CN115427049A - Inhibitors of malaria and plasmodium falciparum hexose transporters and uses thereof - Google Patents

Inhibitors of malaria and plasmodium falciparum hexose transporters and uses thereof Download PDF

Info

Publication number
CN115427049A
CN115427049A CN202180024568.5A CN202180024568A CN115427049A CN 115427049 A CN115427049 A CN 115427049A CN 202180024568 A CN202180024568 A CN 202180024568A CN 115427049 A CN115427049 A CN 115427049A
Authority
CN
China
Prior art keywords
oxy
pyran
tetrahydro
hydroxymethyl
compound
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN202180024568.5A
Other languages
Chinese (zh)
Inventor
颜宁
施一公
尹航
蒋鑫
袁亚飞
黄健
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tsinghua University
Original Assignee
Tsinghua University
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 Tsinghua University filed Critical Tsinghua University
Publication of CN115427049A publication Critical patent/CN115427049A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/18Acyclic radicals, substituted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/26Acyclic or carbocyclic radicals, substituted by hetero rings
    • 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

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biochemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present application provides molecules capable of binding to the binding pocket of Plasmodium falciparum (PfHT) hexose transporter (PfHT) or an analog thereof, and complexes comprising the same. Also provided herein are PfHT inhibitors, pharmaceutical compositions comprising the inhibitors, and methods of using the inhibitors or the pharmaceutical compositions to treat diseases associated with Plasmodium (Plasmodium) or PfHT or to kill or inhibit the growth of Plasmodium (Plasmodium). The present application also provides a set of structural coordinates for such binding pockets and methods of using the set of structural coordinates to screen for and design compounds capable of binding to PfHT or an analog thereof.

