2, 16 mL) at room temperature for 3 hours. After the reaction was complete, it was quenched with acetic acid to pH 4 then it was concentrated. The residue was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0 to 45% acetonitrile and water, using 0.1 % TFA as modifier) to yield the title compound (161 mg, 75%). Ion found by LC/MS [M-H]+ = 773.3.
Step f. Synthesis of Compound 110
A solution of HATU (53.98 mg, 0.14 mmol) in DMF (0.4mL) was added via a syringe pump at a rate of 2.5 mL/hr into a solution of the Step-e Product (50.0 mg, 0.065 mmol), INT-18 (221 .99 mg, 0.14 mmol) and diisopropylethylamine (50 mg, 0.39 mmol) in DMF (1 .3 mL). The reaction mixture was stirred for an additional hour then purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0 to 30 to 00% methanol and water to yield the Boc- protected intermediate. The Boc-protected intermediate was stirred in TFA:DCM (1 :1 , 6 mL) at room temperature for 20 minutes. After the reaction was completed, the reaction mixture was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid
chromatograph eluted with 5 to 45% acetonitrile and water, using 0.1 % TFA as modifier to yield the title compound (1 85.7 mg, 72%). lon(s) found by LC/MS [(M+4H)/4]+ = 71 6.8, [(M+5H)/5]+ = 573.73 and [(M+6H)/6]+ = 478.28.
Example 196: Synthesis of Compound 111
Figure imgf000408_0001
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3] = 1033.6, [M+4H/4] =776.2, [M+5H/5]=620.2, [M+6H/6]=517.8.
Figure imgf000408_0002
The title compound was prepared analogously to Example 37, where Cbz-L-aminodecanoic acid was used in place of Cbz-L-amino-octanoic acid for the first step of that sequence, lon(s) found by LCMS: (M+3H)/3 = 965.3, (M+4H)/4 = 724.3, (M+5H)/5 = 579.7
Figure imgf000409_0001
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3] = 1020.2, [M+4H/4] =776.2, [M+5H/5]=613.1 , [M+6H/6]=51 1 .1 .
Example 199: Synthesis of Compound 114
Figure imgf000409_0002
A solution of HATU (50.06 mg, 0.13 mmol) in DMF (0.4 mL) was added via a syringe pump at a rate of 2.5 mL/hr into a solution of INT-64 (191 .0 mg, 0.13 mmol), INT-66 (48.30 mg, 0.062 mmol) and diisopropylethylamine (48.58 mg, 0.38 mmol) in DMF (1 mL). After the addition, the reaction was stirred for an additional hour at room temperature then purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0 to 30 to100% methanol and water, using 0.1 % TFA as modifier to yield the Boc-protected intermediate. The Boc-intermediate was stirred in TFA:CH2Cl2 (1 :1 , 6 mL) at room temperature for 30 minutes then the volatiles were removed under reduced pressure. The residue was purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5 to 45% acetonitrile and water, using 0.1 % TFA as modifier to yield the title compound as TFA salt (89.4 mg, 38%). lon(s) found by LC/MS [(M+4H)/4]+ = 710.6, [(M+5H)/5]+ = 568.8, [(M+6H)/6]+ = 474.2.
Figure imgf000410_0001
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3] = 1006.3, [M+4H/4] =754.7, [M+5H/5]=604.8, [M+6H/6]=504.1 .
Figure imgf000411_0001
Step a. Synthesis of (2¾-2-{[(benzyloxy)carbonyl]amino}-4-(ureidoureido)butanoic acid
A suspension of (S)-4-amino-2-(benzyloxycarbonylamino)butyric acid (1 .01 g, 4 mmol) in water (4 mL) was heated to dissolved. After cooled to room temperature, the solution was added with potassium cyanate (648.8 mg, 8 mmol). The reaction was stirred overnight, then directly purified by RPLC (1 00 g, 0 to 30 % acetonitrile and water). Yield 966 mg, 81 .8 %. Ion found by LCMS: [M + H]+ = 296. Step b. Synthesis of 4-(ureido)butanamide-INT-11
To a solution of INT-1 1 (618 mg, 0.6 mmol) and the step-a product (212.4 mg, 0.72 mmol) in anhydrous DMF (1 mL) and DIPEA (1 10 mg, 0.85 mmol) was drop-wise added a solution of HATU (273.6 mg, 0.72 mmol) in DMF (1 mL) via syringe pump at a rate of 2 ml/hr. After the addition, the mixture was purified by RPLC (100 g, 1 5 to 85 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid (Ions found by LCMS: [(M - 2 Boc + 2H)/2]+ = 570, [(M - 3 Boc + 2H)/2]+ = 520). The material was dissolved in MeOH (~ 25 mL) and added with Pd/C. The mixture was stirred under hydrogen overnight. Pd/C was filtered off, and the filtrate was concentrated by rotary evaporation to dryness. Yield 520 mg, 71 .9 %. Ions found by LCMS: [(M - Boc + 2H)/2]+ = 553. Step c. Synthesis of octa-Boc-Compound 116 precursor
To a solution of the step-b product (240.5 mg, 0.2 mmol) and INT-40 (124.3 mg, 0.092 mmol) in anhydrous DMF (1 mL) and DIPEA (65 mg, 0.5 mmol) was drop-wise added a solution of HATU (76 mg, 0.2 mmol) in DMF (1 mL) via syringe pump at a rate of 2 ml/hr. After the addition, the mixture was purified by RPLC (100 g, 20 to 95 % MeOH and water). Yield 206.8 mg, 60.3%. Ions found by LCMS: [(M - 2Boc + 3H)/3]+ = 1 175.7, [(M - 3Boc + 3H)/3]+ = 1 142, [(M - 4Boc + 3H)/3]+ = 1 109, [(M - 5Boc + 3H)/3]+ = 1075, [(M - 6Boc + 3H)/3]+ = 1042.
Step d. Removal of the Boc group
Step-c product (206.8 mg, 0.056 mmol) was dissolved in TFA:DCM (1 :1 , ~ 1 .5 mL). The solution was stirred for 15 minutes and purified through HPLC (5 to 25 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 62.8 mg, 29.5 %. Ions found by LCMS: [(M + 3H)/3]+ = 975.3, [(M + 4H)/4]+ = 731 .9, [(M + 5H)/5]+ = 585.7, [(M + 6H)/6]+ = 488.3.
Example 202: Synthesis of Compound 117
Figure imgf000412_0001
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3] = 1006.3, [M+4H/4] =754.7, [M+5H/5]=604.8, [M+6H/6]=504.1 .
Figure imgf000413_0001
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3]=915.5, [M+4H/4]=686.9, [M+5H/5]=572.6, [M+6H/6]=477.3.
Example 204: Synthesis of Compound 119
Figure imgf000413_0002
The title compound was prepared in an analogous fashion to the procedure used to prepare Compound 120. lon(s) found by LCMS: [M+3H/3]=1043.3, [M+4H/4]=783.5, [M+5H/5]=627.0,
[M+6H/6]=522.6, [M+7H/7]=448.1 .
Figure imgf000414_0001
Step a. Preparation of benzyl 12-[2-(benzyloxy)-2-oxoethyl]-1-[(6-deoxy-alpha-L- mannopyranosyl)oxy]-9-{2-[(2-{2-[(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)-amino]-2- oxoethyl}-7-oxo-3-oxa-6,9,12-triazatetradecan-14-oate
2,2'-[(2-{bis[2-(benzyloxy)-2-oxoethyl]amino}ethyl)azanediyl]diacetic acid (200 mg, 0.4 mmol) was dissoved into 10 mL DMF, followed by addition of INT-1 (250 mg, 1 mmol), EDC (200 mg, 1 mmol), HOBT (150 mg, 1 mmol), Hunig's base (0.28 mL, 2 mmol) at room temperature. The solution was stirred overnight. The resultant solution was concentrated and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1 % trifluoroacetic acid as the modifier. The product was obtained as an oil. lon(s) found by LCMS: M+H = 939.4. Step b. Preparation of 12-(carboxymethyl)-1-[(6-deoxy-alpha-L-mannopyranosyl)oxy]-9-{2-[(2-{2- [(6-deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)amino]-2-oxoethyl}-7-oxo-3-oxa-6,9,12- triazatetradecan-14-oic acid
Benzyl 12-[2-(benzyloxy)-2-oxoethyl]-1 -[(6-deoxy-alpha-L-mannopyranosyl)oxy]-9-{2-[(2-{2-[(6- deoxy-alpha-L-mannopyranosyl)oxy]ethoxy}ethyl)-amino]-2-oxoethyl}-7-oxo-3-oxa-6,9,12- triazatetradecan-14-oate (200 mg, 0.02 mmol) was dissolved into 10 mL 5% H20 in MeOH, then 100 mg of 5% Palladium on charcoal was added, and the mixture was stirred at room temperature under a hydrogen atmosphere for 2 hours. The palladium charcoal was removed by filtration, and the filtrate was concentrated and used for next setp without purification, lon(s) found by LCMS: M+H=759.3.
Step c. Preparation of Compound 120
The title compound was prepared analogously using the standard procedure A and B of Example 102. lon(s) found by LCMS: [M+3H/3]=1035.6, [M+4H/4]=776.9, [M+5H/5]=621 .7, [M+6H/6]=517.3, [M+7H/7]=444.4.
Figure imgf000415_0001
The title compound was prepared analogously using the standard procedure A and B of Exampli 102. lon(s) found by LCMS: [M+3H/3]=1048.3, [M+4H/4] =786.4, [M+5H/5]=629.4, [M+6H/6]=524.6, [M+7H/7]=449.8.
Example 207: Synthesis of Compound 121
Figure imgf000416_0001
Step a. Synthesis of methyl 3-(aminomethyl)-5-bromobenzoate
In a sealed tube, a stirring mixture of methyl 3-bromo-5-(bromomethyl)benzoate (3.08 g, 10.00 mmol) and sodium azide (0.715 g, 1 1 .00 mmol) in DMF (10 mL) was heated at 80 °C for 1 h, when HPLC analysis showed full conversion of the bromide. All the volatiles were evaporated per vacuum techniques. The residue was taken up in DCM (40 mL) and water (40 mL). The water layer was washed with additional DCM (2 x 30 mL). The combined organic layers were dried with brine and solid sodium sulfate. Upon filtration, all the volatiles were evaporated per vacuum techniques, affording 2.73 g of the crude azide derivative in high purity, as confirmed per 1 H-NMR and LC-MS analysis. 1 H NMR (DMSO-cfe) δ: 8.03 (t, J = 1 .6 Hz, 1 H), 7.98 - 7.93 (m, 1 H), 7.90 (t, J = 1 .5 Hz, 1 H), 4.60 (s, 2H), 3.88 (s, 3H). This azide (2.73 g) was dissolved under stirring in THF (30 mL) and water (2 mL), and triphenylphosphine (3.148 g, 12.00 mmol) was added. The reaction was stirred at room temperature overnight, allowing gas evolution through a bubbler. All the volatiles were evaporated per vacuum techniques. The desired product was isolated by normal phase liquid chromatography using an Isco CombiFlash liquid chromatograph eluted with 1 % to 20% hexanes and ethylacetate. From the desired fractions, the desired compound was obtained still contaminated with triphenylphenylphosphine oxide. This material was therefore dissolved in DCM (50 mL) and extracted an acidic solution (12 mL of 1 .0 M H2SO4 and 30 mL of water). The water layer was added dropwise to a vigorously stirring solution of saturated sodium bicarbonate (60 mL) and stirring was continued overnight. The obtained mixture was extracted with DCM (3 x 30 mL). The combined organic layers were dried with brine and solid sodium sulfate. Upon filtration, all the volatiles were evaporated per vacuum techniques, affording 1 .31 g, 54%. Ions found by LCMS: (M+H)+ = 244.0; 246.0. Step b. Synthesis of methyl 3-((N-(1 '-(3'-carboxymethyl-5-bromo)phenylene)methylene)methyl)-5- bromobenzoate
A solution of methyl 3-(aminomethyl)-5-bromobenzoate (1 .306 g, 5.350 mmol) and methyl 3- bromo-5-formylbenzoate (1 .238 g, 5.096 mmol) in DCM (10 mL) was evaporated per vacuum techniques and left over high vacuum overnight. The residue was taken up in THF (30 mL) and treated under vigorous stirring with sodium triacetoxyborohydride (3.240 g, 15.29 mmol). This mixture was quenched after 18 h by the addition of a saturated aqueous solution of ammonium chloride (50 mL). The obtained mixture was extracted with DCM (3 x 30 mL). The combined organic layers were dried with brine and solid sodium sulfate. Upon filtration, all the volatiles were evaporated. The desired product was isolated by normal phase liquid chromatography using an Isco CombiFlash liquid chromatograph eluted with 1 % to 50% hexanes and ethylacetate. Yield 1 .61 1 g, 67%. Ions found by LCMS: (M+H)+ = 470.0, 472.1 , 474.0.
Step c. Synthesis of N-Cbz protected methyl 3-((N-(1 '-(3'-carboxymethyl-5- bromo)phenylene)methylene)methyl)-5-bromobenzoate
To a 0°C stirring solution of methyl 3-((N-(1 '-(3'-carboxymethyl-5- bromo)phenylene)methylene)methyl)-5-bromobenzoate (1 .61 1 g, 3.419 mmol) and DIPEA (0.953 mL, 5.471 mmol) in THF (20 mL) it was added Cbz-CI (0.732 mL, 5.129 mmol) dropwise. The temperature was raised to ambient after 10 minutes and stirring was continued until complete disappearance of the starting amine by LC-MS analysis. All the volatiles were removed per rotatory evaporation. The desired product was isolated by normal phase liquid chromatography using an Isco CombiFlash liquid chromatograph eluted with 1 % to 20% hexanes and ethylacetate. Yield 2.070 g, quant. Ions found by LCMS: (M+H)+ = 603.0, 605.0, 607.0. Step d. Synthesis of N-Cbz protected 3-((N-(1 '-(3'-carboxy-5-phenyl)phenylene)methylene)methyl)- 5-phenylbenzoic acid
Under nitrogen, in a capped microwave reaction vessel stirring mixture of N-Cbz protected methyl 3-((N-(1 '-(3'-carboxymethyl-5-bromo)phenylene)methylene)methyl)-5-bromobenzoate (0.299 g, 0.494 mmol), potassium phenyltrifluoroborate (0.200 g, 1 .087 mmol), sodium carbonate (0.209 g, 1 .976 mmol) and bis(triphenylphosphine)palladium (II) dichloride (0.035 g, 0.049 mmol) in MeCN (4 mL) and water (4 mL) were irradiated with microwaves for 10 mim at 130 °C. Upon cooling, the reaction was treated under stirring with SiliaMetS® (0.200 g, 0.240 mmol) for 1 h, and TFA was added (0.380 mL, 5.0 mmol). Upon filtration, all the volatiles were evaporated per vacuum techniques. The desired product was isolated as a dodeca-TFA salt by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% methanol and water, using TFA as the modifier. Yield 0.072 g, 25% yield. 1 H NMR (0.500 mL Chloroform-d + 0.050 mL TFA) δ: 8.1 8 (d, J = 5.5 Hz, 2H), 7.82 (d, J = 16.6 Hz, 2H), 7.60 (d, J = 33.5 Hz, 2H), 7.52 - 7.27 (m, 15H), 5.32 (s, 2H), 4.70 (d, J = 19.9 Hz, 4H).
Step e. Synthesis of N-Cbz-deca-Boc-(Compound 121)-EM
A stirring solution of INT-18 (0.429 g, 0.241 mmol), N-Cbz protected 3-((N-(1 '-(3'-carboxy-5- phenyl)phenylene)methylene)methyl)-5-phenylbenzoic acid (0.069 g, 0.121 mmol), and DIPEA (0.128 mL, 0.736 mmol) in DMF (10 mL), was treated with a solution of HATU (0.094 g, 0.247 mmol in 3.0 mL of DMF), dropwise over 30 minutes. After 1 .5 hour, all the volatiles were evaporated per vacuum techniques. The desired product was isolated by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 50% to 100% methanol and water, using no modifier. Yield 0.1 58 g, 37% yield. Ion found by LCMS: [(M-3Boc)+3H]/4 = 841 .2. Step f. Synthesis of deca-Boc-(Compound 121)-EM
N-Cbz-deca-Boc-(Compound 121 )-EM (0.1 58 g, 0.0431 mmol) was dissolved methanol (10 ml_) and suspended with SiliaCat® Pd(0) (0.043 g; 0.009 mmol) under hydrogen atmosphere (~1 atm). When all the starting material was consumed by HPLC analysis, the mixture was filtered over a short Celite pad, and all the volatiles were evaporated per vacuum techniques. HPLC analysis revealed the desired compound in high purity, and this material was used without further purification. Yield 0.1 51 g, 99% yield. Ion found by LCMS: [(M-3Boc)+3H]/4 = 808.8.
Step g. Synthesis of deca-Boc-Compound 121
Prepared in a similar fashion to "Step e. Synthesis of N-Cbz-deca-Boc-Compound 121 -EM" from deca-Boc-Compound 121 -EM (0.151 g, 0.043 mmol), INT-9 (0.045 g, 0.128 mmol), 2,4,6-trimethylpyridine (0.034 mL, 0.261 mmol) in DMF (5 mL) by dropwise addition of HATU (0.050 g, 0.130 mmol in 1 .5 mL of DMF). Yield 0.062 g, 38% yield. Diagnostic peaks from 1 H NMR (Methanol-ck) δ: 4.71 (br s, 1 H), 2.64 (br s, 2H). Step h. Synthesis of Compound 121
A solution of deca-Boc-Compound 121 (0.062 g, 0.016 mmol), dissolved in DCM (4 mL) and triethylsilane (0.250 mL), was treated with TFA (2 mL), while stirring at room temperature. After 30 minutes, all the volatiles were evaporated per vacuum techniques. The desired product was isolated as the deca-TFA salt by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% methanol and water, using TFA as the modifier. Yield 0.042 g, 65% yield. Main ions found by HRMS: (M+5H)/5 = 573.0.
Example 208: Synthesis of Compound 122
Figure imgf000418_0001
INT-60 (0.0500 g, 0.1 07 mmol), INT-20 (0.366 g, 0.214 mmol) and DIPEA (0.047 ml_, 0.27 mmol) were mixed in 5 mL of DMF. HATU (0.102 g, 0.269 mmol) was added via syringe pump (1 ml_, 1 ml/hr). The reaction solution was stirred for another hour after addition. DMF was removed and the residue was purified by prep-HPLC to give per-Boc-Compound 122, 0.223 g, 54%. Positive mass ions were found as (M - 4Boc + 8H+)/8: 703.6, (M - CeH C - 6Boc + 4H+)/4: 772.4, tr = 3.21 minutes with 7 min (60-95%) method. The Boc groups of Per-Boc-Compound 122 were removed by treating with TFA for less than 1 hour. Most of the TFA was removed and the residue was dissolved in a minimum amount of NMP and loaded onto a Preparative HPLC with ACN/water (0.1 % TFA). The desired product was collected and lyophilized to give the product as a white powder of TFA salt. Positive mass ions were found as [(M + 5H+)/5: 568.3, (M + 4H+)/4: 710.1 ] at tr = 1 .01 min with 5 min (5-95%) method for Compound 122.
Example 209: Synthesis of Compound 123
Figure imgf000419_0001
INT-61 (0.0500 g, 0.1 04 mmol), INT-20 (0.3556 g, 0.2086 mmol) and DIPEA (0.045 mL, 0.26 mmol) were mixed in 5 mL of DMF. HATU (0.0991 g, 0.261 mmol) was added via syringe pump (1 mL, 1 ml/hr). The reaction solution was stirred for another hour after addition. DMF was removed and the residue was purified by prep-HPLC to give per-Boc-Compound 123, 0.208 g, 51 %. Positive mass ions were found as (M - CeH C - 3Boc + 4H+)/4: 851 .6, (M - CeH C - Boc + 4H+)/4: 901 .0, (M - 2Boc + 3H+)/3: 1216.8, tr = 2.44 minutes with 5 min (40-95%) method. The Boc groups of Per-Boc-Compound 123 were removed by treating with TFA for less than 1 hour. Most of the TFA was removed and the residue was dissolved in a minimum amount of NMP and loaded onto a Preparative HPLC with
ACN/water (0.1 % TFA). The desired product was collected and lyophilized to give the product as a white powder of TFA salt. Positive mass ions were found as [(M + 6H+)/6: 475.0, (M + 5H+)/5: 571 .0, (M + 4H+)/4: 713.5] at tr = 0.6 min with 5 min (5-95%) method for Compound 123.
Other Embodiments
While the disclosure has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the disclosure following, in general, the principles of the disclosure and including such departures from the present disclosure come within known or customary practice within the art to which the disclosure pertains and may be applied to the essential features hereinbefore set forth. All publications, patents and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publijyacation, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety.
What is claimed is:

