[go: up one dir, main page]

WO2013142328A1 - Small molecule antagonists of pf4 containing ultra large complexes - Google Patents

Small molecule antagonists of pf4 containing ultra large complexes Download PDF

Info

Publication number
WO2013142328A1
WO2013142328A1 PCT/US2013/032092 US2013032092W WO2013142328A1 WO 2013142328 A1 WO2013142328 A1 WO 2013142328A1 US 2013032092 W US2013032092 W US 2013032092W WO 2013142328 A1 WO2013142328 A1 WO 2013142328A1
Authority
WO
WIPO (PCT)
Prior art keywords
cio
alkyl
optionally substituted
halogen
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.)
Ceased
Application number
PCT/US2013/032092
Other languages
French (fr)
Inventor
Bruce SACHAIS
John RUX
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.)
University of Pennsylvania Penn
Original Assignee
University of Pennsylvania Penn
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 University of Pennsylvania Penn filed Critical University of Pennsylvania Penn
Publication of WO2013142328A1 publication Critical patent/WO2013142328A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/27Esters, e.g. nitroglycerine, selenocyanates of carbamic or thiocarbamic acids, meprobamate, carbachol, neostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/661Phosphorus acids or esters thereof not having P—C bonds, e.g. fosfosal, dichlorvos, malathion or mevinphos
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/662Phosphorus acids or esters thereof having P—C bonds, e.g. foscarnet, trichlorfon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/665Phosphorus compounds having oxygen as a ring hetero atom, e.g. fosfomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/726Glycosaminoglycans, i.e. mucopolysaccharides
    • A61K31/727Heparin; Heparan
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • HIT Heparin-induced thrombocytopenia
  • HITT thrombosis
  • PF4 is a 70 amino acid, lysine-rich 7.8 kDa platelet-specific protein that belongs to the CXC (or beta) chemokine subfamily. PF4 is synthesized by megakaryocytes and comprises 2-3% of the total released protein in mature platelets. PF4 exists as a tetramer in the a-granules of platelets and is secreted in high concentrations when platelets are activated. PF4 tetramer binds avidly to
  • glycosaminoglycans GAGs
  • PF4 glycosaminoglycans
  • methods for (i) preventing formation of, or (ii) disrupting PF4 tetramers include administering a compound to a subject.
  • the compounds useful in such methods bind to PF4 dimers or PF4 monomers.
  • the compounds also disrupt the salt bridges of PF4 tetramers.
  • methods for (i) preventing formation of or (ii) disrupting ultra-large complexes (ULCs) containing a PF4 tetramer and a glycosaminoglycan (GAG) include administering a compound to a subject.
  • ULCs ultra-large complexes
  • GAG glycosaminoglycan
  • the compounds useful in such methods bind to PF4 dimers or PF4 monomers or disrupt the salt bridges of PF4 tetramers.
  • methods for for (i) preventing medical conditions related to the formation of PF4 tetramers, such as HITT and atherosclerosis, (ii) treating medical conditions related to the formation of PF4 tetramers, such as HITT, (iii) correcting a platelet imbalance, (iv) preventing a decrease in platelet production, (v) increasing high density lipoproteins in a subject, (vi) preventing or treating inflammation, or (vii) modulating clotting or hemostasis are provided.
  • the compounds useful in such methods bind to PF4 dimers or PF4 monomers or disrupt the salt bridges of PF4 tetramers.
  • compositions noted above contain a moiety of the following structure, wherein R x ,
  • R Y , R x' , R Y' , X, n, and n' are defined herein:
  • compositions and kits contain (i) a pharmaceutically acceptable carrier and (ii) a compound as described above or a pharmaceutically acceptable salt or prodrug thereof.
  • compositions and kits contain (i) a medication which causes the formation of PF4 tetramers, a medication which disrupts PF4 tetramers, or a combination thereof; and (ii) a compound as described above or a pharmaceutically acceptable salt or prodrug thereof.
  • Figures 1A and IB are molecular modeling diagrams, shown in two orientations, representing the binding of a small molecule to the dimer interface on PF4 (here shown as a dimer), i.e., antagonist, described herein.
  • the antagonistic compound is light gray and the PF4 tetramer is black.
  • Figure IB contains the molecular model of Figure 1A and rotated 90°.
  • Figure 2 is a distribution of DOCK scores over 1.1 million compounds and found to be potential ULC antagonists. Dock scores are empirical with lower values representing higher affinity for PF4. As noted, the docking scores approximate a Gaussian distribution with a mean value of -26.6 and standard deviation (sd) of 3.3. The 100 compounds identified scored more than 10 sd below the mean (-73.8 or less).
  • Figure 3 is a bar graph of data obtained using photon correlation spectroscopy on a DynaPro® DLS instrument and Dynamics® software (V6.7.6; Microsoft) to obtain correlation function.
  • Samples analyzed included PF4 incubated in the absence or presence of unfractionated heparin (UFH) for 20 minutes at room temperature.
  • the bar graph shows the percentage of ULCs and small PF4 particles.
  • the solid bars include data for the small particles representing a population of particles with a mean hydrodynamic diameter of about 1 nm.
  • the striped bars include data for the large particles representing a population of particles with a mean hydrodynamic diameter of about 300 nm. Data are expressed as the percent of total intensity from each measurement, the mean of 10 measurements, and representative of two such experiments.
  • Figures 4A and 4B are bar graphs of data obtained from SDS PAGE of PF4 after BS3 crosslinking and show the ability of compounds 34, 24, 1, 80, 88 and 96 to inhibit formation of PF4 tetramers.
  • Compounds 34, 24, 1, and 96 were tested at 0.1 mM and 1 mM and compounds 80 and 88 were tested at 0.1 mM, 1 mM, and 5 mM.
  • the solid bars represent PF4 tetramer formed; dotted bars represent PF4 trimer formed; striped bars represent PF4 dimer formed; and the brick-like bars represent
  • Figure 5 is a quantitation of data obtained from a dynamic light scattering
  • Figure 5 provides a bar graph mapping the concentration ( ⁇ ) of compound 96 as a function of small PF4 particles (solid bar) and large ULCs (striped bar) formed.
  • Figure 6 is a bar graph demonstrating the ability of compound 96 to inhibit serotonin release. The graph provides the percentage of serotonin released using compound 96, as compared to a sample, i.e., buffer, containing no antagonistic compound.
  • Figure 7 is a line graph illustrating the dose response inhibition of PF4 tetramer formation using compound 1 (filled circles ( ⁇ ) at 250 ⁇ ), 34 (filled triangles (T ) at 500 ⁇ ), 80 (open triangles ( ⁇ ) at 250 ⁇ ), 101 (open circles (o) at 250 ⁇ , and 96 (open diamonds (0) at 500 ⁇ ).
  • the antagonists were incubated at the noted concentrations with PF4 (10 ⁇ g/mL) for 60 minutes at room temperature, followed by the addition of cross-lining reagent BS (0.2 mM).
  • the data are the mean ⁇ SEM of at least three independent experiments. Curves represent fit of data to the indirect Hill equation.
  • Figure 8 A is a line graph which illustrates the ability of compounds of the invention to inhibit the formation and stability of PF4:heparin ULCs as measured by ELISA.
  • Compounds 1 filled circles; ⁇ ), 34 (filled triangles; T), 80 (open triangles; ⁇ ), 101 (open circles; o), and 96 (open diamonds; 0) solutions in varying
  • Figure 8B is a bar graph of compiling data from experiments showing the ability of compounds of the invention at varying concentrations to inhibit ULC formation formed using varying concentrations of heparin.
  • One mM of compounds 1, 34, 80, 101, and 96 were preincubated with PF4 (7.5 ⁇ g/mL) for 1 hour, followed by addition of heparin (0.2 U/mL).
  • One control contained PF4 and heparin (0.2 U/mL) and the other control contained PF4 K50E (7.5 ⁇ g/mL) and heparin (0.2 U/mL).
  • Data are representative of two or more experiments.
  • Figure 8C illustrates the interaction of ULCs, formed using varying concentrations of heparin, with the PF4 antagonists of the invention.
  • 1 mM of compounds 1, 34, 80, and 101 were preincubated with PF4 (7.5 ⁇ g/mL) for 1 hour, followed by addition of varying concentrations of heparin (0.05, 0.1, 0.2, 0.4, 0.8, and 1.6 U/mL).
  • the controls contained PF4 (7.5 ⁇ g/mL) and varying concentrations of heparin (0.05, 0.1, 0.2, 0.4, 0.8, and 1.6 U/mL)
  • Figure 8D demonstrates antibody binding to ULCs in the absence and presence of PF4 antagonists of the invention.
  • ULCs prepared by incubating PF4 (5 ⁇ g/mL) and heparin (0.2 U/mL) and diluting the mixture to a final PF4 concentration (0.1 ⁇ g/mL), were incubated with 1 mM of each of compounds 1, 34, 80, 101, and 96. These mixtures were then incubated in wells pre-coated with KKO. Data are the mean ⁇ SEM of at least three independent experiments performed in triplicate.
  • Figure 8E shows the dose response of the PF4 antagonists of the invention in disrupting preformed ULCs.
  • ULCS were formed by incubating PF4 (5 ⁇ g/mL) and heparin at a 1.5: 1 ratio for 30 minutes.
  • Independent solutions were prepared containing PF4 antagonist 1 (2 runs: 125 ⁇ and 63 ⁇ ), 80 (250 ⁇ ), 101 (500 ⁇ ), 96 (1 mM), and 34 (2 mM) and the ULC presence was measured.
  • Data are mean ⁇ SEM of at least three independent experiments performed in triplicate.
  • Figures 9A-9F demonstrate FcyRIIA-mediated activation of cells and inhibition by PF4 antagonists.
  • Figure 9A is a bar graph showing the activation of DT40 cells transfected with FcyRIIA and a Luc reporter.
  • the basal condition is a buffer only control; Hep is heparin; IV.3 is the anti-FcyRIIA monoclonal antibody in the presence of anti-lgG antibody.
  • Figure 9B is a line graph providing the dose-response of DT40 activation by heparin in the presence of constant amounts of PF4.
  • Figure 9C provides the dose-response of compounds 1 ( ⁇ ), 34 (T), 80 ( ⁇ ),
  • Figure 9D illustrates the inhibition of DT40 activation by plasma obtained from three HIT patients.
  • the control (white bar) represents data obtained in the absence of any PF4 antagonist.
  • the dotted bars are the compilation of data for compound 1 ( ; - the middle bar) and the striped bars (the right bars) are the compilation of data for compound 101.
  • Figure 9E provides a bar graph illustrating activation of platelets as measured by release of 14 C-5HT. Data are representative of at least two experiments.
  • Figure 9F was generated similar to Figure E except is a line graph of activation (% max) vs. concentration of the noted PF4 antagonist ([PF4A]).
  • Activation was by plasma from patients with HIT using compounds 1 (circles; ⁇ ) and 101 (squares; ⁇ ).
  • Figures lOA-C are line graphs and illustrate inhibition of PF4 tetramerization
  • % WT represents the % of total PF4 containing WT PF4, with the remainder comprised of PP4 K50E _
  • Figure 1 OA is a line graph of the presence of PF4 tetramer vs. wt PF4.
  • PF4 and PR4 K50E total mass 10 ⁇ g/mL were incubated for 60 minutes at room temperature, followed by the addition of cross-lining reagent BS (0.2 mM).
  • the data are the mean ⁇ SEM of at least three independent experiments.
  • Figure 10B is a line graph of the presence of ULC vs. wt PF4.
  • PF4 and PF4 K50E total mass 7.5 ⁇ g/mL were for 1 hour, followed by addition of heparin (0.2 U/mL) and detected by ELISA as in figure 8A.
  • Data are the mean ⁇ SEM of at least three independent experiments performed in triplicate.
  • Figure IOC is a line graph of FcyRIIA activation vs. wt PF4.
  • the inventors determined that compounds which prevent formation of or disrupt the PF4 tetramer or ULC containing the same, i.e., antagonists, are useful in treating and/or preventing diseases related to the same.
  • Such conditions include HITT since heparin:PF4 complexes are central to the pathophysiology of HITT as the targets of the pathogenic antibodies.
  • the most pathogenic complexes of heparin and PF4 are very large and are referred to as ultralarge complexes (ULC).
  • ULCs ultralarge complexes
  • Heparin:PF4 complexes smaller than 600 kDa are typically referred to as small complexes (SC).
  • ULCs are 600 kDa or larger.
  • ULCs are 670 kDa or larger.
  • PF4 refers to platelet factor 4 which is a 70 amino acid, lysine-rich, 7.8 kDa platelet-specific protein that belongs to the CXC (or beta) chemokine subfamily, in which the first two of the four conserved cysteine residues are separated by one amino acid residue.
  • PF4 is naturally occurring, i.e., wild-type.
  • PF4 may be synthesized by recombinant or chemical methods.
  • the term PF4 also refers to mutations thereof in which one or more of the amino acids is replaced with a different amino acid.
  • PF4 mutations are described in International Patent Publication No. WO 02/006300, which is incorporated herein by reference.
  • the mutated PF4 contains Arg28 and Glu50 mutations.
  • PF4:heparin ULCs are more pathogenic than heparin:PF4 SCs.
  • ULCs are better recognized by HITT antibodies and lead to more platelet activation in the presence of these antibodies. Therefore, disruption of ULC represents a valid and novel therapeutic target in the treatment of HITT.
  • PF4 tetramers and complexes formed therewith contribute to the onset or development of individuals at risk for certain diseases or medical conditions.
  • antagonists compounds which disrupt PF4 tetramers or prevent their formation, i.e., antagonists.
  • the antagonists are useful in preventing of treating these diseases or medical conditions.
  • the term "antagonist" as used herein refers to any chemical compounds which are capable of binding to a PF4 tetramer, dimer or monomer. By doing so, the compound is thereby capable of preventing formation of or disrupting ULCs formed between the PF4 tetramer.
  • ULCs formed between glycosaminoglycans (GAG) and PF4 tetramers are responsible for the onset or development of individuals at risk for certain diseases or medical conditions. Therefore, desirable compounds are those which disrupt or prevent the formation of the PF4 tetramer and ULCs formed therewith. By doing so, the antagonist compound prevents formation of, or disrupts, ULCs.
  • the inventors determined that compounds having acidic moieties bound directly or indirectly to a heteroatom bind to PF4 monomers, PF4 trimers, PF4 dimers, and PF4 tetramers. These compounds also disrupt the salt bridge of PF4 tetramers and thereby inhibit formation of the PF4 tetramer.
  • the compounds useful herein prevent the formation of the PF4 tetramer by binding to specific sites at the PF4 trimer, dimer or monomer.
  • the compounds are capable of competing with the electrostatic interactions between the PF4 monomers, dimmers, and trimers in the PF4 tetramer and successfully disrupt to the salt bridge of the tetramer. See, Figure 1.
  • the functional groups of the compounds described herein form a stronger bond or interaction with the PF4 monomer, dimer, and trimer than they do with each other.
  • the salt bridge of the PF4 tetramer is formed via electrostatic interactions of a negatively charged amino acid, i.e., glutamic acid of a first PF4 monomer or PF4 dimer and a positively charged amino acid, i.e., lysine, of a second PF4 monomer or PF4 dimer.
  • This salt bridge is typically formed by at least via the interaction of at least one Glu and one Lys on a first PF4 monomer or PF4 dimer and at least one Lys and one Glu on a second PF4 monomer or PF4 dimer.
  • the salt bridge is formed via at least electrostatic interactions between Lys50 in a first PF4 monomer Glu28 in a second PF4 monomer.
  • the salt bridge is formed via at least electrostatic interactions between Glul28, Lys350 of a first PF4 monomer or dimer and Glu328, Lysl50 of a second PF4 monomer or dimer. In another embodiment, the salt bridge is formed via at least electrostatic interactions between Glu228, Lys450 of a first PF4 monomer or dimer and Glu428, Lys250 of a second PF4 monomer or dimer.
  • the compound may contain one or more of an acidic moiety.
  • the term "acidic moiety” as used herein refers to a chemical group on the backbone of a molecule which is capable of losing a hydrogen atom from the moiety. Such acidic moieties are readily identified by one of skill in the art.
  • the acidic moiety is desirably bound directly or indirectly to a heteroatom- containing moiety.
  • the heteroatom-containing moiety contains at least one heteroatom.
  • the acidic moiety is bound directly to the heteroatom.
  • the acidic moiety is bound indirectly to the heteroatom through a hydrocarbon chain which is optionally unsaturated and optionally contains one or more additional heteroatom.
  • the acidic moiety is bound indirectly to the heteroatom through a cyclic hydrocarbon which is optionally unsaturated and optionally contains one or more additional heteroatom.
  • the N-atom may be a primary amine ( ⁇ NH 2 ), secondary amine ( ⁇ NH ⁇ ), tertiary amine ( ⁇ N ⁇ ), or may be doubly bound to another moiety such as a carbon atom.
  • ⁇ NH 2 primary amine
  • ⁇ NH ⁇ secondary amine
  • ⁇ N ⁇ tertiary amine
  • the compound may contain a phosphorus atom in the backbone such as in a moiety of structure (I):
  • g, g', n and n' are, independently, 0 to 10, i.e., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; d is 0, 1, or 2;
  • X and X" are, independently, (i) absent, (ii) O, S, N, or P; or (iii) a ring containing one or more of O, S, N, or P;
  • X' is O or S
  • R u , R v , R u ', R v ', R x , R Y , R x ', and R Y ' are, independently, H, OH, halogen, C 3 to Cio cycloalkyl, Ci to Cio thioalkyl, aryl, heteroaryl, heterocycle, SO2H, OP(0)OH, P(0)OH, Ci to Cio alkyl substituted at one or more carbon-atom by R A , C2 to Cio alkenyl substituted at one or more carbon-atom by R A , or C2 to Cio alkynyl substituted at one or more carbon-atom by R A ; or
  • R u and R v , R u ' and R v ', R x and R Y or R x ' and R Y ' are joined to form a ring containing C3 to Cio cycloalkyl, aryl, heterocycle, or heteroaryl, wherein the ring is optionally substituted by one or more R A ;
  • the compound may contain a ring having at least one phosphorus and one oxygen-atom in the backbone of the ring.
  • the compound may contain a ring having one phosphorus and two oxygen-atoms in the backbone of the ring.
  • the compound may contain a ring having one phosphorus, one oxygen, and one nitrogen-atom in the backbone of the ring.
  • the phosphorus atom forms the backbone of two rings and is the point of attachment for the two rings.
  • the phosphorus atoms may be substituted with one or more of OH, Ci to Cio alkoxy, or Ci to Cio alkyl.
  • the nitrogen-atoms may be substituted with one or more of OH, Ci to C 10 alkoxy, or Ci to C 10 alkyl, may be part of a five to fourteen membered heterocyclic or heteroaryl ring, or may be bound to another nitrogen-atom of the compound. These rings may be optionally substituted with Ci to Cio alkyl, among other substituents.
  • the compound may contain a sulfur atom in the backbone such as a moiety of structure (IA):
  • the compound may contain a carboxylic acid
  • R K , R L , R K' , and R L' are, independently, H, OH, halogen, C 3 to Cio cycloalkyl, Ci to Cio thioalkyl, aryl, heteroaryl, heterocycle, S0 2 H, OP(0)OH, P(0)OH, Ci to Cio alkyl substituted at one or more carbon-atom by R A , C2 to Cio alkenyl substituted at one or more carbon-atom by R A , or C2 to C 10 alkynyl substituted at one or more carbon-atom by R A ; or R K and R L or R K and R L are joined to form a ring containing C3 to C 10 cycloalkyl, aryl, heterocycle, or heteroaryl, wherein the ring is optionally substituted by one or more R A ; or R A is H, Ci to
  • the compound contains an anhydride group to form a moiety of structure (IC):
  • the compound has structure (II):
  • n is 0 to 5, i.e., 0, 1, 2, 3, 4, or 5;
  • R 1 and R 2 are, independently, H, Ci to Cio alkyl, C(0)(Ci to Cio alkyl), C(0)(d to Cio alkyl), benzyl, C 3 to Cio cycloalkyl, C 2 to Cio alkenyl, or C2 to C 10 alkynyl; or R 1 and R 2 together comprise a heterocycle or heteroaryl, optionally substituted by one or more R 5 ;
  • R 3 and R 4 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, benzyl, aryl, heteroaryl, or heterocycle; or R 1 and R 3 together comprise a heterocycle or heteroaryl, each optionally substituted by one or more R 5 ;
  • R 5 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, C2 to Cio alkenyl,
  • the compound has structure (Ila), wherein R 2 is H, Ci to Cio alkyl, C 3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl:
  • the compound is selected from among:
  • the compound is selected from among:
  • the compounds described herein may also have structure (III)
  • R is Ci to C 10 alkyl. heteroaryl or aryl, each optionally substituted by one or more R 19 ;
  • R 16 is H or Ci to C 10 alkyl;
  • R 17 and R 18 are, independently, absent, H, Ci to C 10 alkyl, (Ci to C 10 alkyl)-P-(C 1 to C 10 alkyl), Ci to C 10 alkoxy, halogen, OH, N0 2 , CN, C 2 to C 10 alkenyl, P(0)OH, C 2 to C 10 alkynyl, aryl, heteroaryl, or heterocycle, each optionally substituted by one or more
  • R 19 wherein when R 17 or R18 is absent and q is at least 2, the CR 17 R18 groups are bound through a stable double bond;
  • R 19 is H, Ci to C 10 alkyl, Ci to C 10 alkoxy, halogen, N0 2 , CN, C(0)OH, C(0)(Ci to C 10 alkyl), C 2 to C 10 alkenyl, or C 2 to C 10 alkynyl; or a pharmaceutically acceptable salt or prodrug thereof.
  • the R 10 or R 11 heterocycle is a five or six membered heterocycle and contains one O or S heteroatom.
  • the compound has structure (Ilia):
  • each R is, independently, H, Ci to C 10 alkyl, Ci to C 10 alkoxy, halogen,
  • R 14 and R 15 are, independently, H, Ci to C 10 alkyl, C3 to C 10 cycloalkyl, C 2 to C 10 alkenyl, C 2 to Cio alkynyl, benzyl, aryl, heteroaryl, or heterocycle;
  • R 16 is H or Ci to C 10 alkyl;
  • R 17 and R 18 are, independently, absent, H, Ci to C 10 alkyl, Ci to C 10 alkoxy, halogen, OH, N0 2 , CN, C 2 to Cio alkenyl, P(0)OH, C 2 to C 10 alkynyl, aryl, heteroaryl, or heterocycle; wherein when R 17 or R 18 is absent and q is at least 2, the CR 17 R 18 groups are bound through a stable double bond; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound has structure (Illb), wherein s is 1 to 10: (nib)
  • R 10 is pyridine.
  • the compound has structure (IIIc):
  • t,, tthhee ccooimpound has structure (Hid), wherein R is aryl, heterocycle, or heteroaryl.
  • the compound is of structure (Hid) and R 10 and R 11 are furan.
  • the compound has structure (Hie):
  • the compound is selected from among: or pharmaceutically acceptable salts or prodrugs thereof.
  • the compound may further have structure (Illf): (Illf) wherein, r is 1 to 3, i.e., 1, 2, or 3; X is O, S, or NR 30 ; R 30 is H or Ci to C 10 alkyl; R 31 and R 32 are, independently, H, Ci to Cio alkyl, C3 to C1 0 cycloalkyl, C2 to C1 0 alkenyl, or C2 to Cio alkynyl, benzyl, aryl, heteroaryl, or heterocycle; or a pharmaceutically acceptable salt or prodrug thereof.
  • r is 1 to 3, i.e., 1, 2, or 3
  • X is O, S, or NR 30
  • R 30 is H or Ci to C 10 alkyl
  • R 31 and R 32 are, independently, H, Ci to Cio alkyl, C3 to C1 0 cycloalkyl, C2 to C1 0 alkenyl, or C2 to Cio alkynyl,
  • the comp has structure (Illg), wherein r is 1 to 3 :
  • the compounds useful herein may further have structure (IV):
  • R 20 is H, Ci to C 10 alkyl C3 to C 10 cycloalkyl, C2 to Cio alkenyl, or C2 to C 10 alkynyl, each optionally substituted by one or more R 24 :
  • R , R , and R" are independently, H, Ci to Cio alkyl, or C 3 to Cio cycloalkyl, each optionally substituted by one or more R 24 ;
  • R 24 is H, halogen, OH, CN, N0 2 , d to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R 27 ;
  • R 25 is H, Ci to Cio alkyl C 3 to Cio cycloalkyl, C2 to Cio alkenyl, or C 2 to Cio alkynyl;
  • R 26 is O or S;
  • R 27 is H, P(0)OH, or Ci to Cio alkyl optionally substituted by halogen or OP(0)OH; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound is of structure (IV) and R , R , and R are Ci to Ce alkyl.
  • the compound has structure (IVa):
  • the compound is:
  • R 40 and R 41 are, independently, H, Ci to Cio alkyl optionally substituted by one or more R 42 , aryl optionally substituted by one or more R 42 , or benzyl optionally substituted by one or more R 42 ;
  • R 42 is H, Ci to Cio alkyl, C(0)OH, NH 2 , C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), Ci to Cio aminoalkyl, halogen, aryl, or heteroaryl or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound may alternatively have structure (VI):
  • R is i aarryyll,, hheetteerrooaarryyll,, oorr hheetteerrcocycle, each optionally substituted by one or more R 53 ;
  • R 51 is H, Ci to Cio alkyl, Ci to Cio aminoalkyl, each optionally substituted by halogen or C(0)OH;
  • R 52 is absent, Ci to Cio alkyl optionally substituted by P(0)2, or aryl optionally substituted by 0(Ci to Cio alkyl)-phenyl-C(0)OH;
  • R 53 is Ci to Cio alkyl, halogen, or C(0)OH; or a pharmaceutically acceptable salt or prodrug thereof.
  • R 50 is heteroaryl or heterocycle.
  • the aryl of R 50 contains 5 to 8 carbon atoms, each carbon atom of R 50 contains one R 51 substituent, at least one carbon atom contains a
  • R 51 is H or Ci to C 6 alkyl.
  • R 50 is a benzene ring.
  • ompound is selected from among:
  • the compound may further have structure (VII):
  • t is 1 to 10, i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
  • R 60 is aryl, heteroaryl, or heterocycle;
  • R 61 is Ci to C 10 aminoalkyl, Ci to C 10 alkyl, C2 to C 10 alkenyl, C2 to C 10 alkynyl, C3 to C 10 cycloalkyl, aryl, heteroaryl, or heterocycle;
  • R 65 and R 66 are, independently, H, OH, Ci to C 10 alkyl, C3 to C 10 cycloalkyl, C2 to C 10 alkenyl, C2 to Cio alkynyl benzyl, aryl, heteroaryl, or heterocycle; or a pharmaceutically acceptable salt or prodrug thereof.
  • R 61 is optionally substituted at one or more carbon atom by 0-2 R 63
  • R 63 is Ci to C 10 alkyl or phenyl substituted by 0-5 R 64
  • R 64 is Ci to C 10 alkyl.
  • R is phenyl
  • the phenyl of R is substituted by 0-5 R and R 1 to Cio alkyl, C3 to C 10 cycloalkyl, Ci to C 10 alkoxy, or Ci to C 10 thioalkyl.
  • R 63 is phenyl.
  • R , R , R , and R are, independently, H, Ci to Cio alkyl, C 2 to Cio alkenyl, Ci to Cio aminoalkyl, C(0)(Ci to Cio aminoalkyl), or heteroaryl, each optionally substituted by one or more R 78 ;
  • R 72 is H, Ci to Cio alkyl, 0(Ci to Cio alkyl), 0(Ci to Cio alkoxy), 0(Ci to Cio aminoalkyl), Ci to Cio aminoalkyl, or heteroaryl, each optionally substituted by one or more R 76 ;
  • R 73 is Ci to Cio alkyl, Ci to Cio alkoxy, Ci to Cio aminoalkyl, each optionally substituted by one or more R 76 ;
  • R 76 is C(0)OH, C(0)(Ci to Cio alkyl), NHC(0)(Ci to Cio alkyl), C(0)NH(phenyl optionally substituted by one or more R 77
  • R 70 to R 74 is halogen
  • the compound is selected from among:
  • the compounds may also have structure (IX):
  • w is 1 to 10, i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
  • w' is 1 to 10, i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
  • R 80 is H or Ci to C 10 alkyl;
  • R 81 to R 85 are, independently, H, OH, Ci to Cio alkyl, halogen, or Ci to Cio alkoxy;
  • R 86 to R 89 are, independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl; or a
  • R 80 is H or C 2 to Cio alkyl.
  • the compound has structure (IXa) wherein R 80 to R 81 are defined above:
  • R 81 to R 85 in either compound (IX) or (IXa) is Ci to Ce alkoxy.
  • Ci to Ce alkoxy is OCH 3 .
  • the compound has structure (IXb), wherein R 80 is defined above:
  • the antagonists useful herein may further be a compound of structure (X)
  • x is 1 to 10, i.e. , 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or a pharmaceutically acceptable salt or prodrug thereof. In one embodiment, x is 2 to 10.
  • the compound is 2-amino-3-sulfino-propanoic acid:
  • the compound is (S)-2-amino-3-sulfino-propanoic acid, or a pharmaceutically acceptable salt or prodrug thereof:
  • the compound may fu
  • y is 1 to 10, i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
  • R 90 to R 94 are, independently, H, halogen, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, OH, or Ci to Cio aminoalkyl; and
  • R 95 is heterocycle, heteroaryl, aryl, or Ci to Cio aminoalkyl; or a pharmaceutically acceptable salt or prodrug thereof.
  • R 95 is a five or six membered heterocycle containing one O, S, or NR 92 heteroatom and the carbon atoms of said heteroaryl are substituted by H or Ci to Ce alkyl.
  • the compound has structure (XIa), wherein R 90 , R 95 , and y are defined above:
  • y in either structure (XI) or (XIa) is 2.
  • R 95 in either structure (XI or (XIa) is pyrrolidine.
  • the compound may further have structure (XII):
  • R 105 is aryl, heteroaryl, or heterocycle;
  • z is 1 to 10, i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
  • R 106 to R 110 are, independently, H, Ci to Cio alkyl, C 3 to Cio cycloalkyl, OH, C2 to Cio alkenyl, or C2 to Cio alkynyl; or a pharmaceutically acceptable salt or prodrug thereof.
  • z is 2 to 10.
  • the compound has structure (Xlla):
  • R 105 is aryl, heteroaryl, or heterocycle; z is 1 to 10, i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; R 106 to R 110 are, independently, H, Ci to C 10 alkyl, C 3 to C 10 cycloalkyl, OH, C2 to Cio alkenyl, or C2 to C 10 alkynyl; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound has structure (Xllb):
  • R 100 to R 104 are, independently, H, Ci to Ce alkyl, halogen, or Ci to alkoxy and R 105 , R 106 , and z are defined above.
  • the compound has structure (XIIc):
  • R 100 to R 104 are, independently, H, Ci to Ce alkyl, halogen, or Ci to Ce alkoxy and R 105 , R 106 , and z are defined above.
  • R in either structure (XII) or (Xlla) is H.
  • z is 1 in either structure (XII) or (Xlla).
  • At least one of R 100 to R 104 in either structure (XII) or (Xlla) is Ci to C 6 alkyl.
  • R 100 or R 104 in either structure (XII) or (Xlla) is
  • the compound is the following, or a
  • the compound is the following, or a pharmaceutically acceptable salt or prodrug thereof:
  • a is 0 or 1;
  • R 120 is absent, aryl, C3 to C1 0 cycloalkyl, heteroaryl, or heterocycle, each optionally substituted by one or more R 125 ;
  • R 121 is aryl containing one or more R , Ci to C 10 aminoalkyl optionally containing one or two aryl groups optionally substituted by one or more R 124 , heterocycle optionally substituted by one or more R 125 ;
  • R 123 is H, halogen, Ci to C 10 alkyl, or Ci to C 10 alkoxy;
  • R 124 is H, halogen, Ci to C 10 alkyl, Ci to C 10 alkoxy, or Ci to C 10 aminoalkyl;
  • R 120 and R 121 join to form C 5 to Cio cycloalkyl optionally substituted by one or more
  • the compounds have structure (Xllla), wherein R is a heterocycle having one SO 2 in the heterocycle backbone:
  • the compound is selected from:
  • R 201 is Ci to C 10 alkyl, Ci to C 10 aminoalkyl, C 2 to C 10 alkenyl, or C 2 to C 10
  • R ⁇ is H, CI to C 10 alkyl, halogen, CN, or N0 2 ;
  • R 200 is Ci to C 10 alkyl, Ci to C 10 alkoxy, aryl, C 2 to C 10 alkenyl,
  • Ci to Cio aminoalkyl each optionally substituted by one or more R ;
  • R is halogen, CN, NO 2 , Ci to Cio alkoxy, or aryl optionally substituted by one or more R 204 ;
  • R 204 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, NO 2 , or aryl; or a pharmaceutically acceptable salt or prodrug thereof.
  • R 201 is Ci to Cio aminoalkyl.
  • R 201 is CH 2 CH 2 NH 2 .
  • R 200 is Ci to Cio alkyl optionally substituted by one
  • R and R is Ci to Cio alkyl, Ci to Cio alkoxy, or halogen.
  • R 203 is 1, 2, or 3-tolyl, 1, 2, or 3-methoxyphenyl, or 1, 2, or 3-halogenated phenyl.
  • R 200 is C 2 to Cio alkenyl optionally substituted by
  • R and R is Ci to Cio alkyl, Ci to Cio alkoxy, or halogen.
  • the compound is selected from among:
  • R is Ci to C 10 alkyl, Ci to C 10 aminoalkyl, C 2 to C 10 alkenyl, or C 2 to C 10
  • R Juz is H, CI to C 10 alkyl, halogen, CN, or N0 2 ;
  • R 300 is Ci to C 10 alkyl, Ci to C 10 alkoxy, aryl, C 2 to C 10 alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R ;
  • R is halogen,
  • R 304 R 304.
  • Ci Ci to Cio alkoxy, halogen, CN, NO2, or aryl; or a pharmaceutically acceptable salt or prodrug thereof.
  • R 301 is Ci to Cio aminoalkyl.
  • R is CH 2 CH 2 NH 2 .
  • R 300 is phenyl
  • R 303 is Ci to Cio alkyl or halogen.
  • the compound is selected from among:
  • R 400 is OH or O;
  • R 401 , R 402 , and R 403 are independently, OR 405 or
  • R 405 is absent, Ci to Cio alkyl, heterocycle, aryl, or heteroaryl, optionally substituted by one or more R 406 ;
  • R 406 is H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, C(0)(Ci to Cio alkyl), aryl, heteroaryl, C(0)(aryl optionally substituted by one or more R 404 ;
  • R 407 is absent, H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, aryl, or heteroaryl; with the proviso that when R 400 is O and R 405 or R 407 is absent, then R 400 and R 402 join to form a 5 to 8-membered ring, wherein the 5 to 8-membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-
  • N(R 406 )(R 407 ) and R 405 and R 407 are absent, R 401 and R 402 , and/or R 401 and R 403 , and/or R 402 and R 403 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8- membered ring is optionally substituted by one or more R 404 ; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound is selected from among:
  • R is Ci to C 10 aminoalkyl, R is benzyl optionally substituted by one or more R 503 ;
  • R 501 is H, Ci to C 10 alkyl, C3 to C 10 cycloalkyl, Ci to C 10 alkoxy, or aryl;
  • f is 0 to 4, i.e., 0, 1, 2, 3, or 4;
  • R 503 is H, Ci to C 10 alkyl, Ci to C 10 alkoxy, halogen, or aryl; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
  • antagonistic compounds are those having structure (XVIII):
  • R is H, OH, Ci to C 10 alkyl, Ci to C 10 alkoxy, or C3 to C 10 cycloalkyl
  • R to R 510 are, independently, H, Ci to C 10 alkyl, Ci to C 10 alkoxy, C3 to C 10 cycloalkyl, or halogen; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
  • Additional compounds useful as antagonistic compounds include those of structure (XIX):
  • R bUU is Ci to C 10 alkyl, NH 2 or Ci to C 10 aminoalkyl
  • R bVZ to R are, independently, Ci to C 10 alkyl, C3 to C 10 cycloalkyl, Ci to C 10 alkoxy, or halogen; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound is the following or a pharmaceutically acceptable salt or prodrug the
  • R 606 is H or Ci to C 10 alkyl
  • R 607 to R 610 are, independently, H, Ci to C 10 alkyl, Ci to C 10 alkoxy, or halogen; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
  • Additional compounds useful as antagonistic compounds include those of structure (XXI):
  • R uu to R are, independently, H, Ci to C 10 alkyl, C3 to CIO cycloalkyl, C 3 to Cio spirocycloalkyl, Ci to C 10 alkoxy, halogen, Ci to C 10 aminoalkyl, or CN and R 705 is H or Ci to C 10 alkyl; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
  • R 800 to R 804 and R 806 and R 809 are, independently, H, Ci to C 10 alkyl, halogen, NH 2 , or Ci to C 10 alkoxy and R 805 is H or Ci to C 10 alkyl; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
  • R UU is H or Ci to C 10 alkyl and R U1 to R is H, Ci to C 10 alkyl, C 2 to C 10 alkenyl, C2 to C 10 alkynyl, halogen, Ci to C 10 alkoxy, Ci to C 10 aminoalkyl, or NH 2 ; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound is the following or a pharmaceutically acceptable salt or prodrug the
  • R 906 and R 907 are, independently, Ci to C 10 alkyl or Ci to C 10 alkoxy; e is 0 to 5, i.e., 0, 1, 2, 3, 4, or 5; and R 908 is H, halogen, Ci to C 10 alkyl, Ci to C 10 alkoxy, OH, CN, or NH 2 ; R 909 is H or Ci to C 10 alkyl; or a pharmaceutically acceptable salt or prodrug thereof.
  • R 907 is C2 to C 10 alkyl.
  • R 909 is H.
  • R 909 is Ci to C 10 alkoxy.
  • R 909 is ethoxy.
  • the compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
  • the following compounds may be utilized as PF4 antagonistic compounds 2-acetyl-3 -hydroxy-5-nitro-inden- 1 -one;
  • alkyl is used herein to refer to both straight- and branched-chain saturated aliphatic hydrocarbon groups.
  • an alkyl group has 1 to about 10 carbon atoms (i.e., Ci, C 2 , C3, C 4 , C5 Ce, C 7 , C 8 , C9, or C 10 ).
  • an alkyl group has 1 to about 6 carbon atoms (i.e., Ci, C 2 , C3, C 4 , C5 or Ce).
  • an alkyl group has 1 to about 4 carbon atoms (i.e., Ci, C 2 , C 3 , or C 4 ).
  • alkenyl is used herein to refer to both straight- and branched-chain alkyl groups having one or more carbon-carbon double bonds.
  • an alkenyl group contains 2 to about 10 carbon atoms (i.e., C 2 , C3, C 4 , C5, Ce, C 7 , C 8 , C9, or Cio).
  • an alkenyl group has 1 or 2 carbon-carbon double bonds and 2 to about 6 carbon atoms (i.e., C 2 , C3, C 4 , C5 or Ce).
  • alkynyl is used herein to refer to both straight- and branched-chain alkyl groups having one or more carbon-carbon triple bonds.
  • an alkynyl group has 2 to about 10 carbon atoms (i.e., C 2 , C3, C 4 , C5, Ce, C 7 , C 8 , C9, or Cio).
  • an alkynyl group contains 1 or 2 carbon-carbon triple bonds and 2 to about 6 carbon atoms (i.e., C 2 , C3, C 4 , C5, or Ce).
  • cycloalkyl is used herein to refer to cyclic, saturated aliphatic hydrocarbon groups.
  • the term cycloalkyl may include a single ring or two or more rings fused together to form a multicyclic ring structure.
  • a cycloalkyl group may thereby include a ring system having 1 to about 5 rings.
  • a cycloalkyl group has 3 to about 14 carbon atoms (i.e., C3, C 4 , C5, Ce, C 7 , C 8 , C9, C 10 , C 11 , C 12 , Ci 3 , or C14).
  • a cycloalkyl group has 3 to about 10 carbon atoms (i.e., C3, C 4 , C5 or Ce). In a further embodiment, a cycloalkyl group has 3 to about 6 carbon atoms (i.e., C3, C 4 , C5 or Ce).
  • alkoxy refers to the O-(alkyl) group, where the point of attachment is through the oxygen-atom and the alkyl group is defined above.
  • thioalkyl refers to the S-(alkyl) group, where the point of attachment is through the sulfur-atom and the alkyl group is defined above.
  • aminoalkyl refers to both secondary and tertiary amines where the point of attachment is through the nitrogen-atom and the alkyl groups are defined above.
  • the alkyl groups can be the same or different.
  • halogen refers to CI, Br, F, or I groups.
  • aryl refers to an aromatic, carbocyclic system, e.g., of about 5 to 20 carbon atoms, which can include a single ring or multiple unsaturated rings fused or linked together where at least one part of the fused or linked rings forms the conjugated aromatic system.
  • An aryl group may thereby include a ring system having 1 to about 5 rings.
  • the aryl groups include, but are not limited to, phenyl, naphthyl, biphenyl, anthryl, tetrahydronaphthyl, phenanthryl, indene, benzonaphthyl, and fluorenyl.
  • benzyl refers to a ⁇ CH 2 -phenyl group.
  • heteroaryl refers to a stable, aromatic 5- to 20- membered monocyclic or multicyclic heteroatom-containing ring.
  • the heteroaryl ring has in its backbone carbon atoms and one or more heteroatoms including nitrogen, oxygen, and sulfur atoms.
  • the heteroaryl ring contains 1 to about 4 heteroatoms in the backbone of the ring.
  • the nitrogen or sulfur atoms can be oxidized.
  • the nitrogen atoms may optionally substituted by H, Ci to C 10 alkoxy, C(0)(Ci to Ce alkyl), or C(0)0(Ci to Ce alkyl).
  • the heteroaryl ring can be attached through a heteroatom or carbon atom provided the resultant heterocyclic ring structure is chemically stable.
  • the heteroaryl ring is a multicyclic heteroatom-containing ring, it may contain 2, 3, 4, or 5 rings.
  • heteroaryl groups include, without limitation, oxygen-containing rings, nitrogen-containing rings, sulfur-containing rings, mixed heteroatom-containing rings, fused heteroatom containing rings, and combinations thereof.
  • heteroaryl groups include, without limitation, furyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, azepinyl, thienyl, dithiolyl, oxathiolyl, oxazolyl, thiazolyl, oxadiazolyl, oxatriazolyl, oxepinyl, thiepinyl, diazepinyl, benzofuranyl, thionapthene, indolyl, benzazolyl, purindinyl, pyranopyrrolyl, isoindazolyl, indox
  • heterocycle or “heterocyclic” as used herein can be used interchangeably to refer to a stable, saturated or partially unsaturated 3- to 20- membered monocyclic or multicyclic heterocyclic ring.
  • the heterocyclic ring has carbon atoms and one or more heteroatoms including nitrogen, oxygen, and sulfur atoms in its backbone. In one embodiment, the heterocyclic ring has 1 to about 4 heteroatoms in the backbone of the ring. When the heterocyclic ring contains nitrogen or sulfur atoms in the backbone of the ring, the nitrogen or sulfur atoms can be oxidized.
  • the nitrogen atoms may optionally be substituted with H, Ci to Ce alkyl, substituted Ci to Ce alkyl, C0 2 (Ci to C 6 alkyl), S0 2 (Ci to C 6 alkyl), S0 2 (substituted Ci to C 6 alkyl), S0 2 aryl, S0 2 substituted aryl, CO(Ci to C 6 alkyl), CO(substituted Ci to C 6 alkyl), COaryl or COsubstituted aryl.
  • the heterocyclic ring can be attached through a heteroatom or carbon atom provided the resultant heterocyclic ring structure is chemically stable. When the heterocyclic ring is a multicyclic ring, it may contain 2, 3, 4, or 5 rings.
  • heterocyclic groups include, without limitation, oxygen-containing rings, nitrogen-containing rings, sulfur-containing rings, mixed heteroatom-containing rings, fused heteroatom containing rings, and combinations thereof.
  • heterocyclic groups include, without limitation, tetrahydrofuranyl, piperidinyl, 2-oxopiperidinyl, pyrrolidinyl, morpholinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, pyranyl, pyronyl, dioxinyl, piperazinyl, dithiolyl, oxathiolyl, dioxazolyl, oxathiazolyl, oxazinyl, oxathiazinyl, benzopyranyl, benzoxazinyl and xanthenyl.
  • substituted refers to the substituted of an atom or moiety for another atom of moiety, provided that the substitution results in stable chemical compound.
  • the substitution may occur at a carbon atom and/or heteroatom.
  • the compounds described above may be prepared by known chemical synthesis techniques. These compounds may also be purchased from commercial vendors, e.g., the Sigma-Aldrich Co. Among such preferred techniques known to one of skill in the art are included the synthetic methods described in conventional textbooks relating to the construction of synthetic compounds. III. Compositions Containing the Antagonistic Compounds
  • compositions useful herein contain a compound discussed above in a pharmaceutically acceptable carrier with other optional suitable pharmaceutically inert or inactive ingredients.
  • a compound described above is present in a single composition.
  • a compound described above is combined with one or more excipients and/or other therapeutic agents as described below.
  • the antagonistic compounds discussed above may encompass tautomeric forms of the structures provided herein characterized by the bioactivity of the drawn structures. Further, the compounds may also be used in the form of salts derived from pharmaceutically or physiologically acceptable acids, bases, alkali metals and alkaline earth metals.
  • pharmaceutically acceptable salts can be formed from organic and inorganic acids including, e.g., acetic, propionic, lactic, citric, tartaric, succinic, fumaric, maleic, malonic, mandelic, malic, phthalic, hydrochloric, hydrobromic, phosphoric, nitric, sulfuric, methanesulfonic, napthalenesulfonic, benzenesulfonic, toluenesulfonic, camphorsulfonic, and similarly known acceptable acids.
  • organic and inorganic acids including, e.g., acetic, propionic, lactic, citric, tartaric, succinic, fumaric, maleic, malonic, mandelic, malic, phthalic, hydrochloric, hydrobromic, phosphoric, nitric, sulfuric, methanesulfonic, napthalenesulfonic, benzenesulfonic, toluenesulfonic, camphor
  • pharmaceutically acceptable salts may also be formed from inorganic bases, desirably alkali metal salts including, e.g., sodium, lithium, or potassium, such as alkali metal hydroxides.
  • inorganic bases include, without limitation, sodium hydroxide, potassium hydroxide, calcium hydroxide, and magnesium hydroxide.
  • Pharmaceutically acceptable salts may also be formed from organic bases, such as ammonium salts, mono-, di-, and trimethylammonium, mono-, di- and triethylammonium, mono-, di- and tripropylammonium (iso and normal), ethyldimethylammonium, benzyldimethylammonium, cyclohexylammonium, benzyl- ammonium, dibenzylammonium, piperidinium, morpholinium, pyrrolidinium, piperazinium, 1 -methylpiperidinium, 4-ethylmorpholinium, 1 -isopropylpyrrolidinium, 1,4-dimethylpiperazinium, 1 -n-butyl piperidinium, 2-methylpiperidinium, l-ethyl-2- methylpiperidinium, mono-, di- and triethanolammonium, ethyl diethanolammonium, n-butylmonoethanolammoni
  • salts can be in the form of esters, carbamates and other conventional "pro-drug” forms, which, when administered in such form, convert to the active moiety in vivo.
  • the prodrugs are esters.
  • the prodrugs are carbamates. See, e.g., B. Testa and J. Caldwell, "Prodrugs Revisited: The "Ad Hoc” Approach as a Complement to Ligand Design", Medicinal Research Reviews, 16(3):233-241, ed., John Wiley & Sons (1996), which is incorporated by reference.
  • a therapeutically or prophylactically effective amount of an antagonistic compound is that amount of a compound which prevents PF4 tetramer formation or disrupts PF4 tetramers, optionally present in an ULC.
  • the effective amount of a compound may vary depending upon the formulation and route of delivery. In one embodiment, effective amount (i.e., per unit) of a compound is less than about 10 g/kg.
  • the dosage is less than about 5 g/kg, 500 mg/kg, 400 mg/kg, 300 mg/kg, 200 mg/kg, 100 mg/kg, 50 mg/kg, 25 mg/kg, 10 mg/kg, 1 mg/kg, 0.5 mg/kg, 0.25 mg/kg, 0.1 mg/kg, 100 ⁇ g/kg, 75 ⁇ g/kg, 50 ⁇ g/kg, 25 ⁇ g/kg, 10 ⁇ g/kg, or 1 ⁇ g/kg.
  • the effective amount to be used is subjectively determined by the attending physician.
  • the variables involved include the specific cognitive deficit and the size, age and response pattern of the patient.
  • the effective amount to be administered may vary.
  • the effective amount for the first dose is higher than the effective amount for one or more of the subsequent doses.
  • the effective amount for the first dose is lower than the effective amount for one or more of the subsequent doses.
  • compositions of this invention include compounds described herein formulated neat or with one or more pharmaceutical carriers for administration, the proportion of which is determined by the solubility and chemical nature of the compound, chosen route of administration and standard pharmacological practice.