Description

Inhibitors of malaria and plasmodium falciparum hexose transporters and uses thereof
Technical Field
The present disclosure relates to molecules capable of binding to the binding pocket of Plasmodium falciparum (PfHT) hexose transporter (PfHT) or an analog thereof, and complexes comprising the same. The present disclosure also relates to inhibitors of PfHT or analogs thereof, pharmaceutical compositions comprising the inhibitors, and methods of using the inhibitors or pharmaceutical compositions to treat diseases associated with Plasmodium (Plasmodium) or PfHT, such as malaria, or to kill or inhibit the growth of Plasmodium.
Background
Malaria is an insect-borne infectious disease caused by plasmodium parasites. Glucose is the main source of energy for parasites in the blood stage. The malaria glucose transporter, plasmodium falciparum hexose transporter (PfHT), has proven to be critical for parasite survival and has been chemically validated as an anti-malarial drug target.
As a glucose analog, compound 3361 (i.e., 3-O- (undec-10-en) -1-yl-D-glucose) has been identified as a potent inhibitor of PfHT, which is more selective for PfHT than the human ortholog glucose transporter 1 (GLUT 1) (cfSee Thiierry
Figure BDA0003863898460000012
Et al, PNAS,2003, volume 100, phase 13, pages 7476-7479). However, compound 3361 itself is not considered to be a drug-like property and is therefore not an effective candidate for lead development.
Accordingly, there is a need to develop novel therapeutic agents with improved potency and selectivity that target PfHT or its analogs.
Disclosure of Invention
The present disclosure addresses this need by providing a crystal structure of a complex of PfHT and compound 3361. By interpreting the crystal structure, key structural features of PfHT, particularly its binding pocket and the structure, shape and portion of the molecule to which it binds, can be determined.
Thus, in one aspect, the present disclosure provides a molecule capable of binding to both the R1 binding pocket and the R2 binding pocket of a PfHT polypeptide or analog thereof, the polypeptide having the amino acid sequence of SEQ ID NO:1, the analog having at least 70% sequence identity to SEQ ID NO:1, wherein the R1 binding pocket comprises at least one or more amino acid residues selected from the group consisting of equivalent residues in Q169, Q305, Q306, N311, N341, W412, and N435 of SEQ ID NO:1 or analog thereof, and the R2 binding pocket comprises at least one or more amino acid residues selected from the group consisting of equivalent residues in V44, L47, F85, W436, and V443 of SEQ ID NO:1 or analog thereof;
wherein the molecule is not compound 3361 having the formula:
Figure BDA0003863898460000011
in some embodiments, the R1 binding pocket further comprises one or more additional amino acid residues selected from equivalent residues in F40, I172, I176, I310, F403, and a404 of SEQ ID NO:1, or analogs thereof.
In some embodiments, the R1 binding pocket further comprises one or more additional amino acid residues selected from the group consisting of T145, T173, V314, and I400 of SEQ ID No. 1, or equivalent residues in analogs thereof.
In some embodiments, the R2 binding pocket further comprises one or more additional amino acid residues selected from the group consisting of equivalent residues in L81, V312, a439, and F444 of SEQ ID No. 1, or analogs thereof.
In some embodiments, the molecule is capable of further binding to the R3 binding pocket of a PfHT polypeptide or analog thereof, wherein the R3 binding pocket comprises at least one or more amino acid residues selected from the group consisting of equivalent residues in N48, K51, N52, N311, N316, N318, E319, and D447, or analogs thereof, of SEQ ID No. 1.
In some embodiments, the R3 binding pocket further comprises one or more additional amino acid residues selected from equivalent residues in F85, V312, S315, V443, and F444 of SEQ ID No. 1, or analogs thereof.
In some embodiments, the molecule is capable of binding to a PfHT polypeptide or analog thereof with a Kd value of no more than 20 μ Μ (as determined by microcalorimetry).
In some embodiments, the PfHT analog is selected from the group consisting of Plasmodium vivax (Plasmodium vivax) hexose transporter (PvHT), plasmodium ovale (Plasmodium ovale) hexose transporter (PoHT), plasmodium malariae (Plasmodium malariae) hexose transporter (PmHT), and Plasmodium knowlesi (Plasmodium knowlesi) hexose transporter (PkHT).
In some embodiments, the PfHT analog has an amino acid sequence selected from the group consisting of SEQ ID NO 2, SEQ ID NO 3, SEQ ID NO 4, and SEQ ID NO 5.
In another aspect, the present disclosure provides a complex comprising a PfHT polypeptide having the amino acid sequence of SEQ ID NO:1 or an analog thereof having at least 70% sequence identity to SEQ ID NO:1, bound to a molecule of the present disclosure. In some embodiments, the complex is crystalline.
In another aspect, the present disclosure provides a set of X-ray crystal structure coordinates for at least one allosteric binding pocket of a PfHT polypeptide or analog thereof, the polypeptide having the amino acid sequence of SEQ ID NO:1, the analog having at least 70% sequence identity to SEQ ID NO:1, wherein the allosteric binding pocket is inducible when compound 3361 is complexed with the PfHT polypeptide or analog thereof.
In some embodiments, the at least one allosteric binding pocket comprises:
a) R1 binding pocket comprising at least one or more amino acid residues selected from the group consisting of equivalent residues in F40, Q169, I172, I176, Q305, Q306, I310, N311, N341, F403, A404, W412 and N435 of SEQ ID NO:1 or analogues thereof,
b) An R2 binding pocket comprising at least one or more amino acid residues selected from the group consisting of equivalent residues in V44, L47, L81, F85, V312, W436, A439, V443, and F444 of SEQ ID NO:1 or analogs thereof;
c) An R3 binding pocket comprising at least one or more amino acid residues selected from the group consisting of equivalent residues in N48, K51, N52, F85, N311, V312, S315, N316, N318, E319, V443, F444, and D447 of SEQ ID NO 1 or analogs thereof; or
d) Any combination thereof.
In some embodiments, the R1 binding pocket further comprises one or more additional amino acid residues selected from equivalent residues in T145, T173, V314, and I400 of SEQ ID No. 1, or analogs thereof.
In some embodiments, the set of X-ray crystal structure coordinates is shown in fig. 2.
In another aspect, the present disclosure provides a computer-readable storage medium having stored thereon the set of X-ray crystal structure coordinates provided herein.
In another aspect, the present disclosure provides a method of assessing or predicting the binding characteristics of a compound to a PfHT polypeptide having the amino acid sequence of SEQ ID NO:1 or an analog thereof having at least 70% sequence identity to SEQ ID NO:1, the method comprising the steps of:
a) Generating, on a computer, a representation of the three-dimensional structure of the at least one allosteric binding pocket based on the set of X-ray crystal structure coordinates provided herein,
b) Generating a representation of the compound on a computer, an
c) Fitting the representation of the compound according to step b) with the representation of the three-dimensional structure of the at least one allosteric binding pocket according to step a) to determine the probability that the compound binds to the at least one allosteric binding pocket.
In another aspect, the present disclosure provides a method of identifying a compound as a potential PfHT inhibitor, the method comprising the steps of:
a) Generating on a computer a representation of the three-dimensional structure of the at least one allosteric binding pocket based on the set of X-ray crystal structure coordinates provided herein,
b) A representation of the compound is generated on a computer,
c) Adapting the representation of the compound according to step b) with a computer representation of the three-dimensional structure of the at least one allosteric binding pocket according to step a) to provide an energy minimized configuration of the compound in the at least one allosteric binding pocket; and
d) Evaluating the results of step c) to quantify the binding between the compound and the at least one allosteric binding pocket,
wherein the compound is identified as a potential PfHT inhibitor when the compound binds to the at least one allosteric binding pocket to produce a stable complex of low energy.
In another aspect, the present disclosure provides a virtual screening method for identifying potential PfHT inhibitors, the method comprising the steps of:
a) Generating or accessing on a computer a representation of the three-dimensional structure of the at least one allosteric binding pocket based on the set of X-ray crystal structure coordinates provided herein,
b) Generating or accessing on a computer a representation of a candidate compound from a library of compounds,
c) Adapting the representation of the candidate compound according to step b) with the representation of the three-dimensional structure of the at least one allosteric binding pocket according to step a) to provide an energy minimized configuration of the candidate compound in the at least one allosteric binding pocket; and
d) Evaluating the results of step c) to quantify the binding between the candidate compound and the at least one allosteric binding pocket,
e) The quantitative binding is compared to a predetermined threshold,
wherein the candidate compound is identified as a potential PfHT inhibitor based on the comparison of step e).
In another aspect, the present disclosure provides a method of designing a compound capable of binding to a PfHT polypeptide having an amino acid sequence of SEQ ID NO:1 or an analog thereof having at least 70% sequence identity to SEQ ID NO:1, the method comprising:
a) Generating, on a computer, a representation of the three-dimensional structure of the at least one allosteric binding pocket based on the set of X-ray crystal structure coordinates provided herein,
b) Generating a representation of the candidate compound on a computer,
c) Adapting the representation of the candidate compound according to step b) with the representation of the three-dimensional structure of the at least one allosteric binding pocket according to step a) to provide an energy minimized configuration of the candidate compound in the at least one allosteric binding pocket; and
d) Optionally, modifying the candidate compound based on the results obtained in step c);
e) Optionally, repeating steps b) to c) with the modified candidate compound obtained in step d),
wherein the compound is identified as a potential PfHT inhibitor when the compound binds to the at least one allosteric binding pocket to produce a low energy, stable complex.
In another aspect, the present disclosure provides a compound having formula (I):
A-B-L-D-E (I)
or a pharmaceutically acceptable salt thereof, wherein
A is a hexose moiety linked to B through its atom selected from carbon, nitrogen, oxygen or sulfur;
b is absent, or is selected from-CH 2 C(O)O-、-CH 2 -C (O) NH-and-C (O) -;
l is- (CH) 2 ) m -、-(CH 2 OCH 2 ) q Or- (CH) 2 ) n -W-(CH 2 ) p -, wherein-W-is selected from cyclopropyl, -O-) -S-, -NH-, -C = C-),-C (O) O-and-C (O) NH-, m is an integer from 1 to 12, and n, p and q are each an integer from 1 to 3;
d is absent, or is selected from-O-, -S-, and-NH-;
E is selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein the cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more R groups;
r is selected from halogen, oxo, alkyl, haloalkyl, -OR 1 and-NR 2 R 3
R 1 Selected from the group consisting of hydrogen, alkyl, aryl, alkylaryl, and alkylalkoxy;
R 2 and R 3 Each selected from the group consisting of hydrogen, alkyl, cycloalkyl and aryl, wherein the alkyl, cycloalkyl and aryl are optionally substituted with one or more alkoxy groups.
In some embodiments, the compounds provided herein have the following formula (II)
Figure BDA0003863898460000031
In some embodiments, the compounds provided herein have a formula selected from:
Figure BDA0003863898460000032
Figure BDA0003863898460000041
in another aspect, the present disclosure provides a pharmaceutical composition comprising one or more molecules or compounds provided herein and a pharmaceutically acceptable excipient.
In another aspect, the present disclosure provides a method of treating a malarial parasite or PfHT associated disease in a subject, the method comprising administering to a subject in need thereof a therapeutically effective amount of one or more molecules, one or more compounds or pharmaceutical compositions provided herein.
In another aspect, the present disclosure provides a method of killing or inhibiting the growth of plasmodium by administering an effective amount of one or more molecules, one or more compounds, or a pharmaceutical composition provided herein.
Drawings
FIG. 1 shows an alignment of the sequences of SEQ ID NO 1 to 5.
FIG. 2 shows the atomic structure coordinates of PfHT polypeptide having SEQ ID NO:1 complexed with Compound 3361, as obtained by X-ray diffraction of the crystalline complex. "DRG" in the fourth column refers to Compound 3361.
FIG. 3 shows statistics for data collection and structure refinement of the complex of PfHT polypeptide having SEQ ID NO:1 with compound 3361.
FIG. 4 shows a cross-sectional view of (a) a semi-transparent electrostatic potential of a complex of a PfHT polypeptide having SEQ ID NO:1 and compound 3361; (b) Coordination of the D-glucose moiety in compound 3361 to a representative amino acid residue in the R1-binding pocket of the PfHT polypeptide; (c) An aliphatic moiety in compound 3361, surrounded by representative amino acid residues in the R2 binding pocket of the PfHT polypeptide; and (d) a representative amino acid residue in the induced new R3 binding pocket.
Fig. 5 shows D-glucose uptake mediated by PfHT and GLUT1 in the presence of exemplary compounds 16, 30 and 31 at a concentration of 100 μ M.
FIG. 6 shows the inhibition of glycolytic activity by exemplary compound 29 as observed by the Seahorse extracellular flux analyzer.
Fig. 7 shows that exemplary compounds 29 and 76 inhibit glycolytic activity in a dose-dependent manner on both early (ring stage) and late (trophozoite/schizont stage) and late parasite release from RBCs in RBCs.
FIG. 8 shows a negative correlation of glucose concentration in assay medium with the potency of exemplary Compound 29 on glycolytic activity.
FIG. 9 shows the EC of exemplary Compound 29 at different blood subphases of the parasite 50
FIG. 10 shows a representative schematic of a sub-stage assay. Closely synchronized parasites (Pf 3D 7) were exposed to exemplary compound 29 or DHA for different time durations at the sub-stages shown.
Figure 11 shows representative images of compound-treated parasites. Solid line profile: parasites treated with compound 29; dotted line outline: DHA treated parasites.
Detailed Description
Reference will now be made in detail to certain embodiments of the present disclosure, examples of which are illustrated in the accompanying structures and formulas. While the disclosure will be described in conjunction with the enumerated embodiments, it will be understood that they are not intended to limit the disclosure to those embodiments. On the contrary, the present disclosure is intended to cover all alternatives, modifications, and equivalents, which may be included within the scope of the present disclosure as defined by the appended claims. Those of skill in the art will recognize many methods and materials similar or equivalent to those described herein, which can be used in the practice of the present disclosure. The present disclosure is in no way limited to the methods and materials described. In the event that one or more of the incorporated references and similar materials differ from or contradict this application, including but not limited to defined terms, use of terms, described techniques, etc., the present disclosure controls. All references, patents, and patent applications cited in this disclosure are hereby incorporated by reference in their entirety.
It is appreciated that certain features of the disclosure, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the disclosure which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination. It should be noted that, as used in the specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a compound" includes a plurality of compounds.
Definition of
As used herein, the term "amino acid" broadly refers to any compound and/or substance that can be incorporated into a polypeptide chain, for example, by forming one or more peptide bonds. In some embodiments, the amino acid has the general structure H 2 N-C (H) (R) -COOH. In some embodiments, the amino acid is a naturally occurring amino acid. In some embodiments, the amino acid is a synthetic amino acid; in some embodiments, the amino acid is a D-amino acid; in some embodiments, the amino acid is an L-amino acid; in some embodiments, wherein H 2 R in N-C (H) (R) -COOH is not equal to hydrogen, and the stereocenter of amino acid is in (R) -configuration; in some embodiments, wherein H 2 R in N-C (H) (R) -COOH is not equal to hydrogen, and the stereocenter of amino acid is in (S) -configuration; in some embodiments, the amino acid is a standard amino acid; in some embodiments, the amino acid is a non-standard amino acid. "Standard amino acid" refers to any of the twenty standard L-amino acids commonly found in naturally occurring peptides. "non-standard amino acid" refers to any amino acid other than a standard amino acid, whether synthetically prepared or obtained from a natural source. As will be clear from the context, in some embodiments, the term "amino acid" is used to refer to a free amino acid; in some embodiments, the term is used to refer to amino acid residues of a polypeptide. In this disclosure, the names of amino acids are also indicated by the standard one-letter or three-letter codes, as summarized below.
A = Ala = alanine T = Thr = threonine
V = Val = valine C = Cys = cysteine
L = Leu = leucine Y = Tyr = tyrosine
I = Ile = isoleucine N = Asn = asparagine
P = Pro = proline Q = Gln = glutamine
F = Phe = phenylalanine D = Asp = aspartic acid
W = Trp = tryptophan E = Glu = glutamic acid
M = Met = methionine K = Lys = lysine
G = Gly = glycine R = Arg = arginine
S = Ser = serine H = His = histidine
The terms "protein," "peptide," and "polypeptide" are used interchangeably herein and refer to a polymer of amino acid residues joined by covalent bonds, such as peptide bonds. A protein, peptide, or polypeptide as provided herein can include a natural amino acid, an unnatural amino acid, an analog or mimetic of an amino acid, or any combination thereof, in optically pure form or as a mixture of optical enantiomers. The proteins, peptides or polypeptides described herein may be obtained by any method known in the art, such as, but not limited to, by natural isolation, recombinant expression, chemical synthesis, and the like.
As used herein, the term "conservative substitution" refers to an amino acid residue that is physically or functionally similar to the corresponding reference residue. That is, a conservative substitution will have a similar size, shape, charge, chemical properties, including the ability to form a covalent or hydrogen bond, etc., as its reference residue. Preferred conservative substitutions are those that meet the criteria defined in the following documents for acceptable point mutations: dayhoff et al, atlas of Protein sequences and structures, vol.5, pp.345-352 (1978), which is incorporated herein by reference. Conservative substitutions may be made between amino acid residues having hydrophobic side chains (e.g., met, ala, val, leu, and Ile), between residues having neutral hydrophilic side chains (e.g., cys, ser, thr, asn, and Gln), between residues having acidic side chains (e.g., asp, glu), between amino acids having basic side chains (e.g., his, lys, and Arg), or between residues having aromatic side chains (e.g., trp, tyr, and Phe). As is known in the art, conservative substitutions typically do not result in significant changes in the conformational structure of the protein, and thus may preserve the biological activity of the protein.
As used herein, the term "analog" refers to a candidate amino acid sequence that is similar to a reference sequence, but retains the activity of the reference sequence.
As used herein, the term "percent (%) sequence identity" is defined as the percentage of amino acid (or nucleic acid) residues in a candidate sequence that are identical to the amino acid (or nucleic acid) residues in a reference sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum number of identical amino acids (or nucleic acids). In other words, the percentage (%) of sequence identity of an amino acid sequence (or nucleic acid sequence) can be calculated by dividing the number of identical amino acid residues (or bases) relative to a reference sequence to which it is compared by the total number of amino acid residues (or bases) in the candidate or reference sequence, whichever is shorter. Conservative substitutions of amino acid residues may or may not be considered identical residues. To determine the percent amino acid (or Nucleic acid) sequence identity, the alignment can be accomplished, for example, using publicly available tools such as BLASTN, BLASTp (available on the website of the National Center for Biotechnology Information (NCBI), see also Altschul S.F. et al, J.mol.biol., vol.215: pages 403-410 (1990); stephen F. Et al, nucleic Acids Res., vol.25: pages 3389-3402 (1997)), clustalW2 (available on the website of the European Bioinformatics institute, see also Higgins D.G. et al, methods in Enzymology, vol.266: pages 383-402 (1996)), larkin M.A. et al, bioinformatics (Taurushiga, england), vol.23, vol.21: pages 2947-2948 (2007)), and ALIGN (DNAh) (or Megali). One skilled in the art can use default parameters provided by the tool, or can customize parameters suitable for alignment, such as by selecting an appropriate algorithm.
As used herein, the term "chemical entity" refers to chemical molecules, chemical compounds, and fragments of such molecules or compounds. The chemical entity may be, for example, a ligand, substrate, nucleotide, agonist, antagonist, inhibitor, antibody, peptide, protein, or drug.
As used herein, the term "complex" refers to a polypeptide associated with a chemical entity. For example, "PfHT complex" refers to a complex comprising PfHT or an analog thereof, which is associated with a chemical entity by covalent or non-covalent binding forces at a binding site in a binding pocket disclosed herein. Non-limiting examples of PfHT complexes include PfHT or analogs thereof in combination with any of the compounds listed herein.
As used herein, the term "PfHT" or "PfHT polypeptide" refers to wild-type Plasmodium falciparum hexose transporter ("PfHT 1") whose amino acid sequence is set forth in SEQ ID NO:1 (NCBI accession No.: XP-001349558.1) below:
MTKSSKDICSENEGKKNGKSGFFSTSFKYVLSACIASFIFGYQVSVLNTIKNFIVVEFEWCKGEKDRLNCSNNTIQSSFLLASVFIGAVLGCGFSGYLVQFGRRLSLLIIYNFFFLVSILTSITHHFHTILFARLLSGFGIGLVTVSVPMYISEMTHKDKKGAYGVMHQLFITFGIFVAVMLGLAMGEGPKADSTEPLTSFAKLWWRLMFLFPSVISLIGILALVVFFKEETPYFLFEKGRIEESKNILKKIYETDNVDEPLNAIKEAVEQNESAKKNSLSLLSALKIPSYRYVIILGCLLSGLQQFTGINVLVSNSNELYKEFLDSHLITILSVVMTAVNFLMTFPAIYIVEKLGRKTLLLWGCVGVLVAYLPTAIANEINRNSNFVKILSIVATFVMIISFAVSYGPVLWIYLHEMFPSEIKDSAASLASLVNWVCAIIVVFPSDIIIKKSPSILFIVFSVMSILTFFFIFFFIKETKGGEIGTSPYITMEERQKHMTKSVV(SEQ ID NO:1)
as used herein, the term "wild-type PfHT" or "wild-type PfHT polypeptide" refers to the native sequence of full-length PfHT or a fragment thereof that is expressed in vivo in a subject (e.g., plasmodium falciparum).
As used herein, the term "structural coordinates" refers to cartesian coordinates derived from a mathematical equation relating to a pattern obtained by diffraction of a monochromatic X-ray beam by atoms (scattering centers) of a protein or protein complex in crystalline form. The diffraction data was used to calculate an electron density map of the repeating units of the crystal. These electron density maps are then used to determine the position of the individual atoms of the enzyme or enzyme complex.
Definitions of specific functional groups and chemical terms are described in more detail below. For purposes of this disclosure, chemical elements are identified according to the CAS version of the periodic table of elements in the inner cover of the "Handbook of Chemistry and Physics" 75 th edition, and specific functional groups are generally defined as described herein. In addition, the general principles of Organic Chemistry, as well as specific functional moieties and reactivity, are described in "Organic Chemistry", thomas Sorrell, 2 nd edition, university Science Books, sausalitio, 2006; smith and March, "March's Advanced Organic Chemistry", 6 th edition, john Wiley & Sons, inc., new York, 2007; larock, "Comprehensive Organic Transformations", 3 rd edition, VCH Publishers, inc., new York, 2018; carruthers, "Some Modern Methods of Organic Synthesis", 4 th edition, cambridge University Press, cambridge, 2004; wherein the entire contents of each of these documents is incorporated herein by reference.
In various positions of the disclosure, linking substituents are described. In the case where a linking group is explicitly required for the structure, the Markush (Markush) variables listed for that group should be understood as linking groups. For example, if a structure requires a linking group, and the Markush group definition for this variable lists "alkyl," it will be understood that "alkyl" refers to a linking alkylene group.
When a bond to a substituent is shown to cross a bond connecting two atoms in a ring, then such substituent can be bonded to any atom in the ring. When a substituent is listed without indicating through which atom such substituent is bonded to the remainder of the compound of a given formula, then such substituent may be bonded through any atom in such formula. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
When any variable (e.g. R) i ) When a compound occurs more than one time in any constituent or formula of the compound, its definition on each occurrence is independent of its definition at every other occurrence. Thus, for example, if the radical is shown to be substituted by 0 to 2R i Partially substituted, the radical may then be substituted by up to two R i Part of which is optionally substituted, and R i Independently at each occurrence selected from R i The definition of (1). Furthermore, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
As used herein, the term "C i-j "denotes a range of carbon numbers, where i and j are integers, and the range of carbon numbers includes the endpoints (i.e., i and j) and each integer point therebetween, and where j is greater than i. E.g. C 1-6 Represents a range of one to six carbon atoms, including one carbon atom, two carbon atoms, three carbon atoms, four carbon atoms, five carbon atoms, and six carbon atoms. In some embodiments, the term "C" or "C" refers to a compound having a lower molecular weight 1-12 "denotes 1 to 12, in particular 1 to 10, in particular 1 to 8, in particular 1 to 6, in particular 1 to 5, in particular 1 to 4, in particular 1 to 3 or in particular 1 to 2 carbon atoms.
As used herein, the term "alkyl", whether used as part of another term or independently, refers to a saturated straight or branched hydrocarbon radical, which may be optionally substituted independently with one or more substituents described below. The term "C i-j Alkyl "refers to an alkyl group having i to j carbon atoms. In some embodiments, the alkyl group contains 1 to 10 carbon atomsAnd (4) adding the active ingredients. In some embodiments, the alkyl group contains 1 to 9 carbon atoms. In some embodiments, the alkyl group contains 1 to 8 carbon atoms, 1 to 7 carbon atoms, 1 to 6 carbon atoms, 1 to 5 carbon atoms, 1 to 4 carbon atoms, 1 to 3 carbon atoms, or 1 to 2 carbon atoms. "C 1-10 Examples of alkyl groups "include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, and decyl. ' C 1-6 Examples of the alkyl group "are methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2, 3-dimethyl-2-butyl, 3-dimethyl-2-butyl and the like.
As used herein, the term "alkoxy", whether used as part of another term or independently, refers to an alkyl group, as previously defined, attached to the parent molecule through an oxygen atom. The term "C i-j Alkoxy "means that the alkyl portion of the alkoxy has from i to j carbon atoms. In some embodiments, alkoxy groups contain 1 to 10 carbon atoms. In some embodiments, the alkoxy group contains 1 to 9 carbon atoms. In some embodiments, alkoxy contains 1 to 8 carbon atoms, 1 to 7 carbon atoms, 1 to 6 carbon atoms, 1 to 5 carbon atoms, 1 to 4 carbon atoms, 1 to 3 carbon atoms, or 1 to 2 carbon atoms. "C 1-6 Examples of alkoxy "include, but are not limited to, methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), tert-butoxy, neopentyloxy, n-hexyloxy, and the like.
As used herein, the term "arylalkoxy" whether used as part of another term or independently, refers to an arylalkyl moiety substituted with one or more arylalkoxy moieties.
As used herein, the term "aryl", whether used as part of another term or independently, refers to monocyclic and polycyclic ring systems having a total of 5 to 20 ring members, wherein at least one ring of the ring system is aromatic, and wherein each ring of the ring system contains 3 to 12 ring members. Examples of "aryl" include, but are not limited to, phenyl, diphenyl, naphthyl, anthracenyl, and the like, which may bear one or more substituents. The term "aryl" as used herein also includes within its scope groups in which the aromatic ring is fused to one or more other rings. Where polycyclic ring systems are present, only one of these rings need be aromatic (e.g., 2, 3-indoline), although all of these rings can be aromatic (e.g., quinoline). The second ring may also be fused or bridged. Examples of polycyclic aryl groups include, but are not limited to, benzofuranyl, indanyl, phthalimidyl, naphthalimide, phenanthridinyl, or tetrahydronaphthyl, and the like. The aryl group may be substituted at one or more ring positions with substituents as described above.
As used herein, the term "alkylaryl", whether used as part of another term or independently, refers to an alkyl moiety substituted with one or more aryl moieties. Examples of alkylaryl include, but are not limited to, benzyl, ethylphenyl, and the like.
As used herein, the term "cycloalkyl", whether used as part of another term or independently, refers to monovalent non-aromatic, saturated or partially unsaturated monocyclic and polycyclic ring systems in which all ring atoms are carbon and which contain at least three ring-forming carbon atoms. In some embodiments, a cycloalkyl group can contain 3 to 12 ring-forming carbon atoms, 3 to 10 ring-forming carbon atoms, 3 to 9 ring-forming carbon atoms, 3 to 8 ring-forming carbon atoms, 3 to 7 ring-forming carbon atoms, 3 to 6 ring-forming carbon atoms, 3 to 5 ring-forming carbon atoms, 4 to 12 ring-forming carbon atoms, 4 to 10 ring-forming carbon atoms, 4 to 9 ring-forming carbon atoms, 4 to 8 ring-forming carbon atoms, 4 to 7 ring-forming carbon atoms, 4 to 6 ring-forming carbon atoms, 4 to 5 ring-forming carbon atoms. Cycloalkyl groups may be saturated or partially unsaturated. The cycloalkyl group may be substituted. In some embodiments, the cycloalkyl group can be a saturated cyclic alkyl group. In some embodiments, a cycloalkyl group may be a partially unsaturated cyclic alkyl group containing at least one double or triple bond in its ring system. In some embodiments, a cycloalkyl group can be monocyclic or polycyclic. Examples of monocyclic cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, and cyclododecyl. Examples of polycyclic cycloalkyl groups include, but are not limited to, adamantyl, norbornyl, fluorenyl, spiro-pentadienyl, spiro [3.6] -decyl, bicyclo [1, 1] pentenyl, bicyclo [2, 1] heptenyl, and the like.
As used herein, the term "halogen" refers to an atom selected from fluorine (or fluoro), chlorine (or chloro), bromine (or bromo), and iodine (or iodo).
As used herein, the term "haloalkyl" refers to an alkyl group having one or more halo substituents. Examples of haloalkyl groups include, but are not limited to, -CF 3 、-C 2 F 5 、-CHF 2 、-CCl 3 、-CHCl 2 、-C 2 Cl 5 And the like.
As used herein, the term "heteroatom" refers to nitrogen, oxygen or sulfur and includes any oxidized form of nitrogen or sulfur, as well as any quaternized form of basic nitrogen (including N-oxides).
As used herein, the term "heteroalkyl" refers to an alkyl group in which at least one of the carbon atoms is replaced with a heteroatom selected from N, O, or S. Heteroalkyl groups may be carbon radicals or heteroatom radicals (i.e., heteroatoms may be present in the middle or at the end of the radical), and may be optionally substituted.
As used herein, the term "heteroaryl," whether used as part of another term or independently, refers to an aryl group having one or more heteroatoms in addition to carbon atoms. The heteroaryl group may be monocyclic. Examples of monocyclic heteroaryl groups include, but are not limited to, thienyl, furyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, benzofuryl, and pteridinyl. Heteroaryl also includes polycyclic groups in which a heteroaromatic ring is fused to one or more aryl, alicyclic, or heterocyclic rings, wherein the radical or point of attachment is on the heteroaromatic ring. Examples of polycyclic heteroaryl groups include, but are not limited to, indolyl, isoindolyl, benzothienyl, benzofuranyl, benzo [1,3] dioxolyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzothiazolyl, quinolinyl, isoquinolinyl, dihydroquinolinyl, dihydroisoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and the like.
As used herein, the term "heterocyclyl" refers to a saturated or partially unsaturated carbocyclic group in which one or more ring atoms are heteroatoms independently selected from oxygen, sulfur, nitrogen, phosphorus, and the like, and the remaining ring atoms are carbon, wherein one or more ring atoms may be optionally substituted independently with one or more substituents. In some embodiments, the heterocyclyl is a saturated heterocyclyl. In some embodiments, heterocyclyl is a partially unsaturated heterocyclyl having one or more double bonds in its ring system. In some embodiments, heterocyclyl groups may contain any oxidized form of carbon, nitrogen, or sulfur, as well as any quaternized form of a basic nitrogen. "heterocyclyl" also includes radicals in which a heterocyclyl radical is fused to a saturated, partially unsaturated, or fully unsaturated (i.e., aromatic) carbocyclic or heterocyclic ring. The heterocyclyl radical may be carbon-linked or nitrogen-linked where possible. In some embodiments, the heterocycle is carbon-linked. In some embodiments, the heterocyclic ring is nitrogen-linked. For example, a group derived from pyrrole may be pyrrol-1-yl (nitrogen-linked) or pyrrol-3-yl (carbon-linked). Furthermore, the groups derived from imidazole may be imidazol-1-yl (nitrogen-linked) or imidazol-3-yl (carbon-linked).
The term "hexose" as used herein refers to a monosaccharide having six carbon atoms (monosaccharide @)
simple sugar). The hexose may be in the pyranose or furanose form. For example, glucose may be in the pyranose form (glucopyranose) or the furanose form (glucopyranose). The hexose may be the D-enantiomer or the L-enantiomer. In addition, hexoses include their alpha (α) and beta (β) isomers. Hexoses also include hexose derivatives such as deoxyhexoses, halogen-substituted hexoses, and the like. Examples of hexoses may include D-glucose, L-glucose, D-galactose, L-galactose, D-mannose, L-mannose, D-xylose, L-xylose, D-fructose, L-fructose, D-fucose, L-fucose, D-lyxose, D-altrose, 2-deoxy-D-glucose, 2-deoxy-2-halo-D-glucose, and the like.
As used herein, the term "partially unsaturated" refers to a free radical comprising at least one double or triple bond. The term "partially unsaturated" is intended to encompass rings having multiple sites of unsaturation, but is not intended to encompass aromatic (i.e., fully unsaturated) moieties.
As used herein, the term "substituted," whether preceded by the term "optionally" or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. It will be understood that "substitution" or "substituted" includes the implicit proviso that such substitution complies with the allowed valency of the atom being substituted, and that the substitution results in a stable or chemically feasible compound that, for example, does not spontaneously undergo transformation by means such as rearrangement, cyclization, elimination, and the like. Unless otherwise specified, a "optionally substituted" group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure can be substituted with more than one substituent selected from a specified group, the substituent may be the same or different at each position. It will be appreciated by those skilled in the art that the substituents themselves may be substituted where appropriate. Unless explicitly stated as "unsubstituted," chemical moieties mentioned herein are to be understood as including substituted variants. For example, reference to an "aryl" group or moiety implicitly includes both substituted and unsubstituted variants.
PfHT polypeptides and analogs thereof
Plasmodium falciparum hexose transporter, abbreviated in this disclosure as PfHT, is a glucose transporter essential to the erythrocyte endoparasite. Several PfHT subtypes have been identified in plasmodium falciparum species, which are available to those skilled in the art from published sources, such as those available from NCBI accession numbers: EWC90955.1, ETW46233.1, ETW32528.1, KNG76686.1 and the like. Various hexose transporters have also been identified in different species of plasmodium falciparum, which have a high percentage (e.g., 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) of sequence identity between the different species, particularly within the binding pocket region. Examples of hexose transporters include, but are not limited to, plasmodium vivax hexose transporter (PvHT), plasmodium ovale hexose transporter (PoHT), plasmodium malariae hexose transporter (PmHT), plasmodium falciparum hexose transporter (PkHT), the amino acid sequences of which are set forth in SEQ ID NOs: 2 to 5, respectively, below. FIG. 1 shows an alignment of the sequences of SEQ ID NO 1 to 5. The different hexose transporters between different PfHT polypeptide subtypes within plasmodium falciparum species and different species of plasmodium falciparum are collectively referred to as "PfHT analogs" in this disclosure.
Figure BDA0003863898460000091
As used herein, the term "PfHT analog" refers to a polypeptide having functional or structural characteristics substantially similar to all or part of the wild-type PfHT polypeptide provided herein (i.e., SEQ ID NO: 1), however, the amino acid sequence of the analog differs from the amino acid sequence of the wild-type PfHT polypeptide by at least one amino acid. The PfHT analog may be a partial fragment, derivative, or variant of a wild-type PfHT polypeptide, and may comprise a chemical or biological modification. The PfHT analogs can have conservative substitutions, additions, deletions, insertions, truncations, modifications (e.g., phosphorylation, glycosylation, labeling, etc.) at one or more amino acids of the wild-type PfHT polypeptide, or any combination thereof. PfHT analogs can include naturally occurring variants and artificially generated variants of wild-type PfHT polypeptides, such as artificial polypeptide sequences obtained by recombinant methods or chemical synthesis. PfHT analogs may comprise non-naturally occurring amino acid residues. One skilled in the art will appreciate that the PfHT analogs described herein still retain substantially similar function as the wild-type PfHT polypeptide, e.g., the PfHT analogs may still have the ability to mediate glucose uptake.
In certain embodiments, a PfHT analog as described herein includes a truncated mutant of a full-length wild-type PfHT polypeptide (i.e., a truncated fragment of a wild-type PfHT polypeptide), or a mutant having one or more amino acid mutations, additions, or deletions as compared to the wild-type PfHT polypeptide or a truncated mutant thereof. In certain embodiments, a PfHT analog as described herein can include a full-length wild-type PvHT, poHT, pmHT, pkHT polypeptide (i.e., SEQ ID NOS: 2 through 5), a truncated mutant thereof, or a mutant having one or more amino acid mutations, additions, or deletions as compared to the wild-type PvHT, poHT, pmHT, or PkHT polypeptide, or a truncated mutant thereof.
In certain embodiments, the PfHT analog is capable of transporting glucose at a level comparable to or no less than 30%, 40%, 50%, 60%, 70%, 80%, 90% of the wild-type PfHT polypeptide.
In certain embodiments, pfHT analogs provided herein comprise an amino acid sequence having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID No. 1 while retaining substantial (e.g., at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) biological activity of SEQ ID No. 1.
In certain embodiments, the PfHT analog comprises NO more than 9, 8, 7, 6, 5, 4, 3, or 2 substitutions relative to SEQ ID No. 1 while retaining the essential biological activity of SEQ ID No. 1. In certain embodiments, the PfHT analog comprises at least 2, 3, 4, 5, 6, 7, 8, or 9 substitutions relative to SEQ ID No. 1 while retaining a substantial (e.g., at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) biological activity of SEQ ID No. 1.
Binding pocket for PfHT polypeptides and PfHT analogs
Binding pockets have important uses in areas such as drug discovery. Many drugs exert their biological effects by binding to the binding pocket of a biological macromolecule, such as a receptor or an enzyme. Such binding may involve all or part of the amino acid residues that make up the binding pocket and may therefore aid in the design of drugs with improved biological effects.
The present disclosure is based, at least in part, on the elucidation of the crystal structure of PfHT complexes, which allows for the identification of binding pockets in PfHT-binding molecules. The present inventors have found that compound 3361, previously disclosed as a PfHT inhibitor (see Thierry)
Figure BDA0003863898460000105
Et al, acta Tropica, vol 89 (2004), pp 371-374), are capable of binding to PfHT and inducing conformational changes in PfHT which result in the formation of binding pockets as described herein.
In particular, the inventors have successfully identified at least three binding pockets for PfHT polypeptides and analogs thereof, which are designated in the present disclosure as "R1 binding pockets," R2 binding pockets, "and" R3 binding pockets.
As used herein, the term "binding pocket" refers to a three-dimensional configuration made up of a set of amino acid residues in a macromolecule, typically in the form of a cavity capable of receiving and interacting with one or more functional groups of a chemical entity (e.g., a molecule). The binding pocket of PfHT or an analog thereof may be defined by the coordinates of a set of amino acid residues present in PfHT or a set of equivalent residues present in a PfHT analog, which residues form at least a part of the binding pocket. The coordinates of an amino acid residue as provided herein are expressed in the single letter code for the amino acid residue followed by the position of the amino acid residue in the particular amino acid sequence. For example, F40 of SEQ ID NO:1 refers to the phenylalanine residue at position 40 of SEQ ID NO: 1.
It is contemplated that the set of amino acid residues that make up the R1, R2, and/or R3 binding pocket of PfHT is highly conserved among PfHT and its various analogs. Thus, when the amino acid coordinates of the R1, R2, or R3 binding pocket are referenced to SEQ ID NO:1, the skilled artisan can readily appreciate equivalent residues or coordinates of the R1, R2, or R3 binding pocket in PfHT analogs.
At least one or more of the R1, R2, and R3 binding pockets of PfHT or an analog thereof is an allosteric binding pocket. As used herein, an allosteric binding pocket refers to a binding pocket that is not present in the three-dimensional conformation of an isolated polypeptide, but that can be induced as a result of the polypeptide changing its conformation upon binding to a molecule. That is, these binding pockets are not naturally occurring, but rather the PfHT polypeptide or analog thereof is induced upon binding to a molecule (e.g., compound 3361, etc.). The molecule may be an inhibitor of PfHT activity, including but not limited to the molecules and compounds of the present disclosure and compound 3361.
In some embodiments, the binding pocket of PfHT or an analog thereof may be defined by amino acid residues of PfHT or an analog thereof that comprise any atom in a molecule that is at the time the molecule binds to PfHT or an analog thereof
Figure BDA0003863898460000101
Inside (e.g. in)
Figure BDA0003863898460000102
) Of (c) is used. In some embodiments, the binding pocket of PfHT or an analog thereof may be further defined by amino acid residues of PfHT or an analog thereof that are located near the molecule to which they are bound and that comprise more than one atom from the molecule
Figure BDA0003863898460000103
But less than
Figure BDA0003863898460000104
The atom (c) of (a).
In one aspect, a PfHT polypeptide or analog thereof is found to comprise an R1 binding pocket, the polypeptide having the amino acid sequence of SEQ ID No. 1, the analog having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID No. 1.
In some embodiments, the R1 binding pocket comprises at least one or more (e.g., two, three, four, five, six, seven) amino acid residues selected from the equivalent residues in Q169, Q305, Q306, N311, N341, W412, and N435 of SEQ ID NO:1, or analogs thereof. As used herein, the term "equivalent residue" refers to an amino acid residue in a PfHT analog that corresponds to an amino acid residue of a certain binding pocket of SEQ ID NO:1 when the amino acid sequence of the PfHT analog is aligned with SEQ ID NO:1 (gaps are introduced, if necessary, to achieve the maximum number of identical amino acids), and the position numbering of the equivalent residue is its actual position in the amino acid sequence of the PfHT analog. For example, SEQ ID NO 2 is the full-length amino acid sequence of wild-type PvHT, which has 502 amino acids. When the amino acid sequence of SEQ ID NO. 2 is aligned with SEQ ID NO. 1 having 504 amino acids, two gaps should be introduced in the amino acid sequence of SEQ ID NO. 2 in order to maximize the number of identical amino acids. Thus, Q167 of SEQ ID NO. 2 corresponds to Q169 of SEQ ID NO. 1, such that Q167 of SEQ ID NO. 2 is the equivalent residue of Q169 of SEQ ID NO. 1. Once the amino acid sequences of other PfHT analogs are known and aligned with SEQ ID NO:1, one skilled in the art can readily identify equivalent residues for these other PfHT analogs.
In this case, in some embodiments, the R1 binding pocket may comprise at least one or more (e.g., two, three, four, five, six, seven) amino acid residues selected from Q167, Q303, Q304, N309, N339, W410, and N433 of SEQ ID NO: 2. In some embodiments, the R1 binding pocket may comprise at least one or more (e.g., two, three, four, five, six, seven) amino acid residues selected from Q168, Q304, Q305, N310, N340, W411, and N434 of SEQ ID NO: 3. In some embodiments, the R1 binding pocket may comprise at least one or more (e.g., two, three, four, five, six) amino acid residues selected from Q126, Q262, Q263, N268, N298, H369 of SEQ ID No. 4. In some embodiments, the R1 binding pocket may comprise at least one or more (e.g., two, three, four, five, six, seven) amino acid residues selected from Q166, Q301, Q302, N307, N337, W408, and N431 of SEQ ID NO: 5.
In some embodiments, the R1 binding pocket further comprises one or more (e.g., two, three, four, five, six) additional amino acid residues selected from equivalent residues in F40, I172, I176, I310, F403, and a404 of SEQ ID NO:1, or analogs thereof. In some embodiments, the R1 binding pocket further comprises at least one or more (e.g., two, three, four, five, six) amino acid residues selected from F40, I170, I174, I308, F401, and a402 of SEQ ID NO: 2. In some embodiments, the R1 binding pocket further comprises at least one or more (e.g., two, three, four, five, six) amino acid residues selected from F40, I171, I175, I309, F402, and a403 of SEQ ID NO: 3. In some embodiments, the R1 binding pocket further comprises at least one or more (e.g., two, three, four, five, six) amino acid residues selected from F39, I129, I133, I267, Q360, and K361 of SEQ ID No. 4. In some embodiments, the R1 binding pocket further comprises at least one or more (e.g., two, three, four, five, six) amino acid residues selected from F39, I169, I173, I306, F399, and A400 of SEQ ID NO: 5.
In some embodiments, the R1 binding pocket further comprises one or more (e.g., two, three, four) additional amino acid residues selected from the group consisting of T145, T173, V314, and I400 of SEQ ID NO:1, or equivalent residues in analogs thereof. In some embodiments, the R1 binding pocket further comprises at least one or more (e.g., two, three, four) amino acid residues selected from T143, T171, V312, and I398 of SEQ ID No. 2. In some embodiments, the R1 binding pocket further comprises at least one or more (e.g., two, three, four) amino acid residues selected from T144, T172, V313, and I399 of SEQ ID NO: 3. In some embodiments, the R1 binding pocket further comprises at least one or more (e.g., two, three, four) amino acid residues selected from T102, T130, V271, and L357 of SEQ ID No. 4. In some embodiments, the R1 binding pocket further comprises at least one or more (e.g., two, three, four) amino acid residues selected from T142, T170, V310, and I396 of SEQ ID NO: 5.
In another aspect, it is found that a PfHT polypeptide or analog thereof, which has an amino acid sequence of SEQ ID NO:1, also includes an R2 binding pocket, has at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID NO: 1.
In some embodiments, the R2 binding pocket comprises at least one or more (e.g., two, three, four, five) amino acid residues selected from the group consisting of equivalent residues in V44, L47, F85, W436, and V443 of SEQ ID NO:1 or analogs thereof. In some embodiments, the R2 binding pocket may comprise at least one or more (e.g., two, three, four, five) amino acid residues selected from V44, L47, F83, W434, and V441 of SEQ ID No. 2. In some embodiments, the R2 binding pocket may comprise at least one or more (e.g., two, three, four, five) amino acid residues selected from V44, L47, F84, W435, and V442 of SEQ ID NO: 3. In some embodiments, the R2 binding pocket may comprise at least one or more (e.g., two, three) amino acid residues selected from V43, L46, and F83 of SEQ ID NO: 4. In some embodiments, the R2 binding pocket may comprise at least one or more (e.g., two, three, four, five) amino acid residues selected from V43, L46, F82, W432, and V439 of SEQ ID No. 5.
In some embodiments, the R2 binding pocket further comprises one or more (e.g., two, three, four) additional amino acid residues selected from the group consisting of equivalent residues in L81, V312, a439, and F444 of SEQ ID No. 1, or analogs thereof. In some embodiments, the R2 binding pocket further comprises at least one or more (e.g., two, three, four) amino acid residues selected from L79, V310, A437 and F442 of SEQ ID NO: 2. In some embodiments, the R2 binding pocket further comprises at least one or more (e.g., two, three, four) amino acid residues selected from L80, V311, A438, and F443 of SEQ ID NO: 3. In some embodiments, the R2 binding pocket further comprises at least one or two amino acid residues selected from L79 and V269 of SEQ ID NO 4. In some embodiments, the R2 binding pocket further comprises at least one or more (e.g., two, three, four) amino acid residues selected from L78, V308, a435, and F440 of SEQ ID No. 5.
In another aspect, a PfHT polypeptide or analog thereof is found that further comprises an R3 binding pocket, the polypeptide having the amino acid sequence of SEQ ID No. 1, the analog having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID No. 1.
In some embodiments, the R3 binding pocket comprises at least one or more (e.g., two, three, four, five, six, seven, eight) amino acid residues selected from the equivalent residues of N48, K51, N52, N311, N316, N318, E319, and D447, or analogs thereof, of SEQ ID NO: 1. In some embodiments, the R3 binding pocket may comprise at least one or more (e.g., two, three, four, five, six, seven, eight) amino acid residues selected from N48, K51, N52, N309, N314, N316, E317, and D445 of SEQ ID NO: 2. In some embodiments, the R3 binding pocket may comprise at least one or more (e.g., two, three, four, five, six, seven, eight) amino acid residues selected from N48, K51, N52, N310, N315, N317, E318, and D446 of SEQ ID NO: 3. In some embodiments, the R3 binding pocket may comprise at least one or more (e.g., two, three, four, five, six, seven) amino acid residues selected from N47, K50, N51, N268, N273, N275, and E276 of SEQ ID No. 4. In some embodiments, the R3 binding pocket may comprise at least one or more (e.g., two, three, four, five, six, seven, eight) amino acid residues selected from N47, K50, D51, N307, N312, N314, a315, and D443 of SEQ ID NO: 5.
In some embodiments, the R3 binding pocket further comprises one or more (e.g., two, three, four, five) additional amino acid residues selected from the group consisting of F85, V312, S315, V443, and F444 of SEQ ID NO:1 or an equivalent residue in an analog thereof. In some embodiments, the R3 binding pocket further comprises at least one or more (e.g., two, three, four, five) amino acid residues selected from F83, V310, S313, V441, and F442 of SEQ ID No. 2. In some embodiments, the R3 binding pocket further comprises at least one or more (e.g., two, three, four, five) amino acid residues selected from F84, V311, a314, V442, and F443 of SEQ ID NO: 3. In some embodiments, the R3 binding pocket further comprises at least one or more (e.g., two, three) amino acid residues selected from F83, V269, and S272 of SEQ ID NO: 4. In some embodiments, the R3 binding pocket further comprises at least one or more (e.g., two, three, four, five) amino acid residues selected from F82, V308, S311, V439, and F440 of SEQ ID No. 5.
The group of amino acids listed above represents the residues that define the allosteric binding pocket induced when compound 3361 is bound to a PfHT polypeptide or its equivalent in a PfHT analog. The residues that make up the binding pocket or a portion thereof may be used specifically to define the chemical environment of the binding pocket, or to design a molecule capable of interacting with those residues. For example, such residues may be key residues that play a role in ligand binding, or may be spatially related and define the three-dimensional compartments of the binding pocket. In the primary sequence of the PfHT polypeptide or analog thereof, the residues may be contiguous or non-contiguous.
Molecules capable of binding to the binding pocket of a PfHT polypeptide or PfHT analog
The identified binding pocket of the PfHT polypeptide or analog thereof can be used to design a molecule capable of binding to the binding pocket or a portion thereof. These molecules may be potential inhibitors of PfHT polypeptides or analogs thereof.
As used herein, the term "and 8230; \8230binding (binding to),"' and 8230; (binding to) "or" and 8230; \8230, binding 8230, binding (bind to) "with respect to a binding pocket refers to the proximity between a chemical entity or portion thereof and a binding pocket or binding site of a protein. The chemical entity or portion thereof can be bound to the binding pocket or binding site of the protein covalently (e.g., they share at least one pair of electrons) or non-covalently (e.g., supported by hydrogen bonds or van der waals forces or the energy of electrostatic interactions), or the chemical entity or portion thereof can be surrounded by the binding pocket or binding site of the protein. In some embodiments, the covalent binding is reversible. In some embodiments, the covalent binding is irreversible. In this disclosure, the terms "with 8230; \8230, binding", "with 8230; \8230, interaction", "with 8230; \8230, association (associationwith)" and "with 8230; \8230, association (associatedto)" are used interchangeably.
It will be appreciated by those skilled in the art that the molecule which binds to the binding pocket need not bind to every residue as defined in the binding pocket. For example, depending on the size and functional group of the molecule, binding interactions may or may not occur between the molecule and some of the identified amino acid residues. The calculated length of allowed van der waals interactions is also a factor in determining whether an amino acid residue in the binding pocket binds to a molecule. Thus, it is understood that the molecules of the present disclosure are capable of binding to the identified set of amino acid residues (or a subset thereof) of each binding pocket. A molecule is considered to bind to a binding pocket sufficiently if it binds to at least some of the identified residues in the binding pocket in a manner that allows sufficient interaction or binding between the molecule and the binding pocket. Sufficient interaction orBinding can be measured by methods well known in the art, such as by measuring Kd values, EC bound 50 Competitive binding of IC 50 And so on.
In one aspect, the present disclosure provides a molecule capable of binding to both the R1 binding pocket and the R2 binding pocket of a PfHT polypeptide having the amino acid sequence of SEQ ID NO:1 and an analog thereof having at least 70% sequence identity to SEQ ID NO:1, wherein the R1 binding pocket comprises at least one or more (e.g., two, three, four, five, six, seven) amino acid residues selected from the group consisting of equivalent residues in Q169, Q305, Q306, N311, N341, W412, and N435 of SEQ ID NO:1 or an analog thereof; and the R2 binding pocket comprises at least one or more (e.g., two, three, four, five) amino acid residues selected from the equivalent residues of V44, L47, F85, W436, and V443 of SEQ ID NO 1 or analogs thereof;
Wherein the molecule is not compound 3361 having the formula:
Figure BDA0003863898460000131