Claims

1 . A compound described by formula (I):
Figure imgf000421_0001
(I)
wherein M1 comprises a first cyclic heptapeptide comprising a linking nitrogen and M2 comprises a second cyclic heptapeptide comprising a linking nitrogen ;
each E is, independently, a monosaccharide or oligosaccharide moiety;
L' is a linker covalently attached to E and to the linking nitrogen in each of M1 and M2;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof. 2. The compound of claim 1 , wherein L' is described b formula (L):
Figure imgf000421_0002
wherein L is a remainder of L';
A1 is a 1 -5 amino acid peptide covalently attached to the linking nitrogen in M1 or is absent; and A2 is a 1 -5 amino acid peptide covalently attached to the linking nitrogen in M2 or is absent.
3. The compound of claim 1 or 2, wherein the compound is described by formula (II):
Figure imgf000421_0003
(II)
wherein L is a remainder of L';
n is 1 , 2, 3, or 4;
each of R1 , R12, R'1 , and R'12 is, independently, a lipophilic moiety, a polar moiety, or H;
each of R1 1 , R13, R14, R'1 1 , R'13, and R'14 is, independently, optionally substituted C1 -C5 alkamino, a polar moiety, a positively charged moiety, or H;
each of R15 and R'15 is, independently, a lipophilic moiety or a polar moiety; each of R2, R3, R4, R5, R6, R7, R8, R9, R10, R'2, R'3, R'4, R'5, R'6, R'7, R'8, R'9, and R'10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or two R groups on the same or adjacent atoms join to form an optionally substituted 5-8 membered ring;
each of a', b', c', a, b, and c is, independently, 0 or 1 ;
each of N1 , N2, N3, N4, N'1 , N'2, N'3, and N'4 is a nitrogen atom;
each of C1 , C2, C3, C'1 , C'2, and C'3 is a carbon atom,
or a pharmaceutically acceptable salt thereof.
4. The compound of claim 3, wherein the compound comprises at least one optionally substituted 5-8 membered ring formed by joining (i) R2, R3, and C1 ; (ii) R3, R4, N1 , and C1 ; (iii) R5, R6, and C2; (iv) R6, R7, N2, and C2; (v) R8, R9, and C3; (vi) R9, R10, N3, and C3; (vii) R'2, R'3, and C'1 ; (viii) R'3, R'4, N'1 , and C'1 ; (ix) R'5, R'6, and C'2; (x) R'6, R'7, N'2, and C'2; (xi) R'8, R'9, and C'3; or (xii) R'9, R'10, N'3, and C'3.
5. The compound of any one of claims 1 -4, wherein the compound is described by formula (III):
M2 A2 A1 M1
Figure imgf000422_0001
(III)
or a pharmaceutically acceptable salt thereof.
6. The compound of any one of claims 3-5, wherein
each of R1 , R12, R'1 , and R'12 is, independently, a lipophilic moiety;
each of R1 1 , R13, R14, R'1 1 , R'13, and R'14 is, independently, optionally substituted C1 -C5 alkamino, a polar moiety, or a positively charged moiety; and/or
each of R15 and R'15 is, independently, a polar moiety.
7. The compound of any one of claims 3-6, wherein each of R1 and R12 is a lipophilic moiety.
8. The compound of any one of claims 3-6, wherein each of R'1 and R'12 is a lipophilic moiety.
9. The compound of claim 7 or 8, wherein each lipophilic moiety is, independently, optionally substituted C1 -C20 alkyl, optionally substituted C5-C15 aryl, optionally substituted C6-C35 alkaryl, or optionally C5- C10 substituted heteroaryl.
10. The compound of any one of claims 7-9, wherein each lipophilic moiety is, independently, C1 -C8 alkyl, methyl substituted C2-C4 alkyl, (C1 -C10)alkylene(C6)aryl, phenyl substituted (C1 - C10)alkylene(C6)aryl, or alkyl substituted C4-C9 heteroaryl.
1 1 . The compound of any one of claims 7-10, wherein each lipophilic moiety is, independently, benzyl, isobutyl, sec-butyl, isopropyl, n-propyl, methyl, biphenylmethyl, n-octyl, or methyl substituted indolyl.
12. The compound of any one of claims 3-1 1 , wherein each of R1 1 , R13, R14, R'1 1 , R'13, and R'14 is independently optionally substituted C1 -C5 alkamino.
13. The compound of claim 12, wherein each of R1 1 , R13, R14, R'1 1 , R'13, and R'14 is CH2CH2NH2.
14. The compound of any one of claims 3-13, wherein each of R15 and R'15 is a polar moiety.
15. The compound of claim 14, wherein each polar moiety comprises a hydroxyl group, a carboxylic acid group, an ester group, or an amide group.
16. The compound of claim 14 or 15, wherein each polar moiety is hydroxyl substituted C1 -C4 alkyl.
17. The compound of any one of claims 14-16, wherein each polar moiety is CH(CH3)OH.
18. The compound of claim 3, wherein the compound is described by formula (IV):
Figure imgf000424_0001
(IV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl,
or a pharmaceutically acceptable salt thereof.
19. The compound of claim 18, wherein the compound is described by formula (IV-1 ):
Figure imgf000424_0002
(IV-1 )
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl,
or a pharmaceutically acceptable salt thereof.
20. The compound of claim 18, wherein the compound is described by formula (IV-2):
Figure imgf000424_0003
(IV-2) wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl,
or a pharmaceutically acceptable salt thereof.
21 . The compound of claim 3, wherein the compound is described by formula (V):
Figure imgf000425_0001
(V)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2, R6, R'2, and R'6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyi, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,
or a pharmaceutically acceptable salt thereof.
22. The compound of claim 21 , wherein the compound is described by formula (V-1 ):
Figure imgf000425_0002
(V-1 )
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
23. The compound of claim 22, wherein the compound is described by formula (V-2):
Figure imgf000426_0001
(V-2)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
24. The compound of claim 21 , wherein the compound is described by formula (V-3):
Figure imgf000426_0002
(V-3)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
25. The compound of claim 24, wherein the compound is described by formula (V-4):
Figure imgf000426_0003
(V-4) wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
26. The compound of claim 24 or 25, wherein the compound is described by formula (V-5):
Figure imgf000427_0001
(V-5)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
27. The compound of claim 3, wherein the compound is described by formula (VI):
Figure imgf000427_0002
(VI)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2, R6, R8, R'2, R'6, and R'8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyi, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,
or a pharmaceutically acceptable salt thereof.
28. The compound of claim 27, wherein the compound is described by formula (VI-1 ):
Figure imgf000428_0001
(VI-1 )
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
29. The compound of claim 28, wherein the compound is described by formula (VI-2):
Figure imgf000428_0002
(VI-2)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
30. The compound of claim 27, wherein the compound is described by formula (VI-3):
Figure imgf000428_0003
(VI-3) wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
31 . The compound of claim 30, wherein the compound is described by formula (VI-4):
Figure imgf000429_0001
(VI-4)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
32. The compound of claim 30 or 31 , wherein the compound is described by formula (VI-5):
Figure imgf000429_0002
(VI-5)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
33. The compound of claim 3, wherein the compound is described by formula (VII):
Figure imgf000430_0001
(VII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2, R6, R8, R'2, and R'6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,
or a pharmaceutically acceptable salt thereof.
34. The compound of claim 3, wherein the compound is described by formula (VIII):
Figure imgf000430_0002
(VIM)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2, R6, R8, and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,
or a pharmaceutically acceptable salt thereof.
35. The compound of claim 3, wherein the compound is described by formula (IX):
Figure imgf000431_0001
(IX)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2, R6, and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,
or a pharmaceutically acceptable salt thereof.
36. The compound of claim 35, wherein the compound is described by formula (IX-1 ) :
Figure imgf000431_0002
(IX-1 )
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
37. The compound of claim 36, wherein the compound is described by formula (IX-2):
Figure imgf000432_0001
(IX-2)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
38. The compound of claim 3, wherein the compound is described by formula (X):
Figure imgf000432_0002
(X)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2 and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,
or a pharmaceutically acceptable salt thereof.
39. The compound of claim 3, wherein the compound is described by formula (XI):
Figure imgf000433_0001
(XI)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2 and R6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,
or a pharmaceutically acceptable salt thereof.
40. The compound of claim 39, wherein the compound is described by formula (XI-1 ) :
Figure imgf000433_0002
(XI-1 )
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
41 . The compound of claim 40, wherein the compound is described by formula (XI-2):
Figure imgf000434_0001
(XI-2)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
The compound of claim 1 or 2, wherein the compound is described by formula (XII):
Figure imgf000434_0002
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2 and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
R6, R7, N2, and C2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising an N heteroatom and additional 0-2
heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S; R'6, R'7, N'2, and C'2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S;
or a pharmaceutically acceptable salt thereof.
43. The compound of claim 42, wherein the compound is described by formula (XI 1-1 ):
Figure imgf000435_0001
(XII-1 )
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
44. The compound of claim 1 or 2, wherein the compound described by formula (XIII):
E'1 E1
L !'i1 L Ί1
I I
M2— A2— L A1 M1
(XIII)
wherein A1 is a 1 -5 amino acid peptide covalently attached to the linking nitrogen in M1 ;
A2 is a 1 -5 amino acid peptide covalently attached to the linking nitrogen in M2;
E1 is a first monosaccharide or oligosaccharide moiety;
E'1 is a second monosaccharide or oligosaccharide moiety;
L, L1 , and L'1 are remainders of L';
or a pharmaceutically acceptable salt thereof.
45. The compound of claim 44, wherein the compound is described by formula (XIII-1 ):
Figure imgf000436_0001
(XIII-1 )
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R6 and R'6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each of R2 and R'2 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2- C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene;
or a pharmaceutically acceptable salt thereof.
46. The compound of claim 45, wherein the compound is described by formula (XI 11-2) :
Figure imgf000437_0001
(XIII-2)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
47. The compound of claim 46, wherein the compound is described by formula (XIII-3):
Figure imgf000437_0002
(XIII-3)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
or a pharmaceutically acceptable salt thereof.
48. The compound of claim 1 or 2, wherein the compound described by formula (XIV):
E
L
M2— A2— L A1 M1
(XIV)
wherein A1 is a 1 -5 amino acid peptide covalently attached to the linking nitrogen in M1 ;
A2 is a 1 -5 amino acid peptide covalently attached to the linking nitrogen in M2, or is absent; L and L1 are remainders of L';
or a pharmaceutically acceptable salt thereof.
49. The compound of any one of claims 21 , 27, 33-35, 38, 39, and 42, wherein R2 is optionally substituted C1 -C5 alkamino.
50. The compound of any one of claims 21 , 27, 33-35, 38, and 42, wherein R'2 is optionally substituted C1 -C5 alkamino.
51 . The compound of claim 49 or 50, wherein the optionally substituted C1 -C5 alkamino is CH2NH2 or
52. The compound of any one of claims 21 , 27, 33-35, 38, 39, and 42, wherein R2 is a polar moiety.
53. The compound of any one of claims 21 , 27, 33-35, 38, and 42, wherein R'2 is a polar moiety.
54. The compound of claim 52 or 53, wherein the polar moiety comprises a hydroxyl group, a carboxylic acid group, an ester group, or an amide group.
55. The compound of any one of claims 52-54, wherein the polar moiety is hydroxyl substituted C1 -C4 alkyl.
56. The compound of claim 55, wherein the polar moiety is CH(CH3)OH or CH2OH.
57. The compound of any one of claims 21 , 27, 33-35, 39, and 45, wherein R6 is a polar moiety.
58. The compound of any one of claims 21 , 27, 33, and 45, wherein R'6 is a polar moiety.
59. The compound of claim 57 or 58, wherein the polar moiety comprises a hydroxyl group, a carboxylic acid group, an ester group, or an amide group.
60. The compound of any one of claims 57-59, wherein the polar moiety is hydroxyl substituted C1 -C4 alkyl.
61 . The compound of claim 60, wherein the polar moiety is CH(CH3)OH or CH2OH.
62. The compound of any one of claims 27, 33, and 34, wherein R8 is optionally substituted C1 -C5 alkamino.
63. The compound of claim 27, wherein R'8 is optionally substituted C1 -C5 alkamino.
64. The compound of claim 62 or 63, wherein the optionally substituted C1 -C5 alkamino is CH2NH2 or
65. The compound of any one of claims 27, 33, and 34, wherein R8 is optionally substituted C5-C1 5 aryl.
66. The compound of claim 27, wherein R'8 is optionally substituted C5-C15 aryl.
67. The compound of claim 65 or 66, wherein the optionally substituted C5-C1 5 aryl is naphthyl.