  • the pharmaceutical carrier(s) may be solid or liquid. Formulations may incorporate both solid and liquid carriers.
  • the compounds described herein may be administered to a subject by any desirable route, taking into consideration the specific condition for which it has been selected.
  • the compounds may, therefore, be delivered orally, by injection, inhalation (including orally, intranasally and intratracheally), ocularly, trans dermally, intravenously, subcutaneous ly, intramuscularly, sublingually, intracranially, epiduraly, rectally, and vaginally, among others.
  • the compound is delivered orally, intravenously, subcutaneously, or intramuscularly.
  • the compound may be administered alone, it may also be administered in the presence of one or more pharmaceutical carriers that are physiologically compatible.
  • the carriers may be in dry or liquid form and must be pharmaceutically acceptable. Liquid pharmaceutical compositions are typically sterile solutions or suspensions.
  • liquid carriers When liquid carriers are utilized, they are desirably sterile liquids. Liquid carriers are typically utilized in preparing solutions, suspensions, emulsions, syrups and elixirs.
  • the antagonistic compound is dissolved a liquid carrier.
  • the antagonistic compound is suspended in a liquid carrier.
  • the liquid carrier includes, without limitation, water, organic solvents, oils, fats, or mixtures thereof.
  • the liquid carrier is water containing cellulose derivatives such as sodium carboxymethyl cellulose.
  • the liquid carrier is water and dimethylsulfoxide (DMSO).
  • the liquid carrier contains water and up to 5% of DMSO.
  • organic solvents include, without limitation, alcohols such as monohydric alcohols and polyhydric alcohols, e.g., glycols and their derivatives, among others.
  • oils include, without limitation, fractionated coconut oil, arachis oil, corn oil, peanut oil, and sesame oil and oily esters such as ethyl oleate and isopropyl myristate.
  • the liquid carrier can contain other suitable excipients, including those described below.
  • the antagonistic compounds may be formulated in a solid carrier.
  • the composition may be compacted into a unit dose form, i.e., tablet or caplet.
  • the composition may be added to unit dose form, i.e., a capsule.
  • the composition may be formulated for administration as a powder.
  • the solid carrier may perform a variety of functions, i.e., may perform the functions of two or more of the excipients described below.
  • solid carrier may also act as a flavoring agent, lubricant, solubilizer, suspending agent, filler, glidant, compression aid, binder, disintegrant, or encapsulating material.
  • Suitable solid carriers include, without limitation, calcium phosphate, dicalcium phosphate, magnesium stearate, talc, starch, sugars (including, e.g., lactose and sucrose), cellulose (including, e.g., microcrystalline cellulose, methyl cellulose, sodium carboxymethyl cellulose), polyvinylpyrrolidine, low melting waxes, ion exchange resins, and kaolin.
  • the solid carrier can contain other suitable excipients, including those described below.
  • compositions may also be sub-divided to contain appropriate quantities of the antagonistic compound.
  • the unit dosage can be packaged compositions, e.g., packeted powders, vials, ampoules, prefilled syringes or sachets containing liquids.
  • excipients which may be combined with one or more of the antagonistic compounds include, without limitation, adjuvants, antioxidants, binders, buffers, coatings, coloring agents, compression aids, diluents, disintegrants, emulsifiers, emollients, encapsulating materials, fillers, flavoring agents, glidants, granulating agents, lubricants, metal chelators, osmo-regulators, pH adjustors, preservatives, solubilizers, sorbents, stabilizers, sweeteners, surfactants, suspending agents, syrups, thickening agents, or viscosity regulators. See, the excipients described in the "Handbook of Pharmaceutical Excipients", 5 th Edition, Eds.: Rowe,
  • compositions may be utilized as inhalants.
  • compositions may be prepared as fluid unit doses using an antagonistic compound and a vehicle for delivery by an atomizing spray pump or by dry powder for insufflation.
  • compositions may be utilized as aerosols, i.e., oral or intranasal.
  • the compositions are formulated for use in a pressurized aerosol container together with a gaseous or liquefied propellant, e.g., dichlorodifluoromethane, carbon dioxide, nitrogen, propane, and the like.
  • a gaseous or liquefied propellant e.g., dichlorodifluoromethane, carbon dioxide, nitrogen, propane, and the like.
  • a metered dose in one or more actuations.
  • compositions may be administered by a sustained delivery device.
  • sustained delivery refers to delivery of an antagonistic compound which is delayed.
  • suitable sustained delivery devices For use in such sustained delivery devices, the compounds are formulated as described herein. 3.
  • compositions may contain one or more medication or therapeutic agent.
  • the additional medication causes as an undesired side effect of the formation of PF4 tetramers or ULCs.
  • the medication modulates clotting, i.e., is an anti-coagulant or induces hemostasis.
  • the clotting modulator is a blood thinner such as heparin.
  • the clotting modulator is aspirin or a non-steroidal anti-inflammatory drug.
  • the clotting modulator is dipyridamole.
  • the clotting modulator is ticlopidine or clopidogrel.
  • the clotting modulator is a GPIIB/IIIA inhibitor.
  • the medication causes hemostasis and includes, without limitation, a vasoconstrictor or an agent which induces platelet aggregation.
  • the medication is a chemotherapeutic.
  • the medication is a cardiotonic.
  • the medication is a blood pressure lowering agent.
  • the medication is a cholesterol lowering agent. Examples of chemotherapeutics, cardiotonics, blood pressure lowering agents, and cholesterol lowering agents include those recited in the "Physician's Desk Reference", 64 th Edition, Thomson Reuters, 2010, which is hereby incorporated by reference.
  • the medication has the desirable effect of disrupting PF4 tetramers and/or ULCs.
  • the additional medication works synergistically with the PF4 antagonists discussed herein.
  • Such medications which disrupt PF4 tetramers may be selected by one of skill in the art and may include, without limitation, cyclic peptides which inhibit the interaction of PF4 with CCL5 (CKEY2), carbohydrates such as desulfated heparin (ODSH), or a combination thereof.
  • CKEY2 cyclic peptides which inhibit the interaction of PF4 with CCL5
  • ODSH desulfated heparin
  • the PF4 tetramer disruption agents may be combined with the PF4 antagonist compounds discussed herein either in the compositions set forth above and/or kits and methods for using the same and discussed below.
  • the compounds described herein and/or other therapeutic agents may be administered in a single composition.
  • the present invention is not so limited.
  • the compounds described herein invention may be administered in one or more separate formulations from other compounds described herein, chemotherapeutic agents, or other agents as is desired.
  • kits or packages of pharmaceutical formulations containing the compounds or compositions described herein may be organized to indicate a single formulation or combination of formulations to be taken at each desired time.
  • the kit contains packaging or a container with the compound formulated for the desired delivery route.
  • the kit contains instructions on dosing and an insert regarding the active agent.
  • the kit may further contain instructions for monitoring circulating levels of product and materials for performing such assays including, e.g., reagents, well plates, containers, markers or labels, and the like.
  • Such kits are readily packaged in a manner suitable for treatment of a desired indication.
  • the kit may also contain instructions for use of the spray pump or other delivery device.
  • Other suitable components to include in such kits will be readily apparent to one of skill in the art, taking into consideration the desired indication and the delivery route.
  • the doses are repeated daily, weekly, or monthly, for a predetermined length of time or as prescribed.
  • a package or kit can include the antagonistic compound in each dosage unit (e.g., solution, lotion, tablet, pill, or other unit described above or utilized in drug delivery).
  • a package or kit can include placebos during periods when the compound is not delivered.
  • a package or kit may contain a sequence of dosage units, so varying.
  • kits are known in the art for the use in dispensing pharmaceutical agents for oral use.
  • the package has indicators for each period.
  • the package is a labeled blister package, dial dispenser package, or bottle.
  • the packaging means of a kit may itself be geared for administration, such as an inhalant, syringe, pipette, eye dropper, or other such like apparatus, from which the formulation may be applied to an infected area of the body, such as the lungs, injected into a subject, or even applied to and mixed with the other components of the kit.
  • compositions of these kits also may be provided in dried or lyophilized forms.
  • reagents or components are provided as a dried form, reconstitution generally is by the addition of a suitable solvent. It is envisioned that the solvent also may be provided in another packaging means.
  • kits of the present invention also will typically include a means for containing the vials in close confinement for commercial sale such as, e.g., injection or blow-molded plastic containers into which the desired vials are retained.
  • kits of the invention also may include, or be packaged with a separate instrument for assisting with the
  • Such an instrument may be an inhalant, syringe, pipette, forceps, measuring spoon, eye dropper or any such medically approved delivery means.
  • Other instrumentation includes devices that permit the reading or monitoring of reactions in vitro.
  • a pharmaceutical kit in one embodiment, contains a medication which causes the formation of PF4 tetramers, medication which disrupts PF4 tetramers, or a combination thereof and one or more of an antagonistic compound selected from those described above.
  • the antagonistic compound may be in the presence or absence of one or more of the carriers or excipients described above.
  • the kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer.
  • a pharmaceutical kit in another embodiment, contains a medication which causes the formation of PF4 tetramers in a first dosage unit and one or more of an antagonistic compound selected from those described above in a second dosage unit.
  • the antagonistic compound may be in the presence or absence of one or more of the carriers or excipients described above.
  • the kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer.
  • a pharmaceutical kit is provided and contains a medication which disrupts PF4 tetramers in a first dosage unit and one or more of an antagonistic compound selected from those described above in a second dosage unit.
  • the antagonistic compound may be in the presence or absence of one or more of the carriers or excipients described above.
  • the kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer.
  • a pharmaceutical kit in still a further embodiment, contains a medication which causes the formation of PF4 tetramers in a first dosage unit, a medication which disrupts PF4 tetramers in a second dosage unit, and one or more of an antagonistic compound selected from those described above in a third dosage unit.
  • the antagonistic compound may be in the presence or absence of one or more of the carriers or excipients described above.
  • the kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer.
  • a pharmaceutical kit in a further embodiment, contains a medication which causes the formation of PF4 tetramers in a first dosage unit, one or more of an antagonistic compound selected from those described above in a second dosage unit, and one or more of the carriers or excipients described above in a third dosage unit.
  • the kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer.
  • a pharmaceutical kit in yet another embodiment, contains a medication which disrupts PF4 tetramers in a first dosage unit, one or more of an antagonistic compound selected from those described above in a second dosage unit, and one or more of the carriers or excipients described above in a third dosage unit.
  • the kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer.
  • a pharmaceutical kit in yet a further embodiment, contains a medication which causes the formation of PF4 tetramers in a first dosage unit, a medication which disrupts PF4 tetramers in a second dosage unit, and one or more of an antagonistic compound selected from those described above in a third dosage unit, and one or more of the carriers or excipients described above in a third dosage unit.
  • the kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer.
  • PF4 tetramers are linked to a subject acquiring specific diseases or conditions.
  • patient or “subject” as used herein refer to a mammalian animal. In one embodiment, the patient or subject is a human. In another embodiment, the patient or subject is a veterinary or farm animal, a domestic animal or pet, or animal normally used for clinical research. In a further embodiment, the subject or patient has elevated levels of PF4 tetramer. In yet a further embodiment
  • the subject or patient has elevated levels of PF4 tetramenGAG ULCs. In still another embodiment, the subject or patient has elevated levels of PF4 tetramer :heparin ULCs. In a further embodiment, the subject or patient is asymptomatic for medical conditions related to elevated levels of PF4 tetramer, PF4 tetramenGAG ULCs, or PF4 tetramenheparin ULCs. In another embodiment, the subject shows clinical signs of medical conditions related to elevated levels of PF4 tetramer, PF4 tetramenGAG ULCs, or PF4 tetramenheparin ULCs. In another embodiment, the subject shows clinical signs of developing or has HIT or HITT. In still a further embodiment, the subject or patient has cancer. The subject or patient has either been recognized as having or at risk of having one or more of the diseases or conditions discussed above, i.e., a primary disease or medical condition.
  • the one or more diseases or medical conditions in a subject may be the result of another disease or medical condition, i.e., a secondary disease or condition.
  • disease or “condition” as used herein refers to any disease or medical condition related directly or indirectly to the formation of PF4 tetramers.
  • the disease or medical condition may also be caused by the formation of PF4 tetramers.
  • the methods described herein thereby are useful in treating or preventing these diseases or conditions.
  • the disease or medical condition is heparin-induced thrombocytopenia (HIT).
  • HIT results from the development of thrombocytopenia, i.e., low platelet counts, due to the administration of one of the forms of the anticoagulant.
  • the disease or medical condition is heparin-induced thrombocytopenia and/with thrombosis (HITT).
  • HITT results when HIT precedes thrombosis, i.e., abnormal blood clots form inside a blood vessel and abnormal antibodies form.
  • Atherosclerotic vascular disease involves anti-platelet therapy, i.e., aspirin, the Plavix® drug, among others, and is not tolerated in all patients.
  • anti-platelet therapy i.e., aspirin, the Plavix® drug
  • TPO analogs and mimetics MPL agonists which activate MPL-the TPO receptor
  • these drugs do not work in all patients, and have significant side effects/compliance issues.
  • some lipid lowering therapies are able to increase HDL, but the ability of available drugs to do so is limited.
  • the disease or medical condition is atherosclerosis.
  • atherosclerosis resulting from the formation of a PF4 tetramer may be treated according to the invention described herein.
  • the disease or medical condition is a platelet imbalance.
  • the treatment method thereby includes correcting this platelet imbalance or preventing a platelet imbalance.
  • platelet levels are increased by stimulating platelet production.
  • a decrease in platelet production is prevented.
  • the platelet imbalance i.e., low levels of platelets, results from the formation of PF4 tetramer.
  • the platelet imbalance, i.e., low levels of platelets results from heparin administration to a subject.
  • TPO thrombopoietin
  • the compounds discussed herein may further be useful in inhibiting the activation of nuclear factor ⁇ -light-chain-enhancer of activated B ( F- ⁇ ) cells and thereby treating conditions related to an imbalance NF- ⁇ .
  • Such conditions may include, without limitation, cancer, inflammatory disease, autoimmune diseases, septic shock, viral infection, among others.
  • the compounds discussed herein may also be contemplated for use in preventing or treating inflammation which results from the formation of PF4 tetramers.
  • the inflammation may be the caused by any number of factors.
  • the inflammation is acute or chronic and localized or systemic.
  • the inflammation may be the result of a variety of factors and/or conditions.
  • the inflammation is caused by NF- ⁇ activation.
  • Lys50Glu or non-tetrameric PF4 variant, does not activate NFKB
  • mutation of Lys50 to Glu disrupted these interactions and created a mutant PF4 that readily forms dimers but not tetramers.
  • formation of ULC requires tetrameric PF4, as Lys50Glu does not form ULC with heparin.
  • the importance of PF4 tetramers in complex formation is supported by subsequent biophysical studies as reported in Rauova.
  • the compounds described herein may also be useful in therapies for subjects having atherosclerotic vascular disease in which the patient is intolerant to the conventional treatments, i.e., statins.
  • protein C is upregulated. In another embodiment, protein C is downregulated.
  • upregulated refers to an increase of protein or nucleic acid in level or function. Similarly, the term
  • downregulated refers to a decrease of protein or nucleic acid in level or function.
  • level references the amount of protein or nucleic acid present and the term “function” references the action of the protein or nucleic acid.
  • the compounds discussed above may be useful in patients that are intolerant to these conventional therapies.
  • the compounds discussed above may be synergistic with the conventional therapies.
  • treatment encompasses treatment of a subject clinically diagnosed as having a disease or medical condition.
  • the subject is treated and the disease or medical condition is eradicated, i.e., the subject is cured.
  • the subject is treated for chronic diseases and the symptoms resulting from the disease or medical conditions are relieved.
  • prevention encompasses prevention of symptoms in a subject who has been identified as at risk for the condition, but has not yet been diagnosed with the same and/or who has not yet presented any symptoms thereof.
  • the antagonists described herein be used to prevent or treat diseases and medical conditions, but the same are also useful in increasing high density lipoproteins (HDL) in a subject. Alternatively, the antagonists are useful in preventing a decrease of HDLs.
  • HDL high density lipoproteins
  • GAGs are long unbranched
  • the GAG is selected from among wild- type GAGs or synthetically produced GAGs.
  • the GAG is heparin, hyaluronan, hyaluronic acid, dermatan sulfates, keratan sulfates, and chondroitins, among others or derivatives, salts, or prodrugs thereof.
  • the GAG is heparin. In a further embodiment, the GAG is heparan sulfate. GAGs as described herein may also be variations of the those described above, i.e., they may vary in the type of hexosamine, hexose or hexuronic acid unit they contain (e.g., glucuronic acid, iduronic acid, galactose, galactos amine, glucosamine).
  • the methods described herein may be performed by administering one or more of the compounds described above via a combination therapy in prior to, concurrently with, or subsequent to another medication.
  • Such combination treatment may occur by administering compositions containing multiple active ingredients, as described above.
  • this invention also encompasses a method of
  • a chemotherapeutic agent is administered before treatment with a composition of the invention.
  • a chemotherapeutic agent is administered after treatment with a composition of the invention.
  • a chemotherapeutic agent is administered during treatment with a composition of the invention.
  • a chemotherapeutic agent is administered before treatment with a composition of the invention.
  • a chemotherapeutic agent is administered before treatment with a composition of the invention.
  • chemotherapeutic agent is administered after treatment with a composition of the invention.
  • a chemotherapeutic agent is administered before, during or after treatment with a composition of the invention.
  • the composition of the invention is administered prior to another medication which disrupts PF4 tetramers.
  • the composition of the invention is administered concurrently with another medication which disrupts PF4 tetramers.
  • the composition of the invention is administered subsequent to another medication which disrupts PF4 tetramers.
  • methods for disrupting PF4 tetramers include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject and administering a first effective amount of the compound required to decrease the first PF4 tetramer level.
  • methods for disrupting PF4 tetramers include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject, administering a first effective amount of the compound discussed herein required to decrease the first PF4 tetramer level, and optionally administering a medication which disrupts PF4 tetramers.
  • the medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
  • biological sample refers to a body fluid or tissue.
  • the body fluid can include, without limitation, whole blood, serum, plasma, peripheral blood, synovial fluid, cerebrospinal fluid, saliva, urine, semen, or other fluid secretion.
  • tissue can include, without limitation, bone marrow and lymph node, as well as samples of other tissues.
  • methods for disrupting PF4 tetramers include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject, administering a first effective amount of the compound required to decrease the first PF4 tetramer level, measuring a second level of PF4 tetramer in a second biological sample obtained from the subject, and administering a second effective amount of the compound required to decrease the second PF4 tetramer level.
  • Multiple samples can be obtained from the subject at any interval required to prevent or treat the medical condition.
  • methods for disrupting PF4 tetramers include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject, administering a first effective amount of the compound required to decrease the first PF4 tetramer level, optionally administering a first effective amount of a medication which disrupts PF4 tetramers, measuring a second level of PF4 tetramer in a second biological sample obtained from the subject, administering a second effective amount of the compound required to decrease the second PF4 tetramer level, and optionally administering a second effective amount of a medication which disrupts PF4 tetramers.
  • Multiple samples can be obtained from the subject at any interval required to prevent or treat the medical condition.
  • the medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
  • methods for disrupting ULCs containing PF4 tetramers and heparin include measuring a first level of ULCs in a first biological sample obtained from a subject and administering a first effective amount of the compound required to decrease the first ULC tetramer level.
  • methods for disrupting ULCs containing PF4 tetramers and heparin include measuring a first level of ULCs in a first biological sample obtained from a subject, administering a first effective amount of the compound required to decrease the first ULC tetramer level, and administering a first effective amount of a medication which disrupts PF4 tetramers.
  • the medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
  • methods for disrupting ULCs containing PF4 tetramers and heparin include measuring a first level of ULCs in a first biological sample obtained from a subject, administering a first effective amount of the compound required to decrease the first ULC level, measuring a second level of ULCs in a second biological sample obtained from the subject, administering a second effective amount of the compound required to decrease the second ULC level.
  • methods for disrupting ULCs containing PF4 tetramers and heparin are provided.
  • These methods include measuring a first level of ULCs in a first biological sample obtained from a subject, administering a first effective amount of the compound required to decrease the first ULC level, optionally administering a first effective amount of a medication which disrupts PF4 tetramers, measuring a second level of ULCs in a second biological sample obtained from the subject, administering a second effective amount of the compound required to decrease the second ULC level, and optionally administering a second effective amount of a medication which disrupts PF4 tetramers.
  • the medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
  • methods for preventing the formation of PF4 tetramers include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject and administering a first effective amount of the compound required to prevent formation of the PF4 tetramer.
  • methods for preventing the formation of PF4 tetramers include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject, administering a first effective amount of the compound required to prevent formation of the PF4 tetramer, and administering a first effective amount of a medication which disrupts PF4 tetramers.
  • the medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
  • methods for preventing the formation of PF4 tetramers include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject, administering a first effective amount of the compound required to prevent formation of the PF4 tetramer, measuring a second level of PF4 tetramer in a second biological sample obtained from the subject, and administering a second effective amount of the compound required to prevent formation of PF4 tetramer.
  • methods for preventing the formation of PF4 tetramers include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject, administering a first effective amount of the compound required to prevent formation of the PF4 tetramer, optionally administering a first effective amount of a medication which disrupts PF4 tetramers, measuring a second level of PF4 tetramer in a second biological sample obtained from the subject, administering a second effective amount of the compound required to prevent formation of PF4 tetramer, and optionally administering a second effective amount of a medication which disrupts PF4 tetramers.
  • the medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
  • methods for preventing the formation of ULCs containing PF4 tetramers and heparin include measuring a first level of ULCs in a first biological sample obtained from a subject and administering a first effective amount of the compound required to prevent formation of the ULCs.
  • methods for preventing the formation of ULCs containing PF4 tetramers and heparin include measuring a first level of ULCs in a first biological sample obtained from a subject, administering a first effective amount of the compound required to prevent formation of the ULCs, and administering a first effective amount of a medication which disrupts PF4 tetramers.
  • the medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
  • methods for preventing the formation of ULCs containing PF4 tetramers and heparin include measuring a first level of ULCs in a first biological sample obtained from a subject and administering a first effective amount of the compound required to prevent formation of the ULCs, measuring a second level of ULCs in a second biological sample obtained from the subject, and administering a second effective amount of the compound required to prevent formation of ULCs.
  • methods for preventing the formation of ULCs containing PF4 tetramers and heparin include measuring a first level of ULCs in a first biological sample obtained from a subject and administering a first effective amount of the compound required to prevent formation of the ULCs, optionally administering a first effective amount of a medication which disrupts PF4 tetramers, measuring a second level of ULCs in a second biological sample obtained from the subject, administering a second effective amount of the compound required to prevent formation of ULCs and/or administering a second effective amount of a medication which disrupts PF4 tetramers.
  • the medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
  • biological samples are obtained from subjects and the level of PF4 tetramer and/or PF4 tetramer:GAG (heparin) ULC measured.
  • the screening may be conducted using techniques commonly known and used in the art. Comparison of the levels of PF4 tetramer and/or PF4 tetramenheparin ULC to a control level and/or negative control provides evidence that the patient may be treated using one or more of the antagonistic compounds described herein.
  • antagonistic compound administration may be contemplated if the subject's PF4 tetramer and/or PF4 tetramer :heparin ULC level is higher than the PF4 tetramer and/or PF4 tetramer :heparin ULC level of a healthy subject.
  • Unfractionated heparin was an injectable sodium salt from porcine intestinal mucosa (Sagent Pharmaceuticals, Schaumberg, IL). HiTrap® heparin affinity columns for protein purification were purchased from GE Healthcare (Uppsala, Sweden). Immunochemicals included horseradish peroxidase-conjugated sheep polyclonal anti-human PF4 from Enzyme Research Laboratory (South Bend, ⁇ ), mouse immunoglobulin G 2bK (IgG2bK; MOPC 141) was purchased from Sigma (St. Louis, MO), and sheep anti-mouse IgG and alkaline phosphatase-conjugated goat anti-mouse IgG were from Jackson ImmunoResearch Labs (West Grove, PA).
  • Murine monoclonal antibodies KKO anti-human PF4-heparin complex
  • RTO anti-human PF4
  • IV.3 FcyRIIA-blocking antibody
  • KKO anti-human PF4-heparin complex
  • RTO anti-human PF4
  • IV.3 FcyRIIA-blocking antibody
  • Human plasma samples were obtained from patients with a high clinical suspicion for HIT (see, Cuker, "The HIT expert probability (HEP) score: a novel pre-test probability model for heparin-induced thrombocytopenia based on broad expert opinion", J. Thromb.
  • Bovine serum albumin (BSA), ionomycin, phorbol myristate acetate (PMA) and p- nitrophenyl phosphate (pNPP) tablets were obtained from Sigma and the 3,3,5,5- tetramethylbenzidine (TMB) liquid substrate system for ELISA was purchased from KPL (Gaithersburg, MD).
  • TMB 3,3,5,5- tetramethylbenzidine
  • a set of overlapping spheres which fill the binding site were generated, known as sphere centers, using the SphgenTM program. See, Kuntz, "A geometric approach of macromolecule-ligand interactions", J. Mol. Biol, 161(2):269-288 (1982), which is incorporated by reference.
  • the sphere centers were permitted to touch the surface of the molecule, were not permitted to intersect the surface, and were used to represent the negative image of the binding site on PF4.
  • the coordinates of the sphere centers were used to orient ligands within the target site.
  • Sphere-sphere distances were then compared to ligand atom-atom distances and the compatible orientations of the ligand were found.
  • a scoring grid to evaluate ligand orientations was then produced using the program GRID as described in Shoichet, "Molecular docking using shaper descriptions", J. Comp. Chem., 13(3):380-397 (1992) which is hereby incorporated by reference.
  • Figure 1 provides molecular models of a representative compound from these 101 compounds. As noted, the representative compound fits in the pocket formed by the PF4 tetramer and is adjacent to the salt bridge of the PF4 tetramer.
  • Figure IB is 5 the same molecule model as Figure 1A, but rotated 90°.
  • the 15 compounds of Table 2 were selected from the compounds of Table 1 and were selected to determine their ability to disrupt of inhibit PF4 formation.
  • cDNA encoding human PF4 was cloned into the plasmid pMT/BiPN5-His A (Invitrogen Corp., Carlsbad CA) for expression in the Drosophila Expression System (Invitrogen, Carlsbad, CA). Cloning was performed using Bgl II and Age I cloning sites.
  • PF4 expression was induced by adding copper sulfate (0.5 mM).
  • the induced S2 cells were incubated in serumfree Insect-XpressTM media (Lonza, Walkersville, MD) for 3-5 days; supernatants were collected, sodium azide (0.02% final concentration) and EDTA (2.5 mM final concentration) were added, and the media were filtered through an Express® PLUS 0.22 ⁇ filter (Millipore Corp., Billerica, MA).
  • Wild-type (WT) human PF4 in the pT7-7 vector (Novagen (Madison, WI) was expressed in the Escherichia coli strain BL21DE30 pLysS (Stratagene (La Jolla, CA)), and purified and characterized as described Rauova cited above and Park et al, "Biologic and biochemic properties of recombinant platelet factor 4 demonstrate identity with the native protein", Blood, 75: 1290-1295 (1990), which is hereby incorporated by reference. Briefly, recombinant protein was isolated from the supernatant of the bacterial lysate by affinity chromatography using a HiTrap high- performance (HP) affinity column.
  • HP HiTrap high- performance
  • WT PF4 was purified from the media on a heparin HiTrap® column on an ATKAPrimeTM system (GE Healthcare) at 4°C in Tris (10 mM), EDTA (1 mM), and pH 8 buffer. Media was loaded in buffer containing NaCl (0.5 M) and PF4 was eluted at 1.8 M NaCl using a linear gradient. Fractions containing purified PF4 as detected by silver staining of 12% NuPAGE Bis-Tris gels (Invitrogen) were pooled, concentrated and buffer exchanged into 50 mM HEPES, 0.5 M NaCl, pH - 7.2 using an Amicon® Ultra centrifugal filter (3K NMWL, Millipore Corp). Protein was quantified using a BCA assay. A. Experiment 1
  • the solid bar represent the data for the small particles (a population of particles with a mean hydrodynamic diameter of about 1 nm) and the striped bars represent the data for the large particles (a population of particles with a mean hydrodynamic diameter of about 300 nm). Data are expressed as the percent of total intensity from each measurement, the mean of 10 measurements, and representative of two such experiments. As noted from this figure, larger amounts of heparin result in the disruption of larger particles, i.e., ULCs, demonstrated by the presence of virtually all of the PF4 as small particles ( Figure 3).
  • ULCs were formed as described in Rauova cited above. Specifically, hPF4 (10 ⁇ g/mL) in phosphate-buffered saline (PBS) was incubated at 37°C for 15 minutes in the presence of heparin (0.34 U/mL) - molar ratio of PF4:heparin is 2.5: 1). After removal of three aliquots of this PF4 and heparin solution containing ULCs, the aliquots were incubated with 0.1 ⁇ , 1 ⁇ , or 10 ⁇ of compound 96 at 37°C. The total percentage of ULC (%) was measured and graphed. See, Figure 5 which is a bar graph of DLS data demonstrating ULC antagonistic activity for compound 96 as % inhibition vs. ULC diameter.
  • Example 3 Since the data of Example 3 demonstrated that the PF4 antagonists discussed herein are capable of disrupting preformed ULCs, this example was performed to determine if the PF4 antagonists of the invention were capable of preventing formation of ULCs.
  • Figure 8D demonstrates antibody binding to ULCs in the absence and presence of PF4 antagonists of the invention. Specifically, these data show that compounds 1, 34, 101, and 96 do not directly inhibit antibody binding to preformed ULCs.
  • platelet rich plasma from healthy donors was incubated with 0.5 ⁇ ⁇ carbon- 14 labeled 5-hydroxytryptamine creatinine sulfate (GE Life Sciences, Piscataway, NJ) per milliliter of PRP for 20 minutes at 37°C. Serotonin uptake was inhibited by adding 1 mmol/mL imipramine (Sigma-Aldrich, St. Louis, MO) to the PRP.
  • Negative and positive controls contained sera from patients previously known to have negative or positive serotonin release, respectively. Heparin (1 U/mL) was added to the positive control serum in the absence or presence of compound 96 (3 niM). The percent release of serotonin was calculated for all conditions as previously described and plotted.
  • Figure 6 is a bar graph of % of serotonin release for samples lacking (buffer only) and containing compound 96.These data illustrate that compound 96 is effective in inhibiting serotonin release, thereby inhibiting platelet activation.
  • platelet rich plasma from healthy donors was incubated with 0.5 ⁇ ⁇ 14 C-5-hydroxytryptamine creatinine sulfate (GE Life Sciences, Piscataway, NJ) per milliliter of PRP for 20 minutes at 37°C to produce 14 C-labeled platelets. Serotonin uptake was inhibited by adding 1 mmol/mL imipramine (Sigma-Aldrich, St. Louis, MO) to the PRP.
  • the radiolabeled platelets were mixed with KKO (170 ⁇ g/mL) or with known platelet-activating HIT plasma in the absence (buffer control) or presence of PF4 antagonist compounds 1, 34, 80, 101, and 96 (2.5 mM).
  • the assay was performed in the presence of heparin (1.0 U/mL) and in the absence of heparin (background).
  • NFAT-Luc was prepared as described in Shapiro, "c-rel Regulation of IL-2 gene expression may be mediated through activation of AP-1 ", J. Exp. Med.,
  • the NFAT reporter contained three copies of a composite NFAT-activator protein- 1 (AP-1) element from the human interleukin-2 (IL-2) gene promoter, and is activated by binding of NFAT (nuclear factor of activated T-cells).
  • AP-1 composite NFAT-activator protein- 1
  • IL-2 human interleukin-2
  • pEF6-FcyRIIA which contained the human FcyRIIA coding sequence under the control of the human EF- 1 a promoter, was constructed by cloning a human FcyRIIA IMAGE clone into the multiple cloning site of pEF6c (Invitrogen).
  • DT40 cells (chicken B cells) were cultured in RPMI supplemented with 10% fetal bovine serum, 1% chicken serum (Gibco), 50 ⁇ 13-mercaptoethanol, 2 mM GlutaMAX (Gibco), 100 U/mL penicillin and 100 U/mL streptomycin at 37°C under 5% CO 2 .
  • Cells were co-transfected with pEF6-FcyRIIA (5 ⁇ g) and pEF6-NFAT-Luc (20 ⁇ g) by electroporation.
  • PF4 10 ⁇ g/mL
  • various concentrations of compounds 1, 34, 80, 96, and 101 were independently co-incubated for 60 minutes at 37°C under 5% CO 2 followed the addition of heparin (0.3 U/mL) for 15 minutes at 37°C in 5% CO 2 .
  • KKO 20 ⁇ g/mL
  • plasma from patients with serotonin release assay confirmed HIT (1 :800 dilution) was then added. Fifty ⁇ ⁇ of each mixture was added to the 96 well plate containing 50 ⁇ ⁇ of cells at a concentration of 2 x 10 6 cells/mL in fresh medium without serum.
  • This example provides the results using non-tetramerizing PF4 mutant, PF4 K50E as an antagonist of WT PF4 on solutions containing PF4.
  • PF4 K50E was prepared and purified as described for WT-PF4 in Example 2, but using MES (50 mM), EDTA (1 mM), pH 6.5 as the column buffer system. Media was loaded in buffer containing 0.3 M NaCl and PF4 K50E eluted at NaCl (1.3 M) using a linear gradient.
  • PF4 and PR4 K50E total mass 10 ⁇ g/mL were incubated for 60 minutes at room temperature, followed by the addition of cross-lining reagent BS (0.2 mM). Independent solutions of PF4 and PF4 (total mass 7.5 ⁇ g/mL) were incubated for 1 hour, followed by addition of heparin (0.2 U/mL). The samples were analyzed by ELISA, as described in Example 4 ( Figure 8A), and data generated therefrom compiled.
  • Figures 1 OA- IOC show that while keeping the total PF4 concentration constant, as the percentage of WT PF4 mixed with PF4 K50E is decreased,
  • Example 11 Inhibition of Endothelial Activation using ULC antagonists
  • the 15 compounds from Example 1 are examined for their ability to inhibit HITT related cellular activation. Specifically, the ability of these compounds to inhibit platelet activation from HITT antibodies is measured. These compounds are also analyzed for their ability to inhibit PF4 activation of NFKB in endothelial cells in vitro.
  • Example 1 The 15 compounds from Example 1 are evaluated for their ability to inhibit cellular activation related to HIT using the assays described in Warkentin, "Heparin- induced thrombocytopenia: diagnosis and management", Circulation, 110(18):e454-8 (2004) and Kelton, "Heparin-induced thrombocytopenia: a historical perspective", Blood, 1 12(7):2607-16 (2008).
  • Example 1 The 15 compounds from Example 1 are analyzed for their impact on the ability of PF4 to activate NFKB in endothelial cells as described in Yu, "Endothelial expression of E-selectin is induced by the platelet-specific chemokine platelet factor 4 through LRP in an NF- ⁇ kappa ⁇ B-dependent manner", Blood, 105(9):3545 (May 1, 2005), which is hereby incorporated by reference using human umbilical vein endothelial cells (HUVECs).
  • UAVECs human umbilical vein endothelial cells
  • Example 12 Prevention of Thrombocytopenia and Thrombosis in a Murine Model using ULC Antagonists
  • Example 1 The 15 compounds from Example 1 are evaluated for their ability to decrease thrombocytopenia and thrombosis in the HITT murine model system as described in Reilly I and Reilly II cited above and incorporated herein by reference.
  • the murine model system consists of mice homozygous for FcyRIIa and hPF4 and null for mPF4, hereafter designated IIa +/+ /hPF4 +/+ /mPF4 ⁇ /_ .
  • mice IIa +/+ /hPF4 +/+ /mPF4 -/_ mice are weaned at 3 weeks of age and fed on SD (Lab Rodent Chow, Purina 5020; 4.5% fat).
  • SD Lab Rodent Chow, Purina 5020; 4.5% fat.
  • sex- and weight-matched littermates are either maintained on the SD (normocholesterolemic controls) or switched to a hypercholesterolemic diet (HD).
  • the HD contains 15% cocoa butter, 1% cholesterol and 0.5% sodium cholate, in addition to essential nutrients, vitamins and minerals (TD.88051, Harlan-Teklad, Madison, WI, USA). HD-fed animals remain on the diet for only 4 weeks ⁇ i.e., until 10 weeks of age).
  • Mice are anesthetized by isofluorane inhalation for i.p. and s.c. injections and for blood collections. Blood is collected via the retro-orbital plexus using 50 ⁇ , EDTA- or heparin-coated microcapillary tubes (Microcaps® tubes, Drummond Scientific, Broomall, PA, USA).
  • Plasma cholesterol is measured in all mice at 6 weeks of age (i.e., before initiation of the HD in the experimental group) and weekly thereafter. Mice in both diet groups are fasted for 4 hours before each blood collection. Total plasma cholesterol is determined by enzymatic assay (Thermo Electron, Louisville, CO, USA). Reactivity of platelets from HD-fed mice and SD-fed controls is evaluated at the age of 10 weeks by whole blood impedance aggregometry (the Chronolog® 540- VS instrument, Chronolog Corp., Havertown, PA, USA). Whole blood, collected in heparin, is mixed with 0.9% NaCl to adjust the final platelet count to 150,000 ⁇ /L.
  • Platelet aggregation is induced by collagen (1.5 ⁇ g/mL) and measured as the increase in impedance over time.
  • Plasma levels of soluble vascular cell adhesion molecule (sVCAM) are measured by ELISA (Mouse sVCAM Immunoassay, R & D Systems, Minneapolis, M , USA).
  • each mouse receives i.p. injections of an antagonist compound of Example 1, followed by daily s.c. injections of unfractionated heparin (1200 or 1400 U/kg).
  • Blood samples are collected from each mouse in EDTA, beginning 3 days before antagonist compound administration and are utilized as control samples 1-13, and then on days 1, 2, 3, 4 and 7 after antagonist compound injections.
  • Complete blood counts on all 26 samples are measured using a Hemavet® analyzer (Model 850, CDC Technologies, Oxford, CT, USA). Platelet counts are monitored to assess thrombocytopenia. Red blood cell counts and hematocrit are monitored to ensure against unapparent hemorrhage.
  • Thrombin-antithrombin complex III (TAT) levels are measured by an ELISA (Enzygnost® TATmicro reagents, Dade Behring, Newark, DE, US).
  • mice exhibiting signs of distress subsequent to heparin and antagonist compound injections are sacrificed by CO 2 inhalation.
  • the major organs (lungs, heart, liver, kidneys, and spleen) are immediately harvested and fixed in neutral-buffered 10% formalin.
  • paired SD-fed mice are sacrificed at the same time.
  • Formalin-fixed organs are embedded in paraffin and cut into 5- ⁇ sections. Hematoxylin and eosin-stained sections are evaluated by a comparative pathologist blinded to the experimental conditions. The area of each tissue examined is measured using the NIH ImageJ® software and the number of thrombi per 10 mm 2 are determined.
  • sections are sequentially blocked with 3 ⁇ 4(3 ⁇ 4 in methanol (to inhibit endogenous peroxidase activity) and 10% mouse serum before staining with directly biotinylated anti-hPF4 antibodies.
  • specificity for hPF4 is confirmed by staining mouse spleens, where antibodies react specifically with megakaryocytes.
  • the bound antagonist compounds are detected by incubating biotinylated horseradish peroxidase plus avidin followed by staining with diaminobenzidine. Immunohistochemistry sections are then lightly counterstained with hematoxylin and examined by light microscopy.
  • mice treated with the 15 compounds of Example 1 did not develop thrombocytopenia or that such treatment decreases the severity of thrombocytopenia. It is also expected that the data will show that mice treated with the 15 compounds of Example 1 did not develop thrombosis. Finally, it is expected that these data will demonstrate that the 15 compounds of Example 1 prevent formation of PF4 tetramers and PF4 tetramenheparin ULCs when administered concurrently with heparin administration.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Dermatology (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Emergency Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Description