in some embodiments, the R1 binding pocket further comprises one or more (e.g., two, three, four, five, six) additional amino acid residues selected from equivalent residues in F40, I172, I176, I310, F403, and a404 of SEQ ID NO:1, or analogs thereof.
In some embodiments, the R1 binding pocket further comprises one or more (e.g., two, three, four) additional amino acid residues selected from the group consisting of T145, T173, V314, and I400 of SEQ ID NO:1, or equivalent residues in analogs thereof.
In some embodiments, the R2 binding pocket further comprises one or more (e.g., two, three, four) additional amino acid residues selected from the group consisting of equivalent residues in L81, V312, a439, and F444 of SEQ ID NO:1, or analogs thereof.
In some embodiments, the molecule is capable of further binding to the R3 binding pocket of a PfHT polypeptide or analog thereof, wherein the R3 binding pocket comprises at least one or more (e.g., two, three, four, five, six, seven, eight) amino acid residues selected from the group consisting of equivalent residues in N48, K51, N52, N311, N316, N318, E319, and D447 of SEQ ID NO:1 or an analog thereof.
In some embodiments, the R3 binding pocket further comprises one or more (e.g., two, three, four, five) additional amino acid residues selected from the group consisting of F85, V312, S315, V443, and F444 of SEQ ID NO:1 or an equivalent residue in an analog thereof.
In some embodiments, binding of a molecule of the present disclosure to a PfHT polypeptide or analog thereof can induce the formation of R1, R2, and/or R3 binding pockets, and the molecule binds to these binding pockets.
Binding of the molecule to the PfHT polypeptide or analog thereof can be measured by any method known in the art. In some embodiments, the method is semi-quantitative or quantitative.
In certain embodiments, binding of the molecule to the PfHT polypeptide or analog thereof can be measured directly, e.g., by NMR or surface plasmon resonance.
In some embodiments, if the molecule also serves as a substrate for the enzymatic activity of the PfHT polypeptide or analog thereof, the enzymatic reaction product can be measured (e.g., the amount of substrate transported by the PfHT polypeptide or analog thereof can be measured by a transport/uptake assay; the amount of small molecule substrate can be measured by measuring the amount of cleaved substrate with an analytical tool; the amount of biomolecule substrate can be measured by measuring the amount of cleaved substrate on a Western blot). Alternatively, the molecule itself may exhibit enzymatic properties, and the molecule bound to the peptide can be contacted with a suitable substrate, allowing detection by generation of an intensity signal. For the measurement of enzymatic reaction products, in some embodiments, the amount of substrate is saturated. The substrate may also be labeled with a detectable label prior to the reaction. In some embodiments, the sample is contacted with the substrate for a sufficient period of time. A sufficient amount of time refers to the time necessary to produce a detectable or measurable amount of product. Instead of measuring the amount of product, the time required for a given (e.g., detectable) amount of product to occur can be measured.
In certain embodiments, binding of the molecule to the PfHT polypeptide or analog thereof can be measured by using a labeled form of the molecule. The molecule may be covalently or non-covalently linked to a label, thereby allowing detection and measurement of the molecule. Labeling can be done by direct or indirect methods. Direct labeling involves coupling the label directly (covalently or non-covalently) to the agent. Indirect labeling involves binding (covalently or non-covalently) a secondary reagent to a primary reagent. The secondary reagent should bind specifically to the primary reagent. The secondary agent can be conjugated to a suitable label and/or act as a target (receptor) for a tertiary agent that binds to the secondary agent. Secondary, tertiary or even higher order reagents are often used to increase signal intensity. Suitable secondary and higher reagents may include antibodies, secondary antibodies, and the well-known streptavidin-biotin system (Vector Laboratories, inc.). The reagent or substrate may also be "labeled" with one or more labels as known in the art. Such tags can then be targets for higher-order agents. Suitable tags include biotin, digoxin, his tag, glutathione-S-transferase, FLAG, GFP, myc tag, influenza A virus Hemagglutinin (HA), maltose binding protein, and the like. In the case of peptides or polypeptides, the tag may be located at the N-terminus and/or C-terminus.
In certain embodiments, binding of the molecule to the PfHT polypeptide or analog thereof can be measured by microcalorimetry ("MST"), which is based on detecting a temperature-induced change in fluorescence of the target as a function of non-fluorescent ligand concentration. The observed change in fluorescence is based on two different effects. In one aspect, it is based on the temperature-dependent intensity change (TRIC) of the fluorescent probe, which may be affected by the binding event. On the other hand, it is based on thermophoresis, i.e. the directional movement of particles in microscopic temperature gradients. Any change in the chemical microenvironment of the fluorescent probe, as well as changes in the hydration layer of the biomolecule, results in a relative change in the fluorescence detected upon application of the temperature gradient, and can be used to determine binding affinity. MST allows the measurement of interactions directly in solution without fixation to a surface (immobilizer technique) (see maximian g. Plate et al, bio-Protocol, vol 7, 23: electronic edition, p 2632; moran junerbek-Willemsen et al, journal of Molecular Structure, vol 1077, p 101-113).
In some embodiments, binding of a molecule of the present disclosure to a PfHT polypeptide or analog thereof is measured by the MST method. In certain embodiments, binding of a molecule of the present disclosure to a PfHT polypeptide or analog thereof is measured by the MST method as described in example 13.
In some embodiments, a molecule of the disclosure is capable of binding to a PfHT polypeptide or analog thereof, wherein the Kd value is no more than 20 μ Μ, no more than 15 μ Μ, no more than 10 μ Μ, no more than 5 μ Μ, no more than 4 μ Μ, no more than 3 μ Μ, no more than 2 μ Μ, no more than 1 μ Μ, no more than 900nM, no more than 800nM, no more than 700nM, no more than 600nM, no more than 500nM, no more than 400nM, no more than 300nM, no more than 200nM, no more than 100nM, no more than 90nM, or no more than 80nM, as determined by the MST method described in example 13.
In some embodiments, the molecule capable of binding to a PfHT polypeptide or analog thereof is a peptide, polypeptide, or small molecule compound.
In some embodiments, the molecule capable of binding to a PfHT polypeptide or analog thereof comprises an M1 moiety, an M2 moiety, and an M3 moiety covalently linked together.
In some embodiments, the molecules provided herein bind to a PfHT polypeptide or analog thereof, wherein the M1 portion of the molecule binds to the R1 binding pocket of the PfHT polypeptide or analog thereof.
In some embodiments, the M1 portion of the molecules provided herein binds to one or more amino acid residues of the R1 binding pocket of a PfHT polypeptide or analog thereof through polar interaction.
In some embodiments, the M1 moiety binds by polar interaction to one or more (e.g., two, three, four, five, six, seven) amino acid residues of the R1 binding pocket selected from the equivalent residues in Q169, Q305, Q306, N311, N341, W412, and N435 of SEQ ID NO:1, or an analog thereof.
In some embodiments, the M1 portion of the molecules provided herein binds to one or more amino acid residues of the R1 binding pocket of a PfHT polypeptide or analog thereof through hydrophobic interactions.
In some embodiments, the M1 moiety binds by hydrophobic interaction to one or more (e.g., two, three, four, five, six) amino acid residues of the R1 binding pocket selected from equivalent residues in F40, I172, I176, I310, F403, and a404 of SEQ ID NO:1, or analogs thereof.
In some embodiments, the M1 portion of the molecules provided herein is surrounded by one or more amino acid residues of the R1 binding pocket of a PfHT polypeptide or analog thereof.
In some embodiments, the M1 portion of the molecules provided herein is surrounded by one or more (e.g., two, three, four) amino acid residues of the R1 binding pocket selected from the group consisting of T145, T173, V314, and I400 of SEQ ID No. 1, or equivalent residues in analogs thereof.
In some embodiments, the M1 moiety of the molecules provided herein comprises a hexose moiety. In some embodiments, the M1 portion of the molecules provided herein comprises a hexose moiety in a cyclic form. In some embodiments, the M1 portion of the molecules provided herein includes the D-enantiomer of the hexose moiety. In some embodiments, the M1 portion of the molecules provided herein includes the L-enantiomer of the hexose moiety.
In some embodiments, the M1 moiety of the molecules provided herein comprises a glucose moiety. In some embodiments, the M1 portion of the molecules provided herein comprises a glucose moiety in cyclic form. In some embodiments, the M1 moiety of the molecules provided herein comprises a glucopyranose moiety. In some embodiments, the M1 portion of the molecules provided herein comprises a glucofuranose moiety.
In some embodiments, the M1 moiety of the molecules provided herein comprises a D-glucose, L-glucose, D-galactose, L-galactose, D-mannose, D-xylose, D-fructose, 2-deoxy-D-glucose, or 2-deoxy-2-halo-D-glucose moiety.
In some embodiments, the hexose moiety is covalently attached to the M2 moiety through its carbon, nitrogen, oxygen, or sulfur atom. In some embodiments, the hexose moiety is covalently attached to the M2 moiety through its oxygen atom. In some embodiments, the hexose moiety is covalently linked to the M2 moiety through its nitrogen atom. In some embodiments, the hexose moiety is covalently linked to the M2 moiety through its sulfur atom. In some embodiments, the hexose moiety is covalently attached to the M2 moiety through its carbon atom.
In some embodiments, the hexose moiety is covalently linked to the M2 moiety at a position selected from the group consisting of the 1-position, 2-position, 3-position, 4-position, and 6-position. In some embodiments, the hexose moiety is covalently attached to the M2 moiety through its oxygen atom. In some embodiments, the hexose moiety is covalently linked to the M2 moiety through its oxygen atom at the 2-position. In some embodiments, the hexose moiety is covalently linked to the M2 moiety through its oxygen atom at the 3-position. In some embodiments, the D-glucose moiety is covalently attached to the M2 moiety through its oxygen atom at the 6-position.
In some embodiments, the M1 moiety is a D-glucose moiety. In some embodiments, the D-glucose moiety is a D-glucopyranose moiety. In some embodiments, the D-glucose moiety is a D-glucopyranose moiety.
In some embodiments, the D-glucose moiety is covalently attached to the M2 moiety through its oxygen atom. In some embodiments, the D-glucose moiety is covalently attached to the M2 moiety through its oxygen atom at the 2-position. In some embodiments, the D-glucose moiety is covalently attached to the M2 moiety through its oxygen atom at the 3-position. In some embodiments, the D-glucose moiety is covalently attached to the M2 moiety through its oxygen atom at the 6-position.
In some embodiments, the M1 moiety is an L-glucose moiety. In some embodiments, the L-glucose moiety is an L-glucopyranose moiety. In some embodiments, the L-glucose moiety is an L-glucopyranose moiety.
In some embodiments, the L-glucose moiety is covalently attached to the M2 moiety through its oxygen atom. In some embodiments, the L-glucose moiety is covalently attached to the M2 moiety through its oxygen atom at a position selected from the group consisting of 1-position, 2-position, 3-position, 4-position, and 6-position. In some embodiments, the L-glucose moiety is covalently attached to the M2 moiety through its oxygen atom at the 3-position.
In some embodiments, the M1 moiety is a deoxy-D-glucose moiety. In some embodiments, the deoxy-D-glucose moiety is a deoxy-D-glucopyranose moiety. In some embodiments, the deoxy-D-glucose moiety is a deoxy-D-glucofuranose moiety. In some embodiments, the M1 moiety is a 2-deoxy-D-glucose moiety. In some embodiments, the deoxy-D-glucose moiety is a 2-deoxy-D-glucopyranose moiety. In some embodiments, the deoxy-D-glucose moiety is a 2-deoxy-D-glucofuranose moiety.
In some embodiments, the deoxy-D-glucose moiety is covalently attached to the M2 moiety through its oxygen atom. In some embodiments, the deoxy-D-glucose moiety is covalently attached to the M2 moiety through its oxygen atom at a position selected from the group consisting of 1-position, 3-position, 4-position, and 6-position. In some embodiments, the deoxy-D-glucose moiety is a 2-deoxy-D-glucose moiety covalently linked to the M2 moiety through its oxygen atom at the 3-position.
In some embodiments, the M1 moiety is a D-fructose moiety. In some embodiments, the D-fructose moiety is a D-fructopyranose moiety. In some embodiments, the D-fructose moiety is a D-fructofuranose moiety.
In some embodiments, the D-fructose moiety is covalently attached to the M2 moiety through its oxygen atom. In some embodiments, the D-fructose moiety is covalently attached to the M2 moiety through its oxygen atom at a position selected from the group consisting of the 1-position, 2-position, 3-position, 4-position, and 5-position. In some embodiments, the D-fructose moiety is covalently attached to the M2 moiety through its oxygen atom at the 1-position.
In some embodiments, the molecules provided herein bind to a PfHT polypeptide or analog thereof, wherein the M2 portion of the molecule binds to the R2 binding pocket of the PfHT polypeptide or analog thereof.
In some embodiments, the M2 portion of the molecules provided herein binds to one or more amino acid residues of the R2 binding pocket of a PfHT polypeptide or analog thereof through hydrophobic interactions.
In some embodiments, the M2 moiety interacts with or binds to one or more (e.g., two, three, four, five) amino acid residues of the R2 binding pocket selected from the group consisting of V44, L47, F85, W436, and V443 of SEQ ID NO:1, or equivalent residues in analogs thereof, by hydrophobic interaction.
In some embodiments, the M2 portion of the molecules provided herein is surrounded by one or more amino acid residues of the R2 binding pocket of a PfHT polypeptide or analog thereof.
In some embodiments, the M2 portion of the molecules provided herein is surrounded by one or more (e.g., two, three, four) amino acid residues of the R2 binding pocket selected from the group consisting of L81, V312, a439, and F444 of SEQ ID NO:1, or equivalent residues in analogs thereof.
In some embodiments, the M2 moiety of the molecules provided herein comprises an optionally substituted linear hydrocarbon moiety having a length of 6 to 12 atoms.
In some embodiments, the M2 moiety of the molecules provided herein comprises an optionally substituted straight or heteroalkyl moiety having a length of 6 to 12 atoms.
In some embodiments, the M3 portion of the molecules provided herein binds to one or more amino acid residues of the R3 binding pocket of a PfHT polypeptide or analog thereof through polar interaction.
In some embodiments, the M3 moiety interacts with one or more (e.g., two, three, four, five, six, seven, eight) amino acid residues in the R3 binding pocket selected from the group consisting of N48, K51, N52, N311, N316, N318, E319, and D447 of SEQ ID No. 1, or an equivalent residue thereof, through polar interaction.
In some embodiments, the M3 portion of the molecules provided herein is surrounded by one or more amino acid residues of the R3 binding pocket of a PfHT polypeptide or analog thereof.
In some embodiments, the M3 portion of the molecules provided herein is surrounded by one or more (e.g., two, three, four, five) amino acid residues of the R3 binding pocket selected from the equivalent residues in F85, V312, S315, V443, and F444 of SEQ ID NO:1, or analogs thereof.
In some embodiments, the M3 portion of the molecules provided herein includes an optionally substituted aromatic or non-aromatic cyclic moiety.
In some embodiments, the M3 portion of the molecules provided herein includes an optionally substituted aromatic moiety.
In some embodiments, the M3 portion of the molecules provided herein comprises an optionally substituted aryl or heteroaryl moiety.
In some embodiments, the M3 portion of the molecules provided herein comprises an optionally substituted 5-to 12-membered aryl or heteroaryl moiety.
In some embodiments, the M3 portion of the molecules provided herein comprises an optionally substituted non-aromatic cyclic moiety.
In some embodiments, the M3 portion of the molecules provided herein includes an optionally substituted cycloalkyl or heterocyclyl moiety.
In some embodiments, the M3 portion of the molecules provided herein includes an optionally substituted 5 to 12 membered cycloalkyl or heterocyclyl moiety.
In some embodiments, the molecules provided herein are capable of inhibiting the activity of a PfHT polypeptide or analog thereof upon binding to the binding pocket of the PfHT polypeptide or analog thereof.
In some embodiments, the PfHT analogs used herein are selected from PvHT, poHT, pmHT, and PkHT. In some embodiments, the amino acid sequence of the PfHT analog is selected from the group consisting of SEQ ID NO 2, SEQ ID NO 3, SEQ ID NO 4, and SEQ ID NO 5.
PfHT complexes
In another aspect, the disclosure provides a complex comprising a PfHT polypeptide having the amino acid sequence of SEQ ID NO:1, or an analog thereof having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID NO:1, bound to a molecule provided herein.
In some embodiments, the PfHT analogs used herein are selected from PvHT, poHT, pmHT, and PkHT. In some embodiments, the amino acid sequence of the PfHT analog is selected from the group consisting of SEQ ID NO 2, SEQ ID NO 3, SEQ ID NO 4, and SEQ ID NO 5.
In some embodiments, a complex provided herein comprises a polypeptide having an amino acid sequence of any one of SEQ ID NOs 2 to 5, or an analog thereof having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to a molecule provided herein.
In some embodiments, in a complex, a molecule provided herein binds to the R1 binding pocket and the R2 binding pocket of equivalent residues in SEQ ID No. 1 or an analog thereof, wherein the R1 binding pocket comprises at least one or more (e.g., two, three, four, five, six, seven) amino acid residues selected from the equivalent residues in Q169, Q305, Q306, N311, N341, W412, and N435 of SEQ ID No. 1 or an analog thereof; and the R2 binding pocket comprises at least one or more (e.g., two, three, four, five) amino acid residues selected from the equivalent residues of V44, L47, F85, W436, and V443 of SEQ ID NO:1 or analogs thereof.
In some embodiments, the R1 binding pocket further comprises one or more (e.g., two, three, four, five, six) additional amino acid residues selected from equivalent residues in F40, I172, I176, I310, F403, and a404 of SEQ ID NO:1, or analogs thereof.
In some embodiments, the R1 binding pocket further comprises one or more (e.g., two, three, four) additional amino acid residues selected from the group consisting of T145, T173, V314, and I400 of SEQ ID NO:1, or equivalent residues in analogs thereof.
In some embodiments, the R2 binding pocket further comprises one or more (e.g., two, three, four) additional amino acid residues selected from the group consisting of equivalent residues in L81, V312, a439, and F444 of SEQ ID No. 1, or analogs thereof.
In some embodiments, in the PfHT complex, the molecules provided herein further bind to the R3 binding pocket of the PfHT polypeptide or analog thereof, wherein the R3 binding pocket comprises at least one or more (e.g., two, three, four, five, six, seven, eight) amino acid residues selected from equivalent residues in N48, K51, N52, N311, N316, N318, E319, and D447 of SEQ ID NO:1 or an analog thereof.
In some embodiments, the R3 binding pocket further comprises one or more (e.g., two, three, four, five) additional amino acid residues selected from the equivalent residues in F85, V312, S315, V443, and F444 of SEQ ID NO:1, or analogs thereof.
In some embodiments, a complex of the disclosure is formed between a PfHT polypeptide or analog thereof and another compound (e.g., a compound provided herein and compound 3361). For example, the complex of the present disclosure is formed between a PfHT polypeptide having the amino acid sequence of SEQ ID NO:1 and compound 3361.
In some embodiments, the PfHT complex is crystalline. The crystallized PfHT complex may provide a means to obtain atomic modeling information for particular amino acids and their atoms that form binding pockets and interact with molecules that fit within the binding pockets. The crystallized PfHT complex may also provide modeling information about protein-molecule interactions as well as the molecular structure of the interaction with the protein.
In some embodiments, the crystalline PfHT complexes of the present disclosure are capable of diffracting X-rays to better than
Figure BDA0003863898460000161
Figure BDA0003863898460000162
Or
Figure BDA0003863898460000163
And can be used to determine the three-dimensional structure of the molecule.
The crystallized PfHT complex may be prepared by any method known to those skilled in the art. In some embodiments, the crystalline PfHT complex is prepared by a lipid cubic phase ("LCP") process comprising the steps of:
(a) Pre-incubating a purified PfHT polypeptide or analog thereof with a compound to form a first mixture;
(b) Mixing the first mixture obtained in step (a) with a lipid to form an intermediate phase;
(c) Mixing the intermediate phase obtained in step (b) with a crystallization screening solution to form a second mixture; and
(d) Crystallizing the PfHT complex from the second mixture obtained in step (c) under conditions suitable for crystallization, thereby obtaining a crystallized PfHT complex.
The lipid used in step (b) may be any LCP host lipid known in the art, including but not limited to oleic acid Monoglyceride (MO), palmitoleic acid Monoglyceride (MP), vaccenic acid Monoglyceride (MV), and eicosenoic acid Monoglyceride (ME). In some embodiments, the lipid used in step (b) is oleic acid monoglyceride.
In some embodiments, the first mixture is mixed with the lipid at a protein to lipid ratio (w/w) (e.g., at a protein to lipid ratio (w/w) of 2.
In some embodiments, step (c) is performed using an LCP crystallization robot. The crystallization screening solution used in step (c) may be any known crystallization screening solution suitable for producing macromolecular crystals in the art. In some embodiments, the crystallization screening solution may comprise a buffer, a salt, and a precipitating agent.
In some embodiments, the buffer used in the crystallization screening solution may include sodium malonate, potassium malonate, sodium phosphate, potassium phosphate, sodium acetate, sodium citrate, or sodium succinate. In some embodiments, the buffer used in the crystallization screening solution may include sodium citrate. In some embodiments, the buffer used in the crystallization screening solution may include sodium citrate at a pH of less than about 7.0. In some embodiments, the buffer used in the crystallization screening solution may include sodium citrate at a concentration of about 0.05M to about 0.2M, about 0.06M to about 0.2M, about 0.07M to about 0.2M, about 0.08M to about 0.2M, about 0.09M to about 0.2M, about 0.1M to about 0.2M, about 0.05M to about 0.19M, about 0.05M to about 0.18M, about 0.05M to about 0.17M, about 0.05M to about 0.16M, about 0.05M to about 0.15M, about 0.05M to about 0.14M, about 0.05M to about 0.13M, about 0.05M to about 0.12M, about 0.05M to about 0.11M, or about 0.05M to about 0.1M. In some embodiments, the buffer used in the crystallization screening solution may include sodium citrate at a concentration of about 0.1M.
In some embodiments, the buffer used in the crystallization screening solution can have a pH of about 3.0 to about 6.6, about 3.0 to about 6.4, about 3.0 to about 6.2, about 3.0 to about 6.0, about 3.0 to about 5.8, about 3.0 to about 5.6, about 3.0 to about 5.4, about 3.0 to about 5.2, about 3.0 to about 5.0, about 3.0 to about 4.8, about 3.0 to about 4.6, about 3.0 to about 4.4, about 3.0 to about 4.2, about 3.0 to about 4.0, about 3.0 to about 3.8, about 3.0 to about 3.6, about 3.0 to about 3.4, about 3.0 to about 3.2. In some embodiments, the buffer used in the crystallization screening solution may have a pH of about 5.2.
In some embodiments, the salts used in the crystallization screening solution may include, but are not limited to, ammonium chloride, sodium chloride, potassium chloride, ammonium sulfate, sodium sulfate, potassium sulfate, and the like. In some embodiments, the precipitating agent used in the crystallization screening solution may include ammonium chloride. In some embodiments, the precipitating agent used in the crystallization screening solution may include ammonium chloride at a concentration of about 0.05M to about 0.2M, about 0.06M to about 0.2M, about 0.07M to about 0.2M, about 0.08M to about 0.2M, about 0.09M to about 0.2M, about 0.1M to about 0.2M, about 0.05M to about 0.19M, about 0.05M to about 0.18M, about 0.05M to about 0.17M, about 0.05M to about 0.16M, about 0.05M to about 0.15M, about 0.05M to about 0.14M, about 0.05M to about 0.13M, about 0.05M to about 0.12M, about 0.05M to about 0.11M, or about 0.05M to about 0.1M. In some embodiments, the precipitating agent used in the crystallization screening solution may include ammonium chloride at a concentration of about 0.1M.
In some embodiments, the precipitating agent used in the crystallization screening solution may include, but is not limited to, PEG200, PEG300, PEG400, PEG500MME (monomethyl ether polyethylene glycol 500), PEG550, PEG600MME, PEG1000, PEG2000, PEG3000. In some embodiments, the precipitating agent used in the crystallization screening solution may comprise PEG500MME. In some embodiments, the precipitating agent used in the crystallization screening solution may comprise PEG500MME. In some embodiments, the crystallization screening solution may further comprise PEG500MME at a concentration of about 20% to about 50%, about 25% to about 50%, about 30% to about 50%, about 35% to about 50%, about 40% to about 50%, about 20% to about 45%, about 20% to about 40%, about 20% to about 35%, about 20% to about 30%. In some embodiments, the crystallization screening solution can further comprise PEG500MME at a concentration of 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 40%, etc. In some embodiments, the precipitating agent used in the crystallization screening solution may include PEG500MME at a concentration of about 28%.
In some embodiments, the crystallization screening solution can comprise sodium citrate, ammonium chloride, and PEG500MME. In some embodiments, the crystallization screening solution comprises about 0.1M sodium citrate (pH of about 5.2), 0.1M ammonium chloride, and PEG500MME at a concentration of about 28%.
In some embodiments, the crystallizing in step (d) is performed under standard crystallization conditions. For example, crystallization is carried out at a temperature of about 0 ℃ to about 30 ℃. In some embodiments, the crystallization is performed at a temperature of about 10 ℃ to about 25 ℃, for example at a temperature of about 20 ℃.
In some embodiments, the crystallizing in step (d) is performed at 20 ℃ with a crystallization screen solution comprising about 0.1M sodium citrate (pH 5.2), 0.1M ammonium chloride, and about 28% PEG500MME.
X-ray crystal structure coordinates
The three-dimensional structure of the binding pocket of a PfHT polypeptide or PfHT analog thereof allows the design and identification of compounds that bind to the binding pocket and modulate PfHT-related activity. The three-dimensional structure of the binding pocket of PfHT or an analog thereof also provides a means for studying the mechanism of PfHT action and for identifying inhibitors of its function. For example, knowledge of the three-dimensional structure of the binding pocket of PfHT or an analog thereof allows one to design molecules (preferably agents) capable of binding thereto, including molecules that are thereby capable of inhibiting the interaction of PfHT or an analog thereof with its natural binding partner, thereby blocking glucose transport in plasmodium.
The structural coordinates of the present disclosure enable one to use a variety of molecular design and analysis techniques in order to (i) resolve the three-dimensional structure of the molecule of interest (preferably a molecular complex); and (ii) designing, selecting, and synthesizing a chemical entity that is capable of favorably associating or interacting with the binding pocket of PfHT or an analog thereof, wherein the chemical entity will preferably inhibit PfHT function, including blocking glucose transport in plasmodium.
In another aspect, the present disclosure provides a set of X-ray crystal structure coordinates of at least one allosteric binding pocket of a PfHT polypeptide or analog thereof, having the amino acid sequence of SEQ ID No. 1, the analog having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID No. 1,
wherein the allosteric binding pocket is inducible when compound 3361 is complexed with a PfHT polypeptide or analog thereof, and
wherein compound 3361 has the formula:
Figure BDA0003863898460000181
in some embodiments, the PfHT analogs used herein are selected from PvHT, poHT, pmHT, and PkHT. In some embodiments, the amino acid sequence of the PfHT analog is selected from the group consisting of SEQ ID NO 2, SEQ ID NO 3, SEQ ID NO 4, and SEQ ID NO 5.
In some embodiments, the at least one allosteric binding pocket comprises:
a) An R1 binding pocket comprising at least one or more (e.g., two, three, four, five, six, seven) amino acid residues selected from the equivalent residues in Q169, Q305, Q306, N311, N341, W412, and N435 of SEQ ID No. 1, or analogs thereof;
b) An R2 binding pocket comprising at least one or more (e.g., two, three, four, five, six, seven, eight, nine) amino acid residues selected from the equivalent residues of V44, L47, F85, W436, and V443 of SEQ ID NO:1 or analogs thereof;
c) An R3 binding pocket comprising at least one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen) amino acid residues selected from equivalent residues in N48, K51, N52, N311, N316, N318, E319, and D447, or analogs thereof, of SEQ ID NO: 1; or
d) Any combination thereof.
In some embodiments, the R1 binding pocket further comprises one or more (e.g., two, three, four, five, six) additional amino acid residues selected from equivalent residues in F40, I172, I176, I310, F403, and a404 of SEQ ID NO:1, or analogs thereof.
In some embodiments, the R1 binding pocket further comprises one or more (e.g., two, three, four) additional amino acid residues selected from equivalent residues in T145, T173, V314, and I400 of SEQ ID No. 1, or analogs thereof.
In some embodiments, the R2 binding pocket further comprises one or more (e.g., two, three, four) additional amino acid residues selected from the group consisting of equivalent residues in L81, V312, a439, and F444 of SEQ ID No. 1, or analogs thereof.
In some embodiments, the R3 binding pocket further comprises one or more (e.g., two, three, four, five) additional amino acid residues selected from the group consisting of F85, V312, S315, V443, and F444 of SEQ ID NO:1 or an equivalent residue in an analog thereof.
In some embodiments, the set of X-ray crystal structure coordinates is shown in fig. 2. FIG. 2 shows the X-ray crystal structure coordinates of a PfHT complex comprising compound 3361 and a PfHT polypeptide having the amino acid sequence of SEQ ID NO: 1. In the present disclosure, when referring to fig. 1, these X-ray crystal structure coordinates may refer to a portion of fig. 1 (e.g., the structure coordinates of the amino acid residue portion of fig. 1, or the structure coordinates of the compound portion of fig. 1) or the entire fig. 1.
One skilled in the art will appreciate that the set of structural coordinates of a protein or protein-inhibitor complex or a portion of the complex is a set of relative points that define a three-dimensional shape. Thus, a set of disparate coordinates can define similar or identical shapes and are therefore within the scope of the present invention. Further, a slight change in the individual coordinates does not greatly affect the overall shape. In the case of binding pockets, it is expected that these changes will not significantly alter the properties of the compounds capable of associating with those binding pockets.
It is also noteworthy that modifications in the crystal structure due to mutations, additions, substitutions and/or deletions of amino acids, or other changes in any of the components making up the crystal, may also account for changes in the structural coordinates. If such changes are within acceptable error criteria, such as the root mean square deviation (rmsd) of the conserved backbone atoms comprising the binding pocket as compared to the original coordinates, is no more than about
Figure BDA0003863898460000182
The resulting three-dimensional shape is considered to be the same. In some embodiments, the root mean square deviation is no more than about
Figure BDA0003863898460000183
In some embodiments, the root mean square deviation is no more than about
Figure BDA0003863898460000184
In some embodiments, the root mean square deviation is no more than about
Figure BDA0003863898460000185
Thus, for example, a compound that binds to a binding pocket of PfHT or an analog thereof as described herein is also expected to bind to other binding pockets whose structural coordinates define a shape that falls within acceptable tolerances. As used herein, the term "root mean square deviation" refers to the square root of the arithmetic mean of the squares of the deviations from the mean. In the context of atomic objects, the numerical values are in angstroms
Figure BDA0003863898460000186
It is given.
The above modifications can be made due to the mathematical transformation of the PfHT structure coordinates. For example, the structure coordinates shown in FIG. 2 may be transformed by crystal arrangement of the original structure coordinates, by fractioning the original structure coordinates, by integer addition or subtraction of sets of original structure coordinates, by inversion of the original structure coordinates, or any combination of the above. For example, the PfHT polypeptides and PfHT analogs of the present disclosure preferably comprise a binding pocket characterized by the amino acid residues as shown in fig. 2 ± the rms deviation from the conserved backbone atoms of the amino acids that does not exceed
Figure BDA0003863898460000191
(or more preferably not more than
Figure BDA0003863898460000192
Or more preferably not more than
Figure BDA0003863898460000193
And most preferably not more than
Figure BDA0003863898460000194
)。
Thus, the present disclosure also provides X-ray crystal structure coordinates comprising at least one allosteric binding pocket as provided herein ± the rms deviation from the backbone atoms of the amino acids making up the binding pocket that does not exceed about
Figure BDA0003863898460000195
Not more than about
Figure BDA0003863898460000196
Not more than about
Figure BDA0003863898460000197
Or not more than about
Figure BDA0003863898460000198
In some embodiments, the present disclosure provides a set of X-ray crystal structure coordinates comprising at least one allosteric binding pocket as shown in figure 2 ± the rms deviation of conserved backbone atoms from the amino acids making up the binding pocket that does not exceed about
Figure BDA0003863898460000199
Not more than about
Figure BDA00038638984600001910
Not more than about
Figure BDA00038638984600001911
Or not more than about
Figure BDA00038638984600001912
Computer readable storage medium
It should be noted that in order to use the structural coordinates generated from the PfHT complexes described herein, it may be necessary to display or convert the relevant coordinates as, or otherwise transform, a three-dimensional shape or graphical representation. Typically, such three-dimensional representations of structural coordinates will be used for rational drug design, molecular replacement analysis, homology modeling, and mutation analysis. This is typically accomplished using any of a variety of commercially available software programs capable of generating a three-dimensional graphical representation of a molecule or portion thereof from a set of structural coordinates. The scientific arts have many conventional software programs, which are incorporated by reference herein in their entirety. For example, refer to GRID (Oxford University, oxford, UK) for Oxford; AUTODOCK (Scripps Research Institute, la Jolla, calif.); flo99 (Thistlesoft, morisiston, N.J.) and the like.
Thus, in another aspect, the present disclosure provides a computer-readable storage medium having stored thereon crystal structure coordinates according to the present disclosure, such as shown in fig. 2.
To store, transfer, and use such programs, machines, such as computers, that generate three-dimensional representations of the binding pockets of PfHT and the like are also contemplated. The machine will include a machine readable data storage medium having stored thereon crystal structure coordinates according to the present disclosure. Computer-readable storage media are well known to those skilled in the art and include, for example, hard disks, CD-ROMs, floppy disks, DVDs, thumb drives, etc., as well as other magnetic, magneto-optical, floppy disks and other media that may be suitable for use with a computer. The machine also includes a working memory for storing instructions for processing machine-readable data, and a Central Processing Unit (CPU) coupled to the working memory and the machine-readable data storage medium for processing the machine-readable data into a desired three-dimensional representation. Also, the machine of the present invention further includes a display connected to the CPU so that the three-dimensional representation is visible to the user. Thus, when used with a machine programmed with instructions to use the data (e.g., a computer loaded with one or more programs of the types described above), the machine provided herein is capable of displaying a graphical three-dimensional representation of a PfHT complex described herein.
Methods of assessing or predicting the binding characteristics of a compound to a PfHT polypeptide or analog thereof
In another aspect, the present disclosure provides a method of evaluating or predicting the binding characteristics of a compound to a PfHT polypeptide having an amino acid sequence of SEQ ID NO:1 or an analog thereof having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID NO:1, the method comprising the steps of:
(a) Generating, on a computer, a representation of the three-dimensional structure of at least one allosteric binding pocket based on an X-ray crystal structure coordinate set according to the present disclosure;
(b) Generating a representation of the compound on a computer; and
(c) Fitting the representation of the compound according to step (b) with the representation of the three-dimensional structure of the at least one allosteric binding pocket according to step (a) to determine the probability that the compound binds to the at least one allosteric binding pocket.
In some embodiments, the PfHT analogs used herein are selected from PvHT, poHT, pmHT, and PkHT. In some embodiments, the amino acid sequence of the PfHT analog is selected from the group consisting of SEQ ID NO 2, SEQ ID NO 3, SEQ ID NO 4, and SEQ ID NO 5.
The three-dimensional structure of the at least one allosteric binding pocket and the compound of the present disclosure (i.e., steps (a) and (b) above) can be determined using X-ray crystallography and various computer modeling techniques.
In step (a) provided herein, the representation of the three-dimensional structure of the at least one allosteric binding pocket may be a graphical representation or a map of the coordinates of the amino acid residues in three-dimensional space. This can be achieved by commercially available software. The three-dimensional structure may be used to perform computer modeling, adaptation operations, or display as a three-dimensional graphical representation.
Software for generating a three-dimensional graphical representation of at least one allosteric binding pocket and compounds of the present disclosure is known and commercially available. Examples include Quanta and WebLite Viewer,
Figure BDA00038638984600001913
Suite, AUTODOCK, DOCK, and the like.
The adaptation in step (c) above is a process for assessing complementarity between the representation of the compound according to step (b) and the representation of the three-dimensional structure of the at least one allosteric binding pocket according to step (a). Fitting (or docking) may be performed by methods well known in the art, for example by various computational techniques that evaluate "fit" between a binding pocket and a candidate compound, such as ultra-precision slide docking (Glide XP).
The probability that a compound binds to the at least one allosteric binding pocket can be expressed as the degree of association between the candidate compound and the at least one allosteric binding pocket. In some embodiments, the degree of association can be determined experimentally using standard binding assays. In some embodiments, the degree of association may be determined and ranked by calculation by any number of commercially available software programs (such as Glide XP score, emodel score, etc.). Without being bound by any theory, it is contemplated that a higher degree of association may indicate a higher probability of the compound binding to the at least one allosteric binding pocket. Compounds that are determined to "fit" to a binding pocket as defined herein by some type of association or binding may also block the biological activity of PfHT or an analog thereof, and thus represent potential drug candidates.
Methods for identifying, virtually screening, and designing potential PfHT inhibitors
In another aspect, the present disclosure provides a method of identifying a compound as a potential PfHT inhibitor, the method comprising the steps of:
(a) Generating, on a computer, a representation of the three-dimensional structure of the at least one allosteric binding pocket based on the set of X-ray crystal structure coordinates according to the present disclosure;
(b) Generating a representation of the compound on a computer;
(c) Adapting the representation of the compound according to step (b) to a computer representation of the three-dimensional structure of the at least one allosteric binding pocket according to step (a) to provide an energy minimized configuration of the compound in the at least one allosteric binding pocket; and
(d) Evaluating the results of step (c) to quantify the binding between the compound and the at least one allosteric binding pocket,
wherein the compound is identified as a potential PfHT inhibitor when the compound binds to the at least one allosteric binding pocket to produce a low energy, stable complex.
In another aspect, the present disclosure provides a virtual screening method for identifying potential PfHT inhibitors, the method comprising the steps of:
(a) Generating or accessing on a computer a representation of the three-dimensional structure of the at least one allosteric binding pocket based on an X-ray crystal structure coordinate set according to the present disclosure;
(b) Generating or accessing on a computer a representation of a candidate compound from a library of compounds;
(c) Adapting the representation of the candidate compound according to step (b) to the representation of the three-dimensional structure of the at least one allosteric binding pocket according to step (a) to provide an energy minimized conformation of the candidate compound in the at least one allosteric binding pocket; and
(d) Evaluating the results of step (c) to quantify the binding between the candidate compound and the at least one allosteric binding pocket,
(e) The quantitative binding is compared to a predetermined threshold,
wherein the candidate compound is identified as a potential PfHT inhibitor based on the comparison of step (e).
The comparing step in step (e) above means comparing the quantitative binding of step (d) with a predetermined threshold. The comparison may be performed manually or computer-assisted. For computer-aided comparison, the value of the quantitative binding may be compared by a computer program with predetermined thresholds stored in a database, and the computer program may further evaluate the result of the comparison and automatically provide the desired assessment in a suitable output format. Based on this assessment (e.g., comparing the value of the quantitative binding to a predetermined threshold), one skilled in the art can readily identify potential PfHT inhibitors. According to the computer program for comparison, a value of quantitative binding that is higher or lower than a predetermined threshold may be indicative of a potential PfHT inhibitor. In some embodiments, the predetermined threshold is the Glide XP score, which depends on the hydrophobic shell, hydrogen bonding interactions, internal energy (such as van der waals interactions), electrostatic interactions, and two XP penalties (penalties) (i.e., a desolvation penalty and a ligand-strain penalty). In some embodiments, the predetermined threshold is a Glide XP score of-10, whereas the reference Glide XP score is-12.
In another aspect, the invention provides a method of designing a compound capable of binding to a PfHT polypeptide having the amino acid sequence of SEQ ID No. 1 or an analog thereof having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID No. 1, the method comprising:
(a) Generating, on a computer, a representation of the three-dimensional structure of the at least one allosteric binding pocket based on the set of X-ray crystal structure coordinates according to the present disclosure;
(b) Generating on a computer a representation of a candidate compound;
(c) Fitting the representation of the candidate compound according to step (b) with the representation of the three-dimensional structure of the at least one allosteric binding pocket according to step (a) to provide an energy-minimized configuration of the candidate compound in the at least one allosteric binding pocket, wherein the compound is identified as a potential PfHT inhibitor when the compound binds to the at least one allosteric binding pocket to produce a low-energy, stable complex.
Optionally, in some embodiments, the method further comprises step (d): modifying the candidate compound based on the results obtained in step (c). Optionally, in some embodiments, the method further comprises step (e): repeating steps (b) to (c) with the modified candidate compound obtained in step (d).
Assays can be performed and the results analyzed to determine whether a compound is an inhibitor (i.e., a compound that reduces or prevents the binding affinity between PfHT and its natural binding partner) or has no effect on the interaction between PfHT and its natural binding partner. Compounds identified using the foregoing methods, preferably PfHT inhibitors, can then be tested as therapeutic agents for the treatment and/or prevention of malaria.
The efficiency of binding of selected compounds to the binding pocket of the present disclosure can be tested and optimized by computational evaluation. The quality of fit of a given compound within a PfHT-binding pocket can be assessed, for example, by shape, size, and electrostatic complementarity, as determined qualitatively by visual inspection or quantitatively by using scoring functions such as LUDI, PLP, PMF, SCORE, GOLD, flexX, emodel SCORE, and Glide XP SCORE. These methods of qualitative and quantitative evaluation may be used alone or in combination (e.g., in a consistent scoring manner).
Alternatively, the binding efficiency can be determined based on the interaction energy of the complex formed by the compound binding or associating with PfHT or an analog thereof. For example, compounds that form "low energy, stable complexes" with PfHT or analogs thereof in the manner described herein need further analysis as potential PfHT inhibitors. As used herein, the term "low energy, stable complex" refers to a PfHT complex wherein the interaction energy between the compound, including the hydrophobic shell, hydrogen bonding interactions, internal energy (such as van der waals interactions), and PfHT or an analog thereof, is less than a predetermined value, which may be set by a computer program. Interaction can be determined using software known in the art, for example, ultra-precision sliding docking of Glide program (Glide XP).
The effect of the compound identified by the adaptation step (c) on PfHT activity can be further calculated or experimentally evaluated by competitive binding assays or by contacting the identified compound with PfHT or an analog thereof and measuring the effect of the compound on the target biological activity. Standard enzymatic assays can be performed and the results analyzed to determine whether the agent is an inhibitor of PfHT activity (e.g., prevents glucose transport).
Once the PfHT inhibitor is optimally identified, screened or designed as described above, it may then be substituted on certain atoms or side groups thereof to improve or alter its selectivity and binding properties-i.e., its affinity for the binding pocket disclosed herein. Typically, the initial substitution is conservative, i.e., the substituted group will have approximately the same size, shape, hydrophobicity, and charge as the original group. The efficiency of fit of such substituted compounds to the binding pocket of PfHT can then be analyzed by the same in silico methods as detailed above.
Various molecular analysis and rational drug design techniques are further disclosed in U.S. Pat. Nos. 5,834,228, 5,939,528 and 5,865,116, and PCT application PCT/US98/16879, published as WO 99/09148, the contents of which are hereby incorporated by reference.
Compound (I)
In another aspect, the present disclosure provides a compound having formula (I):
A-B-L-D-E (I)
or a pharmaceutically acceptable salt thereof, wherein
A is a hexose moiety linked to B through its atom selected from carbon, nitrogen, oxygen or sulfur;
b is absent, or is selected from-CH 2 C(O)O-、-CH 2 -C (O) NH-and-C (O) -;
l is- (CH) 2 ) m -、-(CH 2 OCH 2 ) q Or- (CH) 2 ) n -W-(CH 2 ) p <xnotran> -, -W- , -O-, -S-, -NH-, -C = C-, -C (O) O- -C (O) NH-, m 1 12 , n, p q 1 3 ; </xnotran>
D is absent, or is selected from-O-, -S-, and-NH-;
e is selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein the cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more R groups;
r is selected from halogen, oxo, alkyl, haloalkyl, -OR 1 and-NR 2 R 3
R 1 Selected from the group consisting of hydrogen, alkyl, aryl, alkylaryl, and alkylalkoxy;
R 2 and R 3 Each selected from the group consisting of hydrogen, alkyl, cycloalkyl and aryl, wherein the alkyl, cycloalkyl and aryl are optionally substituted with one or more alkoxy groups.
In some embodiments, a is selected from the group consisting of D-glucose, L-glucose, D-galactose, L-galactose, D-mannose, D-xylose, D-fructose, 2-deoxy-D-glucose, and 2-deoxy-2-halo-D-glucose.
In some embodiments, a is a D-glucopyranose moiety.
In some embodiments, A is a D-glucopyranose moiety.
In some embodiments, a is a D-glucose moiety attached to B through its oxygen atom.
In some embodiments, a is a D-glucose moiety attached to B through its nitrogen atom.
In some embodiments, a is a D-glucose moiety attached to B through its sulfur atom.
In some embodiments, a is a D-glucose moiety attached to B through its carbon atom.
In some embodiments, A is a D-glucose moiety attached to B through its oxygen atom at a position selected from the group consisting of 1-position, 2-position, 3-position, 4-position, and 6-position.
In some embodiments, a is a D-glucose moiety attached to B through an oxygen atom at its 2-position.
In some embodiments, a is a D-glucose moiety attached to B through the oxygen atom at its 3-position.
In some embodiments, a is a D-glucose moiety attached to B through an oxygen atom at its 6-position.
In some embodiments, a is an L-glucose moiety. In some embodiments, A is an L-glucopyranose moiety.
In some embodiments, A is an L-glucose moiety attached to B through its oxygen atom at a position selected from the group consisting of 1-position, 2-position, 3-position, 4-position, and 6-position.
In some embodiments, A is an L-glucose moiety attached to B through an oxygen atom at its 3-position.
In some embodiments, a is a deoxy-D-glucose moiety. In some embodiments, the deoxy-D-glucose moiety is a deoxy-D-glucopyranose moiety. In some embodiments, the deoxy-D-glucose moiety is a deoxy-D-glucofuranose moiety.
In some embodiments, a is a 2-deoxy-D-glucose moiety. In some embodiments, a is a 2-deoxy-D-glucopyranose moiety. In some embodiments, a is a 2-deoxy-D-glucofuranose moiety.
In some embodiments, a is a 2-deoxy-D-glucopyranose moiety attached to B through an oxygen atom thereof at a position selected from the group consisting of 1-position, 3-position, 4-position, and 6-position.
In some embodiments, a is a 2-deoxy-D-glucopyranose moiety attached to B through the oxygen atom at its 3-position.
In some embodiments, a is a D-fructose moiety. In some embodiments, a is a D-fructopyranose moiety. In some embodiments, a is a D-fructofuranose moiety.
In some embodiments, A is a D-fructopyranose moiety covalently attached to B through its oxygen atom at a position selected from the group consisting of 1-position, 2-position, 3-position, 4-position and 5-position. In some embodiments, A is a D-fructopyranose moiety covalently attached to B through the oxygen atom in the 1-position thereof.
In some embodiments, B is-CH 2 C(O)O-。
In some embodiments, B is absent.
In some embodiments, B is-CH 2 -C (O) NH-or-C (O) -.
In some embodiments, L is- (CH) 2 ) m -。
In some embodiments, L is- (CH) 2 ) n O(CH 2 ) p -。
In some embodiments, B is-CH 2 C (O) O-and L is- (CH) 2 ) m -or- (CH) 2 ) n O(CH 2 ) p -。
In some embodiments, B is absent and L is- (CH) 2 ) m -。
In some embodiments or wherein B is-CH 2 -C (O) NH-or-C (O) -, and L is- (CH) 2 ) m -。
In some embodiments, D is-O-.
In some embodiments, D is absent.
In some embodiments, D is-NH-.
In some embodiments, B is absent or is-CH 2 C (O) O-or-CH 2 -C (O) NH-, and D is-O-.
In some embodiments, B is absent or is-CH 2 C (O) O-or-C (O) -, and D is absent.
In some embodiments, B is-CH 2 C (O) O-, D is absent or is-O-.
In some embodiments, B is absent and D is absent or is-O-or-NH-.
In some embodiments, B is-C (O) -, and D is absent.
In some embodiments, B is-CH 2 -C (O) NH-and D is-O-.
In some embodiments, B is-CH 2 C (O) O-, L is- (CH) 2 ) m -, and D is absent or is-O-.
In some embodiments, B is absent and L is- (CH) 2 ) m -, and D is absent or is-O-or-NH-.
In some embodiments, B is absent and L is- (CH) 2 ) m -, and D is-O-.
In some embodiments, E is aryl optionally substituted with one or more R groups.
In some embodiments, E is an aryl group selected from:
Figure BDA0003863898460000221
each of which is optionally substituted with one or more R groups.
In certain embodiments, the R group is-OR 1 or-NR 2 R 3
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is an alkyl radical。
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is C 1 -C 6 Alkyl radical, C 1 -C 5 Alkyl radical, C 1 -C 4 Alkyl or C 1 -C 3 An alkyl group.
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is methyl.
In certain embodiments, the R group is-NR 2 R 3 Wherein R is 2 And R 3 Each is hydrogen.
In some embodiments, E is heteroaryl optionally substituted with one or more R groups.
In some embodiments, E is a heteroaryl selected from:
Figure BDA0003863898460000222
each of which is optionally substituted with one or more R groups.
In certain embodiments, the R group is halogen, oxo, alkyl, haloalkyl, -OR 1 and-NR 2 R 3
In certain embodiments, the R group is halogen.
In certain embodiments, the R group is Cl or Br.
In certain embodiments, the R group is oxo.
In certain embodiments, E is
Figure BDA0003863898460000231
R is oxo and/or halogen.
In some embodiments, the R group is alkyl.
In certain embodiments, the R group is C 1 -C 6 Alkyl radical, C 1 -C 5 Alkyl radical, C 1 -C 4 Alkyl or C 1 -C 3 An alkyl group.
In certain embodiments, the R group is methyl.
In some embodiments, the R group is haloalkyl.
In certain embodiments, the R group is C 1 -C 6 Haloalkyl, C 1 -C 5 Haloalkyl, C 1 -C 4 Haloalkyl or C 1 -C 3 A haloalkyl group.
In certain embodiments, the R group is-CF 3
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is hydrogen, alkyl, aryl, alkylalkoxy or alkylaryl.
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is hydrogen.
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is an alkyl group.
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is C 1 -C 6 Alkyl radical, C 1 -C 5 Alkyl radical, C 1 -C 4 Alkyl or C 1 -C 3 An alkyl group.
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is methyl, ethyl or isopropyl.
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is an aryl group.
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is C 3 -C 8 Aryl radical, C 3 -C 7 Aryl radical, C 3 -C 6 Aryl or C 3 -C 5 And (3) an aryl group.
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is phenyl.
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is an alkylalkoxy group.
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is (C) 1 -C 6 Alkyl) (C 1 -C 6 Alkoxy radicalBase), (C) 1 -C 5 Alkyl) (C 1 -C 5 Alkoxy group), (C) 1 -C 4 Alkyl) (C 1 -C 4 Alkoxy) or (C) 1 -C 3 Alkyl) (C) 1 -C 3 Alkoxy groups).
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is an ethyl ethoxy group.
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is an alkylaryl group.
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is (C) 1 -C 6 Alkyl) (C 3 -C 8 Aryl group), (C) 1 -C 5 Alkyl) (C) 3 -C 7 Aryl group), (C) 1 -C 4 Alkyl) (C 3 -C 6 Aryl) or (C) 1 -C 3 Alkyl) (C 3 -C 5 Aryl).