68. The compound of claim 42, wherein R6, R7, N2, and C2 together form a 5- or 6-membered ring comprising C4-C5 heterocycloalkyl comprising an N heteroatom and additional 0 or 1 heteroatom independently selected from N, O, and S; and wherein R'6, R'7, N'2, and C'2 together form a 5- or 6- membered ring comprising C4-C5 heterocycloalkyl comprising an N heteroatom and additional 0 or 1 heteroatom independently selected from N, O, and S.
69. The compound of claim 45, wherein each of R2 and R'2 is C1 -C4 heteroalkylene.
70. The compound of claim 69, wherein each of R2 and R'2 is -(CH2)2NH- or -CH2NH-.
The compound of claim 3, wherein the compound is described by formula (XV):
Figure imgf000439_0001
(XV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl,
or a pharmaceutically acceptable salt thereof.
72. The compound of any one of claims 1 -71 , wherein L', L, L1 , or L'1 comprises one or more optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15 heteroarylene, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino, wherein R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl.
73. The compound of claims 72, wherein the backbone of L', L, L1 , or L'1 consists of one or more optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C1 5 arylene, optionally substituted C2-C15 heteroarylene, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; and wherein R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C1 5 heteroaryl.
74. The compound of 72 or 73, wherein L', L, L1 , or L'1 is oxo substituted.
75. The compound of any one of claims 1 -74, wherein the backbone of L', L, L1 , or L'1 comprises no more than 250 atoms.
76. The compound of any one of claims 1 -75, wherein L', L, L1 , or L'1 is capable of forming an amide, a carbamate, a sulfonyl, or a urea linkage.
77. The compound of any one of claims 2-71 , wherein L, L1 , or L'1 is a bond.
78. The compound of any one of claims 2-43 and 71 , wherein L is described by formula (L-l):
Lc
LB-Q-LA (L-l)
wherein LA is described by formula GA1-(ZA1)gi-(YA1)hi-(ZA2)ii-(YA2)ji -(ZA3)ki -(YA3)ii -(ZA4)mi -
Figure imgf000441_0001
LB is described by formula GB1-(ZB1 )g2-(YB1 )h2-(ZB2)i2-(YB2)j2-(ZB3)k2-(YB3)i2-(ZB4)m2-(YB4)n2-(ZB5)02-
GB2;
Lc is described by formula Gc1 -(Zc1 )g3-(Yc1 )h3-(ZC2)i3-(YC2)j3-(ZC3)k3-(YC3)i3-(ZC4)m3-(YC4)n3-(ZC5)03-
GC2;
GA1 is a bond attached to Q in formula (L-l);
GA2 is a bond attached to A1 or M1 if A1 is absent;
GB1 is a bond attached to Q in formula (L-l);
GB2 is a bond attached to A2 or M2 if A2 is absent;
GC1 is a bond attached to Q in formula (L-l);
GC2 is a bond attached to E;
each of ZA1 , ZA2, ZA3, ZM, ZA5, ZB1 , ZB2, ZB3, ZB4, ZB5, ZC1 , ZC2, ZC3, ZC4 and ZC5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C1 5 arylene, or optionally substituted C2-C15 heteroarylene;
each of YA1 , YA2, YA3, YA4, YB1 , YB2, YB3, YB4, YC1 , YC2, YC3, and YC4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;
R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , o1 , g2, h2, i2, j2, k2, I2, m2, n2, o2, g3, h3, i3, j3, k3, I3, m3, n3, and o3 is, independently, 0 or 1 ;
Q is a nitrogen atom, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20
heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20
heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene.
Figure imgf000442_0001
441
Figure imgf000443_0001
wherein R* is a bond or comprises one or more of optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15
heteroarylene, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, and imino, and
wherein R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl.
80. The compound of any one of claims 2-43 and 71 , wherein L is described by formula (L-lla) or formula (L-llb):
Figure imgf000444_0001
(L-lla) (L-llb)
wherein LA is described by formula GA1-(ZA1)gi-(YA1)hi-(ZA2)ii-(YA2)ji -(ZA3)ki -(YA3)ii -(ZA4)mi -
Figure imgf000444_0002
LB is described by formula GB1-(ZB1 )g2-(YB1 )h2-(ZB2)i2-(YB2)j2-(ZB3)k2-(YB3)i2-(ZB4)m2-(YB4)n2-(ZB5)o2-
GB2;
Lc is described by formula Gc1 -(Zc1 )g3-(Yc1 )h3-(ZC2)i3-(YC2)j3-(ZC3)k3-(YC3)i3-(ZC4)m3-
Figure imgf000444_0003
LD is described by formula GD1 -(ZD1 )g4-(YD1 )h4-(ZD2)i4-(YD2)j4-(ZD3)k4-(YD3)i4-(ZD4)m4-(YD4)n4-(ZD5)o4-
GD2;
GA1 is a bond attached to NA in formula (L-lla) or NA in formula (L-llb);
GA2 is a bond attached to A1 or M1 if A1 is absent;
GB1 is a bond attached to NA in formula (L-lla) or NA in formula (L-llb);
GB2 is a bond attached to A2 or M2 if A2 is absent;
GC1 is a bond attached to NB in formula (L-lla) or C in formula (L-llb);
GC2 is a bond attached to a first monosaccharide or oligosaccharide moiety, E1 ;
GD1 is a bond attached to NB in formula (L-lla) or C in formula (L-llb);
GD2 is a bond attached to a second monosaccharide or oligosaccharide moiety, E'1 ;
each of ZA1 ZA2 ZA3 ZA4 ZA5 ZB1 ZB2 ZB3 ZB4 ZB5 ZC1 ZC2 ZC3 ZC4 ZC5 ZD1 ZD2 ZD3 ZD4 and ZD5 is independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C1 5 arylene, or optionally substituted C2-C15 heteroarylene;
each of YA1 YA2 YA3 YA4 YB1 YB2 YB3 YB4 YC1 YC2 YC3 YC4 YD1 YD2 YD3 and YD4 is independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;
R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; and each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , o1 , g2, h2, i2, j2, k2, I2, m2, n2, o2, g3, h3, i3, j3, k3, I3, m3, n3, o3, g4, h4, i4, j4, k4, I4, m4, n4, and o4 is, independently, 0 or 1 . The compound of claim 80, wherein L is
Figure imgf000445_0001
82. The compound of any one of claims 44-47, wherein L is described by formula (L-lll):
HA1 -(WA1 )gi -(XA1 )hi -(WA2)ii -(XA2)ji -(WA3)ki -(XA3)ii -(WA4)mi -(XA4)m -(WA5)01 -HA2
(L-lll)
wherein HA1 is a bond attached to A2;
HA2 is a bond attached to A1 ;
each of WA1 , WA2, WA3, WM , and WA5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene;
each of XA1 , XA2, XA3, XA4 is, independently, O, S, NR\ P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;
R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; and each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , and o1 is, independently, 0 or 1 .
83. The compound of claim 82, where L is
Figure imgf000446_0001
84. The compound of any one of claims 44-47, 82, and 83, wherein L1 is described by formula (L-IV):
HB1-(WB1 )g2-(XB1 )h2-(WB2)i2-(XB2)j2-(WB3)k2-(XB3)l2-(WB4)m2-(XB4)n2-(WB5)o2-H
(L-IV)
wherein HB1 is a bond attached to A1 ;
HB2 is a bond attached to a first monosaccharide or oligosaccharide moiety, E1 ;
each of WB1 , WB2, WB3, WB4, and WB5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene;
each of XB1 , XB2, XB3, XB4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;
R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; and each of g2, h2, i2, j2, k2, I2, m2, n2, and o2 is, independently, 0 or 1 .
Figure imgf000446_0002
86. The compound of any one of claims 44-47 and 82-85, wherein L'1 is described by formula (L-V):
HC1 -(WC1 )g3-(XC1 )h3-(WC2)i3-(XC2)j3-(WC3)k3-(XC3)l3-(WC )m3-(XC4)n3-(WC5)o3HC2
(L-V)
wherein HC1 is a bond attached to A2;
HC2 is a bond attached to a second monosaccharide or oligosaccharide moiety, E'1 ;
each of WC1 , WC2, WC3, WC4, and WC5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-
C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene;
each of XC1 , XC2, XC3, XC4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;
R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; and each of g3, h3, i3, j3, k3, I3, m3, n3, and o3 is, independently, 0 or 1 .
Figure imgf000447_0001
90. The compound of any one of claims 1 -87 or 342-368, wherein E, E1 , or E'1 is any one of the moieties in Tables 2A and 2B.
91 . The compound of any one of claims 1 -87 or 342-368, wherein E, E1 , or E'1 directly or indirectly activates an immune cell.
92. The compound of any one of claims 1 -87 or 342-368, wherein E, E1 , or E'1 is a ligand to an innate immune receptor.
93. The compound of claim 92, wherein the innate immune receptor is AICL, BDCA2, CLEC2,
Complement receptor 3, Complement receptor 4, DCIR, dectin-1 , dectin-2, DC-SIGN, a C-Type lectin receptor, MMR, langerin, TLR2, Mincle, MBL, or KCR.
94. The compound of claims 1 -87 or 342-368, wherein E, E1 , or E'1 binds to an antibody.
95. The compound of claim 94, wherein the antibody is a natural antibody.
96. The compound of claim 95, wherein the natural antibody is an antibody of the immunoglobulin M (IgM) isotype.
97. The compound of any one of claims 94-96, wherein the antibody binds to a moiety in Tables 2A and 2B.
98. The compound of any one of claims 94-97, wherein the antibody is anti-aGal antibody or anti-aRha antibody.
99. The compound of any one of claims 1 -87 or 342-368, wherein the monosaccharide moiety has one optionally substituted C6-C9 monosaccharide residue.
100. The compound of any one of claims 1 -87 or 342-368, wherein the oligosaccharide moiety has 2-18 optionally substituted C6-C9 monosaccharide residues.
101 . The compound of claim 100, wherein the oligosaccharide moiety has 2-12 optionally substituted C6- C9 monosaccharide residues.
102. The compound of any one of claims 99-101 , wherein each of the optionally substituted C6-C9 monosaccharide residues is, independently, glucose (Glc), galactose (Gal), mannose (Man), allose (All), altrose (Alt), gulose (Gul), idose (Ido), talose (Tal), fucose (Fuc), rhamnose (Rha or L-Rha), thia- rhamnose (thia-Rha or thia-L-Rha), quinovose (Qui), 2-deoxyglucose (2-dGlc), glucosamine (GlcN), galactosamine (GaIN), mannosamine (ManN), fucosamine (FucN), quinovosamine (QuiN), N-Acetyl- glucosamine (GlcNAc), N-Acetyl-galactosamine (GalNAc), N-Acetyl-mannosamine (ManNAc), N-acetyl- fucosamine (FucNAc), N-acetyl-quinovosamine (QuiNAc), glucuronic acid (GlcA), galacturonic acid (GalA), mannuronic acid (ManA), iduronic acid (IdoA), sialic acid (Sia), neuraminic acid (Neu), N-Acetyl- neuraminic acid (Neu5Ac), N-Glycolyl-neuraminic acid (Neu5Gc), glucitol (Glc-ol), galactitol (Gal-ol), mannitol (Man-ol), fructose (Fru), sorbose (Sor), tagatose (Tag), thevetose (The), acofriose (Aco), digitoxose (Dig), cymarose (Cym), abequose (Abe), colitose (Col), tyvelose (Tyv), ascarylose (Asc), paratose (Par), or N-acetyl-muramic acid (MurNAc).
103. The compound of claim 102, wherein each of the optionally substituted C6-C9 monosaccharide residues is, independently, an optionally substituted C6 monosaccharide residue.
104. The compound of claim 103, wherein the optionally substituted C6 monosaccharide residue is Glc, Gal, Man, All, Alt, Gul, Ido, or Tal.
105. The compound of claim 103, wherein the optionally substituted C6 monosaccharide residue is Fuc, Rha or L-Rha, thia-Rha or thia-L-Rha, Qui, or 2-dGlc.
106. The compound of claim 103, wherein the optionally substituted C6 monosaccharide residue is GlcN, GaIN, ManN, FucN, or QuiN.
107. The compound of claim 103, wherein the optionally substituted C6 monosaccharide residue is N- GlcNAc, GalNAc, ManNAc, FucNAc, QuiNAc, GlcA, GalA, ManA, or IdoA.
108. The compound of claim 103, wherein the optionally substituted C6 monosaccharide residue is Glc- ol, Gal-ol, or Man-ol.
109. The compound of claim 103, wherein the optionally substituted C6 monosaccharide residue is Fru, Sor, Tag.
1 10. The compound of claim 103, wherein the optionally substituted C6 monosaccharide residue is The, Aco, Dig, Cym, Abe, Col, Tyv, Asc, Par, or MurNAc.
1 1 1 . The compound of claim 103, wherein the optionally substituted C6 monosaccharide residue is Rha, Gal, Glc, GlcA, GlcNAc, GalNAc, GlcN(Gc) (N-Glycolyl-Glucosamine), Neu5Ac, Neu5Gc, Fuc,
Man, -hbPOsMan (mannose phosphate), 6-H2PO3GIC (glucose phosphate), Mur (muramoyl), Mur-L-Ala-D- i-Gln-Lys, (Mur)-3-0-GlcNAc, sulfate-galactose (Su-Gal), disulfate-galactose (Su2-Gal), sulfate-glucose (Su-Glc), sulfate-GlcNAc (Su-GlcNAc), or sulfate-GalNAc (Su-GalNAc).
1 12. The compound of claim 103, wherein the optionally substituted C6 monosaccharide residue is optionally substituted Rha.
1 13. The compound of claim 1 12, wherein the optionally substituted C6 monosaccharide residue is Rha.
1 14. The compound of claim 1 12 or 1 13, wherein the optionally substituted C6 monosaccharide residue is L-Rha.
1 15. The compound of claim 103, wherein the optionally substituted C6 monosaccharide residue is optionally substituted Gal or optionally substituted Glc.
1 16. The compound of claim 103, wherein the optionally substituted Gal is optionally substituted a1 -3Gal.
1 17. The compound of claim 103, wherein the optionally substituted Glc is an optionally substituted β- glucan having 1 -6 Glc moieties.
1 18. The compound of claim 1 17, wherein the optionally substituted β-glucan is
Figure imgf000450_0001
pustulan.
1 19. The compound of claim 1 18, wherein the optionally substituted β-glucan is laminarin.
120. The compound of claim 102, wherein each of the optionally substituted C6-C9 monosaccharide residues is, independently, an optionally substituted C9 monosaccharide residue, wherein the optionally substituted C9 monosaccharide residue is Sia, Neu, Neu5Ac, or Neu5Gc.
121 . The compound of any one of claims 102-120, wherein at least one optionally substituted C6-C9 monosaccharide residue is substituted with 1 -3 substituents independently selected from sulfate, phosphate, methyl, acetyl, natural amino acids, and non-natural amino acids.
122. The compound of any one of claims 102-120, wherein at least one optionally substituted C6-C9 monosaccharide residue is substituted with 1 -3 substituents independently selected from sulfate, phosphate, methyl, and acetyl.
123. The compound of any one of claims 102-120, wherein the at least one optionally substituted C6-C9 monosaccharide moiety is substituted with 1 -3 substituents independently selected from natural and non- natural amino acids.
124. The compound of claim 123, wherein the natural amino acid is alanine, lysine, serine, glutamine or asparagine.
125. A compound of formula (XVI):
Figure imgf000451_0001
(XVI)
wherein:
each A is an independently selected amino acid;
L is a linker that, when m is 2, 3, 4, or 5, is bound to any of A;
each E is independently selected from a monosaccharide or an oligosaccharide;
m is 0, 1 , 2, 3, 4, or 5;
n is 1 , 2, 3, 4, or 5; and
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof.
126. The compound of claim 125, wherein Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof.
127. The compound of claim 125, wherein at least one of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
128. The compound of claim 125, wherein at least two of Q1 , Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
129. The compound of claim 125, wherein at least three of Q1 , Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
130. The compound of claim 125, wherein at least four of Q1 , Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
131 . The compound of claim 125, wherein at least five of Q1 , Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
132. The compound of claim 125, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
133. The compound of claim 125, wherein each Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof.
134. The compound of claim 125, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyl, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
135. The compound of claim 134, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
136. The compound of claim 135, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000452_0001
Figure imgf000453_0001
or a pharmaceutically acceptable salt thereof.
A compound of formula (XVII):
Figure imgf000454_0001
(XVII)
wherein:
each A1 and A2 is an independently selected amino acid;
L is a linker that, when m is 1 , 2, 3, 4, or 5, is bound to a nitrogen atom in any A1 and a nitrogen atom in any A2;
each E is independently selected from a monosaccharide or an oligosaccharide;
each m is independently 0, 1 , 2, 3, 4, or 5;
n is 1 , 2, 3, 4, or 5; and
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof.
138. The compound of claim 137, wherein Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof.
139. The compound of claim 137, wherein at least one of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
140. The compound of claim 137, wherein at least two of Q1 , Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
141 . The compound of claim 137, wherein at least three of Q1 , Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
142. The compound of claim 137, wherein at least four of Q1 , Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
143. The compound of claim 137, wherein at least five of Q1 , Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
144. The compound of claim 137, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.
145. The compound of claim 137, wherein each Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof.
146. The compound of claim 137, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
147. The compound of claim 137, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
148. The compound of claim 137, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000455_0001
Figure imgf000456_0001
or a pharmaceutically acceptable salt thereof.
149. The compound of any one of claims 137-148, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5- yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2- aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; and
each m is independently selected from 1 , 2, 3, 4, or 5;
or a pharmaceutically acceptable salt thereof.
150. The compound of any one of claims 137-149, wherein each m is independently 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
151 . The compound of any one of claims 137-150, wherein n is 1 , 2, 3, or 4, or a pharmaceutically acceptable salt thereof.
152. The compound of any one of claims 137-151 , wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000456_0002
Figure imgf000457_0001
or a pharmaceutically acceptable salt thereof.
153. The compound of claim 137, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
each m is independently 1 , 2, 3 or 4;
E is a monosaccharide;
n is 1 , 2, 3, or 4; and
the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000457_0003
or a pharmaceutically acceptable salt thereof.
A compound of formula (XVIII):
Figure imgf000457_0002
(XVIII)
wherein:
each A1 and A2 is an independently selected amino acid; X is absent or is -CH2CH2C(0)NH-;
each E is independently selected from a monosaccharide or an oligosaccharide;
each m is independently 0, 1 , 2, 3, 4, or 5;
n is 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 10; and
e is an integer from 0 to 10;
or a pharmaceutically acceptable salt thereof.
155. The compound of claim 154, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
156. The compound of claim 155, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
157. The compound of claim 154, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000458_0001
Figure imgf000459_0001
or a pharmaceutically acceptable salt thereof.
158. The compound of claim 157, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000459_0002
or a pharmaceutically acceptable salt thereof.
159. The compound of any one of claims 154-158, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2 or 3;
d is an integer from 1 to 10;
e is an integer from 1 to 10; and E is a monosaccharide;
or a pharmaceutically acceptable salt thereof.
160. The compound of claim 154, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine; m is 2 or 3;
d is an integer from 1 to 10;
e is an integer from 1 to 10;
E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
161 . The compound of claim 160, wherein m is 2, or a pharmaceutically acceptable salt thereof. 162. The compound of claim 160, wherein m is 3, or a pharmaceutically acceptable salt thereof. 163. The compound of claim 160, wherein:
d is 10;
e is 10; and
X is absent;
or a pharmaceutically acceptable salt thereof.
164. The compound of claim 160, wherein:
m is 2;
d is 1 ;
e is 1 ; and
X is -CH2CH2C(0)NH-;
or a pharmaceutically acceptable salt thereof.
165. The compound of any one of claims 154-164, wherein
E is
Figure imgf000460_0001
or a pharmaceutically acceptable salt thereof.
Figure imgf000461_0001
166. The compound of claim 165, wherein E is OH , or a pharmaceutically acceptable thereof.
A compound of formula (XIX):
Figure imgf000461_0002
(XIX)
wherein:
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
each m is independently 0, 1 , 2, 3, 4, or 5;
n is 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 10;
e is an integer from 0 to 10;
f is an integer from 0 to 10; and
g is an integer from 0 to 10;
or a pharmaceutically acceptable salt thereof.
168. The compound of claim 167, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
169. The compound of claim 168, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
170. The compound of claim 167, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000462_0001
or a p armaceutca y accepta e sat t ereo .
171. The compound of claim 170, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000463_0001
or a pharmaceutically acceptable salt thereof.
172. The compound of any one of claims 167-171 , wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2;
d is 1 ;
e is 1 ;
f is 3;
g is 1 ; and
E is a monosaccharide;
or a pharmaceutically acceptable salt thereof.
173. The compound of claim 167, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine;
m is 2;
d is 1 ;
e is 1 ;
f is 3;
g is 1 ;
E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
1 74. The compound of any one of claims 1 67-1 73, wherein
Figure imgf000464_0001
or a pharmaceutically acceptable salt thereof.
1 75. The compound of claim 1 74, wherein E is
Figure imgf000464_0002
, or a pharmaceutically acceptable salt thereof.
A compound of formula (XX) :
Figure imgf000464_0003
(XX)
wherein :
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 1 0;
e is an integer from 0 to 1 0;
f is an integer from 0 to 1 0; and
g is an integer from 0 to 1 0;
or a pharmaceutically acceptable salt thereof.
1 77. The compound of claim 1 76, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
178. The compound of claim 177, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
179. The compound of claim 178, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000465_0001
Figure imgf000466_0001
180. The compound of claim 179, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000466_0002
or a pharmaceutically acceptable salt thereof.
181 . The compound of any one of claims 176-180, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2;
d is 1 ;
e is 1 ;
f is 1 ;
g is 1 ; and
E is a monosaccharide;
or a pharmaceutically acceptable salt thereof.
182. The compound of claim 176, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine;
m is 2;
d is 1 ;
e is 1 ;
f is 1 ;
g is 1 ;
E is a monosaccharide; and
n is 1 ; or a pharmaceutically acceptable salt thereof.
183. The compound of any one of claims 176-182, wherein
Figure imgf000467_0001
or a pharmaceutically acceptable salt thereof.
Figure imgf000467_0002
184. e compoun o c a m 183, w ere n s , or a p armaceut ca y accepta e thereof.
185. A compound of formula (XXI):
Figure imgf000467_0003
(XXI)
wherein:
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 10;
e is an integer from 0 to 10;
f is an integer from 0 to 10;
g is an integer from 0 to 25;
or a pharmaceutically acceptable salt thereof.
186. The compound of claim 185, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
187. The compound of claim 186, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
188. The compound of claim 187, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000468_0001
Figure imgf000469_0001
189. The compound of claim 188, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000469_0002
or a pharmaceutically acceptable salt thereof.
190. The compound of any one of claims 185-189, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2;
d is 1 ;
e is 1 ;
f is 1 ;
g is 8 to 25; and
E is a monosaccharide;
or a pharmaceutically acceptable salt thereof.
191 . The compound of claim 185, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine;
m is 2;
d is 1 ;
e is 1 ;
f is 1 ;
g is 8 to 25; E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
192. The compound of any one of claims 185-191 , wherein
Figure imgf000470_0001
or a pharmaceutically acceptable salt thereof.
193. The compound of claim 192, wherein E is
Figure imgf000470_0002
, or a pharmaceutically acceptable salt thereof.
A compound of formula (XXII):
Figure imgf000470_0003
(XXII)
wherein:
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and
d is an integer from 0 to 15;
or a pharmaceutically acceptable salt thereof.
195. The compound of claim 194, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
196. The compound of claim 195, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
197. The compound of claim 196, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000471_0001
Figure imgf000472_0001
198. The compound of claim 197, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000472_0002
or a pharmaceutically acceptable salt thereof.
199. The compound of any one of claims 194-198, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2;
d is 10;
E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
200. The compound of claim 194, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid and piperazine-2- carboxylic acid;
m is 2;
d is 10;
E is a monosaccharide; and n is 1 ;
or a pharmaceutically acceptable salt thereof.
201 . The compound of any one of claims 194-200, wherein
Figure imgf000473_0001
or a pharmaceutically acceptable salt thereof.
202. The compound of claim 201 , wherein E is
Figure imgf000473_0002
, or a pharmaceutically acceptable salt thereof.
A compound of formula (XXIII):
Figure imgf000473_0003
(XXIII)
wherein:
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
Y is
Figure imgf000473_0004
, -C(0)CH2CH2-, -CH2-, or is absent;
X is -C(0)CH2CH2- or is absent;
each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 15; e is an integer from 1 to 10;
f is an integer from 1 to 5; and
g is an integer from 1 to 5;
or a pharmaceutically acceptable salt thereof.
204. The compound of claim 203, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
205. The compound of claim 204, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
206. The compound of claim 205, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000474_0001
Figure imgf000475_0001
or a pharmaceutically acceptable salt thereof.
207. The compound of claim 206, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000475_0003
or a pharmaceutically acceptable salt thereof.
208. The compound of any one of claims 203-207, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2, 3 or 4;
d is 1 ;
Figure imgf000475_0002
E is a monosaccharide;
f is 1 or 2; g is 1 or 2; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
209. The compound of claim 203, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 3-aminoalanine, 2- piperazinecarboxylic acid, 2-aminohexanoic acid, 2-aminooctanoic acid, methionine, and threonine; m is 2, 3 or 4;
Figure imgf000476_0001
d is 1 ;
E is a monosaccharide;
f is 1 or 2;
g is 1 or 2; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
210. The compound of any one of claims 203-207, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 3 or 4;
d is 1 ;
Y is absent;
E is a monosaccharide;
f is 1 ;
g is 1 ; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
21 1 . The compound of claim 210, wherein
each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine; and m is 3;
or a pharmaceutically acceptable salt thereof.
212. The compound of any one of claims 203-207, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2, 3 or 4;
d is 1 ;
Y is -C(0)CH2CH2-;
E is a monosaccharide;
f is 1 or 2;
g is 1 or 2; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
213. The compound of claim 212, wherein each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 2,4-diaminobutyric acid, 3-aminoalanine, 2- aminohexanoic acid, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine,
cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine, or a pharmaceutically acceptable salt thereof.
214. The compound of claim 213, wherein m is 4, d is 1 , f is 1 , and g is 1 , or a pharmaceutically acceptable salt thereof.
215. The compound of claim 213, wherein m is 3, f is 2, and g is 1 , or a pharmaceutically acceptable salt thereof.
216. The compound of any one of claims 203-207, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2, 3 or 4;
d is 1 ; E is a monosaccharide;
f is 1 or 2;
g is 1 or 2; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
217. The compound of claim 216, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid and threonine;
f is 1 ; and
g is 1 ;
or a pharmaceutically acceptable salt thereof.
218. The compound of claim 216, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminohexanoic acid, 2- aminooctanoic acid and threonine, or a pharmaceutically acceptable salt thereof;
m is 4;
f is 1 ; and
g is 1 ;
or a pharmaceutically acceptable salt thereof.
219. The compound of claim 217, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminohexanoic acid, 2- aminooctanoic acid and threonine, or a pharmaceutically acceptable salt thereof;
m is 3;
f is 1 ; and
g is 1 ;
or a pharmaceutically acceptable salt thereof.
220. The compound of any one of claims 203-219, wherein
E is
Figure imgf000478_0001
or a pharmaceutically acceptable salt thereof.
Figure imgf000479_0001
221 . The compound of claim 220, wherein E is OH , or a pharmaceutically acceptable thereof.
222. A compound of formula (XXIV):
Figure imgf000479_0002
(XXIV)
wherein:
each A1 and A2 is an independently selected amino acid;
X is -C(0)CH2CH2CH2-Y- or -C(0)CH2CH2C(0)NH-;
Y is heteroaryl ;
each E is independently selected from a monosaccharide or an oligosaccharide;
each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and
d is an integer from 0 to 15;
or a pharmaceutically acceptable salt thereof.
223. The compound of claim 222, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
224. The compound of claim 223, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl. hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
225. The compound of claim 224, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000480_0001
226. The compound of claim 225, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000481_0001
or a pharmaceutically acceptable salt thereof.
227. The compound of any one of claims 222-226, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2 or 3;
d is an integer from 1 to 3;
E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
228. The compound of claim 227, wherein each A1 and A2 is independently selected from 2,4- diaminobutyric acid, 3-(2-naphthyl)alanine, and threonine, or a pharmaceutically acceptable salt thereof.
229. The compound of claim 228, wherein:
X is -C(0)CH2CH2CH2-Y-;
m is 3;
d is 3; and
Y is 1 ,4-triazololyl;
or a pharmaceutically acceptable salt thereof.
230. The compound of claim 228, wherein:
X is -C(0)CH2CH2C(0)-;
m is 2; and
d is 1 ;
or a pharmaceutically acceptable salt thereof.
31 . The compound of any one of claims 222-230, wherein
Figure imgf000482_0001
or a pharmaceutically acceptable salt thereof.
232. The compound of claim 228, wherein E is
Figure imgf000482_0002
, or a pharmaceutically acceptable salt thereof.
A compound of formula (XXV):
Figure imgf000482_0003
(XXV)
wherein:
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 15; and
e is an integer from 0 to 15;
or a pharmaceutically acceptable salt thereof.
234. The compound of claim 233, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
235. The compound of claim 234, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
236. The compound of claim 235, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000483_0001
Figure imgf000484_0001
237. The compound of claim 236, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000484_0002
or a pharmaceutically acceptable salt thereof.
238. The compound of any one of claims 233-237, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2 or 3;
d is an integer from 1 to 3;
e is an integer from 1 to 3;
E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
239. The compound of claim 233, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine;
m is 2 or 3;
d is an integer from 1 to 3;
e is an integer from 1 to 3;
E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
240. The compound of any one of claims 233-239, wherein
E is
Figure imgf000485_0001
;
or a pharmaceutically acceptable salt thereof.
241 . The compound of claim 240, wherein E is
Figure imgf000485_0002
, or a pharmaceutically acceptable salt thereof.
A compound of formula (XXVI):
Figure imgf000485_0003
(XXVI)
wherein:
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
R is C1 -C20 alkyl;
each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 20;
e is an integer from 0 to 20; and
f is an integer from 0 to 20;
or a pharmaceutically acceptable salt thereof.
243. The compound of claim 242, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
244. The compound of claim 243, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
245. The compound of claim 244, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000486_0001
Figure imgf000487_0001
246. The compound of claim 245, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000487_0002
or a pharmaceutically acceptable salt thereof.
247. The compound of any one of claims 242-246, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2 or 3;
d is an integer from 1 to 3;
e is an integer from 1 to 3;
f is an integer from 1 to 3;
E is a monosaccharide;
R is C1 -C10 alkyl; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
248. The compound of claim 242, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine;
m is 2;
d is 1 ;
e is 1 ;
f is 1 ; E is a monosaccharide;
R is C1 -C10 alkyl; and
n is 1 ;
or a pharmaceutically acceptable salt thereof. 249. The compound of any one of claims 242-248, wherein
E is
Figure imgf000488_0001
or a pharmaceutically acceptable salt thereof.
250. The compound of claim 249, wherein E is
Figure imgf000488_0002
, or a pharmaceutically acceptable salt thereof.
251 . A compound of formula (XXVII):
Figure imgf000488_0003
wherein:
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
R is C1 -C20 alkyl;
each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 1 to 20;
e is an integer from 1 to 20; and
f is an integer from 1 to 20;
or a pharmaceutically acceptable salt thereof.
252. The compound of claim 251 , wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
253. The compound of claim 252, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl. hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
254. The compound of claim 253, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000489_0001
Figure imgf000490_0001
or a pharmaceutically acceptable salt thereof.
255. The compound of claim 251 , wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000490_0002
or a pharmaceutically acceptable salt thereof.
256. The compound of any one of claims 251 -255, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2, 3, or 4;
d is an integer from 1 to 3;
e is an integer from 1 to 3;
f is an integer from 1 to 3;
E is a monosaccharide;
R is C1 -C10 alkyl; and
n is 1 ; or a pharmaceutically acceptable salt thereof.
257. The compound of claim 251 , wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine;
m is 2, 3, or 4;
d is 1 ;
e is 1 ;
f is 1 ;
E is a monosaccharide;
R is C1 -C10 alkyl; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
258. The compound of claim 257, wherein m is 2, or a pharmaceutically acceptable salt thereof.
259. The compound of claim 257, wherein m is 3, or a pharmaceutically acceptable salt thereof.
260. The compound of claim 257, wherein m is 4, or a pharmaceutically acceptable salt thereof.
261 . The compound of any one of claims 251 -260, wherein
E is
Figure imgf000491_0001
or a pharmaceutically acceptable salt thereof.
262. The compound of claim 261 , wherein E is
Figure imgf000491_0002
, or a pharmaceutically acceptable salt thereof.
263. A compound of formula (XXVIII):
Figure imgf000492_0001
(XXVIII)
wherein:
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
Y is
Figure imgf000492_0002
, -C(0)CH2CH2-, -CH2-, or is absent;
e is an integer from 1 to 10;
each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and
d is an integer from 0 to 15;
or a pharmaceutically acceptable salt thereof.
264. The compound of claim 263, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
265. The compound of claim 264, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
266. The compound of claim 265, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000492_0003
Figure imgf000493_0001
or a pharmaceutically acceptable salt thereof.
267. The compound of claim 266, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Q1 Q2 Q3 Q4 Q5 Q6
* 3 X Jb X Jb
Figure imgf000494_0001
or a pharmaceutically acceptable salt thereof.
268. The compound of any one of claims 263-267, wherein Y is -C(0)CH2CH2-, or a pharmaceutically acceptable salt thereof.
269. The compound of any one of claims 263-268, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2, 3, or 4;
d is an integer from 1 to 3;
E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
270. The compound of claim 269, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and
d is 1 ;
or a pharmaceutically acceptable salt thereof.
271 . The compound of claim 270, wherein m is 2, or a pharmaceutically acceptable salt thereof.
272. The compound of claim 270, wherein m is 3, or a pharmaceutically acceptable salt thereof.
273. The compound of claim 270, wherein m is 4, or a pharmaceutically acceptable salt thereof.
274. The compound of any one of claims 263-273, wherein
Figure imgf000495_0001
or a pharmaceutically acceptable salt thereof.
Figure imgf000495_0002
275. The compound of claim 273, wherein E is OH , or a pharmaceutically acceptable thereof.
A compound of formula (XXIX):
Figure imgf000495_0003
(XXIX)
wherein:
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
Figure imgf000495_0004
each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and
d is an integer from 0 to 15;
or a pharmaceutically acceptable salt thereof.
277. The compound of claim 276, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
278. The compound of claim 277, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl. hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
279. The compound of claim 278, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000496_0001
Figure imgf000497_0001
280. The compound of claim 279, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000497_0002
or a pharmaceutically acceptable salt thereof.
281 . The compound of any one of claims 276-280, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2, 3, or 4;
d is an integer from 1 to 3;
E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
282. The compound of claim 281 , wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and
d is 1 ;
or a pharmaceutically acceptable salt thereof.
283. The compound of claim 282, wherein m is 2, or a pharmaceutically acceptable salt thereof.
284. The compound of claim 282, wherein m is 3, or a pharmaceutically acceptable salt thereof.
285. The compound of claim 282, wherein m is 4, or a pharmaceutically acceptable salt thereof.
286. The compound of any one of claims 276-285, wherein
E is
Figure imgf000498_0001
;
or a pharmaceutically acceptable salt thereof.
287. The compound of claim 286, wherein E is
Figure imgf000498_0002
, or a pharmaceutically acceptable salt thereof.
A compound of formula (XXX):
Figure imgf000498_0003
(XXX)
wherein:
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
Y is
Figure imgf000498_0004
-C(0)CH2CH2-, -CH2-, or is absent;
each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 0 to 15;
g is an integer from 0 to 15; and
e and e' are independently an integer from 1 to 3;
or a pharmaceutically acceptable salt thereof.
289. The compound of claim 288, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
290. The compound of claim 289, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
291 . The compound of claim 290, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000499_0001
Figure imgf000500_0001
292. The compound of claim 291 , wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000500_0002
or a pharmaceutically acceptable salt thereof.
293. The compound of any one of claims 288-292, wherein Y is -C(0)CH2CH2-, or a pharmaceutically acceptable salt thereof.
294. The compound of any one of claims 288-293, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2, 3, or 4;
d is an integer from 1 to 3;
E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
295. The compound of claim 294, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and d is 1 ; and
e is 1 or 2;
or a pharmaceutically acceptable salt thereof.
296. The compound of claim 295, wherein m is 2, or a pharmaceutically acceptable salt thereof.
297. The compound of claim 295 wherein m is 3, or a pharmaceutically acceptable salt thereof.
298. The compound of claim 295, wherein m is 4, or a pharmaceutically acceptable salt thereof.
299. The compound of any one of claims 288-298, wherein
Figure imgf000501_0001
or a pharmaceutically acceptable salt thereof.
300. The compound of claim 299, wherein E is
Figure imgf000501_0002
, or a pharmaceutically acceptable salt thereof.
301 . A compound of formula (XXXI):
Figure imgf000501_0003
(XXXI)
wherein: each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
each Y is independently selected from
Figure imgf000502_0001
, -C(0)CH2CH2-, and -CH2-, or is absent; each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 1 to 15;
each e is independently from 1 to 15;
each f is independently from 1 to 15; and
g is from 1 to 15;
or a pharmaceutically acceptable salt thereof.
302. The compound of claim 301 , wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
303. The compound of claim 302, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
304. The compound of claim 303, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000502_0002
Figure imgf000503_0001
or a pharmaceutically acceptable salt thereof.
305. The compound of any one of claims 301 -304, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000503_0002
or a pharmaceutically acceptable salt thereof.
306. The compound of any one of claims 301 -305, wherein Y is -C(0)CH2CH2-, or a pharmaceutically acceptable salt thereof.
307. The compound of any one of claims 301 -306, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2, 3, or 4;
d is an integer from 1 to 5;
e is an integer from 1 to 5;
E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
308. The compound of claim 307, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and
d is 4 or 5;
e is 4 or 5; and
f is 1 ;
or a pharmaceutically acceptable salt thereof.
309. The compound of claim 308, wherein m is 2, or a pharmaceutically acceptable salt thereof.
310. The compound of claim 308 wherein m is 3, or a pharmaceutically acceptable salt thereof.
31 1 . The compound of claim 308, wherein m is 4, or a pharmaceutically acceptable salt thereof.
312. The compound of any one of claims 301 -31 1 , wherein
Figure imgf000504_0001
or a pharmaceutically acceptable salt thereof.
Figure imgf000504_0002
313. The compound of claim 312, wherein E is OH , or a pharmaceutically acceptable thereof. A compound of formula (XXXII)
Figure imgf000505_0001
(XXXII)
wherein:
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
each Y is independently selected from
Figure imgf000505_0002
, -C(0)CH2CH2-, -
CH2CH2NHC(0)CH2CH2-, and -CH2-, or is absent;
m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5;
Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 1 to 15;
each e is independently from 1 to 15; and
g is from 1 to 15;
or a pharmaceutically acceptable salt thereof.
315. The compound of claim 314, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
316. The compound of claim 315, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
317. The compound of claim 316, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000506_0001
or a p armaceutca y accepta e sat t ereo .
318. The compound of claim 317, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000507_0001
or a pharmaceutically acceptable salt thereof.
319. The compound of any one of claims 314-318, wherein Y is -CH2CH2NHC(0)CH2CH2-, or a pharmaceutically acceptable salt thereof.
320. The compound of any one of claims 314-319, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2, 3, or 4;
d is an integer from 1 to 5;
e is an integer from 1 to 5;
E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
321 . The compound of claim 320, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and
d is an integer from 1 to 4; and
e is 1 ;
or a pharmaceutically acceptable salt thereof.
322. The compound of claim 321 , wherein d is 2, or a pharmaceutically acceptable salt thereof.
323. The compound of claim 321 , wherein m is 2, or a pharmaceutically acceptable salt thereof.
324. The compound of claim 321 wherein m is 3, or a pharmaceutically acceptable salt thereof.
325. The compound of claim 321 , wherein m is 4, or a pharmaceutically acceptable salt thereof.
326. The compound of any one of claims 314-325, wherein
Figure imgf000508_0001
or a pharmaceutically acceptable salt thereof.
327. The compound of claim 326, wherein E is
Figure imgf000508_0002
, or a pharmaceutically acceptable salt thereof.
A compound of formula (XXXIII):
Figure imgf000508_0003
(XXXIII)
wherein:
each A1 and A2 is an independently selected amino acid;
each E is independently selected from a monosaccharide or an oligosaccharide;
each X is independently selected from
Figure imgf000508_0004
, -C(0)CH2CH2C(0)-, -CH2CH2NHC(0)CH2CH2-, -C(O)-, and -CH2-, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 alkenyl, optionally substituted C1 -C20 alkynyl, optionally substituted C1 - C15 aryl, optionally substituted C1 -C15 heteroaryl, or is absent;
each Y is independently selected from -CH-, or oxygen ;
Z is -NH-, -NHC(O)-, oxygen, or sulfur;
each m is independently 0, 1 , 2, 3, 4, or 5;
each n is independently 1 , 2, 3, 4, or 5; Q1 , Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; d is an integer from 1 to 15;
g is an integer from 1 to 15;
e and e' are independently from 1 to 10 and
or a pharmaceutically acceptable salt thereof.
329. The com
Figure imgf000509_0001
(XXXIII-1 )
or a pharmaceutically acceptable salt thereof. 330. The com
Figure imgf000509_0002
(XXXIII-2)
or a pharmaceutically acceptable salt thereof.
331 . The compound of any one of claims 328-330, wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from C1 -C4 alkyl, C1 -C2 hydroxyalkyi, C1 -C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.
332. The compound of claim 331 , wherein each of Q1 , Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1 -propyl, 2-propyl, 1 -hydroxyethyl, butyl, benzyl, cyclohexylmethyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.
333. The compound of claim 332, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000510_0001
334. The compound of claim 333, wherein the combination of Q1 , Q2, Q3, Q4, Q5 and Q6 is selected from one of
Figure imgf000511_0001
or a pharmaceutically acceptable salt thereof.
335. The compound of any one of claims 328-334, wherein:
each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;
m is 2, 3, or 4;
d is an integer from 1 to 5;
e is 1 ;
X is -C(0)CH2CH2C(0)-;
E is a monosaccharide; and
n is 1 ;
or a pharmaceutically acceptable salt thereof.
336. The compound of claim 335, wherein:
each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminohexanoic acid, and threonine; and
d is 1 ;
or a pharmaceutically acceptable salt thereof.
337. The compound of claim 336, wherein m is 2, or a pharmaceutically acceptable salt thereof.
338. The compound of claim 336 wherein m is 3, or a pharmaceutically acceptable salt thereof.
339. The compound of claim 336, wherein m is 4, or a pharmaceutically acceptable salt thereof.
340. The compound of any one of claims 328-339, wherein
Figure imgf000512_0001
or a pharmaceutically acceptable salt thereof.
^'" 'OH
341 . The compound of claim 340, wherein E is OH , or a pharmaceutically acceptable salt thereof.
342. The compound of claim 3, wherein the compound is described by formula (XXXIV):
Figure imgf000512_0002
(XXXIV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2, R3, R4, R5, R6, R7, R8, R9, R10, R'2, R'3, R'4, R'5, R'6, R'7, R'8, R'9, and R'10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or two R groups on the same or adjacent atoms join to form an optionally substituted 5-8 membered ring;
each of a', b', c', a, b, and c is, independently, 0 or 1 ;
each of N1 , N2, N3, N4, N'1 , N'2, N'3, and N'4 is a nitrogen atom;
each of C1 , C2, C3, C'1 , C'2, and C'3 is a carbon atom ;
L is a linker comprising at least one optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C3-C15 heteroarylene;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
343. The compound of claim 342, wherein the compound comprises at least one optionally substituted 5- 8 membered ring formed by joining (i) R2, R3, and C1 ; (ii) R3, R4, N1 , and C1 ; (iii) R5, R6, and C2; (iv) R6, R7, N2, and C2; (v) R8, R9, and C3; (vi) R9, R10, N3, and C3; (vii) R'2, R'3, and C'1 ; (viii) R'3, R'4, N'1 , and C'1 ; (ix) R'5, R'6, and C'2; (x) R'6, R'7, N'2, and C'2; (xi) R'8, R'9, and C'3; or (xii) R'9, R'10, N'3, and C'3.
344. The compound of claim 342 or 343, wherein L is described by formula (L-VI):
Lc
LB-Q-LA
(L-VI)
wherein LA is described by formula GA1-(ZA1)gi-(YA1)hi-(ZA2)ii-(YA2)ji-(ZA3)ki-(YA3)ii-(ZA4)mi-
Figure imgf000513_0001
LB is described by formula GB1-(ZB1 )g2-(YB1 )h2-(ZB2)i2-(YB2)j2-(ZB3)k2-(YB3)i2-(ZB4)m2-(YB4)n2-(ZB5)02-
GB2;
Lc is described by formula Gc1-(Zc 1 )g3-(Yc1 )h3-(ZC2)i3-(YC2)j3-(ZC3)k3-(YC3)i3-(ZC4)m3-(YC4)n3-(ZC5)03-
GC2;
GA1 is a bond attached to Q in formula (L-VI);
GA2 is a bond attached to A1 or M1 if A1 is absent;
GB1 is a bond attached to Q in formula (L-VI);
GB2 is a bond attached to A2 or M2 if A2 is absent;
GC1 is a bond attached to Q in formula (L-VI);
GC2 is a bond attached to E;
each of ZA1 , ZA2, ZA3, ZA4, ZA5, ZB1 , ZB2, ZB3, ZB4, ZB5, ZC1 , ZC2, ZC3, ZC4 and ZC5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C1 5 arylene, or optionally substituted C2-C15 heteroarylene;
each of YA1 , YA2, YA3, YA4, YB1 , YB2, YB3, YB4, YC1 , YC2, YC3, and YC4 is, independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;
R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ; each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , o1 , g2, h2, i2, j2, k2, I2, m2, n2, o2, g3, h3, i3, j3, k3, I3, m3, n3, and o3 is, independently, 0 or 1 ;
Q comprises at least one optionally substituted C3-C20 cycloalkylene, optionally substituted C3- C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C3-C15 heteroarylene.
345. The compound of claim 342 or 343, wherein L is described by formula (L-VII):
Figure imgf000514_0001
(L-VII)
wherein LA is described by formula GA1-(ZA1)gi-(YA1)hi-(ZA2)ii-(YA2)ji -(ZA3)ki -(YA3)ii -(ZA4)mi -
Figure imgf000514_0002
LB is described by formula GB1-(ZB1 )g2-(YB1 )h2-(ZB2)i2-(YB2)j2-(ZB3)k2-(YB3)i2-(ZB4)m2-(YB4)n2-(ZB5)02-
GB2;
Lc is described by formula Gc1 -(Zc1 )g3-(Yc1 )h3-(ZC2)i3-(YC2)j3-(ZC3)k3-(YC3)i3-(ZC4)m3-(YC4)n3-(ZC5)03-
GC2;
LD is described by formula GD1 -(ZD1 )g4-(YD1 )h4-(ZD2)i4-(YD2)j4-(ZD3)k4-(YD3)i4-(ZD4)m4-(YD4)n4-(ZD5)o4-
GD2;
GA1 is a bond attached to Q in formula (L-VII);
GA2 is a bond attached to A1 or M1 if A1 is absent;
GB1 is a bond attached to Q in formula (L-VII);
GB2 is a bond attached to A2 or M2 if A2 is absent;
GC1 is a bond attached to Q in formula (L-VII);
GC2 is a bond attached to a first monosaccharide or oligosaccharide moiety, E1 ;
GD1 is a bond attached to N in formula (L-VII);
GD2 is a bond attached to a second monosaccharide or oligosaccharide moiety, E2;
each of ZA1 ZA2 ZA3 ZM ZA5 ZB1 ZB2 ZB3 ZB4 ZB5 ZC1 ZC2 ZC3 ZC4 ZC5 ZD1 ZD2 ZD3 ZD4 and ZD5 is, independently, optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20
heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20
heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20
heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene;
each of YA1 YA2 YA3 YA4 YB1 YB2 YB3 YB4 YC1 YC2 YC3 YC4 YD1 YD2 YD3 and YD4 is independently, O, S, NR', P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;
R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl ;
each of g1 , hi , i1 , j1 , k1 , 11 , ml , n1 , o1 , g2, h2, i2, j2, k2, I2, m2, n2, o2, g3, h3, i3, j3, k3, I3, m3, n3, o3, g4, h4, i4, j4, k4, I4, m4, n4, and o4 is, independently, 0 or 1 ;
Q comprises at least one optionally substituted C3-C20 cycloalkylene, optionally substituted C3- C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C3-C15 heteroarylene.
346. The compound of any one of claims 342-345, wherein the compound is described by formula (XXXV):
Figure imgf000515_0001
(XXXV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2, R6, R8, R'2, R'6, and R'8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
347. The compound of any one of claims 342-345, wherein the compound is described by formula (XXXV):
Figure imgf000516_0001
(XXXV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2, R6, R8, R'2, and R'6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyi, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
348. The compound of any one of claims 342-345, wherein the compound is described by formula (XXXVI):
Figure imgf000516_0002
(XXXVI) wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2, R6, R8, and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
349. The compound of any one of claims 342-345, wherein the compound is described by formula (XXXVII):
Figure imgf000517_0001
(XXXVII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2, R6, and R8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
350. The compound of any one of claims 342-345, wherein the compound is described by formula (XXXVIII):
Figure imgf000518_0001
(XXXVIII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2, R6, R'2, and R'6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
351 . The compound of any one of claims 342-345, wherein the compound is described by formula (XXXIX):
Figure imgf000518_0002
(XXXIX)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2, R6, and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
352. The compound of any one of claims 342-345, wherein the compound is described by formula (XXXX):
Figure imgf000519_0001
(XXXX)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2 and R6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
353. The compound of any one of claims 342-345, wherein the compound is described by formula (XXXXI):
Figure imgf000520_0001
(XXXXI)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each of R2 and R'2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
354. The compound of any one of claims 342-345, wherein the compound is described by formula (XXXXI I):
Figure imgf000520_0002
(XXXXI I)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
R2 is a positively charged moiety, a polar moiety, optionally substituted C1 -C5 alkamino, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyi, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyi, optionally substituted C3-C20 heterocycloalkyi, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C3-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
355. The compound of any one of claims 342-345, wherein the compound is described by formula (XXXXIII):
Figure imgf000521_0001
(XXXXIII)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl;
each E is, independently, a monosaccharide or oligosaccharide moiety;
n is 1 , 2, 3, or 4,
or a pharmaceutically acceptable salt thereof.
356. The compound of claim any one of claims 342-355, wherein L comprises at least one an optionally substituted C3 cycloalkylene or an optionally substituted C3 heterocycloalkylene.
357. The compound of claim 356, wherein the compound is described by formula (XXXXIV):
Figure imgf000521_0002
(XXXXIV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl; and L comprises an optionally substituted C3 cycloalkylene or an optionally substituted C3 heterocycloalkylene,
or a pharmaceutically acceptable salt thereof.
358. The compound of claim 356, wherein the compound is described by formula (XXXXV):
Figure imgf000522_0001
(XXXXV)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl; and
L comprises an optionally substituted C3 cycloalkylene or an optionally substituted C3 heterocycloalkylene,
or a pharmaceutically acceptable salt thereof.
359. The compound of any one of claims 356-358, wherein L is
Figure imgf000522_0002
wherein each q is, independently, an integer from 1 to 1 1 , inclusive.
360. The compound of claim 342-355, wherein L comprises at least one an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene. The compound of claim 360, wherein the compound is described by formula (XXXXVI):
Figure imgf000523_0001
(XXXXVI)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl; and
L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene,
or a pharmaceutically acceptable salt thereof.
362. The compound of claim 360, wherein the compound is described by formula (XXXXVM):
Figure imgf000523_0002
(XXXXVM)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl; and
L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene,
or a pharmaceutically acceptable salt thereof.
363. The compound of claim 360, wherein the compound is described by formula (XXXXVMI):
Figure imgf000524_0001
(XXXXVMI)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl; and
L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene,
or a pharmaceutically acceptable salt thereof.
364. The compound of claim 360, wherein the compound is described by formula (XXXXIX):
Figure imgf000524_0002
(XXXXIX)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl; and
L comprises an optionally substituted C5 cycloalkylene or an optionally substituted C5 heterocycloalkylene,
or a pharmaceutically acceptable salt thereof.
65. The compound of any one of claims 360-364, wherein L is
Figure imgf000525_0001
wherein each q is, independently, an integer from 1 to 1 1 , inclusive.
366. The compound of any one of claims 342-355, wherein L comprises at least one an optionally substituted C6 cycloalkylene, at least one an optionally substituted C6 heterocycloalkylene, or at least one an optionally substituted C6 arylene.
367. The compound of claim 366, wherein the compound is described by formula (XXXXX):
Figure imgf000525_0002
(XXXXX)
wherein each of R'1 and R1 is, independently, optionally substituted benzyl, optionally substituted C2-C15 heteroaryl, optionally substituted C1 -C8 alkyl, or cyclohexylmethyl; and
L comprises an optionally substituted C6 arylene,
or a pharmaceutically acceptable salt thereof.
368. The compound of claim 366 or 367, wherein L is
Figure imgf000525_0003
wherein each q is, independently, an integer from 1 to 1 1 , inclusive.
369. The compound of any one of claims 1 -368, wherein a concentration of the compound, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to 10,000 nM.
370. The compound of claim 369, wherein the concentration of the compound, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to equal to 1 ,000 nM.
371 . The compound of claim 370, wherein the concentration of the compound, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to equal to 100 nM.
372. A pharmaceutical composition comprising a compound of any of claims 1 -368, or a
pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
373. The pharmaceutical composition of claim 372, further comprising an antibacterial agent.
374. The pharmaceutical composition of claim 373, wherein the antibacterial agent is selected from the group consisting of linezolid, tedizolid, posizolid, radezolid, retapamulin, valnemulin, tiamulin, azamulin, lefamulin, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem, doripenem, imipenem/cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefepime, ceftaroline fosamil, ceftobiprole, teicoplanin, vancomycin, telavancin, dalbavancin, oritavancin, clindamycin, lincomycin, daptomycin, solithromycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, spiramycin, aztreonam, furazolidone, nitrofurantoin, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin g, penicillin v, piperacillin, penicillin g, temocillin, ticarcillin, amoxicillin clavulanate,
ampicillin/sulbactam, piperacillin/tazobactam, ticarcillin/clavulanate, bacitracin, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethizole, sulfamethoxazole, sulfanamide, sulfasalazine, sulfisoxazole, trimethoprim-sulfamethoxazole (tmp-smx), sulfonamidochrysoidine, eravacycline, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol(bs), ethionamide, isoniazid, pyrazinamide, rifampicin, rifabutin, rifapentine, streptomycin, arsphenamine, chloramphenicol, fosfomycin, fusidic acid, metronidazole, mupirocin, platensimycin, quinupristin/dalfopristin, thiamphenicol, tigecycline, tinidazole, and
trimethoprim, prodrugs thereof, and pharmaceutically acceptable salts thereof.
375. The pharmaceutical composition of claim 374, wherein a prodrug of tedizolid is tedizolid phosphate.
376. The pharmaceutical composition of claim 374, wherein the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, levofloxacin, rifampicin, linezolid, erythromycin, or solithromycin.
377. The pharmaceutical composition of claim 376, wherein the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, or levofloxacin.
378. A method of protecting against or treating a bacterial infection in a subject, the method comprising administering to the subject a compound of any one of claims 1 -368.
379. The method of claim 378, further comprising administering to the subject an antibacterial agent.
380. A method of protecting against or treating a bacterial infection in a subject, the method comprising administering to the subject (1 ) a compound of any one of claims 1 -368 and (2) an antibacterial agent.
381 . The method of any one of claims 378-380, wherein the bacterial infection is caused by Gram- negative bacteria.
382. The method of any one of claims 378-381 , wherein the bacterial infection is caused by a resistant strain of bacteria.
383. The method of claim 382, wherein the resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene, and/or a chromosomal mutation conferring polymyxin resistance.
384. The method of claim 383, wherein the resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, or the mcr-3 gene.
385. The method of claim 383, wherein the resistant strain of bacteria possesses a chromosomal mutation conferring polymyxin resistance.
386. The method of any one of claims 382-385, wherein the resistant strain of bacteria is a resistant strain of E. coli.
387. A method of protecting against or treating sepsis in a subject, comprising administering to the subject a compound of any one of claims 1 -368.
388. A method of preventing lipopolysaccharides (LPS) in Gram-negative bacteria from activating an immune system in a subject, comprsing administering to the subject a compound of any one of claims 1 - 368.
389. The method of claim 388, wherein the method prevents LPS from activating a macrophage.
390. The method of claim 388 or 389, wherein the method prevents LPS-induced nitric oxide (NO) production from a macrophage.
391 . The method of any one of claims 388-390, wherein the Gram-negative bacteria is a resistant strain of Gram-negative bacteria.
392. The method of claim 391 , wherein the resistant strain of Gram-negative bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene, and/or a chromosomal mutation conferring polymyxin resistance.
393. The method of claim 392, wherein the resistant strain of Gram-negative bacteria possesses the mcr- 1 gene, the mcr-2 gene, or the mcr-3 gene.
394. The method of claim 392, wherein the resistant strain of Gram-negative bacteria possesses a chromosomal mutation conferring polymyxin resistance.
395. The method of any one of claims 391 -394, wherein the resistant strain of Gram-negative bacteria is a resistant strain of E. coli.
396. The method of any one of claims 387-395, further comprising administering to the subject an antibacterial agent.
397. The method of claim 379, 380, and 396, wherein the compound and the antibacterial agent are administered substantially simultaneously.
398. The method of claim 379, 380, and 396, wherein the compound and the antibacterial agent are administered separately.
399. The method of claim 398, wherein the compound is administered first, followed by administering of the antibacterial agent alone.
400. The method of claim 398, wherein the antibacterial agent is administered first, followed by administering of the compound alone.
401 . The method of claim 379, 380, and 396, wherein the compound and the antibacterial agent are administered substantially simultaneously, followed by administering of the compound or the antibacterial agent alone.
402. The method of claim 379, 380, and 396, wherein the compound or the antibacterial agent is administered alone first, followed by administering of the compound and the antibacterial agent substantially simultaneously.
403. The method of any one of claims 379, 380, and 396-402, wherein administering the compound and the antibacterial agent together lowers the MIC of each of the compound and the antibacterial agent relative to the MIC of each of the compound and the antibacterial agent when each is used alone.
404. The method of any one of claims 378-403, wherein the compound and/or the antibacterial agent is administered intramuscularly, intravenously, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleural^, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctival, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, orally, locally, by inhalation, by injection, or by infusion.
405. A method of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria, comprising contacting the bacteria or a site susceptible to bacterial growth with a compound of any of claims 1 -368.
406. The method of claim 405, further comprising contacting the bacteria or the site susceptible to bacterial growth with an antibacterial agent.
407. A method of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria, comprising contacting the bacteria or a site susceptible to bacterial growth with (1 ) a compound of any of claims 1 -368 and (2) an antibacterial agent.
408. The method of any one of claims 379, 380, 396, 397, and 398, wherein the antibacterial agent is selected from the group consisting of linezolid, tedizolid, posizolid, radezolid, retapamulin, valnemulin, tiamulin, azamulin, lefamulin, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem, doripenem, imipenem/cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefepime, ceftaroline fosamil, ceftobiprole, teicoplanin, vancomycin, telavancin, dalbavancin, oritavancin, clindamycin, lincomycin, daptomycin, solithromycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, spiramycin, aztreonam, furazolidone, nitrofurantoin, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin g, penicillin v, piperacillin, penicillin g, temocillin, ticarcillin, amoxicillin clavulanate, ampicillin/sulbactam, piperacillin/tazobactam, ticarcillin/clavulanate, bacitracin, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethizole, sulfamethoxazole, sulfanamide, sulfasalazine, sulfisoxazole, trimethoprim-sulfamethoxazole (tmp-smx),
sulfonamidochrysoidine, eravacycline, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol(bs), ethionamide, isoniazid, pyrazinamide, rifampicin, rifabutin, rifapentine, streptomycin, arsphenamine, chloramphenicol, fosfomycin, fusidic acid, metronidazole, mupirocin, platensimycin, quinupristin/dalfopristin, thiamphenicol, tigecycline, tinidazole, and trimethoprim, prodrugs thereof, and pharmaceutically acceptable salts thereof.
409. The method of claim 408, wherein a prodrug of tedizolid is tedizolid phosphate.
410. The method of claim 408, wherein the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, levofloxacin, rifampicin, linezolid, erythromycin, or solithromycin.
41 1 . The method of claim 410, wherein the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, or levofloxacin.
412. The method of any one of claims 405-41 1 , wherein the bacteria are Gram-negative bacteria.
413. The method of any one of claims 405-412, wherein the bacteria are a resistant strain of bacteria.
414. The method of claim 413, wherein the resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, the mcr-3 gene, and/or a chromosomal mutation conferring polymyxin resistance.
415. The method of claim 414, wherein the resistant strain of bacteria possesses the mcr- 1 gene, the mcr-2 gene, or the mcr-3 gene.
416. The method of claim 414, wherein the resistant strain of bacteria possesses a chromosomal mutation conferring polymyxin resistance.
417. The method of any one of claims 413-416, wherein the resistant strain of bacteria is a resistant strain of E. coli.
418. A compound selected from any one of compounds 1 -123, or a pharmaceutically acceptable salt thereof.
419. A compound selected from any one of claims 18-47, 71 -124, or 342-368, wherein each of R'1 and R1 is, independently, benzyl or CH2CH(CH3)2.
PCT/US2017/040467 2016-07-01 2017-06-30 Compositions and methods for the treatment of bacterial infections WO2018006063A1 (en)

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
US201662357825P 2016-07-01 2016-07-01
US62/357,825 2016-07-01
US201662397113P 2016-09-20 2016-09-20
US62/397,113 2016-09-20
US201662427618P 2016-11-29 2016-11-29
US62/427,618 2016-11-29
US201662434296P 2016-12-14 2016-12-14
US62/434,296 2016-12-14
US201762444035P 2017-01-09 2017-01-09
US62/444,035 2017-01-09
US201762482518P 2017-04-06 2017-04-06
US62/482,518 2017-04-06

Publications (1)

Publication Number Publication Date
WO2018006063A1 true WO2018006063A1 (en) 2018-01-04

Family

ID=60787621

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/040467 WO2018006063A1 (en) 2016-07-01 2017-06-30 Compositions and methods for the treatment of bacterial infections

Country Status (1)

Country Link
WO (1) WO2018006063A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108330102A (en) * 2018-03-14 2018-07-27 江南大学 One plant of Tiamulin monoclonal antibody hybridoma cell strain and its application
WO2018203087A1 (en) * 2017-05-04 2018-11-08 Centauri Therapeutics Limited Compounds and therapeutic uses thereof
WO2020014501A1 (en) * 2018-07-11 2020-01-16 Cidara Therapeutics, Inc. Compositions and methods for the treatment of bacterial infections
WO2020074909A1 (en) 2018-10-10 2020-04-16 Centauri Therapeutics Limited Compounds and therapeutic uses thereof
WO2020252396A1 (en) * 2019-06-13 2020-12-17 Cidara Therapeutics, Inc. Compositions and methods for the treatment of respiratory syncytial virus
WO2020252393A1 (en) * 2019-06-13 2020-12-17 Cidara Therapeutics, Inc. Compositions and methods for the treatment of human immunodeficiency virus
CN113332238A (en) * 2021-06-09 2021-09-03 四川恒通动保生物科技有限公司 Preparation method of compound solution with gamithromycin
CN114539048A (en) * 2022-02-18 2022-05-27 新发药业有限公司 Caronic anhydride intermediate and preparation method of caronic anhydride
WO2022232245A3 (en) * 2021-04-27 2022-12-15 The Rockefeller University Macolacins and methods of use thereof
US11833213B2 (en) 2018-09-06 2023-12-05 Cidara Therapeutics, Inc. Compositions and methods for the treatment of viral infections

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000042067A1 (en) * 1999-01-12 2000-07-20 Princeton University Saccharides linked to compounds that bind cell-surface peptides or proteins
US6380356B1 (en) * 1999-12-07 2002-04-30 Advanced Medicine, Inc. Multivalent polymyxin antibiotics
US20100028334A1 (en) * 2006-12-15 2010-02-04 Trustees Of Boston University Compositions and methods to potentiate colistin activity
WO2016083531A1 (en) * 2014-11-26 2016-06-02 New Pharma Licence Holdings Limited Compounds

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000042067A1 (en) * 1999-01-12 2000-07-20 Princeton University Saccharides linked to compounds that bind cell-surface peptides or proteins
US6380356B1 (en) * 1999-12-07 2002-04-30 Advanced Medicine, Inc. Multivalent polymyxin antibiotics
US20100028334A1 (en) * 2006-12-15 2010-02-04 Trustees Of Boston University Compositions and methods to potentiate colistin activity
WO2016083531A1 (en) * 2014-11-26 2016-06-02 New Pharma Licence Holdings Limited Compounds

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MORADI ET AL.: "Glycosylation, an effective synthetic strategy to improve the bioavailability of therapeutic peptides", CHEMICAL SCIENCE, vol. 7, no. 4, 29 January 2016 (2016-01-29), pages 2492 - 2500, XP055306836 *
YOSHIDA ET AL.: "New insight into the mode of action of vancomycin dimers in bacterial cell wall synthesis", MEDCHEMCOMM, vol. 2, no. 4, 11 February 2011 (2011-02-11), pages 278 - 282, XP055447633 *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018203087A1 (en) * 2017-05-04 2018-11-08 Centauri Therapeutics Limited Compounds and therapeutic uses thereof
US11406683B2 (en) 2017-05-04 2022-08-09 Centauri Therapeutics Limited (GB/GB) Compounds and therapeutics uses thereof
CN108330102A (en) * 2018-03-14 2018-07-27 江南大学 One plant of Tiamulin monoclonal antibody hybridoma cell strain and its application
WO2020014501A1 (en) * 2018-07-11 2020-01-16 Cidara Therapeutics, Inc. Compositions and methods for the treatment of bacterial infections
US11833213B2 (en) 2018-09-06 2023-12-05 Cidara Therapeutics, Inc. Compositions and methods for the treatment of viral infections
WO2020074909A1 (en) 2018-10-10 2020-04-16 Centauri Therapeutics Limited Compounds and therapeutic uses thereof
WO2020252396A1 (en) * 2019-06-13 2020-12-17 Cidara Therapeutics, Inc. Compositions and methods for the treatment of respiratory syncytial virus
WO2020252393A1 (en) * 2019-06-13 2020-12-17 Cidara Therapeutics, Inc. Compositions and methods for the treatment of human immunodeficiency virus
CN114390929A (en) * 2019-06-13 2022-04-22 奇达拉治疗公司 Compositions and methods for treating respiratory syncytial virus
CN114401731A (en) * 2019-06-13 2022-04-26 奇达拉治疗公司 Compositions and methods for treating human immunodeficiency virus
WO2022232245A3 (en) * 2021-04-27 2022-12-15 The Rockefeller University Macolacins and methods of use thereof
CN113332238A (en) * 2021-06-09 2021-09-03 四川恒通动保生物科技有限公司 Preparation method of compound solution with gamithromycin
CN114539048B (en) * 2022-02-18 2023-09-08 新发药业有限公司 Carlong anhydride intermediate and preparation method of Carlong anhydride
CN114539048A (en) * 2022-02-18 2022-05-27 新发药业有限公司 Caronic anhydride intermediate and preparation method of caronic anhydride

Similar Documents

Publication Publication Date Title
WO2018006063A1 (en) Compositions and methods for the treatment of bacterial infections
AU2016302940B2 (en) Prodrugs of glutamine analogs
US20230190950A1 (en) Compositions and methods for the treatment of bacterial infections
WO2017218922A2 (en) Compositions and methods for the treatment of bacterial infections
US11046730B2 (en) Antimicrobial compositions
US20220348580A1 (en) Wd40 repeat domain protein 5 (wdr5) degradation / disruption compounds and methods of use
WO2018187574A1 (en) Compositions and methods for the treatment of fungal infections
DK2643342T3 (en) Novel angiotensin type-2 (AT2) receptor agonists and their use
CN113194983A (en) Compositions and methods for treating viral infections
JP6471236B2 (en) Polymyxin derivatives and uses thereof
EP2178552A1 (en) The use of type-b lantibiotic-based compounds having antimicrobial activity
TW201625311A (en) Compositions and methods for the treatment of fungal infections
CN111447941A (en) Polypeptide conjugates for intracellular delivery of binding peptides
US20120238512A1 (en) Antithrombotic dual inhibitors comprising a biotin residue
WO2020014501A1 (en) Compositions and methods for the treatment of bacterial infections
RU2680138C2 (en) Tricyclic gyrase inhibitors
WO2019148194A2 (en) Peptidyl inhibitors of calcineurin-nfat interaction
US20090264345A1 (en) Macrocyclic peptides and methods for making and using them
WO2019126353A2 (en) Compositions and methods for the treatment of bacterial infections
WO2019126341A2 (en) Compositions and methods for the treatment of bacterial infections
US10851099B2 (en) Dipeptide piperidine derivatives
US20160206680A1 (en) Anti-trypanosome compounds and treatments
EP2379589A2 (en) Compounds, compositions and use
WO2020014469A1 (en) Compositions and methods for the treatment of bacterial infectons
JP2023522104A (en) Guanidine modified C-terminal vancomycin compounds, compositions and methods

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17821421

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17821421

Country of ref document: EP

Kind code of ref document: A1