SMALL MOLECULE ANTAGONISTS OF PF4 CONTAINING ULTRA LARGE COMPLEXES
This invention was made with government support under Grant Nos.
5R01HL078726-04 and 3R01HL078726-04-S1 awarded by the National Institutes of Health. The government has certain rights in the invention.
BACKGROUND OF THE INVENTION
Heparin-induced thrombocytopenia (HIT) and thrombosis (HITT) are serious complications of heparin therapy. HITT is a severe prothrombotic disease, with affected individuals having a 20-50% risk of developing new thromboembolic events, and has a mortality rate of about 20% with an additional about 10% of patients requiring amputations or suffering other major morbidity. Since a large number of hospitalized patients are exposed to heparin, HITT is a major iatrogenic cause of morbidity and mortality in this patient population. No specific therapies to treat HITT are reported. Management consists of general measures such as withdrawal of heparin, use of a non-heparin anticoagulant, and supportive care.
PF4 is a 70 amino acid, lysine-rich 7.8 kDa platelet-specific protein that belongs to the CXC (or beta) chemokine subfamily. PF4 is synthesized by megakaryocytes and comprises 2-3% of the total released protein in mature platelets. PF4 exists as a tetramer in the a-granules of platelets and is secreted in high concentrations when platelets are activated. PF4 tetramer binds avidly to
glycosaminoglycans (GAGs), but only a few high affinity protein receptors have been identified to date. The interaction of PF4 with GAGs is central to the pathogenesis of heparin induced thrombocytopenia as well as thrombosis.
Rauova, "Ultralarge Complexes of PF4 and heparin are central to the pathogenesis of heparin-induced thrombocytopenia", Blood, 105(1): 131 (January 1, 2005), which is hereby incorporated by reference, found that heparin:PF4 complexes exist in a dynamic equilibrium and that ULCs are subject to dissociation under certain conditions. Although PF4 crystallizes as a tetramer, in solution it exists in a dynamic equilibrium between monomeric, dimeric and tetrameric forms. The dimers associate to form the tetramer with energy contributed by salt bridges. See, Petersen, "A chondroitin sulfate proteoglycan on human neutrophils specifically binds platelet factor 4 and is involved in cell activation", J. Immunol, 161 :4347 (1998), which is hereby incorporated herein by reference.
Similarly, Zhang, "Crystal structure of recombinant human platelet factor 4",
Biochemistry, 33:8361-8366 (1994), which is hereby incorporated herein by reference, noted the charge-charge interactions (salt bridges) between the positively charged lysine residues (Lys50 in the monomer) and the negatively charged glutamic acid residues (Glu28 in the monomer) stabilize the tetrameric form of PF4.
Krauel, "Heparin-induced thrombocytopenia - therapeutic concentrations of danaparoid, unlike fondaparinux and direct thrombin inhibitors, inhibit formation of platelet factor 4-heparin complexes". J. Thrombosis and Haemostatis, 6:2160-2167 (2008), which is hereby incorporated herein by reference, recently reported that treatment of HITT patients with danaproid (a glycosaminoglycan mixture approved for anticoagulation in HITT) may decrease heparin:PF4 complex size, resulting in less antibody mediated platelet activation.
Reilly, "Prothrombotic factors enhance heparin-induced thrombocytopenia and thrombosis in vivo in a mouse model", J. Thrombosis and Haemostatis, 4:2687-2694 (2006) (Reilly I) and Reilly, "Heparin-induced thrombocytopenia/thrombosis in a transgenic mouse model requires human platelet factor 4 and platelet activation through FcyRIIA", Blood, 98(8):2442-2447 (October 15, 2001) (Reilly II), which are hereby incorporated herein by reference, discussed that the clinical manifestations of HITT are caused by antibodies that recognize a complex composed of heparin and PF4. The transgenic mouse model of HITT conclusively demonstrated that heparin, PF4, antiheparin/PF4 antibody, and platelet FcyRIIa are necessary and sufficient to recapitulate the salient features of HITT in mice.
There is a need in the art for compounds and methods for preventing or treating conditions related to the formation of PF4 tetramers, such as HITT. SUMMARY OF THE INVENTION
In one aspect, methods for (i) preventing formation of, or (ii) disrupting PF4 tetramers, are provided and include administering a compound to a subject. The compounds useful in such methods bind to PF4 dimers or PF4 monomers. The compounds also disrupt the salt bridges of PF4 tetramers.
In another aspect, methods for (i) preventing formation of or (ii) disrupting ultra-large complexes (ULCs) containing a PF4 tetramer and a glycosaminoglycan (GAG) and include administering a compound to a subject. The compounds useful in such methods bind to PF4 dimers or PF4 monomers or disrupt the salt bridges of PF4 tetramers.
In still a further aspect, methods for (i) preventing medical conditions related to the formation of PF4 tetramers, such as HITT and atherosclerosis, (ii) treating medical conditions related to the formation of PF4 tetramers, such as HITT, (iii) correcting a platelet imbalance, (iv) preventing a decrease in platelet production, (v) increasing high density lipoproteins in a subject, (vi) preventing or treating inflammation, or (vii) modulating clotting or hemostasis are provided. The compounds useful in such methods bind to PF4 dimers or PF4 monomers or disrupt the salt bridges of PF4 tetramers.
In yet another aspect, the compounds useful in these methods and
compositions noted above contain a moiety of the following structure, wherein Rx,
RY, Rx', RY', X, n, and n' are defined herein:
O
P— (CRxRY)n(CRxCRY)rf-X^v
HO . Pharmaceutically acceptable salts or prodrugs of such compounds are also useful in the methods and compositions described herein.
In still a further embodiment, compositions and kits are provided and contain (i) a pharmaceutically acceptable carrier and (ii) a compound as described above or a pharmaceutically acceptable salt or prodrug thereof.
In another embodiment, compositions and kits are provided and contain (i) a medication which causes the formation of PF4 tetramers, a medication which disrupts PF4 tetramers, or a combination thereof; and (ii) a compound as described above or a pharmaceutically acceptable salt or prodrug thereof.
Other aspects and advantages of the invention will be readily apparent from the following detailed description of the invention. BRIEF DESCRIPTION OF THE DRAWINGS
Figures 1A and IB are molecular modeling diagrams, shown in two orientations, representing the binding of a small molecule to the dimer interface on PF4 (here shown as a dimer), i.e., antagonist, described herein. The antagonistic compound is light gray and the PF4 tetramer is black. Figure IB contains the molecular model of Figure 1A and rotated 90°.
Figure 2 is a distribution of DOCK scores over 1.1 million compounds and found to be potential ULC antagonists. Dock scores are empirical with lower values representing higher affinity for PF4. As noted, the docking scores approximate a Gaussian distribution with a mean value of -26.6 and standard deviation (sd) of 3.3. The 100 compounds identified scored more than 10 sd below the mean (-73.8 or less).
Figure 3 is a bar graph of data obtained using photon correlation spectroscopy on a DynaPro® DLS instrument and Dynamics® software (V6.7.6; Microsoft) to obtain correlation function. Samples analyzed included PF4 incubated in the absence or presence of unfractionated heparin (UFH) for 20 minutes at room temperature. The bar graph shows the percentage of ULCs and small PF4 particles. The solid bars include data for the small particles representing a population of particles with a mean hydrodynamic diameter of about 1 nm. The striped bars include data for the large particles representing a population of particles with a mean hydrodynamic diameter of about 300 nm. Data are expressed as the percent of total intensity from each measurement, the mean of 10 measurements, and representative of two such experiments.
Figures 4A and 4B are bar graphs of data obtained from SDS PAGE of PF4 after BS3 crosslinking and show the ability of compounds 34, 24, 1, 80, 88 and 96 to inhibit formation of PF4 tetramers. Compounds 34, 24, 1, and 96 were tested at 0.1 mM and 1 mM and compounds 80 and 88 were tested at 0.1 mM, 1 mM, and 5 mM.
The solid bars represent PF4 tetramer formed; dotted bars represent PF4 trimer formed; striped bars represent PF4 dimer formed; and the brick-like bars represent
PF4 monomer present in the samples.
Figure 5 is a quantitation of data obtained from a dynamic light scattering
(DLS) analysis as function of ULC diameter vs. the % inhibition using compound 96.
Specifically, Figure 5 provides a bar graph mapping the concentration (μΜ) of compound 96 as a function of small PF4 particles (solid bar) and large ULCs (striped bar) formed.
Figure 6 is a bar graph demonstrating the ability of compound 96 to inhibit serotonin release. The graph provides the percentage of serotonin released using compound 96, as compared to a sample, i.e., buffer, containing no antagonistic compound.
Figure 7 is a line graph illustrating the dose response inhibition of PF4 tetramer formation using compound 1 (filled circles (·) at 250 μΜ), 34 (filled triangles (T ) at 500 μΜ), 80 (open triangles (Δ) at 250 μΜ), 101 (open circles (o) at 250 μΜ, and 96 (open diamonds (0) at 500 μΜ). The antagonists were incubated at the noted concentrations with PF4 (10 μg/mL) for 60 minutes at room temperature, followed by the addition of cross-lining reagent BS (0.2 mM). The data are the mean ± SEM of at least three independent experiments. Curves represent fit of data to the indirect Hill equation.
Figure 8 A is a line graph which illustrates the ability of compounds of the invention to inhibit the formation and stability of PF4:heparin ULCs as measured by ELISA. Compounds 1 (filled circles; ·), 34 (filled triangles; T), 80 (open triangles; Δ), 101 (open circles; o), and 96 (open diamonds; 0) solutions in varying
concentrations were preincubated with PF4 (7.5 μg/mL) for 1 hour, followed by addition of heparin (0.2 U/mL). Data are the mean ± SEM of at least three independent experiments performed in triplicate. Curves represent fit of data to the indirect Hill equation.
Figure 8B is a bar graph of compiling data from experiments showing the ability of compounds of the invention at varying concentrations to inhibit ULC formation formed using varying concentrations of heparin. One mM of compounds 1, 34, 80, 101, and 96 were preincubated with PF4 (7.5 μg/mL) for 1 hour, followed by addition of heparin (0.2 U/mL). One control contained PF4 and heparin (0.2 U/mL) and the other control contained PF4K50E (7.5 μg/mL) and heparin (0.2 U/mL). Data are representative of two or more experiments.
Figure 8C illustrates the interaction of ULCs, formed using varying concentrations of heparin, with the PF4 antagonists of the invention. Briefly, 1 mM of compounds 1, 34, 80, and 101 were preincubated with PF4 (7.5 μg/mL) for 1 hour, followed by addition of varying concentrations of heparin (0.05, 0.1, 0.2, 0.4, 0.8, and 1.6 U/mL). The controls contained PF4 (7.5 μg/mL) and varying concentrations of heparin (0.05, 0.1, 0.2, 0.4, 0.8, and 1.6 U/mL)
Figure 8D demonstrates antibody binding to ULCs in the absence and presence of PF4 antagonists of the invention. Briefly, ULCs, prepared by incubating PF4 (5 μg/mL) and heparin (0.2 U/mL) and diluting the mixture to a final PF4 concentration (0.1 μg/mL), were incubated with 1 mM of each of compounds 1, 34, 80, 101, and 96. These mixtures were then incubated in wells pre-coated with KKO. Data are the mean ± SEM of at least three independent experiments performed in triplicate.
Figure 8E shows the dose response of the PF4 antagonists of the invention in disrupting preformed ULCs. ULCS were formed by incubating PF4 (5 μg/mL) and heparin at a 1.5: 1 ratio for 30 minutes. Independent solutions were prepared containing PF4 antagonist 1 (2 runs: 125 μΜ and 63 μΜ), 80 (250 μΜ), 101 (500 μΜ), 96 (1 mM), and 34 (2 mM) and the ULC presence was measured. Data are mean ± SEM of at least three independent experiments performed in triplicate.
Figures 9A-9F demonstrate FcyRIIA-mediated activation of cells and inhibition by PF4 antagonists.
Figure 9A is a bar graph showing the activation of DT40 cells transfected with FcyRIIA and a Luc reporter. The basal condition is a buffer only control; Hep is heparin; IV.3 is the anti-FcyRIIA monoclonal antibody in the presence of anti-lgG antibody.
Figure 9B is a line graph providing the dose-response of DT40 activation by heparin in the presence of constant amounts of PF4.
Figure 9C provides the dose-response of compounds 1 (·), 34 (T), 80 (Δ),
101 (o), and 96 (0) in inhibition of DT40 activation. Data are the mean ± SEM of at least three independent experiments performed in triplicate. Curves C represent fit of data to the indirect Hill equation.
Figure 9D illustrates the inhibition of DT40 activation by plasma obtained from three HIT patients. The control (white bar) represents data obtained in the absence of any PF4 antagonist. The dotted bars are the compilation of data for compound 1 ( ; - the middle bar) and the striped bars (the right bars) are the compilation of data for compound 101. Compounds 1 and 101, both at concentration of 100 μΜ, inhibited activation by KKO in patient samples. Data represent the mean ± SEM of at least two independent experiments performed in triplicate.
Figure 9E provides a bar graph illustrating activation of platelets as measured by release of 14C-5HT. Data are representative of at least two experiments.
Compounds 1, 34, 80, 101, and 96 were present at a concentration of 2.5 mM.
Figure 9F was generated similar to Figure E except is a line graph of activation (% max) vs. concentration of the noted PF4 antagonist ([PF4A]).
Activation was by plasma from patients with HIT using compounds 1 (circles; ·) and 101 (squares;□).
Figures lOA-C are line graphs and illustrate inhibition of PF4 tetramerization,
ULC formation, and FcyRIIA using the non-tetramerizing mutant PF4K50E. % WT represents the % of total PF4 containing WT PF4, with the remainder comprised of PP4K50E_
Figure 1 OA is a line graph of the presence of PF4 tetramer vs. wt PF4. PF4 and PR4K50E (total mass 10 μg/mL) were incubated for 60 minutes at room temperature, followed by the addition of cross-lining reagent BS (0.2 mM). The data are the mean ± SEM of at least three independent experiments.
Figure 10B is a line graph of the presence of ULC vs. wt PF4. PF4 and PF4K50E (total mass 7.5 μg/mL) were for 1 hour, followed by addition of heparin (0.2 U/mL) and detected by ELISA as in figure 8A. Data are the mean ± SEM of at least three independent experiments performed in triplicate.
Figure IOC is a line graph of FcyRIIA activation vs. wt PF4. PF4
concentrations were maintained constant by diluting wt PF4 with PF4K50E. Data are the mean ± SEM of at least three independent experiments performed in triplicate.
DETAILED DESCRIPTION OF THE INVENTION
The inventors determined that compounds which prevent formation of or disrupt the PF4 tetramer or ULC containing the same, i.e., antagonists, are useful in treating and/or preventing diseases related to the same. Such conditions include HITT since heparin:PF4 complexes are central to the pathophysiology of HITT as the targets of the pathogenic antibodies. Specifically, the most pathogenic complexes of heparin and PF4 are very large and are referred to as ultralarge complexes (ULC). Heparin:PF4 complexes smaller than 600 kDa are typically referred to as small complexes (SC). In one embodiment, ULCs are 600 kDa or larger. In another embodiment, ULCs are 670 kDa or larger.
The term "PF4" as used herein refers to platelet factor 4 which is a 70 amino acid, lysine-rich, 7.8 kDa platelet-specific protein that belongs to the CXC (or beta) chemokine subfamily, in which the first two of the four conserved cysteine residues are separated by one amino acid residue. In one embodiment, PF4 is naturally occurring, i.e., wild-type. In another embodiment, PF4 may be synthesized by recombinant or chemical methods. The term PF4 also refers to mutations thereof in which one or more of the amino acids is replaced with a different amino acid.
Examples of PF4 mutations are described in International Patent Publication No. WO 02/006300, which is incorporated herein by reference. In one example, the mutated PF4 contains Arg28 and Glu50 mutations.
PF4:heparin ULCs are more pathogenic than heparin:PF4 SCs. Heparin:PF4
ULCs are better recognized by HITT antibodies and lead to more platelet activation in the presence of these antibodies. Therefore, disruption of ULC represents a valid and novel therapeutic target in the treatment of HITT.
PF4 tetramers and complexes formed therewith contribute to the onset or development of individuals at risk for certain diseases or medical conditions.
Therefore, compounds which disrupt PF4 tetramers or prevent their formation, i.e., antagonists, are desirable. By doing so, the antagonists are useful in preventing of treating these diseases or medical conditions. The term "antagonist" as used herein refers to any chemical compounds which are capable of binding to a PF4 tetramer, dimer or monomer. By doing so, the compound is thereby capable of preventing formation of or disrupting ULCs formed between the PF4 tetramer. Specifically, ULCs formed between glycosaminoglycans (GAG) and PF4 tetramers are responsible for the onset or development of individuals at risk for certain diseases or medical conditions. Therefore, desirable compounds are those which disrupt or prevent the formation of the PF4 tetramer and ULCs formed therewith. By doing so, the antagonist compound prevents formation of, or disrupts, ULCs. I. Compounds Useful in the Invention
The inventors determined that compounds having acidic moieties bound directly or indirectly to a heteroatom bind to PF4 monomers, PF4 trimers, PF4 dimers, and PF4 tetramers. These compounds also disrupt the salt bridge of PF4 tetramers and thereby inhibit formation of the PF4 tetramer. The compounds useful herein prevent the formation of the PF4 tetramer by binding to specific sites at the PF4 trimer, dimer or monomer. Alternatively, the compounds are capable of competing with the electrostatic interactions between the PF4 monomers, dimmers, and trimers in the PF4 tetramer and successfully disrupt to the salt bridge of the tetramer. See, Figure 1. In effect, the functional groups of the compounds described herein form a stronger bond or interaction with the PF4 monomer, dimer, and trimer than they do with each other.
The salt bridge of the PF4 tetramer is formed via electrostatic interactions of a negatively charged amino acid, i.e., glutamic acid of a first PF4 monomer or PF4 dimer and a positively charged amino acid, i.e., lysine, of a second PF4 monomer or PF4 dimer. This salt bridge is typically formed by at least via the interaction of at least one Glu and one Lys on a first PF4 monomer or PF4 dimer and at least one Lys and one Glu on a second PF4 monomer or PF4 dimer. In one embodiment, the salt bridge is formed via at least electrostatic interactions between Lys50 in a first PF4 monomer Glu28 in a second PF4 monomer. In a further embodiment, the salt bridge is formed via at least electrostatic interactions between Glul28, Lys350 of a first PF4 monomer or dimer and Glu328, Lysl50 of a second PF4 monomer or dimer. In another embodiment, the salt bridge is formed via at least electrostatic interactions between Glu228, Lys450 of a first PF4 monomer or dimer and Glu428, Lys250 of a second PF4 monomer or dimer.
In one embodiment, the compound may contain one or more of an acidic moiety. The term "acidic moiety" as used herein refers to a chemical group on the backbone of a molecule which is capable of losing a hydrogen atom from the moiety. Such acidic moieties are readily identified by one of skill in the art. In one embodiment, the acidic moiety contains a ~C(=0)~, carboxylic acid (~C(=0)OH), or carboxylic ester group (~C(=0)0~). In another embodiment, the acidic moiety contains a ~C(=S)~, thiocarboxylic acid (~C(=S)OH, ~C(=0)SH, or ~C(=S)SH), or thiocarboxylic ester group (~C(=S)0~, ~C(=0)S~, or ~C(=S)S~). In a further embodiment, the acidic moiety contains a P(=0), phosphinic acid (~P(=0)OH), phosphonic acid (~P(=0)(OH)2), or phosphoric acid (~P(=0)(OH)3) group. In still another embodiment, the acidic moiety contains a P(=S), thiophosphinic acid (~P(=S)OH, ~P(=0)SH, or ~P(=S)SH), thiophosphonic acid (~P(=S)(OH)2, ~P(=0)(OH)(SH), ~P(=0)(SH)2, ~P(=S)(OH)(SH), or ~P(=S)(SH)2), or
thiophosphoric acid (~P(=S)(OH)3, ~P(=0)(SH)(OH)2, ~P(=0)(OH)(SH)2,
~P(=S)(SH)(OH)2, ~P(=S)(OH)(SH)2, or ~P(=S)(SH)3) group. In yet a further embodiment, the acidic moiety contains a S(=0), sulfuric acid (~S(=0)2(OH)0~), sulfonic acid (~S(=0)2(OH)), or sulfinic acid (~S(=0)(OH)) group.
The acidic moiety is desirably bound directly or indirectly to a heteroatom- containing moiety. The heteroatom-containing moiety contains at least one heteroatom. In one embodiment, the acidic moiety is bound directly to the heteroatom. In another embodiment, the acidic moiety is bound indirectly to the heteroatom through a hydrocarbon chain which is optionally unsaturated and optionally contains one or more additional heteroatom. In a further embodiment, the acidic moiety is bound indirectly to the heteroatom through a cyclic hydrocarbon which is optionally unsaturated and optionally contains one or more additional heteroatom. The heteroatom present in the compounds useful herein may be selected by one skilled in the art and may include, O (~OH, (=0), or ~0~), S (~SH, (=S) or ~S~), or N atoms, among others. The N-atom may be a primary amine (~NH2), secondary amine (~NH~), tertiary amine (~N~), or may be doubly bound to another moiety such as a carbon atom. One of skill in the art would readily understand that the use of - in the above-noted description indicates that the atom to which it is attached is bound to another chemical moiety. Compound (I)
In one embodiment, the compound may contain a phosphorus atom in the backbone such as in a moiety of structure (I):
X'
vwX" (CRUCRV)G(CRU'CRV')G. P (CRXRY)N(CRX'CRY')N. ΧΛΛΛ
(OH)D
(I)
wherein,
g, g', n and n' are, independently, 0 to 10, i.e., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; d is 0, 1, or 2;
X and X" are, independently, (i) absent, (ii) O, S, N, or P; or (iii) a ring containing one or more of O, S, N, or P;
X' is O or S;
Ru, Rv, Ru', Rv', Rx, RY, Rx', and RY' are, independently, H, OH, halogen, C3 to Cio cycloalkyl, Ci to Cio thioalkyl, aryl, heteroaryl, heterocycle, SO2H, OP(0)OH, P(0)OH, Ci to Cio alkyl substituted at one or more carbon-atom by RA, C2 to Cio alkenyl substituted at one or more carbon-atom by RA, or C2 to Cio alkynyl substituted at one or more carbon-atom by RA; or
Ru and Rv, Ru' and Rv', Rx and RY or Rx' and RY' are joined to form a ring containing C3 to Cio cycloalkyl, aryl, heterocycle, or heteroaryl, wherein the ring is optionally substituted by one or more RA; and
RA is H, Ci to Cio alkyl, Ci to Cio alkoxy, (=0), (=S), halogen, C(0)(Ci to Cio alkyl), aryl, heteroaryl, heterocycle, OH, C(0)OH, S02H, OP(0)OH, P(0)OH, P(0)(OCi to Cio alkyl)2, Ci to C10 aminoalkyl, Ci to Cio thioalkyl, or SH.
In a further embodiment, the compound may contain a ring having at least one phosphorus and one oxygen-atom in the backbone of the ring. In one example, the compound may contain a ring having one phosphorus and two oxygen-atoms in the backbone of the ring. In another example, the compound may contain a ring having one phosphorus, one oxygen, and one nitrogen-atom in the backbone of the ring. In a further example, the phosphorus atom forms the backbone of two rings and is the point of attachment for the two rings. The phosphorus atoms may be substituted with one or more of OH, Ci to Cio alkoxy, or Ci to Cio alkyl. The nitrogen-atoms may be substituted with one or more of OH, Ci to C10 alkoxy, or Ci to C10 alkyl, may be part of a five to fourteen membered heterocyclic or heteroaryl ring, or may be bound to another nitrogen-atom of the compound. These rings may be optionally substituted with Ci to Cio alkyl, among other substituents.
In another embodiment, the compound may contain a sulfur atom in the backbone such as a moiety of structure (IA):
Figure imgf000013_0001
(IA)
wherein: M is O or N; k and k' are, independently, 0 to 10, i.e. , 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; RD, RE, RD', and RE' are, independently, H, OH, halogen, C3 to C10 cycloalkyl, Ci to C10 thioalkyl, aryl, heteroaryl, heterocycle, SO2H, OP(0)OH,
P(0)OH, Ci to Cio alkyl substituted at one or more carbon-atom by RA, C2 to C10 alkenyl substituted at one or more carbon-atom by RA, or C2 to Cio alkynyl substituted at one or more carbon-atom by RA; or RD and RE or RD and RE are joined to form a ring containing C3 to Cio cycloalkyl, aryl, heterocycle, or heteroaryl, wherein the ring is optionally substituted by one or more RA; or one carbon atom of CRD, CRE, CRD , or CRE is bound to the SO2 to form a C3 to Cio cycloalkyl or aryl optionally substituted by one or more RA; and RA is H, Ci to Cio alkyl, Ci to Cio alkoxy, (=0),
(=S), halogen, C(0)(Ci to Cio alkyl), aryl, heteroaryl, heterocycle, OH, C(0)OH, OS02, S02H, OP(0)OH, P(0)OH, P(0)(OCi to Cio alkyl)2, d to Cio aminoalkyl, Ci to Cio thioalkyl, or SH; or a pharmaceutically acceptable salt or prodrug thereof.
In still a further embodiment, the compound may contain a carboxylic acid
(C(=0)OH) group in the backbone such as a moiety of structure (IA):
o
HO C (CRKRL)S(CRK'RL")S. >A w
(IB)
wherein, s and s' are, independently, 0 to 10, i.e., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; RK, RL, RK', and RL' are, independently, H, OH, halogen, C3 to Cio cycloalkyl, Ci to Cio thioalkyl, aryl, heteroaryl, heterocycle, S02H, OP(0)OH, P(0)OH, Ci to Cio alkyl substituted at one or more carbon-atom by RA, C2 to Cio alkenyl substituted at one or more carbon-atom by RA, or C2 to C10 alkynyl substituted at one or more carbon-atom by RA; or RK and RL or RK and RL are joined to form a ring containing C3 to C10 cycloalkyl, aryl, heterocycle, or heteroaryl, wherein the ring is optionally substituted by one or more RA; or RA is H, Ci to C10 alkyl, Ci to C10 alkoxy, CN, (=0), (=S), halogen, C(0)(Ci to C10 alkyl), aryl, heteroaryl, heterocycle, OH, C(0)OH, OS02, S02H, OP(0)OH, P(0)OH, P(0)(OCi to C10 alkyl)2, d to C10 aminoalkyl, Ci to C10 thioalkyl, or SH; or a pharmaceutically acceptable salt or prodrug thereof.
Alternatively, the compound contains an anhydride group to form a moiety of structure (IC):
Figure imgf000014_0001
(IC)
wherein, RK, RL, and RP are, independently, H, C3 to C10 cycloalkyl, Ci to C10 thioalkyl, aryl, heteroaryl, heterocycle, Ci to C10 alkyl substituted at one or more carbon-atom by RA, C2 to C10 alkenyl substituted at one or more carbon-atom by RA, or C2 to Cio alkynyl substituted at one or more carbon-atom by RA; or RK and RL, RK and Rp, or RL and Rp are joined to form a ring containing C3 to C10 cycloalkyl, aryl, heterocycle, or heteroaryl, wherein the ring is optionally substituted by one or more RA; wherein at one of RK, RL, or Rp contains a heteroatom as described above; and RA is H, Ci to Cio alkyl, Ci to C10 alkoxy, CN, (=0), (=S), halogen, C(0)(Ci to C10 alkyl), aryl, heteroaryl, heterocycle, OH, C(0)OH, OS02, S02H, OP(0)OH, P(0)OH, P(0)(OCi to Cio alkyl)2, Ci to Cio aminoalkyl, Ci to Cio thioalkyl, or SH; or a pharmaceutically acceptable salt or prodrug thereof.
Compound (II)
In another embodiment, the compound has structure (II):
Figure imgf000014_0002
(II)
wherein, n is 0 to 5, i.e., 0, 1, 2, 3, 4, or 5; R1 and R2 are, independently, H, Ci to Cio alkyl, C(0)(Ci to Cio alkyl), C(0)(d to Cio alkyl), benzyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to C10 alkynyl; or R1 and R2 together comprise a heterocycle or heteroaryl, optionally substituted by one or more R5; R3 and R4 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, benzyl, aryl, heteroaryl, or heterocycle; or R1 and R3 together comprise a heterocycle or heteroaryl, each optionally substituted by one or more R5; R5 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, or (=0); or a pharmaceutically acceptable salt or prodrug thereof.
In a further embodiment, the compound has structure (Ila), wherein R2 is H, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl:
HO N— R2
(Ila)
In still another embodiment, the compound is selected from among:
Figure imgf000015_0001
pharmaceutically acceptable salts or prodrugs thereof.
In yet a further embodiment, the compound is selected from among:
Figure imgf000015_0002
, or pharmaceutically acceptable salts or prodrugs thereof
Compound (III)
The compounds described herein may also have structure (III)
OR16
I
0^P (CR17R18)q-R10
(III) wherein, q is 0 to 5, i.e., 0, 1, 2, 3, 4, or 5; R is Ci to C10 alkyl. heteroaryl or aryl, each optionally substituted by one or more R19; R16 is H or Ci to C10 alkyl; R17 and R18 are, independently, absent, H, Ci to C10 alkyl, (Ci to C10 alkyl)-P-(C1 to C10 alkyl), Ci to C10 alkoxy, halogen, OH, N02, CN, C2 to C10 alkenyl, P(0)OH, C2 to C10 alkynyl, aryl, heteroaryl, or heterocycle, each optionally substituted by one or more
R 19 ; wherein when R 17 or R18 is absent and q is at least 2, the CR 17 R18 groups are bound through a stable double bond; R19 is H, Ci to C10 alkyl, Ci to C10 alkoxy, halogen, N02, CN, C(0)OH, C(0)(Ci to C10 alkyl), C2 to C10 alkenyl, or C2 to C10 alkynyl; or a pharmaceutically acceptable salt or prodrug thereof.
In a further embodiment, the R10 or R11 heterocycle is a five or six membered heterocycle and contains one O or S heteroatom.
In a further embodiment, the compound has structure (Ilia):
Figure imgf000016_0001
(Ilia)
wherein, each R is, independently, H, Ci to C10 alkyl, Ci to C10 alkoxy, halogen,
N02, CN, C(0)OH, C(0)(Ci to C10 alkyl), C2 to C10 alkenyl, or C2 to C10 alkynyl; R14 and R15 are, independently, H, Ci to C10 alkyl, C3 to C10 cycloalkyl, C2 to C10 alkenyl, C2 to Cio alkynyl, benzyl, aryl, heteroaryl, or heterocycle; R16 is H or Ci to C10 alkyl; R17 and R18 are, independently, absent, H, Ci to C10 alkyl, Ci to C10 alkoxy, halogen, OH, N02, CN, C2 to Cio alkenyl, P(0)OH, C2 to C10 alkynyl, aryl, heteroaryl, or heterocycle; wherein when R17 or R18 is absent and q is at least 2, the CR17R18 groups are bound through a stable double bond; or a pharmaceutically acceptable salt or prodrug thereof.
In another embodiment, R10 is a pyrrolidine and contains a (=0) in the backbone of said pyrrolidine.
In yet another embodiment, the compound has structure (Illb), wherein s is 1 to 10:
Figure imgf000016_0002
(nib)
In still another embodiment, R10 is pyridine.
In another embodiment, the compound has structure (IIIc):
Figure imgf000017_0001
(IIIIc)
In a further embodiment t,, tthhee ccooimpound has structure (Hid), wherein R is aryl, heterocycle, or heteroaryl.
Figure imgf000017_0002
In another embodiment, the compound is of structure (Hid) and R10 and R11 are furan.
In a further embodiment, the compound has structure (Hie):
Figure imgf000017_0003
(me)
In yet another embodiment, the compound is selected from among:
Figure imgf000017_0004
or pharmaceutically acceptable salts or prodrugs thereof.
The compound may further have structure (Illf):
Figure imgf000017_0005
(Illf) wherein, r is 1 to 3, i.e., 1, 2, or 3; X is O, S, or NR30; R30 is H or Ci to C10 alkyl; R31 and R32 are, independently, H, Ci to Cio alkyl, C3 to C10 cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl, benzyl, aryl, heteroaryl, or heterocycle; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, the comp has structure (Illg), wherein r is 1 to 3 :
Figure imgf000018_0001
or a pharmaceutically acceptable salt or prodrug thereof.
In still another embodiment the compound is selected from among:
Figure imgf000018_0002
Figure imgf000018_0003
, or pharmaceutically acceptable salts or prodrugs thereof. Compound (IV)
The compounds useful herein may further have structure (IV):
Figure imgf000019_0001
(IV)
wherein, m is 0 to 2, i.e., 0, 1, or 2; R20 is H, Ci to C10 alkyl C3 to C10 cycloalkyl, C2 to Cio alkenyl, or C2 to C10 alkynyl, each optionally substituted by one or more R24:
0 00 o~x
R , R , and R" are independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl, each optionally substituted by one or more R24; R24 is H, halogen, OH, CN, N02, d to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R27; R25 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl; R26 is O or S; R27 is H, P(0)OH, or Ci to Cio alkyl optionally substituted by halogen or OP(0)OH; or a pharmaceutically acceptable salt or prodrug thereof.
0 00 o~x
In one embodiment, the compound is of structure (IV) and R , R , and R are Ci to Ce alkyl.
In another embodiment, the compound has structure (IVa):
Figure imgf000019_0002
(IVa)
In another embodiment, the compound is:
Figure imgf000019_0003
acceptable salts Compound (V)
Also provided are compounds of structure (V):
O II
R41' P^R40
(V)
wherein, R40 and R41 are, independently, H, Ci to Cio alkyl optionally substituted by one or more R42, aryl optionally substituted by one or more R42, or benzyl optionally substituted by one or more R42; R42 is H, Ci to Cio alkyl, C(0)OH, NH2, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), Ci to Cio aminoalkyl, halogen, aryl, or heteroaryl or a pharmaceutically acceptable salt or prodrug thereof.
Figure imgf000020_0001
/ or pharmaceutically acceptable salts or prodrugs thereof.
Compound (VI)
The compound may alternatively have structure (VI):
OR51
0=P— OR50
I
R52
(VI)
wherein, R is i aarryyll,, hheetteerrooaarryyll,, oorr hheetteerrcocycle, each optionally substituted by one or more R53; R51 is H, Ci to Cio alkyl, Ci to Cio aminoalkyl, each optionally substituted by halogen or C(0)OH; R52 is absent, Ci to Cio alkyl optionally substituted by P(0)2, or aryl optionally substituted by 0(Ci to Cio alkyl)-phenyl-C(0)OH; R53 is Ci to Cio alkyl, halogen, or C(0)OH; or a pharmaceutically acceptable salt or prodrug thereof. In one embodiment, R50 is heteroaryl or heterocycle.
In one embodiment the aryl of R50 contains 5 to 8 carbon atoms, each carbon atom of R50 contains one R51 substituent, at least one carbon atom contains a
C(=0)OH substituent, and R51 is H or Ci to C6 alkyl.
In a further embodiment, R50 is a benzene ring.
ompound is selected from among:
Figure imgf000021_0001
or pharmaceutically acceptable salts or prodrugs thereof.
Compound (VII)
The compound may further have structure (VII):
P— (CR65R66)tR60R61
HO
(VII)
wherein, t is 1 to 10, i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; R60 is aryl, heteroaryl, or heterocycle; R61 is Ci to C10 aminoalkyl, Ci to C10 alkyl, C2 to C10 alkenyl, C2 to C10 alkynyl, C3 to C10 cycloalkyl, aryl, heteroaryl, or heterocycle; R65 and R66 are, independently, H, OH, Ci to C10 alkyl, C3 to C10 cycloalkyl, C2 to C10 alkenyl, C2 to Cio alkynyl benzyl, aryl, heteroaryl, or heterocycle; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, R61 is optionally substituted at one or more carbon atom by 0-2 R63, R63 is Ci to C10 alkyl or phenyl substituted by 0-5 R64, and R64 is Ci to C10 alkyl.
In a further embodiment, R is phenyl.
In another embodiment, the phenyl of R is substituted by 0-5 R and R 1 to Cio alkyl, C3 to C10 cycloalkyl, Ci to C10 alkoxy, or Ci to C10 thioalkyl.
In still a further embodiment, R63 is phenyl. Compound (VIII)
contemplated as useful antagonists are compounds of structure (VIII):
Figure imgf000022_0001
(VIII)
wherein, R , R , R , and R are, independently, H, Ci to Cio alkyl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, C(0)(Ci to Cio aminoalkyl), or heteroaryl, each optionally substituted by one or more R78;R72 is H, Ci to Cio alkyl, 0(Ci to Cio alkyl), 0(Ci to Cio alkoxy), 0(Ci to Cio aminoalkyl), Ci to Cio aminoalkyl, or heteroaryl, each optionally substituted by one or more R76; R73 is Ci to Cio alkyl, Ci to Cio alkoxy, Ci to Cio aminoalkyl, each optionally substituted by one or more R76; R76 is C(0)OH, C(0)(Ci to Cio alkyl), NHC(0)(Ci to Cio alkyl), C(0)NH(phenyl optionally substituted by one or more R77), C(0)0(Ci to Cio alkyl), heteroaryl optionally substituted by one or more R77, heterocycle optionally substituted by one or more R77, or phenyl optionally substituted by one or more R77; R77 is halogen, Ci to Cio aminoalkyl, Ci to Cio alkoxy, Ci to Cio alkyl, or C2 to Cio alkenyl, each optionally substituted by one or more R78; R78 is CN, C(0)OH, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), C(S)S(Ci to Cio alkyl)C(0)OH, or phenyl optionally substituted by one or more R79; R79 is OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to C10 alkenyl, C2 to Cio alkynyl, halogen, or Ci to e alkoxy; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, R70 to R74 is halogen.
In another embodiment, the compound is selected from among:
Figure imgf000023_0001
Figure imgf000024_0001
, or pharmaceutically acceptable salts or prodrugs thereof.
Compound (IX)
The compounds may also have structure (IX):
Figure imgf000024_0002
(IX)
wherein, w is 1 to 10, i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
w' is 1 to 10, i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; R80 is H or Ci to C10 alkyl; R81 to R85 are, independently, H, OH, Ci to Cio alkyl, halogen, or Ci to Cio alkoxy; and R86 to R89 are, independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl; or a
pharmaceutically acceptable salt or prodrug thereof. In one embodiment, R80 is H or C2 to Cio alkyl.
In one embodiment, the compound has structure (IXa) wherein R80 to R81 are defined above:
Figure imgf000024_0003
(IXa)
In a further embodiment, R81 to R85 in either compound (IX) or (IXa) is Ci to Ce alkoxy.
In another embodiment, two of R81 to R85 in either compound (IX) or (IXa) is
Ci to Ce alkoxy. In a further embodiment, Ci to Ce alkoxy in R to R is OCH3.
In yet another embodiment, the compound has structure (IXb), wherein R80 is defined above:
Figure imgf000025_0001
(IXb)
Compound (X)
The antagonists useful herein may further be a compound of structure (X)
Figure imgf000025_0002
(X)
wherein, x is 1 to 10, i.e. , 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or a pharmaceutically acceptable salt or prodrug thereof. In one embodiment, x is 2 to 10.
In one embodiment, the compound is 2-amino-3-sulfino-propanoic acid:
Figure imgf000025_0003
In one embodiment, the compound is (S)-2-amino-3-sulfino-propanoic acid, or a pharmaceutically acceptable salt or prodrug thereof:
O
HO s
= II
NH2 O
Compound (XI)
The compound may fu
Figure imgf000025_0004
(XI)
wherein, y is 1 to 10, i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; R90 to R94 are, independently, H, halogen, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, OH, or Ci to Cio aminoalkyl; and R95 is heterocycle, heteroaryl, aryl, or Ci to Cio aminoalkyl; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, R95 is a five or six membered heterocycle containing one O, S, or NR92 heteroatom and the carbon atoms of said heteroaryl are substituted by H or Ci to Ce alkyl.
In another embodiment, the compound has structure (XIa), wherein R90, R95, and y are defined above:
Figure imgf000026_0001
(XIa)
In a further embodiment, y in either structure (XI) or (XIa) is 2.
In still another embodiment, R95 in either structure (XI or (XIa) is pyrrolidine.
Compound (XII)
The compound may further have structure (XII):
Figure imgf000026_0002
(XII)
wherein, R105 is aryl, heteroaryl, or heterocycle; z is 1 to 10, i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; R106 to R110 are, independently, H, Ci to Cio alkyl, C3 to Cio cycloalkyl, OH, C2 to Cio alkenyl, or C2 to Cio alkynyl; or a pharmaceutically acceptable salt or prodrug thereof. In one embodiment, z is 2 to 10.
In a further embodiment, the compound has structure (Xlla):
Figure imgf000027_0001
(Xlla)
wherein, R105 is aryl, heteroaryl, or heterocycle; z is 1 to 10, i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; R106 to R110 are, independently, H, Ci to C10 alkyl, C3 to C10 cycloalkyl, OH, C2 to Cio alkenyl, or C2 to C10 alkynyl; or a pharmaceutically acceptable salt or prodrug thereof.
In yet another embodiment, the compound has structure (Xllb):
Figure imgf000027_0002
wherein, R100 to R104 are, independently, H, Ci to Ce alkyl, halogen, or Ci to alkoxy and R105, R106, and z are defined above.
In still a further embodiment, the compound has structure (XIIc):
Figure imgf000027_0003
(Xllb)
wherein, R100 to R104 are, independently, H, Ci to Ce alkyl, halogen, or Ci to Ce alkoxy and R105, R106, and z are defined above. In a further embodiment, R in either structure (XII) or (Xlla) is H.
In another embodiment, z is 1 in either structure (XII) or (Xlla).
In still a further embodiment, at least one of R100 to R104 in either structure (XII) or (Xlla) is Ci to C6 alkyl.
In yet another embodiment, R100 or R104 in either structure (XII) or (Xlla) is
CH3.
In a further embodiment, the compound is the following, or a
pharmaceutically acceptable salt or prodrug thereof such as a propionate salt:
Figure imgf000028_0001
In still another embodiment, the compound is the following, or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000028_0002
Compound (XIII)
Also provided are compounds of structure (XIII):
Figure imgf000028_0003
(XIII)
wherein, a is 0 or 1; R120 is absent, aryl, C3 to C10 cycloalkyl, heteroaryl, or heterocycle, each optionally substituted by one or more R125; R121 is aryl containing one or more R , Ci to C10 aminoalkyl optionally containing one or two aryl groups optionally substituted by one or more R124, heterocycle optionally substituted by one or more R125; R123 is H, halogen, Ci to C10 alkyl, or Ci to C10 alkoxy; R124 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, or Ci to C10 aminoalkyl; R125 is H, halogen, Ci to Cio alkyl, Ci to C10 alkoxy, (=0), or C(0)OH; or R120 and R121 join to form C5 to Cio cycloalkyl optionally substituted by one or more R126; and R126 is H, halo gen, Ci to Cio alkyl, Ci to Cio alkoxy, or Ci to Cio aminoalkyl; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, the compounds have structure (Xllla), wherein R is a heterocycle having one SO2 in the heterocycle backbone:
O
0 S-0R"1
(Xllla)
In another embodiment, the compound is selected from
Figure imgf000029_0001
or pharmaceutically acceptable salts or prodrugs thereof.
Compound (XIV)
Also provided as useful antagonistic compounds are those of structure (XIV):
Figure imgf000030_0001
(XIV)
wherein, R201 is Ci to C10 alkyl, Ci to C10 aminoalkyl, C2 to C10 alkenyl, or C2 to C10
202 202
alkynyl, each optionally substituted by one or more R^; R^ is H, CI to C10 alkyl, halogen, CN, or N02; R200 is Ci to C10 alkyl, Ci to C10 alkoxy, aryl, C2 to C10 alkenyl,
203 203
Ci to Cio aminoalkyl, each optionally substituted by one or more R ; R is halogen, CN, NO2, Ci to Cio alkoxy, or aryl optionally substituted by one or more R204; R204 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, NO2, or aryl; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, R201 is Ci to Cio aminoalkyl.
In another embodiment, R201 is CH2CH2NH2.
In a further embodiment, R200 is Ci to Cio alkyl optionally substituted by one
203 203
or more R and R is Ci to Cio alkyl, Ci to Cio alkoxy, or halogen.
In still another embodiment, R203 is 1, 2, or 3-tolyl, 1, 2, or 3-methoxyphenyl, or 1, 2, or 3-halogenated phenyl.
In a further embodiment, R200 is C2 to Cio alkenyl optionally substituted by
203 203
one or more R and R is Ci to Cio alkyl, Ci to Cio alkoxy, or halogen.
In still another embodiment, the compound is selected from among:
Figure imgf000030_0002
Figure imgf000031_0001
; or pharmaceutically acceptable salts or prodrugs thereof.
Compound (XV)
The compounds having structure (XV) are also useful as described herein:
Figure imgf000031_0002
(XV)
wherein, R is Ci to C10 alkyl, Ci to C10 aminoalkyl, C2 to C10 alkenyl, or C2 to C10
302 302
alkynyl, each optionally substituted by one or more RJUZ; RJuz is H, CI to C10 alkyl, halogen, CN, or N02; R300 is Ci to C10 alkyl, Ci to C10 alkoxy, aryl, C2 to C10 alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R ; R is halogen,
R304. R304 ig C i tQ C io dkylj CN, NO2, or aryl optionally substituted by one or more
Ci to Cio alkoxy, halogen, CN, NO2, or aryl; or a pharmaceutically acceptable salt or prodrug thereof.
In a further embodiment, R301 is Ci to Cio aminoalkyl.
In another embodiment, R is CH2CH2NH2.
In a further embodiment, R300 is phenyl.
In still another embodiment, R303 is Ci to Cio alkyl or halogen.
In yet a further embodiment, the compound is selected from among:
Figure imgf000031_0003
or pharmaceutically acceptable salts or prodrugs thereof Compound (XVI)
Further provided are the compounds having structure (XVI) for use as described herein:
Figure imgf000032_0001
(XVI)
wherein, R400 is OH or O; R401, R402, and R403, are independently, OR405 or
N(R406)(R407);
R404 is H, Ci to Cio alkyl, halogen, (=0), or Ci to Cio alkoxy; R405 is absent, Ci to Cio alkyl, heterocycle, aryl, or heteroaryl, optionally substituted by one or more R406; R406 is H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, C(0)(Ci to Cio alkyl), aryl, heteroaryl, C(0)(aryl optionally substituted by one or more R404; R407 is absent, H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, aryl, or heteroaryl; with the proviso that when R400 is O and R405 or R407 is absent, then R400 and R402 join to form a 5 to 8-membered ring, wherein the 5 to 8-membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404; and with the proviso that when R401 is OR405 and R403 is
N(R406)(R407) and R405 and R407 are absent, R401 and R402, and/or R401 and R403, and/or R402 and R403 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8- membered ring is optionally substituted by one or more R404; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, the compound is selected from among:
Figure imgf000032_0002
NH
Figure imgf000033_0001
or pharmaceutically acceptable salts or prodrugs thereof.
Compounds (XVII)
Also provided for use as described herein are compounds having structure (XVII):
Figure imgf000033_0002
(XVII)
wherein, R is Ci to C10 aminoalkyl, R is benzyl optionally substituted by one or more R503; R501 is H, Ci to C10 alkyl, C3 to C10 cycloalkyl, Ci to C10 alkoxy, or aryl; f is 0 to 4, i.e., 0, 1, 2, 3, or 4; and R503 is H, Ci to C10 alkyl, Ci to C10 alkoxy, halogen, or aryl; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, the compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000033_0003
Compounds (XVIII)
Also provided as antagonistic compounds are those having structure (XVIII):
Figure imgf000033_0004
(XVIII) wherein, R is H, OH, Ci to C10 alkyl, Ci to C10 alkoxy, or C3 to C10 cycloalkyl; R to R510 are, independently, H, Ci to C10 alkyl, Ci to C10 alkoxy, C3 to C10 cycloalkyl, or halogen; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, the compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000034_0001
Compounds (XIX)
Additional compounds useful as antagonistic compounds include those of structure (XIX):
Figure imgf000034_0002
(XIX)
wherein, RbUU is Ci to C10 alkyl, NH2 or Ci to C10 aminoalkyl; RbVZ to R are, independently, Ci to C10 alkyl, C3 to C10 cycloalkyl, Ci to C10 alkoxy, or halogen; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, the compound is the following or a pharmaceutically acceptable salt or prodrug the
Figure imgf000034_0003
Compounds (XX)
Further provided are compounds of structure (XX):
Figure imgf000035_0001
(XX)
wherein, R606 is H or Ci to C10 alkyl; R607 to R610 are, independently, H, Ci to C10 alkyl, Ci to C10 alkoxy, or halogen; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, the compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000035_0002
Compounds (XXI)
Additional compounds useful as antagonistic compounds include those of structure (XXI):
Figure imgf000035_0003
(XXI)
wherein, R uu to R are, independently, H, Ci to C10 alkyl, C3 to CIO cycloalkyl, C3 to Cio spirocycloalkyl, Ci to C10 alkoxy, halogen, Ci to C10 aminoalkyl, or CN and R705 is H or Ci to C10 alkyl; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, the compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000035_0004
Compounds (XXII)
Also provided are comp II):
Figure imgf000036_0001
(XXII)
wherein, R800 to R804 and R806 and R809 are, independently, H, Ci to C10 alkyl, halogen, NH2, or Ci to C10 alkoxy and R805 is H or Ci to C10 alkyl; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, the compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000036_0002
Compounds (XXIII)
Further provided are compounds of structure (XVI) for use as described herein:
Figure imgf000036_0003
(XXIII) wherein, R UU is H or Ci to C10 alkyl and R U1 to R is H, Ci to C10 alkyl, C2 to C10 alkenyl, C2 to C10 alkynyl, halogen, Ci to C10 alkoxy, Ci to C10 aminoalkyl, or NH2; or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, the compound is the following or a pharmaceutically acceptable salt or prodrug the
Figure imgf000037_0001
Compounds XXIV
Also provided are compounds of structure (XXIV):
Figure imgf000037_0002
(XXIV)
wherein, R906 and R907 are, independently, Ci to C10 alkyl or Ci to C10 alkoxy; e is 0 to 5, i.e., 0, 1, 2, 3, 4, or 5; and R908 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, OH, CN, or NH2; R909 is H or Ci to C10 alkyl; or a pharmaceutically acceptable salt or prodrug thereof. In one embodiment, R907 is C2 to C10 alkyl. In another embodiment, R909 is H. In a further embodiment, R909 is Ci to C10 alkoxy. In yet another embodiment, R909 is ethoxy.
In one embodiment, the compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000037_0003
Compendium of Compounds
The following compounds may be utilized as PF4 antagonistic compounds 2-acetyl-3 -hydroxy-5-nitro-inden- 1 -one;
N-(hydroxy-dimethoxy-phosphoranyl)pyrrolidine- 1 -carboxamide;
N-(hydroxy-diisobutoxy-phosphoranyl)-5-methyl-pyridin-2-amine;
3 -(hydroxyphosphonoylmethyl)butanedioic acid;
[4-(hydroxyphosphonoyloxymethyl)cyclohexyl]methoxyphosphinic acid; 2-(2-hydroxyphosphonoylethyl-phenyl-phosphanyl)ethylphosphinic acid;
1- chloro-2-(2-chloroethyoxyphosphonoyloxy)ethane;
4-(4-dimethylaminophenyl)phosphonoyl-N,N-dimethyl-aniline;
2,3 -bis(methoxycarbonyl)propylphosphinic acid;
(2-hydroxyphosphonoyl-2-phenyl-ethyl)phosphinic acid;
1 -ethoxyphosphonoylbutane;
2- diethoxyphosphorylethylphosphinic acid;
2- hydroxyphosphonoylindane- 1 -carboxylic acid;
(l S,2S)-2-hydroxyphosphonoylindane-l-carboxylic acid;
(lR,2R)-2-hydroxyphosphonoylindane-l -carboxylic acid;
(l S,2R)-2-hydroxyphosphonoylindane-l-carboxylic acid;
(lR,2S)-2-hydroxyphosphonoylindane-l-carboxylic acid;
p-tolylphosphinic acid;
3- phenyl-4-sulfinooxy-thiolane- 1 , 1 -dioxide;
(3 S,4R)-3 -phenyl-4-sulfinooxy-thiolane- 1 , 1 -dioxide;
(3 R,4S)-3 -phenyl-4-sulfinooxy-thiolane- 1 , 1 -dioxide;
(3 S,4S)-3 -phenyl-4-sulfinooxy-thiolane- 1 , 1 -dioxide;
(3 S,4R)-3 -phenyl-4-sulfinooxy-thiolane- 1 , 1 -dioxide;
[hydroxy-(3-pyridyl)methyl]phosphinic acid;
styrylphosphinic acid;
2-(2-furyl)ethylphosphinic acid;
l-benzylphosphonoyl-4-phenyl-benzene;
l-hydroxy-3-phenyl-prop-2-enyl]phosphinic acid;
[(E, lR)-l-hydroxy-3-phenyl-prop-2-enyl]phosphinic acid;
[(Z, 1 R)- 1 -hydroxy-3 -phenyl-prop-2-enyl]phosphinic acid; [(E, 1S)-1 -hydroxy-3 -phenyl-prop-2-enyl]phosphinic acid;
[(Z, 1 S)- 1 -hydroxy-3 -phenyl-prop-2-enyl]phosphinic acid