In certain embodiments, the R group is-OR 1 Wherein R is 1 Is benzyl.
In certain embodiments, the R group is-NR 2 R 3 And R is 2 And R 3 Each independently is hydrogen, alkyl, aryl, or cycloalkyl, wherein aryl and cycloalkyl are optionally substituted with one or more alkoxy groups.
In certain embodiments, the R group is-NR 2 R 3 ,R 2 And R 3 One of them is hydrogen and the other is C 3 -C 8 Aryl radical, C 3 -C 7 Aryl radical, C 3 -C 6 Aryl or C 3 -C 5 Aryl radicals bound by one or more C 1 -C 6 Alkoxy radical, C 1 -C 5 Alkoxy radical, C 1 -C 4 Alkoxy or C 1 -C 3 Alkoxy is optionally substituted.
In certain embodiments, the R group is-NR 2 R 3 ,R 2 And R 3 One of which is hydrogen and the other is phenyl optionally substituted with one or more methoxy groups.
At a certain pointIn some embodiments, the R group is-NR 2 R 3 ,R 2 And R 3 One of them is hydrogen and the other is C 3 -C 8 Cycloalkyl radical, C 3 -C 7 Cycloalkyl radical, C 3 -C 6 Cycloalkyl or C 3 -C 5 A cycloalkyl group.
In certain embodiments, the R group is-NR 2 R 3 ,R 2 And R 3 One of which is hydrogen and the other is cyclohexyl.
In certain embodiments, the R group is-NR 2 R 3 And R is 2 And R 3 Both are hydrogen.
In certain embodiments, the R group is-NR 2 R 3 And R is 2 And R 3 Both of which are alkyl groups. In certain embodiments, the R group is-NR 2 R 3 And R is 2 And R 3 Both are C 1 -C 6 Alkyl radical, C 1 -C 5 Alkyl radical, C 1 -C 4 Alkyl or C 1 -C 3 An alkyl group. In certain embodiments, the R group is-NR 2 R 3 And R is 2 And R 3 Both are methyl groups.
In some embodiments, the compounds of the present disclosure have the following formula (II):
Figure BDA0003863898460000232
wherein L and E are as defined above.
In some embodiments, L is- (CH) 2 ) m -, m is an integer from 1 to 12, from 2 to 10 or from 3 to 8.
In some embodiments, E is aryl or heteroaryl, each of which is optionally substituted with one or more R groups.
In some embodiments, E is aryl optionally substituted with one OR more R groups, wherein R group is-OR 1
In certain embodiments, E is
Figure BDA0003863898460000241
Each of which is optionally substituted with one OR more R groups, wherein the R groups are-OR 1 . In certain embodiments, R 1 Is an alkyl group. In certain embodiments, R 1 Is a methyl group.
In some embodiments, E is heteroaryl optionally substituted with one OR more R groups, wherein the R groups are halogen OR-OR 1
In certain embodiments, E is selected from:
Figure BDA0003863898460000242
each of which is optionally substituted with one OR more R groups, wherein the R groups are halogen OR-OR 1
In certain embodiments, the R group is halogen. In certain embodiments, the R group is Cl or Br.
In certain embodiments, the R group is-OR 1 . In certain embodiments, R 1 Is an alkyl group. In certain embodiments, R 1 Is methyl.
In some embodiments, the compounds of the present disclosure have a formula selected from:
Figure BDA0003863898460000243
Figure BDA0003863898460000251
wherein Z is hydrogen or halogen, L and E are as defined above.
In some embodiments, L is- (CH) 2 ) m -, m is an integer from 1 to 12, 4 to 12, 6 to 12 or 8 to 12.
In some embodiments, E is aryl or heteroaryl, each of which is optionally substituted with one or more R groups.
In some embodiments, E is aryl optionally substituted with one OR more R groups, wherein the R group is-OR 1 or-NR 2 R 3
In certain embodiments, E is an aryl group selected from:
Figure BDA0003863898460000252
each of which is optionally substituted with one OR more R groups, wherein the R groups are-OR 1 or-NR 2 R 3
In certain embodiments, the R group is-OR 1 And R is 1 Is an alkyl group. In certain embodiments, the R group is-OR 1 And R is 1 Is methyl.
In certain embodiments, the R group is-NR 2 R 3 . In certain embodiments, the R group is-NR 2 R 3 And R is 2 And R 3 Are all hydrogen.
In some embodiments, E is heteroaryl optionally substituted with one OR more R groups, wherein R groups are halo, alkyl, -OR 1 or-NR 2 R 3
In certain embodiments, E is a heteroaryl selected from:
Figure BDA0003863898460000253
Figure BDA0003863898460000261
each of which is optionally substituted with one OR more R groups, wherein R groups are halogen, alkyl, haloalkyl, -OR 1 or-NR 2 R 3
In certain embodiments, the R group is halogen. In certain embodiments, the R group is Cl or Br.
In certain embodiments, the R group is an alkyl group. In certain embodiments, the R group is C 1 -C 3 An alkyl group. In certain embodiments, the R group is methyl.
In some embodiments, the R group is haloalkyl.
In certain embodiments, the R group is C 1 -C 6 Haloalkyl, C 1 -C 5 Haloalkyl, C 1 -C 4 Haloalkyl or C 1 -C 3 A haloalkyl group.
In certain embodiments, the R group is-CF 3
In certain embodiments, the R group is-OR 1 And R is 1 Is hydrogen, alkyl, aryl, alkylalkoxy or alkylaryl. In certain embodiments, the R group is-OR 1 And R is 1 Is hydrogen. In certain embodiments, the R group is-OR 1 Wherein R is 1 Is an alkyl group. In certain embodiments, the R group is-OR 1 Wherein R is 1 Is methyl, ethyl or isopropyl. In certain embodiments, the R group is-OR 1 Wherein R is 1 Is an aryl group. In certain embodiments, the R group is-OR 1 Wherein R is 1 Is phenyl. In certain embodiments, the R group is-OR 1 Wherein R is 1 Is an alkylalkoxy group. In certain embodiments, the R group is-OR 1 Wherein R is 1 Is an ethyl ethoxy group. In certain embodiments, the R group is-OR 1 Wherein R is 1 Is an alkylaryl group. In certain embodiments, the R group is-OR 1 Wherein R is 1 Is a benzyl group.
In certain embodiments, the R group is-NR 2 R 3 And R is 2 And R 3 Each is hydrogen, alkyl, aryl or cycloalkyl, wherein alkyl, aryl and cycloalkyl are optionally substituted with one or more alkoxy groups. In certain embodiments, the R group is-NR 2 R 3 ,R 2 And R 3 One is hydrogen and the other is phenyl optionally substituted with one or more methoxy groups. In some implementationsIn the scheme, the R group is-NR 2 R 3 ,R 2 And R 3 One is hydrogen and the other is phenyl substituted with three methoxy groups. In certain embodiments, the R group is-NR 2 R 3 ,R 2 And R 3 One of which is hydrogen and the other is cyclohexyl. In certain embodiments, the R group is-NR 2 R 3 And R is 2 And R 3 Both are hydrogen. In certain embodiments, the R group is-NR 2 R 3 And R is 2 And R 3 Both of which are alkyl groups. In certain embodiments, the R group is-NR 2 R 3 And R is 2 And R 3 Both are methyl groups.
In another aspect, the present disclosure provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, selected from:
3- (naphthalen-2-yloxy) propyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (naphthalen-2-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
7- (naphthalen-2-yloxy) heptyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
2- (2- (naphthalen-2-yloxy) ethoxy) ethyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
n- (5- (naphthalen-2-yloxy) pentyl) -2- (((3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetamide;
5- (quinolin-6-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (naphthalen-1-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5-phenoxypentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (pyridin-4-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (benzo [ d ] [1,3] dioxol-5-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (3, 4, 5-trimethoxyphenoxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (3, 4-dimethoxyphenoxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
(3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl-2- (naphthalen-2-yl) acetate;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
5- (isoquinolin-6-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (isoquinolin-7-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (quinolin-7-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (quinolin-3-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (quinolin-8-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (quinolin-2-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (isoquinolin-3-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- ((2-chloroquinolin-6-yl) oxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- ((7-chloroquinolin-4-yl) oxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (7-chloro-4-oxoquinolin-1 (4H) -yl) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- ((7-methoxyquinolin-4-yl) oxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (7-methoxy-4-oxoquinolin-1 (4H) -yl) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
(3R, 4S,5R, 6R) -4- ((8- ((2-chloroquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8-phenoxyoctyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R,4S, 5R,6R) -6- (hydroxymethyl) -4- ((8- (naphthalen-2-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((2-bromoquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2-methylquinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2-methoxyquinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((2-ethoxyquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((2- (2-ethoxyethoxy) quinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2-isopropoxyquinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2-phenoxyquinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2- (phenylamino) quinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2- ((3, 4, 5-trimethoxyphenyl) amino) quinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((2- (cyclohexylamino) quinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (quinolin-4-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
1- (8- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) octyl) quinolin-4 (1H) -one;
(3R, 4S,5R, 6R) -4- ((8- ((7-chloroquinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
7-chloro-1- (8- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) octyl) quinolin-4 (1H) -one;
(3R, 4S,5R, 6R) -4- ((8- ((7-bromoquinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
7-bromo-1- (8- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) octyl) quinolin-4 (1H) -one;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((7-methoxyquinolin-4-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((7- (benzyloxy) quinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((7-aminoquinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((7-hydroxyquinolin-4-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (quinolin-6-ylamino) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((5-aminonaphthalen-2-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (quinolin-3-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((6- (quinolin-6-yloxy) hexyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((10- (quinolin-6-yloxy) decyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((12- (quinolin-6-yloxy) dodecyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3S,4R,5S,6S) -6- (hydroxymethyl) -4- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((7- (trifluoromethyl) quinolin-4-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5S, 6R) -6- (((8- (quinolin-6-yloxy) octyl) oxy) methyl) tetrahydro-2H-pyran-2, 3,4, 5-tetraol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((7-iodoquinolin-4-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((6-bromoquinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((8-bromoquinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((6, 7-dimethoxyquinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2-hydroxyquinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((3-bromoquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((7- (quinolin-6-yloxy) heptyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((9- (quinolin-6-yloxy) nonyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5S, 6R) -6- (hydroxymethyl) -3- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 4, 5-triol;
(2R, 3S,4R, 5R) -2- (((8- (quinolin-6-yloxy) octyl) oxy) methyl) tetrahydro-2H-pyran-2, 3,4, 5-tetraol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((6-methoxyquinolin-4-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ([ 1,1' -biphenyl ] -4-yloxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2-iodoquinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- (benzo [ d ] [1,3] dioxol-5-yloxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ([ 1,1' -biphenyl ] -3-yloxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (naphthalen-1-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- (benzofuran-5-yloxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- (benzo [ b ] thiophen-5-yloxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (quinolin-7-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
6- ((8- (((3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) octyl) oxy) -2H-chroman-2-one;
(3R,4S, 5R,6R) -4- ((8- (anthracen-2-yloxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R,4S, 5R,6R) -6- (hydroxymethyl) -4- ((8- (phenanthren-9-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((9H-carbazol-3-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (pyrene-1-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((11- (quinolin-6-yloxy) undecyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2- (trifluoromethyl) quinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((2-aminoquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((2- (dimethylamino) quinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(2R, 3S,4S, 5R) -2- (hydroxymethyl) -6- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-3, 4, 5-triol;
(3R, 4R,5S, 6R) -6- (hydroxymethyl) -5- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 4-triol;
(4R,5S,6R) -6- (hydroxymethyl) -4- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 5-diol;
(3S,4R,5S,6S) -4- ((9- ((2-chloroquinolin-6-yl) oxy) nonyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5S, 6R) -3- ((8- ((2-chloroquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 4, 5-triol; and
(3R, 4S,5S, 6R) -6- (hydroxymethyl) -3- ((9- (quinolin-6-yloxy) nonyl) oxy) tetrahydro-2H-pyran-2, 4, 5-triol.
Exemplary compounds of the present disclosure are listed in table 1 below.
TABLE 1
Figure BDA0003863898460000281
Figure BDA0003863898460000291
Figure BDA0003863898460000301
Figure BDA0003863898460000311
Figure BDA0003863898460000321
Figure BDA0003863898460000331
Figure BDA0003863898460000341
Figure BDA0003863898460000351
Figure BDA0003863898460000361
Figure BDA0003863898460000371
Figure BDA0003863898460000381
Figure BDA0003863898460000391
Figure BDA0003863898460000401
Figure BDA0003863898460000411
Figure BDA0003863898460000421
Figure BDA0003863898460000431
The compounds provided herein are described with reference to both the general formula and the specific compounds. Further, the compounds of the present disclosure may exist in a variety of different forms or derivatives, including but not limited to prodrugs, soft drugs, active metabolic derivatives (active metabolites), and their pharmaceutically acceptable salts, all of which are within the scope of the present disclosure.
As used herein, the term "prodrug" refers to a compound or a pharmaceutically acceptable salt thereof that when metabolized or converted by solvolysis under physiological conditions to yield the desired active compound. Prodrugs include, but are not limited to, esters, amides, carbamates, carbonates, ureides, solvates or hydrates of the active compound. Typically, a prodrug is inactive, or less active than the active compound, but provides one or more beneficial handling, administration, and/or metabolic properties. For example, some prodrugs are esters of the active compound; during metabolism, the ester group is cleaved to yield the active drug. In addition, some prodrugs are activated enzymatically to yield the active compound, or upon further chemical reaction to yield the compound of the active compound. A prodrug may evolve from a prodrug form to an active form in a single step, or may have one or more intermediate forms that may themselves be active or may be inactive. The preparation and use of prodrugs are discussed in the following documents: higuchi and v.stella, "Pro-drugs as Novel Delivery Systems", volume 14 of a.c.s.symposium Series, edited by Bioreversible Carriers in Drug Delivery ", edward b.roche, american Pharmaceutical Association and Pergamon Press, 1987; in "Prodrugs: challenges and Rewards", V.Stella, R.Borchardt, M.Hageman, R.Oliyai, H.Maag, J.Tilley editors, springer-Verlag New York,2007, the entire contents of which are hereby incorporated by reference.
As used herein, the term "soft drug" refers to a compound that exerts a pharmacological effect but breaks down into inactive metabolic degradants such that the activity has a limited time. See, for example, "Soft drugs: principles and methods for the design of safe drugs", nicholas Bodor, medical Research Reviews, vol.4, no. 4, pp.449-469, 1984, which is hereby incorporated by reference in its entirety.
As used herein, the term "metabolite" (e.g., active metabolite) overlaps with a prodrug as described above. Such metabolites are therefore pharmacologically active compounds or compounds which are further metabolized to pharmacologically active compounds, which are derivatives produced by metabolic processes in the body of a subject. For example, such metabolites may result from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, etc., of the administered compound or salt or prodrug. Wherein the active metabolite is such a pharmacologically active derivative compound. For prodrugs, the prodrug compound is generally inactive or less active than the metabolite. For active metabolites, the parent compound may be an active compound or an inactive prodrug.
Prodrugs and active metabolites may be identified using conventional techniques known in the art. See, e.g., bertolini et al, 1997, J Med Chem, volume 40: 2011-2016; shan et al, J Pharm Sci, volume 86: pages 756-757; bagshawe,1995, drug Dev Res, volume 34: pages 220-230; wermuth, supra.
As used herein, the term "pharmaceutically acceptable" means that the substance or composition is compatible chemically and/or toxicologically with the other ingredients comprising the formulation and/or the subject being treated therewith.
As used herein, unless otherwise indicated, the term "pharmaceutically acceptable salt" includes salts that retain the biological effectiveness of the free acids and bases of the particular compound and are not biologically or otherwise undesirable. Contemplated pharmaceutically acceptable salt forms include, but are not limited to, mono-, di-, tri-, tetra-, and the like. Pharmaceutically acceptable salts are non-toxic in the amounts and concentrations at which they are administered. The preparation of such salts can facilitate pharmacological use by altering the physical properties of the compound without preventing the compound from exerting its physiological effects. Useful physical property changes include lowering the melting point to facilitate transmucosal administration, and increasing solubility to facilitate administration of higher concentrations of drug.
Pharmaceutically acceptable salts include acid addition salts such as those containing sulfate, chloride, hydrochloride, fumarate, maleate, phosphate, sulfamate, acetate, citrate, lactate, tartrate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, cyclohexylsulfamate and quinic acid salts. Pharmaceutically acceptable salts may be obtained from acids such as hydrochloric acid, maleic acid, sulfuric acid, phosphoric acid, sulfamic acid, acetic acid, citric acid, lactic acid, tartaric acid, malonic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, cyclohexylsulfamic acid, fumaric acid, and quinic acid.
Pharmaceutically acceptable salts also include base addition salts when acidic functional groups such as carboxylic acids or phenols are present, such as those containing benzathine, chloroprocaine, choline, diethanolamine, ethanolamine, t-butylamine, ethylenediamine, meglumine, procaine, aluminum, calcium, lithium, magnesium, potassium, sodium, ammonium, alkylamines, and zinc. See, for example, "Remington's Pharmaceutical Sciences", 19 th edition, mack Publishing co., easton, PA, volume 2, page 1457, 1995; "Handbook of Pharmaceutical Salts: properties, selection, and Use", stahl and Wermuth, wiley-VCH, weinheim, germany, 2002. Such salts can be prepared using the appropriate corresponding bases.
Pharmaceutically acceptable salts can be prepared by standard techniques. For example, the free base form of the compound may be dissolved in a suitable solvent, such as an aqueous or aqueous alcoholic solution containing the appropriate acid, and then isolated by evaporation of the solution. Thus, if a particular compound is a base, the desired pharmaceutically acceptable salts can be prepared by any suitable method available in the art, for example, with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or treating the free base with an organic acid such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, pyranosidyl acid (such as glucuronic acid or galacturonic acid), alpha-hydroxy acid (such as citric acid or tartaric acid), amino acid (such as aspartic acid or glutamic acid), aromatic acid (such as benzoic acid or cinnamic acid), sulfonic acid (such as p-toluenesulfonic acid or ethanesulfonic acid), or the like.
Similarly, if a particular compound is an acid, the desired pharmaceutically acceptable salt can be prepared by any suitable method, for example, treating the free acid with an inorganic or organic base such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, and the like. Illustrative examples of suitable salts include organic salts derived from amino acids such as L-glycine, L-lysine and L-arginine, ammonia, primary, secondary and tertiary amines, and cyclic amines such as hydroxyethylpyrrolidine, piperidine, morpholine or piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
It is also to be understood that the compounds of the present disclosure may exist in unsolvated forms, solvated forms (e.g., hydrated forms), and solid forms (e.g., crystalline forms or polymorphs), and the present disclosure is intended to encompass all such forms.
As used herein, the term "solvate" or "solvated form" refers to a form of solvent addition containing a stoichiometric or non-stoichiometric amount of solvent. Some compounds tend to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thereby forming solvates. If the solvent is water, the solvate formed is a hydrate; if the solvent is an alcohol, the solvate formed is an alcoholate. Hydrates are formed by the association of one or more water molecules with a molecule of matter, whichThe reclaimed water keeps the molecular state of H 2 And O. Examples of solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
As used herein, the terms "crystal form", "polymorph (polymorphic forms)" and "polymorph (polymorphs)" are used interchangeably and refer to crystal structures in which a compound (or salt or solvate thereof) may crystallize in different crystal packing arrangements, all of which have the same elemental composition. Different crystalline forms typically have different X-ray diffraction patterns, infrared spectra, melting points, density hardness, crystal shape, optical and electrical properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors may cause one form to predominate. Crystalline polymorphs of a compound may be prepared by crystallization under different conditions.
The present disclosure is also intended to include all isotopes of atoms occurring in the compounds. Isotopes of atoms include atoms having the same atomic number but different mass numbers. For example, unless otherwise specified, hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine, or iodine in the compounds of the present disclosure are intended to also include isotopes thereof, such as, but not limited to 1 H、 2 H、 3 H、 11 C、 12 C、 13 C、 14 C、 14 N、 15 N、 16 O、 17 O、 18 O、 31 P、 32 P、 32 S、 33 S、 34 S、 36 S、 17 F、 18 F、 19 F、 35 Cl、 37 Cl、 79 Br、 81 Br、 124 I、 127 I and 131 I. in some embodiments, hydrogen includes protium, deuterium, and tritium. In some embodiments, the carbon comprises 12 C and 13 C。
those skilled in the art will appreciate that the compounds of the present disclosure may exist in different tautomeric forms, and all such forms are included within the scope of the present disclosure. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which may be interconverted through a low energy barrier. The presence and concentration of isomeric forms will depend on the environment in which the compound is placed and may vary depending on, for example, whether the compound is a solid or in an organic or aqueous solution. For example, proton tautomers (also referred to as proton transfer tautomers) include tautomers that migrate through protons, such as keto-enols, amide-imidic acids, lactam-lactimides, imine-enamine isomerizations, and cyclic forms in which a proton may occupy two or more positions of a heterocyclic ring system. Valence tautomers include interconversion by recombination of some of the bonding electrons. Tautomers can be balanced or sterically locked into one form by appropriate substitutions. Unless otherwise indicated, a compound of the present disclosure identified by name or structure as one particular tautomeric form is intended to include the other tautomeric form.
Synthesis of Compounds
The synthesis of the compounds provided herein, including pharmaceutically acceptable salts thereof, is illustrated in the synthetic schemes of the examples. The compounds provided herein can be prepared using any known organic synthesis technique and can be synthesized according to any of a variety of possible synthetic routes, thus these schemes are merely illustrative and are not meant to limit other possible methods that can be used to prepare the compounds provided herein. In addition, the steps in the schemes are for better illustration and may be changed as appropriate. Embodiments of the compounds in the examples were synthesized for research purposes and for potential submission to regulatory agencies.
The reaction to prepare the disclosed compounds can be carried out in a suitable solvent, which can be readily selected by one skilled in the art of organic synthesis. Suitable solvents may be substantially unreactive with the starting materials (reactants), intermediates, or products at the temperature at which the reaction is carried out, e.g., temperatures which may range from the freezing temperature of the solvent to the boiling temperature of the solvent. A given reaction may be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, one skilled in the art can select a suitable solvent for the particular reaction step.
The preparation of the compounds of the present disclosure may involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of suitable protecting groups, can be readily determined by those skilled in the art. The chemistry of protecting Groups can be found, for example, in T.W.Greene and P.G.M.Wuts, "Protective Groups in Organic Synthesis", 3 rd edition, wiley & Sons, inc., new York (1999); p. kocienski, "Protecting Groups", georg Thieme Verlag, 2003; and Peter g.m.wuts, "green's Protective Groups in Organic Synthesis", 5 th edition, wiley,2014, all of which are incorporated herein by reference in their entirety.
The reaction may be monitored according to any suitable method known in the art. For example, product formation can be by spectroscopic methods such as nuclear magnetic resonance spectroscopy (e.g., nuclear magnetic resonance spectroscopy) 1 H or 13 C) Infrared spectroscopy, spectrophotometry (e.g., UV-visible light), mass spectrometry, or by chromatographic methods such as High Performance Liquid Chromatography (HPLC), liquid chromatography-mass spectrometry (LCMS), or Thin Layer Chromatography (TLC). The compounds can be purified by a person skilled in the art by a variety of methods, including High Performance Liquid Chromatography (HPLC), ("Preparative LC-MS Purification: improved Compound Specific Method Optimization", karl F.Blom, brian Glass, richard spots, andrew P.Combs, J.Combi.chem.2004, vol.6, no. 6, p.874-883, which is incorporated herein by reference in its entirety) and normal phase silica gel chromatography.
The structures of the compounds in the examples were characterized by Nuclear Magnetic Resonance (NMR). NMR spectra were obtained on a Bruker AVANCE III HD 400 nuclear magnetic resonance spectrometer, respectively 1 H is operated at 400MHz for 13 C was run at 101 MHz. 1 H NMR spectra in CHCl 3 -d、(CH 3 ) 2 SO-d 6 And (CH) 3 ) 2 CO-d 6 Recording at 400MHz using residual CHCl 3 (7.26 ppm), DMSO (2.50 ppm) and (CH) 3 ) 2 CO (2.05 ppm) as internal standard. 13 C NMR spectra in CHCl 3 -d、(CH 3 ) 2 SO-d 6 And (CH) 3 ) 2 CO-d 6 Recording at 101MHz using residual CHCl 3 (77.16 ppm), DMSO (39.52 ppm) and (CH) 3 ) 2 CO (29.84 ppm and 206.26 ppm) was used as an internal standard.
Mass spectrometry was performed on a Thermo Scientific QOxctive Mass spectrometer (ESI) of a mass spectrometry apparatus of the institute of medicine, qinghua university.
In Merck Kieselgel
Figure BDA0003863898460000452
Thin layer chromatography was performed on F254 plates, eluted with the indicated solvents, observed with a 254nm UV lamp and stained with 12-molybdophosphoric acid in ethanol. Purification of the Compounds Using flash chromatography (silica gel)
Figure BDA0003863898460000453
230-400 mesh, silicon inc.
Known starting materials of the present disclosure can be synthesized by use or according to methods known in the art, or can be purchased from commercial suppliers. Unless otherwise stated, analytical grade solvents and commercial reagents were used without further purification.
Unless otherwise indicated, the reactions of the present disclosure are carried out under a positive pressure of nitrogen or argon or with a drying tube in anhydrous solvents, and the reaction flasks are typically equipped with rubber septa to introduce substrates and reagents via syringe. The glassware is oven dried and/or heat dried.
For illustrative purposes, the following shows a general synthetic route to the compounds of the present disclosure as well as key intermediates. For a more detailed description of the individual reaction steps, see the examples section below. One skilled in the art will appreciate that other synthetic routes may be used to synthesize the compounds of the invention. Although specific starting materials and reagents are described in the schemes and discussed below, other starting materials and reagents can be readily substituted to provide various derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemical methods well known to those skilled in the art.
General synthetic route
In some embodiments, the compounds provided herein may be prepared according to scheme 1, wherein L, D, and E are as defined above.
Scheme 1
Figure BDA0003863898460000451
Reagents and conditions: (i) 2-Bromoacetic acid tert-butyl ester, naH, bu 4 N + I - DMF, room temperature, 24 hours; (ii) 1M NaOH, refluxing for 2h, then acidifying with 1M HCl; (iii) 5-bromopentan-1-ol, K 2 CO 3 DMF,70 ℃, overnight; (iv) DMAP, EDCI, dichloromethane (DCM), room temperature, overnight; (v) water, TFA, room temperature, 40 min.
In some embodiments, the compounds provided herein may be prepared according to scheme 2, wherein L, D, and E are as defined above.
Scheme 2
Figure BDA0003863898460000461
Reagents and conditions: (i) 1, 5-dibromopentane, bu 4 N + Br - ACN, reflux, 20 hours; (ii) Naphthalene-2-ol, K 2 CO 3 DMF,70 ℃,24 hours; (iii) hydrazine hydrate, etOH, reflux for 4 hours; (iv) 2-Bromoacetic acid tert-butyl ester, naH, bu 4 N + I - DMF, room temperature, 24 hours; (v) 1M NaOH, refluxed for 2 hours, then acidified with 1M HCl; (vi) DMAP, EDCI, DCM, room temperature overnight; (vii) Water, TFA, room temperature, 40 min.
In some embodiments, the compounds provided herein may be prepared according to scheme 3, wherein L, D, and E are as defined above.
Scheme 3
Figure BDA0003863898460000462
Reagents and conditions: (i) DMAP, EDCI, DCM, room temperature overnight; (ii) water, TFA, room temperature, 40 min.
In some embodiments, the compounds provided herein may be prepared according to scheme 4, wherein L, D, and E are as defined above.
Scheme 4
Figure BDA0003863898460000463
Reagents and conditions: (i) 1, 8-dibromooctane, naH, bu 4 N + I - DMF, room temperature, 24 hours; (ii) K is 2 CO 3 DMF,70 ℃ overnight; (iii) water, TFA, room temperature, 40 min.
In some embodiments, the compounds provided herein may be prepared according to scheme 5, wherein L, D, and E are as defined above.
Scheme 5
Figure BDA0003863898460000471
Reagents and conditions: (i) Ph is 3 CCl, pyridine, 75 ℃, overnight; (ii) BnBr, naH, bu 4 N + I - DMF, room temperature, 24 hours; (iii) HBr (48% in water), acOH, ice water bath, 5 min; (iv) 1, 8-dibromooctane, naH, bu 4 N + I - DMF, room temperature, 24 hours; (v) K 2 CO 3 DMF,70 ℃, overnight; (vi) H 2 Pb/C, methanol, room temperature, 24 hours.
In some embodiments, the compounds provided herein may be prepared according to scheme 6, wherein L, D, and E are as defined above.
Scheme 6
Figure BDA0003863898460000472
Reagents and conditions: (i) 1, 8-dibromooctane, naH, bu 4 N + I - DMF, chamberWarming for 24 hours; (ii) H 2 Pb/C, methanol, room temperature, 24 hours; (iii) K 2 CO 3 DMF,70 ℃ overnight.
In some embodiments, the compounds provided herein may be prepared according to scheme 7, wherein L, D, and E are as defined above.
Scheme 7
Figure BDA0003863898460000473
Reagents and conditions: (i) BnBr, naH, bu 4 N + I - DMF, room temperature, 24 hours; (ii) water, TFA, DCM, room temperature, 40 min; (iii) Dibutyltin oxide (IV), meOH,80 ℃,1.5 hours, b.BnBr, K 2 CO 3 DMF,40 ℃,16 hours; (iv) 1, 8-dibromooctane, naH, bu 4 N + I - DMF, room temperature, 24 hours; (v) H 2 Pb/C, methanol, room temperature, 24 hours; (vi) K 2 CO 3 DMF,70 ℃ overnight.
In some embodiments, the compounds provided herein may be prepared according to scheme 8, wherein L, D, and E are as defined above.
Scheme 8
Figure BDA0003863898460000474
Reagents and conditions: (i) BnOH, BF 3 Diethyl ether, DCM, room temperature, overnight; (ii) NaOMe, meOH, room temperature, 3 hours; (iii) PhCH (OMe) 2 TsOH, DMF,80 ℃ for 4 hours; (iv) BnBr, naH, bu 4 N + I - DMF, room temperature, 24 hours; (v) Et (Et) 3 SiH, TFA, DCM, room temperature, 24 hours; (vi) 1, 8-dibromooctane, naH, bu 4 N + I - DMF, room temperature, 24 hours; (vii) H 2 Pb/C, methanol, room temperature, 24 hours; (viii) K 2 CO 3 DMF,70 ℃ overnight.
In some embodiments, the compounds provided herein may be prepared according to scheme 9, wherein L, D, and E are as defined above.
Scheme 9
Figure BDA0003863898460000481
Reagents and conditions: (i) Ac of 2 O, pyridine, room temperature, overnight; (ii) BnOH, BF 3 Diethyl ether, DCM, room temperature, overnight; (iii) NaOMe, meOH, RT, 3 hours; (iv) PhCH (OMe) 2 TsOH, DMF,80 ℃,4 hours; (v) 1, 8-dibromooctane, naH, bu 4 N + I - DMF, room temperature, 24 hours; (vi) K is 2 CO 3 DMF,70 ℃ overnight; (vii) H 2 Pb/C, methanol, room temperature, 24 hours.
Use of compounds
In one aspect, the present disclosure provides compounds of formula (I) through formula (X) or pharmaceutically acceptable salts thereof, capable of binding to a PfHT polypeptide.
In some embodiments, a compound of formula (I) through formula (X), or a pharmaceutically acceptable salt thereof, is capable of binding to a PfHT polypeptide having the amino acid sequence of SEQ ID NO:1 or an analog thereof having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID NO: 1.
In some embodiments, a compound of formula (I) through formula (X), or a pharmaceutically acceptable salt thereof, is capable of binding to a PfHT polypeptide or analog thereof having an amino acid sequence of SEQ ID NO:1, which analog has at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID NO:1, wherein the Kd value is NO more than 20 μ M, NO more than 19 μ M, NO more than 18 μ M, NO more than 17 μ M, NO more than 16 μ M, NO more than 15 μ M, NO more than 14 μ M, NO more than 13 μ M, NO more than 12 μ M, NO more than 11 μ M, NO more than 10 μ M, NO more than 9 μ M, NO more than 8 μ M, NO more than 7 μ M, NO more than 6 μ M, NO more than 5 μ M, NO more than 4 μ M, NO more than 3 μ M, NO more than 2 μ M, NO more than 11 μ M, NO more than 10 μ M, NO more than 1 μ M, NO more than 10 μ M, NO more than 9 μ M, NO more than 8 μ M, NO more than 7 μ M, NO more than 0.0.0.0.0.0.0.0.0.0.0.0 μ M, as measured by microcalorimetry, as described herein.
In some embodiments, the compounds of formula (I) through formula (X), or pharmaceutically acceptable salts thereof, exhibit inhibitory activity against PfHT polypeptide or analogs thereof.
In some embodiments, compounds of formula (I) through formula (X), or pharmaceutically acceptable salts thereof, exhibit inhibitory activity against PfHT polypeptide having an amino acid sequence of SEQ ID NO:1 or an analog thereof having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID NO: 1.
As used herein, the term "inhibitory activity on a PfHT polypeptide" refers to a decrease in the activity of a PfHT polypeptide in direct or indirect response to the presence of a compound of formula (I) to formula (X), or a pharmaceutically acceptable salt, relative to the activity of the PfHT polypeptide in the absence of a compound of formula (I) to formula (X), or a pharmaceutically acceptable salt.
In some embodiments, this decrease in activity may be due to the interaction of a compound of formula (I) through formula (X), or a pharmaceutically acceptable salt thereof, with a PfHT polypeptide.
In some embodiments, the compounds of formulas (I) through (X), or pharmaceutically acceptable salts, can decrease the activity of a PfHT polypeptide by binding to the PfHT polypeptide.
In some embodiments, a compound of formula (I) through formula (X), or a pharmaceutically acceptable salt thereof, can decrease the activity of a PfHT polypeptide by binding to the R1 binding pocket and the R2 binding pocket as described above.
In some embodiments, a compound or pharmaceutically acceptable salt of formula (I) through formula (X) may decrease the activity of a PfHT polypeptide by binding to the R1 binding pocket, the R2 binding pocket, and the R3 binding pocket as described above.
In some embodiments, a compound of formula (I) through formula (X), or a pharmaceutically acceptable salt thereof, is capable of inhibiting a PfHT polypeptide having an amino acid sequence of SEQ ID NO:1, or an analog thereof, having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID NO:1, wherein half of the maximum Effective Concentration (EC) is 50 ) Values of no more than 1000 μ M, no more than 900 μ M, no more than 800 μ M, no more than 700 μ M, no more than 600 μ M, no more than 500 μ M, no more than 400 μ M, no more than 300 μ M, no more than 200 μ M, no more than 100 μ M, no more than 90 μ M, no more than 80 μ M, no more than 70 μ M, no more than 60 μ M, no more than 50 μ M, no more than 40 μ M, no more than 30 μ M, no more than 20 μ M, no more than 10 μ M, no more than 5 μ M, no more than 4 μ M, no more than 3 μ M, no more than 2 μ M, no more than 1 μ M, no more than 900nM, no more than 800nM, no more than 700nM, no more than 600nM, no more than 500nM, no more than 400nM or no more than 300nM.
In some embodiments, a compound of formula (I) through formula (X), or a pharmaceutically acceptable salt thereof, is capable of inhibiting a PfHT polypeptide having the amino acid sequence of SEQ ID NO:1, or an analog thereof, having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID NO:1, wherein half of the maximum Inhibitory Concentration (IC) is the Inhibitory Concentration (IC) 50 ) The value is not more than 100. Mu.M, not more than 90. Mu.M, not more than 80. Mu.M, not more than 70. Mu.M, not more than 60. Mu.MNo more than 50 μ M, no more than 40 μ M, no more than 30 μ M, no more than 20 μ M, no more than 15 μ M, no more than 10 μ M, no more than 5 μ M, no more than 4 μ M, no more than 3 μ M, no more than 2 μ M, no more than 1 μ M, no more than 900nM, no more than 800nM, no more than 700nM, no more than 600nM, no more than 500nM, no more than 400nM or no more than 300nM.
In some embodiments, a compound of formula (I) through formula (X), or a pharmaceutically acceptable salt thereof, exhibits better inhibitory activity against a PfHT polypeptide having the amino acid sequence of SEQ ID NO:1 or an analog thereof having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID NO:1 as compared to compound 3361.
In certain embodiments, a compound of formula (I) through formula (X), or a pharmaceutically acceptable salt thereof, is capable of inhibiting binding of a PfHT polypeptide having the amino acid sequence of SEQ ID NO:1, or an analog thereof, having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID NO:1, wherein the EC is at least one of SEQ ID NO:1, or a pharmaceutically acceptable salt thereof 50 EC of value not exceeding compound 3361 50 The value is obtained.
In certain embodiments, the compounds of formula (I) through formula (X), or pharmaceutically acceptable salts thereof, are capable of inhibiting PfHT polypeptides, wherein EC is 50 Value and EC of Compound 3361 50 The value is at least 5 times, at least 4 times, at least 3 times, or at least 2 times smaller than the value.
In certain embodiments, the compounds of formulae (I) through (X), or pharmaceutically acceptable salts thereof, are capable of inhibiting the binding of a PfHT polypeptide having the amino acid sequence of SEQ ID NO:1, or an analog thereof having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, or C) relative to SEQ ID NO:1, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity, wherein IC is IC 50 IC of value not exceeding compound 3361 50 The value is obtained.
In certain embodiments, the compounds of formula (I) through formula (X), or pharmaceutically acceptable salts thereof, are capable of inhibiting PfHT polypeptides, wherein IC 50 IC of value with Compound 3361 50 The value is at least 65 times, at least 60 times, at least 50 times, at least 40 times, at least 30 times, at least 20 times, at least 15 times, at least 10 times, at least 8 times, at least 6 times, at least 4 times, or at least 2 times smaller than the value.
In some embodiments, the compounds of formulae (I) through (X), or pharmaceutically acceptable salts thereof, exhibit selective inhibitory activity against PfHT polypeptides over glucose transporter type 1 (GLUT 1).
In some embodiments, the compounds of formulae (I) through (X), or pharmaceutically acceptable salts thereof, exhibit selective inhibitory activity over GLUT1 on PfHT polypeptide having the amino acid sequence of SEQ ID NO:1 or an analog thereof having at least 70% (e.g., at least 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%) sequence identity to SEQ ID NO: 1.
As used herein, the term "selective inhibitor of PfHT" or "selective inhibition of PfHT" refers to a compound provided that inhibits PfHT better than GLUT1 as determined by the countercurrent assay described herein. In some embodiments, the term "selective inhibitor of PfHT over GLUT1" or "selective inhibition of PfHT over GLUT1" refers to a compound provided that inhibits PfHT-mediated D-glucose uptake to a greater extent than GLUT 1-mediated D-glucose uptake. In some embodiments, the compounds of formula (I) through formula (X), or pharmaceutically acceptable salts thereof, reduce the percentage of PfHT-mediated D-glucose uptake to a value that is at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, at least 50-fold, at least 60-fold, at least 70-fold, at least 80-fold, at least 90-fold, at least 95-fold less than the percentage of GLUT 1-mediated D-glucose uptake, as determined by the assays described herein.
Accordingly, compounds of formulae (I) to (X), or pharmaceutically acceptable salts thereof, are provided which are highly potent PfHT inhibitors and have high selectivity for PfHT over GLUT 1. Such compounds would allow the treatment of diseases or disorders that can be treated by inhibiting PfHT in a relatively selective manner, thereby minimizing potential side effects associated with the inhibition of GLUT 1.
In some embodiments, the compounds of formula (I) through formula (X) or pharmaceutically acceptable salts exhibit low cytotoxicity, e.g., in human renal epithelial cell lines and human hepatic epithelial cell lines, with half maximal Cytotoxic Concentration (CC) 50 ) Values greater than 1 μ M, greater than 2 μ M, greater than 3 μ M, greater than 4 μ M, greater than 5 μ M, greater than 6 μ M, greater than 7 μ M, greater than 8 μ M, greater than 9 μ M, greater than 10 μ M, greater than 15 μ M, greater than 20 μ M, greater than 25 μ M, greater than 30 μ M, greater than 35 μ M, greater than 40 μ M, greater than 45 μ M, or greater than 50 μ M.
Due to their inhibitory activity on PfHT polypeptides, compounds of formulae (I) to (X), or pharmaceutically acceptable salts thereof, are useful in therapy, for example, for treating diseases or disorders associated with PfHT polypeptides, such as malaria.
As used herein, the term "therapy" is intended to have its normal meaning of treating a disease, in order to alleviate one, some or all of its symptoms, either completely or partially, or to correct or compensate for an underlying pathology, thereby achieving a beneficial or desired clinical outcome. For purposes of this disclosure, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. "therapy" may also refer to an extended survival time as compared to the expected survival time when not receiving treatment. Individuals in need of treatment include individuals already with the disorder or disease as well as individuals susceptible to the disorder or disease or individuals in whom the disorder or disease is to be prevented. The term "therapy" also includes prophylaxis, unless specifically indicated to the contrary. The terms "therapeutic" and "therapeutically" should be interpreted in a corresponding manner.
As used herein, the term "prevention (prophyxiases)" is intended to have its normal meaning and includes primary prevention to prevent the onset of disease and secondary prevention in which disease has occurred and the patient is protected, either temporarily or permanently, to prevent the exacerbation or worsening of disease or the occurrence of new symptoms associated with disease.
The term "treatment" is used synonymously with "therapy". Similarly, the term "treating" can be considered "applying therapy," wherein "therapy" is as defined herein.
According to another aspect, there is provided a compound of formula (I) to formula (X) or a pharmaceutically acceptable salt thereof, for use in therapy, for example in therapy related to PfHT polypeptides.
In some embodiments, compounds of formula (I) through formula (X), or pharmaceutically acceptable salts, are provided for use in therapy for treating malaria.
In some embodiments, compounds of formula (I) through formula (X), or pharmaceutically acceptable salts, are provided for use as a medicament, such as an antimalarial drug.
In some embodiments, compounds of formula (I) through formula (X), or pharmaceutically acceptable salts, are provided for treating a disease or disorder associated with PfHT polypeptides, such as malaria.
In some embodiments, compounds of formula (I) through formula (X), or pharmaceutically acceptable salts thereof, are provided for use in the manufacture of a medicament to treat a disease or disorder associated with a PfHT polypeptide, such as malaria.
Furthermore, due to their inhibitory activity against PfHT polypeptides, compounds of formula (I) through formula (X) or pharmaceutically acceptable salts may be used to kill or inhibit the growth of plasmodium.
Thus, in another aspect, there is provided a compound of formula (I) to formula (X) or a pharmaceutically acceptable salt thereof for use in killing or inhibiting the growth of plasmodium.
Pharmaceutical composition
In another aspect, a pharmaceutical composition is provided comprising one or more molecules or compounds of the present disclosure, or a pharmaceutically acceptable salt thereof.
In another aspect, a pharmaceutical composition is provided comprising one or more molecules or compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
As used herein, the term "pharmaceutical composition" refers to a formulation containing a molecule or compound of the present disclosure in a form suitable for administration to a subject.
As used herein, the term "pharmaceutically acceptable excipient" refers to an excipient that can be used in the preparation of pharmaceutical compositions, which is generally safe, non-toxic, and neither biologically nor otherwise undesirable, and includes excipients that are acceptable for veterinary as well as human pharmaceutical use. As used herein, "pharmaceutically acceptable excipient" includes one and more than one such excipient. The term "pharmaceutically acceptable excipient" also includes "pharmaceutically acceptable carriers" and "pharmaceutically acceptable diluents".
The particular excipients used will depend on the mode and purpose for which the compounds of the present disclosure are used. The solvent is generally selected based on the consideration by those skilled in the art of safe administration to mammals including humans. Generally, the safe solvent is a non-toxic aqueous solvent, such as water and other non-toxic solvents that are soluble or miscible in water. Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycol (e.g., PEG 400, PEG 300), and the like, and mixtures thereof.
In some embodiments, suitable excipients may include buffering agents such as phosphates, citrates and other organic acids; antioxidants, including ascorbic acid and methionine; preservatives (such as octadecyl dimethyl benzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butanol)Or benzyl alcohol; alkyl parabens such as methyl paraben or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose, or sorbitol; salt-forming counterions such as sodium; metal complexes (e.g., zn-protein complexes); and/or nonionic surfactants, such as TWEEN TM 、PLURONICS TM Or polyethylene glycol (PEG).
In some embodiments, suitable excipients may include one or more stabilizers, surfactants, wetting agents, lubricants, emulsifiers, suspending agents, preservatives, antioxidants, opacifiers, glidants, processing aids, colorants, sweeteners, fragrances, flavoring agents and other known additives to provide an elegant appearance of a drug (i.e., a compound of the present disclosure or a pharmaceutical composition thereof) or to aid in the manufacture of a pharmaceutical product (i.e., a drug). The active pharmaceutical ingredient may also be encapsulated in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization (e.g., hydroxymethylcellulose or gelatin microcapsules and poly (methylmethacylate) microcapsules, respectively), in colloidal drug delivery systems (e.g., liposomes, albumin microspheres, microemulsions, nanoparticles, and nanocapsules), or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences, 16 th edition, osol, A. Edition (1980). A "liposome" is a small vesicle composed of various types of lipids, phospholipids and/or surfactants that can be used to deliver drugs (e.g., the compounds disclosed herein and optional chemotherapeutic agents) to mammals, including humans. The components of liposomes are typically arranged in a bilayer, similar to the lipid arrangement of biological membranes.
The pharmaceutical compositions provided herein can be in any form that allows for administration of the composition to a subject (including but not limited to a human) and are formulated to be compatible with the intended route of administration.
The pharmaceutical compositions provided herein contemplate a variety of routes, and thus the pharmaceutical compositions provided herein can be provided in bulk or unit dosage form depending on the intended route of administration. For example, for oral, buccal, and sublingual administration, powders, suspensions, granules, tablets, pills, capsules, soft capsules, and caplets may be accepted as solid dosage forms, and emulsions, syrups, elixirs, suspensions, and solutions may be accepted as liquid dosage forms. For administration by injection, emulsions and suspensions may be received as liquid dosage forms, while powders suitable for reconstitution with an appropriate solution may be received as solid dosage forms. For administration by inhalation, solutions, sprays, dry powders and aerosols may be acceptable dosage forms. For topical (including buccal and sublingual) or transdermal administration, powders, sprays, ointments, pastes, creams, lotions, gels, solutions and patches may be acceptable dosage forms. For vaginal administration, pessaries, tampons, creams, gels, pastes, foams and sprays may be acceptable dosage forms.
The amount of active ingredient in a unit dosage form of the composition is a therapeutically effective amount and will vary depending upon the particular treatment involved. As used herein, the term "therapeutically effective amount" refers to an amount of a molecule, compound, or composition comprising the molecule or compound that treats, ameliorates, or prevents an identified disease or disorder or exhibits a detectable therapeutic or inhibitory effect. This effect can be detected by any assay known in the art. The precise effective amount for a subject will depend upon the weight, size and health of the subject; the nature and extent of the disorder; the rate of administration; selecting a therapeutic agent or combination of therapeutic agents for administration; and the discretion of the prescribing physician. A therapeutically effective amount for a given situation can be determined by routine experimentation within the skill and judgment of the clinician.
In some embodiments, the pharmaceutical compositions of the present disclosure may be in the form of a formulation for oral administration.
In certain embodiments, the pharmaceutical compositions of the present disclosure may be in the form of a tablet formulation. Pharmaceutically acceptable excipients suitable for tablet formulation include, for example, inert diluents such as lactose, sodium carbonate, calcium phosphate or calcium carbonate; granulating and disintegrating agents such as corn starch or alginic acid; binders such as starch; lubricants such as magnesium stearate, stearic acid or talc; preservatives such as ethyl or propyl paraben; and antioxidants such as ascorbic acid. The tablet formulation may be uncoated or coated to modify its disintegration and subsequent absorption of the active ingredient in the gastrointestinal tract, or to improve its stability and/or appearance, in either case using conventional coating agents and procedures well known in the art.
In certain embodiments, the pharmaceutical compositions of the present disclosure may be in the form of hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., calcium carbonate, calcium phosphate, or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil, such as peanut oil, liquid paraffin, or olive oil.
In certain embodiments, the pharmaceutical compositions of the present disclosure may be in the form of an aqueous suspension, which typically contains the active ingredient in finely divided form in combination with one or more suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents such as lecithin or condensation products of alkylene oxides with fatty acids (e.g. polyoxyethylene stearate), or condensation products of ethylene oxide with long chain aliphatic alcohols (e.g. heptadecaethyleneoxycetanol), or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol (e.g. polyoxyethylene sorbitol monooleate), or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides (e.g. polyethylene sorbitan monooleate). Aqueous suspensions may also contain one or more preservatives, such as ethyl or propyl p-hydroxybenzoate, antioxidants, such as ascorbic acid, coloring, flavoring and/or sweetening agents, such as sucrose, saccharin or aspartame.
In certain embodiments, the pharmaceutical compositions of the present disclosure may be in the form of an oily suspension, typically containing the active ingredient suspended in a vegetable oil (such as peanut oil, olive oil, sesame oil or coconut oil) or in a mineral oil (such as liquid paraffin). Oily suspensions may also contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents, such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
In certain embodiments, the pharmaceutical compositions of the present disclosure may be in the form of an oil-in-water emulsion. The oily phase may be a vegetable oil, such as olive oil or arachis oil, or a mineral oil, such as liquid paraffin, or a mixture of any of these. Suitable emulsifying agents may be, for example, naturally-occurring gums such as gum acacia and gum tragacanth, naturally-occurring phosphatides such as soya bean and lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides such as sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide such as polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening, flavouring and preservative agents.
In certain embodiments, the pharmaceutical compositions provided herein can be in the form of syrups and elixirs, which can contain sweetening agents, such as glycerin, propylene glycol, sorbitol, aspartame or sucrose, demulcents, preservatives, flavoring agents and/or coloring agents.
In some embodiments, the pharmaceutical compositions of the present disclosure may be in the form of a formulation for administration by injection.
In certain embodiments, the pharmaceutical compositions of the present disclosure may be in the form of a sterile injectable formulation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension (such as a solution of 1, 3-butanediol) in a non-toxic parenterally acceptable diluent or solvent, or may be prepared as a lyophilized powder. Acceptable vehicles and solvents that may be used are water, ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. In addition, fatty acids (such as oleic acid) are likewise useful in the preparation of injectable preparations.
In some embodiments, the pharmaceutical compositions of the present disclosure may be in the form of a formulation for administration by inhalation.
In certain embodiments, the pharmaceutical compositions of the present disclosure may be in the form of aqueous and non-aqueous (e.g., in fluorocarbon propellants) aerosols comprising any suitable solvent and optionally other compounds, such as, but not limited to, stabilizers, antimicrobials, antioxidants, pH adjusters, surfactants, bioavailability modifiers, and combinations of these. Carriers and stabilizers vary with the requirements of a particular compound, but generally include nonionic surfactants (Tweens, pluronics or polyethylene glycols), innocuous proteins such as serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars or sugar alcohols.
In some embodiments, the pharmaceutical compositions of the present disclosure may be in the form of a formulation for topical or transdermal administration.
In certain embodiments, the pharmaceutical compositions provided herein may be in the form of creams, ointments, gels, and aqueous or oily solutions or suspensions, which are generally obtainable by formulating the active ingredient with conventional, topically acceptable excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
In certain embodiments, the pharmaceutical compositions provided herein can be formulated in the form of transdermal skin patches well known to those of ordinary skill in the art.
Pharmaceutically acceptable excipients and carriers, in addition to those representative dosage forms described above, are generally known to those skilled in the art and are therefore included in this disclosure. Such excipients and carriers are described, for example, in "Remingtons Pharmaceutical Sciences", mack pub.co., new Jersey (1991); "Remington: the Science and Practice of Pharmacy", the University of The Sciences in Philadelphia, 21 st edition, LWW (2005), which is incorporated herein by reference.
In some embodiments, the pharmaceutical compositions of the present disclosure may be formulated in a single dosage form. The amount of a compound provided herein in a single dosage form will vary depending upon the subject being treated and the particular mode of administration.
In some embodiments, the pharmaceutical compositions of the present disclosure may be formulated such that an administrable dose is from 0.001mg/kg body weight/day to 500mg/kg body weight/day, for example, the compound may be administered in an amount of 0.01mg/kg body weight/day to 400mg/kg body weight/day, 0.01mg/kg body weight/day to 300mg/kg body weight/day, 0.1mg/kg body weight/day to 200mg/kg body weight/day, 0.1mg/kg body weight/day to 150mg/kg body weight/day, 0.1mg/kg body weight/day to 100mg/kg body weight/day, 0.5mg/kg body weight/day to 80mg/kg body weight/day, 0.5mg/kg body weight/day to 60mg/kg body weight/day, 0.5mg/kg body weight/day to 50mg/kg body weight/day, 1mg/kg body weight/day to 45mg/kg body weight/day, 1mg/kg body weight/day to 40mg/kg body weight/day, 1mg/kg body weight/day to 35mg/kg body weight/day, 1mg/kg body weight/day to 30mg/kg body weight/day, or the pharmaceutically acceptable salts thereof may be administered in the present invention. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effects, provided that such larger doses are first divided into several small doses for administration throughout the day. For further information on the route of administration and dosage regimen, see "Comprehensive medical Chemistry" (Corwin Hansch; chairman of Editorial Board), vol.5, chapter 25.3, pergamon Press,1990, which is expressly incorporated herein by reference.
In some embodiments, the pharmaceutical compositions of the present disclosure can be formulated as short acting, immediate release, long acting, and sustained release. Thus, the pharmaceutical formulations of the present disclosure may also be formulated for controlled or sustained release.
In another aspect, a veterinary composition is also provided, comprising one or more molecules or compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, and a veterinary carrier. Veterinary carriers are materials used for the purpose of administering the composition and can be solid, liquid or gaseous materials which are inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered parenterally, orally or by any other desired route.
The pharmaceutical or veterinary composition may be packaged in various ways depending on the method used to administer the drug. For example, an article of manufacture for dispensing may comprise a container having deposited therein a composition in a suitable form. Suitable containers are well known to those skilled in the art and include materials such as bottles (plastic and glass), sachets (sachets), ampoules, plastic bags, metal cylinders, and the like. The container may also include a tamper-evident assembly to prevent inadvertent access to the contents of the package. In addition, a label describing the contents of the container is placed on the container. The label may also include appropriate warnings. The compositions may also be packaged in unit-dose or multi-dose containers, for example sealed ampoules and vials (dials), and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described.
In another aspect, there is also provided a pharmaceutical composition comprising one or more compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, as a first active ingredient, and a second active ingredient.
In some embodiments, the second active ingredient has complementary activities to the compounds provided herein such that they do not adversely affect each other. Such ingredients are present in appropriate combinations in amounts effective for the intended purpose.
In some embodiments, the second active ingredient is an antimalarial agent. Exemplary antimalarial agents include, but are not limited to, artemether-lumefantrine (Coartem) TM ) Atovaquone chlorideGuanidine (Malarone) TM ) Mefloquine, chloroquine phosphate (Aralen) TM And biopharmaceuticals), hydroxychloroquine (Plaquenil) TM And biopharmaceuticals), chloroquine phosphate, hydroxychloroquine, primaquine phosphate, tafenoquine (Krintafel) TM ) Quinine sulfate-doxycycline, quinine sulfate-tetracycline, quinine sulfate-clindamycin, chloroquine phosphate-primaquine phosphate, chloroquine phosphate-tafenoquine, hydroxychloroquine phosphate-primaquine, hydroxychloroquine-tafenoquine (Krintafel ™) TM ) Atovaquone-chloroguanidine-primaquine phosphate, atovaquone-chloroguanidine-tafenoquine (Krintafel) TM ) Mefloquine-primaquine phosphate, mefloquine-talfenoquine (Krintafel) TM ) And the like.
Methods of treating diseases
In another aspect, the present disclosure provides a method of treating a disease or disorder associated with a PfHT polypeptide in a subject, the method comprising administering to a subject in need thereof a therapeutically effective amount of one or more molecules, one or more compounds, or a pharmaceutical composition provided herein.
As used herein, the term "subject in need thereof" is a subject having a disease or disorder associated with a PfHT polypeptide, such as malaria. "subject" includes a warm-blooded animal. In some embodiments, the warm-blooded animal is a human.
In this context, the term "therapeutically effective amount" refers to an amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, that is effective to provide "therapy" in a subject, or to "treat" a disease or disorder associated with a PfHT polypeptide (such as malaria) in a subject.
In some embodiments, the methods of treating a disease or disorder associated with a PfHT polypeptide of the present disclosure may be used as monotherapy. As used herein, the term "monotherapy" is directed to the administration of a single active or therapeutic compound to a subject in need thereof. In some embodiments, monotherapy will involve administering to a subject in need of such treatment a therapeutically effective amount of one or more compounds of the present disclosure, or a pharmaceutically acceptable salt thereof.
In some embodiments, the methods of treating a disease or disorder associated with a PfHT polypeptide of the present disclosure may involve, in addition to administering a compound of the present disclosure, one or more additional therapeutic agents, e.g., additional active agents. As used herein, the term "combination therapy" refers to the administration of a combination of multiple active compounds.
The additional therapeutic agent may be administered separately from the compound of the present disclosure as part of a multiple dose regimen. Alternatively, these additional therapeutic agents may be part of a single dosage form, mixed in a single composition with the compounds of the present disclosure.
In some embodiments, the additional therapeutic agent is an additional anti-malarial agent. Additional antimalarial agents may include, but are not limited to, artemether-lumefantrine (Coartem) TM ) Atovaquone-proguanil (Malarone) TM ) Mefloquine, chloroquine phosphate (Aralen) TM And biopharmaceuticals), hydroxychloroquine (plaquinil) TM And biopharmaceuticals), chloroquine phosphate, hydroxychloroquine, primaquine phosphate, and talfenoquine (Krintafel) TM ) Quinine sulfate-doxycycline, quinine sulfate-tetracycline, quinine sulfate-clindamycin, chloroquine phosphate-primaquine phosphate, chloroquine phosphate-tafenoquine, hydroxychloroquine phosphate-primaquine, hydroxychloroquine-tafenoquine (Krintafel ™) TM ) Atovaquone-chloroguanidine-primaquine phosphate, atovaquone-chloroguanidine-tafenoquine (Krintafel) TM ) Mefloquine-primaquine phosphate, mefloquine-talfenoquine (Krintafel) TM ) And the like.
In some embodiments, the compounds of the present disclosure may be administered simultaneously, sequentially or separately with an additional antimalarial agent.
In another aspect, there is provided a method of treating a disease or disorder associated with a PfHT polypeptide in a subject in need thereof, wherein a compound of the disclosure, or a pharmaceutically acceptable salt thereof, is administered simultaneously, separately or sequentially with one or more additional antimalarial agents.
Method of killing or inhibiting the growth of plasmodium
In another aspect, the present disclosure provides a method of killing or inhibiting the growth of plasmodium by administering an effective amount of a molecule, compound or pharmaceutically acceptable salt or pharmaceutical composition of the present disclosure.
In some embodiments, the method of killing or inhibiting the growth of plasmodium is performed in vivo.
In some embodiments, the method of killing or inhibiting the growth of plasmodium is performed in vitro.
Examples
For illustrative purposes, the following examples are included. It should be understood, however, that these examples do not limit the disclosure, and are intended only to set forth ways of practicing the disclosure. One skilled in the art will recognize that the chemical reactions can be readily adapted to prepare many other compounds of the present disclosure, and that alternative methods for preparing the compounds of the present disclosure are considered to be within the scope of the present disclosure. For example, the synthesis of non-exemplified compounds according to the present disclosure may be successfully carried out by modifications apparent to those skilled in the art, e.g., by appropriate protection of interfering groups, by the use of other suitable reagents and building blocks known in the art in addition to those described, and/or by routine modification of reaction conditions. Alternatively, other reactions disclosed herein or known in the art will be considered applicable to the preparation of other compounds of the present disclosure.
Example 1
5- (Naphthalen-2-yloxy) pentyl 2- (((3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate (Compound 3)
Figure BDA0003863898460000541
Reagents and conditions: (i) 2-Bromoacetic acid tert-butyl ester, naH, bu 4 N + I - DMF, room temperature, 24 hours; (ii) 1M NaOH, refluxed for 2h, and then acidified with 1M HCl; (iii) 5-bromopentan-1-ol, K 2 CO 3 DMF,70 ℃, overnight; (iv) DMAP, EDCI, DCM, room temperature overnight; (v) water, TFA, room temperature, 40 min.
Step 1: tert-butyl 2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d ] [1,3] dioxol-6-yl) oxy) acetate
Figure BDA0003863898460000542
In a 100mL round-bottomed flask, diacetone-D-glucose (1.0g, 3.8mmol) was dissolved in anhydrous N, N-Dimethylformamide (DMF) (25 mL), and a mineral oil solution of sodium hydride (60%) (230mg, 5.8mmol), tetra-N-butylammonium iodide (500g, 1.9mmol) was added in small portions under argon over 5 minutes. The mixture was stirred in an ice-water bath for 30 minutes. Tert-butyl 2-bromoacetate (2.235g, 11.5 mmol) was then added slowly. The mixture was allowed to warm to room temperature and stirred for 24 hours. The progress of the reaction was monitored by TLC and after completion of the reaction, part of the solvent was removed in vacuo and precipitated in water. The resulting residue was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by silica gel flash column chromatography (10% ethyl acetate/petroleum ether) to give 1.4g (97%) of 2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d ][1,3]Dioxol-6-yl) oxy) tert-butyl acetate as a pale yellow oil. 1 H NMR (400 MHz, chloroform-d) δ 5.90 (d, J =3.6hz, 1h), 4.72 (d, J =3.6hz, 1h), 4.34 (dt, J =7.5,5.9hz, 1h), 4.20-4.05 (m, 4H), 4.00 (dd, J =8.6,5.6hz, 1h), 3.95 (d, J =2.9hz, 1h), 1.49 (s, 12H), 1.43 (s, 3H), 1.36 (s, 3H), 1.32 (s, 3H) ppm.
And 2, step: 2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d ] [1,3] dioxol-6-yl) oxy) acetic acid
Figure BDA0003863898460000551
In a 100mL round bottom flask, tert-butyl 2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d ] [1,3] dioxol-6-yl) oxy) acetate (1.4 g, 3.7mmol) was dissolved in 1M aqueous sodium hydroxide solution (20 mL). The mixture was heated to reflux for 2 hours. The mixture was then cooled to room temperature and neutralized by 1M hydrochloric acid. The resulting residue was extracted with DCM, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give the crude product, which was used in the next step without further purification.
And 3, step 3:5- (naphthalen-2-yloxy) pentan-1-ol
Figure BDA0003863898460000552
In a 100mL round-bottomed flask, 2-naphthol (500mg, 3.5 mmol) was dissolved in anhydrous DMF (20 mL), and then 5-bromopentan-1-ol (868mg, 5.2 mmol), potassium carbonate (960 mg,6.9 mmol) were added to the solution. The mixture was stirred at 70 ℃ for 24 hours. The progress of the reaction was monitored by TLC and after completion of the reaction, the mixture was cooled to room temperature, part of the solvent was removed in vacuo and precipitated in water. The resulting residue was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by flash column chromatography on silica gel (25% ethyl acetate/petroleum ether) to give 842mg (100%) of 5- (naphthalen-2-yloxy) pentan-1-ol as a pale yellow solid. 1 H NMR (400 MHz, chloroform-d) δ 7.93-7.63 (m, 3H), 7.41 (ddd, J =8.1,6.8,1.3hz, 1h), 7.30 (ddd, J =8.1,6.8,1.2hz, 1h), 7.20-7.05 (m, 2H), 4.03 (t, J =6.4hz, 2h), 3.63 (t, J =6.3hz, 2h), 2.01 (s, 1H), 1.90-1.78 (m, 2H), 1.68-1.58 (m, 2H), 1.54 (dddd, J =12.1,7.3,5.1,1.6hz, 2h) ppm. 13 C NMR(101MHz,CDCl 3 )δ157.04,134.65,129.39,128.95,127.68,126.75,126.37,123.56,119.00,106.62,67.85,62.71,32.46,29.06,22.48ppm.
And 4, step 4:5- (Naphthalen-2-yloxy) pentyl 2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d ] [1,3] dioxol-6-yl) oxy) acetate
Figure BDA0003863898460000553
Adding 5- (naphthalene-2-yloxy) pentan-1-ol (165mg, 0.7 mmol), 2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuranPyrano [2,3-d ]][1,3]Dioxolen-6-yl) oxy) acetic acid (250mg, 0.8mmol), DMAP (20mg, 0.2mmol) and EDCI (165mg, 0.9mmol) were dissolved in dry DCM (5 mL). The mixture was stirred at room temperature overnight. The progress of the reaction was monitored by TLC and after completion of the reaction, part of the solvent was removed in vacuo and precipitated in water. The resulting residue was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by silica gel flash column chromatography (20% ethyl acetate/petroleum ether) to give 329mg (87%) of 5- (naphthalen-2-yloxy) pentyl 2- (((3aR, 5R,6S,6 aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d ] o ][1,3]Dioxol-6-yl) oxy) acetate as a light yellow oil. 1 H NMR (400 MHz, chloroform-d) δ 7.71 (td, J =8.9,8.2,6.2hz, 3h), 7.41 (ddd, J =8.2,6.8,1.3hz, 1h), 7.30 (td, J =7.4,6.8,1.2hz, 1h), 7.20-7.03 (m, 2H), 5.90 (d, J =3.7hz, 1h), 4.72 (d, J =3.7hz, 1h), 4.34 (dt, J =7.7,5.8hz, 1h), 4.24 (s, 2H), 4.22-3.94 (m, 8H), 1.84 (p, J =6.6hz, 2h), 1.73 (p, J =6.8hz, 2h), 1.56 (qd, J =10.1,9.3,6.1hz, 2h), 1.47 (s, 3H), 1.42 (s, 3H), 1.34 (s, 3H), 1.29 (s, 3H) ppm. 13 C NMR(101MHz,CDCl 3 )δ170.32,156.94,134.60,129.38,128.94,127.65,126.70,126.36,123.56,118.93,111.83,109.00,106.53,105.25,83.66,83.32,81.10,72.68,68.37,67.53,67.22,64.94,28.86,28.38,26.85,26.27,25.41,22.66ppm.
And 5:5- (Naphthalen-2-yloxy) pentyl 2- (((3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate (Compound 3)
Figure BDA0003863898460000561
2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2, 3-d)][1,3]Dioxolen-6-yl) oxy) acetate (308mg, 0.6 mmol) was placed in a 50mL round-bottomed flask, followed by trifluoroacetic acid (2 mL), water (0.2 mL). The mixture was stirred at room temperature for 40 minutes. The progress of the reaction was monitored by TLC and after completion of the reaction the solvent was evaporated directly in vacuo. The crude product was purified by flash column chromatography on silica gel (10% formazan)Alcohol in DCM) to give 102mg (38%) of compound 3 as a colorless solid. Yield: 38 percent. ESI-MS m/z:473.1783, [ M ] +Na ] + . 1 H NMR (400 MHz, chloroform-d) δ 7.72 (dd, J =12.5,8.6hz, 3h), 7.42 (t, J =7.5hz, 1h), 7.31 (t, J =7.5hz, 1h), 7.24-7.02 (m, 2H), 6.59-3.20 (m, 17H), 1.88-1.75 (m, 2H), 1.69 (d, J =8.6hz, 2h), 1.51 (dh, J =14.2,8.1,6.3hz, 2h) ppm. 13 C NMR(101MHz,CDCl 3 )δ173.85,173.74,156.89,134.57,129.36,128.90,127.63,126.71,126.33,123.54,118.91,106.56,92.36,86.33,84.29,76.07,74.83,72.26,70.00,69.37,69.20,69.13,67.53,65.65,62.14,61.65,28.81,28.24,22.53ppm.
Example 2
2- (2- (Naphthalen-2-yloxy) ethoxy) ethyl 2- (((3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate (Compound 5)
Figure BDA0003863898460000562
The title compound was prepared in a similar manner as described in example 1. Yield: 23 percent. ESI-MS m/z 475.1573[ m + Na ]] + . 1 H NMR(400MHz,DMSO-d 6 )δ7.90–7.76(m,3H),7.46(ddd,J=8.1,6.7,1.2Hz,1H),7.39–7.30(m,2H),7.18(dd,J=8.9,2.5Hz,1H),6.73(d,J=6.5Hz,1H),5.14–4.28(m,6H),4.26(dt,J=6.6,2.4Hz,2H),4.24–4.17(m,2H),3.88–3.79(m,2H),3.73(dd,J=5.7,3.7Hz,2H),3.68–3.36(m,3H),3.31–2.99(m,3H)ppm. 13 C NMR(101MHz,DMSO)δ172.37,172.18,156.76,134.71,129.78,128.94,127.97,127.16,126.85,124.05,119.17,107.16,97.15,92.56,86.57,83.53,76.92,74.82,72.43,72.39,69.87,69.78,69.33,69.29,68.74,67.55,64.14,61.24ppm.
Example 3
5- (Quinolin-6-Yloxy) pentyl 2- (((3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate (Compound 7)
Figure BDA0003863898460000563
The title compound was prepared in a similar manner as described in example 1. Yield: 26 percent. ESI-MS m/z:452.1916[ 2 ], [ M + H ]] + . 1 H NMR(400MHz,DMSO-d 6 )δ8.73(dd,J=4.2,1.7Hz,1H),8.25(dd,J=8.4,1.6Hz,1H),7.91(d,J=9.0Hz,1H),7.48(dd,J=8.3,4.2Hz,1H),7.44–7.35(m,2H),6.60(dd,J=111.0,5.3Hz,1H),5.23–4.46(m,3H),4.40(d,J=1.3Hz,2H),4.13(tdd,J=10.5,6.5,3.3Hz,4H),3.79–2.88(m,7H),1.83(p,J=6.7Hz,2H),1.69(p,J=6.8Hz,2H),1.56–1.47(m,2H)ppm. 13 C NMR(101MHz,DMSO)δ172.55,172.34,156.99,148.24,144.04,135.36,130.68,129.54,122.75,122.10,106.82,97.16,92.56,86.57,83.55,76.92,74.83,72.44,72.40,69.86,69.79,69.31,69.25,68.17,64.77,61.24,28.63,28.28,28.27,22.50ppm.
The following compounds were prepared using a similar procedure to that in example 1 but using different starting materials.
Figure BDA0003863898460000564
Figure BDA0003863898460000571
Figure BDA0003863898460000581
Example 4
N- (5- (Naphthalen-2-yloxy) pentyl) -2- (((3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetamide (Compound 6)
Figure BDA0003863898460000591
Reagents and conditions: (i) 1, 5-dibromopentane, bu 4 N + Br - ACN, reflux, 20 hours; (ii) 2-naphthalenol, K 2 CO 3 DMF,70 ℃,24 hours; (iii) Hydrazine hydrate, etOH, reflux, 4 hrsWhen the current is in the normal state; (iv) 2-Bromoacetic acid tert-butyl ester, naH, bu 4 N + I - DMF, room temperature, 24 hours; (v) 1M NaOH, refluxing, 2 hours, then acidifying with 1M HCl; (vi) DMAP, EDCI, DCM, room temperature overnight; (vii) Water, TFA, room temperature, 40 min.
Step 1:2- (5-Bromopentyl) isoindoline-1, 3-dione
Figure BDA0003863898460000592
Potassium phthalimide (500mg, 2.7 mmol) was stirred at reflux in 20mL of acetonitrile with a 2.5 fold excess of a mixture of the appropriate 1, 5-dibromopentane (1.551g, 6.7 mmol) and a catalytic amount of TBAB (174mg, 0.5 mmol) for 20 h. Subsequently, the reaction mixture was filtered and the filtrate was concentrated. The crude product was purified by flash column chromatography on silica gel (50% ethyl acetate/petroleum ether) to give 524mg (65%) of 2- (5-bromopentyl) isoindoline-1, 3-dione as a pale yellow oil. 1 H NMR (400 MHz, chloroform-d) δ 7.85 (dt, J =7.0,3.5hz, 2h), 7.73 (dd, J =5.5,3.0hz, 2h), 3.71 (t, J =7.2hz, 2h), 3.41 (t, J =6.7hz, 2h), 1.92 (p, J =6.9hz, 2h), 1.78-1.66 (m, 2H), 1.51 (tt, J =9.8,6.1hz, 2h) ppm.
Step 2:2- (5- (naphthalen-2-yloxy) pentyl) isoindoline-1, 3-dione
Figure BDA0003863898460000593
In a 100mL round-bottomed flask, 2- (5-bromopentyl) isoindoline-1, 3-dione (500mg, 1.7 mmol) was dissolved in anhydrous DMF (10 mL), and then naphthalene-2-ol (200mg, 1.4 mmol), potassium carbonate (383mg, 2.8mmol) were added to the solution. The mixture was stirred at 70 ℃ for 10 hours. The progress of the reaction was monitored by TLC and after completion of the reaction, the mixture was cooled to room temperature, part of the solvent was removed in vacuo and precipitated in water. The resulting residue was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by flash column chromatography on silica gel (50% ethyl acetate/petroleum ether) to give 486mg (80%) of 2- (5- (naphthalen-2-yloxy) pentyl) isoindoline- 1, 3-dione as a pale yellow oil. 1 H NMR (400 MHz, chloroform-d) δ 7.84 (dt, J =7.6,3.7hz, 2h), 7.79-7.65 (m, 5H), 7.42 (ddd, J =8.2,6.8,1.3hz, 1h), 7.31 (ddd, J =8.1,6.8,1.2hz, 1h), 7.11 (s, 2H), 4.07 (t, J =6.4hz, 2h), 3.74 (t, J =7.2hz, 2h), 1.98-1.85 (m, 2H), 1.81 (d, J =7.6hz, 2h), 1.59 (td, J =8.7,4.2hz, 2h) ppm.
And step 3:5- (naphthalen-2-yloxy) pentan-1-amine
Figure BDA0003863898460000601
A solution of 2- (5- (naphthalen-2-yloxy) pentyl) isoindoline-1, 3-dione (486mg, 1.4 mmol) and hydrazine hydrate (80%, 1.6mL, 25mmol) in ethanol (10 mL) was refluxed for 4 hours and then cooled to room temperature. The mixture was filtered and the solid was washed with 95% ethanol. The whole filtrate was concentrated and the solid was dissolved in DCM. The mixture was dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude product, which was used in the next step without further purification. 1 H NMR(400MHz,DMSO-d 6 )δ7.89–7.70(m,3H),7.45(t,J=7.6Hz,1H),7.37–7.25(m,2H),7.15(dd,J=8.9,2.5Hz,1H),4.07(t,J=6.5Hz,2H),2.56(t,J=6.4Hz,2H),2.21(d,J=97.3Hz,0H),1.78(p,J=6.7Hz,2H),1.46(qd,J=13.5,11.8,6.6Hz,4H)ppm.
And 4-5: tert-butyl 2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d ] [1,3] dioxol-6-yl) oxy) acetate and 2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d ] [1,3] dioxol-6-yl) oxy) acetate
Steps 4 and 5 were carried out in the same manner as in steps 1 and 2 of example 1 to give 2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d ] [1,3] dioxol-6-yl) oxy) acetic acid.
And 6:2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d ] [1,3] dioxol-6-yl) oxy) -N- (5- (naphthalen-2-yloxy) pentyl) acetamide
Figure BDA0003863898460000602
Reacting 5- (naphthalen-2-yloxy) pentan-1-amine (303mg, 1.3mmol), 2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2, 3-d-tetrahydrofurane [ -4, 2-d [ -E][1,3]Dioxolen-6-yl) oxy) acetic acid (465mg, 1.5mmol), DMAP (33mg, 0.3mmol) and EDCI (315mg, 1.6 mmol) were dissolved in anhydrous DCM (15 mL). The mixture was stirred at room temperature overnight. The progress of the reaction was monitored by TLC and after completion of the reaction, part of the solvent was removed in vacuo and precipitated in water. The resulting residue was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by silica gel flash column chromatography (25% ethyl acetate/petroleum ether) to give 238mg (35%) of 2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2, 3-d-tetrahydrofurane ] ][1,3]Dioxol-6-yl) oxy) -N- (5- (naphthalen-2-yloxy) pentyl) acetamide as a light yellow oil. 1 H NMR (400 MHz, chloroform-d) δ 7.85-7.63 (m, 3H), 7.48 (t, J =6.0hz, 1h), 7.43 (t, J =7.5hz, 1h), 7.32 (t, J =7.5hz, 1h), 7.11 (s, 2H), 5.90 (d, J =3.7hz, 1h), 5.29 (s, 1H), 4.54 (d, J =3.7hz, 1h), 4.35 (ddd, J =9.8,6.1,4.3hz, 1H), 4.27-3.91 (m, 8H), 3.37 (dt, J =13.8,6.9hz, 1H), 3.26 (dq, J =13.5,6.7hz, 1H), 1.88 (t, J =7.1hz, 2H), 1.73-1.51 (m, 4H), 1.49 (s, 3H), 1.43 (s, 3H), 1.37 (s, 3H), 1.32 (s, 3H) ppm. 13 C NMR(101MHz,CDCl 3 )δ169.09,156.95,134.58,129.34,128.91,127.63,126.68,126.32,123.53,118.91,112.13,109.58,106.53,105.35,82.79,82.03,80.87,72.51,68.40,67.90,67.57,38.93,29.75,28.94,27.10,26.79,26.18,25.40,23.66ppm.
And 7: n- (5- (Naphthalen-2-yloxy) pentyl) -2- (((3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetamide
Figure BDA0003863898460000603
2- (((3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d][1,3]Dioxolen-6-yl) oxy) -N- (5- (naphthalen-2-yloxy) pentyl) acetamide (238mg, 0.4 mmol) was placed in a 50mL round bottom flask followed by trifluoroacetic acid (3.6 mL), water (0.4 mL). The mixture was stirred at room temperature for 40 minutes. The progress of the reaction was monitored by TLC and after completion of the reaction the solvent was evaporated directly in vacuo. The crude product was purified by flash column chromatography on silica gel (10% methanol in DCM) to give 202mg (100%) of compound 6 as a light yellow solid. Yield: 100 percent. ESI-MS m/z 450.2131[ 2 ] M + H ] + . 1 H NMR (400 MHz, acetone-d 6 )δ7.79(dd,J=8.4,3.8Hz,3H),7.47–7.38(m,1H),7.35–7.29(m,1H),7.27(d,J=2.5Hz,1H),7.15(dd,J=9.0,2.5Hz,1H),6.61–2.80(m,16H),2.10–2.04(m,2H),1.85(p,J=6.8Hz,2H),1.66–1.47(m,4H)ppm. 13 C NMR (101 MHz, acetone) delta 171.23,157.15,134.89,129.22,128.97,127.51,126.75,126.22,123.38,118.92,106.52,97.12,85.73,83.60,83.58,76.30,75.04,74.96,72.75,72.68,71.84,71.13,70.96,70.51,67.61,61.62,38.49,38.37,23.34ppm.
Example 5
(3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl 2- (naphthalen-2-yl) acetate (Compound 15)
Figure BDA0003863898460000611
Reagents and conditions: (i) DMAP, EDCI, DCM, room temperature overnight; (ii) water, TFA, room temperature, 40 min.
Step 1: (3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d ] [1,3] dioxol-6-yl 2- (naphthalen-2-yl) acetate
Figure BDA0003863898460000612
diacetone-D-glucose (500mg, 1.9 mmol), 2- (naphthalen-2-yl) acetic acid (357mg, 1.9 mmol), DMAP (50mg, 0.4 mmol) and EDCI (420mg, 2.2 mmol) were dissolved in anhydrous DCM (15 mL). The mixture was stirred at room temperature overnight. The progress of the reaction was monitored by TLC and after completion of the reaction, part of the solvent was removed in vacuo and precipitated in water. The resulting residue was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by silica gel flash column chromatography (25% ethyl acetate/petroleum ether) to give 694mg (85%) (3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2,3-d ] [1,3] dioxol-6-yl 2- (naphthalen-2-yl) acetate as a pale yellow oil.
Step 2: (3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl 2- (naphthalen-2-yl) acetate
Figure BDA0003863898460000613
(3aR, 5R,6S, 6aR) -5- ((R) -2, 2-dimethyl-1, 3-dioxolan-4-yl) -2, 2-dimethyltetrahydrofuro [2, 3-d)][1,3]Dioxolen-6-yl 2- (naphthalen-2-yl) acetate was placed in a 50mL round bottom flask, followed by trifluoroacetic acid (3.6 mL), water (0.4 mL). The mixture was stirred at room temperature for 40 minutes. The progress of the reaction was monitored by TLC and after completion of the reaction the solvent was evaporated directly in vacuo. The crude product was purified by flash column chromatography on silica gel (10% methanol in DCM) to give 355mg (63%) of compound 15 as a light yellow solid. Total yield: and 54 percent. ESI-MS m/z 371.1093[ m ] +Na ]] + . 1 H NMR (400 MHz, acetone-d) 6 )δ7.87(td,J=8.2,7.5,4.0Hz,4H),7.50(dtd,J=14.6,7.9,7.4,3.3Hz,3H),6.75–6.11(m,1H),5.32–3.96(m,5H),3.91(s,2H),3.76–2.90(m,5H)ppm. 13 C NMR (101 MHz, acetone) delta 171.14,170.80,133.54,132.81,132.42,128.11,127.98,127.71,127.70,127.68,127.59,126.01,125.64,97.43,92.71,78.68,76.71,73.42,72.11,71.16,68.80,68.74,61.51,41.03,40.99ppm.
Example 6
3-O- (8- (quinolin-6-yloxy) octyl) -D-glucose (Compound 16)
Figure BDA0003863898460000621
Reagents and conditions: (i) 1, 8-dibromooctane, naH, bu 4 N + I - DMF, room temperature, 24 hours; (ii) K 2 CO 3 DMF,70 ℃ overnight; (iii) water, TFA, room temperature, 40 min.
Step 1:3-O- (8-bromooctyl) -1,2, 5, 6-di-O-isopropylidene- α -D-glucofuranose
Figure BDA0003863898460000622
In a 100mL round bottom flask, diacetone-D-glucose (5.0g, 19.2mmol) was dissolved in anhydrous N, N-Dimethylformamide (DMF) (25 mL), and a mineral oil solution of sodium hydride (60%) (1.2g, 28.7mmol), tetra-N-butylammonium iodide (1.0g, 2.7mmol) was added in small portions over 5 minutes under argon. The mixture was stirred in an ice-water bath for 30 minutes. 1, 8-dibromooctane (5.3 mL,28.7 mmol) was then added slowly. The mixture was allowed to warm to room temperature and stirred for 24 hours. The progress of the reaction was monitored by TLC and after completion of the reaction, part of the solvent was removed in vacuo and precipitated in water. The resulting residue was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by flash column chromatography on silica gel (8% ethyl acetate/petroleum ether) to give 4.2g (48.4%) of 3-O- (8-bromooctyl) -1, 2. The product was of sufficient purity to be used directly in the next step. ESI-MS m/z:451.43,453.46[ 2 ] M + H] + . 1 H NMR (400 MHz, chloroform-d) δ 5.80 (d, J =3.7hz, 1h), 4.46 (d, J =3.7hz, 1h), 4.24 (q, J =6.4hz, 1h), 4.12-3.97 (m, 2H), 3.91 (dd, J =8.5,6.0hz, 1h), 3.79 (d, J =3.0hz, 1h), 3.55 (dt, J =9.4,6.4hz, 1h), 3.45 (dt, J =9.3,6.5hz, 1h), 3.34 (t, J =6.8hz, 2h), 1.79 (p, J =6.9hz, 2h), 1.56-1.46 (m, 2H), 1.43 (s, 3H), 1.37 (d, J = 4.7h), 1.35-12H (m, 12 ppm). 13 C NMR(101MHz,CDCl 3 )δ111.69,108.85,105.26,82.55,82.10,81.19,72.52,70.61,67.22,33.91,32.76,29.67,29.18,28.68,28.09,26.84,26.77,26.26,25.95,25.42ppm.
Step 2:3-O- (8- (quinolin-6-yloxy) octyl) -1,2, 5, 6-di-O-isopropylidene- α -D-glucopyranose
Figure BDA0003863898460000631
In a 100mL round-bottomed flask, 3-O- (8-bromooctyl) -1, 2. The mixture was stirred at 70 ℃ for 10 hours. The progress of the reaction was monitored by TLC and after completion of the reaction, the mixture was cooled to room temperature, part of the solvent was removed in vacuo and precipitated in water. The resulting residue was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by flash column chromatography on silica gel (10% ethyl acetate/petroleum ether) to give 887mg (84.6%) 3-O- (8- (quinolin-6-yloxy) octyl) -1, 2. ESI-MS m/z:516.82[ 2 ], [ M ] +H] + . 1 H NMR (400 MHz, chloroform-d) δ 8.77 (dd, J =4.3,1.7hz, 1h), 8.05 (dd, J =8.2,1.6hz, 1h), 8.01 (d, J =9.2hz, 1h), 7.43-7.32 (m, 2H), 7.07 (d, J =2.8hz, 1h), 5.89 (d, J =3.7hz, 1h), 4.55 (d, J =3.7hz, 1h), 4.33 (dt, J =7.5,6.1hz, 1h), 4.16-4.08 (m, 4H), 4.00 (dd, J =8.5,6.0hz, 1h), 3.87 (d, J =3.1hz, 1h), 3.62 (dt, J =9.2,6.4hz, 1h), 3.53 (dt, J =9.3,6.5hz, 1h), 1.91-1.83 (m, 2H), 1.59 (t, J =6.7hz, 2h), 1.55-1.47 (m, 5H), 1.44 (s, 3H), 1.42-1.35 (m, 9H), 1.33 (s, 3H) ppm. 13 C NMR(101MHz,CDCl 3 )δ157.22,147.83,144.31,134.75,130.76,129.34,122.57,121.32,111.72,108.88,105.77,105.26,82.51,82.09,81.16,72.54,70.64,68.23,67.21,29.72,29.36,29.17,26.85,26.78,26.26,26.07,26.03,25.43ppm.
And step 3:3-O- (8- (quinolin-6-yloxy) octyl) -D-glucose (Compound 16)
Figure BDA0003863898460000632
3-O- (8- (quinolin-6-yloxy) octyl) -1,2D-Glufuranose (887mg, 1.7mmol) was placed in a 50mL round bottom flask and trifluoroacetic acid (7.2 mL) and water (0.8 mL) were added. The mixture was stirred at room temperature for 40 minutes. The progress of the reaction was monitored by TLC and after completion of the reaction, the solvent was evaporated directly in vacuo. The crude product was purified by flash column chromatography on silica gel (10% methanol/DCM) to give 725mg (98%) 3-O- (8- (quinolin-6-yloxy) octyl) -D-glucose (compound 16) as a yellow solid. ESI-MS m/z 436.2328[ m + H ]] + And 458.2145[ M ] +Na] +1 H NMR(400MHz,DMSO-d 6 )δ8.72(dd,J=4.2,1.7Hz,1H),8.23(dd,J=8.3,1.6Hz,1H),7.90(d,J=9.1Hz,1H),7.45(dd,J=8.3,4.2Hz,1H),7.42–7.30(m,2H),6.83–6.14(m,1H),5.03–4.74(m,2H),4.62–4.45(m,1H),4.44–4.22(m,1H),4.08(t,J=6.5Hz,2H),3.75–2.89(m,8H),1.77(p,J=6.8Hz,2H),1.47(dp,J=22.2,6.8Hz,4H),1.31(d,J=8.8Hz,6H). 13 C NMR(101MHz,DMSO)δ156.58,147.83,143.72,134.78,130.32,129.10,122.24,121.62,106.33,96.97,92.37,85.20,81.94,76.76,74.61,72.13,71.98,71.93,69.94,69.77,67.89,61.12,29.99,29.96,29.03,28.86,28.63,25.58ppm.
Example 7
3-O- (8-Phenoxyoctyl) -D-glucose (Compound 30)
Figure BDA0003863898460000633
The title compound was prepared in a similar manner as described in example 6. Yield: 92.6 percent. ESI-MS m/z 407.2037[ 2 ] M + Na] + . 1 H NMR(400MHz,DMSO-d 6 )δ7.38–7.21(m,2H),6.91(d,J=7.5Hz,3H),6.64–6.25(m,1H),4.93–4.74(m,2H),4.52–4.43(m,1H),4.37–4.22(m,1H),3.94(t,J=6.5Hz,2H),3.72–2.90(m,8H),1.70(t,J=7.2Hz,2H),1.49(q,J=6.8Hz,2H),1.40(t,J=7.6Hz,2H),1.31(d,J=9.0Hz,6H). 13 C NMR(101MHz,DMSO)δ158.64,129.42,120.29,114.37,96.91,92.32,85.17,81.89,76.71,74.57,72.07,71.90,69.92,69.75,67.24,61.10,29.91,28.97,28.81,28.71,25.52ppm.
Example 8
3-O- (8- (Naphthalen-2-yloxy) octyl) -D-glucose (Compound 31)
Figure BDA0003863898460000641
The title compound was prepared in a similar manner as described in example 6. Yield: 83.2 percent. ESI-MS m/z:457.2202 2[ 2 ] M + Na] + . 1 H NMR(400MHz,DMSO-d 6 )δ7.80(dt,J=8.4,3.9Hz,3H),7.44(ddd,J=8.1,6.8,1.3Hz,1H),7.39–7.27(m,2H),7.15(dd,J=9.0,2.5Hz,1H),6.64–6.24(m,1H),4.93–4.73(m,2H),4.56–4.41(m,1H),4.40–4.22(m,1H),4.07(t,J=6.5Hz,2H),3.70–2.91(m,8H),1.77(p,J=6.8Hz,2H),1.48(dp,J=22.6,6.9Hz,4H),1.32(q,J=7.3,6.3Hz,6H). 13 C NMR(101MHz,DMSO)δ156.57,134.31,129.19,128.37,127.45,126.62,126.29,123.41,118.73,106.59,96.91,92.32,85.17,81.89,76.71,74.58,72.07,71.90,71.87,69.92,69.75,67.50,61.10,29.95,29.91,28.99,28.83,28.67,25.57,25.54ppm.
The following compounds were prepared using a similar procedure to that in example 6 but using different starting materials.
Compound 29: yield: 81.6 percent; ESI-MS m/z:470.1950[ 2 ], [ M + H ] ] +1 H NMR(400MHz,DMSO-d 6 )δ8.31(dd,J=8.7,0.7Hz,1H),7.91–7.80(m,1H),7.51(d,J=8.6Hz,1H),7.48–7.37(m,2H),6.74–6.25(m,1H),5.01–4.73(m,2H),4.61–4.46(m,1H),4.45–4.23(m,1H),4.08(t,J=6.5Hz,2H),3.73–2.89(m,8H),1.77(p,J=6.7Hz,2H),1.46(dq,J=26.6,6.6Hz,4H),1.38–1.22(m,6H). 13 C NMR(101MHz,DMSO)δ157.08,147.02,142.99,138.75,129.23,128.05,123.36,122.48,106.69,96.95,92.36,85.20,81.93,76.75,74.61,72.12,71.99,71.94,69.93,69.76,68.04,61.11,29.99,29.96,29.03,28.85,28.58,25.61,25.58,25.55.
Compound 32: yield: 93.2 percent; ESI-MS m/z 536.1258[ 2 ] M + Na] +1 H NMR(400MHz,DMSO-d 6 )δ8.20(d,J=8.6Hz,1H),7.91–7.80(m,1H),7.62(d,J=8.6Hz,1H),7.44(dd,J=7.0,2.9Hz,2H),6.73–6.24(m,1H),4.93–4.74(m,2H),4.56–4.43(m,1H),4.41–4.23(m,1H),4.09(t,J=6.5Hz,2H),3.72–2.91(m,8H),1.78(p,J=6.7Hz,2H),1.47(dp,J=22.6,6.8Hz,4H),1.38–1.27(m,6H). 13 C NMR(101MHz,DMSO)δ157.10,143.79,138.26,138.24,129.26,128.19,125.81,123.29,106.79,96.92,92.32,85.17,81.89,76.71,74.57,72.08,71.91,71.87,69.91,69.74,68.04,61.09,29.94,29.91,28.98,28.79,28.53,25.56,25.53,25.50.
Compound 33: yield: 91.2 percent; ESI-MS m/z:450.2491[ deg. ] M +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.11(d,J=8.4Hz,1H),7.80(d,J=9.0Hz,1H),7.32(ddd,J=15.0,7.2,3.4Hz,3H),6.79–6.23(m,1H),5.00–4.73(m,2H),4.59–4.44(m,1H),4.42–4.19(m,1H),4.07(t,J=6.5Hz,2H),3.77–2.89(m,8H),2.60(s,3H),1.77(p,J=6.7Hz,2H),1.47(dp,J=22.8,7.5,6.8Hz,4H),1.40–1.25(m,6H). 13 C NMR(101MHz,DMSO)δ155.96,155.79,143.16,134.96,129.54,127.13,122.21,121.77,106.33,96.94,92.33,85.17,81.90,76.72,74.57,72.09,71.92,71.88,69.91,69.74,67.78,61.09,29.96,29.92,28.99,28.83,28.64,25.56,24.51.
Compound 34: yield: 88.0 percent; ESI-MS m/z:466.2432[ 2 ], [ M ] +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.12(dd,J=9.0,0.7Hz,1H),7.67(d,J=8.9Hz,1H),7.32(d,J=2.8Hz,1H),7.29(dd,J=8.9,2.8Hz,1H),6.97(d,J=8.8Hz,1H),6.66–6.25(m,1H),4.93–4.74(m,2H),4.54–4.44(m,1H),4.39–4.24(m,1H),4.04(t,J=6.5Hz,2H),3.94(s,3H),3.73–2.88(m,8H),1.84–1.70(m,2H),1.50(t,J=6.8Hz,2H),1.47–1.39(m,2H),1.39–1.25(m,6H). 13 C NMR(101MHz,DMSO)δ160.48,155.02,141.04,138.32,127.94,125.54,121.28,112.90,107.49,96.92,92.33,85.17,81.90,76.72,74.58,72.08,71.92,71.88,69.91,69.75,67.77,61.09,52.92,29.95,29.92,28.99,28.83,28.68,25.56,25.54.
Compound 35: yield: 84.0 percent; ESI-MS m/z:480.2594[ 2 ], [ M ] +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.11(d,J=8.9Hz,1H),7.64(d,J=8.9Hz,1H),7.37–7.21(m,2H),6.94(d,J=8.8Hz,1H),6.68–6.28(m,1H),4.91–4.79(m,2H),4.56–4.25(m,4H),4.03(t,J=6.5Hz,2H),3.67–2.93(m,8H),1.75(p,J=6.6Hz,2H),1.55–1.39(m,4H),1.38–1.26(m,9H). 13 C NMR(101MHz,DMSO)δ160.18,154.98,141.13,138.30,127.96,125.48,121.27,113.06,107.44,96.95,92.36,85.19,81.92,76.75,74.61,72.12,71.98,71.93,69.93,69.76,67.78,61.11,61.01,29.99,29.96,29.04,28.88,28.72,25.60,14.52.
Compound 37: yield: 65.0 percent; ESI-MS m/z:494.2742[ m ] +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.09(d,J=8.9Hz,1H),7.63(d,J=8.9Hz,1H),7.34–7.21(m,2H),6.88(d,J=8.8Hz,1H),6.68–6.28(m,1H),5.50–5.34(m,1H),4.96–4.75(m,2H),4.57–4.44(m,1H),4.42–4.23(m,1H),4.03(t,J=6.5Hz,2H),3.71–2.91(m,9H),1.75(p,J=6.5Hz,2H),1.54–1.38(m,4H),1.38–1.26(m,12H). 13 C NMR(101MHz,DMSO)δ159.69,154.92,141.17,138.28,127.93,125.32,121.22,113.51,107.42,96.94,92.36,85.19,81.92,76.75,74.60,72.11,71.97,71.93,69.93,69.76,67.77,67.14,61.11,29.99,29.95,29.03,28.87,28.72,25.59,21.92.
Compound 38: yield: 60.2 percent; ESI-MS m/z 528.2602[ m ] +H ]] +1 H NMR(400MHz,DMSO-d 6 )δ8.29(d,J=8.8Hz,1H),7.54(d,J=9.1Hz,1H),7.48–7.41(m,2H),7.38(d,J=2.8Hz,1H),7.28(dd,J=9.1,2.8Hz,1H),7.26–7.16(m,4H),6.70–6.25(m,1H),4.98–4.75(m,2H),4.57–4.47(m,1H),4.43–4.23(m,1H),4.05(t,J=6.5Hz,2H),3.70–2.89(m,8H),1.76(p,J=6.7Hz,2H),1.55–1.39(m,4H),1.37–1.25(m,6H). 13 C NMR(101MHz,DMSO)δ159.79,155.61,153.72,140.91,139.43,129.66,128.32,126.42,124.57,121.95,121.36,113.05,107.19,96.94,92.35,85.19,81.92,76.75,74.60,72.11,71.97,71.92,69.92,69.76,67.85,61.10,29.98,29.95,29.03,28.86,28.66,25.59,25.57.
Compound 39: yield: 78.6 percent; ESI-MS m/z 527.2756[ 2 ], [ M ] +H] +1 H NMR(400MHz,DMSO-d 6 )δ9.23(s,1H),8.07–7.87(m,3H),7.60(d,J=8.9Hz,1H),7.38–7.26(m,2H),7.26–7.15(m,2H),7.03(d,J=8.9Hz,1H),6.91(tt,J=7.3,1.2Hz,1H),6.72–6.26(m,1H),4.97–4.74(m,2H),4.55–4.43(m,1H),4.39–4.25(m,1H),4.03(t,J=6.5Hz,2H),3.75–2.88(m,8H),1.76(p,J=6.7Hz,2H),1.51(p,J=6.5Hz,2H),1.47–1.39(m,2H),1.39–1.24(m,6H). 13 C NMR(101MHz,DMSO)δ154.19,152.76,142.06,141.73,136.05,128.57,127.61,124.04,120.82,120.48,117.91,114.21,107.56,96.92,92.33,85.18,81.90,76.72,74.58,72.09,71.93,71.89,69.92,69.75,67.72,61.11,29.96,29.93,29.01,28.85,28.75,25.58,25.55.
Compound 40: yield: 69.0 percent; ESI-MS m/z 617.3066[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ9.22(s,1H),7.95(d,J=8.9Hz,1H),7.59(d,J=8.9Hz,1H),7.42(s,2H),7.22(dd,J=8.9,2.8Hz,1H),7.18(d,J=2.8Hz,1H),6.98(d,J=8.9Hz,1H),6.78–6.22(m,1H),4.99–4.74(m,2H),4.50(dd,J=6.8,4.2Hz,1H),4.41–4.23(m,1H),4.02(t,J=6.5Hz,2H),3.82(s,6H),3.73–2.87(m,11H),1.82–1.69(m,2H),1.57–1.48(m,2H),1.48–1.39(m,2H),1.39–1.26(m,6H). 13 C NMR(101MHz,DMSO)δ154.20,152.81,152.77,152.66,141.91,137.94,135.96,131.41,127.72,123.94,120.89,114.22,107.47,96.95,95.62,92.33,85.17,81.90,76.72,74.57,72.09,71.92,71.88,69.91,69.74,67.72,61.09,60.13,55.55,29.96,29.93,29.02,29.01,28.86,28.75,25.59,25.55.
Compound 42: yield: 78.2 percent; ESI-MS m/z 436.2327[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.71(d,J=5.2Hz,1H),8.14(dd,J=8.4,1.4Hz,1H),7.94(d,J=8.3Hz,1H),7.74(ddd,J=8.5,6.8,1.5Hz,1H),7.57(dd,J=8.2,6.8Hz,1H),7.02(d,J=5.3Hz,1H),6.77–6.25(m,1H),5.06–4.20(m,6H),3.81–2.89(m,8H),1.86(p,J=6.6Hz,2H),1.50(p,J=7.2Hz,4H),1.42–1.26(m,6H). 13 C NMR(101MHz,DMSO)δ161.34,151.98,148.90,130.24,128.89,126.20,121.96,121.23,119.24,116.26,102.01,97.38,92.79,85.59,82.34,77.17,75.03,72.54,72.40,72.36,70.34,70.19,68.85,63.23,61.51,30.42,30.38,29.43,29.25,28.77,26.01.
Compound 44: yield: 71.2 percent; ESI-MS m/z:470.1939[ m + H ]] +1 H NMR(400MHz,DMSO-d 6 )δ8.74(d,J=5.2Hz,1H),8.15(d,J=8.9Hz,1H),7.98(d,J=2.1Hz,1H),7.59(dd,J=8.9,2.2Hz,1H),7.05(d,J=5.3Hz,1H),6.72–6.25(m,1H),4.95–4.75(m,2H),4.52–4.34(m,1H),4.26(dt,J=9.7,6.5Hz,3H),3.73–2.90(m,8H),1.90–1.77(m,2H),1.55–1.42(m,4H),1.42–1.26(m,6H). 13 C NMR(101MHz,DMSO)δ160.86,153.11,149.16,134.34,127.27,126.24,123.75,119.43,102.10,96.93,85.18,81.90,76.72,74.57,72.08,71.92,71.87,69.92,69.75,68.60,61.09,29.94,29.90,28.94,28.76,28.26,25.55,25.52,25.50.
Compound 46: yield: 69.6 percent; ESI-MS m/z 514.1440[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.73(d,J=5.2Hz,1H),8.14(d,J=2.0Hz,1H),8.07(d,J=8.9Hz,1H),7.70(dd,J=8.9,2.0Hz,1H),7.06(d,J=5.3Hz,1H),6.74–6.24(m,1H),4.98–4.74(m,2H),4.58–4.16(m,4H),3.75–2.89(m,8H),1.92–1.78(m,2H),1.49(qt,J=7.5,4.7Hz,4H),1.43–1.26(m,6H). 13 C NMR(101MHz,DMSO)δ160.92,153.02,149.41,130.50,128.77,123.77,123.06,119.67,102.17,96.92,92.32,85.18,81.90,76.71,74.57,72.08,71.91,71.87,69.92,69.75,68.61,61.09,29.94,29.90,28.94,28.76,28.25,25.55,25.52,25.50.
Compound 48: yield: 89.2 percent; ESI-MS m/z 466.2442[ m + H ], [ m ], [ solution of calcium ] and calcium] +1 H NMR(400MHz,DMSO-d 6 )δ8.62(d,J=5.3Hz,1H),8.02(d,J=9.1Hz,1H),7.31(d,J=2.5Hz,1H),7.18(dd,J=9.1,2.6Hz,1H),6.87(d,J=5.3Hz,1H),6.72–6.21(m,1H),4.97–4.73(m,2H),4.57–4.12(m,4H),3.90(s,3H),3.74–2.89(m,8H),1.85(p,J=6.6Hz,2H),1.57–1.42(m,4H),1.41–1.26(m,6H). 13 C NMR(101MHz,DMSO)δ160.81,160.31,151.97,150.63,122.77,117.82,115.30,107.31,100.08,96.94,92.33,85.18,81.91,76.72,74.57,72.09,71.92,71.88,69.91,69.75,68.18,61.09,55.36,29.95,29.91,28.96,28.78,28.35,25.56,25.53.
Compound 52: yield: 46.8 percent; ESI-MS m/z 452.2287[ m + H ]] +1 H NMR(400MHz,DMSO-d 6 )δ10.07(s,1H),8.54(d,J=5.2Hz,1H),7.97(d,J=9.0Hz,1H),7.16(d,J=2.4Hz,1H),7.08(dd,J=9.0,2.4Hz,1H),6.78(d,J=5.3Hz,1H),6.73–6.17(m,1H),5.05–4.70(m,2H),4.63–4.08(m,4H),3.72–2.88(m,8H),1.83(h,J=8.5,7.5Hz,2H),1.49(h,J=7.3Hz,4H),1.41–1.27(m,6H). 13 C NMR(101MHz,DMSO)δ160.83,158.61,151.76,150.70,122.85,117.81,114.48,110.03,99.27,96.92,92.33,85.18,81.90,76.72,74.58,72.08,71.92,71.88,69.92,69.75,68.04,61.10,29.96,29.92,28.98,28.79,28.38,25.55.
Compound 53: yield: 78.2 percent; ESI-MS m/z:435.2489[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.45(dd,J=4.2,1.7Hz,1H),7.97(dd,J=8.3,1.6Hz,1H),7.68(d,J=9.1Hz,1H),7.27(dd,J=8.3,4.2Hz,1H),7.19(dd,J=9.1,2.6Hz,1H),6.80–6.23(m,2H),6.10(t,J=5.3Hz,1H),4.97–4.73(m,2H),4.50(dd,J=6.7,4.3Hz,1H),4.41–4.24(m,1H),3.69–2.92(m,10H),1.66–1.56(m,2H),1.50(p,J=6.7Hz,2H),1.40(dq,J=11.6,6.8Hz,2H),1.31(d,J=5.0Hz,6H). 13 C NMR(101MHz,DMSO)δ146.99,144.79,142.16,133.03,130.09,129.30,121.64,121.24,100.77,96.94,92.33,85.17,81.90,76.72,74.57,72.09,71.93,71.89,69.91,69.74,61.09,42.92,29.98,29.95,29.07,28.96,28.41,26.80,25.60,25.57.
Compound 54: yield: 64.8 percent; ESI-MS m/z:472.2299[ M + Na ]] +1 H NMR(400MHz,DMSO-d 6 )δ7.95(d,J=9.2Hz,1H),7.16–7.06(m,2H),7.01–6.91(m,2H),6.68–6.29(m,2H),5.61(s,2H),4.95–4.78(m,2H),4.55–4.47(m,1H),4.43–4.24(m,1H),4.03(t,J=6.5Hz,2H),3.73–2.90(m,8H),1.75(p,J=6.7Hz,2H),1.56–1.39(m,4H),1.39–1.23(m,6H). 13 C NMR(101MHz,DMSO)δ156.79,145.23,136.12,127.77,124.48,118.35,116.39,114.97,107.22,106.12,97.38,92.79,85.64,82.36,77.19,75.04,72.57,72.54,72.43,72.39,70.36,70.19,67.75,61.55,30.44,30.41,29.50,29.36,29.23,26.09,26.06,26.03.
Compound 55: yield: 86.0 percent; ESI-MS m/z 436.2323 2[ m + H ]] +1 H NMR(400MHz,DMSO-d 6 )δ8.62(d,J=2.9Hz,1H),8.00–7.91(m,1H),7.91–7.84(m,1H),7.75(d,J=2.9Hz,1H),7.60–7.52(m,2H),6.71–6.21(m,1H),4.95–4.74(m,2H),4.55–4.43(m,1H),4.41–4.23(m,1H),4.13(t,J=6.5Hz,2H),3.74–2.90(m,8H),1.80(p,J=6.8Hz,2H),1.59–1.40(m,4H),1.40–1.28(m,6H). 13 C NMR(101MHz,DMSO)δ152.17,144.26,142.80,128.69,128.57,127.00,126.97,126.49,113.20,96.92,92.33,85.17,81.90,76.72,74.58,72.08,71.92,71.88,69.91,69.75,67.99,61.10,29.95,29.92,28.98,28.80,28.47,25.56,25.54,25.48.
Compound 60: yield: 87.0 percent; ESI-MS m/z 504.2193 solution [ M + H ]] +1 H NMR(400MHz,DMSO-d 6 )δ8.86(d,J=5.2Hz,1H),8.35(d,J=8.7Hz,1H),8.27(d,J=1.8Hz,1H),7.83(dd,J=8.8,1.8Hz,1H),7.18(d,J=5.3Hz,1H),6.72–6.28(m,1H),4.98–4.76(m,2H),4.56–4.19(m,4H),3.73–2.89(m,8H),1.94–1.81(m,2H),1.50(p,J=7.2Hz,4H),1.43–1.24(m,6H). 13 C NMR(101MHz,DMSO)δ160.69,153.54,147.58,129.87( 2 J=32.3Hz),126.09( 3 J=5.1Hz),124.03( 1 J=273.7Hz),123.75,122.85,121.02,120.99,103.47,96.93,92.34,85.19,81.92,76.73,74.58,72.10,71.95,71.90,69.92,69.75,68.82,61.09,39.52,29.96,29.92,28.97,28.78,28.25,25.57,25.54,25.51.
Compound 62: yield: 29.4 percent; ESI-MS m/z:562.1308 2[ 2 ] M + H ] +1 H NMR(400MHz,DMSO-d 6 )δ8.70(d,J=5.2Hz,1H),8.34(d,J=1.6Hz,1H),7.90(d,J=8.7Hz,1H),7.84(dd,J=8.7,1.7Hz,1H),7.05(d,J=5.3Hz,1H),6.71–6.27(m,1H),4.96–4.76(m,2H),4.58–4.16(m,4H),3.73–2.89(m,8H),1.85(p,J=6.6Hz,2H),1.48(p,J=7.0Hz,4H),1.40–1.26(m,6H). 13 C NMR(101MHz,DMSO)δ160.94,152.63,149.51,136.95,134.04,123.35,119.95,102.19,96.93,96.72,92.33,85.18,81.91,76.73,74.58,72.10,71.95,71.90,69.91,69.74,68.57,61.09,29.96,29.92,28.97,28.78,28.28,25.57,25.54,25.52.
Compound 64: yield: 82.4 percent; ESI-MS m/z 514.1441[ 2 ], [ M ] +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.74(d,J=5.2Hz,1H),8.24(d,J=2.1Hz,1H),7.92–7.81(m,2H),7.07(d,J=5.3Hz,1H),6.79–6.23(m,1H),5.01–4.73(m,2H),4.60–4.14(m,4H),3.75–2.84(m,8H),1.86(p,J=6.7Hz,2H),1.48(p,J=6.8Hz,4H),1.42–1.24(m,6H). 13 C NMR(101MHz,DMSO)δ159.83,152.32,147.27,132.77,131.06,123.58,122.05,118.67,102.41,96.94,92.34,85.18,81.91,76.74,74.58,72.10,71.95,71.90,69.91,69.74,68.73,61.10,29.98,29.94,28.97,28.80,28.19,25.60,25.57,25.53.
Compound 66: yield: 78.0 percent; ESI-MS m/z 514.1446[ m ] +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.82(d,J=5.2Hz,1H),8.17(dd,J=8.3,1.3Hz,1H),8.12(dd,J=7.5,1.4Hz,1H),7.47(dd,J=8.3,7.5Hz,1H),7.13(d,J=5.3Hz,1H),6.69–6.27(m,1H),4.94–4.76(m,2H),4.55–4.17(m,4H),3.72–2.87(m,8H),1.93–1.78(m,2H),1.50(p,J=8.5,7.5Hz,4H),1.42–1.24(m,6H). 13 C NMR(101MHz,DMSO)δ161.06,152.54,145.34,133.43,126.39,123.96,122.20,121.66,102.46,96.93,92.34,85.18,81.91,76.73,74.58,72.10,71.94,71.90,69.91,69.74,68.85,61.09,29.96,29.93,28.97,28.78,28.28,25.58,25.54,25.53.
Compound 68: yield: 89.2 percent; ESI-MS m/z:496.2547, M + H] +1 H NMR(400MHz,DMSO-d 6 )δ7.86(d,J=7.6Hz,1H),7.54(s,1H),7.02(s,1H),6.76–6.27(m,1H),5.96(d,J=7.6Hz,1H),5.00–4.76(m,2H),4.58–4.17(m,4H),3.93(s,3H),3.83(s,3H),3.70–2.88(m,8H),1.73(t,J=7.5Hz,2H),1.45(q,J=6.8Hz,2H),1.35–1.16(m,8H). 13 C NMR(101MHz,DMSO)δ175.07,152.85,146.35,143.03,135.16,120.80,107.84,105.23,98.31,96.95,92.34,85.17,81.90,76.73,74.58,72.10,71.91,71.86,69.90,69.74,61.08,56.00,55.48,51.93,29.91,29.88,28.92,28.59,28.39,25.93,25.48,25.45.
Compound 69: yield: 43.6 percent; ESI-MS m/z:452.2281[ m + H ], [ solution of calcium ] and calcium] +1 H NMR(400MHz,DMSO-d 6 )δ11.62(s,1H),7.83(d,J=9.5Hz,1H),7.28–7.17(m,2H),7.13(dd,J=8.9,2.7Hz,1H),6.69–6.26(m,2H),4.95–4.75(m,2H),4.57–4.45(m,1H),4.33(dt,J=44.4,6.1Hz,1H),3.96(t,J=6.5Hz,2H),3.73–2.86(m,8H),1.71(p,J=6.7Hz,2H),1.49(q,J=6.1Hz,2H),1.44–1.36(m,2H),1.36–1.23(m,6H). 13 C NMR(101MHz,DMSO)δ161.50,153.47,139.81,133.24,122.24,119.90,119.69,116.33,110.04,96.93,92.34,85.18,81.91,76.73,74.59,72.10,71.95,71.91,69.91,69.75,67.86,61.10,29.97,29.93,29.02,28.84,28.72,25.58,25.55.
Compound 72: yield: 76.4 percent; ESI-MS m/z:514.1438[ mu ] M +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.75(d,J=2.3Hz,1H),8.55(d,J=2.2Hz,1H),7.91(d,J=9.1Hz,1H),7.43(dd,J=9.2,2.7Hz,1H),7.35(d,J=2.8Hz,1H),6.72–6.27(m,1H),5.00–4.75(m,2H),4.54–4.47(m,1H),4.33(dt,J=43.3,6.2Hz,1H),4.08(t,J=6.5Hz,2H),3.74–2.86(m,8H),1.78(p,J=6.7Hz,2H),1.54–1.47(m,2H),1.47–1.39(m,2H),1.38–1.27(m,6H). 13 C NMR(101MHz,DMSO)δ157.43,148.11,141.83,136.01,130.33,130.31,122.86,117.18,105.74,96.93,92.34,85.18,81.91,76.73,74.59,72.10,71.94,71.90,69.92,69.75,68.03,61.10,29.96,29.93,29.00,28.84,28.53,25.58,25.55,25.53.
Compound 73: yield: 89.0 percent; ESI-MS m/z 422.2166[ m + H ])] +1 H NMR(400MHz,DMSO-d 6 )δ8.72(dd,J=4.2,1.7Hz,1H),8.23(dd,J=8.4,1.6Hz,1H),7.90(d,J=9.0Hz,1H),7.46(dd,J=8.3,4.2Hz,1H),7.41–7.33(m,2H),6.78–6.25(m,1H),5.02–4.74(m,2H),4.61–4.47(m,1H),4.43–4.24(m,1H),4.09(t,J=6.5Hz,2H),3.75–2.88(m,8H),1.86–1.70(m,2H),1.58–1.48(m,2H),1.44(q,J=6.9Hz,2H),1.40–1.26(m,4H). 13 C NMR(101MHz,DMSO)δ156.57,147.82,143.71,134.76,130.32,129.08,122.22,121.61,106.32,96.96,92.35,85.19,81.92,76.74,74.59,72.11,71.92,71.88,69.92,69.75,67.89,61.10,29.94,29.90,28.77,28.59,25.60,25.57.
Compound 74: yield: 86.2 percent; ESI-MS m/z:450.2483 2[ M ] +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.72(dd,J=4.3,1.7Hz,1H),8.23(dd,J=8.4,1.6Hz,1H),7.90(d,J=9.0Hz,1H),7.46(dd,J=8.3,4.2Hz,1H),7.42–7.31(m,2H),6.71–6.27(m,1H),4.97–4.77(m,2H),4.58–4.44(m,1H),4.43–4.23(m,1H),4.09(t,J=6.5Hz,2H),3.71–2.91(m,8H),1.78(p,J=6.7Hz,2H),1.47(dt,J=18.8,7.1Hz,4H),1.38–1.25(m,8H). 13 C NMR(101MHz,DMSO)δ156.56,147.82,143.71,134.75,130.32,129.08,122.22,121.61,106.33,96.96,92.35,85.18,81.91,76.74,74.58,72.11,71.96,71.92,69.91,69.74,67.86,61.09,29.99,29.95,29.07,29.02,28.81,28.61,25.58.
Compound 78: yield: 87.5 percent; ESI-MS m/z 466.2429[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.85(d,J=2.2Hz,1H),8.76(s,1H),7.98(dd,J=9.3,1.9Hz,1H),7.49(dd,J=9.1,2.5Hz,1H),7.43(s,1H),6.66–6.23(m,1H),4.93–4.71(m,2H),4.51–4.09(m,4H),3.74–2.91(m,8H),2.50(s,3H),1.80(p,J=6.2Hz,2H),1.48(dt,J=16.3,7.2Hz,4H),1.40–1.28(m,6H). 13 C NMR(101MHz,DMSO)δ159.65,145.55,143.98,142.90,138.25,130.20,123.26,107.41,96.93,92.34,85.18,81.91,76.73,74.59,72.10,71.95,71.91,69.91,69.75,68.28,61.10,29.97,29.93,29.00,28.82,28.50,25.58,25.54.
Compound 79: yield: 93.6 percent; ESI-MS m/z:483.2362[ 2 ] M + Na] +1 H NMR(400MHz,DMSO-d 6 )δ7.65–7.55(m,4H),7.42(t,J=7.7Hz,2H),7.35–7.26(m,1H),7.06–6.96(m,2H),6.68–6.27(m,1H),4.97–4.75(m,2H),4.56–4.45(m,1H),4.41–4.22(m,1H),3.99(t,J=6.5Hz,2H),3.74–2.87(m,8H),1.78–1.64(m,2H),1.50(t,J=6.5Hz,2H),1.45–1.37(m,2H),1.36–1.25(m,6H). 13 C NMR(101MHz,DMSO)δ158.79,140.32,132.82,129.31,128.19,127.12,126.60,115.32,97.39,92.80,85.65,82.37,77.20,75.05,72.56,72.42,72.37,70.38,70.21,67.96,61.56,30.43,30.40,29.47,29.31,29.18,26.04,26.01.
Compound 82: yield: 84.7 percent; ESI-MS m/z:562.1279[ m + H ]] +1 H NMR(400MHz,DMSO-d 6 )δ7.97–7.92(m,1H),7.84(d,J=8.7Hz,1H),7.79(d,J=8.5Hz,1H),7.43–7.35(m,2H),6.69–6.25(m,1H),4.98–4.73(m,2H),4.58–4.22(m,2H),4.08(t,J=6.5Hz,2H),3.73–2.90(m,8H),1.77(p,J=6.7Hz,2H),1.56–1.47(m,2H),1.43(q,J=7.3,6.8Hz,2H),1.39–1.24(m,6H). 13 C NMR(101MHz,DMSO)δ157.03,145.02,136.79,131.90,129.43,128.26,123.02,116.19,106.74,96.92,92.34,85.18,81.90,76.73,74.58,72.09,71.94,71.90,69.91,69.74,68.02,61.09,29.96,29.92,28.99,28.82,28.56,25.57,25.54,25.53.
Compound 83: yield: 91.8 percent; ESI-MS m/z:451.1942[ 2 ], [ M ] +Na ]] +1 H NMR(400MHz,DMSO-d 6 )δ6.78(d,J=8.5Hz,1H),6.70–6.38(m,2H),6.33(dd,J=8.5,2.5Hz,1H),5.94(s,2H),4.95–4.79(m,2H),4.57–4.47(m,1H),4.42–4.23(m,1H),3.85(t,J=6.5Hz,2H),3.72–2.90(m,8H),1.71–1.59(m,2H),1.55–1.43(m,2H),1.42–1.24(m,8H). 13 C NMR(101MHz,DMSO)δ154.15,147.91,140.94,108.04,105.61,100.93,97.74,96.94,85.20,76.75,74.60,71.99,69.76,68.19,61.11,29.95,29.03,28.87,28.77,25.57,25.56.
Compound 84: yield: 82.0 percent; ESI-MS m/z 483.2349[ 2 ] M + Na] +1 H NMR(400MHz,DMSO-d 6 )δ7.70–7.61(m,2H),7.50–7.41(m,2H),7.40–7.31(m,2H),7.20(dt,J=7.6,1.2Hz,1H),7.16(t,J=2.1Hz,1H),6.92(ddd,J=8.2,2.6,0.9Hz,1H),6.71–6.27(m,1H),4.95–4.79(m,2H),4.56–4.49(m,1H),4.44–4.23(m,1H),4.02(t,J=6.5Hz,2H),3.70–2.91(m,8H),1.73(p,J=6.6Hz,2H),1.45(dq,J=30.8,6.6Hz,4H),1.37–1.26(m,6H). 13 C NMR(101MHz,DMSO)δ159.20,141.69,140.11,129.99,128.89,127.55,126.80,118.89,113.55,112.67,96.95,92.35,85.15,81.89,76.75,74.60,72.12,71.97,71.93,69.89,69.73,67.46,61.07,30.00,29.96,29.05,28.88,28.79,25.59.
Compound 85: yield: 94.2 percent; ESI-MS m/z:457.2205[ m ] +Na ]] +1 H NMR(400MHz,DMSO-d 6 )δ8.20–8.11(m,1H),7.90–7.81(m,1H),7.55–7.36(m,4H),6.94(dd,J=7.5,1.2Hz,1H),6.71–6.27(m,1H),5.00–4.77(m,2H),4.57–4.46(m,1H),4.43–4.22(m,1H),4.13(t,J=6.3Hz,2H),3.74–2.91(m,8H),1.93–1.77(m,2H),1.51(h,J=6.5Hz,4H),1.43–1.27(m,6H). 13 C NMR(101MHz,DMSO)δ154.13,134.03,127.47,126.42,126.30,125.26,124.98,121.48,119.73,105.08,96.95,92.36,85.21,81.94,76.76,74.61,72.13,72.12,72.00,71.96,69.93,69.76,67.74,61.12,30.02,29.98,29.06,28.91,28.75,25.79,25.63,25.60.
Compound 86: yield: 45.8 percent; ESI-MS m/z:447.1988[ 2 ] M + Na] +1 H NMR(400MHz,DMSO-d 6 )δ7.92(t,J=1.7Hz,1H),7.46(d,J=8.9Hz,1H),7.18–7.11(m,1H),6.91–6.83(m,2H),6.66–6.27(m,1H),4.92–4.76(m,2H),4.53–4.45(m,1H),4.41–4.24(m,1H),3.96(t,J=6.5Hz,2H),3.69–2.91(m,8H),1.72(p,J=6.8Hz,2H),1.50(p,J=6.8Hz,2H),1.41(q,J=7.1Hz,2H),1.36–1.27(m,6H). 13 C NMR(101MHz,DMSO)δ154.95,149.11,146.56,127.82,113.35,111.64,106.87,104.40,96.92,92.33,85.17,81.90,76.72,74.58,72.09,71.93,71.89,69.92,69.76,68.05,61.11,29.96,29.92,29.00,28.84,28.80,25.58,25.54.
Compound 87: yield: 52.6 percent; ESI-MS m/z:463.1766[ 2 ] M + Na] +1 H NMR(400MHz,DMSO-d 6 )δ7.83(d,J=8.8Hz,1H),7.72(d,J=5.4Hz,1H),7.40(d,J=2.3Hz,1H),7.35(d,J=5.4Hz,1H),6.98(dd,J=8.7,2.2Hz,1H),6.66–6.27(m,1H),4.95–4.74(m,2H),4.54–4.44(m,1H),4.41–4.23(m,1H),4.00(t,J=6.5Hz,2H),3.73–2.89(m,8H),1.73(p,J=6.5Hz,2H),1.45(dq,J=31.3,6.8Hz,4H),1.38–1.26(m,6H). 13 C NMR(101MHz,DMSO)δ156.46,140.66,131.21,128.31,123.80,123.16,114.81,106.51,96.93,92.34,85.18,81.90,76.73,74.59,72.09,71.93,71.90,69.92,69.76,67.69,61.11,29.97,29.93,29.01,28.85,28.74,25.57,25.55.
Compound 88: yield: 87.6 percent; ESI-MS m/z 436.2331[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.80(dd,J=4.3,1.8Hz,1H),8.26(dd,J=8.2,1.7Hz,1H),7.87(d,J=9.0Hz,1H),7.41–7.32(m,2H),7.24(dd,J=8.9,2.5Hz,1H),6.68–6.28(m,1H),5.01–4.78(m,2H),4.57–4.46(m,1H),4.43–4.22(m,1H),4.12(t,J=6.5Hz,2H),3.72–2.91(m,8H),1.78(p,J=6.6Hz,2H),1.48(dp,J=22.2,7.2,6.7Hz,4H),1.40–1.28(m,6H). 13 C NMR(101MHz,DMSO)δ159.54,150.61,149.46,135.64,129.21,123.00,119.53,119.17,107.78,96.94,85.19,76.75,74.60,71.98,69.75,67.79,61.10,29.95,29.02,28.86,28.59,25.58,25.57.
Compound 89: yield: 89.4 percent; ESI-MS m/z 475.1937[ m ] +Na] +1 H NMR(400MHz,DMSO-d 6 )δ8.00(d,J=9.6Hz,1H),7.32(d,J=9.0Hz,1H),7.27(d,J=3.0Hz,1H),7.18(dd,J=9.0,3.0Hz,1H),6.71–6.28(m,2H),4.93–4.78(m,2H),4.55–4.47(m,1H),4.41–4.25(m,1H),3.98(t,J=6.5Hz,2H),3.69–2.91(m,8H),1.72(p,J=6.6Hz,2H),1.48(q,J=6.5Hz,2H),1.45–1.36(m,2H),1.36–1.25(m,7H). 13 C NMR(101MHz,DMSO)δ160.21,155.04,147.76,144.17,119.88,119.24,117.36,116.55,111.32,96.95,92.37,85.20,81.93,76.75,74.62,72.13,71.99,71.94,69.93,69.77,68.16,61.11,29.99,29.96,29.03,28.84,28.65,25.60,25.57,25.52.
Compound 90: yield: 85.3 percent; ESI-MS m/z 507.2361 2[ M ] +Na ] +1 H NMR(400MHz,DMSO-d 6 )δ8.48(s,1H),8.37(s,1H),8.09–7.92(m,3H),7.45(dddd,J=18.8,8.0,6.5,1.3Hz,2H),7.36(d,J=2.4Hz,1H),7.18(dd,J=9.1,2.4Hz,1H),6.70–6.28(m,1H),4.99–4.77(m,2H),4.51(t,J=5.9Hz,1H),4.44–4.23(m,1H),4.32(s,1H),4.11(t,J=6.5Hz,2H),3.75–2.90(m,8H),1.80(p,J=6.8Hz,2H),1.58–1.41(m,4H),1.33(q,J=10.4,6.5Hz,6H). 13 C NMR(101MHz,DMSO)δ156.18,132.47,131.69,129.77,128.13,127.75,127.45,126.02,125.66,124.51,123.90,120.72,104.33,96.95,85.21,76.76,74.61,71.99,69.77,67.54,61.11,29.97,29.05,28.91,28.68,25.66,25.59.
Compound 92: yield: 42.5 percent; ESI-MS m/z:496.2317[ 2 ], [ M ] +Na ]] +1 H NMR(400MHz,DMSO-d 6 )δ11.24(s,1H),8.14(d,J=7.8Hz,1H),7.48–7.41(m,1H),7.37–7.25(m,2H),7.18–7.11(m,1H),7.05(d,J=8.0Hz,1H),6.68(d,J=8.0Hz,1H),6.65–6.29(m,1H),4.91–4.78(m,2H),4.51(t,J=5.9Hz,1H),4.42–4.25(m,1H),4.19(t,J=6.3Hz,2H),3.70–2.92(m,9H),1.90(p,J=6.5Hz,2H),1.64–1.46(m,4H),1.45–1.30(m,6H). 13 C NMR(101MHz,DMSO)δ155.03,141.09,138.91,126.54,124.52,122.10,121.77,118.63,111.44,110.43,103.72,100.40,96.94,85.21,81.93,76.76,74.60,72.13,72.00,69.76,67.35,61.11,29.98,29.08,28.94,28.88,25.79,25.58.
Compound 93: yield: 54.2 percent; ESI-MS m/z:531.2360[ m + [ Na ]] +1 H NMR(400MHz,DMSO-d 6 )δ8.37(d,J=9.2Hz,1H),8.24(d,J=8.5Hz,1H),8.19(ddd,J=7.6,6.2,1.1Hz,2H),8.13(d,J=9.2Hz,1H),8.09–7.93(m,3H),7.75(d,J=8.5Hz,1H),6.71–6.27(m,1H),5.00–4.77(m,2H),4.55–4.48(m,1H),4.41–4.25(m,3H),3.74–2.92(m,8H),1.93(p,J=6.5Hz,2H),1.62–1.47(m,4H),1.45–1.30(m,6H). 13 C NMR(101MHz,DMSO)δ153.17,131.71,131.58,127.78,126.87,126.77,126.50,125.42,125.11,124.93,124.80,124.63,124.59,121.29,119.81,110.27,97.39,92.81,85.66,82.39,77.21,75.05,72.57,72.45,72.40,70.38,70.22,68.98,61.56,30.46,30.42,29.51,29.36,26.22,26.08,26.05.
Compound 94: yield: 92.4 percent; ESI-MS m/z 478.2798[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.72(dd,J=4.2,1.7Hz,1H),8.23(ddd,J=8.5,1.8,0.8Hz,1H),7.90(d,J=9.0Hz,1H),7.46(dd,J=8.3,4.2Hz,1H),7.43–7.31(m,2H),6.76–6.26(m,1H),5.00–4.75(m,2H),4.58–4.45(m,1H),4.43–4.22(m,1H),4.08(t,J=6.5Hz,2H),3.72–2.89(m,8H),1.83–1.69(m,2H),1.55–1.39(m,4H),1.36–1.21(m,12H). 13 C NMR(101MHz,DMSO)δ156.57,147.84,143.72,134.78,130.33,129.10,122.24,121.64,106.33,96.94,92.36,85.20,76.75,74.60,72.12,72.01,69.92,69.76,67.86,61.11,29.98,29.13,29.10,29.08,29.06,28.83,28.62,25.62,25.59.
Compound 95: yield: 68.7 percent; ESI-MS m/z 504.2196[ 2 ], [ M ] +H ]] +1 H NMR(400MHz,DMSO-d 6 )δ8.54(d,J=8.6Hz,1H),8.05(d,J=9.2Hz,1H),7.89(dd,J=8.6,1.3Hz,1H),7.54(d,J=8.4Hz,2H),6.71–6.21(m,1H),4.95–4.75(m,2H),4.55–4.44(m,1H),4.41–4.22(m,1H),4.14(t,J=6.6Hz,2H),3.73–2.91(m,8H),1.80(p,J=6.8Hz,2H),1.47(dq,J=21.4,7.0Hz,4H),1.40–1.26(m,6H). 13 C NMR(101MHz,DMSO)δ158.33,144.02,143.69,142.39,137.47,130.74,130.38,124.41,123.19,117.17,117.16,106.18,96.93,92.34,85.18,81.91,76.72,74.59,72.09,71.93,71.89,69.92,69.76,68.20,61.10,29.96,29.92,28.99,28.81,28.49,25.57,25.54,25.51.
Compound 97: yield: 34.2 percent; ESI-MS m/z:475.1935[ M + Na ]] +1 H NMR(400MHz,DMSO-d 6 )δ7.92(d,J=9.3Hz,1H),7.47(d,J=8.7Hz,1H),7.15(d,J=10.1Hz,2H),7.04(dd,J=9.1,1.3Hz,1H),6.67–6.26(m,1H),4.95–4.74(m,2H),4.55–4.43(m,1H),4.42–4.21(m,1H),3.99(t,J=6.5Hz,2H),3.72–2.91(m,14H),1.74(p,J=6.8Hz,2H),1.47(dh,J=29.9,6.6Hz,4H),1.38–1.24(m,6H). 13 C NMR(101MHz,DMSO)δ156.27,153.27,142.86,136.22,127.13,122.48,120.95,109.68,107.21,96.92,92.33,85.17,81.90,76.72,74.58,72.09,71.93,71.89,69.92,69.75,67.64,61.10,37.76,29.97,29.93,29.01,28.86,28.78,25.60,25.55.
Compound 101: yield: 78.2 percent; ESI-MS m/z:506.1917[ 2 ], [ M + ] Na] +1 H NMR(400MHz,DMSO-d 6 )δ8.31(d,J=8.6Hz,1H),7.89–7.79(m,1H),7.52(d,J=8.6Hz,1H),7.48–7.37(m,2H),6.77–6.25(m,1H),4.98–4.77(m,2H),4.56–4.46(m,1H),4.38–4.25(m,1H),4.13–4.04(m,2H),3.67–2.90(m,8H),1.77(p,J=6.7Hz,2H),1.46(dp,J=22.6,6.9Hz,4H),1.38–1.19(m,8H). 13 C NMR(101MHz,DMSO)δ157.08,147.01,142.99,138.74,129.23,128.04,123.35,122.48,106.69,96.94,85.20,76.75,74.60,71.99,69.76,68.03,61.11,29.97,29.09,29.03,28.82,28.58,25.60,25.57.
Figure BDA0003863898460000691
Figure BDA0003863898460000701
Example 9
(3R, 4S,5S, 6R) -6- (((8- (quinolin-6-yloxy) octyl) oxy) methyl) tetrahydro-2H-pyran-2, 3,4, 5-tetraol (Compound 61)
Figure BDA0003863898460000702
Reagents and conditions: (i) Ph is 3 CCl, pyridine, 75 ℃, overnight; (ii) BnBr, naH, bu 4 N + I - DMF, room temperature, 24 hours; (iii) HBr (48% aq), acOH, ice water bath for 5 min; (iv) 1, 8-dibromooctane, naH, bu 4 N + I - DMF, room temperature, 24 hours; (v) K is 2 CO 3 DMF,70 ℃ overnight; (vi) H 2 Pb/C, methanol, room temperature, 24 hours.
Step 1: (3R, 4S,5S, 6R) -6- ((trityloxy) methyl) tetrahydro-2H-pyran-2, 3,4, 5-tetraol
Figure BDA0003863898460000703
In a 100ml round-bottom flask, D-glucose (5.0 g, 27.75mmol) was dissolved in 30ml of anhydrous pyridine, and triphenylchloromethane (8.5 g, 30.49mmol) was added to the solution. The mixture was stirred at 75 ℃ overnight. The reaction mixture was then cooled to room temperature. Pyridine was removed under reduced pressure to give a yellow gum. Dissolving the obtained precipitate in dichlorine In methane, it was washed three times with aqueous sodium bicarbonate solution. The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by flash column chromatography on silica gel (10% methanol/dichloromethane) to give 9.56g (81.5%) (3R, 4S,5S, 6R) -6- ((trityloxy) methyl) tetrahydro-2H-pyran-2, 3,4, 5-tetraol as a white solid. 1 H NMR (400 MHz, chloroform-d) delta 7.37 (t, J =8.0Hz, 6H), 7.22-6.91 (m, 9H), 5.52-4.27 (m, 4H), 3.86-3.53 (m, 2H), 3.48-2.94 (m, 5H). 13 C NMR(101MHz,CDCl 3 )δ143.75,143.56,128.72,127.93,127.90,127.15,127.07,96.36,92.31,87.17,86.78,76.20,74.45,73.40,71.96,71.41,71.21,70.30,64.09,63.54.