benzylphosphonic acid;
2-chloroethylphosphonoylbenzene;
(2,4-dimethoxyphenyl)phosphinic acid;
2-chloroethoxyphosphonoyloxybenzene;
2-butoxyphosphonoyl-2-methyl-propane;
1 -(2-phenylacetyl)aminoethylphosphinic acid;
a compound having the following structure:
Figure imgf000039_0001
1 -butylphosphonoylbutane;
( 1 -benzhydrylamino-3 -methyl-butyl)phosphinic
phenoxyphosphinic acid;
1 ,4-dichloro-2-sulfino-benzene;
styrylphosphinic acid;
diethoxy-thioxo-phosphorane;
(3 -chloro-4-methyl-phenyl)phosphinic acid;
2-carbazol-9-ylethylphosphinic acid;
a compound having the following structure:
Figure imgf000039_0002
[4-[2-(2-chlorophenoxy)ethoxy]-3-methoxy-phenyl]methanamine; 2-[4-(2-diethylaminoethoxy)phenyl]thiazole-4-carboxylic acid;
N-(bis(dimethylamino)-hydroxy-phosphoranyl)-4-chloro-benzamide; 1 -ethoxyphosphonoylbutane;
2-cyano-3-[4-(2-diethylaminoethoxy)-3-methoxy-phenyl]prop-2-enoic (E)-2-cyano-3-[4-(2-diethylaminoethoxy)-3-methoxy-phenyl]prop-2-enoic acid;
(Z)-2-cyano-3-[4-(2-diethylaminoethoxy)-3-methoxy-phenyl]prop-2-enoic acid;
2- [2-(3-methoxyphenyl)thiazolo[2,3-e][l,2,4]triazol-6-yl]ethanamine;
4-(ethoxycarbonylmethylsulfanyl)naphthalene-l,8-dicarboxylic acid;
3- [2-[2-[(2-methoxy-5-methyl-phenyl)amino]-2-oxo-ethoxylphenyl]propanoic acid;
2-[2-[2-(4-fluorophenyl)ethyl]thiazolo[2,3-e][l,2,4]triazol-6-yl]ethanamine; 3 - [2- [2 - [(2-methoxy-5 -methyl-phenyl)amino] -2-oxo-ethoxy]phenyl]propionic acid;
2- [2-[2-(4-fluorophenyl)ethyl]thiazolo[2,3-e][l,2,4]triazol-6-yl]ethanamine;
3- [3-(2-diethylaminoethoxy)-4-methoxy-phenyl]propanoic acid;
2-[2-[2-(o-tolyl)vinyl]thiazolo[2,3-e][l,2,4]triazol-6-yl]ethanamine;
2-[2-[(E}-2-(o-tolyl)vinyl]thiazolo[2,3-e][l,2,4]triazol-6-yl]ethanamine; 2-[2-[(Z}-2-(o-tolyl)vinyl]thiazolo[2,3-e][l,2,4]triazol-6-yl]ethanamine;
4- (aminomethyl)-8-(3-fluorophenyl)-7-thia-l,5,9-triazabicyclo[4.3.0]nona- 3,5,8-trien-2-one;
4-(aminomethyl)-8-(4-chlorophenyl)-7-thia-l,5,9-triazabicyclo[4.3.0]nona- 3,5,8-trien-2-one;
[5-(4-benzyl-l-piperidyl)-l,3,4-oxadiazol-2-yl]methanamine;
2-cyano-N-[2-[4-(2-diethylaminoethoxy)-3-ethoxy-phenyl]ethyl]acetamide;
2-(4-ethylpiperazin- 1 -yl)- 1 -( 1 H-indol-3 -yl)-2-phenyl-ethanone;
2-( 1 -cyclopropylethylcarbamoylmethyl-methyl-amino)-N-(2,4,6- trimethylphenyl)-acetamide;
2-cyano-3-[3-(2-diethylaminoethoxy)-4-methoxy-phenyl]prop-2-enoic acid;
(E)-2-cyano-3-[3-(2-diethylaminoethoxy)-4-methoxy-phenyl]prop-2-enoic acid;
(Z)-2-cyano-3-[3-(2-diethylaminoethoxy)-4-methoxy-phenyl]prop-2-enoic acid;
2- chloroethylphosphonoylbenzene;
3- indolin-7-yl-2-(phosphonomethyl)prop-2-enoic acid; (Z)-3-indolin-7-yl-2-(phosphonomethyl)prop-2-enoic acid;
(E)-3-indolin-7-yl-2-(phosphonomethyl)prop-2-enoic acid;
methyl 2-[4-(aminomethyl)phenyl]-4-methyl-thiazole-5-carboxylate;
2-cyano-3-[4-(3-dimethylaminopropoxy)-3-ethoxy-phenyl]prop-2-enoic acid;
(Z)-2-cyano-3-[4-(3-dimethylaminopropoxy)-3-ethoxy-phenyl]prop-2-enoic acid;
(E)-2-cyano-3-[4-(3-dimethylaminopropoxy)-3-ethoxy-phenyl]prop-2-enoic acid;
5-[l-aminoethyl]-N-[l, l-dioxothiolan-3-yl]-N-methyl-furan-2-carboxamide; 5-[(lR)-l-aminoethyl]-N-[(3R)-l, l-dioxothiolan-3-yl]-N-methyl-furan-2- carboxamide;
5-[(l S)- l-aminoethyl]-N-[(3R)- 1 , 1 -dioxothiolan-3-yl]-N-methyl-furan-2- carboxamide;
5-[(lR)-l-aminoethyl]-N-[(3S)-l, l-dioxothiolan-3-yl]-N-methyl-furan-2- carboxamide;
5- [( 1 S)- 1 -aminoethyl]-N- [(3 S)- 1 , 1 -dioxothiolan-3 -yl] -N-methyl-furan-2- carboxamide;
2-[2-[2-(p-tolyl)vinyl]thiazolo[2,3-e][l,2,4]triazol-6-yl]ethanamine;
2-[2-[(E)-2-(p-tolyl)vinyl]thiazolo[2,3-e][l,2,4]triazol-6-yl]ethanamine; 2-[2-[(Z)-2-(p-tolyl)vinyl]thiazolo[2,3-e][l,2,4]triazol-6-yl]ethanamine; 4-(aminomethyl)-8-(3-methoxyphenyl)-7-thia-l,5,9-triazabicyclo[4.3.0]nona- 3,5,8-trien-2-one;
4-[2-[2-(3-dimethylaminopropoxy)phenyl]ethylamino]-4-oxo-butanoic acid;
2-[2-[2-(m-tolyl)ethyl]thiazolo[2,3-e][l,2,4]triazol-6-yl]ethanamine;
2- [ [3 -methyl-2-[(2-methylbenzoyl)amino]butanoyl] amino] -3 -sulfidopropionic acid;
(2S)-2-[[(2S)-3-methyl-2-[(2-methylbenzoyl)amino]butanoyl]amino]-3- sulfidopropionic acid;
(2R)-2-[[(2R)-3-methyl-2-[(2-methylbenzoyl)amino]butanoyl]amino]-3- sulfidopropionic acid;
(2R)-2-[[(2S)-3-methyl-2-[(2-methylbenzoyl)amino]butanoyl]amino]-3- sulfidopropionic acid; (2S)-2-[[(2R)-3-methyl-2-[(2-methylbenzoyl)amino]butanoyl]amino]-3- sulfidopropionic acid;
2- cyano-3-[3-[2-(2-methoxyphenoxy)ethoxy]phenyl]prop-2-enoic acid; (Z)-2-cyano-3-[3-[2-(2-methoxyphenoxy)ethoxy]phenyl]prop-2-enoic acid; (E)-2-cyano-3-[3-[2-(2-methoxyphenoxy)ethoxy]phenyl]prop-2-enoic acid;
[4-[(2-methoxyphenoxy)methyl]phenyl]methanamine;
4-[(2-phosphonophenoxy}methyl]benzoic acid;
[5-(aminomethyl)-2-furyl]-[2-ethyl-l-piperidyl]methanone;
[5-(aminomethyl)-2-furyl]-[(2R)-2-ethyl-l-piperidyl]methanone;
[5-(aminomethyl)-2-furyl]-[(2S)-2-ethyl-l-piperidyl]methanone;
[4-[2-(3,5-dimethylphenoxy)ethoxy]-3-methoxy-phenyl]methanamine;
1 - [2-(4-tert-butylphenoxy)ethy 1 ]piperazine;
[3-methoxy-4-(o-tolylmethoxy)phenyl]methanamine;
4-[[4-(dimethylaminomethyl)phenyl]methylsulfamoyl]benzoic acid; and 3-[(3,5-dimethoxybenzoyl)amino]-3-(p-tolyl)propanoic acid;
(3R)-3-[(3,5-dimethoxybenzoyl)amino]-3-(p-tolyl)propanoic acid;
(3S)-3-[(3,5-dimethoxybenzoyl)amino]-3-(p-tolyl)propanoic acid; and
3- [l-(ethoxycarbonylmethyl}-5-(4-chlorophenyl)-pyrrol-2-yl]propanoic acid. The term "alkyl" is used herein to refer to both straight- and branched-chain saturated aliphatic hydrocarbon groups. In one embodiment, an alkyl group has 1 to about 10 carbon atoms (i.e., Ci, C2, C3, C4, C5 Ce, C7, C8, C9, or C10). In another embodiment, an alkyl group has 1 to about 6 carbon atoms (i.e., Ci, C2, C3, C4, C5 or Ce). In a further embodiment, an alkyl group has 1 to about 4 carbon atoms (i.e., Ci, C2, C3, or C4).
The term "alkenyl" is used herein to refer to both straight- and branched-chain alkyl groups having one or more carbon-carbon double bonds. In one embodiment, an alkenyl group contains 2 to about 10 carbon atoms (i.e., C2, C3, C4, C5, Ce, C7, C8, C9, or Cio). In another embodiment, an alkenyl group has 1 or 2 carbon-carbon double bonds and 2 to about 6 carbon atoms (i.e., C2, C3, C4, C5 or Ce).
The term "alkynyl" is used herein to refer to both straight- and branched-chain alkyl groups having one or more carbon-carbon triple bonds. In one embodiment, an alkynyl group has 2 to about 10 carbon atoms (i.e., C2, C3, C4, C5, Ce, C7, C8, C9, or Cio). In another embodiment, an alkynyl group contains 1 or 2 carbon-carbon triple bonds and 2 to about 6 carbon atoms (i.e., C2, C3, C4, C5, or Ce).
The term "cycloalkyl" is used herein to refer to cyclic, saturated aliphatic hydrocarbon groups. The term cycloalkyl may include a single ring or two or more rings fused together to form a multicyclic ring structure. A cycloalkyl group may thereby include a ring system having 1 to about 5 rings. In one embodiment, a cycloalkyl group has 3 to about 14 carbon atoms (i.e., C3, C4, C5, Ce, C7, C8, C9, C10, C11, C12, Ci3, or C14). In another embodiment, a cycloalkyl group has 3 to about 10 carbon atoms (i.e., C3, C4, C5 or Ce). In a further embodiment, a cycloalkyl group has 3 to about 6 carbon atoms (i.e., C3, C4, C5 or Ce).
The term "alkoxy" as used herein refers to the O-(alkyl) group, where the point of attachment is through the oxygen-atom and the alkyl group is defined above.
The term "thioalkyl" as used herein refers to the S-(alkyl) group, where the point of attachment is through the sulfur-atom and the alkyl group is defined above.
The term "aminoalkyl" as used herein refers to both secondary and tertiary amines where the point of attachment is through the nitrogen-atom and the alkyl groups are defined above. The alkyl groups can be the same or different.
The term "halogen" as used herein refers to CI, Br, F, or I groups.
The term "aryl" as used herein refers to an aromatic, carbocyclic system, e.g., of about 5 to 20 carbon atoms, which can include a single ring or multiple unsaturated rings fused or linked together where at least one part of the fused or linked rings forms the conjugated aromatic system. An aryl group may thereby include a ring system having 1 to about 5 rings. The aryl groups include, but are not limited to, phenyl, naphthyl, biphenyl, anthryl, tetrahydronaphthyl, phenanthryl, indene, benzonaphthyl, and fluorenyl.
The term "benzyl" as used herein refers to a ~CH2-phenyl group.
The term "heteroaryl" as used herein refers to a stable, aromatic 5- to 20- membered monocyclic or multicyclic heteroatom-containing ring. The heteroaryl ring has in its backbone carbon atoms and one or more heteroatoms including nitrogen, oxygen, and sulfur atoms. In one embodiment, the heteroaryl ring contains 1 to about 4 heteroatoms in the backbone of the ring. When the heteroaryl ring contains nitrogen or sulfur atoms in the backbone of the ring, the nitrogen or sulfur atoms can be oxidized. Further, when the heteroaryl ring contains nitrogen atoms, the nitrogen atoms may optionally substituted by H, Ci to C10 alkoxy, C(0)(Ci to Ce alkyl), or C(0)0(Ci to Ce alkyl). The heteroaryl ring can be attached through a heteroatom or carbon atom provided the resultant heterocyclic ring structure is chemically stable. When the heteroaryl ring is a multicyclic heteroatom-containing ring, it may contain 2, 3, 4, or 5 rings.
A variety of heteroaryl groups are known in the art and include, without limitation, oxygen-containing rings, nitrogen-containing rings, sulfur-containing rings, mixed heteroatom-containing rings, fused heteroatom containing rings, and combinations thereof. Examples of heteroaryl groups include, without limitation, furyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, azepinyl, thienyl, dithiolyl, oxathiolyl, oxazolyl, thiazolyl, oxadiazolyl, oxatriazolyl, oxepinyl, thiepinyl, diazepinyl, benzofuranyl, thionapthene, indolyl, benzazolyl, purindinyl, pyranopyrrolyl, isoindazolyl, indoxazinyl, benzoxazolyl, quinolinyl, isoquinolinyl, benzodiazonyl, napthylridinyl, benzothienyl, pyridopyridinyl, acridinyl, carbazolyl, and purinyl rings.
The term "heterocycle" or "heterocyclic" as used herein can be used interchangeably to refer to a stable, saturated or partially unsaturated 3- to 20- membered monocyclic or multicyclic heterocyclic ring. The heterocyclic ring has carbon atoms and one or more heteroatoms including nitrogen, oxygen, and sulfur atoms in its backbone. In one embodiment, the heterocyclic ring has 1 to about 4 heteroatoms in the backbone of the ring. When the heterocyclic ring contains nitrogen or sulfur atoms in the backbone of the ring, the nitrogen or sulfur atoms can be oxidized. Further, when the heterocyclic ring contains nitrogen atoms, the nitrogen atoms may optionally be substituted with H, Ci to Ce alkyl, substituted Ci to Ce alkyl, C02 (Ci to C6 alkyl), S02(Ci to C6 alkyl), S02(substituted Ci to C6 alkyl), S02aryl, S02substituted aryl, CO(Ci to C6 alkyl), CO(substituted Ci to C6 alkyl), COaryl or COsubstituted aryl. The heterocyclic ring can be attached through a heteroatom or carbon atom provided the resultant heterocyclic ring structure is chemically stable. When the heterocyclic ring is a multicyclic ring, it may contain 2, 3, 4, or 5 rings.
A variety of heterocyclic groups are known in the art and include, without limitation, oxygen-containing rings, nitrogen-containing rings, sulfur-containing rings, mixed heteroatom-containing rings, fused heteroatom containing rings, and combinations thereof. Examples of heterocyclic groups include, without limitation, tetrahydrofuranyl, piperidinyl, 2-oxopiperidinyl, pyrrolidinyl, morpholinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, pyranyl, pyronyl, dioxinyl, piperazinyl, dithiolyl, oxathiolyl, dioxazolyl, oxathiazolyl, oxazinyl, oxathiazinyl, benzopyranyl, benzoxazinyl and xanthenyl.
The term "substituted" as used herein refers to the substituted of an atom or moiety for another atom of moiety, provided that the substitution results in stable chemical compound. The substitution may occur at a carbon atom and/or heteroatom. One of skill in the art would readily be able to determine if a chemical compound is "stable".
II. Methods of Production
The compounds described above may be prepared by known chemical synthesis techniques. These compounds may also be purchased from commercial vendors, e.g., the Sigma-Aldrich Co. Among such preferred techniques known to one of skill in the art are included the synthetic methods described in conventional textbooks relating to the construction of synthetic compounds. III. Compositions Containing the Antagonistic Compounds
Pharmaceutical compositions useful herein, in one embodiment, contain a compound discussed above in a pharmaceutically acceptable carrier with other optional suitable pharmaceutically inert or inactive ingredients. In another embodiment, a compound described above is present in a single composition. In a further embodiment, a compound described above is combined with one or more excipients and/or other therapeutic agents as described below.
1. Salts
The antagonistic compounds discussed above may encompass tautomeric forms of the structures provided herein characterized by the bioactivity of the drawn structures. Further, the compounds may also be used in the form of salts derived from pharmaceutically or physiologically acceptable acids, bases, alkali metals and alkaline earth metals. In one embodiment, pharmaceutically acceptable salts can be formed from organic and inorganic acids including, e.g., acetic, propionic, lactic, citric, tartaric, succinic, fumaric, maleic, malonic, mandelic, malic, phthalic, hydrochloric, hydrobromic, phosphoric, nitric, sulfuric, methanesulfonic, napthalenesulfonic, benzenesulfonic, toluenesulfonic, camphorsulfonic, and similarly known acceptable acids.
In another embodiment, pharmaceutically acceptable salts may also be formed from inorganic bases, desirably alkali metal salts including, e.g., sodium, lithium, or potassium, such as alkali metal hydroxides. Examples of inorganic bases include, without limitation, sodium hydroxide, potassium hydroxide, calcium hydroxide, and magnesium hydroxide. Pharmaceutically acceptable salts may also be formed from organic bases, such as ammonium salts, mono-, di-, and trimethylammonium, mono-, di- and triethylammonium, mono-, di- and tripropylammonium (iso and normal), ethyldimethylammonium, benzyldimethylammonium, cyclohexylammonium, benzyl- ammonium, dibenzylammonium, piperidinium, morpholinium, pyrrolidinium, piperazinium, 1 -methylpiperidinium, 4-ethylmorpholinium, 1 -isopropylpyrrolidinium, 1,4-dimethylpiperazinium, 1 -n-butyl piperidinium, 2-methylpiperidinium, l-ethyl-2- methylpiperidinium, mono-, di- and triethanolammonium, ethyl diethanolammonium, n-butylmonoethanolammonium, tris(hydroxymethyl)methylammonium, phenylmono- ethanolammonium, diethanolamine, ethylenediamine, and the like. In one example, the base is selected from among sodium hydroxide, lithium hydroxide, potassium hydroxide, and mixtures thereof.
These salts, as well as other compounds, can be in the form of esters, carbamates and other conventional "pro-drug" forms, which, when administered in such form, convert to the active moiety in vivo. In one embodiment, the prodrugs are esters. In another embodiment, the prodrugs are carbamates. See, e.g., B. Testa and J. Caldwell, "Prodrugs Revisited: The "Ad Hoc" Approach as a Complement to Ligand Design", Medicinal Research Reviews, 16(3):233-241, ed., John Wiley & Sons (1996), which is incorporated by reference.
The compounds discussed herein also encompass "metabolites" which are unique products formed by processing the compounds in vivo, i.e., by the cell or subject. Desirably, metabolites are formed in vivo. As described herein, a therapeutically or prophylactically effective amount of an antagonistic compound is that amount of a compound which prevents PF4 tetramer formation or disrupts PF4 tetramers, optionally present in an ULC. The effective amount of a compound may vary depending upon the formulation and route of delivery. In one embodiment, effective amount (i.e., per unit) of a compound is less than about 10 g/kg. In another embodiment, the dosage is less than about 5 g/kg, 500 mg/kg, 400 mg/kg, 300 mg/kg, 200 mg/kg, 100 mg/kg, 50 mg/kg, 25 mg/kg, 10 mg/kg, 1 mg/kg, 0.5 mg/kg, 0.25 mg/kg, 0.1 mg/kg, 100 μg/kg, 75 μg/kg, 50 μg/kg, 25 μg/kg, 10 μg/kg, or 1 μg/kg. However, the effective amount to be used is subjectively determined by the attending physician. The variables involved include the specific cognitive deficit and the size, age and response pattern of the patient. These effective amounts may be provided on regular schedule, i.e., daily, weekly, monthly, or yearly basis or on an irregular schedule with varying administration days, weeks, months, etc. Alternatively, the effective amount to be administered may vary. In one embodiment, the effective amount for the first dose is higher than the effective amount for one or more of the subsequent doses. In another embodiment, the effective amount for the first dose is lower than the effective amount for one or more of the subsequent doses.
2. Carriers
The pharmaceutical compositions of this invention include compounds described herein formulated neat or with one or more pharmaceutical carriers for administration, the proportion of which is determined by the solubility and chemical nature of the compound, chosen route of administration and standard pharmacological practice. The pharmaceutical carrier(s) may be solid or liquid. Formulations may incorporate both solid and liquid carriers.
The compounds described herein may be administered to a subject by any desirable route, taking into consideration the specific condition for which it has been selected. The compounds may, therefore, be delivered orally, by injection, inhalation (including orally, intranasally and intratracheally), ocularly, trans dermally, intravenously, subcutaneous ly, intramuscularly, sublingually, intracranially, epiduraly, rectally, and vaginally, among others. In one embodiment, the compound is delivered orally, intravenously, subcutaneously, or intramuscularly. Although the compound may be administered alone, it may also be administered in the presence of one or more pharmaceutical carriers that are physiologically compatible. The carriers may be in dry or liquid form and must be pharmaceutically acceptable. Liquid pharmaceutical compositions are typically sterile solutions or suspensions.
When liquid carriers are utilized, they are desirably sterile liquids. Liquid carriers are typically utilized in preparing solutions, suspensions, emulsions, syrups and elixirs. In one embodiment, the antagonistic compound is dissolved a liquid carrier. In another embodiment, the antagonistic compound is suspended in a liquid carrier. One of skill in the art of formulations would be able to select a suitable liquid carrier, depending on the route of administration. In one embodiment, the liquid carrier includes, without limitation, water, organic solvents, oils, fats, or mixtures thereof. In another embodiment, the liquid carrier is water containing cellulose derivatives such as sodium carboxymethyl cellulose. In a further embodiment, the liquid carrier is water and dimethylsulfoxide (DMSO). In yet another embodiment, the liquid carrier contains water and up to 5% of DMSO. Examples of organic solvents include, without limitation, alcohols such as monohydric alcohols and polyhydric alcohols, e.g., glycols and their derivatives, among others. Examples of oils include, without limitation, fractionated coconut oil, arachis oil, corn oil, peanut oil, and sesame oil and oily esters such as ethyl oleate and isopropyl myristate. The liquid carrier can contain other suitable excipients, including those described below.
Alternatively, the antagonistic compounds may be formulated in a solid carrier. In one embodiment, the composition may be compacted into a unit dose form, i.e., tablet or caplet. In another embodiment, the composition may be added to unit dose form, i.e., a capsule. In a further embodiment, the composition may be formulated for administration as a powder. The solid carrier may perform a variety of functions, i.e., may perform the functions of two or more of the excipients described below. For example, solid carrier may also act as a flavoring agent, lubricant, solubilizer, suspending agent, filler, glidant, compression aid, binder, disintegrant, or encapsulating material. Suitable solid carriers include, without limitation, calcium phosphate, dicalcium phosphate, magnesium stearate, talc, starch, sugars (including, e.g., lactose and sucrose), cellulose (including, e.g., microcrystalline cellulose, methyl cellulose, sodium carboxymethyl cellulose), polyvinylpyrrolidine, low melting waxes, ion exchange resins, and kaolin. The solid carrier can contain other suitable excipients, including those described below.
The composition may also be sub-divided to contain appropriate quantities of the antagonistic compound. For example, the unit dosage can be packaged compositions, e.g., packeted powders, vials, ampoules, prefilled syringes or sachets containing liquids.
Examples of excipients which may be combined with one or more of the antagonistic compounds include, without limitation, adjuvants, antioxidants, binders, buffers, coatings, coloring agents, compression aids, diluents, disintegrants, emulsifiers, emollients, encapsulating materials, fillers, flavoring agents, glidants, granulating agents, lubricants, metal chelators, osmo-regulators, pH adjustors, preservatives, solubilizers, sorbents, stabilizers, sweeteners, surfactants, suspending agents, syrups, thickening agents, or viscosity regulators. See, the excipients described in the "Handbook of Pharmaceutical Excipients", 5th Edition, Eds.: Rowe,
Sheskey, and Owen, APhA Publications (Washington, DC), December 14, 2005, which is incorporated herein by reference.
In one embodiment, the compositions may be utilized as inhalants. For this route of administration, compositions may be prepared as fluid unit doses using an antagonistic compound and a vehicle for delivery by an atomizing spray pump or by dry powder for insufflation.
In another embodiment, the compositions may be utilized as aerosols, i.e., oral or intranasal. For this route of administration, the compositions are formulated for use in a pressurized aerosol container together with a gaseous or liquefied propellant, e.g., dichlorodifluoromethane, carbon dioxide, nitrogen, propane, and the like. Also provided is the delivery of a metered dose in one or more actuations.
In another embodiment, the compositions may be administered by a sustained delivery device. "Sustained delivery" as used herein refers to delivery of an antagonistic compound which is delayed. Those of skill in the art know suitable sustained delivery devices. For use in such sustained delivery devices, the compounds are formulated as described herein. 3. Pharmaceutical Compositions Containing Multiple Compounds
In addition to the components described above for use in the composition and the antagonistic compound, the compositions may contain one or more medication or therapeutic agent.
In one aspect, the additional medication causes as an undesired side effect of the formation of PF4 tetramers or ULCs. In one embodiment, the medication modulates clotting, i.e., is an anti-coagulant or induces hemostasis. In one example, the clotting modulator is a blood thinner such as heparin. In a further example, the clotting modulator is aspirin or a non-steroidal anti-inflammatory drug. In another example, the clotting modulator is dipyridamole. In yet a further example, the clotting modulator is ticlopidine or clopidogrel. In still another example, the clotting modulator is a GPIIB/IIIA inhibitor. In still another embodiment, the medication causes hemostasis and includes, without limitation, a vasoconstrictor or an agent which induces platelet aggregation. In another embodiment, the medication is a chemotherapeutic. In a further embodiment, the medication is a cardiotonic. In another embodiment, the medication is a blood pressure lowering agent. In yet a further embodiment, the medication is a cholesterol lowering agent. Examples of chemotherapeutics, cardiotonics, blood pressure lowering agents, and cholesterol lowering agents include those recited in the "Physician's Desk Reference", 64th Edition, Thomson Reuters, 2010, which is hereby incorporated by reference.
In another aspect, the medication has the desirable effect of disrupting PF4 tetramers and/or ULCs. In one embodiment, the additional medication works synergistically with the PF4 antagonists discussed herein. Such medications which disrupt PF4 tetramers may be selected by one of skill in the art and may include, without limitation, cyclic peptides which inhibit the interaction of PF4 with CCL5 (CKEY2), carbohydrates such as desulfated heparin (ODSH), or a combination thereof. The PF4 tetramer disruption agents may be combined with the PF4 antagonist compounds discussed herein either in the compositions set forth above and/or kits and methods for using the same and discussed below.
In one embodiment, the compounds described herein and/or other therapeutic agents may be administered in a single composition. However, the present invention is not so limited. In other embodiments, the compounds described herein invention may be administered in one or more separate formulations from other compounds described herein, chemotherapeutic agents, or other agents as is desired.
IV. Kits Containing the Antagonistic Compound
Also provided herein are kits or packages of pharmaceutical formulations containing the compounds or compositions described herein. The kits may be organized to indicate a single formulation or combination of formulations to be taken at each desired time.
Suitably, the kit contains packaging or a container with the compound formulated for the desired delivery route. Suitably, the kit contains instructions on dosing and an insert regarding the active agent. Optionally, the kit may further contain instructions for monitoring circulating levels of product and materials for performing such assays including, e.g., reagents, well plates, containers, markers or labels, and the like. Such kits are readily packaged in a manner suitable for treatment of a desired indication. For example, the kit may also contain instructions for use of the spray pump or other delivery device. Other suitable components to include in such kits will be readily apparent to one of skill in the art, taking into consideration the desired indication and the delivery route. The doses are repeated daily, weekly, or monthly, for a predetermined length of time or as prescribed.
The compounds or compositions described herein can be a single dose or for continuous or periodic discontinuous administration. For continuous administration, a package or kit can include the antagonistic compound in each dosage unit (e.g., solution, lotion, tablet, pill, or other unit described above or utilized in drug delivery). When the compound is to be delivered with periodic discontinuation, a package or kit can include placebos during periods when the compound is not delivered. When varying concentrations of a composition, of the components of the composition, or of relative ratios of the antagonist compounds or agents within a composition over time is desired, a package or kit may contain a sequence of dosage units, so varying.
A number of packages or kits are known in the art for the use in dispensing pharmaceutical agents for oral use. In one embodiment, the package has indicators for each period. In another embodiment, the package is a labeled blister package, dial dispenser package, or bottle. The packaging means of a kit may itself be geared for administration, such as an inhalant, syringe, pipette, eye dropper, or other such like apparatus, from which the formulation may be applied to an infected area of the body, such as the lungs, injected into a subject, or even applied to and mixed with the other components of the kit.
The compositions of these kits also may be provided in dried or lyophilized forms. When reagents or components are provided as a dried form, reconstitution generally is by the addition of a suitable solvent. It is envisioned that the solvent also may be provided in another packaging means.
The kits of the present invention also will typically include a means for containing the vials in close confinement for commercial sale such as, e.g., injection or blow-molded plastic containers into which the desired vials are retained.
Irrespective of the number or type of packages, the kits of the invention also may include, or be packaged with a separate instrument for assisting with the
injection/administration or placement of the ultimate complex composition within the body of an animal. Such an instrument may be an inhalant, syringe, pipette, forceps, measuring spoon, eye dropper or any such medically approved delivery means. Other instrumentation includes devices that permit the reading or monitoring of reactions in vitro.
In one embodiment, a pharmaceutical kit is provided and contains a medication which causes the formation of PF4 tetramers, medication which disrupts PF4 tetramers, or a combination thereof and one or more of an antagonistic compound selected from those described above. The antagonistic compound may be in the presence or absence of one or more of the carriers or excipients described above. The kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer.
In another embodiment, a pharmaceutical kit is provided and contains a medication which causes the formation of PF4 tetramers in a first dosage unit and one or more of an antagonistic compound selected from those described above in a second dosage unit. The antagonistic compound may be in the presence or absence of one or more of the carriers or excipients described above. The kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer. In a further embodiment, a pharmaceutical kit is provided and contains a medication which disrupts PF4 tetramers in a first dosage unit and one or more of an antagonistic compound selected from those described above in a second dosage unit. The antagonistic compound may be in the presence or absence of one or more of the carriers or excipients described above. The kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer.
In still a further embodiment, a pharmaceutical kit is provided and contains a medication which causes the formation of PF4 tetramers in a first dosage unit, a medication which disrupts PF4 tetramers in a second dosage unit, and one or more of an antagonistic compound selected from those described above in a third dosage unit. The antagonistic compound may be in the presence or absence of one or more of the carriers or excipients described above. The kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer.
In a further embodiment, a pharmaceutical kit is provided and contains a medication which causes the formation of PF4 tetramers in a first dosage unit, one or more of an antagonistic compound selected from those described above in a second dosage unit, and one or more of the carriers or excipients described above in a third dosage unit. The kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer.
In yet another embodiment, a pharmaceutical kit is provided and contains a medication which disrupts PF4 tetramers in a first dosage unit, one or more of an antagonistic compound selected from those described above in a second dosage unit, and one or more of the carriers or excipients described above in a third dosage unit. The kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer.
In yet a further embodiment, a pharmaceutical kit is provided and contains a medication which causes the formation of PF4 tetramers in a first dosage unit, a medication which disrupts PF4 tetramers in a second dosage unit, and one or more of an antagonistic compound selected from those described above in a third dosage unit, and one or more of the carriers or excipients described above in a third dosage unit. The kit may optionally contain instructions for administering the medication(s) and the compound to a subject having cancer. V. Methods of Treatment Using the Antagonistic Compounds
As noted above, the formation of PF4 tetramers is linked to a subject acquiring specific diseases or conditions. The terms "patient" or "subject" as used herein refer to a mammalian animal. In one embodiment, the patient or subject is a human. In another embodiment, the patient or subject is a veterinary or farm animal, a domestic animal or pet, or animal normally used for clinical research. In a further embodiment, the subject or patient has elevated levels of PF4 tetramer. In yet a further
embodiment, the subject or patient has elevated levels of PF4 tetramenGAG ULCs. In still another embodiment, the subject or patient has elevated levels of PF4 tetramer :heparin ULCs. In a further embodiment, the subject or patient is asymptomatic for medical conditions related to elevated levels of PF4 tetramer, PF4 tetramenGAG ULCs, or PF4 tetramenheparin ULCs. In another embodiment, the subject shows clinical signs of medical conditions related to elevated levels of PF4 tetramer, PF4 tetramenGAG ULCs, or PF4 tetramenheparin ULCs. In another embodiment, the subject shows clinical signs of developing or has HIT or HITT. In still a further embodiment, the subject or patient has cancer. The subject or patient has either been recognized as having or at risk of having one or more of the diseases or conditions discussed above, i.e., a primary disease or medical condition.
Alternatively, the one or more diseases or medical conditions in a subject may be the result of another disease or medical condition, i.e., a secondary disease or condition.
The term "disease" or "condition" as used herein refers to any disease or medical condition related directly or indirectly to the formation of PF4 tetramers. The disease or medical condition may also be caused by the formation of PF4 tetramers. The methods described herein thereby are useful in treating or preventing these diseases or conditions.
In one embodiment, the disease or medical condition is heparin-induced thrombocytopenia (HIT). As known in the art, HIT results from the development of thrombocytopenia, i.e., low platelet counts, due to the administration of one of the forms of the anticoagulant. In another embodiment, the disease or medical condition is heparin-induced thrombocytopenia and/with thrombosis (HITT). As known in the art, HITT results when HIT precedes thrombosis, i.e., abnormal blood clots form inside a blood vessel and abnormal antibodies form.
Although there are treatment options for atherosclerotic vascular disease and thrombocytopenia and increasing HDL, none of the therapies are directed toward PF4 tetramer formation/interactions. Specifically, treatment of atherosclerotic vascular disease involves anti-platelet therapy, i.e., aspirin, the Plavix® drug, among others, and is not tolerated in all patients. For the treatment of thrombocytopenia, TPO analogs and mimetics (MPL agonists which activate MPL-the TPO receptor) may be used, but these drugs do not work in all patients, and have significant side effects/compliance issues. Finally, some lipid lowering therapies are able to increase HDL, but the ability of available drugs to do so is limited.
In a further embodiment, the disease or medical condition is atherosclerosis. Typically, atherosclerosis resulting from the formation of a PF4 tetramer may be treated according to the invention described herein. In still another embodiment, the disease or medical condition is a platelet imbalance. The treatment method thereby includes correcting this platelet imbalance or preventing a platelet imbalance. In one example, platelet levels are increased by stimulating platelet production. In another example, a decrease in platelet production is prevented. In a further example, the platelet imbalance, i.e., low levels of platelets, results from the formation of PF4 tetramer. In yet another example, the platelet imbalance, i.e., low levels of platelets, results from heparin administration to a subject.
The compounds discussed herein may also be an alternative therapy utilized to treat diseases related to thrombopoietin (TPO).
The compounds discussed herein may further be useful in inhibiting the activation of nuclear factor κ-light-chain-enhancer of activated B ( F-κΒ) cells and thereby treating conditions related to an imbalance NF-κΒ. Such conditions may include, without limitation, cancer, inflammatory disease, autoimmune diseases, septic shock, viral infection, among others.
The compounds discussed herein may also be contemplated for use in preventing or treating inflammation which results from the formation of PF4 tetramers. The inflammation may be the caused by any number of factors. In one embodiment, the inflammation is acute or chronic and localized or systemic. The inflammation may be the result of a variety of factors and/or conditions. In another embodiment, the inflammation is caused by NF-κΒ activation.
Given that Lys50Glu, or non-tetrameric PF4 variant, does not activate NFKB, the inventors hypothesized that compounds which inhibit tetramer formation will inhibit the activation of NFKB. TO test this hypothesis, mutation of Lys50 to Glu disrupted these interactions and created a mutant PF4 that readily forms dimers but not tetramers. Further, formation of ULC requires tetrameric PF4, as Lys50Glu does not form ULC with heparin. The importance of PF4 tetramers in complex formation is supported by subsequent biophysical studies as reported in Rauova.
The compounds described herein may also be useful in therapies for subjects having atherosclerotic vascular disease in which the patient is intolerant to the conventional treatments, i.e., statins.
The compounds discussed herein may further be useful for modulating clotting or hemostasis. In one embodiment, protein C is upregulated. In another embodiment, protein C is downregulated. The term "upregulated" as used herein refers to an increase of protein or nucleic acid in level or function. Similarly, the term
"downregulated" as used herein refers to a decrease of protein or nucleic acid in level or function. The term "level" references the amount of protein or nucleic acid present and the term "function" references the action of the protein or nucleic acid.
The compounds discussed above may be useful in patients that are intolerant to these conventional therapies. Alternatively, the compounds discussed above may be synergistic with the conventional therapies.
As used herein, "treatment" encompasses treatment of a subject clinically diagnosed as having a disease or medical condition. In one embodiment, the subject is treated and the disease or medical condition is eradicated, i.e., the subject is cured.
In another embodiment, the subject is treated for chronic diseases and the symptoms resulting from the disease or medical conditions are relieved.
As used herein, "prevention" encompasses prevention of symptoms in a subject who has been identified as at risk for the condition, but has not yet been diagnosed with the same and/or who has not yet presented any symptoms thereof.
Not only can the antagonists described herein be used to prevent or treat diseases and medical conditions, but the same are also useful in increasing high density lipoproteins (HDL) in a subject. Alternatively, the antagonists are useful in preventing a decrease of HDLs.
As known to those skilled in the art, GAGs are long unbranched
polysaccharides having a repeating disaccharide unit (a hexose (six-carbon sugar) or a hexuronic acid, linked to a hexosamine (six-carbon sugar containing nitrogen)). One of skill in the art would readily be able to determine the GAGs which are capable of binding to PF4 tetramers. In one embodiment, the GAG is selected from among wild- type GAGs or synthetically produced GAGs. In one embodiment, the GAG is heparin, hyaluronan, hyaluronic acid, dermatan sulfates, keratan sulfates, and chondroitins, among others or derivatives, salts, or prodrugs thereof. In another embodiment, the GAG is heparin. In a further embodiment, the GAG is heparan sulfate. GAGs as described herein may also be variations of the those described above, i.e., they may vary in the type of hexosamine, hexose or hexuronic acid unit they contain (e.g., glucuronic acid, iduronic acid, galactose, galactos amine, glucosamine).
The methods described above, either for treating or preventing the stated medical conditions or disease, may be performed in vivo or ex vivo as determined by the attending physician or clinician.
The methods described herein may be performed by administering one or more of the compounds described above via a combination therapy in prior to, concurrently with, or subsequent to another medication. Such combination treatment may occur by administering compositions containing multiple active ingredients, as described above. However, this invention also encompasses a method of
administration of anti-cancer agents or therapies in conjunction with a composition containing an antagonistic compound. In one embodiment of such a method, the antagonistic compound and additional medications are administered to the patient by one or more selected routes of administration sequentially. In one embodiment, a chemotherapeutic agent is administered before treatment with a composition of the invention. In another embodiment, a chemotherapeutic agent is administered after treatment with a composition of the invention. In still another embodiment, a chemotherapeutic agent is administered during treatment with a composition of the invention. In one embodiment, a chemotherapeutic agent is administered before treatment with a composition of the invention. In another embodiment, a
chemotherapeutic agent is administered after treatment with a composition of the invention. In still another embodiment, a chemotherapeutic agent is administered before, during or after treatment with a composition of the invention. In a further embodiment, the composition of the invention is administered prior to another medication which disrupts PF4 tetramers. In another embodiment, the composition of the invention is administered concurrently with another medication which disrupts PF4 tetramers. In yet further embodiment, the composition of the invention is administered subsequent to another medication which disrupts PF4 tetramers.
In one embodiment, methods for disrupting PF4 tetramers are provided and include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject and administering a first effective amount of the compound required to decrease the first PF4 tetramer level.
In another embodiment, methods for disrupting PF4 tetramers are provided and include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject, administering a first effective amount of the compound discussed herein required to decrease the first PF4 tetramer level, and optionally administering a medication which disrupts PF4 tetramers. The medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
As used herein, the term "biological sample" refers to a body fluid or tissue. The body fluid can include, without limitation, whole blood, serum, plasma, peripheral blood, synovial fluid, cerebrospinal fluid, saliva, urine, semen, or other fluid secretion. The term "tissue" can include, without limitation, bone marrow and lymph node, as well as samples of other tissues.
In another embodiment, methods for disrupting PF4 tetramers are provided and include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject, administering a first effective amount of the compound required to decrease the first PF4 tetramer level, measuring a second level of PF4 tetramer in a second biological sample obtained from the subject, and administering a second effective amount of the compound required to decrease the second PF4 tetramer level. Multiple samples can be obtained from the subject at any interval required to prevent or treat the medical condition.
In yet another embodiment, methods for disrupting PF4 tetramers are provided and include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject, administering a first effective amount of the compound required to decrease the first PF4 tetramer level, optionally administering a first effective amount of a medication which disrupts PF4 tetramers, measuring a second level of PF4 tetramer in a second biological sample obtained from the subject, administering a second effective amount of the compound required to decrease the second PF4 tetramer level, and optionally administering a second effective amount of a medication which disrupts PF4 tetramers. Multiple samples can be obtained from the subject at any interval required to prevent or treat the medical condition. The medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
In a further embodiment, methods for disrupting ULCs containing PF4 tetramers and heparin are provided. These methods include measuring a first level of ULCs in a first biological sample obtained from a subject and administering a first effective amount of the compound required to decrease the first ULC tetramer level.
In still a further embodiment, methods for disrupting ULCs containing PF4 tetramers and heparin are provided. These methods include measuring a first level of ULCs in a first biological sample obtained from a subject, administering a first effective amount of the compound required to decrease the first ULC tetramer level, and administering a first effective amount of a medication which disrupts PF4 tetramers. The medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
In yet another embodiment, methods for disrupting ULCs containing PF4 tetramers and heparin are provided. These methods include measuring a first level of ULCs in a first biological sample obtained from a subject, administering a first effective amount of the compound required to decrease the first ULC level, measuring a second level of ULCs in a second biological sample obtained from the subject, administering a second effective amount of the compound required to decrease the second ULC level. In a further embodiment, methods for disrupting ULCs containing PF4 tetramers and heparin are provided. These methods include measuring a first level of ULCs in a first biological sample obtained from a subject, administering a first effective amount of the compound required to decrease the first ULC level, optionally administering a first effective amount of a medication which disrupts PF4 tetramers, measuring a second level of ULCs in a second biological sample obtained from the subject, administering a second effective amount of the compound required to decrease the second ULC level, and optionally administering a second effective amount of a medication which disrupts PF4 tetramers. The medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
In still a further embodiment, methods for preventing the formation of PF4 tetramers are provided and include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject and administering a first effective amount of the compound required to prevent formation of the PF4 tetramer.
In another embodiment, methods for preventing the formation of PF4 tetramers are provided and include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject, administering a first effective amount of the compound required to prevent formation of the PF4 tetramer, and administering a first effective amount of a medication which disrupts PF4 tetramers. The medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
In still a further embodiment, methods for preventing the formation of PF4 tetramers are provided. These methods include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject, administering a first effective amount of the compound required to prevent formation of the PF4 tetramer, measuring a second level of PF4 tetramer in a second biological sample obtained from the subject, and administering a second effective amount of the compound required to prevent formation of PF4 tetramer.
In yet a further embodiment, methods for preventing the formation of PF4 tetramers are provided. These methods include measuring a first level of PF4 tetramer in a first biological sample obtained from a subject, administering a first effective amount of the compound required to prevent formation of the PF4 tetramer, optionally administering a first effective amount of a medication which disrupts PF4 tetramers, measuring a second level of PF4 tetramer in a second biological sample obtained from the subject, administering a second effective amount of the compound required to prevent formation of PF4 tetramer, and optionally administering a second effective amount of a medication which disrupts PF4 tetramers. The medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
In a further embodiment, methods for preventing the formation of ULCs containing PF4 tetramers and heparin are provided. These methods include measuring a first level of ULCs in a first biological sample obtained from a subject and administering a first effective amount of the compound required to prevent formation of the ULCs.
In another embodiment, methods for preventing the formation of ULCs containing PF4 tetramers and heparin are provided. These methods include measuring a first level of ULCs in a first biological sample obtained from a subject, administering a first effective amount of the compound required to prevent formation of the ULCs, and administering a first effective amount of a medication which disrupts PF4 tetramers. The medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
In yet another embodiment, methods for preventing the formation of ULCs containing PF4 tetramers and heparin are provided. These methods include measuring a first level of ULCs in a first biological sample obtained from a subject and administering a first effective amount of the compound required to prevent formation of the ULCs, measuring a second level of ULCs in a second biological sample obtained from the subject, and administering a second effective amount of the compound required to prevent formation of ULCs.
In yet a further embodiment, methods for preventing the formation of ULCs containing PF4 tetramers and heparin are provided. These methods include measuring a first level of ULCs in a first biological sample obtained from a subject and administering a first effective amount of the compound required to prevent formation of the ULCs, optionally administering a first effective amount of a medication which disrupts PF4 tetramers, measuring a second level of ULCs in a second biological sample obtained from the subject, administering a second effective amount of the compound required to prevent formation of ULCs and/or administering a second effective amount of a medication which disrupts PF4 tetramers. The medication which disrupts PF4 tetramers may be administered prior to, concurrent with, or subsequent to the PF4 antagonists discussed herein.
VI. Screening Methods Using the Antagonistic Compounds
Also provided are methods for using the antagonistic compounds described herein for determining a subject's sensitivity to side effects or secondary medical conditions related to heparin administration. Further provided are methods for determining the likelihood of a subject to acquire a medical condition related to the formation of PF4 tetramers or ULCs containing PF4 tetramers and heparin. In one embodiment, these screening methods are useful in monitoring cancer patients. In another embodiment, these screening methods are useful in determining the likelihood of cancer patients being administered heparin in developing HIT or HITT.