And 2, step: (3R, 4S,5S, 6R) -2,3,4, 5-tetrakis (benzyloxy) -6- ((trityloxy) methyl) tetrahydro-2H-pyran
Figure BDA0003863898460000711
In a 50ml round-bottom flask, compound (3R, 4S,5S, 6R) -6- ((trityloxy) methyl) tetrahydro-2H-pyran-2, 3,4, 5-tetraol (1.03g, 2.44mmol) was dissolved in 10ml anhydrous N, N-Dimethylformamide (DMF). A solution of sodium hydride (426mg, 10.65mmol) in mineral oil (60%) and tetra-n-butylammonium iodide (100mg, 0.27mmol) were then added in portions over a period of 5 minutes under argon. The mixture was stirred in an ice-water bath for 30 minutes. Benzyl bromide (2ml, 16.84mmol) was then added slowly. The mixture was allowed to warm to room temperature and stirred for 24 hours. The progress of the reaction was monitored by TLC. After the reaction was completed, the organic solvent was removed in vacuo, and a precipitate was generated in water. The resulting residue was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by flash column chromatography on silica gel (10% ethyl acetate/petroleum ether) to give 1.69g (88.7%) (3R, 4S,5R, 6R) -2,3,4, 5-tetrakis (benzyloxy) -6- ((trityloxy) methyl) tetrahydro-2H-pyran as a pale yellow oil. 1 H NMR (400 MHz, chloroform-d) δ 7.64-7.40 (m, 8H), 7.40-7.18 (m, 25H), 6.87 (dd, J =7.4,1.8hz, 2h), 5.13-4.49 (m, 8H), 4.34 (dd, J =27.3,10.4hz, 1h), 4.07-3.54 (m, 4H), 3.52-3.37 (m, 1H), 3.23 (ddd, J =31.6,10.1, 4.21h). 13 C NMR(101MHz,CDCl 3 )δ144.03,138.83,138.61,138.60,138.35,137.97,137.91,137.44,137.08,128.91,128.88,128.77,128.57,128.48,128.46,128.43,128.27,128.22,128.19,128.12,127.93,127.88,127.85,127.79,127.72,127.66,127.04,127.02,102.32,94.69,86.45,86.37,84.82,82.65,82.43,80.29,78.21,77.97,76.07,76.03,75.15,75.10,75.04,74.71,73.03,70.76,70.62,68.67,62.49.
And 3, step 3: ((2R, 3R,4S, 5R) -3,4,5, 6-tetrakis (benzyloxy) tetrahydro-2H-pyran-2-yl) methanol
Figure BDA0003863898460000712
In a 25ml round-bottom flask, compound (3R, 4S,5R, 6R) -2,3,4, 5-tetrakis (benzyloxy) -6- ((trityloxy) methyl) tetrahydro-2H-pyran (659mg, 0.84mmol) was dissolved in 3ml of acetic acid in an ice-water bath, and hydrobromic acid (48% aqueous solution, 3 ml) was added to the solution. The mixture was stirred in an ice-water bath for 4 minutes. The progress of the reaction was monitored by TLC. After completion of the reaction, the mixture was filtered through a celite pad and washed three times with dichloromethane. The filtrate was concentrated in vacuo. The crude product was purified by silica gel flash column chromatography (20% ethyl acetate/petroleum ether) to give 324mg (71.2%) ((2R, 3R,4S, 5R) -3,4,5, 6-tetrakis (benzyloxy) tetrahydro-2H-pyran-2-yl) methanol as a pale yellow oil (α -form/β -form: 35%/65%). α -form: 1 h NMR (400 MHz, chloroform-d) δ 7.41-7.26 (m, 20H), 5.01 (d, J =10.9hz, 1h), 4.89 (d, J =11.0hz, 1h), 4.84 (d, J =10.9hz, 1h), 4.80 (d, J =3.7hz, 1h), 4.71-4.61 (m, 3H), 4.55 (dd, J =12.1,4.2hz, 2h), 4.07 (t, J =9.3hz, 1h), 3.75-3.62 (m, 3H), 3.58-3.46 (m, 2H), 1.58 (s, 1H). 13 C NMR(101MHz,CDCl 3 ) δ 138.81,138.13,137.09,128.52,128.43,128.13,127.96,127.83,127.63,95.60,81.98,80.04,75.75,75.13,73.10,71.00,69.29,61.82.β -form: 1 h NMR (400 MHz, chloroform-d) δ 7.39-7.26 (m, 20H), 4.98-4.89 (m, 3H), 4.83 (dd, J =22.8,10.9hz, 2h), 4.76-4.61 (m, 3H), 4.57 (d, J =7.8hz, 1h), 3.87 (ddd, J =12.0,5.8,2.8hz, 1h), 3.74-3.64 (m, 2H), 3.57 (t, J =9.3hz, 1h), 3.49 (dd, J =9.1,7.8hz, 1h), 3.36 (ddd, J =9.6,4.7,2.8hz, 1h), 1.84 (dd, J =9.1,7, 2.8hz, 1h)=7.6,5.9Hz,1H). 13 C NMR(101MHz,CDCl 3 )δ138.51,138.31,137.97,137.28,128.52,128.43,128.40,128.17,128.10,127.97,127.91,127.74,127.69,102.86,84.56,82.37,77.58,75.76,75.10,75.07,75.03,71.70,62.10.
And 4, step 4: (3R, 4S,5S, 6R) -2,3,4, 5-tetrakis (benzyloxy) -6- (((8-bromooctyl) oxy) methyl) tetrahydro-2H-pyran
Figure BDA0003863898460000713
In a 50ml round-bottom flask, compound ((2R, 3R,4S, 5R) -3,4,5, 6-tetrakis (benzyloxy) tetrahydro-2H-pyran-2-yl) methanol (910mg, 1.68mmol) was dissolved in 15ml of anhydrous N, N-Dimethylformamide (DMF). A solution of sodium hydride (150mg, 3.75mmol) in mineral oil (60%) and tetra-n-butylammonium iodide (310mg, 0.84mmol) were then added in portions over a period of 5 minutes under argon. The mixture was stirred in an ice-water bath for 30 minutes. 1, 8-dibromooctane (645. Mu.l, 3.49 mmol) was then added slowly. The mixture was allowed to warm to room temperature and stirred for 24 hours. The progress of the reaction was monitored by TLC. After the reaction was completed, the organic solvent was removed in vacuo, and a precipitate was generated in water. The resulting residue was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by silica gel flash column chromatography (10% ethyl acetate/petroleum ether) to give 455mg (36.9%) (3R, 4S,5R, 6R) -2,3,4, 5-tetrakis (benzyloxy) -6- (((8-bromooctyl) oxy) methyl) tetrahydro-2H-pyran as a light yellow oil. 1 H NMR (400 MHz, chloroform-d) δ 7.65-7.01 (m, 20H), 5.07-4.43 (m, 9H), 4.08-3.39 (m, 8H), 3.34 (td, J =6.8,2.7hz, 2h), 1.80 (pd, J =6.9,4.4hz, 2h), 1.58 (ddt, J =14.2,10.3,6.9hz, 2h), 1.47-1.25 (m, 8H). 13 C NMR(101MHz,CDCl 3 )δ138.97,138.70,138.54,138.51,138.39,138.27,137.56,137.29,128.48,128.42,128.38,128.22,128.01,127.95,127.92,127.87,127.80,127.68,127.65,127.62,102.66,95.69,84.82,82.39,82.22,80.00,78.08,77.87,75.80,75.76,75.15,75.02,74.92,73.07,71.84,71.73,71.15,70.44,69.72,69.25,69.15,34.00,32.85,29.75,29.68,29.36,28.79,28.75,28.18,26.19.
And 5:6- ((8- (((2R, 3R,4S, 5R) -3,4,5, 6-tetrakis (benzyloxy) tetrahydro-2H-pyran-2-yl) methoxy) octyl) oxy) quinoline
Figure BDA0003863898460000721
In a 50ml round bottom flask, compound (3R, 4S,5R, 6R) -2,3,4, 5-tetrakis (benzyloxy) -6- (((8-bromooctyl) oxy) methyl) tetrahydro-2H-pyran (455mg, 0.62mmol) was dissolved in 10ml anhydrous DMF before quinolin-6-ol (136mg, 0.94mmol), potassium carbonate (172mg, 1.24mmol) were added to the solution. The mixture was stirred at 70 ℃ for 10 hours. The progress of the reaction was monitored by TLC. After completion of the reaction, the mixture was cooled to room temperature and the organic solvent was removed in vacuo. The resulting precipitate was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by flash column chromatography on silica gel (30% ethyl acetate/petroleum ether) to give 368mg (74.4%) of 6- ((8- (((2r, 3r,4s, 5r) -3,4,5, 6-tetrakis (benzyloxy) tetrahydro-2H-pyran-2-yl) methoxy) octyl) oxy) quinoline as an orange oil. 1 H NMR (400 MHz, chloroform-d) δ 8.78 (dd, J =4.3,1.7hz, 1H), 8.06-8.00 (m, 2H), 7.64-7.09 (m, 22H), 7.06 (d, J =2.8hz, 1H), 5.09-4.43 (m, 9H), 4.07 (td, J =6.4,2.3hz, 2h), 3.86-3.32 (m, 8H), 1.99-1.74 (m, 2H), 1.72-1.56 (m, 2H), 1.51 (q, J =7.2hz, 2h), 1.39 (d, J =16.3hz, 6H). 13 C NMR(101MHz,CDCl 3 )δ157.26,147.80,144.34,138.90,138.62,138.48,138.43,138.32,138.20,137.49,137.22,134.74,130.76,129.36,128.45,128.43,128.39,128.37,128.33,128.17,127.98,127.96,127.90,127.88,127.83,127.75,127.73,127.64,127.61,127.57,122.59,121.29,105.84,102.60,95.63,84.76,82.34,82.16,79.94,78.03,77.81,75.76,75.72,75.11,74.98,74.97,74.88,73.01,71.84,71.73,71.11,70.38,69.67,69.19,69.10,68.28,29.74,29.67,29.46,29.38,29.34,29.18,26.19,26.07.
Step 6: (3R, 4S,5S, 6R) -6- (((8- (quinolin-6-yloxy) octyl) oxy) methyl) tetrahydro-2H-pyran-2, 3,4, 5-tetraol (Compound 61)
Figure BDA0003863898460000722
In a 25ml round bottom flask, compound 6- ((8- (((2R, 3R,4S, 5R) -3,4,5, 6-tetrakis (benzyloxy) tetrahydro-2H-pyran-2-yl) methoxy) octyl) oxy) quinoline (368mg, 0.46mmol) was dissolved in 5ml methanol. Palladium (10%)/carbon (100mg, 0.09mmol) was then added, and the flask was carefully evacuated and purged 3 times with hydrogen using a balloon. The mixture was stirred at room temperature under an atmosphere of hydrogen (using a balloon) for 24 hours. After completion of the reaction, pd/C was filtered off using a Celite pad and the filtrate was concentrated. The crude product was purified by silica gel flash column chromatography (8% methanol/dichloromethane) to give 123mg (51.6%) (3R, 4S,5S, 6R) -6- (((8- (quinolin-6-yloxy) octyl) oxy) methyl) tetrahydro-2H-pyran-2, 3,4, 5-tetraol (Compound 61) as a white solid. ESI-MS m/z 436.2330[ 2 ], [ M + H ]] + .
Example 10
(3R, 4S,5S, 6R) -6- (hydroxymethyl) -3- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 4, 5-triol (Compound 76)
Figure BDA0003863898460000731
Reagents and conditions: (i) BnBr, naH, bu 4 N + I - DMF, room temperature, 24 hours; (ii) water, TFA, DCM, room temperature, 40 min; (iii) Dibutyl tin oxide (IV), meOH,80 ℃ for 1.5 hours, b. BnBr, K 2 CO 3 DMF,40 ℃,16 hours; (iv) 1, 8-dibromooctane, naH, bu 4 N + I - DMF, room temperature, 24 hours; (v) H 2 Pb/C, methanol, room temperature, 24 hours; (vi) K is 2 CO 3 DMF,70 ℃ overnight.
Step 1-3: (2S, 3R,4R, 5R) -2, 4-bis (benzyloxy) -5- ((R) -1, 2-bis (benzyloxy) ethyl) tetrahydrofuran-3-ol
Figure BDA0003863898460000732
Following the procedure of the previously established method, from 1, 2-O-isopropylidene-alpha-D-furanThe title compound was synthesized starting with Glucose and the characterization data matched well with previously reported data (see Patra M, awuah S G, lipprard S J et al, "Chemical Aproach to position instruments of Glucose-Platinum Conjugates receptors; scientific Cancer Targeting high Glucose-Transporter-media Uptake in Vitro and Vivo" [ J]Journal of the American Chemical Society,2016, vol 138, no. 38: pages 12541-12551). Yield: 56.2 percent. 1 H NMR (400 MHz, chloroform-d) δ 7.43-7.18 (m, 20H), 5.22 (d, J =4.6hz, 1h), 4.79 (dd, J =13.0,11.6hz, 2h), 4.70 (d, J =11.7hz, 1h), 4.62-4.46 (m, 5H), 4.36 (dd, J =8.4,4.4hz, 1h), 4.25 (ddd, J =6.2,4.6,1.9hz, 1h), 4.09-3.99 (m, 2H), 3.84 (dd, J =10.6,2.1hz, ddh), 3.68 (J, J =10.6,5.7hz, 1h), 2.96 (d, J =5.8hz, 1h). 13 C NMR(101MHz,CDCl 3 )δ138.90,138.62,137.92,137.09,128.58,128.52,128.37,128.33,128.25,128.17,128.04,127.66,127.63,127.57,127.56,127.46,127.39,100.28,83.99,77.86,76.39,76.12,73.46,72.65,71.66,71.16,70.13.
And 4-6: (2R, 3R,4S,5S, 6R) -6- (hydroxymethyl) -3- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 4, 5-triol (Compound 76)
The title compound (compound 76) was synthesized from (2s, 3r,4r, 5r) -2, 4-bis (benzyloxy) -5- ((R) -1, 2-bis (benzyloxy) ethyl) tetrahydrofuran-3-ol following an analogous procedure to that described in example 12. Yield: 51.0 percent; ESI-MS m/z 436.2328[ m ] +H] +1 H NMR(400MHz,DMSO-d6)δ8.72(dd,J=4.2,1.7Hz,1H),8.23(dd,J=8.4,1.6Hz,1H),7.90(d,J=9.1Hz,1H),7.46(dd,J=8.3,4.2Hz,1H),7.38(dd,J=9.1,2.8Hz,1H),7.35(d,J=2.8Hz,1H),6.75–6.15(m,1H),5.14–4.79(m,2H),4.77–4.44(m,1H),4.44–4.30(m,1H),4.08(t,J=6.5Hz,2H),3.74–3.37(m,5H),3.19–2.69(m,3H),1.78(p,J=6.7Hz,2H),1.53–1.40(m,4H),1.38–1.25(m,6H). 13 C NMR(101MHz,DMSO)δ156.57,147.84,143.72,134.77,130.33,129.09,122.24,121.63,106.33,96.66,89.98,83.23,80.41,76.59,76.11,72.04,71.79,71.71,70.74,70.53,69.68,67.88,61.19,61.18,29.84,29.76,28.98,28.96,28.84,28.62,25.56.
Example 11
(2R, 3S,4S, 5R) -2- (hydroxymethyl) -6- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-3, 4, 5-triol (Compound 98)
Figure BDA0003863898460000733
Reagents and conditions: (i) 1, 8-dibromooctane, naH, bu 4 N + I - DMF, room temperature, 24 hours; (ii) H 2 Pb/C, methanol, room temperature, 24 hours; (iii) K is 2 CO 3 DMF,70 ℃ overnight.
Step 1-3: (2R, 3S,4S, 5R) -2- (hydroxymethyl) -6- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-3, 4, 5-triol (Compound 98)
The title compound was synthesized from (3r, 4s,5r, 6r) -3,4, 5-tris (benzyloxy) -6- ((benzyloxy) methyl) tetrahydro-2H-pyran-2-ol (compound 98) following a similar procedure as described in example 12.
Example 12
(3R, 4R,5S, 6R) -6- (hydroxymethyl) -5- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 4-triol (Compound 99)
Figure BDA0003863898460000741
Reagents and conditions: (i) BnOH, BF 3 Diethyl ether, DCM, room temperature, overnight; (ii) NaOMe, meOH, rt, 3 hours; (iii) PhCH (OMe) 2 TsOH, DMF,80 ℃ for 4 hours; (iv) BnBr, naH, bu 4 N + I - DMF, room temperature, 24 hours; (v) Et (Et) 3 SiH, TFA, DCM, room temperature, 24 hours; (vi) 1, 8-dibromooctane, naH, bu 4 N + I - DMF, room temperature, 24 hours; (vii) H 2 Pb/C, methanol, room temperature, 24 hours; (viii) K is 2 CO 3 DMF,70 ℃ overnight.
Step 1-5: (2R, 3R,4S,5R, 6R) -4,5, 6-tris (benzyloxy) -2- ((benzyloxy) methyl) tetrahydro-2H-pyran-3-ol
Figure BDA0003863898460000742
This compound was synthesized starting from beta-D-Glucose-pentaacetate following a modified procedure of the previously established method (see Patra M, awuah S G, lipprard S J et al, "Chemical Approach to position instruments of Glucose-Platinum Conjugates Specific Cancer Targeting through Glucose-Transporter-media update in Vitro and in Vivo" [ J M, awuah S G, lipprard S J et al "[ J]Journal of the American Chemical Society,2016, vol 138, no. 38: pages 12541-12551). Yield: 44.2 percent. 1 H NMR (400 MHz, chloroform-d) δ 7.44-7.27 (m, 20H), 4.95 (t, J =11.4hz, 3h), 4.80-4.59 (m, 5H), 4.53 (d, J =7.2hz, 1h), 3.80 (dd, J =10.5,3.8hz, 1h), 3.73 (dd, J =10.4,5.3hz, 1h), 3.65-3.58 (m, 1H), 3.53-3.42 (m, 3H), 2.44 (s, 1H). 13 C NMR(101MHz,CDCl 3 )δ138.61,138.35,137.96,137.35,128.56,128.44,128.43,128.36,128.18,127.99,127.97,127.85,127.81,127.74,127.70,102.61,84.08,81.77,75.30,74.81,74.10,73.69,71.58,71.19,70.28.
Step 6: (2R, 3R,4S,5R, 6R) -2,3, 4-tris (benzyloxy) -6- ((benzyloxy) methyl) -5- ((8-bromooctyl) oxy) tetrahydro-2H-pyran
Figure BDA0003863898460000743
In a 50ml round-bottom flask, compound (2R, 3R,4S,5R, 6R) -4,5, 6-tris (benzyloxy) -2- ((benzyloxy) methyl) tetrahydro-2H-pyran-3-ol (720mg, 1.33mmol) was dissolved in 10ml of anhydrous N, N-Dimethylformamide (DMF). A solution of sodium hydride (80mg, 2mmol) in mineral oil (60%) and tetra-n-butylammonium iodide (246mg, 0.67mmol) were then added in small portions over a period of 5 minutes under argon. The mixture was stirred in an ice-water bath for 30 minutes. 1, 8-dibromooctane (490. Mu.l, 2.66 mmol) was then added slowly. The mixture was allowed to warm to room temperature and stirred for 24 hours. The progress of the reaction was monitored by TLC. After the reaction was completed, the organic solvent was removed in vacuo, and a precipitate was generated in water. The resulting residue was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. Crude productThe material was purified by flash column chromatography on silica gel (8% ethyl acetate/petroleum ether) to give 509mg (52.2%) (2R, 3R,4S,5R, 6R) -2,3, 4-tris (benzyloxy) -6- ((benzyloxy) methyl) -5- ((8-bromooctyl) oxy) tetrahydro-2H-pyran as a pale yellow oil. 1 H NMR (400 MHz, chloroform-d) δ 7.79-6.99 (m, 20H), 5.03-4.53 (m, 8H), 4.49 (d, J =7.5hz, 1h), 3.81-3.61 (m, 3H), 3.57-3.33 (m, 7H), 1.48-1.17 (m, 12H). 13 C NMR(101MHz,CDCl 3 )δ139.04,138.75,138.50,138.33,137.54,128.39,128.34,128.31,128.16,127.95,127.78,127.73,127.69,127.59,127.58,127.54,102.59,84.70,82.23,78.20,75.61,75.16,74.89,73.53,73.13,71.12,69.11,33.94,32.78,30.33,29.31,28.68,28.11,26.03.
And 7: (2R, 3R,4R,5S, 6R) -5- ((8-bromooctyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 4-triol
Figure BDA0003863898460000751
In a 25ml round bottom flask, compound (2R, 3R,4S,5R, 6R) -2,3, 4-tris (benzyloxy) -6- ((benzyloxy) methyl) -5- ((8-bromooctyl) oxy) tetrahydro-2H-pyran (509mg, 0.70mmol) was dissolved in 5ml methanol. Palladium (10%)/carbon (172mg, 0.16mmol) was then added and the flask carefully evacuated and purged 3 times with hydrogen using a balloon. The mixture was stirred at room temperature under an atmosphere of hydrogen (using a balloon) for 24 hours. After completion of the reaction, pd/C was filtered off using a Celite pad and the filtrate was concentrated. The crude product was purified by flash column chromatography on silica gel (8% methanol/dichloromethane) to give 230mg (89.0%) (2r, 3r,4r,5s, 6r) -5- ((8-bromooctyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 4-triol as a white solid. 1 H NMR(400MHz,DMSO-d 6 )δ6.74–6.14(m,1H),5.01–4.19(m,4H),3.76(dt,J=9.3,6.4Hz,1H),3.67–3.38(m,6H),3.29–2.84(m,3H),1.85–1.73and 0.92–0.81(m,2H),1.45(q,J=6.6Hz,2H),1.42–1.31(m,2H),1.26(d,J=7.3Hz,6H). 13 C NMR(101MHz,DMSO)δ97.24,92.59,78.83,78.58,77.16,76.01,75.56,73.51,73.07,72.04,72.01,71.27,61.17,35.66,32.71,31.73,30.36,30.32,30.30,29.37,29.24,29.23,29.18,28.58,27.97,26.11,26.09,26.00,25.98,22.56,14.42.
And step 8: (2R, 3R,4R,5S, 6R) -6- (hydroxymethyl) -5- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 4-triol (Compound 99)
Figure BDA0003863898460000752
In a 50ml round-bottom flask, compound (2R, 3R,4R,5S, 6R) -5- ((8-bromooctyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 4-triol (216mg, 0.58mmol) was dissolved in 10ml anhydrous DMF, and then quinolin-6-ol (488mg, 3.36mmol), potassium carbonate (512mg, 3.70mmol) were added to the solution. The mixture was stirred at 70 ℃ for 10 hours. The progress of the reaction was monitored by TLC. After completion of the reaction, the mixture was cooled to room temperature and the organic solvent was removed in vacuo. The resulting precipitate was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by silica gel flash column chromatography (10% ethyl acetate/petroleum ether) to give (2r, 3r,4r,5s, 6r) -6- (hydroxymethyl) -5- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 4-triol (compound 99) as a white solid. Yield: 46.8 percent; ESI-MS m/z 436.2331[ 2 ] M + H ] +1 H NMR(400MHz,DMSO-d 6 )δ8.72(dd,J=4.2,1.7Hz,1H),8.23(dd,J=8.4,1.6Hz,1H),7.90(d,J=9.0Hz,1H),7.46(dd,J=8.3,4.2Hz,1H),7.42–7.32(m,2H),6.72–6.18(m,1H),5.02–4.67(m,2H),4.65–4.52(m,1H),4.52–4.22(m,1H),4.08(t,J=6.5Hz,2H),3.82–3.40(m,5H),3.27–2.86(m,3H),1.77(p,J=6.8Hz,2H),1.44(p,J=6.8Hz,4H),1.39–1.24(m,6H). 13 C NMR(101MHz,DMSO)δ156.58,147.85,143.73,134.79,130.33,129.11,122.25,121.64,106.33,96.80,92.17,78.38,78.13,76.71,75.57,75.10,73.07,72.62,71.64,70.84,67.88,60.72,29.93,29.92,28.95,28.84,28.62,25.64,25.63,25.57.
Example 13
(2R, 3S,4S, 5R) -2- (hydroxymethyl) -6- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-3, 4, 5-triol (Compound 98)
Figure BDA0003863898460000753
The title compound was prepared in a similar manner as described in example 12. Yield: 52.4 percent; ESI-MS m/z 436.2326[ m + H ]] +1 H NMR (400 MHz, methanol-d) 4 )δ8.66(d,J=4.1Hz,1H),8.26(d,J=8.3Hz,1H),7.91(d,J=9.2Hz,1H),7.47(dd,J=8.4,4.3Hz,1H),7.41(dd,J=9.2,2.8Hz,1H),7.28(d,J=2.7Hz,1H),4.25(d,J=7.8Hz,1H),4.12(t,J=6.4Hz,2H),3.94–3.84(m,2H),3.67(dd,J=12.1,4.9Hz,1H),3.54(dt,J=9.6,6.8Hz,1H),3.35(t,J=8.0Hz,1H),3.27(t,J=5.9Hz,2H),3.17(t,J=8.4Hz,1H),1.86(p,J=6.7Hz,2H),1.63(q,J=6.9Hz,2H),1.53(q,J=7.1Hz,2H),1.42(dd,J=11.0,5.3Hz,6H). 13 C NMR(101MHz,MeOD)δ158.97,148.39,144.66,137.17,131.22,130.31,124.23,122.68,107.19,104.37,78.14,77.92,75.13,71.68,70.86,69.46,62.79,30.77,30.52,30.46,30.27,27.15,27.05.
Example 14
(3R, 4S,5S, 6R) -3- ((8- ((2-chloroquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 4, 5-triol (Compound 102)
Figure BDA0003863898460000754
The title compound was prepared in a similar manner as described in example 10. Yield: 68.2 percent; ESI-MS m/z 492.1754[ m ] +Na] +1 H NMR(400MHz,DMSO-d 6 )δ8.31(d,J=8.5Hz,1H),7.93–7.78(m,1H),7.52(d,J=8.6Hz,1H),7.48–7.36(m,2H),6.69–6.15(m,1H),5.13–4.78(m,2H),4.75–4.34(m,2H),4.08(t,J=6.5Hz,2H),3.73–3.38(m,6H),3.15–2.90(m,2H),1.77(p,J=6.7Hz,2H),1.52–1.39(m,4H),1.37–1.26(m,6H). 13 C NMR(101MHz,DMSO)δ157.07,147.01,142.99,138.74,129.23,128.04,123.34,122.47,106.69,96.66,89.97,83.23,80.40,76.59,76.11,72.04,71.79,71.70,70.74,70.53,69.68,68.03,61.19,29.75,28.96,28.82,28.57,25.56,25.53.
Example 15
(3R, 4S,5S, 6R) -6- (hydroxymethyl) -3- ((9- (quinolin-6-yloxy) nonyl) oxy) tetrahydro-2H-pyran-2, 4, 5-triol (Compound 103)
Figure BDA0003863898460000761
The title compound was prepared in a similar manner as described in example 10. Yield: 56.0 percent; ESI-MS m/z 450.2510[ 2 ], [ M ] +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.72(dd,J=4.2,1.7Hz,1H),8.23(ddd,J=8.4,1.7,0.8Hz,1H),7.90(d,J=9.0Hz,1H),7.46(dd,J=8.3,4.2Hz,1H),7.42–7.33(m,2H),6.71–6.13(m,1H),5.11–4.77(m,2H),4.73–4.26(m,2H),4.09(t,J=6.5Hz,2H),3.74–3.38(m,5H),3.20–2.67(m,3H),1.78(p,J=6.7Hz,2H),1.51–1.40(m,4H),1.38–1.26(m,8H). 13 C NMR(101MHz,DMSO)δ156.56,147.83,143.71,134.76,130.32,129.08,122.22,121.62,106.33,96.64,89.96,83.22,80.40,76.58,76.10,72.02,71.77,71.70,70.72,70.52,69.68,67.87,61.17,29.84,29.76,29.06,28.98,28.95,28.80,28.62,25.58.
Example 16
(4R,5S,6R) -6- (hydroxymethyl) -4- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 5-diol (Compound 100)
Figure BDA0003863898460000762
Reagents and conditions: (i) Ac 2 O, pyridine, room temperature, overnight; (ii) BnOH, BF 3 Diethyl ether, DCM, room temperature, overnight; (iii) NaOMe, meOH, room temperature, 3 hours; (iv) PhCH (OMe) 2 TsOH, DMF,80 ℃,4 hours; (v) 1, 8-dibromooctane, naH, bu 4 N + I - DMF, room temperature, 24 hours; (vi) K 2 CO 3 DMF,70 ℃ overnight; (vii) H 2 Pb/C, methanol, room temperature, 24 hours.
Step 1: (4R, 5S, 6R) -6- (acetoxymethyl) tetrahydro-2H-pyran-2, 4, 5-triacetic acid triester
Figure BDA0003863898460000763
In a 100ml round-bottom flask, 2-deoxy-D-glucose (2.10g, 12.79mmol) was dissolved in 20ml of pyridine. Acetic anhydride (11.52ml, 121.85mmol) was then added gradually and the mixture was stirred at room temperature under a nitrogen atmosphere overnight. The progress of the reaction was monitored by TLC. After completion of the reaction, pyridine was evaporated in vacuo. The resulting precipitate was diluted with dichloromethane, neutralized with sodium bicarbonate, washed with water and aqueous sodium chloride, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by silica gel flash column chromatography (33% ethyl acetate/petroleum ether) to give 4.154g (97.7%) (4R, 5S, 6R) -6- (acetoxymethyl) tetrahydro-2H-pyran-2, 4, 5-triacetic acid triester as a pale yellow oil (α -form/β -form: 25%/75%). 1 H NMR (400 MHz, chloroform-d) δ 6.26 (dd, J =3.7,1.5hz,1H for α), 5.79 (dd, J =9.9,2.3hz,1H for α 0), 5.32 (ddd, J =11.6,9.5,5.3hz,1H for α 2), 5.15-4.98 (m, 1H for α 3 and 2H for α 1), 4.31 (dt, J =12.9,4.9hz,1H for α 5and 1H for α 4), 4.17-4.00 (m, 3H for α 7and 2H for α 6), 3.75 (d, J =9.4,4.6,2.2hz,1H for α 8), 2.35 (ddd, J =12.6,4.9,2.3H for α 9, 3H for β 0), 2.13H for α 0, 13H for β 2, 13H for β 13H, 3H for α 2, 13H for α 0, 13H for α 2, 13H for β 13, 13H for α 8, 2H for β 13H, 3H for α 2, 13H. 13 C NMR(101MHz,CDCl 3 )δ171.10,170.68,170.24,170.07,169.72,169.67,168.91,168.77,91.07,90.89,72.87,70.22,70.14,68.71,68.47,68.29,61.98,61.95,34.72,33.87,21.01,20.93,20.91,20.84,20.75,20.71,20.68.
Step 2: (2R, 3S,4R, 6R) -2- (acetoxymethyl) -6- (benzyloxy) tetrahydro-2H-pyran-3, 4-diacetic acid diester
Figure BDA0003863898460000771
In a 250ml round bottom flask, (4R, 5S, 6R) -6- (acetoxymethyl) tetrahydro-2H-pyran-2, 4, 5-triacetoxy triester (4.154g, 12.50mmol) was dissolved in 50ml dichloromethane. Then benzyl alcohol (3.0ml, 28.85mmol) and BF were added 3 -Et 2 O (2.5ml, 20.25mmol). The mixture was stirred at room temperature under a nitrogen atmosphere overnight. With 5% carbonic acidThe reaction was quenched with aqueous sodium bicarbonate. The organic layer was separated, washed with aqueous sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by silica gel flash column chromatography (20% ethyl acetate/petroleum ether) to afford 4.342g (91.3%) (2r, 3s,4r, 6r) -2- (acetoxymethyl) -6- (benzyloxy) tetrahydro-2H-pyran-3, 4-diacetic acid diester as a light yellow oil. 1 H NMR (400 MHz, chloroform-d) δ 7.40-7.30 (m, 5H), 5.36 (ddd, J =11.5,9.5,5.3hz, 1h), 5.07-4.98 (m, 2H), 4.68 (d, J =12.0hz, 1h), 4.51 (d, J =12.0hz, 1h), 4.36-4.25 (m, 1H), 4.06-3.93 (m, 2H), 2.29 (ddd, J =12.9,5.4,1.2hz, 1h), 2.11 (s, 3H), 2.04 (s, 3H), 2.01 (s, 3H), 1.85 (ddd, J =13.1,11.7,3.8hz, 1h). 13 C NMR(101MHz,CDCl 3 )δ170.73,170.17,169.90,137.09,128.49,127.97,127.95,96.21,69.42,69.31,69.13,68.06,62.35,35.01,20.97,20.78,20.73.
And step 3: (2R, 3S,4R, 6R) -6- (benzyloxy) -2- (hydroxymethyl) tetrahydro-2H-pyran-3, 4-diol
Figure BDA0003863898460000772
In a 250ml round bottom flask, (2R, 3S,4R, 6R) -2- (acetoxymethyl) -6- (benzyloxy) tetrahydro-2H-pyran-3, 4-diacetic acid diester (4.280g, 11.25mmol) was dissolved in 100ml methanol. Sodium methoxide (120mg, 2.22mmol) was then added. The mixture was stirred at room temperature for 3 hours. The organic solvent was concentrated in vacuo. The resulting precipitate was purified by flash column chromatography on silica gel (10% methanol/dichloromethane) to give 2.686g (93.9%) (2R, 3S,4R, 6R) -6- (benzyloxy) -2- (hydroxymethyl) tetrahydro-2H-pyran-3, 4-diol as a white solid. 1 H NMR(400MHz,DMSO-d 6 )δ7.39–7.26(m,5H),4.97–4.89(m,1H),4.87(d,J=5.3Hz,1H),4.76(d,J=4.9Hz,1H),4.64(d,J=11.9Hz,1H),4.49(t,J=6.0Hz,1H),4.38(d,J=11.9Hz,1H),3.67(dddd,J=17.3,13.7,8.0,3.6Hz,2H),3.50(dt,J=11.7,5.9Hz,1H),3.42(ddd,J=9.8,5.8,2.1Hz,1H),3.07(td,J=9.2,5.3Hz,1H),1.94(ddd,J=13.1,5.1,1.3Hz,1H),1.50(ddd,J=13.0,11.5,3.6Hz,1H). 13 C NMR(101MHz,DMSO)δ138.59,128.71,128.12,127.87,96.40,73.91,72.22,68.54,68.01,61.55,38.31.
And 4, step 4: (4aR, 6R,8R, 8aS) -6- (benzyloxy) -2-phenylhexahydropyrano [3,2-d ] [1,3] dioxin-8-ol
Figure BDA0003863898460000773
In a 100ml round-bottom flask, compound (2R, 3S,4R, 6R) -6- (benzyloxy) -2- (hydroxymethyl) tetrahydro-2H-pyran-3, 4-diol (2.604g, 10.24mmol) was dissolved in 25ml DMF. Benzaldehyde dimethyl acetal (2.1ml, 13.07mmol) and p-toluenesulfonic acid (500mg, 2.63mmol) were then added. The mixture was stirred at 80 ℃ for 4 hours. The progress of the reaction was monitored by TLC. After completion of the reaction, the mixture was cooled to room temperature and then concentrated in vacuo. The resulting precipitate was diluted in dichloromethane, washed with aqueous sodium bicarbonate, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by flash column chromatography on silica gel (20% ethyl acetate/petroleum ether) to give 1.263g (36.0%) (4aR, 6R,8R, 8aS) -6- (benzyloxy) -2-phenylhexahydropyrano [3, 2-d) ][1,3]Dioxin-8-ol, which is a white solid. 1 H NMR (400 MHz, chloroform-d) δ 7.54-7.27 (m, 10H), 5.57 (s, 1H), 5.01 (dd, J =3.8,1.1hz, 1h), 4.70 (d, J =11.9hz, 1h), 4.48 (d, J =11.9hz, 1h), 4.24 (tdd, J =7.9,6.5,4.1hz, 2h), 3.87 (td, J =9.8,4.8hz, 1h), 3.75 (t, J =10.3hz, 1h), 3.49 (t, J =9.2hz, 1h), 2.27 (ddd, J =13.3,5.2,1.2hz, 2ddh), 1.81 (J =13.3,11.3, 3.9h). 13 C NMR(101MHz,CDCl 3 )δ137.42,137.30,129.24,128.49,128.36,127.89,127.87,126.25,102.05,97.27,83.92,69.21,69.06,65.96,62.88,37.36.
And 5-7: (4R,5S,6R) -6- (hydroxymethyl) -4- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 5-diol (Compound 100)
Figure BDA0003863898460000781
The title compound (compound 100) was synthesized from (4 aR,6R,8R, 8aS) -6- (benzyloxy) -2-phenylhexahydropyrano [3,2-d ] [1,3] dioxin-8-ol according to a similar procedure to that described in example 9.
Example 17
Protein expression and purification
Synthesis of full-Length wild-type PfHT codon-optimized cDNA from Plasmodium falciparum and subcloning it into a plasmid with N-terminal His 10 The modified pFastBac1 vector of the tag (Invitrogen). Recombinant PfHT and mutants were expressed using the pFastBac baculovirus system (Invitrogen). Bacmid was generated in DH10Bac cells, followed by transfection of Sf9 insect cells to generate and amplify baculovirus.
72 hours after viral infection, sf9 insect cells were harvested and resuspended in a mixture containing 25mM MES pH 6.0, 150mM NaCl and protease inhibitors (aprotinin 5. Mu.g mL) -1 1. Mu.g/mL of pepstatin -1 And leupeptin 5. Mu.g mL -1 (ii) a Amresco). The overexpressed PfHT and mutants were extracted with 2% (w/v) n-dodecyl- β -D-maltopyranoside (DDM, anatrace) at 4 ℃ for 2 hours. Cell membrane debris was pelleted by high speed (18,700g) centrifugation at 4 ℃ for 30 minutes and the supernatant was treated with Ni at 4 ℃ 2+ Nitrilotriacetate affinity resin (Ni-NTA, qiagen) incubation for 30 min. The resin was washed with a buffer containing 25mM MES pH 6.0, 150mM NaCl, 30mM imidazole and 0.02% (w/v) DDM. The protein was eluted with wash buffer plus 270mM imidazole. For crystallization, wild-type PfHT was concentrated to 15mg mL -1 And using 10mg mL at 18 DEG C -1 Proteinase K (Sigma) was digested for 30 min at a protein to proteinase K ratio (v/v) of 32. The proteolytic reaction was terminated by addition of 1mM PMSF prior to size exclusion chromatography (Superdex 200Increatase 10/300GL column, GE Healthcare) pre-equilibrated with a buffer containing 25mM MES pH 6.0, 150mM NaCl and the selected detergent. Peak fractions were collected and flash frozen in liquid nitrogen for further experiments. For biochemical analysis, neither wild-type PfHT nor the mutant was directly applied to gel filtration without proteinase K digestion.
Example 18
Crystallization of PfHT-compound 3361 complex
Purified wild-type PfHT was preincubated with 50mM compound 3361 for 1 hour at 4 ℃ and then with glycerol monooleate (Sigm)a) Protein to lipid ratio (w/w) of 2. Then, 45nL of the intermediate phase was mixed with 400nL of well buffer under each condition on a glass sandwich plate using an LCP crystallization robot (Gryphon, art Robbins Instruments). In the presence of 0.1M NH 4 Cl, 0.1M sodium citrate pH 5.2 and 28% PEG500MME, crystals suitable for X-ray data collection were obtained at 20 ℃. Crystals were collected using MicroMesh (M3-L18 SP-50 MiTeGen) and immediately snap frozen in liquid nitrogen.
Example 19
Data collection and structure determination
The crystals were tested at the Shanghai Synchrontron Radiation Facility beam line station BL18U and diffraction data were collected at the SPring-8 beam line BL32 XU. All datasets were integrated and scaled using XDS, and then anisotropic analysis was performed by a UCLA diffraction anisotropy server to generate anisotropic truncated datasets. Further processing was performed using the CCP4 kit. This phase was resolved by molecular replacement using PHASER and using a modified GLUT3 structure (PDB code: 4ZW 9), the residues of which were modified to the corresponding sequence of PfHT with CHAINSAW in the CCP4 suite as a search model. The structural model was reconstructed in COOT and refined using pheenix. Fig. 2 shows the atomic structure coordinates of a PfHT polypeptide having SEQ ID NO:1 complexed to compound 3361, as obtained by X-ray diffraction of crystalline PfHT-compound 3361 complex. Fig. 3 shows statistics of data collection and structure refinement. Fig. 4 shows an overall view of the complex structure of the PfHT-compound 3361 complex.
Example 20
Rational design of molecules based on computational-assisted structures
All compounds designed to bind pockets allosterically are delivered by
Figure BDA0003863898460000782
Suite 2018-1 interfaces with PfHT. All compounds were constructed using Maestro and the ligaprep program was used to switch the compounds from 2D to 3D structures under OPLS3 force field. Using the coordinates of PfHT-glucose complex as input in the protein preparation guideProcessing of protein structures, including the addition of hydrogen atoms, minimizes the retained energy of the protein structure with an optimized potential-full atomic (OPLS-AA) force field simulated with a liquid. Molecular docking was performed by Glide program using ultra-precision slide docking (Glide XP).
For the results, the poses of a given ligand were ranked using the Emodel score. For the Glide XP score, it includes all hydrophobic outer shells, hydrogen bonding interactions, internal energies (such as van der waals interactions), electrostatic interactions, and two XP penalties (i.e., a desolvation penalty and a ligand-strain penalty).
Example 21
Binding affinity
Method
Micro-calorimetry (MST) analysis was performed using a Nano-tester Monolith nt. The PfHT polypeptide having the amino acid sequence of SEQ ID NO. 1 was purified in a buffer containing 25mM MES pH 6.0, 150mM NaCl and 0.06% (w/v) Cymal-6. The different concentrations of test compound were then incubated with PfHT polypeptide for 2 minutes at room temperature and then loaded into MO-Z002 capillaries. MST measurements were carried out at 10% LED power and 60% MST power. Data sets were analyzed using mo. Affinity Analysis v2.2.4 (Nano Temper Technologies GmbH).
Example 22
Mediated by PfHT and GLUT1 3 Inhibition of H-D-glucose uptake
Method
I. Preparation of liposomes and proteoliposomes.
Coli (E.coli) polar lipid extract (Avanti) was purified in KPM 6.5 buffer (50 mM potassium phosphate pH 6.5,2mM MgSO 4 ) And 50mM D-glucose to 20mg mL -1 . After incubation with 1% n-octyl- β -D-glucopyranoside (. Beta. -OG, anarrace) for 30 minutes at 4 deg.C, liposomes were incubated with 200. Mu.g mL -1 The indicated proteins (PfHT or GLUT 1) were mixed and incubated for a further 1 hour at 4 ℃. beta-OG was removed by overnight incubation with 400mg/mL Bio-Beads SM2 (Bio-Rad). The proteoliposomes were frozen and thawed 5 times in liquid nitrogen and passed through a 0.4 μm membrane filter (Millipo)re) extrusion. To remove the remaining glucose, the homogenized proteoliposomes were ultracentrifuged at 100,000g for 1 hour and the pellet was washed twice with ice-cold KPM 6.5 buffer. Finally, the lipoprotein body was resuspended to 100mg mL with ice-cold KPM 6.5 buffer -1 For countercurrent measurement.
Countercurrent assay
The countercurrent measurement is carried out at 25 ℃. For each assay, 2. Mu.L of 100mg mL -1 The 50mM D-glucose-preloaded proteoliposome of (a) was mixed with 100. Mu.L of KPM 6.5 buffer containing 1. Mu. Ci of D2- 3 H]Glucose (Perkinelmer), corresponding to a specific radioactivity of 23.4Ci mmol -1 0.427 μ M D2- 3 H]-glucose. After 30 seconds, the mixed solution was filtered with a 0.22 μm membrane filter (Millipore). The filter was washed with ice cold KPM 6.5 buffer. The membrane filters were placed in vials containing Optiphase hisake 3 (PerkinElmer) and incubated overnight, and radioactivity was quantified by liquid scintillation counting using MicroBeta JET (PerkinElmer).
The time course experiment shows that D2- 3 H]The accumulation of glucose was approximately linear within the first 30 seconds. K for determining transport of D-glucose by PfHT m And V max The initial velocity was measured at 15 seconds. The glucose concentration in the external solution is adjusted using a non-radiolabeled substrate. In GraphPad prism8.0, the data were fitted to the michaelis equation V = (V) max [ D-glucose ]])/(K m + [ D-glucose])。
To perform transport assays using test compounds, the proteoliposomes were preincubated with different concentrations (25 μ M and 100 μ M, respectively) of test compounds on ice for 30 minutes. Then 2. Mu.L of the proteoliposomes were added to 98. Mu.L of KPM buffer containing 1. Mu. Ci of D2- 3 H]Glucose and test compounds (25. Mu.M and 100. Mu.M, respectively). The remainder of the protocol is the same as described above. To determine the IC of a test Compound 50 The concentration of test inhibitor was varied as indicated.
All experiments were repeated three times. All data were processed in GraphPad Prism 8.0. Data are expressed as relative activity normalized to radioactivity, 100% without test compound and 0% with empty liposomes. Error bars indicate standard deviation.
Results
The compounds synthesized in examples 1-16 were tested at a concentration of 100. Mu.M. As shown in fig. 5, at 100 μ M concentration, exemplary compounds 16, 30, and 31 significantly inhibited PfHT-mediated D-glucose uptake, while compounds 16, 30, and 31 reduced GLUT 1-mediated D-glucose uptake by less than 10%. This indicates that compounds 16, 30 and 31 are highly selective inhibitors of PfHT over its human ortholog GLUT 1.
The results of D-glucose uptake (results not shown) for other compounds of the present disclosure were comparable or even better than those of exemplary compounds 30 and 31.
Table 2 shows the IC's of exemplary compounds of the present disclosure, MMV009085 (the only reported PfHT1 inhibitor in the malaria drug service (MMV) database, see Kraft, T.E. et al, "A novel fluorescence sensitivity energy transfer-based detector in high-through product formation to identification inhibitors of malarial and human glucose transporters," anti-microbial Agents Chemothers, 2016, vol.60, pp.7407-7414), and Compound 3361 50 The value is obtained.
TABLE 2
Figure BDA0003863898460000791
Figure BDA0003863898460000801
As can be seen from table 2, exemplary compounds of the present disclosure exhibit inhibitory activity against PfHT.
Other compounds of the present disclosure, whose results are not shown, exhibit comparable or even better inhibitory activity against PfHT than the exemplified compounds.
Example 23
In vitro plasmodium falciparum blood stage culture and inhibition assay
Method
Plasmodium falciparum Dd2 strain (which is a multidrug resistant strain, e.g., chloroquine resistant) and 3D7 strain (which is a drug susceptible strain) were gifted by the shanghai pasteur institute of china academy of sciences. At 1% of 2 、5%CO 2 The parasites were cultured in complete medium (10.4 g/L RPMI 1640,5.94g/L HEPES,5g/L albumax II, 50mg/L hypoxanthine, 2.1g/L sodium bicarbonate and 25mg/L gentamicin) at 37 ℃ in human blood.
For in vitro potency assays, test compounds in DMSO were printed together with control compounds (quinine and DHA) into 384-well black transparent plates by Tecan D300e digital dispensers. Parasite suspensions of 1.0% parasitemia and 0.8% hematocrit in culture medium were dispensed into assay plates. The assay plates were incubated at 37 ℃ for 72 hours. mu.L of a detection reagent consisting of 10 XSSYBR Green I (Invitrogen; provided at 10,000. Times. Concentration) in lysis buffer (20 mM Tris-HCl, 5mM EDTA, 0.16% (w/v) saponin, 1.6% (v/v) Triton X-100) was dispensed to the assay plate. To obtain optimal staining, the assay plates were left at room temperature for 24 hours in the dark. The assay plates were read in Envision (PerkinElmer) with 485nm excitation and 530nm emission settings. EC was determined using nonlinear regression curve fitting in Prism software version 8 (GraphPad) 50 The value is obtained. The reported values are the result of two technical replicates and at least three biological replicates.
Results
The results of potency assays for exemplary compounds of the present disclosure, MMV009085, reference compound 1 (oct-7-en-1-yl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate), and compound 3361 as a positive control are shown in table 3.
Example 24
Cytotoxicity assays of mammalian cells
Method
Mammalian cells (human renal epithelial cell line HEK293T/17 and human hepatic epithelial cell line HepG 2) were obtained from ATCC and were rated at 5% 2 In an atmosphere containing 10% of FBS (Gibco) and 1% (v/v) of Penicillium at 37%The hormone-streptomycin (Thermo Fisher) was cultured routinely in DMEM.
For cytotoxicity assays, test compounds in DMSO were printed together with control compounds (puromycin) by Tecan D300e digital dispensers into 384-well white solid plates.
HEK293T/17 cells or HepG2 cells were seeded at 2,000 cells/well and 2,500 cells/well, respectively, into assay plates and incubated for 72 hours. Viability was measured in Envision (PerkinElmer) with US LUM 384 using Cell-Titer Glo (Promega). CC's were generated using Prism software version 8 (GraphPad) 50 The values were fitted to a nonlinear regression curve. The reported values are the result of two technical replicates and at least three biological replicates.
As a result, the
The cytotoxicity assay results for exemplary compounds of the present disclosure, MMV009085, reference compound 1 and compound 3361 are shown in table 3. As can be seen from table 3, the compounds of the present disclosure exhibit high potency and mild toxicity, providing a wide therapeutic window.
TABLE 3
Figure BDA0003863898460000811
Figure BDA0003863898460000821
Figure BDA0003863898460000831
Example 25
Extracellular flux analysis of Plasmodium falciparum Using XFP Analyzer
Method
All assays were performed according to the manufacturer's manual with some modifications. The sensor cartridge was hydrated overnight at 37 ℃ in XF calibration solution. Two assay media were used for parasite analysis: RPMI 1640 medium (Thermo Fisher Scientific, containing 11mM glucose) and Seahorse XF RPMI medium (Agilent Technologies). Injection solutions containing the test compound at a final concentration of 10 x were prepared in assay medium and loaded into the reagent delivery chamber of the sensor (20 μ L, 22 μ L, 24.5 μ L and 27 μ L for the first, second, third and fourth injections, respectively). Late Dd2 parasites in RBCs were magnetically purified from 5% sorbitol sync cultures using MACS LD columns (Miltenyi Biotec) and seeded at 80 million RBCs/well in Seahorse microplates pre-coated with Cell-Tak cells and tissue adhesive (Corning). The ring stage Dd2 parasites in RBCs were obtained at approximately 20% parasitemia by overnight culturing MACS purified schizonts with small amounts of fresh blood and inoculating with 80 million RBCs per well. Free merozoites were prepared by saponin lysis (see Sakata-Kato, T. And Wirth, D.F. "A Novel method for Bioenergetic Analysis of plasmid falciparum revealals a Glucose-Regulated Metabolic Shift and Enables model of Action analytes of mitochon drial inhibitors." ACS infection Dis, vol.2, pp.903-916 (2016), and seeded with 250 ten thousand RBC/well. After centrifugation at 500rpm for 5 minutes with slow acceleration and no brake, the assay medium was added to all wells (180. Mu.L as final volume) and the microplate was loaded into the flux analyzer to start the measurement (mixing time: 30 seconds; waiting time: 1 minute 30 seconds; measuring time: 3 minutes). In the assay plate, two wells were used for background correction.
As a result, the
To assess whether the compounds provided herein disrupt glycolytic activity of plasmodium falciparum at a blood stage, glycolysis and mitochondrial respiration in living cells were simultaneously monitored by extracellular acidification rate (ECAR) and Oxygen Consumption Rate (OCR), respectively, using a Seahorse extracellular flux analyzer. The Dd2 schizont stage parasites in Red Blood Cells (RBC) were inoculated in glucose-free medium and exposed to either glucose (final concentration of 11 mM) or fructose (final concentration of 40 mM) at 15 minutes (first vertical dashed line), resulting in a significant increase in ECAR (fig. 6). This increased ECAR was eliminated by adding exemplary compound 29 and 2-deoxyglucose, a well-known glycolytic inhibitor, at 61 minutes (second vertical line), clearly demonstrating the glycolytic inhibitory activity of compound 29. It should be noted that 2-DG requires a higher concentration (50 mM) than compound 29 (20. Mu.M) to reduce the increased ECAR.
Extracellular flux analysis showed that exemplary compound 29 and compound 76 inhibited glycolytic activity in a dose-dependent manner on the early (ring stage) and late (trophozoite/schizont stage) of parasites in RBCs and parasites released late from RBCs (fig. 7). Pfdd2 parasites were inoculated into assay medium containing glucose (11 mM) and compound 29 or compound 76 was added four times in sequence to final concentrations of 0.4. Mu.M, 1. Mu.M, 2.5. Mu.M and 6.75. Mu.M or 0.2. Mu.M, 0.5. Mu.M, 1.25. Mu.M and 3.13. Mu.M, respectively. The glycolytic inhibitor 2-DG was added once at 50 mM. The ECAR values were normalized with the value before the first compound addition as 100% and the background value as 0%.
In addition, the decrease in ECAR of exemplary compound 29 in media containing three different glucose concentrations was measured (fig. 8). A decrease in ECAR was observed that correlated negatively with glucose concentration.
All these findings indicate that the compounds provided herein abolish glycolytic activity of parasites at the blood stage.
Example 26
Sub-phase selectivity and time course determination
Method
Tightly synchronized 3D7 cultures were obtained by a combination of sorbitol synchronization and heparin treatment. After sorbitol synchronization, the ring stage parasites obtained were cultured in the presence of sodium salt of heparin (230. Mu.g/mL; sigma-Aldrich) from porcine intestinal mucosa until most of the parasites had progressed to the late schizont stage. Heparin is then removed from the culture, allowing the merozoites to invade the red blood cells. After 6 hours, the cultures were sorbitol synchronized to produce early ring stage parasites (0-6 hpi) and resuspended in culture medium at 1.0% parasitemia and 0.8% hematocrit. Test compounds were prepared in the same manner as described above and the resulting closely synchronized cultures were exposed to the test compounds. After incubation with test compounds, the cultures were freeze-thawed and drug sensitivity was determined by LDH assay (see Gamo, f.j. Et al, "methods of chemical starting points for anti-pharmaceutical lead identification", nature, volume 465, pages 305-310 (2010)).
Results
The blood stage plasmodium falciparum has a 2-day life cycle including merozoite invasion, proliferation from the ring stage to trophozoite and then to multicellular schizonts, and elimination from erythrocytes (see White, n.j. Et al, malaria. Lancet, volume 383, pages 723-735 (2014)). The ring stage parasite (p. Falciparum 3D 7) was first treated with the exemplary compound 29 or DHA (control) for 24, 36, 48, and 72 hours. Discovery of EC 50 The values were very similar to the 72 hour growth inhibition assay described in example 23 (see (i) in fig. 9 and 10). Next, the early-ring stage, late-ring stage, trophozoite stage, or schizont stage was treated with compound 29 for 12 hours. The parasites were then washed with growth medium and incubated for an additional 36 hours without compound. It was observed that the ring-stage parasite was less sensitive to compound 29 than the late parasites (trophozoites and schizonts) (see (ii) in fig. 9 and 10). Conversely, DHA was found to be less potent on late stages. Finally, the parasites were treated with the test compounds for 24, 36, 48 and 72 hours starting from the early ring stage and then incubated for another 36 hours without test compounds (see (iii) in fig. 9 and 10). Obtained EC 50 Values also show that the ring stage parasites are less sensitive to PfHT1 inhibitors than late stage parasites (24 hour treatment), but treatment over 36 hours shows similar efficacy to the 72 hour assay. Light microscopic observations of compound-treated parasites showed that exposure to compound 29 induced the ring-stage parasites to stop their development, but to resume growth after removal of compound 29 (fig. 11).
The foregoing description is considered as illustrative only of the principles of the disclosure. Further, since numerous modifications and changes will readily occur to those skilled in the art, it is not desired to limit the invention to the exact construction and process shown and described. Accordingly, all suitable modifications and equivalents may be resorted to, falling within the scope of the invention as defined by the appended claims.
Sequence listing
<110> Qinghua university
<120> inhibitors of malaria and plasmodium falciparum hexose transporters and uses thereof
<130> 077013-8001WO03
<160> 5
<170> SIPOSequenceListing 1.0
<210> 1
<211> 504
<212> PRT
<213> Plasmodium falciparum (Plasmodium falciparum)
<400> 1
Met Thr Lys Ser Ser Lys Asp Ile Cys Ser Glu Asn Glu Gly Lys Lys
1 5 10 15
Asn Gly Lys Ser Gly Phe Phe Ser Thr Ser Phe Lys Tyr Val Leu Ser
20 25 30
Ala Cys Ile Ala Ser Phe Ile Phe Gly Tyr Gln Val Ser Val Leu Asn
35 40 45
Thr Ile Lys Asn Phe Ile Val Val Glu Phe Glu Trp Cys Lys Gly Glu
50 55 60
Lys Asp Arg Leu Asn Cys Ser Asn Asn Thr Ile Gln Ser Ser Phe Leu
65 70 75 80
Leu Ala Ser Val Phe Ile Gly Ala Val Leu Gly Cys Gly Phe Ser Gly
85 90 95
Tyr Leu Val Gln Phe Gly Arg Arg Leu Ser Leu Leu Ile Ile Tyr Asn
100 105 110
Phe Phe Phe Leu Val Ser Ile Leu Thr Ser Ile Thr His His Phe His
115 120 125
Thr Ile Leu Phe Ala Arg Leu Leu Ser Gly Phe Gly Ile Gly Leu Val
130 135 140
Thr Val Ser Val Pro Met Tyr Ile Ser Glu Met Thr His Lys Asp Lys
145 150 155 160
Lys Gly Ala Tyr Gly Val Met His Gln Leu Phe Ile Thr Phe Gly Ile
165 170 175
Phe Val Ala Val Met Leu Gly Leu Ala Met Gly Glu Gly Pro Lys Ala
180 185 190
Asp Ser Thr Glu Pro Leu Thr Ser Phe Ala Lys Leu Trp Trp Arg Leu
195 200 205
Met Phe Leu Phe Pro Ser Val Ile Ser Leu Ile Gly Ile Leu Ala Leu
210 215 220
Val Val Phe Phe Lys Glu Glu Thr Pro Tyr Phe Leu Phe Glu Lys Gly
225 230 235 240
Arg Ile Glu Glu Ser Lys Asn Ile Leu Lys Lys Ile Tyr Glu Thr Asp
245 250 255
Asn Val Asp Glu Pro Leu Asn Ala Ile Lys Glu Ala Val Glu Gln Asn
260 265 270
Glu Ser Ala Lys Lys Asn Ser Leu Ser Leu Leu Ser Ala Leu Lys Ile
275 280 285
Pro Ser Tyr Arg Tyr Val Ile Ile Leu Gly Cys Leu Leu Ser Gly Leu
290 295 300
Gln Gln Phe Thr Gly Ile Asn Val Leu Val Ser Asn Ser Asn Glu Leu
305 310 315 320
Tyr Lys Glu Phe Leu Asp Ser His Leu Ile Thr Ile Leu Ser Val Val
325 330 335
Met Thr Ala Val Asn Phe Leu Met Thr Phe Pro Ala Ile Tyr Ile Val
340 345 350
Glu Lys Leu Gly Arg Lys Thr Leu Leu Leu Trp Gly Cys Val Gly Val
355 360 365
Leu Val Ala Tyr Leu Pro Thr Ala Ile Ala Asn Glu Ile Asn Arg Asn
370 375 380
Ser Asn Phe Val Lys Ile Leu Ser Ile Val Ala Thr Phe Val Met Ile
385 390 395 400
Ile Ser Phe Ala Val Ser Tyr Gly Pro Val Leu Trp Ile Tyr Leu His
405 410 415
Glu Met Phe Pro Ser Glu Ile Lys Asp Ser Ala Ala Ser Leu Ala Ser
420 425 430
Leu Val Asn Trp Val Cys Ala Ile Ile Val Val Phe Pro Ser Asp Ile
435 440 445
Ile Ile Lys Lys Ser Pro Ser Ile Leu Phe Ile Val Phe Ser Val Met
450 455 460
Ser Ile Leu Thr Phe Phe Phe Ile Phe Phe Phe Ile Lys Glu Thr Lys
465 470 475 480
Gly Gly Glu Ile Gly Thr Ser Pro Tyr Ile Thr Met Glu Glu Arg Gln
485 490 495
Lys His Met Thr Lys Ser Val Val
500
<210> 2
<211> 502
<212> PRT
<213> Plasmodium vivax
<400> 2
Met Lys Lys Ser Ser Lys Glu Ile Ser Ser Ser Gln Ser Leu Lys Asn
1 5 10 15
Gly Gly Ser Asp His Phe Phe Asn Thr Ser Leu Met Tyr Val Leu Ala
20 25 30
Ala Cys Leu Ala Ser Phe Ile Phe Gly Tyr Gln Val Ser Val Leu Asn
35 40 45
Thr Ile Lys Asn Phe Ile Val Ile Glu Phe Gly Trp Cys Thr Gly Asn
50 55 60
Lys Val Glu Cys Asp Asp Ser Thr Leu Lys Ser Ser Phe Leu Leu Ala
65 70 75 80
Ser Val Phe Ile Gly Ala Val Val Gly Ser Gly Phe Ser Gly Tyr Leu
85 90 95
Val Gln His Gly Arg Arg Phe Ser Leu Leu Val Ile Tyr Asn Phe Phe
100 105 110
Ile Leu Val Ser Ile Leu Thr Ser Ile Thr His His Phe His Thr Ile
115 120 125
Leu Phe Ser Arg Leu Leu Ser Gly Phe Gly Ile Gly Leu Ile Thr Val
130 135 140
Ser Val Pro Met Tyr Ile Ser Glu Met Thr His Lys Asp Lys Lys Gly
145 150 155 160
Ala Tyr Gly Val Leu His Gln Leu Phe Ile Thr Phe Gly Ile Phe Val
165 170 175
Ala Val Leu Leu Gly Met Ala Met Gly Glu Ala Pro Asp Ala Lys Ser
180 185 190
Val Asp Ala Leu Gly Glu Phe Gln Lys Ile Trp Trp Arg Leu Met Phe
195 200 205
Phe Phe Pro Cys Leu Ile Ser Ile Leu Gly Ile Val Leu Leu Thr Phe
210 215 220
Phe Tyr Lys Glu Glu Thr Pro Tyr Tyr Leu Phe Glu Asn Gly Lys Ile
225 230 235 240
Glu Glu Ser Lys Lys Ile Leu Lys Lys Ile Tyr Gly Thr Asp Asn Val
245 250 255
Asp Glu Pro Leu Lys Ala Ile Lys Asp Ala Val Glu Gln Asn Glu Ala
260 265 270
Ala Lys Lys Asn Ser Ile Ser Leu Met Arg Ala Met Gln Ile Pro Ser
275 280 285
Tyr Arg Asn Val Ile Leu Leu Gly Cys Ile Leu Ser Gly Leu Gln Gln
290 295 300
Phe Thr Gly Ile Asn Val Leu Val Ser Asn Ser Asn Glu Leu Tyr Lys
305 310 315 320
Glu Phe Leu Ser Asn Lys Leu Ile Thr Thr Leu Ser Val Ile Met Thr
325 330 335
Val Val Asn Phe Leu Met Thr Phe Pro Ala Ile Tyr Ile Val Glu Lys
340 345 350
Leu Gly Arg Lys Thr Leu Leu Leu Cys Gly Cys Ala Gly Val Ile Cys
355 360 365
Ala Phe Leu Pro Thr Ala Ile Ala Asn Gln Ile Asp Ser Thr Ser Ala
370 375 380
Phe Val Lys Asn Leu Ser Ile Ala Ala Thr Phe Val Met Ile Ile Ser
385 390 395 400
Phe Ala Val Ser Tyr Gly Pro Val Leu Trp Ile Tyr Leu His Glu Met
405 410 415
Phe Pro Ser Glu Ile Lys Asp Ser Ala Ala Ser Leu Ala Ser Leu Val
420 425 430
Asn Trp Val Cys Ala Ile Ile Val Val Phe Pro Ser Asp Ile Ile Ile
435 440 445
Lys Lys Ser Pro Thr Ile Leu Phe Phe Ile Phe Ser Gly Met Ser Ile
450 455 460
Leu Ser Phe Leu Phe Ile Phe Phe Phe Ile Lys Glu Thr Lys Gly Gly
465 470 475 480
Glu Ile Gly Thr Ser Pro Tyr Ile Thr Met Glu Glu Arg Gln Lys His
485 490 495
Met Gly Lys Ser Ala Val
500
<210> 3
<211> 503
<212> PRT
<213> Plasmodium ovale (Plasmodium ovale)
<400> 3
Met Lys Lys Ser Ser Lys Glu Ala Ser Ser Ser Cys Ser Ala Lys Gly
1 5 10 15
Glu Val Ser Asn Ser Phe Phe Asn Thr Ser Phe Leu Tyr Val Leu Ser
20 25 30
Ala Cys Ile Ala Ser Phe Ile Phe Gly Tyr Gln Val Ser Val Leu Asn
35 40 45
Thr Ile Lys Asn Phe Ile Val Ile Glu Phe Gln Trp Cys Lys Gly Glu
50 55 60
Asp Arg Leu Asp Cys Asp Glu Asn Thr Ile Arg Ser Ser Phe Leu Leu
65 70 75 80
Ala Ser Val Phe Ile Gly Ala Val Phe Gly Ser Gly Phe Ser Gly Tyr
85 90 95
Leu Val Gln Phe Gly Arg Arg Phe Ser Leu Leu Val Ile Tyr Val Phe
100 105 110
Phe Val Phe Val Ser Ile Leu Thr Ser Ile Thr His His Phe Asp Thr
115 120 125
Ile Leu Phe Ala Arg Leu Phe Ser Gly Phe Gly Ile Gly Leu Ile Thr
130 135 140
Val Ser Val Pro Met Tyr Ile Ser Glu Met Thr His Lys Asp Lys Lys
145 150 155 160
Gly Ala Tyr Gly Val Leu His Gln Leu Phe Ile Thr Phe Gly Ile Phe
165 170 175
Ile Ala Val Leu Leu Gly Leu Ala Met Gly Glu Gly Pro Asp Gly Lys
180 185 190
Ser Thr Lys Pro Leu Ser Glu Phe Glu Lys Ile Trp Trp Arg Leu Met
195 200 205
Phe Phe Phe Pro Thr Leu Ile Ser Leu Leu Gly Ile Phe Leu Leu Val
210 215 220
Ile Phe Phe Lys Glu Glu Thr Pro Tyr Phe Leu Phe Glu Lys Gly Lys
225 230 235 240
Ile Glu Glu Ser Lys Lys Ile Leu Met Lys Ile Tyr Gly Thr Asn Asp
245 250 255
Val Glu Glu Pro Met Lys Ala Ile Lys Glu Ala Ile Glu Gln Asn Glu
260 265 270
Ser Ala Lys Asn Asn Ser Leu Ser Leu Leu Arg Ala Met Lys Ile Pro
275 280 285
Ser Tyr Arg Tyr Val Ile Leu Leu Gly Cys Ile Leu Ser Gly Phe Gln
290 295 300
Gln Phe Thr Gly Ile Asn Val Leu Val Ala Asn Ser Asn Glu Leu Tyr
305 310 315 320
Lys Gly Phe Leu Gly Lys Asp Leu Ile Thr Ile Leu Ser Val Ile Met
325 330 335
Thr Ala Val Asn Phe Leu Met Thr Phe Pro Ala Ile Tyr Ile Val Glu
340 345 350
Lys Leu Gly Arg Lys Thr Leu Leu Leu Gly Gly Cys Ile Gly Val Ile
355 360 365
Cys Ala Tyr Leu Pro Thr Ala Ile Ala Asn Glu Leu Tyr Lys Asp Ser
370 375 380
Tyr Ser Val Lys Ile Met Ser Val Val Gly Thr Phe Val Met Ile Ile
385 390 395 400
Ser Phe Ala Val Ser Tyr Gly Pro Val Leu Trp Ile Tyr Leu His Glu
405 410 415
Met Phe Pro Ser Glu Ile Lys Asp Ser Ala Ala Ser Leu Ala Ser Leu
420 425 430
Val Asn Trp Val Cys Ala Ile Val Val Val Phe Pro Ser Asp Ile Ile
435 440 445
Ile Lys Lys Ser Pro Ser Ile Leu Phe Ile Leu Phe Ser Val Met Cys
450 455 460
Ile Ile Ser Phe Leu Phe Ile Phe Phe Phe Ile Lys Glu Thr Lys Gly
465 470 475 480
Gly Glu Ile Gly Thr Ser Pro Tyr Ile Ser Ile Glu Glu Arg Gln Lys
485 490 495
His Met Thr Lys Ser Ala Val
500
<210> 4
<211> 379
<212> PRT
<213> Plasmodium malariae (Plasmodium malariae)
<400> 4
Met Asn Lys Ser Ser Lys Glu Ile Ser Ser Gly Ser Cys Ala Ile Asn
1 5 10 15
Glu Glu Asp Lys Phe Phe Asn Thr Ser Phe Lys Tyr Val Leu Ser Ala
20 25 30
Cys Ile Ala Ser Phe Ile Phe Gly Tyr Gln Val Ser Val Leu Asn Thr
35 40 45
Ile Lys Asn Phe Ile Val Val Glu Phe Glu Trp Cys Thr Gly Asp Asn
50 55 60
Arg Leu Asp Cys Arg Glu Asn Thr Ile Lys Ser Ser Phe Leu Leu Ala
65 70 75 80
Ser Val Phe Ile Gly Ala Val Ile Gly Ser Gly Phe Ser Gly Gly Phe
85 90 95
Gly Ile Gly Leu Ile Thr Val Ser Val Pro Leu Tyr Ile Ser Glu Met
100 105 110
Thr His Lys Asp Lys Lys Gly Ala Tyr Gly Val Leu His Gln Leu Ser
115 120 125
Ile Thr Phe Gly Ile Phe Ile Ala Val Leu Leu Gly Leu Ala Met Gly
130 135 140
Glu Gly Pro Lys Gly Glu Ser Lys Glu Pro Leu Gly Asn Phe Glu Lys
145 150 155 160
Ile Trp Trp Arg Leu Met Phe Phe Met Pro Ser Ile Ile Ser Ile Val
165 170 175
Gly Ile Phe Leu Leu Val Val Phe Phe Lys Glu Glu Thr Pro Tyr Phe
180 185 190
Leu Tyr Glu Lys Gly Lys Leu Glu Glu Ser Lys Lys Ile Leu Lys Lys
195 200 205
Ile Tyr Gly Ser Asp Asp Val Asp Glu Pro Leu Asn Ala Ile Lys Glu
210 215 220
Ala Ile Glu Gln Thr Glu Ser Ala Lys Arg Asn Ser Leu Ser Leu Leu
225 230 235 240
Asn Ala Leu Lys Ile Pro Cys Tyr Arg Tyr Val Ile Leu Leu Gly Cys
245 250 255
Ile Leu Ser Ala Phe Gln Gln Phe Thr Gly Ile Asn Val Leu Val Ser
260 265 270
Asn Ser Asn Glu Leu Tyr Lys Glu Phe Leu Pro Ser Glu Trp Ile Thr
275 280 285
Ile Leu Ser Val Ile Met Thr Ile Val Asn Phe Leu Met Thr Phe Pro
290 295 300
Ala Ile Tyr Ile Val Glu Lys Leu Gly Arg Lys Thr Leu Leu Leu Gly
305 310 315 320
Gly Cys Phe Gly Ile Ile Cys Ala Tyr Val Pro Thr Ala Ile Ala Asn
325 330 335
Leu Ile Asn Lys Asn Ser Asn Pro Val Lys Ser Leu Leu Ser Tyr Leu
340 345 350
Phe Ser Phe Leu Leu Lys Lys Gln Lys Val Glu Lys Leu Glu Gln Val
355 360 365
His Ile Phe Arg Trp Lys Lys Gly Lys Asn Thr
370 375
<210> 5
<211> 500
<212> PRT
<213> Plasmodium knowlesi >
<400> 5
Met Lys Asn Ser Asn Glu Ile Ser Ser Ser Gln Ser Leu Lys Asn Asn
1 5 10 15
Gly Ser Asp Gly Phe Phe Asn Thr Ser Leu Met Tyr Val Leu Ala Ala
20 25 30
Cys Leu Ala Ser Phe Leu Phe Gly Tyr Gln Val Ser Val Leu Asn Thr
35 40 45
Ile Lys Asp Phe Ile Val Ile Glu Phe Gly Trp Cys Ala Gly Lys Glu
50 55 60
Val Asn Cys Asp Asp Ser Thr Leu Lys Ser Ser Phe Leu Leu Ala Ser
65 70 75 80
Val Phe Ile Gly Ala Val Val Gly Ser Gly Phe Ser Gly Phe Leu Val
85 90 95
Gln His Gly Arg Arg Phe Ser Leu Leu Val Ile Tyr Asn Phe Phe Ile
100 105 110
Leu Val Ser Ile Leu Thr Ser Ile Thr His His Phe His Thr Ile Leu
115 120 125
Phe Ser Arg Leu Leu Ser Gly Phe Gly Ile Gly Leu Ile Thr Val Ser
130 135 140
Val Pro Met Tyr Ile Ser Glu Met Thr His Lys Asp Lys Lys Gly Ala
145 150 155 160
Tyr Gly Val Leu His Gln Leu Phe Ile Thr Phe Gly Ile Phe Ile Ala
165 170 175
Val Leu Leu Gly Met Ala Met Gly Asn Val Pro Glu Glu Val Asn Asn
180 185 190
Pro Leu Gly Thr Phe Gln Gln Ile Trp Trp Arg Leu Met Phe Phe Phe
195 200 205
Pro Cys Ile Ile Ser Ile Leu Gly Ile Val Leu Leu Thr Phe Phe Phe
210 215 220
Lys Glu Glu Thr Pro Tyr Tyr Leu Phe Glu Lys Gly Lys Val Glu Glu
225 230 235 240
Ser Lys Glu Ile Leu Lys Lys Ile Tyr Gly Ser Asp Asp Val Asp Glu
245 250 255
Pro Leu Lys Ala Ile Lys Asp Ala Val Glu Gln Asn Glu Ala Ala Lys
260 265 270
Lys Asn Ser Ile Ser Leu Met Arg Ala Met Lys Ile Pro Ser Tyr Arg
275 280 285
Tyr Val Ile Leu Leu Gly Cys Ile Leu Ser Gly Leu Gln Gln Phe Thr
290 295 300
Gly Ile Asn Val Leu Val Ser Asn Ser Asn Ala Leu Tyr Lys Gly Phe
305 310 315 320
Leu Thr Asn Glu Trp Ile Thr Thr Leu Ser Val Ile Met Thr Val Val
325 330 335
Asn Phe Leu Met Thr Phe Pro Ala Ile Tyr Ile Val Glu Lys Leu Gly
340 345 350
Arg Lys Thr Leu Leu Leu Cys Gly Cys Ala Gly Ile Val Cys Ala Phe
355 360 365
Leu Pro Thr Ala Ile Ala Asn Leu Ile Asn Asn Thr Ser Asp Val Val
370 375 380
Lys Lys Leu Ser Ile Ser Ala Thr Phe Val Met Ile Val Ser Phe Ala
385 390 395 400
Val Ser Tyr Gly Pro Val Leu Trp Ile Tyr Leu His Glu Met Phe Pro
405 410 415
Ser Glu Ile Lys Asp Ser Ala Ala Ser Leu Ala Ser Leu Val Asn Trp
420 425 430
Met Cys Ala Ile Ile Val Val Phe Pro Ser Asp Ile Ile Ile Lys Gln
435 440 445
Ser Pro Thr Ile Leu Phe Phe Ile Phe Ser Gly Met Ser Ile Val Ala
450 455 460
Phe Leu Phe Ile Phe Phe Phe Ile Lys Glu Thr Lys Gly Gly Glu Ile
465 470 475 480
Gly Thr Ser Pro Tyr Ile Thr Leu Glu Glu Arg Gln Lys His Met Gly
485 490 495
Lys Ser Val Val
500

Claims (66)

1. A molecule capable of binding to both the R1 binding pocket and the R2 binding pocket of a Plasmodium falciparum hexose transporter (PfHT) polypeptide having an amino acid sequence of SEQ ID NO:1 or an analog thereof having at least 70% sequence identity to SEQ ID NO:1,
Wherein the R1 binding pocket comprises at least one or more amino acid residues selected from the group consisting of the equivalent residues in Q169, Q305, Q306, N311, N341, W412, and N435 of SEQ ID NO:1 or analogs thereof, and the R2 binding pocket comprises at least one or more amino acid residues selected from the group consisting of the equivalent residues in V44, L47, F85, W436, and V443 of SEQ ID NO:1 or analogs thereof;
wherein the molecule is not compound 3361 having the formula:
Figure FDA0003863898450000011
2. the molecule of claim 1, wherein the R1 binding pocket further comprises one or more additional amino acid residues selected from equivalent residues in F40, I172, I176, I310, F403, and A404 of SEQ ID NO:1 or analogs thereof.
3. The molecule of any one of the preceding claims, wherein the R1 binding pocket further comprises one or more additional amino acid residues selected from equivalent residues in T145, T173, V314, and I400 of SEQ ID No. 1, or analogs thereof.
4. The molecule of any one of the preceding claims, wherein the R2 binding pocket further comprises one or more additional amino acid residues selected from the group consisting of equivalent residues in L81, V312, a439 and F444 of SEQ ID No. 1 or an analog thereof.
5. The molecule of any one of the preceding claims, wherein the molecule is capable of further binding to the R3 binding pocket of the PfHT polypeptide or analog thereof, wherein the R3 binding pocket comprises at least one or more amino acid residues selected from the group consisting of equivalent residues in N48, K51, N52, N311, N316, N318, E319, and D447 or an analog thereof of SEQ ID No. 1.
6. The molecule of claim 5, wherein the R3 binding pocket further comprises one or more additional amino acid residues selected from equivalent residues in F85, V312, S315, V443, and F444 of SEQ ID NO 1 or analogs thereof.
7. The molecule of claim 1, wherein the molecule is capable of binding to the PfHT polypeptide or analog thereof with a Kd value of no more than 20 μ M as determined by the MST method.
8. The molecule of any one of the preceding claims, wherein the molecule comprises an M1 moiety, an M2 moiety, and an M3 moiety covalently linked together, wherein the molecule is capable of binding to the PfHT polypeptide or analog thereof in a manner such that, upon binding, the M1 moiety fits the R1 binding pocket, the M2 moiety fits the R2 binding pocket, and the M3 moiety fits the R3 binding pocket.
9. The molecule of claim 8, wherein the M1 moiety comprises a hexose moiety.
10. The molecule of claim 9, wherein the M1 moiety comprises a D-glucose, L-glucose, D-galactose, L-galactose, D-mannose, D-xylose, D-fructose, 2-deoxy-D-glucose, or 2-deoxy-2-halo-D-glucose moiety.
11. The molecule of claim 8, wherein the M2 moiety comprises an optionally substituted linear hydrocarbon moiety having a length of 6 to 12 atoms.
12. A molecule according to claim 8, wherein the M3 moiety comprises an optionally substituted aromatic or non-aromatic cyclic moiety.
13. The molecule of any one of the preceding claims, wherein the PfHT analog is selected from the group consisting of Plasmodium vivax hexose transporter (PvHT), plasmodium ovale hexose transporter (PoHT), plasmodium malariae hexose transporter (PmHT), and Plasmodium knowlesi sugar transporter (PkHT).
14. The molecule of claim 13, wherein the PfHT analog has an amino acid sequence selected from the group consisting of SEQ ID NO 2, SEQ ID NO 3, SEQ ID NO 4, and SEQ ID NO 5.
15. A complex comprising a PfHT polypeptide having the amino acid sequence of SEQ ID No. 1, or an analog thereof having at least 70% sequence identity to SEQ ID No. 1, bound to a molecule according to any one of claims 1 to 14.
16. The complex of claim 15, which is crystalline.
A set of X-ray crystal structure coordinates of at least one allosteric binding pocket of a PfHT polypeptide having the amino acid sequence of SEQ ID NO:1 or an analogue thereof having at least 70% sequence identity with SEQ ID NO:1,
wherein the allosteric binding pocket is inducible when compound 3361 is complexed with the PfHT polypeptide or analog thereof, and
wherein the compound 3361 has the formula:
Figure FDA0003863898450000021
18. the set of X-ray crystal structure coordinates of claim 17, wherein the at least one allosteric binding pocket comprises:
a) R1 binding pocket comprising at least one or more amino acid residues selected from the group consisting of equivalent residues in F40, Q169, I172, I176, Q305, Q306, I310, N311, N341, F403, A404, W412 and N435 of SEQ ID NO:1 or analogues thereof,
b) An R2 binding pocket comprising at least one or more amino acid residues selected from the group consisting of equivalent residues in V44, L47, L81, F85, V312, W436, A439, V443, and F444 of SEQ ID NO:1 or analogs thereof;
c) An R3 binding pocket comprising at least one or more amino acid residues selected from the group consisting of equivalent residues in N48, K51, N52, F85, N311, V312, S315, N316, N318, E319, V443, F444, and D447 of SEQ ID NO 1 or analogs thereof; or
d) Any combination thereof.
19. The set of X-ray crystal structure coordinates of claim 18, wherein the R1 binding pocket further comprises one or more additional amino acid residues selected from the group consisting of T145, T173, V314, and I400 of SEQ ID No. 1, or an equivalent residue in an analog thereof.
20. The set of X-ray crystal structure coordinates of any one of claims 17 to 19, as shown in figure 2.
21. A computer readable storage medium having stored thereon the set of X-ray crystal structure coordinates of any one of claims 17 to 20.
22. A method of assessing or predicting the binding characteristics of a compound to a PfHT polypeptide having the amino acid sequence of SEQ ID NO:1 or an analogue thereof having at least 70% sequence identity to SEQ ID NO:1, comprising the steps of:
a) Generating on a computer a representation of the three-dimensional structure of the at least one allosteric binding pocket based on the set of X-ray crystal structure coordinates according to any one of claims 17 to 20,
b) Generating a representation of said compound on a computer, an
c) Adapting the representation of the compound according to step b) to the representation of the three-dimensional structure of the at least one allosteric binding pocket according to step a) to determine the probability that the compound binds to the at least one allosteric binding pocket.
23. A method of identifying a compound as a potential PfHT inhibitor, the method comprising the steps of:
a) Generating on a computer a representation of the three-dimensional structure of the at least one allosteric binding pocket based on the set of X-ray crystal structure coordinates of any one of claims 17 to 20,
b) Generating a representation of the compound on a computer,
c) Adapting the representation of the compound according to step b) to a computer representation of the three-dimensional structure of the at least one allosteric binding pocket according to step a) to provide an energy minimized configuration of the compound in the at least one allosteric binding pocket; and
d) Evaluating the results of step c) to quantify the binding between the compound and the at least one allosteric binding pocket,
wherein the compound is identified as a potential PfHT inhibitor when the compound binds to the at least one allosteric binding pocket to produce a low-energy, stable complex.
24. A virtual screening method for identifying potential PfHT inhibitors, the method comprising the steps of:
a) Generating or accessing on a computer a representation of the three-dimensional structure of the at least one allosteric binding pocket based on the set of X-ray crystal structure coordinates according to any of the claims 17 to 20,
b) Generating or accessing on a computer a representation of a candidate compound from a library of compounds,
c) Adapting the representation of the candidate compound according to step b) to the representation of the three-dimensional structure of the at least one allosteric binding pocket according to step a) to provide an energy minimized conformation of the candidate compound in the at least one allosteric binding pocket; and
d) Evaluating the results of step c) to quantify the binding between the candidate compound and the at least one allosteric binding pocket,
e) The quantitative binding is compared to a predetermined threshold,
wherein the candidate compound is identified as a potential PfHT inhibitor based on the comparison of step e).
25. A method of designing a compound capable of binding to a PfHT polypeptide having the amino acid sequence of SEQ ID No. 1 or an analog thereof having at least 70% sequence identity to SEQ ID No. 1, the method comprising:
a) Generating on a computer a representation of the three-dimensional structure of the at least one allosteric binding pocket based on the set of X-ray crystal structure coordinates according to any one of claims 17 to 20,
b) Generating a representation of the candidate compound on a computer,
c) Fitting the representation of the candidate compound according to step b) with the representation of the three-dimensional structure of the at least one allosteric binding pocket according to step a) to provide an energy minimized configuration of the candidate compound in the at least one allosteric binding pocket; and
d) Optionally, modifying the candidate compound based on the results obtained in step c);
e) Optionally, repeating steps b) to c) with the modified candidate compound obtained in step d),
wherein the compound is identified as a potential PfHT inhibitor when the compound binds to the at least one allosteric binding pocket to produce a low-energy, stable complex.
26. A compound having the formula (I):
A-B-L-D-E (I)
or a pharmaceutically acceptable salt thereof, wherein
A is a hexose moiety linked to B through its atom selected from carbon, nitrogen, oxygen or sulfur;
b is absent, or is selected from-CH 2 C(O)O-、-CH 2 -C (O) NH-and-C (O) -;
l is- (CH) 2 ) m -、-(CH 2 OCH 2 ) q -or- (CH) 2 ) n -W-(CH 2 ) p <xnotran> -, -W- , -O-, -S-, -NH-, -C = C-, -C (O) O- -C (O) NH-, m 1 12 , n, p q 1 3 ; </xnotran>
D is absent, or is selected from-O-, -S-, and-NH-;
e is selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, wherein said cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more R groups;
r is selected from halogen, oxo, alkyl, haloalkyl, -OR 1 and-NR 2 R 3
R 1 Selected from the group consisting of hydrogen, alkyl, aryl, alkylaryl, and alkylalkoxy;
R 2 and R 3 Each selected from the group consisting of hydrogen, alkyl, cycloalkyl and aryl, wherein said alkyl, cycloalkyl and aryl are optionally substituted with one or more alkoxy groups.
27. The compound of claim 26, wherein a is selected from the group consisting of D-glucose, L-glucose, D-galactose, L-galactose, D-mannose, D-xylose, D-fructose, 2-deoxy-D-glucose, and 2-deoxy-2-halo-D-glucose moieties.
28. The compound of claim 26, wherein B is-CH 2 C(O)O-。
29. The compound of claim 28, wherein L is- (CH) 2 ) m -or- (CH) 2 ) n O(CH 2 ) p -。
30. The compound of claim 26, wherein B is absent, or-CH 2 -C (O) NH-or-C (O) -.
31. The compound of claim 26 or 30, wherein L is- (CH) 2 ) m -。
32. The compound of claim 26, wherein D is-O-.
33. The compound of claim 26, wherein E is aryl optionally substituted with one or more R groups.
34. The compound of claim 33, wherein the aryl group is selected from:
Figure FDA0003863898450000031
35. the compound of claim 33 OR 34, wherein R is-OR 1 or-NR 2 R 3
36. The compound of claim 35, wherein R is-OR 1 And R is 1 Is an alkyl group.
37. The compound of claim 35, wherein R is-NR 2 R 3 And R is 2 And R 3 Is hydrogen.
38. The compound of claim 26, wherein E is heteroaryl optionally substituted with one or more R groups.
39. The compound of claim 38, wherein the heteroaryl is selected from:
Figure FDA0003863898450000032
40. the compound of claim 38 OR 39, wherein R is halogen, oxo, alkyl, haloalkyl, -OR 1 or-NR 2 R 3
41. The compound of claim 38 OR 39, wherein R is-OR 1 And R is 1 Is hydrogen, alkyl, aryl, alkylalkoxy or alkylaryl.
42. The compound of claim 38 or 39, wherein R is-NR 2 R 3 And R is 2 And R 3 Each hydrogen, alkyl, aryl or cycloalkyl.
43. The compound of claim 26, wherein D is absent.
44. The compound of claim 43, wherein E is aryl optionally substituted with one or more R groups.
45. The compound of claim 44, wherein the aryl is
Figure FDA0003863898450000041
46. The compound of claim 43, wherein E is heteroaryl optionally substituted with one or more R groups.
47. The compound of claim 46, wherein the heteroaryl is
Figure FDA0003863898450000042
48. The compound of claim 46 OR 47, wherein R is halogen, oxo, OR-OR 1
49. The compound of claim 46 OR 47, wherein R is-OR 1 And R is 1 Is an alkyl group.
50. The compound of claim 26, having the formula (II)
Figure FDA0003863898450000043
Wherein L and E are as defined in claim 26.
51. The compound of claim 50, wherein L is- (CH) 2 ) m -m is an integer from 1 to 12, preferably from 2 to 10, or from 3 to 8.
52. The compound of claim 50 or 51, wherein E is aryl or heteroaryl, and said aryl and said heteroaryl are optionally substituted with one or more R groups.
53. The compound of claim 52, wherein R is halogen OR-OR 1
54. The compound of claim 26, having a formula selected from:
Figure FDA0003863898450000044
Figure FDA0003863898450000051
wherein Z is hydrogen or halogen, L and E are as defined in claim 26.
55. The compound of claim 54, wherein L is- (CH) 2 ) m And m is an integer from 1 to 12, preferably an integer from 4 to 12, an integer from 6 to 12 or an integer from 8 to 12.
56. The compound of claim 54 or 55, wherein E is aryl or heteroaryl, and said aryl and said heteroaryl are optionally substituted with one or more R groups.
57. The compound of claim 56, wherein R is halogen, alkyl, haloalkyl, -OR 1 or-NR 2 R 3
58. The compound of claim 26, wherein the compound is capable of binding to a PfHT polypeptide having the amino acid sequence of SEQ ID No. 1 or an analog thereof having at least 70% sequence identity to SEQ ID No. 1 with a Kd value of NO more than 20 μ Μ, as determined by the MST method.
59. The compound of claim 26, wherein the compound is capable of an EC of no more than at least 5-fold that of compound 3361 50 Value inhibits a PfHT polypeptide having the amino acid sequence of SEQ ID NO. 1 or an analog thereof having at least 70% sequence identity to SEQ ID NO. 1.
60. The compound of claim 26, wherein the compound is selected from the group consisting of:
3- (naphthalen-2-yloxy) propyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (naphthalen-2-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
7- (naphthalen-2-yloxy) heptyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
2- (2- (naphthalen-2-yloxy) ethoxy) ethyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
n- (5- (naphthalen-2-yloxy) pentyl) -2- (((3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetamide;
5- (quinolin-6-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (naphthalen-1-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5-phenoxypentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (pyridin-4-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (benzo [ d ] [1,3] dioxol-5-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (3, 4, 5-trimethoxyphenoxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (3, 4-dimethoxyphenoxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
(3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl-2- (naphthalen-2-yl) acetate;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
5- (isoquinolin-6-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (isoquinolin-7-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (quinolin-7-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (quinolin-3-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (quinolin-8-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (quinolin-2-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (isoquinolin-3-yloxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- ((2-chloroquinolin-6-yl) oxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- ((7-chloroquinolin-4-yl) oxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (7-chloro-4-oxoquinolin-1 (4H) -yl) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- ((7-methoxyquinolin-4-yl) oxy) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
5- (7-methoxy-4-oxoquinolin-1 (4H) -yl) pentyl 2- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) acetate;
(3R, 4S,5R, 6R) -4- ((8- ((2-chloroquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8-phenoxyoctyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (naphthalen-2-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((2-bromoquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2-methylquinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2-methoxyquinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((2-ethoxyquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((2- (2-ethoxyethoxy) quinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2-isopropoxyquinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2-phenoxyquinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2- (phenylamino) quinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2- ((3, 4, 5-trimethoxyphenyl) amino) quinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((2- (cyclohexylamino) quinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (quinolin-4-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
1- (8- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) octyl) quinolin-4 (1H) -one;
(3R, 4S,5R, 6R) -4- ((8- ((7-chloroquinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
7-chloro-1- (8- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) octyl) quinolin-4 (1H) -one;
(3R, 4S,5R, 6R) -4- ((8- ((7-bromoquinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
7-bromo-1- (8- (((3r, 4s,5r, 6r) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) octyl) quinolin-4 (1H) -one;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((7-methoxyquinolin-4-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((7- (benzyloxy) quinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((7-aminoquinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((7-hydroxyquinolin-4-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (quinolin-6-ylamino) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((5-aminonaphthalen-2-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (quinolin-3-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((6- (quinolin-6-yloxy) hexyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((10- (quinolin-6-yloxy) decyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((12- (quinolin-6-yloxy) dodecyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3S,4R, 5S,6S) -6- (hydroxymethyl) -4- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((7- (trifluoromethyl) quinolin-4-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5S, 6R) -6- (((8- (quinolin-6-yloxy) octyl) oxy) methyl) tetrahydro-2H-pyran-2, 3,4, 5-tetraol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((7-iodoquinolin-4-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((6-bromoquinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((8-bromoquinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((6, 7-dimethoxyquinolin-4-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2-hydroxyquinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((3-bromoquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((7- (quinolin-6-yloxy) heptyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((9- (quinolin-6-yloxy) nonyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5S, 6R) -6- (hydroxymethyl) -3- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 4, 5-triol;
(2R, 3S,4R, 5R) -2- (((8- (quinolin-6-yloxy) octyl) oxy) methyl) tetrahydro-2H-pyran-2, 3,4, 5-tetraol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((6-methoxyquinolin-4-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ([ 1,1' -biphenyl ] -4-yloxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2-iodoquinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- (benzo [ d ] [1,3] dioxol-5-yloxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ([ 1,1' -biphenyl ] -3-yloxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R,4S, 5R,6R) -6- (hydroxymethyl) -4- ((8- (naphthalen-1-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- (benzofuran-5-yloxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- (benzo [ b ] thiophen-5-yloxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- (quinolin-7-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
6- ((8- (((3R, 4S,5R, 6R) -2,3, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-4-yl) oxy) octyl) oxy) -2H-chroman-2-one;
(3R,4S,5R,6R) -4- ((8- (anthracen-2-yloxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R,4S, 5R,6R) -6- (hydroxymethyl) -4- ((8- (phenanthren-9-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((9H-carbazol-3-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R,4S, 5R,6R) -6- (hydroxymethyl) -4- ((8- (pyrene-1-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((11- (quinolin-6-yloxy) undecyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -6- (hydroxymethyl) -4- ((8- ((2- (trifluoromethyl) quinolin-6-yl) oxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((2-aminoquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5R, 6R) -4- ((8- ((2- (dimethylamino) quinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(2R, 3S,4S, 5R) -2- (hydroxymethyl) -6- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-3, 4, 5-triol;
(3R, 4R,5S, 6R) -6- (hydroxymethyl) -5- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 3, 4-triol;
(4R, 5S, 6R) -6- (hydroxymethyl) -4- ((8- (quinolin-6-yloxy) octyl) oxy) tetrahydro-2H-pyran-2, 5-diol;
(3S,4R, 5S,6S) -4- ((9- ((2-chloroquinolin-6-yl) oxy) nonyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 3, 5-triol;
(3R, 4S,5S, 6R) -3- ((8- ((2-chloroquinolin-6-yl) oxy) octyl) oxy) -6- (hydroxymethyl) tetrahydro-2H-pyran-2, 4, 5-triol; and
(3R, 4S,5S, 6R) -6- (hydroxymethyl) -3- ((9- (quinolin-6-yloxy) nonyl) oxy) tetrahydro-2H-pyran-2, 4, 5-triol.
61. A pharmaceutical composition comprising one or more molecules according to any one of claims 1 to 14, or one or more compounds according to any one of claims 26 to 60, and a pharmaceutically acceptable excipient.
62. A method of treating a disease associated with Plasmodium infection or a PfHT polypeptide having the amino acid sequence of SEQ ID No. 1 or an analog thereof having at least 70% sequence identity to SEQ ID No. 1 in a subject, comprising administering to the subject in need thereof a therapeutically effective amount of one or more molecules of any one of claims 1 to 14, one or more compounds of any one of claims 26 to 60, or a pharmaceutical composition of claim 61.
63. The method of claim 62, wherein the disease is malaria.
64. The method of claim 62, wherein the subject is a human.
65. A method of killing or inhibiting the growth of Plasmodium by administering an effective amount of one or more molecules according to any one of claims 1 to 14, one or more compounds according to any one of claims 26 to 60 or a pharmaceutical composition according to claim 61.
66. The method of claim 65, wherein the killing or inhibiting the growth of Plasmodium is performed in vivo or in vitro.
CN202180024568.5A 2020-02-04 2021-01-26 Inhibitors of malaria and plasmodium falciparum hexose transporters and uses thereof Pending CN115427049A (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
CNPCT/CN2020/074258 2020-02-04
CN2020074258 2020-02-04
CN2020111218 2020-08-26
CNPCT/CN2020/111218 2020-08-26
PCT/CN2021/073697 WO2021155748A1 (en) 2020-02-04 2021-01-26 Inhibitors of malarial and plasmodium falciparum hexose transporter and uses thereof