According to this method, biological samples are obtained from subjects and the level of PF4 tetramer and/or PF4 tetramer:GAG (heparin) ULC measured. The screening may be conducted using techniques commonly known and used in the art. Comparison of the levels of PF4 tetramer and/or PF4 tetramenheparin ULC to a control level and/or negative control provides evidence that the patient may be treated using one or more of the antagonistic compounds described herein. In one embodiment, if the subject's PF4 tetramer and/or PF4 tetramer :heparin ULC level is higher than the PF4 tetramer and/or PF4 tetramer :heparin ULC level of a healthy subject, then antagonistic compound administration may be contemplated.
The following examples are illustrative of methods using the antagonistic compounds described herein. It will be readily understood by one of skill in the art that the specific conditions described herein for producing these compounds can be varied without departing from the scope of the present invention. It will be further understood that other compounds encompassed by the structures described herein, as well as other salts, hydrates, and/or prodrugs thereof, are within the scope of the invention.
EXAMPLES
Unfractionated heparin was an injectable sodium salt from porcine intestinal mucosa (Sagent Pharmaceuticals, Schaumberg, IL). HiTrap® heparin affinity columns for protein purification were purchased from GE Healthcare (Uppsala, Sweden). Immunochemicals included horseradish peroxidase-conjugated sheep polyclonal anti-human PF4 from Enzyme Research Laboratory (South Bend, ΓΝ), mouse immunoglobulin G 2bK (IgG2bK; MOPC 141) was purchased from Sigma (St. Louis, MO), and sheep anti-mouse IgG and alkaline phosphatase-conjugated goat anti-mouse IgG were from Jackson ImmunoResearch Labs (West Grove, PA). Murine monoclonal antibodies KKO (anti-human PF4-heparin complex), RTO (anti-human PF4), and IV.3 (FcyRIIA-blocking antibody) may be prepared according to Arepally, "Characterization of a murine monoclonal antibody that mimics heparin-induced thrombocytopenia antibodies", Blood, 95(5): 1533-1540 (2000), which is hereby incorporated by reference. Human plasma samples were obtained from patients with a high clinical suspicion for HIT (see, Cuker, "The HIT expert probability (HEP) score: a novel pre-test probability model for heparin-induced thrombocytopenia based on broad expert opinion", J. Thromb. Haemost, 8(12):2642-2650 (2010), which is hereby incorporated by reference) and who had positive heparin/PF4 ELISA (GTI X- HAT45; Waukesha, WI) and serotonin release assay results. The Bicinchoninic acid (BCA) Protein Assay Reagent Kit and bis(sulfosuccinimidyl)suberate (BS3) crosslinker were obtained from Pierce (Rockford, IL). Immulon® 4 HBX microtiter plates for ELISA were purchased from Fisher Scientific (Pittsburgh, PA). Bovine serum albumin (BSA), ionomycin, phorbol myristate acetate (PMA) and p- nitrophenyl phosphate (pNPP) tablets were obtained from Sigma and the 3,3,5,5- tetramethylbenzidine (TMB) liquid substrate system for ELISA was purchased from KPL (Gaithersburg, MD). Dose response data were analyzed using the indirect Hill relationship using GraphPad® Prism® software (version 5.03, sigmoidal dose- response, variable slope model). Example 1: Identification of ULC antagonists
To identify potential ULC antagonists, the Dock® program, as described in Moustakas et al, "Development and validation of a modular extensible docking program: DOCK 5", J. Comput. Aided Mol. Des., 20:601-619 (2006), which is hereby incorporated by reference, was utilized to screen a library of about 1.1 million leadlike compounds from the Zinc® database. See, Irwin and Shoichet, "ZINC— a free data base of commercially available compounds for virtual screening", J. Chem. Inf. Model, 45: 177-182 (2005), which is hereby incorporated by reference. Specifically, these compounds were screened in an effort to identify those having the potential to bind in the cavity containing the salt bridge described above, thereby impeding the formation of PF4 tetramers.
A set of overlapping spheres which fill the binding site were generated, known as sphere centers, using the Sphgen™ program. See, Kuntz, "A geometric approach of macromolecule-ligand interactions", J. Mol. Biol, 161(2):269-288 (1982), which is incorporated by reference. The sphere centers were permitted to touch the surface of the molecule, were not permitted to intersect the surface, and were used to represent the negative image of the binding site on PF4. The coordinates of the sphere centers were used to orient ligands within the target site. Sphere-sphere distances were then compared to ligand atom-atom distances and the compatible orientations of the ligand were found. A scoring grid to evaluate ligand orientations was then produced using the program GRID as described in Shoichet, "Molecular docking using shaper descriptions", J. Comp. Chem., 13(3):380-397 (1992) which is hereby incorporated by reference.
The docking calculations were run in parallel, using 56 simultaneous processes, on a 14-node compute cluster as described in Papadopoulos, "NPACI Rocks: tools and techniques of easily deploying manageable Linux clusters", Concurrency Computat: Pract. Exper., 15:707-725 (2003), which is hereby incorporated by reference. Rigid docking with automated matching, a maximum of 1000 orientations per ligand, grid scoring and ligand conformation optimization required about 10 days (at about 1 compound/sec) to perform. Dock scores were empirical with lower values representing higher affinity. The docking scores approximated a Gaussian distribution (Figure 2) with a mean value of -26.6 and a standard deviation of 3.3. Of the over 1.1 million compounds modeled, the 101 compounds of Table 1 bound PF4 near Lys50. These 100 compounds scored greater than 5 standard deviations below the mean (-73.8 or less).
Table 1
Figure imgf000065_0001
Name # Name
4-(aminomethyl)-8-(3-fluorophenyl)-7-
[hydro xy-(3 -pyridyl)methyl]phosphinic acid 71 thia-l,5,9-triazabicyclo[4.3.0]nona-3,5,8- trien-2-one
4-(aminomethyl)-8-(4-chlorophenyl)-7-
72 thia-l,5,9-triazabicyclo[4.3.0]nona-3,5,8- trien-2-one
[5-(4-benzyl- 1 -piperidyl)- 1 ,3 ,4-oxadiazol- styrylphosphinic acid 73
2-yl]methanamine
2-cyano-N-[2-[4-(2-diethylaminoethoxy)-
(acetyl-allyl-amino)methylphosphinic acid 74
3-ethoxy-phenyl]ethyl]acetamide
2-(4-ethylpiperazin- 1 -yl)- 1 -( 1 H-indol-3 -
2-(2-furyl)ethylphosphinic acid 75
yl)-2-phenyl-ethanone
2-( 1 -cyclopropylethylcarbamoylmethyl- l-benzylphosphonoyl-4-phenyl-benzene 76 methyl-amino)-N-(2,4,6- trimethylphenyl)-acetamide
(E)-2-cyano-3-[3-(2-diethylaminoethoxy)-
77
4 -methoxy-phenyl]prop -2 -enoic acid
[(E, 1 R)- 1 -hydro xy-3 -phenyl-prop-2-
78 2-chloroethylphosphonoylbenzene enyllphosphinic acid
(Z)-3-indolin-7-yl-2-
79
(pho sphonomethyl)prop -2 -enoic acid
3 -[ 1 -(carboxymethyl} -5-(4-
80
chlorophenyl)-pyrrol-2-yl]propanoic acid methyl 2 - [4-(aminomethyl)pheny 1] -4 - benzylphosphonic acid 81
methyl -thiazole-5 -carboxylate
(Z)-2-cyano-3-[4-(3-
(2-hydroxyphosphonoyl-2-phenyl-
82 dimethylaminopropoxy)-3-ethoxy- ethyl)phosphinic acid
phenyl]prop-2-enoic acid
5 - [( 1 R)- 1 -aminoethyl] -N-[(3 R)- 1 , 1 -
2-chloroethylphosphonoylbenzene 83 dioxothiolan-3 -yl] -N-methyl-furan-2- carboxamide
2-[2-[(E)-2-(p-tolyl)vinyl]thiazolo[2,3-
(5-oxotetrahydrofuran-3 -yl)phosphinic acid 84
e] [ 1 ,2,4]triazol-6-yl]ethanamine
N-methyl- 1 -[4-(2-pyrrolidin- 1 -
(2,4-dimethoxyphenyl)phosphinic acid 85
ylethoxy)phenyl]-propan-2 -amine
4-(aminomethyl)-8-(3-methoxyphenyl)-7-
86 thia-l,5,9-triazabicyclo[4.3.0]nona-3,5,8- trien-2-one
4-[2-[2-(3-
2-chloroethoxyphosphonoyloxybenzene 87 dimethylaminopropoxy)phenyl] ethylaminol-4-oxo-butanoic acid
2-[2-(3,4-dimethoxyphenyl)ethyl-methyl-
2 -butoxypho sphonoyl-2 -methyl-prop ane 88
thiocarbamoyl] sulfanylacetic acid
2-[2-[2-(m-tolyl)ethyl]thiazolo[2,3-
1 -(2-phenylacetyl)aminoethylphosphinic acid 89
e] [ 1 ,2,4]triazol-6-yl]ethanamine a compound having the following
(2S)-2-[[(2S)-3-methyl-2-[(2-
90 methylbenzoyl)amino]butanoyl] amino] -3 - sulfidopropionic acid structure 0— '
(Z)-2-cyano-3-[3-[2-(2-
1 -butylphosphonoylbutane 91 methoxyphenoxy)ethoxy]phenyl]prop-2- enoic acid
[4-[(2-methoxyphenoxy)methyl]phenyl]
92
methanamine # Name # Name
4 - [(2 -pho sphonophenoxy } methyljbenzoic
43 93
acid
( 1 -benzhydrylamino-3 -methyl - [5-(aminomethyl)-2-furyl]-[(2R)-2-ethyl-
44 94
butyl)phosphinic acid 1 -piperidyllmethanone
[4-[2-(3,5-dimethylphenoxy)ethoxy]-3-
45 phenoxyphosphinic acid 95
methoxy-phenyllmethanamine
(benzylamino-phenyl-methyl)phosphinic
46 96 2-phosphonooxybenzoic acid acid
l-[2-(4-tert-
47 97
butylphenoxy)ethy 1 Jpiperazine
[3-methoxy-4-(o-
48 98
tolylmethoxy)phenyl]methanamine
4 - [ [4 -(dimethyl aminome thy l)pheny 1]
49 2-amino-3-sulfino-propanoic acid 99
methylsulfamoyl]benzoic acid
(3R)-3-[(3,5-dimethoxybenzoyl)amino]-
50 100
3-(p-tolyl)propanoic acid
3-[5-(4-chlorophenyl)-l-(2-ethoxy-2-
101
oxyethyl)-lH-pyrrol-2-yl] propanoic acid
Figure 1 provides molecular models of a representative compound from these 101 compounds. As noted, the representative compound fits in the pocket formed by the PF4 tetramer and is adjacent to the salt bridge of the PF4 tetramer. Figure IB is 5 the same molecule model as Figure 1A, but rotated 90°.
The 15 compounds of Table 2 were selected from the compounds of Table 1 and were selected to determine their ability to disrupt of inhibit PF4 formation.
Table 2
Figure imgf000067_0001
Figure imgf000068_0001
Example 2: Prevention of PF4 tetramer Formation using Antagonistic Compounds
The 15 compounds from Example 1 in Table 2 were then tested using SDS 5 PAGE analysis of BS3 crosslinked PF4 for their ability to prevent PF4 tetramer
formation.
As is known in the art, DLS is useful in distinguishing PF4 ULCs from PF4 alone. See, Suvarna and Greinacher et al, "Heparin-induced thrombocytopenia: a stoichiometry-based model to explain the differing immunogenicities of
10 unfractionated heparin, low-molecular-weight heparin, and fondaparinux in difference clinical settings", Throm. Res, 122:21 1-220 (2008), which are hereby incorporated by reference, for a detailed description of this technique. cDNA encoding human PF4 was cloned into the plasmid pMT/BiPN5-His A (Invitrogen Corp., Carlsbad CA) for expression in the Drosophila Expression System (Invitrogen, Carlsbad, CA). Cloning was performed using Bgl II and Age I cloning sites. A hexanucleotide encoding Bgl II site was then eliminated by site-directed mutagenesis so that the expressed protein contained full-length wild type (wt) PF4 or PF4K50E with an identical sequence as their counterparts expressed in E. coli. See, Park described above. PF4 expression was induced by adding copper sulfate (0.5 mM). The induced S2 cells were incubated in serumfree Insect-Xpress™ media (Lonza, Walkersville, MD) for 3-5 days; supernatants were collected, sodium azide (0.02% final concentration) and EDTA (2.5 mM final concentration) were added, and the media were filtered through an Express® PLUS 0.22 μιη filter (Millipore Corp., Billerica, MA).
Wild-type (WT) human PF4 in the pT7-7 vector (Novagen (Madison, WI)) was expressed in the Escherichia coli strain BL21DE30 pLysS (Stratagene (La Jolla, CA)), and purified and characterized as described Rauova cited above and Park et al, "Biologic and biochemic properties of recombinant platelet factor 4 demonstrate identity with the native protein", Blood, 75: 1290-1295 (1990), which is hereby incorporated by reference. Briefly, recombinant protein was isolated from the supernatant of the bacterial lysate by affinity chromatography using a HiTrap high- performance (HP) affinity column.
WT PF4 was purified from the media on a heparin HiTrap® column on an ATKAPrime™ system (GE Healthcare) at 4°C in Tris (10 mM), EDTA (1 mM), and pH 8 buffer. Media was loaded in buffer containing NaCl (0.5 M) and PF4 was eluted at 1.8 M NaCl using a linear gradient. Fractions containing purified PF4 as detected by silver staining of 12% NuPAGE Bis-Tris gels (Invitrogen) were pooled, concentrated and buffer exchanged into 50 mM HEPES, 0.5 M NaCl, pH - 7.2 using an Amicon® Ultra centrifugal filter (3K NMWL, Millipore Corp). Protein was quantified using a BCA assay. A. Experiment 1
Compounds 34, 24, 1, 80, 88, and 96 were then tested for their ability to inhibit PF4 tetramer formation using a cross-linking assay as described in Rauova cited above. Specifically, hPF4 (10 μg/mL) in phosphate-buffered saline (PBS) was incubated at 37°C for 15 minutes in the absence of the antagonistic compounds (control sample) and presence of compounds 34, 24, 1, 80, 88 and 96. See, the following Table 3, which provides the reagents and amounts. Samples 1 and 18 were the molecular weight markers.
Table 3
Figure imgf000070_0001
Cross linking and SDS-PAGE analysis were then performed as described in "Analysis of PF4 multimerization" of Rauova. The relative amounts of PF4 in each of the monomer, dimer, trimer and tetramer form were calculated by analyzing the samples on a 12% SDS-polyacrylamide gel under reducing conditions. Bands were quantified using the Kodak ID Image Analysis system (Kodak, Rochester, NY). The SigmaMarker™ reagent served as the molecular weight standard. Data was compiled and bar graphed as % PF4 for the tetramer, trimer, dimer, and monomer as a function of concentration for the test and control samples. See, Figures 4A and 4B.
These data show that all of the compounds were effective in inhibiting PF4 tetramer formation at tested concentrations. However, compound 96 had the best ability to inhibit tetramer formation at 1 mM. B. Experiment 2
This experiment was performed in a manner similar to experiment 1, but PF4 (10 μg/mL) in PBS was independently incubated with compound 1 (250 μΜ), 34 (500 μΜ), 80 (250 μΜ), 101 (250 μΜ), and 96 (500 μΜ) for 60 minutes at room temperature, followed by the addition of cross-linking reagent
bis(sulfosuccinimidyl)suberate (0.2 mM) for 30 minutes at room temperature. The reaction was stopped by adding NuPAGE® lithium dodecyl sulfate (LDS) sample buffer, and denatured by heating to 70°C for 10 minutes according to the
manufacturer's instructions. The analysis and compilation of the data also was performed as described above in Experiment 1.
These data show that all of compounds 1, 80, 101, and 96 demonstrated a dose-dependent and ultimately complete inhibition of PF4 tetramer formation. See, Figure 7 and the following Table 4.
Table 4
Figure imgf000071_0001
Example 3: Disruption of ULC and/or PF4 tetramer Formation using
Antagonistic Compounds
Since ULCs are formed as part of a dynamic equilibrium between heparin and PF4, dynamic light scattering (DLS) was performed to measure the ability of compound 34, 1, 80, 96, and 100 to disrupt preformed ULCs.
In an effort to validate the following findings, samples were analyzed and included PF4 incubated in the absence or presence of unfractionated heparin (UFH) for 20 minutes at room temperature at PF4:heparin ratios (PHR) of 1 :0 (no heparin), 2: 1, and 0.64: 1. The samples were then analyzed using photon correlation spectroscopy on a DynaPro® DLS instrument and Dynamics® software (V6.7.6; Microsoft) to obtain correlation function. Figure 3 shows the results of the DLS analysis and is a bar graph showing the percentage of ULCs and small PF4 particles. The solid bar represent the data for the small particles (a population of particles with a mean hydrodynamic diameter of about 1 nm) and the striped bars represent the data for the large particles (a population of particles with a mean hydrodynamic diameter of about 300 nm). Data are expressed as the percent of total intensity from each measurement, the mean of 10 measurements, and representative of two such experiments. As noted from this figure, larger amounts of heparin result in the disruption of larger particles, i.e., ULCs, demonstrated by the presence of virtually all of the PF4 as small particles (Figure 3). A. Analysis of Compound 96
ULCs were formed as described in Rauova cited above. Specifically, hPF4 (10 μg/mL) in phosphate-buffered saline (PBS) was incubated at 37°C for 15 minutes in the presence of heparin (0.34 U/mL) - molar ratio of PF4:heparin is 2.5: 1). After removal of three aliquots of this PF4 and heparin solution containing ULCs, the aliquots were incubated with 0.1 μΜ, 1 μΜ, or 10 μΜ of compound 96 at 37°C. The total percentage of ULC (%) was measured and graphed. See, Figure 5 which is a bar graph of DLS data demonstrating ULC antagonistic activity for compound 96 as % inhibition vs. ULC diameter.
These data show that the sample lacking compound 96 results in exclusively ULCs. However, regardless of concentration, compound 96 is effective in disruption the preformed ULCs. Specifically, even at 0.1 μΜ, compound 96 reduces the amount of ULC and increases the formation of small particles. In fact, the use of 10 μΜ of compound 96 results in almost complete disruption of all ULCs. PF4 is present as small particles (similar to PF4 alone) when ULC are disrupted.
B. Analysis of Compounds 34, 1, 80, 96, and 101
This experiment was performed to evaluate the ability of compounds 1, 34, 80, 101, and 96 to facilitate the disruption of preformed ULC. ULC were formed as described above and then independently incubated with compounds 1 (125 μΜ and 63 μΜ), 34 (2 mM), 80 (250 μΜ), 101 (500 μΜ), and 96 (1 mM) or buffer for 24 hours at 37°C. Figure 8E shows that compounds 1, 80 and 101 facilitated dissociation of preformed ULC in a dose dependent manner. Under the conditions examined, breakdown was incomplete (some ULC were still present). However, for compounds 1 and 101, less than 50% of ULC remained as compared to the baseline. Of interest, compound 96 did not promote breakdown of preformed ULC despite successfully preventing de novo ULC formation, as discussed in Example 4. Compound 34 did not inhibit the stability of preformed ULC.
Example 4: Inhibition of ULC Formation
Since the data of Example 3 demonstrated that the PF4 antagonists discussed herein are capable of disrupting preformed ULCs, this example was performed to determine if the PF4 antagonists of the invention were capable of preventing formation of ULCs.
A. ELISA Measurement of ULC Inhibition
In this experiment, solutions containing varying concentrations of compound 1 (1, 2, 4, 8, 16, 31, 62, 125, 250, 500, and 100 μΜ), 34 (125, 250, 500, and 1000 μΜ),
80 (31, 62, 125, 250, 500, and 1000 μΜ), 101 (8, 16, 31, 62, 125, 250, and 500 μΜ), and 96 (16, 31, 62, 125, 250, 500, 1000, 2500, and 5000 μΜ) were preincubated with
PF4 (7.5 μg/mL) for 1 hour at 37°C. Heparin (0.2 U/mL) was then added to each sample and the mixtures (final PF4 concentration of 5 μg/mL) were incubated for an additional 15 minutes at 37 °C. The total percentage of ULC (%) was measured by
ELISA and the data plotted. See, Figure 8A which is line graph demonstrating ULC antagonistic activity for the tested compounds.
The data of Figure 8 A show that compounds 1, 80, 101, and 96 inhibited ULC formation in a dose-dependent matter. Inhibition was complete at the highest concentrations tested. Further, the relative potency of the tested PF4 antagonists, as measured by IC50 and provided in the following Table 5 is similar to that in Example
2.
Table 5
Figure imgf000073_0001
PF4 Antagonist ICso (μΜ)
101 190
96 1300
B. Gel Filtration Measurement of ULC Inhibition
In this experiment, independent 1 mM solutions of compound 1, 34, 80, 101, and 96 were preincubated with PF4 (7.5 μg/mL) for 1 hour at 37°C. Heparin (0.2 U/mL) was then added to each sample and the mixtures (final PF4 concentration of 5 μg/mL) were incubated for an additional 15 minutes at 37 °C. One control contained PF4K50E and the other control contained PF4 (5 μg/mL) and heparin (0.2 U/mL). The total percentage of ULC (%) was measured using gel filtration as described in Ranuova, Blood, 10591): 131-138 (2005) cited above and the percentage of ULC provided for the tested compounds. See, Figure 8B which is a bar graph
demonstrating ULC antagonistic activity for the tested compounds.
The data of Figure 8B shows that compounds 1, 80, 101, and 96 substantially inhibited ULC formation. C. Varying Heparin Concentrations on ULC Inhibition
In this experiment, the ability of PF4 antagonists to inhibit ULC formation over a range of heparin concentrations was determined. Specifically, 1 mM of each of compounds 1, 34, 80, and 101 were independently preincubated with PF4 (7.5 μg/mL) for 1 hour, followed by the addition of varying concentrations of heparin (0.05, 0.1, 0.2, 0.4, 0.8, and 1.6 U/mL). The controls contained PF4 (7.5 μg/mL) and varying concentrations of heparin (0.05, 0.1, 0.2, 0.4, 0.8, and 1.6 U/mL). Absorbance of each solution was measure at 450 nm in a Packard SpectraCount™ plate reader. See, Figure 8C which provides the absorbance for each of the varying heparin containing solutions.
The data of Figure 8C illustrates that 1, 80, and 101 inhibited ULC formation over a range of heparin concentrations that span the parabolic dose response curve. These data show that inhibition of ULC by PF4 antagonists is not dependent on the amount of heparin present. Example 5: Effect of PF4 Antagonists on Antibody Binding to ULCs
The effect of the PF4 antagonists of the invention on antibody binding to intact ULCs was evaluated. ULC were formed as described in Part B of Example 2, with the exception that the PF4 was incubated with heparin for 30 minutes.
Compounds 1, 34, 80, 101, and 96 (1 mM) were then added to wells pre-coated with KKO. These solutions were then incubated in the KKO wells overnight at 37°C. Antibody binding was detected by adding HRP -conjugated sheep polyclonal anti- human PF4 antibody and developed with TMB substrate. After stopping the enzymatic reaction with 1 M H3PO4, absorbance was measured at 450 nm in a Packard SpectraCount™ plate reader. A direct ELISA was then performed as described in Arepally.
Figure 8D demonstrates antibody binding to ULCs in the absence and presence of PF4 antagonists of the invention. Specifically, these data show that compounds 1, 34, 101, and 96 do not directly inhibit antibody binding to preformed ULCs.
Example 6: Inhibition of Platelet Activation
Since one of the therapeutic goals of using ULC antagonists in patients with HITT is to prevent activation, compound 96 was evaluated for its ability to inhibit platelet activation. These data were generated using a serotonin release assay as described in Hirschman, "The use of platelet serotonin release as a sensitive method for detecting anti-platelet antibodies and a plasma anti-platelet factor in patients with idiopathic thrombocytopenic purpura", Br. J. Haematol, 24(6):793-802 (1973), which is hereby incorporated by reference.
In this assay, platelet rich plasma (PRP) from healthy donors was incubated with 0.5 μϊ^ carbon- 14 labeled 5-hydroxytryptamine creatinine sulfate (GE Life Sciences, Piscataway, NJ) per milliliter of PRP for 20 minutes at 37°C. Serotonin uptake was inhibited by adding 1 mmol/mL imipramine (Sigma-Aldrich, St. Louis, MO) to the PRP.
Negative and positive controls contained sera from patients previously known to have negative or positive serotonin release, respectively. Heparin (1 U/mL) was added to the positive control serum in the absence or presence of compound 96 (3 niM). The percent release of serotonin was calculated for all conditions as previously described and plotted. Figure 6 is a bar graph of % of serotonin release for samples lacking (buffer only) and containing compound 96.These data illustrate that compound 96 is effective in inhibiting serotonin release, thereby inhibiting platelet activation.
Example 7: Serotonin Release Assay
This example assesses the activity of certain PF4 antagonists described herein as described in Hirschman, "The use of platelet serotonin release as a sensitive method for detecting anti-platelet antibodies and a plasma anti-platelet factor in patients with idiopathic thrombocytopenic purpura", Br. J. Haematol, 24(6):793-802 (1973), and Rauova and Cines, "Heparin-associated thrombocytopenia", N. Engl. J. Med., 303 :788-795 (1980), which are hereby incorporated by reference.
Briefly, platelet rich plasma (PRP) from healthy donors was incubated with 0.5 μΐ^ 14C-5-hydroxytryptamine creatinine sulfate (GE Life Sciences, Piscataway, NJ) per milliliter of PRP for 20 minutes at 37°C to produce 14C-labeled platelets. Serotonin uptake was inhibited by adding 1 mmol/mL imipramine (Sigma-Aldrich, St. Louis, MO) to the PRP.
The radiolabeled platelets were mixed with KKO (170 μg/mL) or with known platelet-activating HIT plasma in the absence (buffer control) or presence of PF4 antagonist compounds 1, 34, 80, 101, and 96 (2.5 mM). The assay was performed in the presence of heparin (1.0 U/mL) and in the absence of heparin (background).
Negative controls without antibody were studied in parallel. 14C-5-hydroxytryptamine released from platelets is measured by scintillation counter. Data are expressed as % maximal release of radioactivity with release by the positive control plasma and 1.0 U/mL heparin defined as 100%.
Compounds 1, 80, 101 and 96 inhibited platelet activation by KKO as measured by the serotonin release assay (Figure 9E). The rank order of potency for inhibition of cellular activation by these compounds paralleled their ability to inhibit tetramer formation. Further, compounds 1 and 101 inhibited serotonin release from platelets activated by plasma from patients with HIT (Figure 9F). Example 8: HIT antibodies bound to ULC on platelets initiate activation by engaging FcyRIIA
This example was performed using the described below. Further, the assays described in the application entitled "Methods of Identifying HIT -Antibodies and PF4 Antagonists and Cells Lines for Use Therein", i.e., US Provisional Patent Application No. 61/614,829, filed March 23, 2012 and hereby incorporated by reference.
A. Preparation of B cells containing NFAT-Luc and FcyRIIA plasmids.
NFAT-Luc was prepared as described in Shapiro, "c-rel Regulation of IL-2 gene expression may be mediated through activation of AP-1 ", J. Exp. Med.,
184(5): 1663-1669 (1996), hereby incorporated by reference. The NFAT reporter contained three copies of a composite NFAT-activator protein- 1 (AP-1) element from the human interleukin-2 (IL-2) gene promoter, and is activated by binding of NFAT (nuclear factor of activated T-cells). pEF6-FcyRIIA, which contained the human FcyRIIA coding sequence under the control of the human EF- 1 a promoter, was constructed by cloning a human FcyRIIA IMAGE clone into the multiple cloning site of pEF6c (Invitrogen).
DT40 cells (chicken B cells) were cultured in RPMI supplemented with 10% fetal bovine serum, 1% chicken serum (Gibco), 50 μΜ 13-mercaptoethanol, 2 mM GlutaMAX (Gibco), 100 U/mL penicillin and 100 U/mL streptomycin at 37°C under 5% CO2. Cells were co-transfected with pEF6-FcyRIIA (5 μg) and pEF6-NFAT-Luc (20 μg) by electroporation.
B. Validation of B Cells
This experiment validated that the transfected cells were useful in measuring inhibition of FcyRIIA mediated activation by HIT antibodies in the presence of PF4 and heparin.
On the day following transfection as described in Pary A, cells were pelleted by centrifugation and re-suspended at 2 x 106 cells/mL in fresh medium without serum. Cell aliquots (50 μΚ) were placed into each well of a 96 well plate. To establish basal expression, 50 μΐ^ media was added. The cells were also incubated, independently, with the following: PF4, heparin, RTO antibody, KKO antibody, ULCs alone, ULCs and RTO; ULCs and KKO and IV.3 antibody. Monoclonal antibody IV.3 (FcyRIIA-blocking antibody) was used as a positive control for FcyRIIA signaling. IV.3 (8 μg/mL) was added for 15 minutes at 37°C under 5% CO2, followed by sheep anti-mouse-lgG secondary antibody (50 uL) at 15 μg/mL to crosslink and activate these receptors. As a positive control for an intact intracellular signaling pathway, phorbol myristate acetate (PMA; 25 ng/mL) with ionomycin (1 μΜ) was added.
Data show that the cells are activated by the antiFcyRIIA monoclonal antibody IV.3, as well as by the HIT-like monoclonal antibody KKO in the presence of ULC (Figure 9A). However, RTO, an isotype control that binds avidly to PF4 but does not mimic the pathological properties of HIT, failed to activate these cells. Importantly, KKO/ULC did not activate cells lacking FcyRIIA (transfected with pEF6-NFAT-Luc alone) which demonstrated that activation in this system is FcyRIIA dependent.
Furthermore, addition of PF4, heparin, KKO, or RTO alone did not activate the FcyRIIA-expressing cells, demonstrating the relevance of this assay in measuring the functional activity of HIT antibodies (Figure 9A).
It was next examined whether FcyRIIA activation was heparin-dependent. To do so, activation by KKO was measured using a constant amount of PF4 and increasing amounts of heparin. As seen in Figure 9B, the previously described bell- shaped dose-dependence of heparin concentration on platelet activation was observed. Taken together, the data support the use of the DT40 cell activation system as an assay of cellular activation via FcyRIIA dependent on PF4, heparin and HIT antibody and therefore as a means to assess the effect of PF4 antagonists discussed herein.
See, Kelton, "Heparin-induced thrombocytopenia: Laboratory studies", Blood, 72(3):925-930 (1988) and Reilly, "Heparin-induced thrombocytopenia/thrombosis in a transgenic mouse model requires human platelet factor 4 and platelet activation through FcyRIIA", 98(8):2442-2447 (2001), which are hereby incorporated by reference. Example 9: Measurement of FcyRIIA activation in DT40 cells
This example measured the inhibitory capacity of the PF4 antagonist compounds described herein. PF4 (10 μg/mL) and various concentrations of compounds 1, 34, 80, 96, and 101 were independently co-incubated for 60 minutes at 37°C under 5% CO2 followed the addition of heparin (0.3 U/mL) for 15 minutes at 37°C in 5% CO2. KKO (20 μg/mL) or plasma from patients with serotonin release assay confirmed HIT (1 :800 dilution) was then added. Fifty μϊ^ of each mixture was added to the 96 well plate containing 50 μΐ^ of cells at a concentration of 2 x 106 cells/mL in fresh medium without serum. Plates were incubated for 6 hours at 37°C under 5% CO2, and then frozen at -80°C. To measure activation, cells were thawed and lysed with 5X Passive Lysis Buffer (Promega) for 15 minutes. Luciferase activity was measured on a Berthold MultiLumat™ LB 9506 Luminometer (10 second readings) using Promega's Luciferase Assay Reagent following the manufacturer's instructions.
These data illustrate that compounds 1 and 101 were the most potent inhibitors of cellular activation (Table 6 and Figure 9C), causing complete inhibition at the highest doses tested. Compounds 80 and 96 were less potent and compound 34 was not inhibitory. Importantly, compounds 1 and 101 both inhibited activation by human HIT plasma with a similar potency as activation by KKO (Figure 9D).
Table 6
Figure imgf000079_0001
Example 10: Effect of PF4 Mutant, PF4 , on PF4 Solutions
This example provides the results using non-tetramerizing PF4 mutant, PF4K50E as an antagonist of WT PF4 on solutions containing PF4.
PF4K50E was prepared and purified as described for WT-PF4 in Example 2, but using MES (50 mM), EDTA (1 mM), pH 6.5 as the column buffer system. Media was loaded in buffer containing 0.3 M NaCl and PF4K50E eluted at NaCl (1.3 M) using a linear gradient.
PF4 and PR4K50E (total mass 10 μg/mL) were incubated for 60 minutes at room temperature, followed by the addition of cross-lining reagent BS (0.2 mM). Independent solutions of PF4 and PF4 (total mass 7.5 μg/mL) were incubated for 1 hour, followed by addition of heparin (0.2 U/mL). The samples were analyzed by ELISA, as described in Example 4 (Figure 8A), and data generated therefrom compiled.
Figures 1 OA- IOC show that while keeping the total PF4 concentration constant, as the percentage of WT PF4 mixed with PF4K50E is decreased,
tetramerization (Figure 10A), ULC formation (Figure 10B) and FcyRIIA activation (Figure IOC) are all inhibited in a similar dose dependent manner. These data provide independent evidence that inhibition of PF4 tetramerization leads to inhibition of FcyRIIA activation by decreasing ULC formation.
Example 11 : Inhibition of Endothelial Activation using ULC antagonists
The 15 compounds from Example 1 are examined for their ability to inhibit HITT related cellular activation. Specifically, the ability of these compounds to inhibit platelet activation from HITT antibodies is measured. These compounds are also analyzed for their ability to inhibit PF4 activation of NFKB in endothelial cells in vitro.
A. Inhibition of cellular activation as related to HITT
The 15 compounds from Example 1 are evaluated for their ability to inhibit cellular activation related to HIT using the assays described in Warkentin, "Heparin- induced thrombocytopenia: diagnosis and management", Circulation, 110(18):e454-8 (2004) and Kelton, "Heparin-induced thrombocytopenia: a historical perspective", Blood, 1 12(7):2607-16 (2008).
It is anticipated that the data will show that the 15 compounds inhibit cellular activation as related to HITT.
B. Inhibition of PF4 activation of NFKB
The 15 compounds from Example 1 are analyzed for their impact on the ability of PF4 to activate NFKB in endothelial cells as described in Yu, "Endothelial expression of E-selectin is induced by the platelet-specific chemokine platelet factor 4 through LRP in an NF- {kappa} B-dependent manner", Blood, 105(9):3545 (May 1, 2005), which is hereby incorporated by reference using human umbilical vein endothelial cells (HUVECs).
The data are anticipated to provide support that the antagonists discussed herein inhibit PF4 tetramerization and interfere with PF4 activation of endothelial cells, which is directly relevant to HITT.
Example 12: Prevention of Thrombocytopenia and Thrombosis in a Murine Model using ULC Antagonists
The 15 compounds from Example 1 are evaluated for their ability to decrease thrombocytopenia and thrombosis in the HITT murine model system as described in Reilly I and Reilly II cited above and incorporated herein by reference.
The murine model system consists of mice homozygous for FcyRIIa and hPF4 and null for mPF4, hereafter designated IIa+/+/hPF4+/+/mPF4 ~/_.
IIa+/+/hPF4+/+/mPF4 -/_ mice are weaned at 3 weeks of age and fed on SD (Lab Rodent Chow, Purina 5020; 4.5% fat). At 6 weeks of age, sex- and weight-matched littermates are either maintained on the SD (normocholesterolemic controls) or switched to a hypercholesterolemic diet (HD). The HD contains 15% cocoa butter, 1% cholesterol and 0.5% sodium cholate, in addition to essential nutrients, vitamins and minerals (TD.88051, Harlan-Teklad, Madison, WI, USA). HD-fed animals remain on the diet for only 4 weeks {i.e., until 10 weeks of age).
Mice are anesthetized by isofluorane inhalation for i.p. and s.c. injections and for blood collections. Blood is collected via the retro-orbital plexus using 50 μΐ, EDTA- or heparin-coated microcapillary tubes (Microcaps® tubes, Drummond Scientific, Broomall, PA, USA).
Plasma cholesterol is measured in all mice at 6 weeks of age (i.e., before initiation of the HD in the experimental group) and weekly thereafter. Mice in both diet groups are fasted for 4 hours before each blood collection. Total plasma cholesterol is determined by enzymatic assay (Thermo Electron, Louisville, CO, USA). Reactivity of platelets from HD-fed mice and SD-fed controls is evaluated at the age of 10 weeks by whole blood impedance aggregometry (the Chronolog® 540- VS instrument, Chronolog Corp., Havertown, PA, USA). Whole blood, collected in heparin, is mixed with 0.9% NaCl to adjust the final platelet count to 150,000 μ/L. Platelet aggregation is induced by collagen (1.5 μg/mL) and measured as the increase in impedance over time. Plasma levels of soluble vascular cell adhesion molecule (sVCAM) are measured by ELISA (Mouse sVCAM Immunoassay, R & D Systems, Minneapolis, M , USA).
After four weeks of HD feeding, each mouse receives i.p. injections of an antagonist compound of Example 1, followed by daily s.c. injections of unfractionated heparin (1200 or 1400 U/kg). Blood samples are collected from each mouse in EDTA, beginning 3 days before antagonist compound administration and are utilized as control samples 1-13, and then on days 1, 2, 3, 4 and 7 after antagonist compound injections. Complete blood counts on all 26 samples are measured using a Hemavet® analyzer (Model 850, CDC Technologies, Oxford, CT, USA). Platelet counts are monitored to assess thrombocytopenia. Red blood cell counts and hematocrit are monitored to ensure against unapparent hemorrhage. Thrombin-antithrombin complex III (TAT) levels are measured by an ELISA (Enzygnost® TATmicro reagents, Dade Behring, Newark, DE, US).
Experimental mice exhibiting signs of distress subsequent to heparin and antagonist compound injections are sacrificed by CO2 inhalation. The major organs (lungs, heart, liver, kidneys, and spleen) are immediately harvested and fixed in neutral-buffered 10% formalin. Similarly treated, paired SD-fed mice are sacrificed at the same time. Formalin-fixed organs are embedded in paraffin and cut into 5-μιη sections. Hematoxylin and eosin-stained sections are evaluated by a comparative pathologist blinded to the experimental conditions. The area of each tissue examined is measured using the NIH ImageJ® software and the number of thrombi per 10 mm2 are determined.
For immunohistochemistry, sections are sequentially blocked with ¾(¾ in methanol (to inhibit endogenous peroxidase activity) and 10% mouse serum before staining with directly biotinylated anti-hPF4 antibodies. Specificity for hPF4 is confirmed by staining mouse spleens, where antibodies react specifically with megakaryocytes.
The bound antagonist compounds are detected by incubating biotinylated horseradish peroxidase plus avidin followed by staining with diaminobenzidine. Immunohistochemistry sections are then lightly counterstained with hematoxylin and examined by light microscopy.
The data are anticipated to show that mice treated with the 15 compounds of Example 1 did not develop thrombocytopenia or that such treatment decreases the severity of thrombocytopenia. It is also expected that the data will show that mice treated with the 15 compounds of Example 1 did not develop thrombosis. Finally, it is expected that these data will demonstrate that the 15 compounds of Example 1 prevent formation of PF4 tetramers and PF4 tetramenheparin ULCs when administered concurrently with heparin administration.
All publications and priority application including US Provisional Patent Application No. 61/614,709, filed March 23, 2012, cited in this specification are incorporated herein by reference. While the invention has been described with reference to particular embodiments, it will be appreciated that modifications can be made without departing from the spirit of the invention. Such modifications are intended to fall within the scope of the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A compound for use in preventing formation of platelet factor-4 (PF4) tetramers in a subject, wherein said compound is selected from the group consisting of:
(a) a compound of formula (II):
Figure imgf000084_0001
(II)
wherein:
n is 0 to 5;
R1 and R2 are, independently, H, Ci to C10 alkyl, C(0)(C1 to C10 alkyl), C(0)(Ci to Cio alkyl), benzyl, C3 to C10 cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl; or
R1 and R2 together comprise a heterocycle or heteroaryl, optionally substituted by one or more R5;
R3 and R4 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, benzyl, aryl, heteroaryl, or heterocycle; or
R1 and R3 together comprise a heterocycle or heteroaryl, each optionally substituted by one or more R5;
R5 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, or (=0);
(b) a compound of formula (III):
OR16
I
0^P^(CR17R18)Q-R10
(HI)
wherein:
q is 0 to 5;
R10 is Ci to Cio alkyl. heteroaryl or aryl, each optionally substituted by one or more R19;
R16 is H or Ci to Cio alkyl; R17 and R18 are, independently, absent, H, Ci to C10 alkyl, (Ci to C10 alkyl)-P-(Ci to C10 alkyl), Ci to C10 alkoxy, halogen, OH, N02, CN, C2 to C10 alkenyl, P(0)OH, C2 to C10 alkynyl, aryl, heteroaryl, or heterocycle, each optionally substituted by one or more R19;
17 18 17 18 wherein when R or R is absent and q is at least 2, the CR R groups are bound through a stable double bond;
R19 is H, Ci to Cio alkyl, Ci to C10 alkoxy, halogen, N02, CN, C(0)OH, C(0)(Ci to Cio alkyl), C2 to Cio alkenyl, oxo, or C2 to Cio alkynyl; (c) a compound of formula (IV):
Figure imgf000085_0001
(IV)
wherein:
m is 0 to 2;
R20 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R24;
R21, R22, and R23 are independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl, each optionally substituted by one or more R24;
R24 is H, halogen, OH, CN, N02, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R27;
R25 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl;
R26 is O or S;
R27 is H, P(0)OH, or Ci to Cio alkyl optionally substituted by halogen or OP(0)OH;
(d) a compound of formula (V):
O II
. P.
(V)
wherein: R and R are, independently, H, Ci to C10 alkyl optionally substituted by one or more R42, aryl optionally substituted by one or more or benzyl optionally substituted by one or more R42;
R42 is H, Ci to Cio alkyl, C(0)OH, NH2, C(0)(d to C10 alkyl), C(0)0(Ci to Cio alkyl), Ci to Cio aminoalkyl, halogen, aryl, or heteroaryl;
(e) a compound of formula (VI):
OR51
0=P— OR50
I
R52
(VI)
wherein:
R50 is heteroaryl or heterocycle, each optionally substituted by one or more R53;
R51 is H, Ci to Cio alkyl, Ci to Cio aminoalkyl, each optionally substituted by halogen or C(0)OH;
R52 is absent, Ci to Cio alkyl optionally substituted by P(0)2, or aryl optionally substituted by 0(Ci to Cio alkyl)-phenyl-C(0)OH;
R53 is Ci to Cio alkyl, halogen, or C(0)OH;
(f) a compound of formula (VII):
O
\\
P— (CR65R66)tR60R61
HO
(VII)
wherein:
t is 1 to 10;
R60 is aryl, heteroaryl, or heterocycle;
R61 is Ci to Cio aminoalkyl, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, heteroaryl, or heterocycle;
R65 and R66 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl benzyl, aryl, heteroaryl, or heterocycle;
(g) a compound of formula (VIII):
Figure imgf000087_0001
(VIII)
wherein:
R , R , R/4, and R/5 are, independently, H, Ci to C10 alkyl, C2 to C10 alkenyl, Ci to Cio aminoalkyl, C(0)(Ci to Cio aminoalkyl), or heteroaryl, each optionally substituted by one or more R78;
R72 is H, Ci to Cio alkyl, 0(d to Cio alkyl), 0(d to Cio alkoxy), 0(d to Cio aminoalkyl), Ci to Cio aminoalkyl, or heteroaryl, each optionally substituted by one or more R76;
R73 is Ci to Cio alkyl, Ci to Cio alkoxy, Ci to Cio aminoalkyl, each optionally substituted by one or more R76;
R76 is C(0)OH, C(0)(Ci to Cio alkyl), NHC(0)(Ci to Cio alkyl), C(0)NH(phenyl optionally substituted by one or more R77), C(0)0(C1 to Cio alkyl), heteroaryl optionally substituted by one or more R77, heterocycle optionally substituted by one or more R77, or phenyl optionally substituted by one or more R77;
R77 is halogen, Ci to Cio aminoalkyl, Ci to Cio alkoxy, Ci to Cio alkyl, or C2 to Cio alkenyl, each optionally substituted by one or more R78;
R78 is CN, C(0)OH, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), or phenyl optionally substituted by one or more R79;
R79 is OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, halogen, or Ci to Ce alkoxy;
(h) a compound of formula (IX):
Figure imgf000087_0002
(IX)
wherein:
w is 1 to 10; w' is 1 to 10;
R80 is H or C2 to Cio alkyl;
R81 to R85 are, independently, H, OH, Ci to Cio alkyl, halogen, or Ci to Cio alkoxy; and
R86 to R89 are, independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl;
(i) a compound of formula (X):
Figure imgf000088_0001
(X)
wherein:
x is 2 to 10;
(j) a compound of formula (XI):
Figure imgf000088_0002
(XI)
wherein:
y is 1 to 10;
R90 to R94 are, independently, H, halogen, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, OH, or Ci to Cio aminoalkyl; and
R95 is heterocycle, heteroaryl, aryl, or Ci to Cio aminoalkyl;
(k) a compound of formula (XII):
Figure imgf000088_0003
(XII)
wherein: R is aryl, heteroaryl, or heterocycle;
z is 2 to 10;
R106 to R110 are, independently, H, Ci to C10 alkyl, C3 to C10 cycloalkyl, OH, C2 to Cio alkenyl, or C2 to C10 alkynyl;
(1) a compound of formula (XIII):
Figure imgf000089_0001
(XIII)
wherein:
a is 0 or 1;
R120 is absent, aryl, C3 to C10 cycloalkyl, heteroaryl, or heterocycle, each optionally substituted by one or more R125;
R121 is aryl containing one or more R123, Ci to C10 aminoalkyl optionally containing one or two aryl groups optionally substituted by one or more R124, heterocycle optionally substituted by one or more R125;
R123 is H, halogen, Ci to C10 alkyl, or Ci to C10 alkoxy;
R124 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, or Ci to C10 aminoalkyl;
R125 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, (=0), or C(0)OH; or
R and R join to form C5 to Cio cycloalkyl optionally substituted by one or more R126; and
R126 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, or Ci to Cio aminoalkyl;
(m) a compound of fo
Figure imgf000089_0002
(XIV)
wherein: R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R202;
R202 is H, CI to Cio alkyl, halogen, CN, or 02; R200 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally
203 203
substituted by one or more R ;R is halogen, CN, NO2, Ci to Cio alkoxy, or aryl optionally substituted by one or more R204;
R204 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl; (n) a compound of formula (XV):
Figure imgf000090_0001
wherein:
R301 is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, or C2 to
R302
Cio alkynyl, each optionally substituted by one or more
R302 is H, Ci to Cio alkyl, halogen, CN, or N02; R300 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R303;
R303 is halogen, CN, NO2, or aryl optionally substituted by one or more
304.
R
R is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl;
(0) a compound of formula (XVI):
Figure imgf000090_0002
(XVI)
wherein:
R400 is OH or O;
R401, R402, and R403, are independently, OR405 or N(R406)(R407);
R404 is H, Ci to Cio alkyl, halogen, (=0), or Ci to Cio alkoxy; R405 is absent, Ci to C10 alkyl, heterocycle, aryl, or heteroaryl,
R406
optionally substituted by one or more
R406 is H, Ci to Cio alkyl, halogen, Ci to C10 alkoxy, C(0)(Ci to C10 alkyl), aryl, heteroaryl, C(0)(aryl optionally substituted by one or more R404
R407 is absent, H, Ci to C10 alkyl, halogen, Ci to C10 alkoxy, aryl, or heteroaryl;
with the proviso that when R400 is O and R405 or R407 is absent, then R400 and R402 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404; and
with the proviso that when R401 is OR405 and R403 is N(R406)(R407) and R405 and R407 are absent, R401 and R402, and/or R401 and R403, and/or R402 and R403 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404;
(p) a compound of formula (XVII):
Figure imgf000091_0001
(XVII)
wherein:
R is Ci to Cio aminoalkyl;
R502 is benzyl optionally substituted by one or more R503; R501 is H, Ci to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or aryl;
f is 0 to 4; and
R503 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, or aryl;
(q) a compound of formula (XVIII):
Figure imgf000091_0002
(XVIII)
wherein:
R507 is H, OH, C i to Cio alkoxy, or C3 to C10 cycloalkyl;
R508 tQ R5io are^ independently, H, Ci to Cio alkyl, d to Cio alkoxy, to Cio cycloalkyl, or halogen; and
(r) a compound of formula (XIX):
Figure imgf000092_0001
(XIX)
wherein:
R600 is Ci to Cio alkyl, NH2 or Ci to Cio aminoalkyl;
R602 to R605 are, independently, Ci to Cio alkyl, d to Cio cycloalkyl, Ci to Cio alkoxy, or halogen;
(s) a compound of formula (XX):
Figure imgf000092_0002
(XX)
wherein:
R606 is H or Ci to Cio alkyl;
R607 to R610 are, independently, H, d to Cio alkyl, d to Cio alkoxy, or halogen;
(t) a compound of formula (XXI):
Figure imgf000092_0003
(XXI) wherein:
R7oo tQ R704 ar6j independently, H, Ci to C10 alkyl, C3 to CIO cycloalkyl, C3 to C10 spirocycloalkyl, Q to C10 alkoxy, halogen, Ci to C10 aminoalkyl, or CN; and
R705 is H or Ci to C10 alkyl;
(u) a compound of formula (XXII):
Figure imgf000093_0001
(XXII)
wherein:
R8oo tQ R804 and R806 ^ R809 ^ independently, H, Ci to C10 alkyl, halogen, NH2, or Ci to C10 alkoxy and
805 is H or Ci to C10 alkyl;
Figure imgf000093_0002
(XXIII)
wherein:
RJUU is H or Ci to C10 alkyl; and
R901 to R905 is H, Ci to Cio alkyl, C2 to C10 alkenyl, C2 to C10 alkynyl, halogen, Ci to C10 alkoxy, Ci to C10 aminoalkyl, or H2; and
(w) a compound of formula (XXIV):
Figure imgf000094_0001
(XXIV)
wherein:
R906 is Ci to Cio alkyl or Ci to C10 alkoxy;
R907 is C2 to Cio alkyl or Ci to Cio alkoxy;
e is 0 to 5;
R908 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, OH, CN, or NH2; and
R909 is H or Ci to Cio alkyl;
or a pharmaceutically acceptable salt or prodrug thereof.
2. The compound according to claim 1, wherein said compound binds to a PF4 dimer or PF4 monomer.
3. A compound for use in disrupting platelet factor-4 (PF4) tetramers in a subject, wherein said compound is selected from the group consisting of:
(a) a compound of formula (II):
Figure imgf000094_0002
(II)
wherein:
n is 0 to 5;
R1 and R2 are, independently, H, Ci to Cio alkyl, C(0)(Ci to Cio alkyl), C(0)(Ci to Cio alkyl), benzyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl; or
R1 and R2 together comprise a heterocycle or heteroaryl, optionally substituted by one or more R5;
R3 and R4 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, benzyl, aryl, heteroaryl, or heterocycle; or
R1 and R3 together comprise a heterocycle or heteroaryl, each optionally substituted by one or more R5; R5 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, or (=0);
(b) a compound of formula (III):
OR 16
O' (CR17R18)q-R 10
(III)
wherein:
q is 0 to 5;
R10 is Ci to Cio alkyl. heteroaryl or aryl, each optionally substituted by one or more R19;
R16 is H or Ci to Cio alkyl;
R17 and R18 are, independently, absent, H, Ci to Cio alkyl, (Ci to Cio alkyl)-P-(Ci to Cio alkyl), Ci to Cio alkoxy, halogen, OH, N02, CN, C2 to Cio alkenyl, P(0)OH, C2 to Cio alkynyl, aryl, heteroaryl, or heterocycle, each optionally substituted by one or more R19;
wherein when R17 or R18 is absent and q is at least 2, the CR17R18 groups are bound through a stable double bond;
R19 is H, Ci to Cio alkyl, Ci to C10 alkoxy, halogen, N02, CN, C(0)OH, C(0)(Ci to Cio alkyl), C2 to Cio alkenyl, oxo, or C2 to Cio alkynyl; (c) a compound of formula (IV):
Figure imgf000095_0001
(IV)
wherein:
m is 0 to 2;
R20 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R24;
R21, R22, and R23 are independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl, each optionally substituted by one or more R24; R is H, halogen, OH, CN, N02, d to C10 alkyl C3 to C10 cycloalkyl, C2 to Cio alkenyl, or C2 to C10 alkynyl, each optionally substituted by one or more R27;
R25 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl;
R26 is O or S;
R27 is H, P(0)OH, or Ci to Cio alkyl optionally substituted by halogen or OP(0)OH;
(d) a compound of formula (V):
O II
R41' P^R40
(V)
wherein:
R40 and R41 are, independently, H, Ci to Cio alkyl optionally substituted by one or more R42, aryl optionally substituted by one or more R42, or benzyl optionally substituted by one or more R42;
R42 is H, Ci to Cio alkyl, C(0)OH, NH2, C(0)(d to Cio alkyl), C(0)0(Ci to Cio alkyl), Ci to Cio aminoalkyl, halogen, aryl, or heteroaryl;
(e) a compound of formula (VI):
OR51
0=P— OR50
I
R52
(VI)
wherein:
R50 is heteroaryl or heterocycle, each optionally substituted by one or more R53;
R51 is H, Ci to Cio alkyl, Ci to Cio aminoalkyl, each optionally substituted by halogen or C(0)OH;
R52 is absent, Ci to Cio alkyl optionally substituted by P(0)2, or aryl optionally substituted by 0(Ci to Cio alkyl)-phenyl-C(0)OH;
R53 is Ci to Cio alkyl, halogen, or C(0)OH;
(f) a compound of formula (VII):
Figure imgf000097_0001
(VII)
wherein:
t is 1 to 10;
R60 is aryl, heteroaryl, or heterocycle;
R61 is Ci to Cio aminoalkyl, Ci to C10 alkyl, C2 to C10 alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, heteroaryl, or heterocycle;
R65 and R66 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl benzyl, aryl, heteroaryl, or heterocycle;
(g) a compound of formula (VIII):
Figure imgf000097_0002
(VIII)
wherein:
R , R , R , and R/5 are, independently, H, Ci to Cio alkyl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, C(0)(Ci to Cio aminoalkyl), or heteroaryl, each optionally substituted by one or more R78;
R72 is H, Ci to Cio alkyl, 0(Ci to Cio alkyl), 0(Ci to Cio alkoxy), 0(Ci to Cio aminoalkyl), Ci to Cio aminoalkyl, or heteroaryl, each optionally substituted by one or more R76;
R73 is Ci to Cio alkyl, Ci to Cio alkoxy, Ci to Cio aminoalkyl, each optionally substituted by one or more R76;
R76 is C(0)OH, C(0)(Ci to Cio alkyl), NHC(0)(Ci to Cio alkyl), C(0)NH(phenyl optionally substituted by one or more R77), C(0)0(Ci to Cio alkyl), heteroaryl optionally substituted by one or more R77, heterocycle optionally substituted by one or more R77, or phenyl optionally substituted by one or more R77; R is halogen, Ci to C10 aminoalkyl, Ci to C10 alkoxy, Ci to C10 alkyl, or C2 to Cio alkenyl, each optionally substituted by one or more R78;
R78 is CN, C(0)OH, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), or phenyl optionally substituted by one or more R79;
R79 is OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, halogen, or Ci to Ce alkoxy;
(h) a compound of formula (IX):
Figure imgf000098_0001
(IX)
wherein:
w is 1 to 10;
w' is 1 to 10;
R80 is H or C2 to Cio alkyl;
R81 to R85 are, independently, H, OH, Ci to Cio alkyl, halogen, or Ci to Cio alkoxy; and
R86 to R89 are, independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl;
(i) a compound of formula (X):
Figure imgf000098_0002
(X)
wherein:
x is 2 to 10;
G) a compound of formula (XI):
Figure imgf000099_0001
(XI)
wherein:
y is 1 to 10;
R90 to R94 are, independently, H, halogen, Ci to C10 alkyl, C2 to C10 alkenyl, C2 to C10 alkynyl, OH, or Ci to C10 aminoalkyl; and
R95 is heterocycle, heteroaryl, aryl, or Ci to C10 aminoalkyl;
(k) a compound of formula (XII):
Figure imgf000099_0002
(XII)
wherein:
R105 is aryl, heteroaryl, or heterocycle;
z is 2 to 10;
R106 to R110 are, independently, H, Ci to C10 alkyl, C3 to C10 cycloalkyl, OH, C2 to Cio alkenyl, or C2 to C10 alkynyl;
(1) a compound of formula (XIII):
Figure imgf000099_0003
(XIII)
wherein:
a is 0 or 1;
R120 is absent, aryl, C3 to Cio cycloalkyl, heteroaryl, or heterocycle, each optionally substituted by one or more R125; R is aryl containing one or more R , Ci to C10 aminoalkyl optionally containing one or two aryl groups optionally substituted by one or more R124, heterocycle optionally substituted by one or more R125;
R123 is H, halogen, Ci to C10 alkyl, or Ci to C10 alkoxy;
R124 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, or Ci to Cio aminoalkyl;
R125 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, (=0), or C(0)OH; or
R and R join to form C5 to Cio cycloalkyl optionally substituted by one or more R126; and
R126 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, or Ci to Cio aminoalkyl;
(m) a compound of formula (XIV):
j2
Figure imgf000100_0001
(XIV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R202;
R202 is H, CI to Cio alkyl, halogen, CN, or 02; R200 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally
203 203
substituted by one or more R ;R is halogen, CN, NO2, Ci to Cio alkoxy, or aryl optionally substituted by one or more R204;
R204 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl; (n) a compound of fo
Figure imgf000100_0002
(XV)
wherein:
R301 is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to C10 alkenyl, or C2 to
R302
Cio alkynyl, each optionally substituted by one or more
R302 is H, Ci to Cio alkyl, halogen, CN, or 02; R300 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally
303.
substituted by one or more R
R is halogen, CN, NO2, or aryl optionally substituted by one or more
R 304.
R304 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl; (0) a compound of formula (XVI):
Figure imgf000101_0001
(XVI)
wherein:
R400 is OH or O;
R401, R402, and R403, are independently, OR405 or N(R406)(R407);
R404 is H, Ci to Cio alkyl, halogen, (=0), or Ci to Cio alkoxy;
R405 is absent, Ci to Cio alkyl, heterocycle, aryl, or heteroaryl,
R406
optionally substituted by one or more
R406 is H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, C(0)(Ci to Cio alkyl), aryl, heteroaryl, C(0)(aryl optionally substituted by one or more R404
R407
R is absent, H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, aryl, or heteroaryl;
with the proviso that when R400 is O and R405 or R407 is absent, then R400 and R402 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404; and
with the proviso that when R401 is OR405 and R403 is N(R406)(R407) and R405 and R407 are absent, R401 and R402, and/or R401 and R403, and/or R402 and R join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404;
(p) a compound of formula (XVII):
Figure imgf000102_0001
(XVII)
wherein:
R is Ci to Cio aminoalkyl;
R502 is benzyl optionally substituted by one or more R503; R501 is H, Ci to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or aryl;
f is 0 to 4; and
R503 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, or aryl;
(q) a compound of formula (XVIII):
Figure imgf000102_0002
(XVIII)
wherein:
R507 is H, OH, Ci to Cio alkoxy, or C3 to Cio cycloalkyl;
R508 to R510 are, independently, H, Ci to Cio alkyl, Ci to C10 alkoxy, C3 to Cio cycloalkyl, or halogen; and
(r) a compound of formula (XIX):
Figure imgf000102_0003
(XIX)
wherein:
R600 is Ci to Cio alkyl, NH2 or Ci to Cio aminoalkyl; R to R are, independently, Ci to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or halogen;
(s) a compound of formula (XX):
Figure imgf000103_0001
(XX)
wherein:
R606 is H or Ci to Cio alkyl;
R607 10 R6io are^ independently, H, Ci to Cio alkyl, d to Cio alkoxy, or halogen;
(t) a compound of formula (XXI):
Figure imgf000103_0002
(XXI)
wherein:
R7oo tQ R704 are^ independently, H, Ci to Cio alkyl, C3 to C I O cycloalkyl, C3 to Cio spirocycloalkyl, d to Cio alkoxy, halogen, d to Cio aminoalkyl, or CN; and
R705 is H or Ci to Cio alkyl;
(u) a compound of
Figure imgf000103_0003
(XXII)
wherein:
R8oo t0 R804 and R806 ω(1 R809 ^ independently; H> Q to Cl0 alkyl, halogen, NH2, or Ci to C10 alkoxy and
R805 is H or Ci to C10 alkyl;
(v) a compound o
Figure imgf000104_0001
wherein:
R900 is H or Ci to Cio alkyl; and
R9oi tQ R905 is Hj C i to alkyli C2 10 alkenyl5 Cl t0 C l0 aikyny^ halogen, Ci to Cio alkoxy, Ci to Cio aminoalkyl, or H2; and
(w) a compoun
Figure imgf000104_0002
(XXIV)
wherein:
R906 is Ci to Cio alkyl or d to C10 alkoxy;
R907 is C2 to Cio alkyl or Ci to C10 alkoxy;
e is 0 to 5;
R908 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, OH, CN, or NH2; and
RJUJ is H or Ci to Cio alkyl;
or a pharmaceutically acceptable salt or prodrug thereof
4. The compound according to claim 3, wherein said compound disrupts the salt bridge of said PF4 tetramer.
5. A compound for use in preventing formation of ultra-large complexes (ULCs) comprising a PF4 tetramer and a glycosaminoglycan (GAG) in a subject, wherein said compound is selected from the group consisting of:
(a) a compound of formula (II):
Figure imgf000105_0001
(II)
wherein:
n is 0 to 5;
R1 and R2 are, independently, H, Ci to C10 alkyl, C(0)(C1 to C10 alkyl), C(0)(Ci to Cio alkyl), benzyl, C3 to C10 cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl; or
R1 and R2 together comprise a heterocycle or heteroaryl, optionally substituted by one or more R5;
R3 and R4 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, benzyl, aryl, heteroaryl, or heterocycle; or
R1 and R3 together comprise a heterocycle or heteroaryl, each optionally substituted by one or more R5;
R5 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, or (=0);
(b) a compound of formula (III):
OR16
I
O ^(CR17R18)q-R10
(HI)
wherein:
q is 0 to 5;
R10 is Ci to Cio alkyl. heteroaryl or aryl, each optionally substituted by one or more R19;
R16 is H or Ci to Cio alkyl;
R17 and R18 are, independently, absent, H, Ci to Cio alkyl, (Ci to Cio alkyl)-P-(Ci to Cio alkyl), Ci to Cio alkoxy, halogen, OH, N02, CN, C2 to Cio alkenyl, P(0)OH, C2 to C10 alkynyl, aryl, heteroaryl, or heterocycle, each optionally substituted by one or more R19;
wherein when R17 or R18 is absent and q is at least 2, the CR17R18 groups are bound through a stable double bond;
R19 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, N02, CN, C(0)OH, C(0)(Ci to Cio alkyl), C2 to Cio alkenyl, oxo, or C2 to Cio alkynyl; (c) a compound of formula (IV):
Figure imgf000106_0001
(IV)
wherein:
m is 0 to 2;
R20 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R24;
R21, R22, and R23 are independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl, each optionally substituted by one or more R24;
R24 is H, halogen, OH, CN, N02, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R27;
R25 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl;
R26 is O or S;
R27 is H, P(0)OH, or Ci to Cio alkyl optionally substituted by halogen or OP(0)OH;
(d) a compound of formula (V):
O II
. P.
(V)
wherein: R and R are, independently, H, Ci to C10 alkyl optionally substituted by one or more R42, aryl optionally substituted by one or more or benzyl optionally substituted by one or more R42;
R42 is H, Ci to Cio alkyl, C(0)OH, NH2, C(0)(d to C10 alkyl), C(0)0(Ci to Cio alkyl), Ci to Cio aminoalkyl, halogen, aryl, or heteroaryl;
(e) a compound of formula (VI):
OR51
0=P— OR50
I
R52
(VI)
wherein:
R50 is heteroaryl or heterocycle, each optionally substituted by one or more R53;
R51 is H, Ci to Cio alkyl, Ci to Cio aminoalkyl, each optionally substituted by halogen or C(0)OH;
R52 is absent, Ci to Cio alkyl optionally substituted by P(0)2, or aryl optionally substituted by 0(Ci to Cio alkyl)-phenyl-C(0)OH;
R53 is Ci to Cio alkyl, halogen, or C(0)OH;
(f) a compound of formula (VII):
O
\\
P— (CR65R66)tR60R61
HO
(VII)
wherein:
t is 1 to 10;
R60 is aryl, heteroaryl, or heterocycle;
R61 is Ci to Cio aminoalkyl, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, heteroaryl, or heterocycle;
R65 and R66 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl benzyl, aryl, heteroaryl, or heterocycle;
(g) a compound of formula (VIII):
Figure imgf000108_0001
(VIII)
wherein:
R70, R71, R74, and R75 are, independently, H, Ci to C10 alkyl, C2 to C10 alkenyl, Ci to C10 aminoalkyl, C(0)(Ci to C10 aminoalkyl), or heteroaryl, each optionally substituted by one or more R78:
R72 is H, Ci to Cio alkyl, 0(d to C10 alkyl), 0(d to C10 alkoxy), 0(d to Cio aminoalkyl), Ci to C10 aminoalkyl, or heteroaryl, each optionally substituted by one or more R76;
R73 is Ci to Cio alkyl, Ci to Cio alkoxy, Ci to Cio aminoalkyl, each optionally substituted by one or more R76:
R76 is C(0)OH, C(0)(Ci to Cio alkyl), NHC(0)(Ci to Cio alkyl), C(0)NH(phenyl optionally substituted by one or more R77), C(0)0(C1 to Cio alkyl), heteroaryl optionally substituted by one or more R77, heterocycle optionally substituted by one or more R77, or phenyl optionally substituted by one or more R77;
R77 is halogen, Ci to Cio aminoalkyl, Ci to Cio alkoxy, Ci to Cio alkyl, or C2 to Cio alkenyl, each optionally substituted by one or more R78;
R78 is CN, C(0)OH, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), or phenyl optionally substituted by one or more R79;
R79 is OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, halogen, or Ci to Ce alkoxy;
(h) a compound of formula (IX):
Figure imgf000108_0002
(IX)
wherein: w is 1 to 10;
w' is 1 to 10;
R80 is H or C2 to Cio alkyl;
R81 to R85 are, independently, H, OH, Ci to Cio alkyl, halogen, or Ci to Cio alkoxy; and
R86 to R89 are, independently, H, Ci to Cio alkyl, or C3 to Cio
cycloalkyl;
(i) a compound of formula (X):
Figure imgf000109_0001
wherein:
x is 2 to 10;
(j) a compound of formula (XI)
Figure imgf000109_0002
(XI)
wherein:
y is 1 to 10;
R90 to R94 are, independently, H, halogen, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, OH, or Ci to Cio aminoalkyl; and
R95 is heterocycle, heteroaryl, aryl, or Ci to Cio aminoalkyl;
(k) a compound of formula (XII):
Figure imgf000109_0003
(XII)
wherein:
R105 is aryl, heteroaryl, or heterocycle;
z is 2 to 10;
R106 to R110 are, independently, H, Ci to Cio alkyl, C3 to Cio cycloalkyl, OH, C2 to Cio alkenyl, or C2 to Cio alkynyl;
(1) a compound of formula (XIII):
Figure imgf000110_0001
(XIII)
wherein:
a is 0 or 1;
R120 is absent, aryl, C3 to Cio cycloalkyl, heteroaryl, or heterocycle, each optionally substituted by one or more R125;
R121 is aryl containing one or more R123, Ci to Cio aminoalkyl optionally containing one or two aryl groups optionally substituted by one or more R124, heterocycle optionally substituted by one or more R125;
R123 is H, halogen, Ci to Cio alkyl, or Ci to Cio alkoxy;
R124 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, or Ci to Cio aminoalkyl;
R125 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, (=0), or C(0)OH; or
R and R join to form C5 to Cio cycloalkyl optionally substituted by one or more R126; and
R126 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, or Ci to Cio aminoalkyl;
(m) a compound of formula (XIV):
j2
Figure imgf000110_0002
(XIV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R202;
R202 is H, CI to Cio alkyl, halogen, CN, or 02; R200 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally
203 203
substituted by one or more R ;R is halogen, CN, NO2, Ci to Cio alkoxy, or aryl optionally substituted by one or more R204;
R204 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl; (n) a compound of fo
Figure imgf000111_0001
(XV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, or C2 to
R302
Cio alkynyl, each optionally substituted by one or more
R302 is H, Ci to Cio alkyl, halogen, CN, or N02; R300 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R303;
R303 is halogen, CN, NO2, or aryl optionally substituted by one or more
R304;
R304 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl; (0) a compound of formula (XVI):
Figure imgf000111_0002
(XVI)
wherein:
4UU is OH or O;
Figure imgf000111_0003
R is H, Ci to Cio alkyl, halogen, (=0), or Ci to Cio alkoxy;
R405 is absent, Ci to Cio alkyl, heterocycle, aryl, or heteroaryl,
R406
optionally substituted by one or more
R406 is H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, C(0)(Ci t
404. alkyl), aryl, heteroaryl, C(0)(aryl optionally substituted by one or more R RR40 is absent, H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, aryl, or heteroaryl;
with the proviso that when R400 is O and R405 or R407 is absent, then R400 and R402 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404; and
with the proviso that when R401 is OR405 and R403 is N(R406)(R407) and R405 and R407 are absent, R401 and R402, and/or R401 and R403, and/or R402 and R403 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404;
(p) a compound of formula (XVII):
Figure imgf000112_0001
(XVII)
wherein:
R500 is Ci to Cio aminoalkyl;
R502 is benzyl optionally substituted by one or more R503; R5 to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or aryl;
f is 0 to 4; and
R503 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, or aryl;
(q) a compound of formula (XVIII):
Figure imgf000112_0002
(XVIII)
wherein:
R507 is H, OH, Ci to Cio alkoxy, or C3 to Cio cycloalkyl;
R508 to R510 are, independently, H, Ci to do alkyl, Ci to C10 alkoxy, C3 to Cio cycloalkyl, or halogen; and
(r) a compound of
Figure imgf000113_0001
(XIX)
wherein:
R600 is Ci to Cio alkyl, NH2 or Ci to Cio aminoalkyl;
R 602 tQ R 6os are^ mcjepen(jently, Ci to C10 alkyl, C3 to C10 cycloalkyl, Ci to Cio alkoxy, or halogen;
(s) a compound of formula (XX):
Figure imgf000113_0002
(XX)
wherein:
R606 is H or Ci to Cio alkyl;
R607 to R610 are, independently, H, Ci to do alkyl, Ci to do alkoxy, or halogen;
(t) a compound of formula (XXI):
Figure imgf000113_0003
(XXI) wherein:
R7oo tQ R704 ar6j independently, H, Ci to C10 alkyl, C3 to CIO cycloalkyl, C3 to C10 spirocycloalkyl, Q to C10 alkoxy, halogen, Ci to C10 aminoalkyl, or CN; and
R70S is H or Ci to C10 alkyl;
(u) a compound of formula (XXII):
Figure imgf000114_0001
(XXII)
wherein:
R8oo tQ R804 and R806 ^ R809 ^ independently, H, Ci to C10 alkyl, halogen, NH2, or Ci to C10 alkoxy and
805 is H or Ci to C10 alkyl;
Figure imgf000114_0002
(XXIII)
wherein:
RJUU is H or Ci to do alkyl; and
R901 to R905 is H, Ci to Cio alkyl, C2 to C10 alkenyl, C2 to C10 alkynyl, halogen, Ci to C10 alkoxy, Ci to C10 aminoalkyl, or H2; and
(w) a compound of formula (XXIV):
Figure imgf000115_0001
(XXIV)
wherein:
R906 is Ci to Cio alkyl or Ci to Cio alkoxy;
R907 is C2 to Cio alkyl or Ci to Cio alkoxy;
e is 0 to 5;
R908 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, OH, CN, or NH2; and
R909 is H or Ci to Cio alkyl;
or a pharmaceutically acceptable salt or prodrug thereof.
6. The compound according to claim 5, wherein said compound binds to a PF4 dimer or PF4 monomer.
7. A compound for use in disrupting ultra-large complexes (ULCs) comprising a PF4 tetramer and a glycosaminoglycan (GAG) in a subject, wherein said compound is selected from the group consisting of:
(a) a compound of formula (II):
Figure imgf000115_0002
(II)
wherein:
n is 0 to 5;
R1 and R2 are, independently, H, Ci to Cio alkyl, C(0)(Ci to Cio alkyl), C(0)(Ci to Cio alkyl), benzyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl; or
R1 and R2 together comprise a heterocycle or heteroaryl, optionally substituted by one or more R5;
R3 and R4 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, benzyl, aryl, heteroaryl, or heterocycle; or R1 and R3 together comprise a heterocycle or heteroaryl, each optionally substituted by one or more R5;
R5 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, or (=0);
(b) a compound of formula (III):
OR16
I
^(CR17R18)q-R10
(III)
wherein:
q is 0 to 5;
R10 is Ci to Cio alkyl. heteroaryl or aryl, each optionally substituted by one or more R19;
R16 is H or Ci to Cio alkyl;
R17 and R18 are, independently, absent, H, Ci to Cio alkyl, (Ci to Cio alkyl)-P-(Ci to Cio alkyl), Ci to Cio alkoxy, halogen, OH, N02, CN, C2 to Cio alkenyl, P(0)OH, C2 to Cio alkynyl, aryl, heteroaryl, or heterocycle, each optionally substituted by one or more R19;
17 18 17 18 wherein when R or R is absent and q is at least 2, the CR R groups are bound through a stable double bond;
R19 is H, Ci to Cio alkyl, Ci to C10 alkoxy, halogen, N02, CN, C(0)OH, C(0)(Ci to Cio alkyl), C2 to Cio alkenyl, oxo, or C2 to Cio alkynyl;
(c) a compound of formula (IV):
Figure imgf000116_0001
(IV)
wherein:
m is 0 to 2;
R20 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R24; R , R , and R are independently, H, Ci to C10 alkyl, or C3 to C10 cycloalkyl, each optionally substituted by one or more R24;
R24 is H, halogen, OH, CN, N02, d to C10 alkyl C3 to C10 cycloalkyl, C2 to Cio alkenyl, or C2 to C10 alkynyl, each optionally substituted by one or more R27;
R25 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl;
R26 is O or S;
R27 is H, P(0)OH, or Ci to Cio alkyl optionally substituted by halogen or OP(0)OH;
(d) a compound of formula (V):
O II
R41' P^R40
(V)
wherein:
R40 and R41 are, independently, H, Ci to Cio alkyl optionally substituted by one or more R42, aryl optionally substituted by one or more R42, or benzyl optionally substituted by one or more R42;
R42 is H, Ci to Cio alkyl, C(0)OH, NH2, C(0)(d to Cio alkyl), C(0)0(Ci to Cio alkyl), Ci to Cio aminoalkyl, halogen, aryl, or heteroaryl;
(e) a compound of formula (VI):
OR51
0=P— OR50
I
R52
(VI)
wherein:
R50 is heteroaryl or heterocycle, each optionally substituted by one or more R53;
R51 is H, Ci to Cio alkyl, Ci to Cio aminoalkyl, each optionally substituted by halogen or C(0)OH;
R52 is absent, Ci to Cio alkyl optionally substituted by P(0)2, or aryl optionally substituted by 0(Ci to Cio alkyl)-phenyl-C(0)OH; R is Ci to Cio alkyl, halogen, or C(0)OH;
(f) a compound of formula (VII):
Figure imgf000118_0001
(VII)
wherein:
t is 1 to 10;
R60 is aryl, heteroaryl, or heterocycle;
R61 is Ci to Cio aminoalkyl, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, heteroaryl, or heterocycle;
R65 and R66 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl benzyl, aryl, heteroaryl, or heterocycle;
(g) a compound of formula (VIII):
Figure imgf000118_0002
(VIII)
wherein:
R , R , R , and R/5 are, independently, H, Ci to Cio alkyl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, C(0)(Ci to Cio aminoalkyl), or heteroaryl, each optionally substituted by one or more R78;
R72 is H, Ci to Cio alkyl, 0(Ci to Cio alkyl), 0(Ci to Cio alkoxy), 0(Ci to Cio aminoalkyl), Ci to Cio aminoalkyl, or heteroaryl, each optionally substituted by one or more R76;
R73 is Ci to Cio alkyl, Ci to Cio alkoxy, Ci to Cio aminoalkyl, each optionally substituted by one or more R76;
R76 is C(0)OH, C(0)(Ci to Cio alkyl), NHC(0)(Ci to Cio alkyl), C(0)NH(phenyl optionally substituted by one or more R77), C(0)0(Ci to Cio alkyl), heteroaryl optionally substituted by one or more R77, heterocycle optionally substituted by one or more R , or phenyl optionally substituted by one or more R77;
R77 is halogen, Ci to C10 aminoalkyl, Ci to C10 alkoxy, Ci to C10 alkyl, or C2 to Cio alkenyl, each optionally substituted by one or more R78;
R78 is CN, C(0)OH, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), or phenyl optionally substituted by one or more R79;
R79 is OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, halogen, or Ci to Ce alkoxy;
(h) a compound of formula (IX):
Figure imgf000119_0001
(IX)
wherein:
w is 1 to 10;
w' is 1 to 10;
R80 is H or C2 to Cio alkyl;
R81 to R85 are, independently, H, OH, Ci to Cio alkyl, halogen, or Ci to Cio alkoxy; and
R86 to R89 are, independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl;
(i) a compound of formula (X):
Figure imgf000119_0002
(X)
wherein:
x is 2 to 10;
G) a compound of formula (XI):
Figure imgf000120_0001
(XI)
wherein:
y is 1 to 10;
R90 to R94 are, independently, H, halogen, Ci to C10 alkyl, C2 to C10 alkenyl, C2 to C10 alkynyl, OH, or Ci to C10 aminoalkyl; and
R95 is heterocycle, heteroaryl, aryl, or Ci to C10 aminoalkyl;
(k) a compound of formula (XII):
Figure imgf000120_0002
(XII)
wherein:
R105 is aryl, heteroaryl, or heterocycle;
z is 2 to 10;
R106 to R110 are, independently, H, Ci to C10 alkyl, C3 to C10 cycloalkyl, OH, C2 to Cio alkenyl, or C2 to C10 alkynyl;
(1) a compound of formula (XIII):
Figure imgf000120_0003
(XIII)
wherein:
a is 0 or 1;
R120 is absent, aryl, C3 to Cio cycloalkyl, heteroaryl, or heterocycle, each optionally substituted by one or more R125; R is aryl containing one or more R , Ci to C10 aminoalkyl optionally containing one or two aryl groups optionally substituted by one or more R124, heterocycle optionally substituted by one or more R125;
R123 is H, halogen, Ci to C10 alkyl, or Ci to C10 alkoxy;
R124 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, or Ci to Cio aminoalkyl;
R125 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, (=0), or C(0)OH; or
R and R join to form C5 to Cio cycloalkyl optionally substituted by one or more R126; and
R126 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, or Ci to Cio aminoalkyl;
(m) a compound of formula (XIV):
j2
Figure imgf000121_0001
(XIV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R202;
R202 is H, CI to Cio alkyl, halogen, CN, or 02; R200 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally
203 203
substituted by one or more R ;R is halogen, CN, NO2, Ci to Cio alkoxy, or aryl optionally substituted by one or more R204;
R204 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl; (n) a compound of formula (XV):
Figure imgf000121_0002
(XV)
wherein:
R301 is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to C10 alkenyl, or C2 to
R302
Cio alkynyl, each optionally substituted by one or more
R302 is H, Ci to Cio alkyl, halogen, CN, or 02; R300 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally
303.
substituted by one or more R
R is halogen, CN, NO2, or aryl optionally substituted by one or more
R 304.
R304 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl; (0) a compound of formula (XVI):
Figure imgf000122_0001
(XVI)
wherein:
R400 is OH or O;
R401, R402, and R403, are independently, OR405 or N(R406)(R407);
R404 is H, Ci to Cio alkyl, halogen, (=0), or Ci to Cio alkoxy;
R405 is absent, Ci to Cio alkyl, heterocycle, aryl, or heteroaryl,
R406
optionally substituted by one or more
R406 is H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, C(0)(Ci to Cio alkyl), aryl, heteroaryl, C(0)(aryl optionally substituted by one or more R404
R407
R is absent, H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, aryl, or heteroaryl;
with the proviso that when R400 is O and R405 or R407 is absent, then R400 and R402 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404; and
with the proviso that when R401 is OR405 and R403 is N(R406)(R407) and R405 and R407 are absent, R401 and R402, and/or R401 and R403, and/or R402 and R join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404;
(p) a compound of formula (XVII):
Figure imgf000123_0001
(XVII)
wherein:
R500 is Ci to Cio aminoalkyl;
R502 is benzyl optionally substituted by one or more R503; R5 to Cio alkyl, C3 to C10 cycloalkyl, Ci to C10 alkoxy, or aryl;
f is 0 to 4; and
R503 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, or aryl;
(q) a compound of formula (XVIII):
Figure imgf000123_0002
(XVIII)
wherein:
R507 is H, OH, Ci to Cio alkoxy, or C3 to Cio cycloalkyl;
R508 to R510 are, independently, H, Ci to Cio alkyl, Ci to C10 alkoxy, C3 to Cio cycloalkyl, or halogen; and
(r) a compound of formula (XIX):
Figure imgf000123_0003
(XIX)
wherein:
R600 is Ci to Cio alkyl, NH2 or Ci to Cio aminoalkyl; R to R are, independently, Ci to Cio alkyl, C3 to Cio cycloalkyl, Cio alkoxy, or halogen;
Figure imgf000124_0001
(XX)
wherein:
R606 is H or Ci to Cio alkyl;
R607 to R610 are, independently, H, Ci to Cio alkyl, Ci to Cio alkoxy, or halogen;
(t) a compound of formula (XXI):
Figure imgf000124_0002
(XXI)
wherein:
R7oo tQ R704 are^ independentlyj Hj Cl t0 Cl0 aiky^ C3 to CIO cycloalkyl, C3 to Cio spirocycloalkyl, Ci to Cio alkoxy, halogen, d to C io aminoalkyl, or CN; and
R705 is H or Ci to Cio alkyl;
(u) a compound of formula (XXII):
Figure imgf000124_0003
(XXII)
wherein:
R 8oo tQ R 804 and R 806 and R 809 ^ independently5 H, C[ to C10 alkyl, halogen, NH2, or Ci to Cio alkoxy and
R80S is H or Ci to Cio alkyl;
(v) a compound of formula (XXIII):
Figure imgf000125_0001
wherein:
R900 is H or Ci to Cio alkyl; and
R9oi tQ R905 is Hj Ci tQ alkyli C2 10 alkenyl; Cl t0 C l0 aikynyi, halogen, Ci to Cio alkoxy, Ci to Cio aminoalkyl, or H2; and
(w) a compound of formula (XXIV):
Figure imgf000125_0002
wherein:
R906 is Ci to Cio alkyl or Ci to Cio alkoxy;
R907 is C2 to Cio alkyl or Ci to C10 alkoxy;
e is 0 to 5;
R908 is H, halogen, d to C io alkyl, Ci to Cio alkoxy, OH, CN, or NH2; and
R909 is H or Ci to Cio alkyl;
or a pharmaceutically acceptable salt or prodrug thereof.
8. The compound according to claim 7, wherein said compound disrupts the salt bridge of said PF4 tetramer.
9. The compound according to claim 5 or 7, wherein said GAG is heparin.
10. The compound according to claim 5 or 7, wherein said ULC is greater than about 600 kD.
11. A compound for use in preventing or treating heparin induced
thrombocytopenia and thrombosis (HITT) in a subject, wherein said compound is selected from the group consisting of:
(a) a compound of formula (II):
Figure imgf000126_0001
(II)
wherein:
n is 0 to 5;
R1 and R2 are, independently, H, Ci to C10 alkyl, C(0)(C1 to C10 alkyl), C(0)(Ci to Cio alkyl), benzyl, C3 to C10 cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl; or
R1 and R2 together comprise a heterocycle or heteroaryl, optionally substituted by one or more R5;
R3 and R4 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, benzyl, aryl, heteroaryl, or heterocycle; or
R1 and R3 together comprise a heterocycle or heteroaryl, each optionally substituted by one or more R5;
R5 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, or (=0);
(b) a compound of formula (III):
OR16
I
0^P (CR17R18)q-R10
(HI)
wherein:
q is 0 to 5;
R10 is Ci to Cio alkyl. heteroaryl or aryl, each optionally substituted by one or more R19; Rlb is H or Ci to C10 alkyl;
R17 and R18 are, independently, absent, H, Ci to C10 alkyl, (Ci to C10 alkyl)-P-(Ci to C10 alkyl), Ci to C10 alkoxy, halogen, OH, N02, CN, C2 to C10 alkenyl, P(0)OH, C2 to C10 alkynyl, aryl, heteroaryl, or heterocycle, each optionally substituted by one or more R19;
17 18 17 18 wherein when R or R is absent and q is at least 2, the CR R groups are bound through a stable double bond;
R19 is H, Ci to Cio alkyl, Ci to C10 alkoxy, halogen, N02, CN, C(0)OH, C(0)(Ci to Cio alkyl), C2 to Cio alkenyl, oxo, or C2 to Cio alkynyl; (c) a compound of formula (IV):
Figure imgf000127_0001
(IV)
wherein:
m is 0 to 2;
R20 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R24;
R21, R22, and R23 are independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl, each optionally substituted by one or more R24;
R24 is H, halogen, OH, CN, N02, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R27;
R25 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl;
R26 is O or S;
R27 is H, P(0)OH, or Ci to Cio alkyl optionally substituted by halogen or OP(0)OH;
(d) a compound of formula (V):
O II
- P. (V)
wherein:
R40 and R41 are, independently, H, Ci to C10 alkyl optionally substituted by one or more R42, aryl optionally substituted by one or more R42, or benzyl optionally substituted by one or more R42;
R42 is H, Ci to Cio alkyl, C(0)OH, NH2, C(0)(Ci to C10 alkyl), C(0)0(Ci to Cio alkyl), Ci to Cio aminoalkyl, halogen, aryl, or heteroaryl;
(e) a compound of formula (VI):
OR51
0=P— OR50
I
R52
(VI)
wherein:
R50 is heteroaryl or heterocycle, each optionally substituted by one or more R53;
R51 is H, Ci to Cio alkyl, Ci to Cio aminoalkyl, each optionally substituted by halogen or C(0)OH;
R52 is absent, Ci to Cio alkyl optionally substituted by P(0)2, or aryl optionally substituted by 0(Ci to Cio alkyl)-phenyl-C(0)OH;
R53 is Ci to Cio alkyl, halogen, or C(0)OH;
(f) a compound of formula (VII):
P— (CR65R66)tR60R61
HO
(VII)
wherein:
t is 1 to 10;
R60 is aryl, heteroaryl, or heterocycle;
R61 is Ci to Cio aminoalkyl, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, heteroaryl, or heterocycle;
R65 and R66 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl benzyl, aryl, heteroaryl, or heterocycle; (g) a compound of fo
Figure imgf000129_0001
(VIII)
wherein:
R , R , R , and R s are, independently, H, Ci to C10 alkyl, C2 to C10 alkenyl, Ci to C10 aminoalkyl, C(0)(Ci to C10 aminoalkyl), or heteroaryl, each optionally substituted by one or more R78:
R72 is H, Ci to Cio alkyl, 0(d to C10 alkyl), 0(d to C10 alkoxy), 0(d to Cio aminoalkyl), Ci to C10 aminoalkyl, or heteroaryl, each optionally substituted by one or more R76;
R73 is Ci to Cio alkyl, Ci to Cio alkoxy, Ci to Cio aminoalkyl, each optionally substituted by one or more R76:
R76 is C(0)OH, C(0)(Ci to Cio alkyl), NHC(0)(Ci to Cio alkyl), C(0)NH(phenyl optionally substituted by one or more R77), C(0)0(C1 to Cio alkyl), heteroaryl optionally substituted by one or more R77, heterocycle optionally substituted by one or more R77, or phenyl optionally substituted by one or more R77;
R77 is halogen, Ci to Cio aminoalkyl, Ci to Cio alkoxy, Ci to Cio alkyl, or C2 to Cio alkenyl, each optionally substituted by one or more R78;
R78 is CN, C(0)OH, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), or phenyl optionally substituted by one or more R79;
R79 is OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, halogen, or Ci to Ce alkoxy;
(h) a compound of formula (IX):
Figure imgf000129_0002
(IX)
wherein: w is 1 to 10;
w' is 1 to 10;
R80 is H or C2 to Cio alkyl;
R81 to R85 are, independently, H, OH, Ci to Cio alkyl, halogen, Cio alkoxy; and
R86 to R89 are, independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl;
(i) a compound of formula (X):
Figure imgf000130_0001
(X)
wherein:
x is 2 to 10;
G) a compound of formula (XI):
Figure imgf000130_0002
(XI)
wherein:
y is 1 to 10;
R90 to R94 are, independently, H, halogen, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, OH, or Ci to Cio aminoalkyl; and
R95 is heterocycle, heteroaryl, aryl, or Ci to Cio aminoalkyl;
(k) a compound of formula (XII):
Figure imgf000130_0003
(XII) wherein:
R105 is aryl, heteroaryl, or heterocycle;
z is 2 to 10;
R106 to R110 are, independently, H, Ci to C10 alkyl, C3 to C10 cycloalkyl, OH, C2 to Cio alkenyl, or C2 to C10 alkynyl;
(1) a compound of formula (XIII):
Figure imgf000131_0001
(XIII)
wherein:
a is 0 or 1;
R120 is absent, aryl, C3 to C10 cycloalkyl, heteroaryl, or heterocycle, each optionally substituted by one or more R125;
R121 is aryl containing one or more R123, Ci to Cio aminoalkyl optionally containing one or two aryl groups optionally substituted by one or more R124, heterocycle optionally substituted by one or more R125;
R123 is H, halogen, Ci to Cio alkyl, or Ci to Cio alkoxy;
R124 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, or Ci to Cio aminoalkyl;
R125 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, (=0), or C(0)OH; or
R120 and R121 join to form C5 to Cio cycloalkyl optionally substituted by one or more R126; and
R126 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, or Ci to Cio aminoalkyl;
(m) a compound of formula (XIV):
Figure imgf000131_0002
(XIV) wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R202;
R202 is H, CI to Cio alkyl, halogen, CN, or 02; R200 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R 203 ;R 203 is halogen, CN, NO2, Ci to Cio alkoxy, or aryl optionally substituted by one or more R204;
R204 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl; (n) a compound of fo
Figure imgf000132_0001
(XV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R302
R302 is H, Ci to Cio alkyl, halogen, CN, or N02; R300 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R303;
R303 is halogen, CN, NO2, or aryl optionally substituted by one or more
R304;
R304 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl; (0) a compound of formula (XVI):
Figure imgf000132_0002
(XVI)
wherei
Figure imgf000132_0003
R405 is absent, Ci to C10 alkyl, heterocycle, aryl, or heteroaryl,
R406
optionally substituted by one or more
R406 is H, Ci to Cio alkyl, halogen, Ci to C10 alkoxy, C(0)(Ci to C10 alkyl), aryl, heteroaryl, C(0)(aryl optionally substituted by one or more R404
R407 is absent, H, Ci to C10 alkyl, halogen, Ci to C10 alkoxy, aryl, or heteroaryl;
with the proviso that when R400 is O and R405 or R407 is absent, then R400 and R402 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404; and
with the proviso that when R401 is OR405 and R403 is N(R406)(R407) and R405 and R407 are absent, R401 and R402, and/or R401 and R403, and/or R402 and R403 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R"
(p) a compound of formula (XVII):
Figure imgf000133_0001
(XVII)
wherein:
R500 is Ci to Cio aminoalkyl;
R502 is benzyl optionally substituted by one or more R503; R5 to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or aryl;
f is 0 to 4; and
R503 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, or aryl;
(q) a compound of formula (XVIII):
Figure imgf000133_0002
wherein:
R is H, OH, Ci to Cio alkoxy, or C3 to C10 cycloalkyl;
R508 tQ R5io are^ independently, H, Ci to Cio alkyl, Ci to Cio alkoxy, C3 to Cio cycloalkyl, or halogen; and
(r) a compound of formula (XIX):
Figure imgf000134_0001
(XIX)
wherein:
R is Ci to Cio alkyl, NH? or Ci to Cio aminoalkyl;
R602 to R605 are, independently, Ci to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or halogen;
(s) a compound of formula (XX):
Figure imgf000134_0002
(XX)
wherein:
R6Ub is H or Ci to Cio alkyl;
R607 to R610 are, independently, H, d to Cio alkyl, d to Cio alkoxy, or halogen;
(t) a compound of formula (XXI):
Figure imgf000134_0003
(XXI)
wherein: R /uu tQ R/u4 are^ independentlyj Hj Cl t0 Cl0 alkylj C3 to CIO cycloalkyl, C3 to C10 spirocycloalkyl, Ci to C10 alkoxy, halogen, Ci to C10 aminoalkyl, or CN; and
R705 is H or Ci to do alkyl;
(u) a compound of
Figure imgf000135_0001
(XXII)
wherein:
R8oo tQ R804 and R806 and R809 ^ independently5 H, Ci to C10 alkyl, halogen, NH2, or Ci to C10 alkoxy and
R80S is H or Ci to C10 alkyl;
(v) a compound of formula (XXIII):
Figure imgf000135_0002
(XXIII)
wherein:
R900 is H or Ci to do alkyl; and
R9oi to R905 is H^ C i tQ alkyl) C2 10 alkenyl) Cl t0 Cl0 alkynyl^ halogen, Ci to Cio alkoxy, Ci to Cio aminoalkyl, or H2; and
(w) a compound of formula (XXIV):
Figure imgf000136_0001
(XXIV)
wherein:
R906 is Ci to Cio alkyl or Ci to C10 alkoxy;
R907 is C2 to Cio alkyl or Ci to C10 alkoxy;
e is 0 to 5;
R908 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, OH, CN, or H2; and
R909 is H or Ci to Cio alkyl;
or a pharmaceutically acceptable salt or prodrug thereof.
12. A compound for use in preventing atherosclerosis in a subject, wherein said compound binds to PF4 dimers or PF4 monomers or disrupts the salt bridge of PF4 tetramers.
13. The compound according to claim 12, wherein said atherosclerosis results from PF4 tetramer formation.
14. The compound according to claim 12, wherein the formation of GAG-PF4 complex results in said atherosclerosis.
15. A compound for use in correcting a platelet imbalance in a subject, wherein said compound binds to PF4 dimers or PF4 monomers or disrupts the salt bridge of PF4 tetramers.
16. The compound according to claim 15, wherein said platelet imbalance results from said PF4 tetramer formation.
17. The compound according to claim 15, wherein said platelet imbalance results from heparin administration to said subject.
18. The compound according to any one of claims 15 to 17, wherein platelet production is stimulated.
19. A compound for use in preventing a decrease in platelet production in a subject, wherein said compound binds to PF4 dimers or PF4 monomers or disrupts the salt bridge of PF4 tetramers.
20. The compound according to claim 19, wherein said decrease in platelets results from PF4 tetramer formation.
21. The compound according to claim 19, wherein said decrease in platelets results from heparin administration to said subject.
22. A compound for use in preventing a decrease of or increasing high density lipoproteins in a subject, wherein said compound binds to PF4 dimers or PF4 monomers or disrupts the salt bridge of PF4 tetramers.
23. A compound for use in preventing or treating inflammation in a subject, wherein said compound binds to PF4 dimers or PF4 monomers or disrupts the salt bridge of PF4 tetramers.
24. The compound according to claim 23, wherein said inflammation is caused by NF-KB activation.
25. A compound for use in modulating clotting or hemostasis in a subject, wherein said compound binds to PF4 dimers or PF4 monomers or disrupts the salt bridge of PF4 tetramers.
26. The compound according to claim 25, wherein protein C is upregulated.
27. The compound according to claim 25, wherein protein C is downregulated.
28. The compound according to any one of claims 1 to 27, wherein said PF4 is wild-type PF4.
29. The compound according to any one of claims 1 to 27, wherein said compound binds to Lys50 in said monomer or dimer, Glu28 in said monomer or dimer, or a combination thereof.
30. The compound according to claim 29, wherein said salt bridge comprises Glul28, Lys350 of a first PF4 monomer and Glu328, Lysl50 of a second PF4 monomer.
31. The compound according to claim 29, wherein said salt bridge comprises Glu228, Lys450 of a first PF4 monomer and Glu428, Lys250 of a second PF4 monomer.
32. The compound according to any one of claims 12 to 27, wherein said compound has structure (II):
Figure imgf000138_0001
(II)
wherein:
n is 0 to 5;
R1 and R2 are, independently, H, Ci to C10 alkyl, C(0)(C1 to C10 alkyl),
C(0)(Ci to Cio alkyl), benzyl, C3 to C10 cycloalkyl, C2 to C10 alkenyl, or C2 to C10 alkynyl; or
R1 and R2 together comprise a heterocycle or heteroaryl, optionally substituted by one or more R5;
R3 and R4 are, independently, H, OH, Ci to C10 alkyl, C3 to C10 cycloalkyl, benzyl, aryl, heteroaryl, or heterocycle; or
R1 and R3 together comprise a heterocycle or heteroaryl, each optionally substituted by one or more R5;
R5 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, or (=0); or a pharmaceutically acceptable salt or prodrug thereof.
33. The compound according to any one of claims 1 to 27, wherein said compound is selected from the group consisting of:
Figure imgf000138_0002
0r a pharmaceutically acceptable salt or prodrug thereof.
34. The compound according to any one of claims 1 to 27, wherein said compound is selected from the group consisting of:
Figure imgf000138_0003
, or a pharmaceutically acceptable salt or prodrug thereof.
35. The compound according to any one of claims 12 to 27, wherein said compound has structure (III):
OR16
I
^(CR17R18)q-R10
(III)
wherein:
q is 0 to 5;
R10 is Ci to Cio alkyl. heteroaryl or aryl, each optionally substituted by one or more R19;
R16 is H or Ci to Cio alkyl;
R17 and R18 are, independently, absent, H, Ci to Cio alkyl, (Ci to Cio alkyl)-P- (Ci to Cio alkyl), Ci to C10 alkoxy, halogen, OH, N02, CN, C2 to C10 alkenyl, P(0)OH, C2 to Cio alkynyl, aryl, heteroaryl, or heterocycle, each optionally substituted by one or more R19;
wherein when R17 or R18 is absent and q is at least 2, the CR17R18 groups are bound through a stable double bond;
R19 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, N02, CN, C(0)OH, C(0)(Ci to Cio alkyl), C2 to Cio alkenyl, or C2 to Cio alkynyl; or a pharmaceutically acceptable salt or prodrug thereof.
36. The compound according to any one of claims 1 to 27, wherein said compound is:
Figure imgf000139_0001
(Ilia)
wherein:
each R19 is, independently, H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, NO2, CN, C(0)OH, C(0)(Ci to Cio alkyl), C2 to Cio alkenyl, oxo, or C2 to Cio alkynyl; R17 and R18 are, independently, absent, H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, N02, CN, C2 to Cio alkenyl, P(0)OH, C2 to C10 alkynyl, aryl, heteroaryl, or heterocycle;
wherein when R17 or R18 is absent and q is at least 2, the CR17R18 groups are bound through a stable double bond;
or a pharmaceutically acceptable salt or prodrug thereof.
37. The compound according to claim any one of claims 1 to 27, wherein said compound is:
Figure imgf000140_0001
(Hid).
38. The compound according to any one of claims 1 to 27, wherein said compound is selected from the group consisting of:
Figure imgf000140_0002
or a pharmaceutically acceptable salt or prodrug thereof.
39. The compound according to any one of claims 1 to 27, wherein said compound is:
Figure imgf000140_0003
(Illf)
wherein:
r is 1 to 3;
X is O, S, or R30;
R30 is H or Ci to C10 alkyl;
R31 and R32 are, independently, H, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, benzyl, aryl, heteroaryl, or heterocycle;
or a pharmaceutically acceptable salt or prodrug thereof. The compound according to any one of claims 12 to 27, wherein said
Figure imgf000141_0001
or a pharmaceutically acceptable salt or prodrug thereof.
41. The compound according to any one of claims 1 to 27, wherein said compound is selected from the group consisting of:
Figure imgf000141_0002
or pharmaceutically acceptable salt or prodrug thereof.
42. The compound according to any one of claims 12 to 27, wherein said compound has structure (IV):
Figure imgf000141_0003
(IV)
wherein:
m is 0 to 2; R is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R24:
R21, R22, and R23 are independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl, each optionally substituted by one or more R24:
R24 is H, halogen, OH, CN, N02, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R27;
R25 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl;
R26 is O or S;
R27 is H, P(0)OH, or Ci to Cio alkyl optionally substituted by halogen or OP(0)OH; or a pharmaceutically acceptable salt or prodrug thereof.
43. The compound according to any one of claims 1 to 27, wherein said compound is:
Figure imgf000142_0001
(IVa).
44. The compound according to any one of claims 1 to 27, wherein said compound is selected from the group consisting of:
Figure imgf000142_0002
, and or a pharmaceutically acceptable salt or prodrug thereof.
45. The compound according to any one of claims 12 to 27, wherein said compound has structure (V):
O II
R41 - P^ R40 (V)
wherein:
R40 and R41 are, independently, H, Ci to C10 alkyl optionally substituted by one or more R42, aryl optionally substituted by one or more R42, or benzyl optionally substituted by one or more R42:
R42 is H, Ci to Cio alkyl, C(0)OH, NH2, C(0)(Ci to C10 alkyl), C(0)0(Ci to Cio alkyl), Ci to Cio aminoalkyl, halogen, aryl, or heteroaryl or a pharmaceutically acceptable salt or prodrug thereof.
46. The compound according to any one of claims 1 to 27, wherein said
Figure imgf000143_0001
47. The compound according to any one of claims 12 to 27, wherein said compound has structure (VI):
OR51
0=P— OR50
I
R52
(VI)
wherein:
R50 is aryl, heteroaryl= or heterocycle, each optionally substituted by one or more R ;
R51 is H, Ci to Cio alkyl, Ci to Cio aminoalkyl, each optionally substituted by halogen or C(0)OH; R is absent, Ci to C10 alkyl optionally substituted by P(0)2, or aryl optionally substituted by 0(Ci to C10 alkyl)-phenyl-C(0)OH;
R53 is Ci to Cio alkyl, halogen, or C(0)OH; or a pharmaceutically acceptable salt or prodrug thereof.
48. The compound according to any one of claims 12 to 27, wherein said compound is selected from the group consisting of:
Figure imgf000144_0001
or a pharmaceutically acceptable salt or prodrug thereof.
49. The compound according to claims 1 to 27, wherein said compound is
o— P— o
II
o or a pharmaceutically acceptable salt of prodrug thereof.
50. The compound according to any one of claims 12 to 27, wherein said compound has structure (VIII):
Figure imgf000144_0002
(VIII)
wherein: R , R , R , and R are, independently, H, Ci to C10 alkyl, C2 to C10 alkenyl, Ci to Cio aminoalkyl, C(0)(Ci to Cio aminoalkyl), or heteroaryl, each optionally substituted by one or more R78:
R72 is H, Ci to Cio alkyl, 0(d to Cio alkyl), 0(d to Cio alkoxy), 0(d to Cio aminoalkyl), Ci to Cio aminoalkyl, or heteroaryl, each optionally substituted by one or more R ;
R73 is Ci to Cio alkyl, Ci to Cio alkoxy, Ci to Cio aminoalkyl, each optionally substituted by one or more R76; R/b is C(0)OH, C(0)(Ci to C10 alkyl), NHC(0)(Ci to C10 alkyl),
C(0)NH(phenyl optionally substituted by one or more R77), C(0)0(C1 to C10 alkyl), heteroaryl optionally substituted by one or more R77, heterocycle optionally substituted by one or more R77, or phenyl optionally substituted by one or more R77;
R77 is halogen, Ci to Cio aminoalkyl, Ci to Cio alkoxy, Ci to Cio alkyl, or C2 to Cio alkenyl, each optionally substituted by one or more R78:
R78 is CN, C(0)OH, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), C(S)S(d to Cio alkyl)C(0)OH, or phenyl optionally substituted by one or more
R79 is OH, Ci to Cio alkyl, C3 to C10 cycloalkyl, C2 to Cio alkenyl, C2 to C10 alkynyl, halogen, or Ci to Ce alkoxy; or a pharmaceutically acceptable salt or prodrug thereof.
51. The compound according to any one of claims 1 to 27, wherein said compound is selected from the group consisting of:
Figure imgf000145_0001
Figure imgf000146_0001
pharmaceutically acceptable salt or prodrug thereof.
52. The compound according to any one of claims 12 to 27, wherein said
compound is
Figure imgf000146_0002
or a pharmaceutically acceptable salt or prodrug thereof.
53. The compound according to any one of claims 12 to 27, wherein said compound has structure (IX):
Figure imgf000147_0001
(IX)
wherein:
w is 1 to 10;
w' is 1 to 10;
R80 is H or Ci to C10 alkyl;
R81 to R85 are, independently, H, OH, Ci to Cio alkyl, halogen, or Ci to Cio alkoxy; and
R86 to R89 are, independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl; or a pharmaceutically acceptable salt or prodrug thereof.
54. The compound according to any one of claims 12 to 27, wherein said compound has structure (X):
Figure imgf000147_0002
(X)
wherein:
x is 1 to 10; or a pharmaceutically acceptable salt or prodrug thereof.
55. The compound according to any one of claims 12 to 27, wherein said compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
O
„OH
HO' >r ^s'
= II
NH2 O
56. The compound according to any one of claims 12 to 27, wherein said compound has structure (XI):
Figure imgf000147_0003
(XI)
wherein:
y is 1 to 10;
R90 to R94 are, independently, H, halogen, Ci to C10 alkyl, C2 to C10 alkenyl, C2 to Cio alkynyl, OH, or Ci to C10 aminoalkyl; and
R95 is heterocycle, heteroaryl, aryl, or Ci to C10 aminoalkyl;
or a pharmaceutically acceptable salt or prodrug thereof.
57. The compound according to any one of claims 12 to 27, wherein said compound has structure (XII):
Figure imgf000148_0001
(XII)
wherein:
R105 is aryl, heteroaryl, or heterocycle;
z is 1 to 10;
R io6 tQ R iio ar6j independently, H, Ci to C10 alkyl, C3 to C10 cycloalkyl, OH, C2 to Cio alkenyl, or C2 to C10 alkynyl;
or a pharmaceutically acceptable salt or prodrug thereof.
58. The compound according to any one of claims 12 to 27, wherein said compound is the following, or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000148_0002
59. The compound according to any one of claims 12 to 27, wherein said compound has structure (XIII):
Figure imgf000149_0001
(XIII)
wherein:
a is 0 or 1 ;
R120 is absent, aryl, C3 to C10 cycloalkyl, heteroaryl, or heterocycle, each optionally substituted by one or more R125;
R121 is aryl containing one or more R123, Ci to C10 aminoalkyl optionally containing one or two aryl groups optionally substituted by one or more R124, heterocycle optionally substituted by one or more R125;
R123 is H, halogen, Ci to C10 alkyl, or Ci to C10 alkoxy;
R124 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, or Ci to C10 aminoalkyl;
R125 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, (=0), or C(0)OH; or
R120 and R121 join to form C5 to C10 cycloalkyl optionally substituted by one or more R126; and
R126 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, or Ci to C10 aminoalkyl; or a pharmaceutically acceptable salt or prodrug thereof.
60. The compound according to any one of claims 1 to 27, wherein said compound has structure (XHIa), wherein R121 is a heterocycle having one SO2 in the heterocycle backbone:
O II
^S-OR 121
O"
(XHIa).
61. The compound according to any one of claims 1 to 27, wherein said compound is selected from the group consisting of:
pharmaceutically acceptable salt or prodrug thereof.
62. The compound according to any one of claims 12 to 27, wherein said compound structure (XIV):
Figure imgf000150_0002
(XIV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to C10 alkenyl, or C2 to C10 alkynyl, each optionally substituted by one or more R202;
R202 is H, CI to Cio alkyl, halogen, CN, or 02; R200 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R ;R is halogen, CN, NO2, Ci to Cio alkoxy, or aryl optionally substituted by one or more R204;
R204 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl;
or a pharmaceutically acceptable salt or prodrug thereof.
63. The compound according to any one of claims 1 to 27, wherein said compound is selected from the group consisting of:
Figure imgf000150_0003
Figure imgf000151_0001
thereof.
64. The compound according to any one of claims 12 to 27, wherein said compound is:
Figure imgf000151_0002
(XV)
wherein:
R301 is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to C10 alkenyl, or C2 to C10 alkynyl, each optionally substituted by one or more R302;
R302 is H, CI to Cio alkyl, halogen, CN, or 02; R300 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R303;
R303 is halogen, CN, NO2, or aryl optionally substituted by one or more R304:
R304 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl;
or a pharmaceutically acceptable salt or prodrug thereof.
65. The compound according to any one of claims 1 to 27, wherein said compound is selected from the group consisting of:
Figure imgf000152_0001
pharmaceutically acceptable salt or prodrug thereof
66. The compound according to any one of claims 12 to 27, wherein said compound has structure (XVI):
Figure imgf000152_0002
(XVI)
wherein:
R400 is OH or O;
R401, R402, and R403, are independently, OR405 or N(R406)(R407);
R404 is H, Ci to Cio alkyl, halogen, (=0), or Ci to C10 alkoxy;
R405 is aabbsseenntt,, CCii ttoo C Cio alkyl, heterocycle, aryl, or heteroaryl, optionally
.406.
substituted by one or more R
RR440U6b iiss HH,, CCii ttoo CCiioo aallkyl, halogen, Ci to Cio alkoxy, C(0)(Ci to Cio alkyl),
404.
aryl, heteroaryl, C(0)(aryl optionally substituted by one or more R
RR40 is absent, H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, aryl, or heteroaryl;
with the proviso that when R4lJlJ is O and R4U5 or R4lJ / is absent, then R4lJlJ and R402 join to form a 5 to 8-membered ring, wherein the 5 to 8-membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8- membered ring is optionally substituted by one or more R404; and
with the proviso that when R401 is OR405 and R403 is N(R406)(R407) and R405 and R407 are absent, R401 and R402, and/or R401 and R403, and/or R402 and R403 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404; or a pharmaceutically acceptable salt or prodrug thereof.
67. The compound according to any one of claims 1 to 27, wherein said
Figure imgf000153_0001
68. The compound according to any one of claims 12 to 27, wherein said compound is:
Figure imgf000153_0002
(XVII)
wherein:
R is Ci to Cio aminoalkyl;
R502 is benzyl optionally substituted by one or more R503; R501 is H, Ci to C10 alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or aryl;
f is 0 to 4; and
R503 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, or aryl;
or a pharmaceutically acceptable salt or prodrug thereof.
69. The compound according to any one of claims 1 to 27, wherein said compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000153_0003
70. The compound according to any one of claims 12 to 27, wherein said compound has structure (XVIII)
Figure imgf000154_0001
(XVIII)
wherein:
R507 is H, OH, Ci to Cio alkyl, Ci to C10 alkoxy, or C3 to C10 cycloalkyl;
R508 to R510 are, independently, H, Ci to C10 alkyl, Ci to C10 alkoxy, C3 to C10 cycloalkyl, or halogen;
or a pharmaceutically acceptable salt or prodrug thereof.
71. The compound according to any one of claims 12 to 27, wherein said compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000154_0002
72. The compound according to any one of claims 12 to 27, wherein said compound has structure (XIX)
Figure imgf000154_0003
wherein:
R600 is Ci to Cio alkyl, NH2 or Ci to C10 aminoalkyl;
R602 to R605 are, independently, Ci to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or halogen;
or a pharmaceutically acceptable salt or prodrug thereof.
73. The compound according to any one of claims 1 to 27, wherein said compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000155_0001
74. The compound according to any one of claims 12 to 27, wherein said compound has structure (XX):
Figure imgf000155_0002
(XX)
wherein:
R606 is H or Ci to C10 alkyl;
R607 to R610 are, independently, H, Ci to C10 alkyl, Ci to C10 alkoxy, or halogen;
or a pharmaceutically acceptable salt or prodrug thereof.
75. The compound according to any one of claims 1 to 27, wherein said compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000155_0003
76. The compound according to any one of claims 12 to 27, wherein said compound has structure (XXI):
Figure imgf000155_0004
(XXI)
wherein: R uu to R are, independently, H, Ci to C10 alkyl, C3 to C10 cycloalkyl, C3 to Cio spirocycloalkyl, Ci to Cio alkoxy, halogen, Ci to Cio aminoalkyl, or CN; and R705 is H or Ci to Cio alkyl;
or a pharmaceutically acceptable salt or prodrug thereof.
77. The compound according to any one of claims 1 to 27, wherein said compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000156_0001
78. The compound according to any one of claims 12 to 27, wherein said compound has structure (XXII):
Figure imgf000156_0002
(XXII)
wherein:
R8uu tQ R8U4 and R8ub and R8uy ^ independentlyj Hj Cl to Cl0 alkylj halogen,
NH2, or Ci to Cio alkoxy and
R805 is H or Ci to Cio alkyl;
or a pharmaceutically acceptable salt or prodrug thereof.
79. The compound according to any one of claims 1 to 27, wherein said compound is the following or a ph eptable salt or prodrug thereof:
Figure imgf000156_0003
80. The compound according to any one of claims 12 to 27, wherein said compound has structure (XVI
Figure imgf000157_0001
wherein:
R900 is H or Ci to C10 alkyl; and
R901 to R905 is H, Ci to Cio alkyl, C2 to C10 alkenyl, C2 to C10 alkynyl, halogen, Ci to Cio alkoxy, Ci to Cio aminoalkyl, or NH2;
or a pharmaceutically acceptable salt or prodrug thereof.
81. The compound according to any one of claims 1 to 27, wherein said compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000157_0002
82. The compound according to any one of claims 12 to 26, wherein said compound has structure (XXIV):
Figure imgf000157_0003
R and R are, independently, Ci to Cio alkyl or Ci to Cio alkoxy;
e is 0 to 5; and
R908 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, OH, CN, or NH2 R909 is H or Ci to Cio alkyl; or a pharmaceutically acceptable salt or prodrug thereof.
83. The compound according to any one of claims 12 to 27, wherein said compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000158_0001
84. The compound according to any one of claims 1 to 27, wherein said compound is the following or a pharmaceutically acceptable salt or prodrug thereof:
Figure imgf000158_0002
85. The compound according to any one of claims 1 to 84, wherein the pharmaceutically acceptable salt is of a base.
86. The compound according to claim 85, wherein said base is selected from the group consisting of sodium hydroxide, lithium hydroxide, potassium hydroxide, and mixtures thereof.
87. The compound according to any one of claims 1 to 85, wherein the pharmaceutically acceptable salt is of an acid.
88. The compound according to claim 87, wherein said acid is selected from the group consisting of acetic acid, propionic acid, lactic acid, citric acid, tartaric acid, succinic acid, fumaric acid, maleic acid, malonic acid, mandelic acid, malic acid, phthalic acid, hydrochloric acid, hydrobromic acid, phosphoric acid, nitric acid, sulfuric acid, methanesulfonic acid, napthalenesulfonic acid, benzenesulfonic acid, toluenesulfonic acid, and camphorsulfonic acid.
89. The compound according to any one of claims 1 to 84, wherein said compound is delivered to said subject orally, ocularly, by injection, by inhalation, transdermally or by suppository.
90. The compound according to any one of claims 3 or 7 to 89, further comprising: (a) measuring a first level of PF4 tetramer in a first biological sample obtained from said subject; and
(b) administering a first effective amount of said compound required to decrease said PF4 tetramer level.
91. The compound according to claim 90, further comprising:
(c) measuring a second level of PF4 tetramer in a second biological sample obtained from said subject; and
(d) administering a second effective amount of said compound required to decrease said PF4 tetramer level.
92. The compound according to any one of claims 1, 5, or 7 to 89, further comprising:
(a) measuring a first level of PF4 tetramer in a first biological sample obtained from said subject; and
(b) administering a first effective amount of said compound required to prevent formation of said PF4 tetramer.
93. The compound according to claim 92, further comprising:
(c) measuring a second level of PF4 tetramer in a second biological sample obtained from said subject; and
(d) administering a second effective amount of said compound required to prevent formation of said PF4 tetramer.
94. The compound according to any one of claims 90 to 93, wherein said effective amount is about 500 mg/kg or lower.
95. The compound according to any one of claims 1 to 94, wherein said subject has cancer.
96. A composition comprising:
(i) a medication which causes the formation of PF4 tetramers, a medication which disrupts PF4 tetramers, or a combination thereof; and
(ii) a compound selected from the group consisting of:
(a) a compound of formula (II):
Figure imgf000160_0001
(II)
wherein:
n is 0 to 5;
R1 and R2 are, independently, H, Ci to C10 alkyl, C(0)(C1 to Cio alkyl), C(0)(C1 to C10 alkyl), benzyl, C3 to C10 cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl; or
R1 and R2 together comprise a heterocycle or heteroaryl, optionally substituted by one or more R5;
R3 and R4 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, benzyl, aryl, heteroaryl, or heterocycle; or
R1 and R3 together comprise a heterocycle or heteroaryl, each optionally substituted by one or more R5;
R5 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, or (=0);
(b) a compound of formula (III):
OR16
I
0^P (CR17R18)q-R10
(HI)
wherein:
q is 0 to 5;
R10 is Ci to Cio alkyl. heteroaryl or aryl, each optionally substituted by one or more R19;
R16 is H or Ci to Cio alkyl;
R17 and R18 are, independently, absent, H, Ci to Cio alkyl, (Ci to Cio alkyl)-P-(Ci to Cio alkyl), Ci to Cio alkoxy, halogen, OH, N02, CN, C2 to Cio alkenyl, P(0)OH, C2 to Cio alkynyl, aryl, heteroaryl, or heterocycle, each optionally substituted by one or more R19;
wherein when R17 or R18 is absent and q is at least 2, the CR17R18 groups are bound through a stable double bond; R is H, Ci to Cio alkyl, Ci to C10 alkoxy, halogen, N02, CN, C(0)OH, C(0)(Ci to Cio alkyl), C2 to C10 alkenyl, oxo, or C2 to C10 alkynyl;
(c) a compound of formula (IV):
Figure imgf000161_0001
(IV)
wherein:
m is 0 to 2;
R20 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R24;
R21, R22, and R23 are independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl, each optionally substituted by one or more R24;
R24 is H, halogen, OH, CN, N02, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R27;
R25 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl;
R26 is O or S;
R27 is H, P(0)OH, or Ci to Cio alkyl optionally substituted by halogen or OP(0)OH;
(d) a compound of formula (V):
Figure imgf000161_0002
(V)
wherein:
R and R are, independently, H, Ci to Cio alkyl optionally substituted by one or more R42, aryl optionally substituted by one or more R42, or benzyl optionally substituted by one or more R42; R42 is H, Ci to Cio alkyl, C(0)OH, NH2, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), Ci to Cio aminoalkyl, halogen, aryl, or heteroaryl;
(e) a compound of formula (VI):
OR51
0=P— OR50
I
R52
(VI)
wherein:
R50 is heteroaryl or heterocycle, each optionally substituted by one or more R53;
R51 is H, Ci to Cio alkyl, Ci to Cio aminoalkyl, each optionally substituted by halogen or C(0)OH;
R52 is absent, Ci to Cio alkyl optionally substituted by P(0)2, or aryl optionally substituted by 0(Ci to Cio alkyl)-phenyl-C(0)OH;
R53 is Ci to Cio alkyl, halogen, or C(0)OH;
(f) a compound of formula (VII):
P— (CR65R66)tR60R61
HO
(VII)
wherein:
t is 1 to 10;
R60 is aryl, heteroaryl, or heterocycle;
R61 is Ci to Cio aminoalkyl, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, heteroaryl, or heterocycle;
R65 and R66 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl benzyl, aryl, heteroaryl, or heterocycle;
(g) a compound of formula (VIII):
Figure imgf000163_0001
(VIII)
wherein:
R70, R71, R74, and R75 are, independently, H, Ci to C10 alkyl, C2 to Cio alkenyl, Ci to C10 aminoalkyl, C(0)(Ci to C10 aminoalkyl), or heteroaryl, each optionally substituted by one or more R78:
R72 is H, Ci to Cio alkyl, 0(d to Cio alkyl), 0(d to Cio alkoxy), 0(Ci to Cio aminoalkyl), Ci to Cio aminoalkyl, or heteroaryl, each optionally substituted by one or more R76:
R73 is Ci to Cio alkyl, Ci to Cio alkoxy, Ci to Cio aminoalkyl, each optionally substituted by one or more R76:
R76 is C(0)OH, C(0)(Ci to Cio alkyl), NHC(0)(Ci to Cio alkyl), C(0)NH(phenyl optionally substituted by one or more R77), C(0)0(Ci to Cio alkyl), heteroaryl optionally substituted by one or more R77, heterocycle optionally substituted by one or more R77, or phenyl optionally substituted by one or more R77;
R77 is halogen, Ci to Cio aminoalkyl, Ci to Cio alkoxy, Ci to Cio alkyl, or C2 to Cio alkenyl, each optionally substituted by one or
78
more R ;
R78 is CN, C(0)OH, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), or phenyl optionally substituted by one or more R79;
R79 is OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, halogen, or Ci to Ce alkoxy;
(h) a compound of formula (IX):
Figure imgf000163_0002
(IX)
wherein:
w is 1 to 10;
w' is 1 to 10;
R80 is H or C2 to Cio alkyl;
R81 to R85 are, independently, H, OH, Ci to Cio alkyl, halogen, or Ci to Cio alkoxy; and
R86 to R89 are, independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl;
(i) a compound of formula (X):
Figure imgf000164_0001
(X)
wherein:
x is 2 to 10;
(j) a compound of formula (XI):
Figure imgf000164_0002
(XI)
wherein:
y is 1 to 10;
R90 to R94 are, independently, H, halogen, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, OH, or Ci to Cio aminoalkyl; and
R95 is heterocycle, heteroaryl, aryl, or Ci to Cio aminoalkyl; (k) a compound of formula (XII):
Figure imgf000165_0001
(XII)
wherein:
R10S is aryl, heteroaryl, or heterocycle;
z is 2 to 10;
Rio6 10 R i io ar6j independently, H, Ci to C10 alkyl, C3 to Cio cycloalkyl, OH, C2 to C10 alkenyl, or C2 to C10 alkynyl;
(1) a compound of formula (XIII):
Figure imgf000165_0002
(XIII)
wherein:
a is 0 or 1;
R120 is absent, aryl, C3 to C10 cycloalkyl, heteroaryl, or heterocycle, each optionally substituted by one or more R125;
R121 is aryl containing one or more R123, Ci to C10 aminoalkyl optionally containing one or two aryl groups optionally substituted by one or more R124, heterocycle optionally substituted by one or more R125;
R12~ is H, halogen, Ci to C10 alkyl, or Ci to C10 alkoxy;
R124 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, or Ci to Cio aminoalkyl;
R12S is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, (=0), or C(0)OH; or
R and R join to form C5 to Cio cycloalkyl optionally substituted by one or more R126; and R is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, or Ci to Cio aminoalkyl;
(m) a compound of formula (XIV):
Figure imgf000166_0001
(XIV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R202;
R202 is H, CI to Cio alkyl, halogen, CN, or 02; R200 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R 203 ;R 203 is halogen, CN, NO2, Ci to Cio alkoxy, or aryl optionally substituted by one or more R204;
R204 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl;
(n) a compound of formula (XV):
Figure imgf000166_0002
(XV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R302;
R302 is H, Ci to Cio alkyl, halogen, CN, or N02; R300 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R303; R303 is halogen, CN, O2, or aryl optionally substituted by one or more R304;
R304 is Ci to Cio alkyl, Ci to C10 alkoxy, halogen, CN, N02, or aryl;
(0) a compound of formula (XVI):
Figure imgf000167_0001
(XVI)
wherein:
R400 is OH or O;
R401, R402, and R403, are independently, OR405 or N(R406)(R407); R404 is H, Ci to Cio alkyl, halogen, (=0), or Ci to Cio alkoxy; R405 is absent, Ci to Cio alkyl, heterocycle, aryl, or heteroaryl,
R406
optionally substituted by one or more
R406 is H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, C(0)(Ci to Cio alkyl), aryl, heteroaryl, C(0)(aryl optionally substituted by one or more R404;
R407 is absent, H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, aryl, or heteroaryl;
with the proviso that when R400 is O and R405 or R407 is absent, then R400 and R402 join to form a 5 to 8-membered ring, wherein the 5 to 8-membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404; and
with the proviso that when R401 is OR405 and R403 is
N(R406)(R407) R405 ^ R407 ^ R401 ^ R402j R401 and R403, and/or R402 and R403 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404;
(p) a compound of formula (XVII):
Figure imgf000168_0001
(XVII)
wherein:
R500 is Ci to Cio aminoalkyl;
R502 is benzyl optionally substituted by one or more R503; R5 is H, Ci to Cio alkyl, C? to Cio cycloalkyl, Ci to Cio alkoxy, or aryl; f is 0 to 4; and
R503 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, or aryl; (q) a compo
Figure imgf000168_0002
(XVIII)
wherein:
R507 is H, OH, Ci to do alkoxy, or C3 to C10 cycloalkyl;
Rso8 tQ R5 io are^ independently, H, Ci to Cio alkyl, Ci to Cio alkoxy, C3 to Cio cycloalkyl, or halogen; and
(r) a compound of formula (XIX):
Figure imgf000168_0003
(XIX)
wherein:
R600 is Ci to Cio alkyl, NH2 or Ci to Cio aminoalkyl;
R602 tQ R605 are^ independently, Ci to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or halogen;
(s) a compound of formula (XX):
Figure imgf000169_0001
(XX)
wherein:
R606 is H or Ci to Cio alkyl;
R607 tQ R6io ar6j independently, H, Ci to Cio alkyl, Ci to Cio alkoxy, or halogen;
(t) a compou
Figure imgf000169_0002
(XXI)
wherein:
R /oo to R/o4 are^ independently, H, Ci to Cio alkyl, C3 to CIO cycloalkyl, C3 to Cio spirocycloalkyl, Ci to Cio alkoxy, halogen, Ci to Cio aminoalkyl, or CN; and
R705 is H or Ci to Cio alkyl;
(u) a compo
Figure imgf000169_0003
(XXII)
wherein: R800 tQ R804 RWb RS0y ar6j independentlyj H, Cl tO C alkyl, halogen, NH2, or Ci to Cio alkoxy and
805 is H or Ci to do alkyl;
(v) a compound of formula (XXIII):
Figure imgf000170_0001
(XXIII)
wherein:
R9UU is H or Ci to Cio alkyl; and
R901 to R905 is H, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, halogen, Ci to Cio alkoxy, Ci to Cio aminoalkyl, or H2; and (w) a compound of formula (XXIV):
Figure imgf000170_0002
(XXIV)
wherein:
RJU6 is Ci to Cio alkyl or Ci to Cio alkoxy;
R907 is C2 to Cio alkyl or Ci to Cio alkoxy;
e is 0 to 5;
R908 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, OH, CN, or NH2; and
R909 is H or Ci to Cio alkyl;
or a pharmaceutically acceptable salt or prodrug thereof.
97. The composition according to claim 96, wherein said medication is a chemotherapeutic, cardiotonic, blood pressure lowering agent, or cholesterol lowering agent.
98. The composition according to claim 96, wherein said medication which disrupts PF4 tetramers is CKEY2, ODSH (o-desulfated heparin), or a combination thereof.
99. A composition comprising:
(i) a pharmaceutically acceptable carrier; and
(ii) a compound selected from the group consisting of:
(a) a compound of formula (II):
Figure imgf000171_0001
(II)
wherein:
n is 0 to 5;
R1 and R2 are, independently, H, Ci to C10 alkyl, C(0)(C1 to Cio alkyl), C(0)(C1 to C10 alkyl), benzyl, C3 to C10 cycloalkyl, C2 to Cio alkenyl, or C2 to C10 alkynyl; or
R1 and R2 together comprise a heterocycle or heteroaryl, optionally substituted by one or more R5;
R3 and R4 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, benzyl, aryl, heteroaryl, or heterocycle; or
R1 and R3 together comprise a heterocycle or heteroaryl, each optionally substituted by one or more R5;
R5 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, or (=0);
(b) a compound of formula (III):
OR16
I
0^P (CR17R18)q-R10
(HI)
wherein:
q is 0 to 5;
R10 is Ci to Cio alkyl. heteroaryl or aryl, each optionally substituted by one or more R19; Rlb is H or Ci to C10 alkyl;
R17 and R18 are, independently, absent, H, Ci to C10 alkyl, (Ci to Cio alkyl)-P-(Ci to C10 alkyl), Ci to C10 alkoxy, halogen, OH, N02, CN, C2 to Cio alkenyl, P(0)OH, C2 to Cio alkynyl, aryl, heteroaryl, or heterocycle, each optionally substituted by one or more R19;
wherein when R17 or R18 is absent and q is at least 2, the CR17R18 groups are bound through a stable double bond;
R19 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, N02, CN, C(0)OH, C(0)(Ci to Cio alkyl), C2 to Cio alkenyl, oxo, or C2 to Cio alkynyl;
(c) a compound of formula (IV):
Figure imgf000172_0001
(IV)
wherein:
m is 0 to 2;
R20 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R24;
R21, R22, and R23 are independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl, each optionally substituted by one or more R24;
R24 is H, halogen, OH, CN, N02, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R27;
R25 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl;
R26 is O or S;
R27 is H, P(0)OH, or Ci to Cio alkyl optionally substituted by halogen or OP(0)OH;
(d) a compound of formula (V): O
II
R41- P^R40
(V)
wherein:
R40 and R41 are, independently, H, Ci to C10 alkyl optionally substituted by one or more R42, aryl optionally substituted by one or more R42, or benzyl optionally substituted by one or more R42;
R42 is H, Ci to Cio alkyl, C(0)OH, NH2, C(0)(Ci to C10 alkyl), C(0)0(Ci to Cio alkyl), Ci to Cio aminoalkyl, halogen, aryl, or heteroaryl;
(e) a compound of formula (VI):
OR51
0=P— OR50
I
R52
(VI)
wherein:
R50 is heteroaryl or heterocycle, each optionally substituted by one or more R53;
R51 is H, Ci to Cio alkyl, Ci to Cio aminoalkyl, each optionally substituted by halogen or C(0)OH;
R52 is absent, Ci to Cio alkyl optionally substituted by P(0)2, or aryl optionally substituted by 0(Ci to Cio alkyl)-phenyl-C(0)OH;
R53 is Ci to Cio alkyl, halogen, or C(0)OH;
(f) a compound of formula (VII):
O
\\
P— (CR65R66)tR60R61
HO
(VII)
wherein:
t is 1 to 10;
R60 is aryl, heteroaryl, or heterocycle;
R61 is Ci to Cio aminoalkyl, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, heteroaryl, or heterocycle; RM and Br are, independently, H, OH, Ci to C10 alkyl, C3 to Cio cycloalkyl, C2 to C10 alkenyl, C2 to C10 alkynyl benzyl, aryl, heteroaryl, or heterocycle;
(g) a compound of formula (VIII):
Figure imgf000174_0001
(VIII)
wherein:
R , R , R/4, and R/5 are, independently, H, Ci to Cio alkyl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, C(0)(Ci to Cio aminoalkyl), or heteroaryl, each optionally substituted by one or more R78;
R72 is H, Ci to Cio alkyl, 0(d to Cio alkyl), 0(d to Cio alkoxy), 0(Ci to Cio aminoalkyl), Ci to Cio aminoalkyl, or heteroaryl, each optionally substituted by one or more R76;
R73 is Ci to Cio alkyl, Ci to Cio alkoxy, Ci to Cio aminoalkyl, each optionally substituted by one or more R76;
R76 is C(0)OH, C(0)(Ci to Cio alkyl), NHC(0)(Ci to Cio alkyl), C(0)NH(phenyl optionally substituted by one or more R77), C(0)0(Ci to Cio alkyl), heteroaryl optionally substituted by one or more R77, heterocycle optionally substituted by one or more R77, or phenyl optionally substituted by one or more R77;
R77 is halogen, Ci to Cio aminoalkyl, Ci to Cio alkoxy, Ci to Cio alkyl, or C2 to Cio alkenyl, each optionally substituted by one or
78
more R ;
R78 is CN, C(0)OH, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), or phenyl optionally substituted by one or more R79;
R79 is OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, halogen, or Ci to Ce alkoxy;
(h) a compound of formula (IX):
Figure imgf000175_0001
(IX)
wherein:
w is 1 to 10;
w' is 1 to 10;
R80 is H or C2 to C io alkyl;
R81 to R85 are, independently, H, OH, d to C10 alkyl, halogen, or Ci to Cio alkoxy; and
R86 to R89 are, independently, H, d to Cio alkyl, or C3 to Cio cycloalkyl;
(i) a compound of formula (X):
Figure imgf000175_0002
(X)
wherein:
x is 2 to 10;
G) a compound of formula (XI):
Figure imgf000175_0003
wherein:
y is 1 to 10;
R90 to R94 are, independently, H, halogen, d to Cio alkyl, C2 to Cio alkenyl, to Cio alkynyl, OH, or d to Cio aminoalkyl; and
R95 is heterocycle, heteroaryl, aryl, or d to Cio aminoalkyl; (k) a compound of formula (XII):
Figure imgf000176_0001
(XII)
wherein:
R105 is aryl, heteroaryl, or heterocycle;
z is 2 to 10;
Rio6 tQ R i io ar6j independently, H, Ci to C10 alkyl, C3 to C10 cycloalkyl, OH, C2 to C10 alkenyl, or C2 to C10 alkynyl;
(1) a compound of formula (XIII):
Figure imgf000176_0002
(XIII)
wherein:
a is 0 or 1;
R120 is absent, aryl, C3 to C10 cycloalkyl, heteroaryl, or heterocycle, each optionally substituted by one or more R125;
R121 is aryl containing one or more R123, Ci to C10 aminoalkyl optionally containing one or two aryl groups optionally substituted by one or more R124, heterocycle optionally substituted by one or more R125;
R123 is H, halogen, Ci to C10 alkyl, or Ci to C10 alkoxy;
R124 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, or Ci to Cio aminoalkyl;
R125 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, (=0), or C(0)OH; or
R120 and R121 join to form C5 to C10 cycloalkyl optionally substituted by one or more R126; and R is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, or Ci to Cio aminoalkyl;
(m) a compound of formula (XIV):
Figure imgf000177_0001
(XIV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R202;
R202 is H, CI to Cio alkyl, halogen, CN, or 02; R200 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R 203 ;R 203 is halogen, CN, NO2, Ci to Cio alkoxy, or aryl optionally substituted by one or more R204;
R204 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl;
(n) a compound of formula (XV):
Figure imgf000177_0002
(XV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R302;
R302 is H, Ci to Cio alkyl, halogen, CN, or N02; R300 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R303; R303 is halogen, CN, O2, or aryl optionally substituted by one or more R304;
R304 is Ci to Cio alkyl, Ci to C10 alkoxy, halogen, CN, N02, or aryl;
(0) a compound of formula (XVI):
Figure imgf000178_0001
(XVI)
wherein:
R400 is OH or O;
R401, R402, and R403, are independently, OR405 or N(R406)(R407); R404 is H, Ci to Cio alkyl, halogen, (=0), or Ci to Cio alkoxy; R405 is absent, Ci to Cio alkyl, heterocycle, aryl, or heteroaryl,
R406
optionally substituted by one or more
R406 is H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, C(0)(Ci to Cio alkyl), aryl, heteroaryl, C(0)(aryl optionally substituted by one or more R404;
R407 is absent, H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, aryl, or heteroaryl;
with the proviso that when R400 is O and R405 or R407 is absent, then R400 and R402 join to form a 5 to 8-membered ring, wherein the 5 to 8-membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404; and
with the proviso that when R401 is OR405 and R403 is
N(R406)(R407) R405 ^ R407 ^ R401 ^ R402j R401 and R403, and/or R402 and R403 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404;
(p) a compound of formula (XVII):
Figure imgf000179_0001
(XVII)
wherein:
R5UU is Ci to Cio aminoalkyl;
R502 is benzyl optionally substituted by one or more R503; R5 is H, Ci to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or aryl; f is 0 to 4; and
R503 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, or aryl; (q) a compound of formula (XVIII):
Figure imgf000179_0002
(XVIII)
wherein:
R is H, OH, Ci to Cio alkoxy, or C3 to Cio cycloalkyl;
R508 10 R5 io are^ independently, H, Ci to Cio alkyl, Ci to Cio alkoxy, C? to Cio cycloalkyl, or halogen; and
(r) a compound of formula (XIX):
Figure imgf000179_0003
(XIX)
wherein:
R600 is Ci to Cio alkyl, NH2 or Ci to Cio aminoalkyl;
R602 tQ R605 are^ indeperideritlyi Ci to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or halogen;
(s) a compound of formula (XX):
Figure imgf000180_0001
(XX)
wherein:
RWJb is H or Ci to Cio alkyl;
R607 10 R6io are^ independently, H, Ci to Cio alkyl, Ci to Cio alkoxy, or halogen;
(t) a compou
Figure imgf000180_0002
(XXI)
wherein:
R to R/04 are, independently, H, Ci to Cio alkyl, C3 to C IO cycloalkyl, C3 to Cio spirocycloalkyl, Ci to Cio alkoxy, halogen, Ci to Cio aminoalkyl, or CN; and
R705 is H or Ci to Cio alkyl;
(u) a compou
Figure imgf000180_0003
(XXII)
wherein: Raoo tQ R8U4 and Rwb and ar6i independently5 H, Ci to ClO alkyl, halogen, NH2, or Ci to C10 alkoxy and
R805 is H or Ci to C10 alkyl;
(v) a compound of formula (XXIII):
Figure imgf000181_0001
(XXIII)
wherein:
R900 is H or Ci to C10 alkyl; and
R9oi t0 R9os is Hj C i tQ alky]) C2 10 Ci o alkenyl > Cl t0 C l 0 alkynyl, halogen, Ci to Cio alkoxy, Ci to Cio aminoalkyl, or NH2; and (w) a compound of formula (XXIV):
Figure imgf000181_0002
(XXIV)
wherein:
R906 is Ci to Cio alkyl or Ci to Cio alkoxy;
R907 is C2 to Cio alkyl or d to C10 alkoxy;
e is 0 to 5;
R908 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, OH, CN, or NH2; and
R909 is H or Ci to Cio alkyl;
or a pharmaceutically acceptable salt or prodmg thereof.
100. The composition according to any one of claims 94 to 99, wherein said compound is selected from the group consisting of:
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
101. The composition according to any of claims 96 to 100, further comprising an adjuvant, syrup, elixir, diluent, excipient, binder, lubricant, surfactant, granulating agent, emollient, metal chelator, pH adjustor, filler, and disintegrant.
102. The composition according to claim 101, wherein said diluent is water.
103. A kit comprising:
(i) a medication which causes the formation of PF4 tetramers, a medication which disrupts PF4 tetramers, or a combination thereof;
(ii) a compound selected from the group consisting of:
(a) a compound of formula (II):
Figure imgf000185_0002
(II) wherein:
n is 0 to 5;
R1 and R2 are, independently, H, Ci to C10 alkyl, C(0)(C1 to Cio alkyl), C(0)(C1 to C10 alkyl), benzyl, C3 to C10 cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl; or
R1 and R2 together comprise a heterocycle or heteroaryl, optionally substituted by one or more R5;
R3 and R4 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, benzyl, aryl, heteroaryl, or heterocycle; or
R1 and R3 together comprise a heterocycle or heteroaryl, each optionally substituted by one or more R5;
R5 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, or (=0);
(b) a compound of formula (III):
OR16
I
0^P (CR17R18)q-R10
(III)
wherein:
q is 0 to 5;
R10 is Ci to Cio alkyl. heteroaryl or aryl, each optionally substituted by one or more R19;
R16 is H or Ci to Cio alkyl;
R17 and R18 are, independently, absent, H, Ci to Cio alkyl, (Ci to Cio alkyl)-P-(Ci to Cio alkyl), Ci to Cio alkoxy, halogen, OH, N02, CN, C2 to Cio alkenyl, P(0)OH, C2 to Cio alkynyl, aryl, heteroaryl, or heterocycle, each optionally substituted by one or more R19;
wherein when R17 or R18 is absent and q is at least 2, the CR17R18 groups are bound through a stable double bond;
R19 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, N02, CN, C(0)OH, C(0)(Ci to Cio alkyl), C2 to Cio alkenyl, oxo, or C2 to Cio alkynyl; a compound of formula (IV):
Figure imgf000187_0001
(IV)
wherein:
m is 0 to 2;
R2U is H, Ci to Cio alkyl C3 to C10 cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R24:
R21, R22, and R23 are independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl, each optionally substituted by one or more R24:
R24 is H, halogen, OH, CN, N02, d to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R27;
R25 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl;
R26 is O or S;
R27 is H, P(0)OH, or Ci to Cio alkyl optionally substituted by halogen or OP(0)OH;
(d) a compound of formula (V):
O
R41 - ^ R40
(V)
wherein:
R40 and R41 are, independently, H, Ci to Cio alkyl optionally substituted by one or more R42, aryl optionally substituted by one or more R42, or benzyl optionally substituted by one or more R42:
R42 is H, Ci to Cio alkyl, C(0)OH, NH2, C(0)(d to Cio alkyl), C(0)0(Ci to Cio alkyl), Ci to Cio aminoalkyl, halogen, aryl, or heteroaryl;
(e) a compound of formula (VI): OR51
0=P— OR50
I
R52
(VI)
wherein:
R50 is heteroaryl or heterocycle, each optionally substituted by one or more R53;
R51 is H, Ci to Cio alkyl, Ci to Cio aminoalkyl, each optionally substituted by halogen or C(0)OH;
R52 is absent, Ci to Cio alkyl optionally substituted by P(0)2, or aryl optionally substituted by 0(Ci to Cio alkyl)-phenyl-C(0)OH;
R53 is Ci to Cio alkyl, halogen, or C(0)OH;
(f) a compound of formula (VII):
P— (CR65R66)tR60R61
HO
(VII)
wherein:
t is 1 to 10;
R60 is aryl, heteroaryl, or heterocycle;
R61 is Ci to Cio aminoalkyl, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, heteroaryl, or heterocycle;
R65 and R66 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl benzyl, aryl, heteroaryl, or heterocycle;
(g) a compound of formula (VIII):
Figure imgf000188_0001
(VIII)
wherein: R , R , R , and R are, independently, H, Ci to C10 alkyl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, C(0)(Ci to Cio aminoalkyl), or heteroaryl, each optionally substituted by one or more R78;
R72 is H, Ci to Cio alkyl, 0(Ci to Cio alkyl), 0(Ci to Cio alkoxy), 0(Ci to Cio aminoalkyl), Ci to Cio aminoalkyl, or heteroaryl, each optionally substituted by one or more R76;
R73 is Ci to Cio alkyl, Ci to Cio alkoxy, Ci to Cio aminoalkyl, each optionally substituted by one or more R76;
R76 is C(0)OH, C(0)(Ci to Cio alkyl), NHC(0)(Ci to Cio alkyl), C(0)NH(phenyl optionally substituted by one or more R77), C(0)0(Ci to Cio alkyl), heteroaryl optionally substituted by one or more R77, heterocycle optionally substituted by one or more R77, or phenyl optionally substituted by one or more R77;
R77 is halogen, Ci to Cio aminoalkyl, Ci to Cio alkoxy, Ci to Cio alkyl, or C2 to Cio alkenyl, each optionally substituted by one or more R78;
R78 is CN, C(0)OH, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), or phenyl optionally substituted by one or more
R79 is OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, halogen, or Ci to Ce alkoxy;
(h) a compound of formula (IX):
Figure imgf000189_0001
(IX)
wherein:
w is 1 to 10;
w' is 1 to 10;
R80 is H or C2 to Cio alkyl;
R81 to R85 are, independently, H, OH, Ci to Cio alkyl, halogen, or Ci to Cio alkoxy; and R to R are, independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl;
(i) a compound of formula (X):
Figure imgf000190_0001
(X)
wherein:
x is 2 to 10;
(j) a compound of formula (XI):
Figure imgf000190_0002
(XI)
wherein:
y is 1 to 10;
R90 to R94 are, independently, H, halogen, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, OH, or Ci to Cio aminoalkyl; and
R95 is heterocycle, heteroaryl, aryl, or Ci to Cio aminoalkyl; (k) a compound of formula (XII):
Figure imgf000190_0003
(XII)
wherein:
R105 is aryl, heteroaryl, or heterocycle;
z is 2 to 10; Rio6 tQ R i io ar6j independently, H, Ci to C10 alkyl, C3 to C10 cycloalkyl, OH, C2 to C10 alkenyl, or C2 to C10 alkynyl;
(1) a compound of formula (XIII):
Figure imgf000191_0001
(XIII)
wherein:
a is 0 or 1;
R120 is absent, aryl, C3 to C10 cycloalkyl, heteroaryl, or heterocycle, each optionally substituted by one or more R125;
R121 is aryl containing one or more R123, Ci to C10 aminoalkyl optionally containing one or two aryl groups optionally substituted by one or more R124, heterocycle optionally substituted by one or more R125;
R123 is H, halogen, Ci to C10 alkyl, or Ci to C10 alkoxy;
R124 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, or Ci to Cio aminoalkyl;
R125 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, (=0), or C(0)OH; or
R120 and R121 join to form C5 to C10 cycloalkyl optionally substituted by one or more R126; and
R126 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, or Ci to Cio aminoalkyl;
(m) a compound of formula (XIV):
Figure imgf000191_0002
(XIV)
wherein: R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R202;
R202 is H, CI to Cio alkyl, halogen, CN, or 02; R200 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio
203 203 aminoalkyl, each optionally substituted by one or more R ;R is halogen, CN, NO2, Ci to Cio alkoxy, or aryl optionally substituted by one or more R204;
R204 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl;
(n) a compound of formula (XV):
Figure imgf000192_0001
(XV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl,
R302 or C2 to Cio alkynyl, each optionally substituted by one or more
R302 is H, Ci to Cio alkyl, halogen, CN, or N02; R300 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R303;
R303 is halogen, CN, NO2, or aryl optionally substituted by one or more R304;
R304 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl;
(0) a compound of formula (XVI):
Figure imgf000192_0002
(XVI)
wherein:
R4UU is OH or O; R401, R402, and R403, are independently, OR405 or N(R406)(R407); R404 is H, Ci to Cio alkyl, halogen, (=0), or Ci to C10 alkoxy; R405 is absent, Ci to Cio alkyl, heterocycle, aryl, or heteroaryl,
R406
optionally substituted by one or more
R406 is H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, C(0)(Ci to Cio alkyl), aryl, heteroaryl, C(0)(aryl optionally substituted by one or more R404;
R407 is absent, H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, aryl, or heteroaryl;
with the proviso that when R400 is O and R405 or R407 is absent, then R400 and R402 join to form a 5 to 8-membered ring, wherein the 5 to 8-membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404; and
with the proviso that when R401 is OR405 and R403 is
N(R406)(R407) R405 ^ R407 ^ R401 ^ R402j R401 and R403, and/or R402 and R403 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered
R404
ring is optionally substituted by one or more
(p) a compound of formula (XVII):
Figure imgf000193_0001
(XVII)
wherein:
R500 is Ci to Cio aminoalkyl;
R502 is benzyl optionally substituted by one or more is H, Ci to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or aryl; f is 0 to 4; and
R503 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, or aryl; (q) a compound of formula (XVIII):
Figure imgf000194_0001
(XVIII)
wherein:
R is H, OH, Ci to Cio alkoxy, or C3 to do cycloalkyl;
R508 tQ R5 io are> independently, H, Ci to Cio alkyl, Ci to Cio alkoxy, C3 to Cio cycloalkyl, or halogen; and
(r) a comp
Figure imgf000194_0002
(XIX)
wherein:
R600 is Ci to Cio alkyl, NH2 or Ci to Cio aminoalkyl;
R 602 tQ R 605 are> independently, Ci to Cio alkyl, C3 to Cio cycloalkyl, Ci to C io alkoxy, or halogen;
(s) a compound of formula (XX):
Figure imgf000194_0003
(XX)
wherein:
R^ is H or Ci to Cio alkyl;
R607 to R610 are, independently, H, Ci to Cio alkyl, Ci to Cio alkoxy, or halogen;
(t) a compound of formula (XXI):
Figure imgf000195_0001
(XXI)
wherein:
R7oo tQ R704 ar6j independently, H, Ci to C10 alkyl, C3 to CIO cycloalkyl, C3 to C10 spirocycloalkyl, Ci to C10 alkoxy, halogen, d to Cio aminoalkyl, or CN; and
R705 is H or Ci to C10 alkyl;
(u) a compound of formula (XXII):
Figure imgf000195_0002
(XXII)
wherein:
R8UU to R8U and R8UO and Rsuv are, independently, H, Ci to C10 alkyl, halogen, NH2, or Ci to C10 alkoxy and
R805 is H or Ci to Cio alkyl;
(v) a compound of formula (XXIII):
Figure imgf000195_0003
(XXIII)
wherein:
R900 is H or Ci to Cio alkyl; and Ryui to Ryu3 is H, Ci to Cio alkyl, C2 to C10 alkenyl, C2 to C10 alkynyl, halogen, Ci to C10 alkoxy, Ci to C10 aminoalkyl, or NH2; and (w) a compound of formula (XXIV):
Figure imgf000196_0001
(XXIV)
wherein:
R is Ci to Cio alkyl or Ci to Cio alkoxy;
R907 is C2 to Cio alkyl or Ci to Cio alkoxy;
e is 0 to 5;
R908 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, OH, CN, or NH2; and
R909 is H or Ci to Cio alkyl;
or a pharmaceutically acceptable salt or prodrug thereof; and
(iii) instructions for administering said medication and said compound to a subject having cancer.
104. The kit according to claim 103, wherein said medication which causes the formation of PF4 tetramers is a chemotherapeutic, cardiotonic, blood pressure lowering agent, or cholesterol lowering agent.
105. The kit according to claim 103, wherein said medication which disrupts PF4 tetramers is CKEY2, ODSH (o-desulfated heparin), or a combination thereof
106. A kit comprising:
(i) a pharmaceutically acceptable carrier;
(ii) a compound selected from the group consisting of:
(a) a compound of formula (II):
Figure imgf000196_0002
(II)
wherein:
n is 0 to 5; R1 and R2 are, independently, H, Ci to C10 alkyl, C(0)(C1 to Cio alkyl), C(0)(C1 to C10 alkyl), benzyl, C3 to C10 cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl; or
R1 and R2 together comprise a heterocycle or heteroaryl, optionally substituted by one or more R5;
R3 and R4 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, benzyl, aryl, heteroaryl, or heterocycle; or
R1 and R3 together comprise a heterocycle or heteroaryl, each optionally substituted by one or more R5;
R5 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, OH, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, or (=0);
(b) a compound of formula (III):
OR16
I
^(CR17R18)q-R10
(III)
wherein:
q is 0 to 5;
R10 is Ci to Cio alkyl. heteroaryl or aryl, each optionally substituted by one or more R19;
R16 is H or Ci to Cio alkyl;
R17 and R18 are, independently, absent, H, Ci to Cio alkyl, (Ci to Cio alkyl)-P-(Ci to Cio alkyl), Ci to Cio alkoxy, halogen, OH, N02, CN, C2 to Cio alkenyl, P(0)OH, C2 to Cio alkynyl, aryl, heteroaryl, or heterocycle, each optionally substituted by one or more R19;
wherein when R17 or R18 is absent and q is at least 2, the CR17R18 groups are bound through a stable double bond;
R19 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, N02, CN, C(0)OH, C(0)(Ci to Cio alkyl), C2 to Cio alkenyl, oxo, or C2 to Cio alkynyl;
(c) a compound of formula (IV):
Figure imgf000198_0001
(IV)
wherein:
m is 0 to 2;
R20 is H, Ci to Cio alkyl C3 to C10 cycloalkyl, C2 to C10 alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R24:
R21, R22, and R23 are independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl, each optionally substituted by one or more R24:
R24 is H, halogen, OH, CN, N02, d to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl, each optionally substituted by one or more R27;
R25 is H, Ci to Cio alkyl C3 to Cio cycloalkyl, C2 to Cio alkenyl, or C2 to Cio alkynyl;
R26 is O or S;
R27 is H, P(0)OH, or Ci to Cio alkyl optionally substituted by halogen or OP(0)OH;
(d) a compound of formula (V):
O II
R41 ' P^ R40
(V)
wherein:
R40 and R41 are, independently, H, Ci to Cio alkyl optionally substituted by one or more R42, aryl optionally substituted by one or more R42, or benzyl optionally substituted by one or more R42:
R42 is H, Ci to Cio alkyl, C(0)OH, NH2, C(0)(d to Cio alkyl), C(0)0(Ci to Cio alkyl), Ci to Cio aminoalkyl, halogen, aryl, or heteroaryl;
(e) a compound of formula (VI): OR51
0=P— OR50
I
R52
(VI)
wherein:
R50 is heteroaryl or heterocycle, each optionally substituted by one or more R53;
R51 is H, Ci to Cio alkyl, Ci to Cio aminoalkyl, each optionally substituted by halogen or C(0)OH;
R52 is absent, Ci to Cio alkyl optionally substituted by P(0)2, or aryl optionally substituted by 0(Ci to Cio alkyl)-phenyl-C(0)OH;
R53 is Ci to Cio alkyl, halogen, or C(0)OH;
(f) a compound of formula (VII):
P— (CR65R66)tR60R61
HO
(VII)
wherein:
t is 1 to 10;
R60 is aryl, heteroaryl, or heterocycle;
R61 is Ci to Cio aminoalkyl, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, C3 to Cio cycloalkyl, aryl, heteroaryl, or heterocycle;
R65 and R66 are, independently, H, OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl benzyl, aryl, heteroaryl, or heterocycle;
(g) a compound of formula (VIII):
Figure imgf000199_0001
(VIII)
wherein: R , R , R , and R are, independently, H, Ci to C10 alkyl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, C(0)(Ci to Cio aminoalkyl), or heteroaryl, each optionally substituted by one or more R78;
R72 is H, Ci to Cio alkyl, 0(Ci to Cio alkyl), 0(Ci to Cio alkoxy), 0(Ci to Cio aminoalkyl), Ci to Cio aminoalkyl, or heteroaryl, each optionally substituted by one or more R76;
R73 is Ci to Cio alkyl, Ci to Cio alkoxy, Ci to Cio aminoalkyl, each optionally substituted by one or more R76;
R76 is C(0)OH, C(0)(Ci to Cio alkyl), NHC(0)(Ci to Cio alkyl), C(0)NH(phenyl optionally substituted by one or more R77), C(0)0(Ci to Cio alkyl), heteroaryl optionally substituted by one or more R77, heterocycle optionally substituted by one or more R77, or phenyl optionally substituted by one or more R77;
R77 is halogen, Ci to Cio aminoalkyl, Ci to Cio alkoxy, Ci to Cio alkyl, or C2 to Cio alkenyl, each optionally substituted by one or more R78;
R78 is CN, C(0)OH, C(0)(Ci to Cio alkyl), C(0)0(Ci to Cio alkyl), or phenyl optionally substituted by one or more
R79 is OH, Ci to Cio alkyl, C3 to Cio cycloalkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, halogen, or Ci to Ce alkoxy;
(h) a compound of formula (IX):
Figure imgf000200_0001
(IX)
wherein:
w is 1 to 10;
w' is 1 to 10;
R80 is H or C2 to Cio alkyl;
R81 to R85 are, independently, H, OH, Ci to Cio alkyl, halogen, or Ci to Cio alkoxy; and R to R are, independently, H, Ci to Cio alkyl, or C3 to Cio cycloalkyl;
(i) a compound of formula (X):
Figure imgf000201_0001
(X)
wherein:
x is 2 to 10;
(j) a compound of formula (XI):
Figure imgf000201_0002
wherein:
y is 1 to 10;
R90 to R94 are, independently, H, halogen, Ci to Cio alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, OH, or Ci to Cio aminoalkyl; and
R95 is heterocycle, heteroaryl, aryl, or Ci to Cio aminoalkyl; a compound of formula (XII)
Figure imgf000201_0003
(XII)
wherein:
R105 is aryl, heteroaryl, or heterocycle;
z is 2 to 10;
R io6 tQ R i io ar6j independently, H, Ci to Cio alkyl, C3 to Cio cycloalkyl, OH, C2 to Cio alkenyl, or C2 to Cio alkynyl; (1) a compound of formula (XIII):
Figure imgf000202_0001
(XIII)
wherein:
a is 0 or 1;
R120 is absent, aryl, C3 to C10 cycloalkyl, heteroaryl, or heterocycle, each optionally substituted by one or more R125;
R121 is aryl containing one or more R123, Ci to C10 aminoalkyl optionally containing one or two aryl groups optionally substituted by one or more R124, heterocycle optionally substituted by one or more R125;
R123 is H, halogen, Ci to C10 alkyl, or Ci to C10 alkoxy;
R124 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, or Ci to Cio aminoalkyl;
R125 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, (=0), or C(0)OH; or
R120 and R121 join to form C5 to C10 cycloalkyl optionally substituted by one or more R126; and
R126 is H, halogen, Ci to C10 alkyl, Ci to C10 alkoxy, or Ci to Cio aminoalkyl;
(m) a compound of formula (XIV):
j2
Figure imgf000202_0002
(XIV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl, to Cio alkynyl, each optionally substituted by one or more R202; Rzvz is H, CI to Cio alkyl, halogen, CN, or 02; R is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R ;R is halogen, CN, NO2, Ci to Cio alkoxy, or aryl optionally substituted by one or more R204;
R204 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl;
(n) a compound of formula (XV):
Figure imgf000203_0001
(XV)
wherein:
R is Ci to Cio alkyl, Ci to Cio aminoalkyl, C2 to Cio alkenyl,
R302 or C2 to Cio alkynyl, each optionally substituted by one or more
R302 is H, Ci to Cio alkyl, halogen, CN, or N02; R300 is Ci to Cio alkyl, Ci to Cio alkoxy, aryl, C2 to Cio alkenyl, Ci to Cio aminoalkyl, each optionally substituted by one or more R303;
R303 is halogen, CN, NO2, or aryl optionally substituted by one or more R304;
R304 is Ci to Cio alkyl, Ci to Cio alkoxy, halogen, CN, N02, or aryl;
(0) a compound of formula (XVI):
Figure imgf000203_0002
(XVI)
wherein:
4UU is OH or O;
Figure imgf000203_0003
R is H, Ci to Cio alkyl, halogen, (=0), or Ci to Cio alkoxy; R405 is absent, Ci to Cio alkyl, heterocycle, aryl, or heteroaryl,
R406
optionally substituted by one or more
R406 is H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, C(0)(Ci to Cio alkyl), aryl, heteroaryl, C(0)(aryl optionally substituted by one or more R404;
R407 is absent, H, Ci to Cio alkyl, halogen, Ci to Cio alkoxy, aryl, or heteroaryl;
with the proviso that when R400 is O and R405 or R407 is absent, then R400 and R402 join to form a 5 to 8-membered ring, wherein the 5 to 8-membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered ring is optionally substituted by one or more R404; and
with the proviso that when R401 is OR405 and R403 is
N(R406)(R407) R405 ^ R407 ^ R401 ^ R402j R401 and R403, and/or R402 and R403 join to form a 5 to 8-membered ring, wherein the 5 to 8- membered ring contains carbon atoms and optionally one or more heteroatom and wherein the 5 to 8-membered
R404
ring is optionally substituted by one or more
(p) a compound of formula (XVII):
Figure imgf000204_0001
(XVII)
wherein:
R500 is Ci to Cio aminoalkyl;
R502 is benzyl optionally substituted by one or more R503; R5 is H, Ci to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or aryl; f is 0 to 4; and
R503 is H, Ci to Cio alkyl, Ci to Cio alkoxy, halogen, or aryl; (q) a compound of formula (XVIII):
Figure imgf000205_0001
(XVIII)
wherein:
R507 is H, OH, Ci to Cio alkoxy, or C3 to Cio cycloalkyl;
R508 10 R5 io are^ independently, H, Ci to Cio alkyl, Ci to Cio alkoxy, C3 to Cio cycloalkyl, or halogen; and
(r) a compo
Figure imgf000205_0002
wherein:
R600 is Ci to Cio alkyl, NH2 or Ci to Cio aminoalkyl;
R602 tQ R605 are> independently, Ci to Cio alkyl, C3 to Cio cycloalkyl, Ci to Cio alkoxy, or halogen;
(s) a compound of formula (XX):
Figure imgf000205_0003
(XX)
wherein:
R606 is H or Ci to Cio alkyl;
R607 tQ R6io are> independently, H, Ci to Cio alkyl, Ci to Cio alkoxy, or halogen;
(t) a compound of formula (XXI):
Figure imgf000206_0001
(XXI)
wherein:
R7oo tQ R704 ar6j independentlyj Hj Cl t0 Cl0 aiky^ C3 t0 C 10 cycloalkyl, C3 to C10 spirocycloalkyl, Ci to C10 alkoxy, halogen, Ci to Cio aminoalkyl, or CN; and
R705 is H or Ci to C10 alkyl;
(u) a compo
Figure imgf000206_0002
(XXII)
wherein:
R800 tQ R804 R806 ^ R809 ^ independentlyj H, Cl tO C alkyl, halogen, NH2, or Ci to C10 alkoxy and
805 is H or Ci to C10 alkyl;
Figure imgf000206_0003
(XXIII)
wherein: Ryuu is H or Ci to C10 alkyl; and
R901 to R905 is H, Ci to Cio alkyl, C2 to C10 alkenyl, C2 to C10 alkynyl, halogen, d to Cio alkoxy, Ci to C10 aminoalkyl, or H2; and (w) a compound of formula (XXIV):
Figure imgf000207_0001
(XXIV)
wherein:
R9lJb is Ci to Cio alkyl or Ci to Cio alkoxy;
R is C2 to Cio alkyl or Ci to Cio alkoxy;
e is 0 to 5;
R908 is H, halogen, Ci to Cio alkyl, Ci to Cio alkoxy, OH, CN, or NH2; and
R909 is H or Ci to Cio alkyl;
or a pharmaceutically acceptable salt or prodrug thereof,
(iii) instructions for administering said medication and said compound to a ect having cancer.
107. The kit according to claim any one of claims 103 to 106, wherein said compound is selected from the group consisting of:
Figure imgf000207_0002
Figure imgf000208_0001
-207-
Figure imgf000209_0001
-208-
Figure imgf000210_0001
-209-
Figure imgf000211_0001
-210-
PCT/US2013/032092 2012-03-23 2013-03-15 Small molecule antagonists of pf4 containing ultra large complexes Ceased WO2013142328A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261614709P 2012-03-23 2012-03-23
US61/614,709 2012-03-23

Publications (1)

Publication Number Publication Date
WO2013142328A1 true WO2013142328A1 (en) 2013-09-26

Family

ID=49223254

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/032092 Ceased WO2013142328A1 (en) 2012-03-23 2013-03-15 Small molecule antagonists of pf4 containing ultra large complexes

Country Status (1)

Country Link
WO (1) WO2013142328A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113025562A (en) * 2021-03-18 2021-06-25 浙江大学 Application of R406 in promotion of somatic cell reprogramming, reprogramming medium and method thereof
CN116710440A (en) * 2020-12-31 2023-09-05 赛诺哈勃药业(成都)有限公司 A kind of plasmin inhibitor, its preparation method and application

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060247213A1 (en) * 2002-03-21 2006-11-02 Schering Aktiengesellschaft Plasma carboxypeptidase b inhibitors
US20080305041A1 (en) * 2004-11-19 2008-12-11 Bioquanta Corp. Pf4 Pharmacophores and Their Uses
US20090311344A1 (en) * 2008-06-03 2009-12-17 Yurkow Edward J Dosing Regimen

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060247213A1 (en) * 2002-03-21 2006-11-02 Schering Aktiengesellschaft Plasma carboxypeptidase b inhibitors
US20080305041A1 (en) * 2004-11-19 2008-12-11 Bioquanta Corp. Pf4 Pharmacophores and Their Uses
US20090311344A1 (en) * 2008-06-03 2009-12-17 Yurkow Edward J Dosing Regimen

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
AIDOUDI ET AL.: "Interaction of PF4 (CXCL4) with the vasculature: A role in atherosclerosis and angiogenesis", THROMB HAEMOST., vol. 104, 2010, pages 941 - 948, Retrieved from the Internet <URL:http://www.thrombosis-online.com> [retrieved on 20130512] *
RAUOVA ET AL.: "Ultralarge complexes of PF4 and heparin are central to the pathogenesis of heparin-induced thrombocytopenia", BLOOD, vol. 105, 2005, pages 131 - 138, XP055223350, Retrieved from the Internet <URL:bloodjoumal.hematologylibrary.org> [retrieved on 20130423], DOI: doi:10.1182/blood-2004-04-1544 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116710440A (en) * 2020-12-31 2023-09-05 赛诺哈勃药业(成都)有限公司 A kind of plasmin inhibitor, its preparation method and application
JP2024507433A (en) * 2020-12-31 2024-02-20 シノハブ ファーマシューティカルズ カンパニー リミテッド Plasmin inhibitor, its preparation method and its application
JP7651708B2 (en) 2020-12-31 2025-03-26 シノハブ ファーマシューティカルズ カンパニー リミテッド Plasmin inhibitors, preparation method therefor and their applications
CN116710440B (en) * 2020-12-31 2025-10-28 赛诺哈勃药业(成都)有限公司 A plasmin inhibitor, its preparation method and application
CN113025562A (en) * 2021-03-18 2021-06-25 浙江大学 Application of R406 in promotion of somatic cell reprogramming, reprogramming medium and method thereof

Similar Documents

Publication Publication Date Title
US12128106B2 (en) ASGPR-binding compounds for the degradation of extracellular proteins
Xie et al. Allosteric inhibitors of SHP2 with therapeutic potential for cancer treatment
Henise et al. Irreversible Nek2 kinase inhibitors with cellular activity
ES2632975T3 (en) C5aR antagonists
AU2009256253B2 (en) Stat3 inhibitors
Kuang et al. Computational insight into the binding profile of the second-generation PET tracer PI2620 with tau fibrils
JP6291132B2 (en) Bruton tyrosine kinase inhibitor
Luo et al. Targeting of the FOXM1 oncoprotein by E3 ligase-assisted degradation
Matts et al. A systematic protocol for the characterization of Hsp90 modulators
Kong et al. Optimization of P2Y12 antagonist ethyl 6-(4-((benzylsulfonyl) carbamoyl) piperidin-1-yl)-5-cyano-2-methylnicotinate (AZD1283) led to the discovery of an oral antiplatelet agent with improved druglike properties
Brown et al. Structure-based design of stapled peptides that bind GABARAP and inhibit autophagy
KR102586710B1 (en) 5-5 fused ring as C5a inhibitor
Ni et al. Allosteric modulators of protein–protein interactions (PPIs)
MX2012007872A (en) Methods and compositions of targeted drug development.
EP2027087A2 (en) Intracellular kinase inhibitors
Lei et al. Structure-guided design of the first noncovalent small-molecule inhibitor of CRM1
EA027603B1 (en) Piperazine derivatives which reverse anticoagulant effects
Chen et al. Discovery of ddo-2213 as a potent and orally bioavailable inhibitor of the wdr5–mixed lineage leukemia 1 protein–protein interaction for the treatment of mll fusion leukemia
Gomez-Monterrey et al. Heat shock protein 90 inhibitors as therapeutic agents
Yang et al. Discovery of novel small-molecule-Based potential PD-L1/EGFR dual inhibitors with high druggability for glioblastoma immunotherapy
Cavalier et al. Small molecule inhibitors of Ca2+-S100B reveal two protein conformations
Bonomo et al. The role of fluorine in stabilizing the bioactive conformation of dihydroorotate dehydrogenase inhibitors
WO2013142328A1 (en) Small molecule antagonists of pf4 containing ultra large complexes
Fu et al. Discovery of potent, specific, and orally available NLRP3 inflammasome inhibitors based on pyridazine scaffolds for the treatment of septic shock and peritonitis
Epstein et al. Molecular determinants of binding of non-oxime bispyridinium nerve agent antidote compounds to the adult muscle nAChR

Legal Events

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

Ref document number: 13763866

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13763866

Country of ref document: EP

Kind code of ref document: A1