Publications (1)

Publication Number Publication Date
CN115427049A true CN115427049A (en) 2022-12-02

Family

ID=77200740

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180024568.5A Pending CN115427049A (en) 2020-02-04 2021-01-26 Inhibitors of malaria and plasmodium falciparum hexose transporters and uses thereof

Country Status (3)

Country Link
CN (1) CN115427049A (en)
AU (1) AU2021215743B2 (en)
WO (1) WO2021155748A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006037807A2 (en) * 2004-10-06 2006-04-13 Institut Pasteur Identification of regions of apical membrane antigen 1 of apicomplexan parasites suitable for targeting by vaccines and drugs by derivation and analysis of the three-dimensional structure
CN101820879A (en) * 2007-10-08 2010-09-01 疟疾药物投资公司 Antimalarial compounds with flexible side-chains
US20170307590A1 (en) * 2016-04-20 2017-10-26 Washington University In St. Louis Inhibitors of malarial and human glucose transporters and methods for identifying the inhibitors
CN109810153A (en) * 2019-04-08 2019-05-28 大理大学 Fragrance replaces the preparation method of glucose compounds and its pharmaceutical composition and analgesia to apply

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR102019004351A2 (en) * 2019-03-01 2020-09-29 Universidade Federal De Minas Gerais D-GLYCOSAMINE DERIVATIVES, PHARMACEUTICAL COMPOSITIONS AND USES

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006037807A2 (en) * 2004-10-06 2006-04-13 Institut Pasteur Identification of regions of apical membrane antigen 1 of apicomplexan parasites suitable for targeting by vaccines and drugs by derivation and analysis of the three-dimensional structure
CN101820879A (en) * 2007-10-08 2010-09-01 疟疾药物投资公司 Antimalarial compounds with flexible side-chains
US20170307590A1 (en) * 2016-04-20 2017-10-26 Washington University In St. Louis Inhibitors of malarial and human glucose transporters and methods for identifying the inhibitors
CN109810153A (en) * 2019-04-08 2019-05-28 大理大学 Fragrance replaces the preparation method of glucose compounds and its pharmaceutical composition and analgesia to apply

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ABDUL AZIZ QURESHI,ET AL.: "The molecular basis for sugar import in malaria parasites", NATURE, vol. 578, pages 321 - 325, XP037019020, DOI: 10.1038/s41586-020-1963-z *
KRYSTYNA BACZKO,ET AL.: "SUGAR CHEMISTRY WITHOUT PROTECTING GROUPS-III. A FACILE CHEMICAL SYNTHESIS OF 6-O-ACYL-D-GLYCOPYRANOSES AND METHYL-6-O-ACYL-D-GLYCOPYRANOSIDES", TERMHEDRON, vol. 47, no. 23, 31 December 1991 (1991-12-31), pages 3817 - 3828 *
RALF T. OTTO,ET AL.: "Substrate specificity of lipase B from Candida antarctica in the synthesis of arylaliphatic glycolipids", JOURNAL OF MOLECULAR CATALYSIS B: ENZYMATIC, vol. 8, 31 December 2000 (2000-12-31), pages 201 - 211 *
李海姣等: "脑靶向脂质体配体葡萄糖-胆甾偶联物的设计与合成", 华西药学杂志, vol. 25, no. 3, 31 March 2010 (2010-03-31), pages 251 - 253 *

Also Published As

Publication number Publication date
WO2021155748A1 (en) 2021-08-12
AU2021215743B2 (en) 2024-06-13
AU2021215743A1 (en) 2022-09-22

Similar Documents

Publication Publication Date Title
TWI580668B (en) Hepatitis b antiviral agents
KR102626669B1 (en) Compounds and their uses for the prevention and treatment of diseases
JP5766617B2 (en) Novobiocin analogs having modified sugar moieties
KR20200066292A (en) ENPP1 inhibitors and their use for the treatment of cancer
EP3786167A1 (en) Diaryl macrocyclic compound and pharmaceutical composition, and use thereof
US20230134986A1 (en) Glucose triptolide conjugates and uses thereof
KR20130100300A (en) Substituted 2-oxy-quinoline-3-carboxamides as kcnq2/3 modulators
EP3560928B1 (en) Fused imidazole compound having indoleamine 2,3-dioxygenase inhibitory activity
US7432272B2 (en) Antiviral analogs
CA2919068A1 (en) Triazine derivative
AU2021215743B2 (en) Inhibitors of malarial and Plasmodium falciparum hexose transporter and uses thereof
WO2023030335A1 (en) Compound as tyk2/jak1 pseudokinase domain inhibitor, and synthesis and use methods
US9181251B2 (en) Anti-malarial compounds from marine natural products
EP3772355A1 (en) Bifunctional compound and its use in immunotherapy
Shan et al. 6-Heterocyclic carboxylic ester derivatives of gliotoxin lead to LSD1 inhibitors in gastric cancer cells
CN111825695B (en) Oxazolidinone compound, preparation method, application and pharmaceutical composition thereof
US20220110962A1 (en) Analogues of oleuropein and oleacein and uses thereof
Miranda et al. Bifunctional compound and its use in immunotherapy
WO2023211930A1 (en) Dual functioning immune modulating compounds, formulations, and uses thereof
WO2021204423A1 (en) Use of 15-membered azalides as active agents in the treatment of viral infections
US8440634B2 (en) Bicyclic derivatives of morphine-6-glucuronide, preparation method thereof and use of same in therapeutics
WO2022253316A1 (en) Quinoxaline derivative, preparation therefor and use thereof
Li et al. 6-Heterocyclic Carboxylic Ester Derivatives of Gliotoxin Lead to LSD1 Inhibitors in Gastric Cancer Cells
JP2006151849A (en) Therapeutic and preventive agent for cryptosporidiosis

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination