WO2005007672A2 - Small molecule toll-like receptor (tlr) antagonists - Google Patents

Small molecule toll-like receptor (tlr) antagonists Download PDF

Info

Publication number
WO2005007672A2
WO2005007672A2 PCT/US2004/019714 US2004019714W WO2005007672A2 WO 2005007672 A2 WO2005007672 A2 WO 2005007672A2 US 2004019714 W US2004019714 W US 2004019714W WO 2005007672 A2 WO2005007672 A2 WO 2005007672A2
Authority
WO
WIPO (PCT)
Prior art keywords
group
aryl
tlr
hydrogen atom
substituted
Prior art date
Application number
PCT/US2004/019714
Other languages
French (fr)
Other versions
WO2005007672A8 (en
WO2005007672A3 (en
Inventor
Grayson B. Lipford
Alexandra Forsbach
Charles M. Zepp
Original Assignee
Coley Pharmaceutical Gmbh
Coley Pharmaceutical Group, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to CN200480017064.7A priority Critical patent/CN1809357B/en
Priority to EA200600069A priority patent/EA200600069A1/en
Priority to AU2004257149A priority patent/AU2004257149A1/en
Priority to CA002528774A priority patent/CA2528774A1/en
Priority to BRPI0411514-7A priority patent/BRPI0411514A/en
Priority to JP2006517471A priority patent/JP2007524615A/en
Application filed by Coley Pharmaceutical Gmbh, Coley Pharmaceutical Group, Inc. filed Critical Coley Pharmaceutical Gmbh
Priority to EP04776820A priority patent/EP1635846A4/en
Priority to MXPA05013922A priority patent/MXPA05013922A/en
Publication of WO2005007672A2 publication Critical patent/WO2005007672A2/en
Publication of WO2005007672A3 publication Critical patent/WO2005007672A3/en
Priority to IL172580A priority patent/IL172580A0/en
Publication of WO2005007672A8 publication Critical patent/WO2005007672A8/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/382Heterocyclic compounds having sulfur as a ring hetero atom having six-membered rings, e.g. thioxanthenes
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4462Non condensed piperidines, e.g. piperocaine only substituted in position 3
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/5415Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with carbocyclic ring systems, e.g. phenothiazine, chlorpromazine, piroxicam
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/04Ortho- or peri-condensed ring systems
    • C07D221/06Ring systems of three rings
    • C07D221/10Aza-phenanthrenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/94Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/95Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in positions 2 and 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the invention relates to generally to the field of immunology. More particularly, the invention relates to compositions and methods for altering immune function. More specifically, the invention relates to compositions and methods for affecting immune stimulation mediated through Toll-like receptor (TLR) molecules.
  • TLR Toll-like receptor
  • TLRs Toll-like receptors
  • compositions and methods useful for modulating innate immunity are therefore of great interest, as they may affect therapeutic approaches to conditions involving autoimmunity, inflammation, allergy, asthma, graft rejection, graft versus host disease (GvHD), infection, cancer, and immunodeficiency.
  • TLR9 Toll-like receptor 9 (TLR9) recognizes bacterial DNA and CpG DNA. Hemmi H et al. (2000) Nature 408:740-5; Bauer S et al. (2001) Proc Natl Acad Sci USA 98:9237-42.
  • Chlroroquines have been recognized as useful not only as anti-malarial agents but also as anti-inflammatory agents. Although its mechanism of action is not well understood, chloroquine has been used effectively in the treatment of various autoimmune diseases, including rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). For a review, see Wallace DJ (1996) Lupus 5 Suppl 1 :S59-64.
  • RA rheumatoid arthritis
  • SLE systemic lupus erythematosus
  • the present invention is based, in part, on the discovery by the applicants that a number of small molecules, some previously known but distinct from those described by Macfarlane et al. and by Tobe et al., can alter TLR-mediated immunostimulatory signaling. Many of the compounds inhibit TLR signaling and are useful as inhibitors of immune stimulation. Compositions and methods described herein are useful for inhibiting immune stimulation in vitro and in vivo. Such compositions and methods thus will find use in a number of clinical applications, including as pharmaceutical agents and methods for treating conditions involving unwanted immune activity, including inflammatory and autoimmune disorders.
  • compositions of the invention can also be used in methods for the preparation of medicaments for use in the treatment of conditions involving unwanted imnlune activity, including a variety of inflammatory and autoimmune disorders.
  • molecules having similar substituents but different core structures compared to those related to chloroquine and quinacrine described by Macfarlane et al. are potent immunomodulatory small molecules.
  • the small molecules described by tjhe present invention affect immune stimulation via interaction with a TLR. More particularly, it is believed that many of the small molecules described by the present invention inhibit immune stimulation via TLR antagonism. In particular, it is believed that many of the small molecules described by the present invention inhibit immune stimulation via TLR9 antagonism.
  • the invention in certain aspects provides methods useful for altering TLR-mediated signaling.
  • the methods of the invention are useful whenever it is desirable to alter TLR- mediated signaling in response to a suitable TLR ligand or TLR signaling agonist.
  • the methods can be used to treat any of a variety of conditions involving autoimmunity, inflammation, allergy, asthma, graft rejection, graft-versus-host disease (GvHD), infection, sepsis, cancer, and immunodeficiency.
  • GvHD graft-versus-host disease
  • methods useful in the treatment of conditions involving autoimmunity, inflammation, allergy, asthma, graft rejection, and GvHD will employ small molecules that inhibit TLR-mediated signaling in response to a suitable TLR ligand or TLR signaling agonist.
  • method$ useful in the treatment of conditions involving infection, cancer, and immunodeficiency will lemploy small molecules that augment TLR-mediated signaling in response to a suitable TLR ligand.
  • the methods can be used to inhibit or promote TLR-mediated signaling in response to a TLR ligand or TLR signaling agonist.
  • the methods can be used to inhibit TLR-mediated immunostimulatory signaling in response to a TLR ligand or TLR signaling agonist.
  • the methods can be used to inhibit or promote TLR-mediated immunostimulation in a subject.
  • the methods can be used to inhibit TLR-mediated immunostimulation in a subject.
  • the methodss ' can be used to inhibit an immunostimulatory nucleic acid-associated response in a subject.
  • the invention in certain aspects provides novel small molecule compositions useful for altering TLR-mediated signaling.
  • the compositions of the invention are useful whenever it is desirable to alter TLR-mediated signaling in response to a suitable TLR ligand or TLR signaling agonist.
  • the small molecules can be used in methods td treat any of a variety of conditions involving autoimmunity, inflammation, allergy, asthma, graft rejection, GvHD, infection, sepsis, cancer, and immunodeficiency.
  • methods ⁇ seful in the treatment of conditions involving autoimmunity, inflammation, allergy, asthma, graft rejection, and GvHD will employ small molecules that inhibit TLR-mediated signaling in response to a suitable TLR ligand or TLR signaling agonist.
  • methods useful in the treatment of conditions involving infection, cancer, and immunodeficiency will employ small molecules that augment TLR-mediated signaling in response to a suitable TLR ligand.
  • the molecules can be used in a method to inhibit or promote TLR-mediated signaling in response to a TLR ligand or TLR signaling agonist.
  • the small molecules can be used in a method to inhibit TLR-mediated immunostimulatory signaling in response to a TLR ligand or TLR signaling agonist. In some instances the small molecules can be used in a method to inhibit or promote TLR-mediated immunostimulatioii in a subject. In some instances the small molecules can be used in a method to inhibit TLR-mediated immunostimulation in a subject. In some instances the small molecules can be uised to inhibit an immunostimulatory nucleic acid-associated response in a subject. As a feature of the present invention, the methods of the invention can be combined with administration of additional agents to achieve synergistic effect on TLR-m diated immunostimulation.
  • TLR-bearing cells e.g., antigen-presenting cells
  • agents described herein can be used in conjunction with additional agents which affect non-APC immune cells, e.g., T lymphocytes (T cells).
  • T cells T lymphocytes
  • a method of affecting TLR-mediated signaling in response to a TLR ligand involves contacting a cell expressing a TLR with an effective amount of a compound of Formula I
  • 2 is a five- to seven-membered homocyclic or heterocyclic ring, wherein each of X, Y, and Z is independently chosen from a carbon atom (C), a nitrogen atom (N), an oxygen atom (O), and a sulfur atom (S), and wherein B2 optionally includes at least one atom selected from C, N, O, and S; wherein 1 and 2 are optionally bridged by B3 to form a five- to seven-membered ring (3), wherein B3 optionally includes at least one atom selected from C, N, O, and S, and wherein when A is carbon, (3) is not pyridine; wherein 2 optionally includes an unsaturated bond; wherein (3) optionally includes an unsaturated bond; wherejin R2, when present, is a hydrogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, optionally linked to Z via N, O, or S
  • a method of inhibiting TLR-mediated immunostimulatory signaling involves contacting a cell expressing a TLR with an effective amount of a compound of Formula I, as provided above, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
  • the invention provides a method of affecting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula I, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject.
  • the invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula I, as provided above, to inhibit TLR-mediated immunostimulation in the subject.
  • the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject.
  • the method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula I, as provided above, to inhibit an immunostimulatory nucleic acid- associated response in the subject.
  • the subject is otherwise free of symptoms calling for treatment with a compound of Formula I.
  • A is nitrogen and (3) is a five-membered ring.
  • the compound is Formula II
  • A is chosen from C and N; and Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S.
  • the compound is any one of compounds 455, 470, 564, 568, 593, 607, 612-614, 619-621, 636, 685, 875, 878, 890, 904, 918, 939, 944, 1039, 1050, 1132, 1241, and 1243 listed in Table 5 below.
  • A is nitrogen
  • (3) is a six-membered ring
  • B3 is one atom selected from C, N, O, and S.
  • A is nitrogen
  • 3 is a six-membered ring
  • B3 is S.
  • the compound is Formula III
  • Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked viaN, O, or S, and wherein X is chosen from N, O, and S. In certain embodiments X is S.
  • the compound is any one of compounds 53, 64, 125, 149, 313, 399, 529, 576, 693, 797, 840, and 842 listed in Table 6 below.
  • A is carbon
  • (3) is a six-membered ring
  • B3 is C or S.
  • the compound is Formula IV
  • X is C or S and Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S.
  • Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S.
  • the compound is any one of compounds 43, 294, 340, 346, 348, 413, 491, 917, 1015, 1042, 1158, 1287, and 1337 listed in Table 7 below.
  • A is nitrogen
  • (3) is a seven-membered ring
  • B3 includes two carbon atoms.
  • the compound is Formula V
  • Formula V wherein X and Y are each independently C, N, O, or S; and Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S.
  • the compound is any one of compounds 72, 74, 99, 109, 343, 488, 1013, and 1352 listed in Table 8 below.
  • 1 and 2 are unbridged by B3, A is carbon, and A' is -OR9.
  • the compound is Formula VI
  • Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and R9 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group.
  • R9 and R4 are united to form a lactone.
  • the compound is any one of compounds 65, 229, 239, 306, 386, 707, 793, 957, 970, 974, 1161, and 1308 listed in Table 9 below.
  • 1 and 2 are unbridged by B3, A is carbon, and A' is -NR9R10.
  • the compound is Formula VII
  • Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked vi N, O, or S.
  • the compound is any one of compounds 133, 267, 312, 457, and 510 listed in Table 10 below.
  • 1 and 2 are unbridged by B3, A is carbon, and A' is -CR9R10.
  • the compound is Formula VIII
  • Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S.
  • the compound is any one of compounds 93, 108, 138, 144, 267, 270, 308, 381, 560, 778, 799, 822, 833, 884, 965, and 1187 listed in Table 11 below.
  • the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand.
  • the method according to this aspect of the inventioni involves contacting a cell expressing a TLR with an effective amount of a compound of Formula IX
  • R2 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein R10 is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated signaling in response to a ligand for the
  • a method of inhibiting TLR-mediated immunostimulatory signaling involves contacting a cell expressing a TLR with an effective amount of a compound of Formula IX, as provided above, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
  • the invention provides a method of affecting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula IX, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject.
  • the invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula IX, as provided above, to inhibit TLR-mediated immunostimulation in the subject.
  • the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject.
  • the method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula IX, as provided above, to inhibit an immunostimulatory nucleic acid- associated response in the subject.
  • the subject is otherwise free of symptoms calling for treatment with a compound of Formula IX.
  • the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand.
  • the method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula X
  • each of Rl, R2, R3, R4, R5, R6, R7, and R8 is independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S, to inhibit or promote TLR- mediated signaling in response to a ligand for the TLR.
  • the compound is any one of compounds 431, 583, 586, 792, and 830 listed in Table 13 below.
  • a method of inhibiting TLR-mediated immunostimulatory signaling involves contacting a cell expressing a TLR with an effective amount of a compound of Formula X, as provided above, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
  • the invention provides a method of affecting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula X, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject.
  • the invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula X, as provided above, to inhibit TLR-mediated immunostimulation in the subject.
  • the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject.
  • the method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula X, as provided above, to inhibit an immunostimulatory nucleic acid- associated response in the subject.
  • the subject is otherwise free of symptoms calling for treatment with a compound of Formula X.
  • the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand.
  • the method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XI
  • each of Rl, R2, R5, R6, R7, R8, Rl 1, and R12 is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and X is C, N, O, or S, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR.
  • R2 includes a cycloalkyl, benzyl or phenyl group.
  • X is N.
  • Rl 1 and R12 are linked as a spiro group in a five- or six-membered ring.
  • the compound is any one of compounds 891, 926, 1137, 1213, 1248, 1320, and 1322 listed in Table 14 below.
  • a method of inhibiting TLR-mediated immunostimulatory signaling involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XI, as provided above, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
  • the invention provides a method of affecting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XI, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject.
  • the invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XI, as provided above, to inhibit TLR-mediated immunostimulation in the subject.
  • the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject.
  • the method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula XI, as provided above, to inhibit an immunostimulatory nucleic acid- associated response in the subject.
  • the subject is otherwise free of symptoms calling for treatment with a compound of Formula XI.
  • the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XII
  • Rl, R2, R3, R4, R5, R6, R7, R8, R9, RIO, and Rl 1 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR.
  • the compound is any one of compounds 101, 600, and 609 listed in Table 15 below.
  • the method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XII, as provided above, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
  • the invention provides a method of affecting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XII, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject.
  • the invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XII, as provided above, to inhibit TLR-mediated immunostimulation in the subject.
  • the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject.
  • the method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula XII, as provided above, to inhibit an immunostimulatory nucleic acid-associated response in the subject.
  • the subject is otherwise free of symptoms calling for treatment with a compound of Formula XII.
  • the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand.
  • the method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XIII
  • 2' is a five- to seven-membered heterocyclic ring, wherein each of X, Y, and Z is independently chosen from C, N, O, and S, and wherein B2' optionally includes at least one atom selected from C, N, O, and S; wherein 2' optionally includes an unsaturated bond; wherein R4, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocycl
  • a method of inhibiting TLR-mediated immunostimulatory signaling involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XIII, as provided above, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
  • the invention provides a method of affecting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XIII, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject.
  • the invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XIII, as provided above, to inhibit TLR-mediated immunostimulation in the subject.
  • the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject.
  • the method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula XIII, as provided above, to inhibit an immunostirnulatory nucleic acid-associated response in the subject.
  • A" is a substituted alkyl group selected from the group consisting of a cyclic amino group, an alkylamino group, a dialkylamino group, furyl, phenyl, thienyl, azabicyclooctyl, azabicycloheptyl, and any combination thereof.
  • the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group.
  • the subject is otherwise free of symptoms calling for treatment with a compound of Formula XIII.
  • the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand.
  • the method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XIV or Formula XV
  • Rl, R2, R3, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR.
  • the compound is any one of compounds 807, 1163, and 1367 listed in Table 17 below.
  • a method of inhibiting TLR-mediated immunostimulatory signaling involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XIV or Formula XV, as provided above, to inhibit TLR-rhediated immunostimulatory signaling in response to a ligand for the TLR.
  • the invention provides a method of affecting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XIV or Forrhula XV, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject.
  • the invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject.
  • the method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XIV or Formula XV, as provided above, to inhibit TLR-mediated immunostimulation in the subject.
  • the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject.
  • the method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula XIV or Formula XV, as provided above, to inhibit an immunostimulatory nucleic acid-associated response in the subject.
  • R2 is a substituted or unsubstituted phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group.
  • R2 is a phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group substituted with one or more substituent groups selected from the group consisting of an alkyl group, an alkoxy group, an alkoxyalkyl group, an ester group, an alkylamino group, a dialkylamino group, a cyclic amino group, a halogen atom, and any combination thereof.
  • R2 is a phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group substituted with one or more substituent groups selected from the group consisting of an alkylamino group, a dialkylamino group, and a cyclic amino group.
  • the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group.
  • R9 is a substituted alkyl group selected from the group consisting of a cyclic amino group, an alkylamino group, a dialkylamino group, furyl, phenyl, thienyl, azabicyclooctyl, azabicycloheptyl, and any combination thereof.
  • the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group.
  • each of R4 and R5 is hydrogen atom.
  • each of R5 and R8 is a hydrogen atom. In some embodiments each of R4, R5, and R8 is a hydrogen atom. In some embodiments RIO is a hydrogen atom.
  • the TLR is TLR9. In one embodiment the ligand for the TLR is an immunostimulatory nucleic acid. In one embodiment the immunostimulatory nucleic acid is a CpG nucleic acid. In each of the foregoing aspects of the invention, in one embodiment the TLR is
  • the ligand for the TLR is a natural ligand for TLR8. In one embodiment the ligand for the TLR is resiquimod (R848). In each of the foregoing aspects of the invention, in one embodiment the TLR is TLR7. In one embodiment the ligand for the TLR is a natural ligand for TLR7. In one embodiment the ligand for the TLR is R848. In each of the foregoing aspects of the invention, in one embodiment the TLR is TLR3. In one embodiment the ligand for the TLR is a double stranded RNA. In one embodiment the ligand for the TLR is poly(LC).
  • the present invention is based, in part, on the appreciation by the applicants of a potential connection between the antimalarial activity of known metabolites of chloroquine and the biological activity of related small molecules with respect to inhibition of TLR9.
  • the major metabolites are mono de-ethylated chloroquine and bis de ethylated chloroquine.
  • chloroquine and hydroxychloroquine derive a substantial portion of their efficacy from the long lived de-ethyl metabolite and that this relationship may carry over to the binding to TLR9. It turns out that chloroquine, hydroxychloroquine, mono de-ethylchloroqiliine, and bis de-ethylchloroquine have been examined by Macfarlane.
  • the IC50 of these compounds by 7 1 his assay were: chloroquine, 1.1x10 " M; hydroxychloroquine, 4.1x10 " M; mono de- ethylchloroquine, 7.08X10 '7 M; and bis de-ethylchloroquine, 18.6xlO '7 M. So by this data the loss of one ethyl group from chloroquine reduced the potency by about 7-fold and the loss of both ethyl groups reduced the potency by about 18-fold.
  • the present invention in certain aspects is related to compositions and methods involving certain 4-primary amino quinoline compounds which are useful for inhibiting TLR9 signaling and immune activation.
  • the present invention is also related to compositions and methods involving certain quinazoline compounds, some of which are structurally related to the 4-primary amino quinoline compounds of the invention, which are also useful for inhibiting TLR9 signaling and immune activation. It was surprisingly discovered according to the invention that the 4-primary amino quinoline compounds and the quinazoline compounds of the invention are highly and similarly active as inhibitors of TLR9 signaling activity.
  • the quinazoline molecules exhibit a significantly improved toxicity profile in vivo compared with the quinoline compounds.
  • the invention provides novel substituted 4-primary amino quinoline compositions having a structural Formula XVI
  • Formula XVI wherein X is absent or is an aryl, alkyl, heterocyclic, or styryl group; R t and R are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group, wherein Ri and R 2 optionally are combined to form ' a heterocycle; R 3 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein Ri and R 3 optionally are combined to form a heterocycle; Y is absent or is an oxygen atom, a sulfur atom, CRgR 9 , or NR 10 , where Rs, R 9 , and R 10 are each independently a hydrogen atom
  • R 5 and R 6 are each independently a halogen atom or an alkoxy group. In one embodiment R 5 and R_ are each independently a chlorine atom or a methoxy group.
  • X is absent or is an aryl group;
  • NRiR 2 is a heterocyclic amine or is NRg(CH 2 ) n NR 9 R 10 , wherein n is an integer from 2 to 6, inclusive, and R 8 , R , and R 10 are each independently a hydrogen atom or an alkyl group;
  • R 3 is a hydrogen atom;
  • Y is an aryl group or is NR ⁇ where R ⁇ is a hydrogen atom or an aryl or alkyl group;
  • L is absent or is a C 2 -C 6 alkyl group; and
  • t , R 5 , R_, and R 7 are each independently a hydrogen atom, a halogen atom, or an alkoxy group.
  • X is absent or is an aryl group;
  • NRs(CH 2 ) n NR 9 R 10 wherein n is an integer from 2 to 6, inclusive, R 8 is a hydrogen atom, and R 9 and R 10 are each independently an alkyl group; R 3 is a hydrogen atom; Y is NH; L is a C 2 -C 6 alkyl group; and Rj, R 5 , R_, and R are each independently a hydrogen atom, a halogen atom, or an alkoxy group.
  • X is a phenyl group;
  • NR t R 2 is attached to a para position of the phenyl group X; Y is NH; L is -(CH 2 ) n - where n is an integer between 2 and 6, inclusive; and each of R 3 , R 4 , R 5 , R and R is a hydrogen atom.
  • the composition is optionally in the form of a pharmaceutically acceptable hydrate or salt.
  • the invention provides a method for inhibiting signaling by a TLR.
  • the method according to this aspect of the invention involves contacting a cell expressing a functional TLR with an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII
  • X is absent or is a nitrogen, oxygen, or sulfur atom or an SO or SO 2 group
  • Ri is a hydrogen atom or a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group
  • R 2 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein R] and R 2 optionally are combined to form a heterocycle or carbocycle;
  • Y is absent or is an oxygen atom, a sulfur atom, CR Rs, or NR , where R , R 8 , and R 9 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or an SO
  • the invention provides a method for inhibiting signaling by a TLR.
  • the method according to this aspect of the invention involves contacting an immune cell expressing a functional TLR with (a) an effective amount of a TLR signal agonist to stimulate signaling by the TLR in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to inhibit signaling by the TLR in response to the TLR signal agonist compared with the signaling by the TLR in response to the TLR signal agonist in absence of the substituted 4-primary amino quinoline composition.
  • the TLR is TLR9 and the TLR signal agonist is a TLR9 signal agonist.
  • the TLR signal agonist in one embodiment is CpG DNA, which can be an oligodeoxynucleotide (ODN).
  • ODN oligodeoxynucleotide
  • the TLR signal agonist is an immune complex.
  • the TLR signal agonist is an immune complex that includes a nucleic acid.
  • the TLR signal agonist is an immune complex that includes a nucleic acid that is self-DNA.
  • the TLR signal agonist is an immune complex that includes a nucleic acid that is self-RNA.
  • the invention provides a method for inhibiting an immune response to an antigenic substance.
  • the method according to this aspect of the invention involves contacting an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the substituted 4-primary amino quinoline composition.
  • the immune response is an innate immune response.
  • the immune response includes an adaptive immune response.
  • a method of treating an autoimmune disorder in a subject involves the step of administering to a subject having an autoimmune disorder an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to treat the autoimmune disorder.
  • the autoimmune disorder is chosen from systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Sjogren's syndrome, polymyositis, vasculitis, Wegener's granulomatosis, sarcoidosis, ankylosing spondylitis, Reiter's syndrome, psoriatic arthritis, and Behcet's syndrome.
  • the autoimmune disease is systemic lupus erythematosus. In one particular embodiment the autoimmune disease is rheumatoid arthritis. In one embodiment the subject is a human. In one embodiment the autoimmune disorder is an immune complex associated disease.
  • the invention provides novel quinazoline compositions of the invention having a structural Formula XVIII
  • Fo ⁇ nula XVIII wherein X is absent or is an aryl, alkyl, heterocyclic or styryl group, provided that if X is a phenyl group, NR ⁇ R 2 is part if a heterocycle or is a diamine; R !
  • R are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein Ri and R 2 optionally are combined to form a heterocycle
  • Y is an oxygen atom, a sulfur atom, CR 9 R 10 , or NR ⁇ , where R 9 , R 10 , and R ⁇ are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R 9 , R 10 , and R ⁇ optionally is combined with R 3 or t to form a substituted or unsubstituted heterocycle
  • L is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group
  • R 3 and t are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R 3 and t optionally are combined to form a heterocycle
  • R 5 , Re, R 7 , and R 8 are each independently a hydrogen atom, a hal
  • R and R 7 are each independently a halogen atom or an alkoxy group.
  • R_ and R are each independently a chlorine atom or a methoxy group.
  • X is absent or is an aryl group;
  • NRiR 2 is a heterocyclic amine or NR (CH 2 ) n NR 10 R 11 , wherein n is an integer from 2 to 6, inclusive, and
  • R 9 , R 10 , and R ⁇ are each independently a hydrogen atom or ain alkyl group;
  • Y is an aryl group or is NR 12 where R 12 is a hydrogen atom or an aryl or alkyl group;
  • L is absent or is a C 2 -C 6 alkyl group;
  • R 3 and R t are each independently a hydrogen atom or an alkyl group, wherein R 3 and R 4 optionally are combined to form a heterocycle; and
  • R 5 , R_, R , and R 8 are each independently a hydrogen atom,
  • X is absent or is an aryl group
  • NRiR 2 is a substituted or unsubstituted piperazino or morpholino group or is NR (CH 2 ) n NR 1 oR ⁇ , wherein n is an integer from 2 to 6, inclusive, R 9 is a hydrogen atom, and R 10 and R ⁇ are each independently an alkyl group
  • Y is NH
  • L is a C 2 -C 6 alkyl group
  • R 3 and R 4 are each independently a hydrogen atom or an alkyl group, wherein R 3 and R t optionally are combined to form a heterocycle
  • R 5 , R_, R , and R 8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group.
  • X is an aryl group
  • NRiR 2 is substituted or unsubstituted piperazino or morpholino group
  • Y is NH
  • L is a C 2 -C 6 alkyl group
  • R 3 and t are each independently a methyl or ethyl group or R 3 and t optionally are combined to form a morpholino group
  • R 5 , R_, R , and R 8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group.
  • X is a phenyl group; NRiR 2 is N-methylpiperazine; Y is NH; L is -CH 2 CH 2 -; R 3 and t are each a methyl group; and R 5 , R , R , and R 8 are each a hydrogen atom.
  • X is a phenyl group; NRiR 2 is N-methylpiperazine; Y is NH; L is -CH 2 CH 2 -; R 3 and R t are combined as a morpholino group; and R 5 , Re, R , and Rs are each a hydrogen atom.
  • X is absent;
  • NR ⁇ is a substituted or unsubstituted piperazino or morpholino group;
  • Y is NH;
  • L is a C 2 -C 6 alkyl group;
  • R 3 and t are each independently a methyl or ethyl group or R 3 and t optionally are combined to form a morpholino group;
  • R 5 , R_, R , and R 8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group.
  • X is absent; NR t R is N-methylpiperazine; Y is NH; L is -CH 2 CH 2 -; R 3 and t are each a methyl group; R_ and R are each a methoxy group; and R 5 and R 8 are each a hydrogen atom.
  • X is absent; NRiR 2 is N-phenylpiperazine; Y is NH; L is -CH 2 CH 2 -; R 3 and R t are each a methyl group; Re and R are each a methoxy group; and R 5 and Rs are each a hydrogen atom.
  • the composition is optionally in the form of a pharmaceutically acceptable hydrate or salt.
  • the invention provides a method for inhibiting signaling by a TLR. The method according to this aspect of the invention involves contacting a cell expressing a functional TLR with an effective amount of quinazoline composition having structural Formula XIX
  • X is a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group, optionally attached to the quinazoline by a nitrogen, oxygen, or sulfur atom or by a SO or SO 2 group;
  • Y is absent or is an oxygen atom, a sulfur atom, CR R 10 , or NR ⁇ , where R 9 , R 10 , and R ⁇ are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R , R 10 , and R ⁇ optionally is combined with R 3 or R t to form a heterocycle;
  • L is absent or is a hydrogen atom, an alkyl or alkenyl group containing from 1 to 10 carbons, or an aryl group;
  • R 3 and R 4 are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R 3 and t optionally are combined to form a heterocycle; and R
  • the invention provides a method for inhibiting signaling by a TLR.
  • the method according to this aspect of the invention involves contacting an immune cell expressing a functional TLR with (a) an effective amount of a TLR signal agonist to stimulate signaling by the TLR in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having structural Formula XIX, as defined above, to inhibit signaling by the TLR in response to the TLR signal agonist compared with the signaling by the TLR in response to the TLR signal agonist in absence of the quinazoline composition.
  • the TLR is TLR9 and the TLR signal agonist is a TLR9 signal agonist.
  • the TLR signal agonist in one embodiment is CpG DNA, which can be an oligodeoxynucleotide (ODN).
  • ODN oligodeoxynucleotide
  • the TLR signal agonist is an immune complex that includes a nucleic acid.
  • the invention provides a method for inhibiting an immune response to an antigenic substance.
  • the method according to this aspect of the invention involves contacting an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having structural Formula XIX, as defined above, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the quinazoline composition.
  • the immune response is an innate immune response.
  • the immune response includes an adaptive immune response.
  • a method of treating an autoimmune disorder in a subject involves the step of administering to a subject having an autoimmune disorder an effective amount of a quinazoline composition having structural Formula XIX, as defined above, to treat the autoimmune disorder.
  • the autoimmune disorder is chosen from systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Sjogren's syndrome, polymyositis, vasculitis, Wegener' s granulomatosis, sarcoidosis, ankylosing spondylitis,
  • the autoimmune disease is systemic lupus erythematosus. In one particular embodiment the autoimmune disease is rheumatoid arthritis. In one embodiment the subject is a human. In one embodiment the autoimmune disorder is an immune complex associated disease, as described above. BRIEF DESCRIPTION OF THE DRAWINGS FIG.
  • FIG. 1 is a graph depicting the concentration-dependent inhibition of TLR9 signaling in response to CpG ODN 2006 (5*-TCGTCGTTTTGTCGTTTTGTCGTT-3'; SEQ ID NO:l) by compounds 613 (harmine hydrochloride), 878 (3-hydroxymethyl-beta-carboline, and 470 (norharman), molecules representative of compounds of Formula II.
  • FIG. 2 is a pair of graphs depicting results of an in vivo toxicity assay in mice for specific quinoline and quinazoline compounds.
  • FIG. 2A shows change in mean body weight (grams) and FIG 2B shows mean body weight (grams) over 5 days for mice treated with dimethysulfoxide (DMSO) control (closed circles), compound CMZ 203-43 (also referred to herein as 203-43) administered at 1 mg and 4 mg (open and closed triangles, respectively), compound CMZ 203-34 at 1 mg and 4 mg (open and closed diamonds, respectively), and compound CMZ 203-49 at 1 mg and 4 mg (open and closed inverted triangles, respectively).
  • N 3 for each treatment group.
  • the group receiving CMZ 203-43 at 4 mg were sick and had to be sacrificed on day 3.
  • FIG. 5 is a bar graph depicting in vivo inhibition of TNF- ⁇ induction (pg/ml) by CpG
  • ODN 2006 following pretreatment with PBS control or a quinoline compound (203-43) or various indicated quinazoline compounds (203-34, 203-49, or 203-51). N 5 for each group.
  • the present invention provides novel compositions and methods for inhibiting immune responses, including immune responses involved in clinical disorders characterized as autoimmune disorders and immune complex associated diseases.
  • novel compositions disclosed herein which include 4-primary amino quinolines and structurally similar quinazolines
  • the invention also provides methods for use of previously known compositions within these and other classes of compounds in inhibiting immune responses, including immune responses involved in clinical disorders characterized as autoimmune disorders and immune complex associated diseases.
  • the present invention is based in part on discoveries made by the applicants arising from screening a library of small molecule compounds in an in vitro assay of TLR9 signaling in response to immunostimulatory CpG oligodeoxynucleotides (ODN).
  • ODN immunostimulatory CpG oligodeoxynucleotides
  • a core structure including at least two rings, which can be fused or unfused, wherein at least one of the rings contains a nitrogen atom and/or has a side chain that contains a nitrogen atom. Certain of the molecules are effective even without presence of a nitrogen atom. It has been observed that fused three-ring structures such as quinacrine are typically an order of magnitude more potent as inhibitors of TLR-mediated immunostimulation than are fused two-ring structures such as chloroquine.
  • quinacrine is reported to have an EC 50 of about 8 nM, where EC 50 is the concentration required for half-maximal inhibition of CpG-ODN effect on thymidine uptake by WEHI 231 B-cells in the presence of anti-surface IgM.
  • U.S. Pat. No. 6,479,504 Bl It has now also been discovered according to the present invention that fused two-ring core structures to which are attached a nitrogen- containing ring substituent can be as potent as fused three-ring structures such as quinacrine. In particular, such compounds can exhibit EC 50 in the low nanomolar range.
  • the nitrogen-containing ring substituent is a C 2 -C 6 alkyl group terminated by a tertiary amine, such as methylethylamine, diethyl amine, dimethyl amine, or a ring containing at least one nitrogen.
  • a tertiary amine such as methylethylamine, diethyl amine, dimethyl amine, or a ring containing at least one nitrogen.
  • the phenothiazine chlorpromazine has been reported to inhibit messenger RNA expression for interleukin 2 (IL-2), tumor necrosis factor alpha (TNF- ⁇ ), and gamma interferon (IFN- ⁇ ). Schleuning MJ et al. (1989) Eur J Immunol 19: 1491-6. In a separate study, chlorpromazine was reported to depress contact hypersensitivity to dinitrochlorobenzene in guinea-pigs. Descotes J et al. (1982) J Neuroimmunol 2:21-5.
  • H1R histamine HI receptor
  • H2R histamine H2 receptor
  • H3R histamine H3 receptor
  • Pharmacologic inhibitors for these receptors include pyrilamine and tripelennamine (H1R); cimetidine, famotidine, and ranitidine (H2R); and thioperamide and clobenpropit (H3R).
  • H1R pyrilamine and tripelennamine
  • H2R cimetidine
  • ranitidine H2R
  • H3R thioperamide and clobenpropit
  • H1R " ⁇ ) mice T cells and B cells derived from HlR-deficient (H1R " ⁇ ) mice were reported to have normal responses to LPS. Banu et al. also reported that administration of the H2R-specific antagonist famotidine further depressed T-cell and B-cell antigen receptor-mediated responses in H1R "A mice. Jutel and coworkers recently reported that in their study of T cells, the Thl subset of CD4+ T cells preferentially express H1R, while Th2 cells preferentially express H2R.
  • adaptive immune response refers to any type of antigen- specific immune response. Adaptive immune responses, which are also known in the art as specific immune responses, involve lymphocytes are also characterized by immunological memory, whereby response to a second or subsequent exposure to antigen is more vigorous than the response to a first exposure to the antigen.
  • adaptive immune response encompasses both humoral (antibody) immunity and cell-mediated (cellular) immunity.
  • allergy refers to acquired hypersensitivity to a substance (allergen).
  • Allergic conditions include eczema, allergic rhinitis or coryza, hay fever, asthma, urticaria (hives) and food allergies, and other atopic conditions.
  • antigenic substance refers to any substance that induces an adaptive (specific) immune response.
  • An antigen typically is any substance that can be specifically bound by a T-cell antigen receptor, antibody, or B-cell antigen receptor.
  • Antigenic substances include, without limitation, peptides, proteins, carbohydrates, lipids, phospholipids, nucleic acids, autacoids, and hormones. Antigenic substances further specifically include antigens that are classified as allergens, cancer antigens, and microbial antigens.
  • autoimmune disease refers to a disorder of the respiratory system characterized by inflammation, narrowing of the airways and increased reactivity of the airways to inhaled agents. Asthma is frequently, although not exclusively associated with atopic or allergic symptoms. For example, asthma can be precipitated by exposure to an allergen, exposure to cold air, respiratory infection, and exertion.
  • autoimmune disease and, equivalently, “autoimmune disorder” and “ autoimmunity”, refer to immunologically mediated acute or chronic injury to a tissue or organ derived from the host. The terms encompass both cellular and antibody- mediated autoimmune phenomena, as well as organ-specific and organ-nonspecific autoimmunity.
  • Autoimmune diseases include insulin-dependent diabetes mellitus, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, atherosclerosis, and inflammatory bowel disease. Autoimmune diseases also include, without limitation, ankylosing spondylitis, autoimmune hemolytic anemia, Behcet's syndrome, Goodpasture's syndrome, Graves' disease, Guillain-Barre syndrome, Hashimoto's thyroiditis, idiopathic thrombocytopenia, myasthenia gravis, pernicious anemia, polyarteritis nodosa, polymyositis/dermatomyositis, primary biliary sclerosis, psoriasis, sarcoidosis, sclerosing cholangitis, Sjogren's syndrome, systemic sclerosis (scleroderma and CREST syndrome), Takayasu's arteritis, temporal arteritis, and Wege
  • Autoimmune diseases also include certain immune complex-associated diseases.
  • cancer and, equivalently, “tumor” refer to a condition in which abnormally replicating cells of host origin are present in a detectable amount in a subject.
  • the cancer can be a malignant or non-malignant cancer.
  • Cancers or tumors include but are not limited to biliary tract cancer; brain cancer; breast cancer; cervical cancer; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric (stomach) cancer; intraepithelial neoplasms; leukemias; lymphomas; liver cancer; lung cancer (e.g., small cell and non-small cell); melanoma; neuroblastomas; oral cancer; ovarian cancer; pancreatic cancer; prostate cancer; rectal cancer; renal (kidney) cancer; sarcomas; skin cancer; testicular cancer; thyroid cancer; as well as other carcinomas and sarcomas. Cancers can be primary or metastatic.
  • CpG DNA refers to an immunostimulatory nucleic acid which contains a cytosine-guanine (CG) dinucleotide, the C residue of which is unmethylated.
  • CG cytosine-guanine
  • the effects of CpG nucleic acids on immune modulation have been described extensively in U.S. patents such as U.S. Pat. Nos. 6,194,388; 6,207,646; 6,239,116; and 6,218,371, and published international patent applications, such as WO98/37919, WO98/40100, WO98/52581, and WO99/56755. The entire contents of each of these patents and published patent applications is hereby incorporated by reference.
  • CpG DNA includes both naturally occurring immunostimulatory nucleic acids, as found in bacterial DNA and plasmids, as well as synthetic oligodeoxynucleotides (ODN).
  • ODN synthetic oligodeoxynucleotides
  • the CpG DNA is a CpG ODN that has a base sequence provided by 5'-TCGTCGTTTTGTCGTTTTGTCGTT-3' (ODN 2006; SEQ ID NO: 1).
  • B-class CpG ODN such as ODN 2006 include the originally described immunostimulatory CpG ODN and characteristically activate B cells and NK cells but do not induce or only weakly induce expression of type I interferon (e.g., IFN- ⁇ ).
  • A- class CpG ODN described in published PCT international application WO 01/22990, incorporate a CpG motif, include a chimeric phosphodiester/phosphorothioate backbone, and characteristically activate NK cells and induce plasmacytoid dendritic cells to express large amounts of IFN- ⁇ but do not activate or only weakly activate B cells.
  • An example of an A- class CpG ODN is 5'-G*G*G_G_G_A_C_G_A_T_C_G_T_C_G*G*G*G*G-3' (ODN 2216, SEQ ID NO:2), wherein "*" represents phosphorothioate and "_" represents phosphodiester.
  • C-class CpG ODN incorporate a CpG, include a wholly phosphorothioate backbone, include a GC-rich palindromic or nearly-palindromic region, and are capable of both activating B cells and inducing expression of IFN- ⁇ .
  • C-class CpG ODN have been described, for example, in published U.S. patent application 2003/0148976.
  • An example of a C-class CpG ODN is 5'-TCGTCGTTTTCGGCGCGCGCCG-3' (ODN 2395; SEQ ID NO:3).
  • cytokine refers to any of a number of soluble proteins or glycoproteins that act on immune cells through specific receptors to affect the state of activation and function of the immune cells. Cytokines include interferons, interleukins, tumor necrosis factor, transforming growth factor beta, colony-stimulating factors (CSFs), chemokines, as well as others. Various cytokines affect innate immumty, acquired immunity, or both.
  • Cytokines specifically include, without limitation, IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12, IL-13, IL-18, TNF- ⁇ , TGF- ⁇ , granulocyte colony-stimulating factor (G-CSF), and granulocyte-macrophage colony-stimulating factor (GM-CSF).
  • Chemokines specifically include, without limitation, IL-8, IP- 10, 1-TAC, • RANTES, MlP-l ⁇ , MlP-l ⁇ , Gro- ⁇ , Gro- ⁇ , Gro- ⁇ , MCP-1, MCP-2, and MCP-3.
  • Thl cells are associated with IL-2, IL-3, IFN, GM-CSF and high levels of TNF- ⁇ .
  • Th2 cells are associated with IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-13, GM-CSF and low levels of TNF- ⁇ .
  • the Thl subset promotes both cell-mediated immunity and humoral immunity that is characterized by immunoglobulin class switching to IgG2ain mice. Thl responses can also be associated with delayed-type hypersensitivity and autoimmune disease.
  • the Th2 subset induces primarily humoral immunity and induces immunoglobulin class switching to IgE and IgGl in mice.
  • the antibody isotypes associated with Thl responses generally have good neutralizing and opsonizing capabilities, whereas those associated with Th2 responses are associated more with allergic responses.
  • Several factors have been shown to influence commitment to Thl or Th2 profiles.
  • the best characterized regulators are cytokines.
  • IL-12 and IFN- ⁇ are positive Thl and negative Th2 regulators.
  • IL-12 promotes IFN- ⁇ production, and IFN- ⁇ provides positive feedback for IL-12.
  • IL-4 and IL-10 appear to be required for the establishment of the Th2 cytokine profile and to down-regulate Thl cytokine production; the effects of IL-4 are in some cases dominant over those of IL-12.
  • IL-13 was shown to inhibit expression of inflammatory cytokines, including IL-12 and TNF- ⁇ by LPS-induced monocytes, in a way similar to IL-4.
  • "effective amount” refers to any amount that is necessary or sufficient for achieving or promoting a desired outcome. In some instances an effective amount is a therapeutically effective amount. A therapeutically effective amount is any amount that is necessary or sufficient for promoting or achieving a desired biological response in a subject.
  • graft rejection refers to immunologically mediated hyperacute, acute, or chronic injury to a tissue or organ derived from a source other than the host. The term thus encompasses both cellular and antibody-mediated rejection, as well as rejection of both allografts and xenografts.
  • immune cell refers to a cell belonging to the immune system.
  • Immune cells include T lymphocytes (T cells), B lymphocytes (B cells), natural killer (NK) cells, granulocytes, neutrophils, macrophages, monocytes, dendritic cells, and specialized forms of any of the foregoing, e.g., plasmacytoid dendritic cells, plasma cells, NKT, T helper, and cytotoxic T lymphocytes (CTL).
  • T cells T lymphocytes
  • B cells B lymphocytes
  • NK natural killer cells
  • granulocytes neutrophils
  • macrophages macrophages
  • monocytes monocytes
  • dendritic cells e.g., monocytes, dendritic cells
  • specialized forms of any of the foregoing e.g., plasmacytoid dendritic cells, plasma cells, NKT, T helper, and cytotoxic T lymphocytes (CTL).
  • CTL cytotoxic T lymphocytes
  • the term "immune complex comprising a nucleic acid” refers to any conjugate including an antibody and a nucleic acid-containing antigen specifically bound by the antibody.
  • the nucleic acid-containing antigen can include chromatin, ribosomes, small nuclear proteins, histones, nucleosomes, DNA, RNA, or any combination thereof.
  • the antibody can but need not necessarily bind specifically to a nucleic acid component of the nucleic acid-containing antigen.
  • immune complex-associated disease refers to any disease characterized by the production and/or tissue deposition of immune complexes, including, but not limited to systemic lupus erythematosus (SLE) and related connective tissue diseases, rheumatoid arthritis, hepatitis C- and hepatitis B-related immune complex disease (e.g., cryoglobulinemia), Behcet's syndrome, autoimmune glomerulonephritides, and vasculopathy associated with the presence of LDL/anti-LDL immune complexes.
  • SLE systemic lupus erythematosus
  • connective tissue diseases e.g., hepatitis C- and hepatitis B-related immune complex disease (e.g., cryoglobulinemia), Behcet's syndrome, autoimmune glomerulonephritides, and vasculopathy associated with the presence of LDL/anti-LDL immune complexes.
  • immunodeficiency refers to a disease or disorder in which the subject's immune system is not functioning in normal capacity or in which it would be useful to boost a subject's immune response for example to eliminate a tumor or cancer (e.g., tumors of the brain, lung (e.g., small cell and non-small cell), ovary, breast, prostate, colon, as well as other carcinomas and sarcomas) or an infection in a subject.
  • the immunodeficiency can be acquired or it can be congenital.
  • immunonostimulatory nucleic acid-associated response in a subject refers to a measurable response in a subject associated with administration to the subject of an immunostimulatory nucleic acid.
  • Such responses include, without limitation, elaboration of cytokines, chemokines, growth factors, or immunoglobulin; expression of immune cell surface activation markers; Thl/Th2 skewing; and clinical disease activity.
  • infection and, equivalently, "infectious disease” refer to a condition in which an infectious organism or agent is present in a detectable amount in the blood or in a normally sterile tissue or normally sterile compartment of a subject. Infectious organisms and agents include viruses, bacteria, fungi, and parasites. The terms encompass both acute and chronic infections, as well as sepsis.
  • the term “innate immune response” refers to any type of immune response to certain pathogen-associated molecular patterns (PAMPs).
  • Innate immunity which is also known in the art as natural or native immunity, involves principally neutrophils, granulocytes, mononuclear phagocytes, dendritic cells, NKT cells, and NK cells. Innate immune responses can include, without limitation, type I interferon production (e.g., IFN- ⁇ ), neutrophil activation, macrophage activation, phagocytosis, opsonization, complement activation, and any combination thereof.
  • type I interferon production e.g., IFN- ⁇
  • neutrophil activation e.g., macrophage activation, phagocytosis, opsonization, complement activation, and any combination thereof.
  • self-DNA refers to any DNA derived from the genome of a host subject.
  • self-DNA includes complementary DNA (cDNA) derived from a host subject. Self-DNA includes intact and degraded DNA.
  • self-RNA refers to any RNA derived from the genome of a host subject.
  • self-RNA is a messenger RNA (mRNA) derived from a host subject.
  • self-RNA includes ribosomal RNA (rRNA) derived from a host subject.
  • Self-RNA includes intact and degraded RNA.
  • the term "subject” refers to a vertebrate animal. In one embodiment the subject is a mammal. In one embodiment the subject is a human. In other embodiments the subject is a non-human vertebrate animal, including, without limitation, non-human primates, laboratory animals, livestock, domesticated animals, and non-domesticated animals.
  • subject having or at risk of developing TLR-mediated immunostimulation refers to a subject exposed to or at risk of exposure to a PAMP or other TLR ligand.
  • TLR Toll-like receptor
  • TLRl - TLR10 pathogen-associated molecular patterns
  • TLR polypeptides share a characteristic structure that includes an extracellular (extracytoplasmic) domain that has leucine-rich repeats, a transmembrane domain, and an intracellular (cytoplasmic) domain that is involved in TLR signaling.
  • TLRs include but are not limited to human TLRs. Nucleic acid and amino acid sequences for all ten currently known human TLRs are available from public databases such as GenBank. Similarly, nucleic acid and amino acid sequences for various TLRs from numerous non-human species are also available from public databases including GenBank.
  • nucleic acid and amino acid sequences for human TLR9 can be found as GenBank accession numbers AF245704 (coding region spanning nucleotides 145-3243) and AAF78037, respectively.
  • Nucleic acid and amino acid sequences for murine TLR9 can be found as GenBank accession numbers AF348140 (coding region spanning nucleotides 40-3138) and AAK29625, respectively.
  • the deduced human TLR9 protein contains 1,032 amino acids and shares an overall amino acid identity of 75.5% with mouse TLR9.
  • human TLR9 contains extracellular leucine-rich repeats (LRRs) and a cytoplasmic Toll interleukin- 1R (TIR) domain. It also has a signal peptide (residues 1-25) and a transmembrane domain (residues 819-836).
  • LRRs leucine-rich repeats
  • TIR Toll interleukin- 1R
  • hTLR8 Nucleic acid and amino acid sequences for human TLR8 (hTLR8) can be found as GenBank accession numbers AF245703 (coding region spanning nucleotides 49-3174) and AAF78036, respectively.
  • Nucleic acid and amino acid sequences for murine TLR8 can be found as GenBank accession numbers AY035890 (coding region spanning nucleotides 59-3157) and AAK62677, respectively.
  • Nucleic acid and amino acid sequences for human TLR7 can be found as GenBank accession numbers AF240467 (coding region spanning nucleotides 135-3285) and AAF60188, respectively.
  • Nucleic acid and amino acid sequences for murine TLR7 (mTLR7) can be found as GenBank accession numbers AY035889 (coding region spanning nucleotides 49-3201) and AAK62676, respectively.
  • Nucleic acid and amino acid sequences for human TLR3 can be found as GenBank accession numbers NM_003265 (coding region spanning nucleotides 102-2816) and NP_003256, respectively.
  • Nucleic acid and amino acid sequences for murine TLR3 can be found as GenBank accession numbers AF355152 (coding region spanning nucleotides 44-2761 ) and AAK26117, respectively.
  • hTLRl is ubiquitously expressed, hTLR2, hTLR4 and hTLR5 are present in monocytes, polymorphonuclear phagocytes, and dendritic cells. Muzio M et al. (2000) J Leukoc Biol 67:450-6.
  • TLR7 and hTLR9 are present in human B cells.
  • Human TLR7 and hTLR9 are present in plasmacytoid dendritic cells (pDCs), while myeloid dendritic cells express hTLR7 and hTLR8 but not hTLR9.
  • IL-1R pro-inflammatory interleukin- 1 receptor
  • TLRs share homologies in their cytoplasmic domains called Toll/IL-IR homology (TIR) domains.
  • TIR Toll/IL-IR homology
  • MyD88 is believed to act as an adapter molecule between the TIR domain of a TLR or IL-1R and IRAK (which includes at least any one of IRAK- 1, IRAK-2, IRAK-4, and IRAK-M).
  • MyD88 includes a C-terminal Toll homology domain and an N-terminal death domain.
  • the Toll homology domain of MyD88 binds the TIR domain of TLR or IL- 1R, and the death domain of MyD88 binds the death domain of the serine kinase IRAK.
  • IRAK interacts with TRAF6, which acts as an entryway into at least two pathways, one leading to activation of the transcription factor NF- ⁇ B and another leading to activation of Jun and Fos, members of the activator protein- 1 (AP-1) transcription factor family.
  • Activation of NF- ⁇ B involves the activation of TAK-1, a member of the MAP 3 kinase (MAPK) family, and I ⁇ B kinases.
  • the I ⁇ B kinases phosphorylate I ⁇ B, leading to its degradation and the translocation of NF- ⁇ B to the nucleus.
  • MAPKKs MAP kinase kinases
  • ERK p38
  • JNK/SAPK MAP kinase kinases
  • TLR ligand and, equivalently, "ligand for a TLR” and “TLR signaling agonist”, refer to a molecule, other than a small molecule according to Formula I-XIX described herein or a 4-primary amino quinoline or quinazoline molecule according to the invention, that interacts, directly or indirectly, with a TLR through a TLR domain other than a TIR domain and induces TLR-mediated signaling.
  • a TLR ligand is a natural ligand, i.e., a TLR ligand that is found in nature.
  • a TLR ligand refers to a molecule other than a natural ligand of a TLR, e.g., a molecule prepared by human activity.
  • the TLR is TLR9 and the TLR signal agonist is a CpG nucleic acid.
  • Ligands for many but not all of the TLRs have been described. For instance, it has been reported that TLR2 signals in response to peptidoglycan and lipopeptides. Yoshimura A et al. (1999) J Immunol 163:1-5; Brightbill HD et al. (1999) Science 285:732-6; Aliprantis AO et al.
  • TLR4 has been reported to signal in response to lipopolysaccharide (LPS). Hoshino K et al. (1999) J Immunol 162:3749-52; Poltorak A et al. (1998) Science 282:2085-8; Medzhitov R et al. (1997) Nature 388:394-7. Bacterial flagellin has been reported to be a natural ligand for TLR5. Hayashi F et al. (2001) Nature 410:1099- 1103.
  • TLR6 in conjunction with TLR2, has been reported to signal in response to proteoglycan.
  • TLR9 is a receptor for CpG DNA.
  • CpG DNA which includes bacterial DNA and synthetic DNA with CG dinucleotides in which cytosine is unmethylated, is described in greater detail elsewhere herein.
  • Imidazoquinolines are potent synthetic activators of immune cells with antiviral and antitumor properties.
  • TNF tumor necrosis factor
  • IL-12 interleukin- 12
  • Macrophages from mice deficient in TLR7 but not other TLRs produced no detectable cytokines in response to these imidazoquinolines.
  • the imidazoquinolines induced dose-dependent proliferation of splenic B cells and the activation of intracellular signaling cascades in cells from wildtype but not TLR7 -/- mice.
  • Luciferase analysis established that expression of human TLR7, but not TLR2 or TLR4, in human embryonic kidney cells results in NF- ⁇ B activation in response to resiquimod.
  • the findings of Hemmi et al. thus s ⁇ ggested that these imidazoquinoline compounds are non-natural ligands of TLR7 that jean induce signaling through TLR7.
  • R848 is also a ligand for human TLR8.
  • TLR3 ligands of TLR3 include poly( C) and double-stranded R ⁇ A (dsR ⁇ A).
  • poly(LC) and double-stranded R A (dsR ⁇ A) are classified as oligonucleotide molecules.
  • Alexopoulou et al. By stimulating kidney cells expressing one of a range of TLRs with poly(I:C), Alexopoulou et al. reported that only cells expressing TLR3 respond by activating ⁇ F- ⁇ B. Alexopoulou L et al. (2001) Nature 413: 732-8. Alexopoulou et al.
  • TLR3 '7 cells responded equivalently to wildtype cells in response to lipopolysaccharide, peptidoglycan, and CpG dinucleotides.
  • MyD88 _/" cells indicated that this adaptor protein is involved in dsRNA-induced production of cytokines and proliferative responses, although activation of NF- ⁇ B and MAP kinases are not affected, indicating distinct pathways for these cellular responses. Alexopoulou et al.
  • TLR3 may have a role in host defense against viruses.
  • a "cell expressing a TLR” refers to any cell which expresses, either naturally or artificially, a functional TLR.
  • a functional TLR is a full-length TLR protein or a fragment thereof capable of inducing a signal in response to interaction with its ligand.
  • the functional TLR will include at least a TLR ligand-binding fragment of the extracellular domain of the full-length TLR and at least a fragment of a TIR domain capable of interacting with another Toll homology domain-containing polypeptide, e.g., lv_yD88.
  • the functional TLR is a full-length TLR selected from TLRl , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, and TLR10.
  • the functional TLR is naturally expressed by a cell.
  • the cell naturally expresses functional TLR and is an isolated cell from human multiple myeloma cell line RPMI 8226 (ATCC CCL-155). This cell line was established from the peripheral blood of a 61 year old man at the time of diagnosis of multiple myeloma (IgG lambda type). Matsuoka Y et al. (1967) Proc Soc Exp Biol Med 125:1246-50.
  • RPMI 8226 was previously reported as responsive to CpG nucleic acids as evidenced by the induction of IL-6 protein and IL-12p40 mRNA. Takeshita F et al. (2000) Eur J Immunol 30:108-16; Takeshita F et al. (2000) Eur J Immunol 30:1967-76. Takeshita et al. used the cell line solely to study promoter constructs in order to identify transcription factor binding sites important for CpG nucleic acid signaling. It is now known that RPMI 8226 cells secrete a number of other chemokines and cytokines including IL-8, IL-10 and IP- 10 in response to immunostimulatory nucleic acids.
  • this cell line expresses TLR9, through which immunostimulatory nucleic acids such as for example CpG nucleic acids mediate their effects, it is a suitable cell line for use in the methods of the invention relating to CpG nucleic acids as reference and test compounds, as well as to other TLR9 ligands.
  • PBMCs peripheral blood mononuclear cells
  • the RPMI 8226 cell line has been observed to upregulate its cell surface expression of markers such as CD71, CD86 and HLA-DR in response to CpG nucleic acid exposure. This has been observed by flow cytometric analysis of the cell line.
  • the methods provided herein can be structured to use appropriately selected cell surface marker expression as a readout, in addition to or in place of chemokine or cytokine production or other readouts described elsewhere herein.
  • the RPMI 8226 cell line has also been found to respond to certain small molecules including imidazoquinoline compounds. For example, incubation of RPMI 8226 cells with the imidazoquinoline compound R848 (resiquimod) induces IL-8, IL-10, and IP-10 production. It has recently been reported that R848 mediates its immunostimulatory effects through TLR7 and TLR8.
  • the ability of RPMI 8226 to respond to R848 suggests that the RPMI 8226 cell line also expresses TLR7, as previously reported for normal human B cells.
  • the RPMI cell line can be used in unmodified form or in a modified form.
  • the RPMI 8226 cell is transfected with a reporter construct.
  • the cell is stably transfected with the reporter construct.
  • the reporter construct generally includes a promoter, a coding sequence and a polyadenylation signal.
  • the coding sequence can include a reporter sequence selected from the group consisting of an enzyme (e.g., luciferase, alkaline phosphatase, beta-galactosidase, chloramphenicol acetyltransferase (CAT), secreted alkaline phosphatase, etc.), a bioluminescence marker (e.g., green fluorescent protein (GFP, U.S. Pat. No. 5,491,084), etc.), a surface-expressed molecule (e.g., CD25), a secreted molecule (e.g., IL-8, IL-12 p40, TNF- ⁇ , etc.), and other detectable protein products known to those of skill in the art.
  • an enzyme e.g., luciferase, alkaline phosphatase, beta-galactosidase, chloramphenicol acetyltransferase (CAT), secreted alkaline phosphatase, etc.
  • the coding sequence encodes a protein having a level or an activity tha is quantifiable.
  • the functional TLR is artificially expressed (including over- expressed) by a cell, for example by introduction into the "cell of an expression vector bearing a coding sequence for the functional TLR wherein the coding sequence is operably linked to a gene expression sequence.
  • a coding sequence and the gene expression sequence are said to be operably linked when they are covalently linked in such a way as to place the expression or transcription and/or translation of the coding sequence under the influence or control of the gene expression sequence.
  • Two DNA sequences are said to be operably linked if induction of a promoter in the 5' gene expression sequence results in the transcription of the coding sequence and if the nature of the linkage between the two DNA sequences does not (1) result in the introduction of a frame-shift mutation, (2) interfere with the ability of the promoter region to direct the transcription of the coding sequence, or (3) interfere with the ability of the corresponding RNA transcript to be translated into a protein.
  • a gene expression sequence would be operably linked to a coding sequence if the gene expression sequence were capable of effecting transcription of that coding sequence such that the resulting transcript is translated into the desired protein or polypeptide.
  • a coding sequence refers to a nucleic acid sequence coding for a functional TLR.
  • a coding sequence refers to a nucleic acid sequence coding for a reporter.
  • a cell that artificially expresses a functional TLR can be a cell that does not express the functional TLR but for the TLR expression vector.
  • human 293 fibroblasts (ATCC CRL-1573) do not express TLR3, TLR7, TLR8, or TLR9.
  • suitable expression vector or vectors
  • a cell that artificially expresses a functional TLR can be a cell that expresses the functional TLR at a significantly higher level with the TLR expression vector than it does without the TLR expression vector.
  • a cell that artificially expresses a functional TLR is preferably a stably transfected cell that expresses the function ⁇ l TLR.
  • Such a cell can also be stably transfected with a suitable reporter construct.
  • TLR-mediated signaling refers to any portion of the intracellular signal transduction pathway involving interaction of a TLR with a suitable TLR ligand and engagement by the TLR of MyD88 or any aspect downstream of MyD88 engagement, described above in relation to the structure and function of the TIR domain of a TLR.
  • TLR-mediated immunostimulatory signaling refers to TLR- mediated signaling that results in a measurable immunostimulatory response. This term applies to such signaling both in vitro and in vivo.
  • TLR-mediated immunostimulation in a subject refers to TLR- mediated immunostimulatory signaling as it applies in vivo.
  • treat as used in reference to a disorder, disease, or condition means to intervene in such disorder, disease, or condition so as to prevent or slow the development of, to prevent, slow or halt the progression of, or to eliminate the disorder, disease, or condition.
  • Immunostimulatory nucleic acid refers to a nucleic acid molecule that, when contacted with a cell of the immune system, induces the cell of the immune system to become activated. Immune cell activation can be determined using any suitable means known to one of skill in the art, including but not limited to measurement of stimulated growth, proliferation, differentiation, migration, transcription or expression of gene products that are expressed or secreted in association with immune activation, activity of intracellular signaling pathways, and the like.
  • markers of immune cell activation include, without limitation, secretion of cytokines (e.g., IL-6), secretion of immunoglobulin (e.g., IgG), expression of cell surface antigens (e.g., CD86), induction of cytolytic activity, and induction and nuclear translocation of transcription factors (e.g., NF- ⁇ B).
  • cytokines e.g., IL-6
  • immunoglobulin e.g., IgG
  • CD86 cell surface antigens
  • induction of cytolytic activity e.g., NF- ⁇ B
  • a "nucleic acid” as used herein with respect to the methods of the invention shall refer to any polymer of two or more individual nucleoside or nucleotide units. Nucleic acids can be single- or double-stranded.
  • individual nucleoside or nucleotide units will include any one or combination of deoxyribonucleosides, ribonucleosides, deoxyribonucleotides, and ribonucleotides.
  • the individual nucleotide or nucleoside units of the nucleic acid can be naturally occurring or not naturally occurring.
  • the individual nucleotide units can include deoxyadenosine, deoxycytidine, deoxyguanosine, thymidine, and uracil.
  • individual nucleosides also include synthetic nucleosides having modified base moieties and/or modified sugar moieties, e.g., as described in Uhlmann E et al. (1990) Chem Rev 90:543-84.
  • the linkages between individual nucleotide or nucleoside units can be naturally occurring or not naturally occurring.
  • the linkages can be phosphodiester, phosphorothioate, phosphorodithioate, phosphoramidate, as well as peptide linkages and other covalent linkages, known in the art, suitable for joining adjacent nucleoside or nucleotide units.
  • Immunostimulatory nucleic acids typically range in size from 3-4 units to a few tens of units, e.g., 18-40 units, although longer nucleic acids are also contemplated by the invention.
  • the nucleic acids are oligonucleotides made up of 2 to about 100 nucleotides, and more typically 4 to about 40 nucleotides. Oligonucleotides composed exclusively of deoxyribonucleotides are termed oligodeoriboxynucleotides or, equivalently, oligodeoxynucleotides (ODN).
  • An immunostimulatory nucleic acid includes any of a number of different types of immunostimulatory nucleic acids, including specifically immunostimulatory CpG nucleic acids (CpG nucleic acids), including but not limited to types A, B, and C; and immunostimulatory non-CpG nucleic acids, including without limitation methylated CpG nucleic acids, T-rich nucleic acids, and poly-G nucleic acids. Certain of these various classes of immunostimulatory nucleic acids can coexist in a given nucleic acid molecule.
  • CpG nucleic acid and, equivalently, “CpG ODN” refer to an immunostimulatory nucleic acid which contains a cytosine-guanine (CG) dinucleotide, the C residue of which is unmethylated.
  • CG cytosine-guanine
  • the effects of CpG nucleic acids on immune modulation have been described extensively in U.S. patents such as U.S. Pat. Nos. 6,194,388; 6,207,646; 6,218,371; and 6,239,116, and published international patent applications, such as WO 98/37919, WO 98/40100, WO 98/52581, and WO 99/56755.
  • the entire immunostimulatory nucleic acid can be unmethylated or portions may be unmethylated but at least the C of the 5'-CG-3' must be unmethylated.
  • the CpG nucleic acid sequences of the invention include those broadly described above as well as disclosed in U.S. Pat. Nos. 6,207,646 B 1 and 6,239, 116 B 1.
  • the CpG nucleic acid has a base sequence provided by 5'-TCGTCGTTTTGTCGTTTTGTCGTT-3' (ODN 2006; SEQ ID NO:l).
  • Type B CpG nucleic acids such as ODN 2006 include the earliest described CpG nucleic acids and characteristically activate B cells but do not induce or only weakly induce expression of IFN- ⁇ .
  • Type A CpG nucleic acids described in published international application PCT/US00/26527 (WO 01/22990), incorporate a CpG motif, include a hybrid phosphodiester/phosphorothioate backbone, and characteristically induce plasmacytoid dendritic cells to express large amounts of IFN- ⁇ but do not activate or only weakly activate B cells.
  • Type C oligonucleotides incorporate a CpG, include a chimeric backbone, include a GC-rich palindromic or nearly-palindromic region, and are capable of both activating B cells and inducing expression of IFN- ⁇ . These have been described, for example, in published U.S. patent application 2003/0148976.
  • a non-CpG nucleic acid is used.
  • a non-CpG nucleic acid is an immunostimulatory nucleic acid which either does not have a CpG motif in its sequence, or has a CpG motif which contains a methylated C residue.
  • the non-CpG nucleic acid may still be immunostimulatory by virtue of its having other immunostimulatory motifs such as those described herein and known in the art.
  • the non-CpG nucleic acid is a methylated CpG nucleic acid. In som
  • non-CpG nucleic acid is a methylated CpG nucleic acid having a base sequence provided by 5'-TZGTZGTTTTGTZGTTTTGTZGTT-3' (ODN 2117; SEQ ID NO:4, wherein Z represents 5-methylcytidine).
  • Non-CpG nucleic acids include T-rich immunostimulatory nucleic acids.
  • the T-rich immunostimulatory nucleic acids include those disclosed in published PCT patent application PCT/USOO/26383, the entire contents of which are incorporated herein by reference. In some embodiments, T-rich nucleic acids 24 bases in length are used.
  • a T-rich nucleic acid is a nucleic acid which includes at least one poly T sequence and/or which has a nucleotide composition of greater than 25% T nucleotide residues.
  • a nucleic acid having a poly-T sequence includes at least four Ts in a row, such as 5'-TTTT-3'.
  • the T- rich nucleic acid includes more than one poly T sequence.
  • the T- rich nucleic acid may have 2, 3, 4, or more poly T sequences, such as ODN 2006.
  • Non-CpG nucleic acids also include poly-G immunostimulatory nucleic acids. A variety of references describe the immunostimulatory properties of poly-G nucleic acids. Pisetsky DS et al.
  • immunostimulatory nucleic acids of the invention can also be those which do not possess CpG, methylated CpG, T-rich, or poly-G motifs.
  • Exemplary immunostimulatory nucleic acid sequences further include but are not limited to those immunostimulatory sequences described and listed in published PCT patent application WO 01/22972.
  • the invention provides novel compounds that fall within Formula VI and Formula VII as disclosed herein.
  • These compounds include various diarylmethane TLR9 antagonists denoted herein as CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91. Syntheses for each of these compounds are provided in Examples 12-17, respectively. Similar to other compounds of Formula VI and Formula VII, these specific compounds were tested in vitro and found to inhibit TLR9 signaling. See Example 18.
  • CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91 are believed to be useful in the methods of the invention. More specifically, in one aspect the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand.
  • the method includes the step of contacting a cell expressing a TLR with an effective amount of any one or combination of compounds CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR.
  • the invention provides a method of inhibiting TLR-mediated signaling in response to a TLR ligand.
  • the method includes the step of contacting a cell expressing a TLR with an effective amount of any one or combination of compounds CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
  • the invention provides a method of affecting TLR-mediated immunostimulation in a subject.
  • the method includes the step of administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of any one or combination of compounds CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91, to inhibit or promote TLR- mediated immunostimulation in the subject.
  • the invention provides a method of inhibiting TLR-mediated immunostimulation in a subject.
  • the method includes the step of administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of any one or combination of compounds CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91, to inhibit TLR-mediated immunostimulation in the subject.
  • the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject.
  • the method according to this aspect includes the step of administering to a subject in need of such treatment an effective amount of any one or combination of compounds CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91, to inhibit an immunostimulatory nucleic acid-associated response in the subject.
  • the invention provides novel substituted 4-primary amino quinoline compositions. As described further below, these compositions and other substituted 4-primary amino quinoline compositions have been discovered to be useful in methods for inhibiting an immune response, both in vitro and in vivo, including methods for treating immune complex associated diseases and autoimmune disorders.
  • novel substituted 4-primary amino quinoline compositions of the invention will also be useful for prevention and treatment of malaria, as well as for treatment of other diseases which have been described to be responsive to treatment with chloroquines.
  • novel substituted 4-primary amino quinoline compositions of the invention have a structural Formula XVI
  • R ! and R 2 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group, wherein R ⁇ and R optionally are combined to form a heterocycle;
  • R 3 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein R ⁇ and R 3 optionally are combined to form a heterocycle or a carbocycle;
  • Y is absent or is an oxygen atom, a sulfur atom, CRgR , or NRio, where R 8 , R 9 , and R 10
  • R 5 and Rg are each independently a halogen atom or an alkoxy group. In one embodiment R 5 and Rg are each independently a chlorine atom or a methoxy group.
  • X is absent or is an aryl group;
  • NR]R 2 is a heterocyclic amine or is NR 8 (CH 2 ) n NR R ⁇ o, wherein n is an integer from 2 to 6, inclusive, and Rg, R , and R 10 are each independently a hydrogen atom or an alkyl group;
  • R 3 is a hydrogen atom;
  • Y is an aryl group or is NR ⁇ where R ⁇ is a hydrogen atom or an aryl or alkyl group;
  • L is absent or is a C -C 6 alkyl group; and
  • Rt, R 5 , Rg, and R are each independently a hydrogen atom, a halogen atom, or an alkoxy group.
  • X is absent or is an aryl group;
  • NR t R 2 is a substituted or unsubstituted piperazino or morpholino group or is wherein n is an integer from 2 to 6, inclusive, R 8 is a hydrogen atom, and R and R 10 are each independently an alkyl group;
  • R 3 is a hydrogen atom;
  • Y is NH;
  • L is a C 2 -Cg alkyl group; and
  • R t , R 5 , R_, and R 7 are each independently a hydrogen atom, a halogen atom, or an alkoxy group.
  • X is a phenyl group;
  • NR t R 2 is attached to a para position of the phenyl group X; Y is NH; L is -(CH 2 ) n - where n is an integer between 2 and 6, inclusive; and each of R 3 , R t , R , R ⁇ 5 , and R 7 is a hydrogen atom.
  • the composition is optionally in the form of a pharmaceutically acceptable hydrate or salt.
  • Representative, non-limiting examples of substituted 4-primary amino quinoline compositions of Formula XVI of the invention are compounds 101-104, 106-109, 111, 113- 116, and 118-119, presented in Table 1.
  • the invention provides a method for inhibiting signaling by a TLR.
  • the method according to this aspect of the invention involves contacting a cell expressing a functional TLR with an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII
  • X is absent or is a nitrogen, oxygen, or sulfur atom or an SO or SO 2 group
  • Ri is a hydrogen atom or a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group
  • R 2 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein R] and R 2 optionally are combined to form a heterocycle or carbocycle;
  • Y is absent or is an oxygen atom, a sulfur atom, CR Rs, or NR , where R 7 , R 8 , and R 9 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl,
  • the substituted 4-primary amino quinoline composition in this and all other aspects of the invention involving the use of a substituted 4-primary amino quinoline composition can be in the form a hydrate or pharmaceutically acceptable salt.
  • the method according to this aspect of the invention can be performed in vitro or it can be performed in vivo.
  • the cell expressing the functional TLR can, but need not necessarily, be an immune cell.
  • the TLR is TLR9 and the method is thus a method for inhibiting intracellular signaling by TLR9.
  • the invention provides a method for inhibiting signaling by a TLR.
  • the method according to this aspect of the invention involves contacting an immune cell expressing a functional TLR with (a) an effective amount of a TLR signal agonist to stimulate signaling by the TLR in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to inhibit signaling by the TLR in response to the TLR signal agonist compared with the signaling by the TLR in response to the TLR signal agonist in absence of the substituted 4-primary amino quinoline composition.
  • the substituted 4-primary amino quinoline composition in this aspect of the invention can be in the form a hydrate or pharmaceutically acceptable salt.
  • the method according to this aspect of the invention can be performed in vitro or it can be performed in vivo.
  • the TLR is TLR9 and the TLR signal agonist is a TLR9 signal agonist, and the method in one embodiment is thus a method of inhibiting intracellular signaling by TLR9 in response to a TLR9 signal agonist.
  • the TLR signal agonist in one embodiment is CpG DNA, for example a CpG ODN such as ODN 2006.
  • the CpG ODN can belong to any class of CpG ODN, including A-class (e.g., ODN 2216), B-class (e.g., ODN 2006), or C-class (e.g., ODN 2395).
  • the TLR signal agonist is an immune complex that includes a nucleic acid. Immune complexes that include a nucleic acid are known by those of skill in the art to include immunoglobulin complexed with nucleic acids that are either naked or, more commonly, that are associated with proteins or other non-nucleic acid components.
  • the nucleic acids that are associated with proteins or other non-nucleic acid components can include, for example, chromatin, ribosomes, small nuclear proteins, ribonuclear proteins (RNP), histones, nucleosomal protein-DNA complexes, and nucleosomes.
  • RNP ribonuclear proteins
  • nucleosomal protein-DNA complexes and nucleosomes.
  • clinically important antibodies specific for nucleic acids and for nucleic acid-containing material include antinuclear antibodies (ANA), anti-dsDNA, anti-ssDNA, anti-Sm, anti-RNP, anti-Ro (SS-A), anti-La (SS-B), and antihistone antibodies.
  • Immune complexes that include a nucleic acid include, without limitation, immunoglobulin complexed with DNA, including specifically double-stranded DNA, and immunoglobulin complexed with nucleosomal material, both characteristic of systemic lupus erythematosus, and immunoglobulin complexed with RNA.
  • the invention provides a method for inhibiting an immune response to an antigenic substance.
  • the method according to this aspect of the invention involves contacting an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the substituted 4-primary amino quinoline composition.
  • the substituted 4-primary amino quinoline composition in this aspect of the invention can be in the form a hydrate or pharmaceutically acceptable salt.
  • the method according to this aspect of the invention can be performed in vitro or it can be performed in vivo.
  • the immune response is an innate immune response. In one embodiment the immune response includes an adaptive immune response. ; In one embodiment the method involves contacting, in a subject, an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response in the subject to the antigenic substance in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to inhibit an immune response in the subject to the antigenic substance compared with the immune response to the antigenic substance in absence of the substituted 4-primary amino quinoline composition. In one embodiment the step of contacting an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response in the subject to the antigenic substance in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as
  • Toll-like receptor with an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a substituted 4-primary amino quinoline composition involves the active step of administering an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a substituted 4-primary amino quinoline composition.
  • the antigenic substance can be administered using any effective route or means for administering the antigenic substance to effect the contacting.
  • the administering can be by local or systemic injection, inhalation, oral ingestion, topical administration, mucosal administration, or any combination thereof.
  • the step of contacting an immune cell expressing a functional Toll-like receptor with an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a substituted 4-primary amino quinoline composition is passive.
  • the immune cell is already in contact or has already been in contact with an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance, at the time of or prior to the step of contacting the immune cell expressing a functional Toll-like receptor with an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the substituted 4-primary amino quinoline composition.
  • the antigenic substance can be an allergen.
  • An allergen is a substance that can induce an allergic or asthmatic response in a susceptible subject.
  • the list of allergens is enormous and can include pollens, insect venoms, animal dander, dust, fungal spores and drugs (e.g., penicillin).
  • Examples of natural animal and plant allergens include proteins specific to the following genera: Canis (Ca is familiaris); Dermatophagoides (e.g., Dermatophagoides farinae); Felis (Felis domesticus); Ambrosia ⁇ Ambrosia artemiisfolia); Lolium (e.g., Lolium perenne or Lolium multiflorum); Cryptomeria (Cryptomeriajaponicd); Alternaria (Alternaria alternata); Alder; Alnus (Alnus gultinosa); Betula (Betula verrucosd); Quercus (Quercus alba); Olea (Olea europa); Artemisia (Artemisia vulgaris); Plantago (e.g., Plantago lanceolata); Parietaria (e.g., Parietaria officinalis or Par ietaria judaica); Blattella (e.g., Blattella germanica); Apis
  • allergy refers to acquired hypersensitivity to a substance (allergen).
  • An "allergic reaction” is the response of an immune system to an allergen in a subject allergic to the allergen. Allergic conditions include eczema, allergic rhinitis or coryza, hay fever, bronchial asthma, urticaria (hives) and food allergies, and other atopic conditions.
  • the antigenic substance can be an antigen that is or is derived from an infectious microbial agent, including a bacterium, a virus, a fungus, or a parasite. Examples of infectious bacteria include: Helicobacter pyloris, Borrelia burgdorferi, Legionella pneumophilia, Mycobacteria sps (such as. M.
  • tuberculosis M. avium, M. intracellulare, M. kansasii, and M. gordonae
  • Staphylococcus aureus Neisseria gonorrhoeae, Neisseria meningitidis, Listeria monocytogenes, Streptococcus pyogenes (Group A Streptococcus), Streptococcus agalactiae (Group B Streptococcus), Streptococcus (viridans group), Streptococcus faecalis, Streptococcus bovis, Streptococcus (anaerobic sps.), Streptococcus pneumoniae, pathogenic Campylobacter sp., Enterococcus sp., Haemophilus influenzae, Bacillus anthracis, Corynebacterium diphtheriae, Corynebacterium sp., Erysipelo
  • Retro viridae including but not limited to human immunodeficiency virus (HIV)
  • Picornaviridae for example, polio viruses, hepatitis A virus; enteroviruses, human coxsackie viruses, rhinoviruses, echoviruses); Calciviridae (such as strains that cause gastroenteritis); Togaviridae (for example, equine encephalitis viruses, rubella viruses); Flaviviridae (for example, dengue viruses, encephalitis viruses, yellow fever viruses); Coronaviridae (for example, coronaviruses); Rhabdoviridae (for example, vesicular stomatitis viruses, rabies viruses); Filoviridae (for example, ebola viruses); Paramyxoviridae (for example, parainfluenza viruses, mumps virus, measles virus, respiratory syncytial virus); Orthomyxoviridae (for example, influenza viruses); Buny
  • infectious fungi examples include, but are not limited to, Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis, and Candida albicans.
  • the antigenic substance can be a cancer antigen.
  • a cancer antigen as used herein is a compound, such as a peptide or protein, associated with a tumor or cancer cell surface and which is capable of provoking an immune response when expressed on the surface of an antigen-presenting cell in the context of a major histocompatibility complex (MHC) molecule.
  • MHC major histocompatibility complex
  • Cancer antigens can be prepared from cancer cells either by preparing crude extracts of cancer cells, for example, as described in Cohen PA et al.
  • Cancer antigens include but are not limited to antigens that are recombinantly expressed, an immunogenic portion thereof, or a whole tumor or cancer cell. Such antigens can be isolated or prepared recombinantly or by any other means known in the art.
  • cancer antigen and “tumor antigen” are used interchangeably and refer to antigens which are differentially expressed by cancer cells and can thereby be exploited in order to target cancer cells.
  • Cancer antigens are antigens which can potentially stimulate apparently tumor-specific immune responses. Some of these antigens are encoded, although not necessarily expressed, by normal cells.
  • cancer antigens can be characterized as those which are normally silent (i.e., not expressed) in normal cells, those that are expressed only at certain stages of differentiation and those that are temporally expressed such as embryonic and fetal antigens.
  • Other cancer antigens are encoded by mutant cellular genes, such as oncogenes (e.g., activated ras oncogene), suppressor genes (e.g., mutant p53), fusion proteins resulting from internal deletions or chromosomal translocations.
  • Still other cancer antigens can be encoded by viral genes such as those carried on RNA and DNA tumor viruses.
  • tumor antigens examples include MAGE, MART-1/Melan-A, gplOO, Dipeptidyl peptidase IV (DPPIV), adenosine deaminase-binding protein (ADAbp), cyclophilin b, Colorectal associated antigen (CRC) ⁇ C017-1A/GA733, Carcinoembryonic Antigen (CEA) and its immunogenic epitopes CAP-1 and CAP-2, etv6, amll, Prostate Specific Antigen (PSA) and its immunogenic epitopes PSA-1, PSA-2, and PSA-3, prostate-specific membrane antigen (PSMA), T-cell receptor/CD3-zeta chain, MAGE-family of tumor antigens (e.g., MAGE-A1 , MAGE-A2, MAGE-A3, MAGE-A4, MAGE-A5, MAGE-A6, MAGE-A7, MAGE-A8, MAGE-A9, MAGE-A10, MAGE-A11,
  • Cancers or tumors and tumor antigens associated with such tumors include acute lymphoblastic leukemia (etv6; amll; cyclophilin b), B cell lymphoma (Ig-idiotype), glioma (E-cadherin; ⁇ -catenin; ⁇ -catenin; ⁇ -catenin; pl20ctn), bladder cancer (p21ras), biliary cancer (p21ras), breast cancer (MUC family; HER2/neu; c- erbB-2), cervical carcinoma (p53; p21ras), colon carcinoma (p21ras; HER2/neu; c-erbB-2; MUC family), colorectal cancer (Colorectal associated antigen (CRC) ⁇ C017-1A/GA733; APC), choriocarcinoma (CEA), epithelial cell cancer (cyclophilin b), gastric cancer (HER2/neu; c-erbB-2; ga733 glyco
  • a method of treating an autoimmune disorder in a subject involves the step of administering to a subject having an autoimmune disorder an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to treat the autoimmune disorder.
  • the substituted 4-primary amino quinoline composition in this aspect of the invention can be in the form a hydrate or pharmaceutically acceptable salt.
  • the autoimmune disorder is chosen from systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Sjogren's syndrome, polymyositis, vasculitis, Wegener' s granulomatosis, sarcoidosis, ankylosing spondylitis, Reiter's syndrome, psoriatic arthritis, and Behcet's syndrome.
  • the autoimmune disease is systemic lupus erythematosus.
  • the autoimmune disease is rheumatoid arthritis.
  • the subject is a human. The method can also be applied to animal models of any of the autoimmune disorders listed above.
  • the autoimmune disorder is an immune complex associated disease.
  • Immune complex associated diseases specifically include, without limitation, systemic lupus erythematosus, rheumatoid arthritis, polyarteritis nodosa, poststreptococcal glomerulonephritis, cryoglobulinemia, and acute and chronic serum sickness.
  • the substituted 4-primary amino quinoline composition can be administered to the subject by any suitable route of administration, including, without limitation, oral and parenteral.
  • Parenteral routes of administration include, without limitation, intravenous, intramuscular, intraperitoneal, subcutaneous, intranasal, intrapulmonary, transdermal, topical, and mucosal.
  • Parenteral routes of administration also include direct injection into a specific tissue or other site of injection, including, for example, lymphoid tissue and a site of inflammation. It has been discovered according to the invention that quinzoline compounds structurally similar to quinolines of the invention are unexpectedly useful in methods for inhibiting an immune response, both in vitro and in vivo, including methods for treating immune complex associated diseases and autoimmune disorders.
  • the quinazoline compounds and their methods of use according to the invention were unexpectedly found to retain much or all of the immunoinhibitory features of corresponding quinoline compounds and their methods of use, but with the additional feature of having reduced toxicity, at least in vivo.
  • the invention provides novel quinazoline compositions.
  • these compositions and other quinazoline compositions have been discovered to be useful in methods for inhibiting an immune response, both in vitro and in vivo, including methods for treating immune complex associated diseases and autoimmune disorders. Due to their similarity to certain known antimalarial agents, it is also believed that the novel quinazoline compositions of the invention will also be useful for prevention and treatment of malaria, as well as for treatment of other diseases which have been described to be responsive to treatment with chloroquines.
  • the novel quinazoline compositions of the invention have a structural Formula XVIII
  • X is absent or is an aryl, alkyl, heterocyclic or styryl group, provided that if X is a phenyl group, NRjR . is part if a heterocycle or is a diamine; R ⁇ and R 2 are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R ⁇ and R 2 optionally are combined to form a heterocycle; Y is an oxygen atom, a sulfur atom, CR 9 R 10 , or NR ⁇ , where R , R 10 , and R ⁇ are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R , R 10 , and R ⁇ optionally is combined with R 3 or R f to form a substituted or unsubstituted heterocycle; L is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; R 3 and t are each independently independently
  • Rg and R are each independently a halogen atom or an alkoxy group. In one embodiment Rg and R are each independently a chlorine atom or a methoxy group.
  • X is absent or is an aryl group;
  • NRiR 2 is a heterocyclic amine or NR (CH 2 ) n NR 10 R ⁇ , wherein n is an integer from 2 to 6, inclusive, and R , R 10 , and R ⁇ are each independently a hydrogen atom or an alkyl group;
  • Y is an aryl group or is NR 12 where R 12 is a hydrogen atom or an aryl or alkyl group;
  • L is absent or is a C 2 -Cg alkyl group;
  • R 3 and t are each independently a hydrogen atom or an alkyl group, wherein R 3 and R t optionally are combined to form a heterocycle; and
  • R 5 , Rg, R 7 , and Rg are each independently a hydrogen atom, a hal
  • X is absent or is an aryl group
  • NR ! R 2 is a substituted or unsubstituted piperazino or morpholino group or is NR 9 (CH 2 ) n NR 1 oR 11 , wherein n is an integer from 2 to 6, inclusive, R 9 is a hydrogen atom, and R 10 and R ⁇ are each independently an alkyl group
  • Y is NH
  • L is a C 2 -Cg alkyl group
  • R 3 and t are each independently a hydrogen atom or an alkyl group, wherein R 3 and R t optionally are combined to form a heterocycle
  • R 5 , Rg, R , and R 8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group.
  • X is an aryl group
  • NRjR 2 is substituted or unsubstituted piperazino or morpholino group
  • Y is NH
  • L is a C 2 -Cg alkyl group
  • R 3 and R t are each independently a methyl or ethyl group or R 3 and R ⁇ t optionally are combined to form a morpholino group
  • R 5 , Rg, R 7 , and R 8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group.
  • X is a phenyl group; NR t R 2 is N-methylpiperazine; Y is NH; L is -CH 2 CH 2 -; R 3 and f are each a methyl group; and R 5 , Rg, R 7 , and R 8 are each a hydrogen atom.
  • X is a phenyl group; NRiR 2 is N-methylpiperazine; Y is NH; L is -CH 2 CH 2 -; R 3 and R 4 are combined as a morpholino group; and R 5 , Rg, R 7 , and R 8 are each a hydrogen atom.
  • X is absent; NRiR .
  • R 5 , Rg, R , and R 8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group.
  • X is absent; NRjR 2 is N-methylpiperazine; Y is NH; L is -CH 2 CH 2 -; R 3 and t are each a methyl group; Rg and R are each a methoxy group; and R 5 and Rs are each a hydrogen atom.
  • X is absent; NR t R 2 is N-phenylpiperazine; Y is NH; L is -CH 2 CH 2 -; R 3 and R f are each a methyl group; Rg and R are each a methoxy group; and R 5 and Rs are each a hydrogen atom.
  • the composition is optionally in the form of a pharmaceutically acceptable hydrate or salt.
  • Representative, non-limiting examples of quinazoline compositions of Formula XVIII of the invention are compounds 201-214, presented in Table 3.
  • CMZ 203-34 CMZ 203-44, CMZ 203-45, CMZ 203-49, CMZ 203-51, CMZ 203-76, CMZ 203-78, CMZ 203-87, CMZ 203-93, and CMZ 203-95. Syntheses for each of these novel compounds are provided in Examples 19-28, respectively. Certain of these specific compounds have already been tested in vitro and found to inhibit TLR9 signaling. See Example 29.
  • CMZ 203-34, CMZ 203-44, CMZ 203-45, CMZ 203-49, CMZ 203-51, CMZ 203-76, CMZ 203-78, CMZ 203-87, CMZ 203-93, and CMZ 203-95 are believed to be useful in the methods of the invention.
  • the invention provides a method for inhibiting signaling by a TLR.
  • the method according to this aspect of the invention involves contacting a cell expressing a functional TLR with an effective amount of quinazoline composition having structural Formula XIX
  • X is a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group, optionally attached to the quinazoline by a nitrogen, oxygen, or sulfur atom or by a SO or SO 2 group;
  • Y is absent or is an oxygen atom, a sulfur atom, CR R 10 , or NR ⁇ , where R , R 10 , and R ⁇ are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R 9 , R 10 , and R ⁇ optionally is combined with R 3 or t to form a heterocycle;
  • L is absent or is a hydrogen atom, an alkyl or alkenyl group containing from 1 to 10 carbons, or an aryl group;
  • R 3 and R 4 are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R 3 and R 4 optionally are combined to form a heterocycle; and Rs,
  • the method according to this aspect of the invention can be performed in vitro or it can be performed in vivo.
  • the cell expressing the functional TLR can, but need not necessarily, be an immune cell.
  • the cell expressing the functional TLR can be a cell transfected with an expression vector that directs expression of the TLR by the cell.
  • the TLR is TLR9 and the method is thus a method for inhibiting intracellular signaling by TLR9.
  • compounds 201-214, presented in Table 3 the following additional representative and non-limiting quinazoline compounds, compounds 215-229, presented in Table 4 with reference to Formula XIX, can be used in the method according to this and all other aspects of the invention directed to methods involving use of substituted 4-primary amino quinoline compounds. Additional examples are presented in the Examples below.
  • the invention provides a method for inhibiting signaling by a TLR.
  • the method according to this aspect of the invention involves contacting an immune cell expressing a functional TLR with (a) an effective amount of a TLR signal agonist to stimulate signaling by the TLR in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having structural Formula XIX, as defined above, to inhibit signaling by the TLR in response to the TLR signal agonist compared with the signaling by the TLR in response to the TLR signal agonist in absence of the quinazoline composition.
  • the quinazoline composition in this aspect of the invention can be in the form a hydrate or pharmaceutically acceptable salt.
  • the TLR is TLR9 and the TLR signal agonist is a TLR9 signal agonist, and the method in one embodiment is thus a method of inhibiting intracellular signaling by TLR9 in response to a TLR9 signal agonist.
  • the TLR signal agonist in one embodiment is CpG DNA, for example a CpG ODN such as ODN 2006.
  • the CpG ODN can belong to any class of CpG ODN, including A-class (e.g., ODN 2216), B-class (e.g., ODN 2006), or C-class (e.g., ODN 2395).
  • the TLR signal agonist is an immune complex that includes a nucleic acid, as described above.
  • the invention provides a method for inhibiting an immune response to an antigenic substance.
  • the method according to this aspect of the invention involves contacting an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having structural Formula XIX, as defined above, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the quinazoline composition.
  • the quinazoline composition in this aspect of the invention can be in the form a hydrate or pharmaceutically acceptable salt.
  • the method according to this aspect of the invention can be performed in vitro or it can be performed in vivo.
  • the immune response is an innate immune response.
  • the immune response includes an adaptive immune response.
  • the step of contacting an immune cell expressing a functional Toll-like receptor with an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a quinazoline composition involves the active step of administering an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a quinazoline composition.
  • the antigenic substance can be administered using any effective route or means for administering the antigenic substance to effect the contacting.
  • the administering can be by local or systemic injection, inhalation, oral ingestion, topical administration, mucosal administration, or any combination thereof.
  • the method involves contacting, in a subject, an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response in the subject to the antigenic substance in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having structural Formula XVII, as defined above, to inhibit an immune response in the subject to the antigenic substance compared with the immune response to the antigenic substance in absence of the quinazoline composition.
  • the step of contacting an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response in the subject to the antigenic substance in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having structural Formula XVII, as defined above, to inhibit an immune response in the subject to the antigenic substance compared with the immune response to the antigenic substance in absence of the quinazoline composition.
  • Toll-like receptor with an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a quinazoline composition is passive.
  • the immune cell is already in contact or has already been in contact with an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance, at the time of or prior to the step of contacting the immune cell expressing a functional Toll-like receptor with an effective amount of a quinazoline composition having structural Formula XVII, as defined above, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the quinazoline composition.
  • the antigenic substance can be an allergen, as described above.
  • the antigenic substance can be an antigen that is or is derived from an infectious microbial agent, including a bacterium, a virus, a fungus, or a parasite, as described above.
  • the antigenic substance can be a cancer antigen, as described above.
  • a method of treating an autoimmune disorder in a subject involves the step of administering to a subject having an autoimmune disorder an effective amount of a quinazoline composition having structural Formula XIX, as defined above, to treat the autoimmune disorder.
  • the quinazoline composition in this aspect of the invention can be in the form a hydrate or pharmaceutically acceptable salt.
  • the autoimmune disorder is chosen from systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Sj ⁇ gren's syndrome, polymyositis, vasculitis, Wegener's granulomatosis, sarcoidosis, ankylosing spondylitis, Reiter's syndrome, psoriatic arthritis, and Behcet's syndrome.
  • the autoimmune disease is systemic lupus erythematosus.
  • the autoimmune disease is rheumatoid arthritis.
  • the subject is a human. The method can also be applied to animal models of any of the autoimmune disorders listed above.
  • the autoimmune disorder is an immune complex associated disease, as described above.
  • the quinazoline composition can be administered to the subject by any suitable route of administration, including, without limitation, oral and parenteral. Parenteral routes of administration are as described above with respect to substituted 4-primary amino quinolines.
  • the methods of the invention can be assessed using any of a number of possible readout systems based upon a TLR/IL-1R signal transduction pathway.
  • the readout for the method is based on the use of native genes or, alternatively, transfected or otherwise artificially introduced reporter gene constructs which are responsive to the TLR/IL-1R signal transduction pathway involving MyD88, TRAP, p38, and/or ERK.
  • TRAP TRAP
  • p38 p38
  • ERK ERK
  • reporter genes and reporter gene constructs particularly useful for the assays include, e.g., a reporter gene operatively linked to a promoter sensitive to NF- ⁇ B.
  • promoters include, without limitation, those for NF-KB, IL-l ⁇ , IL-6, IL-8, IL-12 p40, IP-10, CD80, CD86, and TNF- ⁇ .
  • the reporter gene operatively linked to the TLR-sensitive promoter can include, without limitation, an enzyme (e.g., luciferase, alkaline phosphatase, ⁇ -galactosidase, chloramphenicol acetyltransferase (CAT), etc.), a bioluminescence marker (e.g., green- fluorescent protein (GFP, e.g., U.S. Pat. No. 5,491,084), blue fluorescent protein (BFP, e.g., U.S. Pat. No.
  • an enzyme e.g., luciferase, alkaline phosphatase, ⁇ -galactosidase, chloramphenicol acetyltransferase (CAT), etc.
  • CAT chloramphenicol acetyltransferase
  • bioluminescence marker e.g., green- fluorescent protein (GFP, e.g., U.S. Pat. No. 5,491,08
  • the reporter is selected from IL-8, TNF- ⁇ , NF- ⁇ B-luciferase (NF- ⁇ B-luc; hacker H et al. (1999) EMBOJ 18:6973-82), IL-12 p40-luc (Murphy TL et al. (1995) Mol Cell Biol 15:5258-67), and TNF-luc (Hacker H et al. (1999) EMBO J 18:6973-82).
  • substrate can be supplied as part of the assay, and detection can involve measurement of chemiluminescence, fluorescence, color development, incorporation of radioactive label, drug resistance, or other marker of enzyme activity.
  • detection can be accomplished using flow cytometry (FACS) analysis or functional assays.
  • Secreted molecules can be assayed using enzyme- linked immunosorbent assay (ELISA) or bioassays. Many of these and other suitable readout systems are well known in the art and are commercially available.
  • a cell expressing a functional TLR and useful for the methods of the invention has, in some embodiments, an expression vector including an isolated nucleic acid which encodes a reporter construct useful for detecting TLR signaling.
  • the expression vector including an isolated nucleic acid which encodes a reporter construct useful for detecting TLR signaling can include a reporter gene under control of a promoter response element (enhancer element).
  • the promoter response element is associated with a minimal promoter responsive to a transcription factor believed by the applicant to be activated as a consequence of TLR signaling. Examples of such minimal promoters include, without limitation, promoters for the following genes: AP-1, NF- ⁇ B, ATF2, IRF3, and IRF7.
  • the expression vector including an isolated nucleic acid which encodes a reporter construct useful for detecting TLR signaling can include a gene under control of a promoter response element selected from response elements sensitive to IL-6, IL-8, IL-12 p40 subunit, a type I IFN, RANTES, TNF, IP-10, 1-TAC, and interferon-stimulated response element (ISRE).
  • the promoter response element generally will be present in multiple copies, e.g., as tandem repeats.
  • coding sequence for luciferase is under control of an upstream 6X tandem repeat of NF- ⁇ B response element.
  • an ISRE-luciferase reporter construct useful in the invention is available from Sfratagene (catalog no. 219092) and includes a 5x ISRE tandem repeat joined to a TATA box upstream of a luciferase reporter gene.
  • the reporter itself can be any gene product suitable for detection by methods recognized in the art. Such methods for detection can include, for example, measurement of spontaneous or stimulated light emission, enzyme activity, expression of a soluble molecule, expression of a cell surface molecule, etc.
  • Readouts typically involve usual elements of Toll/IL-IR signaling, e.g., MyD88, TRAF, and IRAK molecules, although in the case of TLR3 the role of MyD88 is less clear than for other TLR family members.
  • Such responses include the induction of a gene under control of a specific promoter such as a NF- ⁇ B promoter, increases in particular cytokine levels, increases in particular chemokine levels, etc.
  • the gene under the control of the NF- ⁇ B promoter can be a gene which naturally includes an NF- ⁇ B promoter or it can be a gene in a construct in which an NF- ⁇ B promoter has been inserted.
  • Genes and constructs which include the NF- ⁇ B promoter include but are not limited to IL-8, IL-12 p40, NF- ⁇ B-luc, IL-12 p40-luc, and TNF-luc. Increases in cytokine levels can result from increased production, increased stability, increased secretion, or any combination of the forgoing, of the cytokine in response to the TLR-mediated signaling.
  • Cytokines generally include, without limitation, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-10, IL-11, IL-12, IL-13, IL-15, IL-18, IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , TNF- ⁇ , GM-CSF, G-CSF, M-CSF.
  • Thl cytokines include but are not limited to IL-2, IFN- ⁇ , and IL-12.
  • Th2 cytokines include but are not limited to IL-4, IL-5, and IL-10.
  • chemokines of particular significance in the invention include but are not limited to CCL5 (RANTES), CXCL9 (Mig), CXCL10 (IP- 10), and CXCL11 (I-TAC), IL-8, and MCP-1.
  • Administration to a Subject Some aspects of the invention involve administering an effective amount of a composition to a subject to achieve a specific outcome.
  • the small molecule compositions useful according to the methods of the present invention thus can be formulated in any manner suitable for pharmaceutical use.
  • compositions of the invention are administered in pharmaceutically acceptable solutions, which may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic ingredients.
  • an effective amount of the compound can be administered to a subject by any mode allowing the compound to be taken up by the appropriate target cells.
  • administering the pharmaceutical composition of the present invention can be accomplished by any means known to the skilled artisan.
  • Specific routes of administration include but are not limited to oral, transdermal (e.g., via a patch), parenteral injection (subcutaneous, intradermal, intramuscular, intravenous, intraperitoneal, intrathecal, etc.), or mucosal (intranasal, intratracheal, inhalation, intrarectal, intravaginal, etc.).
  • An injection can be in a bolus or a continuous infusion.
  • the pharmaceutical compositions according to the invention are often administered by intravenous, intramuscular, or other parenteral means, or by biolistic "gene- gun" application to the epidermis.
  • liquid or solid pharmaceutical preparation forms are, for example, aqueous or saline solutions for injection or inhalation, microencapsulated, encochleated, coated onto microscopic gold particles, contained in liposomes, nebulized, aerosols, pellets for implantation into the skin, or dried onto a sharp object to be scratched into the skin.
  • the pharmaceutical compositions also include granules, powders, tablets, coated tablets, (micro)capsules, suppositories, syrups, emulsions, suspensions, creams, drops or preparations with protracted release of active compounds, in whose preparation excipients and additives and/or auxiliaries such as disintegrants, binders, coating agents, swelling agents, lubricants, flavorings, sweeteners or solubilizers are customarily used as described above.
  • the pharmaceutical compositions are suitable for use in a variety of drug delivery systems. For a brief review of present methods for drug delivery, see Langer R (1990) Science 249:1527-33, which is incorporated herein by reference.
  • the concentration of compounds included in compositions used in the methods of the invention can range from about 1 nM to about 100 ⁇ M. Effective doses are believed to range from about 10 picomole/kg to about 100 micromole/kg.
  • the pharmaceutical compositions are preferably prepared and administered in dose units. Liquid dose units are vials or ampoules for injection or other parenteral administration. Solid dose units are tablets, capsules, powders, and suppositories. For treatment of a patient, depending on activity of the compound, manner of administration, purpose of the administration (i.e., prophylactic or therapeutic), nature and severity of the disorder, age and body weight of the patient, different doses may be necessary.
  • compositions can be administered per se (neat) or in the form of a pharmaceutically acceptable salt.
  • salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts can conveniently be used to prepare pharmaceutically acceptable salts thereof.
  • Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic.
  • such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
  • Suitable buffering agents include: acetic acid and a salt (1-2% w/v); citric acid and a salt (1-3% w/v); boric acid and a salt (0.5-2.5% w/v); and phosphoric acid and a salt (0.8-2% w/v).
  • Suitable preservatives include benzalkonium chloride (0.003-0.03% w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004-0.02% w/v).
  • the pharmaceutical compositions of the invention optionally include an active ingredient and optionally a pharmaceutically-acceptable carrier.
  • compositions suitable for parenteral administration conveniently include sterile aqueous preparations, which can be isotonic with the blood of the recipient.
  • the acceptable vehicles and solvents are water, Ringer's solution, phosphate buffered saline, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed mineral or non-mineral oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Carrier formulations suitable for subcutaneous, intramuscular, intraperitoneal, intravenous, etc. administrations can be found in Remingto 's Pharmaceutical Sciences, Mack Publishing Company, Easton, PA.
  • the compounds useful in the invention can be delivered in mixtures of more than two such compounds.
  • a mixture can further include one or more adjuvants in addition to the combination of compounds.
  • a variety of administration routes is available. The particular mode selected will depend, of course, upon the particular compound selected, the age land general health status of the subject, the particular condition being treated, and the dosage required for therapeutic efficacy.
  • the methods of this invention generally speaking, can be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of response without causing clinically unacceptable adverse effects. Preferred modes of administration are discussed above.
  • the compositions can conveniently be presented in unit dosage form and can be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the compounds into association with a carrier which constitutes one or more accessory ingredients.
  • compositions are prepared by uniformly and intimately bringing the compounds into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product.
  • Other delivery systems can include time-release, delayed release or sustained release delivery systems. Such systems can avoid repeated administrations of the compounds, increasing convenience to the subject and the physician.
  • Many types of release delivery systems are available and known to those of ordinary skill in the art. They include polymer base systems such as poly(lactide-glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides. Microcapsules of the foregoing polymers containing drugs are described in, for example, U.S. Pat. No.
  • Delivery systems also include non-polymer systems that are: lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di-and tri-glycerides; hydrogel release systems; silastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially fused implants; and the like.
  • lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di-and tri-glycerides
  • hydrogel release systems silastic systems
  • peptide based systems such as wax, but are not limited to: (a) erosional systems in which an agent of the invention is contained in a form within a matrix such as those described in U.S. Pat. Nos.
  • TLR9 activity was assayed in terms of induction of a 6x NF- ⁇ B-luciferase reporter construct cotransfected in the cells. Results were measured as fold induction over baseline luciferase activity measured in the absence of ODN 2006 and compared to fold induction over baseline in the presence of the EC 50 concentration of ODN 2006 alone. From this initial screening a number of small molecules were identified as lead compounds and were categorized by structure and activity. Additional screening was performed in like manner using additional small molecules selected from another library or specifically prepared for this purpose. Results of some of these assays are presented in FIG. l and in Tables 5-16. The method is described in greater detail as follows.
  • the cells for each individual cell clone (hTLR3-NF ⁇ B-293, hTLR7-NF ⁇ B-293, hTLR8-NF ⁇ B-293 and hTLR9-NF ⁇ B-293) were counted and seeded one day before the stimulation at 1.5x10 4 cells per well in 96-well cell culture plates. To become adherent the cells were incubated after seeding overnight at 37°C in 5% CO 2 humidified air. The screen for each compound was performed at the same time for all of the receptors TLR3, TLR7, TLR8 and TLR9.
  • test compounds Up to fifteen test compounds, each assayed in duplicate at each of three different final concentrations (0.5 ⁇ g/ml, 5 ⁇ g/ml, and 50 ⁇ g/ml), were used for 16h cell stimulation on a single multiwell culture plate. A 4X final concentration master plate was created to prepare the different test compounds before they were added to the cells. Controls were added as duplicates individually for each cell clone on the cell culture plate.
  • Positive controls were used as follows (final concentrations): 50 ⁇ g/ml poly(I:C) for hTLR3-NF ⁇ B-293; 8 ⁇ M resiquimod (R848) for hTLR7-NF ⁇ B-293; 30 ⁇ M R848 for hTLR8-NF ⁇ B-293; and 2.5 ⁇ M CpG-ODN 2006 for hTLR9-NF ⁇ B-293. Media alone was used as negative control. After addition of controls and small molecules, the cell clones were additionally stimulated with the EC 50 concentration of the appropriate receptor-specific ligand. To calculate the baseline response to the EC 50 concentration of the appropriate receptor-specific ligand, wells H3 and H6 did not receive receptor-specific ligand.
  • Antagonists were scored as follows: 13: compounds for which there was clear down regulation at 0.5 ⁇ g/ml, 5 ⁇ g/ml, and 50 ⁇ g/ml; 12: compounds for which there was clear down regulation at 5 ⁇ g/ml and 50 ⁇ g/ml; 11 : compounds for which there was clear down regulation only at 50 ⁇ g/ml; — : compounds for which there was no clear down regulation even at 50 ⁇ g/ml.
  • Agonists To detect agonist activity the cells were treated in the above described manner but without adding the EC 50 concentration of receptor-specific ligand after treatment of the cells with small molecules. Usually the baseline in the agonist screen is taken to be 1 because these cells have not been additionally stimulated with the EC 50 concentration of the receptor- specific ligand. Agonist activity was present when there was above-baseline activity. Agonists were scored as follows:
  • Toxicity Measuring of "toxicity" of the small molecules was performed by analysing below- baseline activity within the agonist screen. "Toxicity” can have different explanations - either differences between the cell clones, the compound are effecting the signal transduction pathway or the TLR within the cell clone, or the compounds do re ⁇ illy have a toxic effect on the cells. Each compound was screened for "toxicity" on each of the four cell lines. Usually the baseline in the agonist screen is taken to be 1 because these cells have not been additionally stimulated with the EC50 concentration of the receptor-specific ligand. Apparent toxicity was scored as follows:
  • T3 compounds for which there was clear down regulation at 0.5 ⁇ g/ml, 5 ⁇ g/ml, and 50 ⁇ g/ml;
  • T2 compounds for which there was clear down regulation at 5 ⁇ g/ml and 50 ⁇ g/ml
  • TI compounds for which there was clear down regulation only at 50 ⁇ g/ml.
  • Example 2 In Vitro Screening of a Library of Small Molecules for Inhibitors of Human TLR8
  • HEK293 cells were stably transfected with a human TLR8 (hTLR8) expression vector and incubated overnight in the presence of 500 nM R848 (EC 50 of R848 for TLR8) and selected small molecule candidate compounds at different concentrations ranging from 5x10 "7 M to 5xl0 "5 M.
  • TLR8 activity was assayed in terms of induction of a 6x NF- ⁇ B- luciferase reporter construct cotransfected in the cells.
  • Results were expressed as fold induction over baseline luciferase activity measured in the absence of R848. From this initial screening a number of small molecules were identified as lead compounds and were categorized by structure and activity. Additional screening was performed in like manner using additional small molecules selected from another library or specifically prepared for this purpose. Results of some of these assays are presented in Tables 5-16. Scoring is reported as above but as applied to TLR8.
  • Example 3 In Vitro Screening of a Library of Small Molecules for Inhibitors of Human TLRl
  • HEK293 cells were stably transfected with a human TLR7 (hTLR7) expression vector and incubated overnight in the presence of 2 ⁇ M R848 (EC 50 of R848 for TLR7) and selected small molecule candidate compounds at different concentrations ranging 7 ⁇ from 5x10 " M to 5x10 " M.
  • TLR7 activity was assayed in terms of induction of a 6x NF- ⁇ B- luciferase reporter construct cotransfected in the cells.
  • Results were expressed as fold induction over baseline luciferase activity measured in the absence of R848. From this initial screening a number of small molecules were identified as lead compounds and were categorized by structure and activity. Additional screening was performed in like manner using additional small molecules selected from another library or specifically prepared for this purpose. Results of some of these assays are presented in Tables 5-16. Scoring is reported as above but as applied to TLR7.
  • Example 4 In Vitro Screening of a Library of Small Molecules for Inhibitors of Human TLRS
  • Candidate small molecules for initial screening were identified in an assay similar to that in Example 1 except HEK293 cells were stably transfected with a human TLR3 (hTLR3) expression vector and incubated overnight in the presence of 2.5 ⁇ g/ml poly(I:C) (EC 50 of poly(LC) for TLR3) and selected small molecule candidate compounds at different concentrations ranging from 5x10 "7 M to 5xl0 "5 M.
  • TLR3 activity was assayed in terms of induction of a 6x NF- ⁇ B -luciferase reporter construct cotransfected in the cells.
  • Results were expressed as fold induction over baseline luciferase activity measured in the absence of poly(I:C). From this initial screening a number of small molecules were identified as lead compounds and were categorized by structure and activity. Additional screening was performed in like manner using additional small molecules selected from another library or specifically prepared for this purpose. Results of some of these assays are presented in Tables 5-16. Scoring is reported as above but as applied to TLR3.
  • mice are administered known amounts of candidate small molecules and a source of PAMP or other suitable TLR ligand, e.g., CpG nucleic acid.
  • Negative control mice receive the source of PAMP or other suitable TLR ligand, e.g., CpG nucleic acid, alone.
  • blood samples are obtained from control and experimental mice and evaluated for serum concentration of cytokine using a suitable method, e.g., enzyme- linked immunosorbent assay (ELISA).
  • ELISA enzyme- linked immunosorbent assay
  • PBMC peripheral blood mononuclear cells
  • FACS fluorescence activated cell sorting
  • Control and experimental results are compared in pairwise fashion.
  • Reduced expression of activation marker or reduced concentration of cytokine in an experimental animal compared to a negative control animal indicates the small molecule inhibits TLR-mediated signaling in response to a ligand for the TLR.
  • Increased expression of activation marker or increased concentration of cytokine in an experimental animal compared to a negative control animal indicates the small molecule promotes TLR-mediated signaling in response to a ligand for the TLR.
  • Example 6 In Vivo Screening of Selected Small Molecules Experimental mice are administered candidate small molecules. Negative control mice are administered carrier alone.
  • PBMC are isolated and then exposed in vitro to CpG nucleic acid under conditions in which the PBMC, in the absence of small molecule, are stimulated to express an activation marker such as CD86 or secrete a product such as cytokine (e.g., IFN- ⁇ , IL-6, TNF- ⁇ ) or chemokine (e.g., IP- 10).
  • an activation marker such as CD86 or secrete a product such as cytokine (e.g., IFN- ⁇ , IL-6, TNF- ⁇ ) or chemokine (e.g., IP- 10).
  • cytokine e.g., IFN- ⁇ , IL-6, TNF- ⁇
  • chemokine e.g., IP- 10
  • Reduced expression of activation marker or reduced concentration of cytokine in an experimental animal compared to a negative control animal indicates the small molecule inhibits TLR-mediated signaling in response to a ligand for the TLR.
  • Increased expression of activation marker or increased concentration of cytokine in an experimental animal compared to a negative control animal indicates the small molecule promotes TLR-mediated signaling in response to a ligand for the TLR.
  • Example 7 Human TLR9 Antagonism by 4-Primary Amino Quinolines Stably co-transfecting the human TLR9 receptor (hTLR9) and an NF- ⁇ B promoter- driven luciferase gene into HEK-293 cells created the cell line hTLR9-NFkB-293.
  • hTLR9- NFkB-293 cells were counted and seeded at 1.5xl0 4 cells per well in 96-well cell culture plates one day before assaying and cultured at 37°C / 5% CO 2 . The day of the assay, antagonist was added at 50 ⁇ M, 5.0 ⁇ M or 0.5 ⁇ M.
  • the cells were then stimulated with the EC50 dose of the TLR9 agonist, CpG-ODN 2006, and cultured for 16 h in a humidified incubator at 37°C. The culture supernatant was removed and the cells were treated with lysis buffer and stored at -80°C before measuring luciferase activity. The luciferase readout was measured according to manufacturer's instructions using a luciferase assay system available from Promega, USA. Results are summarized in Table 18.
  • the antagonist dose listed in Table 18 is the minimum effective dose ( ⁇ M) for blockade of the TLR9 agonist. As evident from the results presented in Table 18, the minimum effective dose ( ⁇ M) for blockade of the TLR9 agonist was 0.5 ⁇ M or 5 ⁇ M for all of the compounds tested, as measured by this three-point assay.
  • Example 8 Human TLR9 Antagonism by Quinazolines hTLR9-NFkB-293 cells were counted and seeded at 1.5xl0 4 cells per well in 96-well cell culture plates one day before assaying and cultured at 37°C / 5% CO 2 , as described in Example 7. The day of the assay, antagonist was added at varying concentrations starting at 50 ⁇ M with a 1/5 to 1/6 dilution for 7 steps. The cells were then stimulated with the EC50 dose of the TLR9 agonist, CpG-ODN 2006, and cultured for 16 h in a humidified incubator at 37°C.
  • hTLR9 are the IC50 dose ( ⁇ M) for blockade of a CpG ODN-generated signal in hTLR9-NFkB-293 cells.
  • TLR9 ligand antagonism in human peripheral blood mononuclear cells (PBMC) was monitored.
  • PBMC peripheral blood buffy coat preparations from healthy male and female human donors were obtained from the Blood Bank of the University of Diisseldorf (Germany) and from these, PBMC were purified by centrifugation over Ficoll-Hypaque (Sigma). The purified PBMC were resuspended in RPMI 1640 culture medium supplemented with 5% (v/v) heat-inactivated human AB serum (BioWhittaker, Belgium) or 10% (v/v) heat-inactivated fetal calf serum (FCS), 1.5 mM L-glutamine, 100 U/ml penicillin and 100 ⁇ g/ml streptomycin (all from Sigma).
  • PBMC fresh PBMC at a concentration of 3x107ml to 5x107ml were added to 96-well round-bottomed plates (150 ⁇ l/well). After cell plating, antagonist was added at varying concentrations starting at 50 ⁇ M with a 1/5 to 1/6 dilution for 7 steps. The cells were then stimulated with the TLR9 agonist, CpG-ODN 2006, and cultured for 16 h in a humidified incubator at 37°C. Culture supernatants were collected and, if not used immediately, frozen at -20°C until required. Interleukin 6 (IL-6) in the supernatants was quantitatively assessed using commercially available ELISA Kits (IL6, Diaclone, USA). Sigmoidal antagonism curves were generated and IC50 calculated. Results shown in Table 19 under the heading IL-6 are the IC50 dose ( ⁇ M) for blockade of CpG ODN-generated IL-6 production by human PBMC.
  • Example 9 Reduced In Vivo Toxicity of Quinazoline Compounds Compared with a Structurally Similar Quinoline
  • Female BALB/c mice (n 3 per group) were given a single intraperitoneal bolus injection of CMZ 203-43 (1.0 or 4.0 mg), CMZ 203-34 (1.0 or 4.0 mg) or CMZ 203-49 (1.0 or 4.0 mg) in a volume of 0.2 ml.
  • Compound CMZ 203-43 has the structural formula
  • mice received 10 % dimethyl sulfoxide (DMSO) in an identical manner. Animals were weighed immediately prior to injection (day 1) and then daily until day 5. On day 5, blood was collected by cardiac puncture and analyzed for hematological and biochemical parameters. Animals were monitored daily for morbidity and mortality. Results are shown in
  • FIG. 2 and FIG. 3. shows change in body weight (relative to body weight prior to first injection)
  • FIG. 2B shows mean body weight ⁇ standard error of the means for different groups. Mice receiving 1.0 mg CMZ 203-43 had significant weight loss, while mice receiving quinazoline at either dose did not. All of the mice receiving 4.0 mg CMZ 203-43 group were dead on day 4. Necropsy of mice in the 4.0 mg CMZ 203-43 group revealed bowel obstruction.
  • FIG. 3 shows white blood cell (WBC) differential as mean percentage of total white cells ⁇ standard error of the means for different groups. Mice treated with 1.0 mg CMZ 203- 43 had a significant neutrophilia while mice receiving quinazoline at either dose did not.
  • WBC white blood cell
  • mice treated with 4.0 mg CMZ 203-43 was not available because all these mice were dead before day 5.
  • - I l l - Example 11 In Vivo Inhibition ofTNF-a by 4-Primary Amino Quinoline and by Quinazolines
  • Groups of BALB/c mice treated as in Example 10 were also analyzed for TNF- ⁇ . Animals were bled at 1 hr post injection with CpG ODN 2006 and TNF- ⁇ levels in plasma measured by TNF- ⁇ -specific ELISA. Results are presented in FIG. 5.
  • Example 13 Synthesis of CMZ 203-85 A mixture of the benzophenone (15 gm, 0.056 moles) and powdered sodium hydroxide (15 gm) in ethanol (150 mL) was stirred and heated to reflux. The heat was removed and zinc dust (15 gm) was added in portions at a rate that kept the mixture at reflux. After the addition was complete, the mixture was heated at reflux for one hour. The reaction mixture was allowed to cool and was then filtered to remove zinc. The zinc was washed with ethanol (20 mL) and the combined filtrates were added to water (500 mL). The crystalline white product was isolated by filtration, washed with water and dried. The carbinol was obtained in a yield of 12.1 gm, 80%.
  • CMZ 203-91 was 0.58 gm, 77% as an oil which crystallized upon standing.
  • hTLR9-NFkB-293 cells were counted and seeded at 1.5xl0 4 cells per well in 96-well cell culture plates one day before assaying and cultured at 37°C / 5% CO 2 , as described in Example 7.
  • the solution was cooled to room temperature and was diluted with water (500 mL) and tert- butylmethyl ether (TBME, 500 mL).
  • TBME tert- butylmethyl ether
  • the mixture was stirred and made basic by the addition of solid sodium carbonate. After stirring for 15 minutes the precipitated product was isolated by filtration.
  • the crude product was re-suspended in warm water, stirred for 5 minutes and collected by filtration. After washing with water followed by TBME, the solid was air dried.
  • the dried quinazolinone was recrystallized from n-butanol to give 11.1 gm (50%) of product as colorless needles.
  • NMP N-methylpyrrolidinone
  • N,N-dimethylethylenediamine (1.32 gm, 0.015 moles) was added and the solution was then warmed at 100°C for 30 minutes.
  • the NMP solution was diluted with water (200 mL) and concentrated ammonium hydroxide (50 mL).
  • the product, which had precipitated, was extracted into methylene chloride (2 X 100 mL). The extracts were combined, dried over magnesium sulfate and then, after filtration, were stripped to give the product as a light brown oil.
  • N,N-dimethylethylenediamine (3.5 gm, 0.04 moles) and 2-phenyl-4- chloroquinazoline (2.4 gm, 0.01 moles) were combined in n-butanol (50 mL) and were brought to reflux. Once reflux had been achieved, TLC (silica, 10% methanol in dichloromethane) showed that the reaction was complete. The solution was cooled and stripped. The residue was dissolved in t-butylmethyl ether (TBME) and the solution was washed with water. After drying over magnesium sulfate the solution was filtered and stripped to give 1.37 gm (0.0047 moles, 47%) of the crude product as an oil.
  • TBME t-butylmethyl ether
  • the 4-chloroquinazoline (3.7 gm, 0.015 moles) was refluxed in ethanol with N-2-aminoethylmorpholine (3.99 gm, 0.031 moles) for one hour. Once cooled, the solution was stripped and the residue was dissolved in water (400 mL). The solution was made basic by the addition of sodium carbonate and the product was extracted into methylene chloride (2 X 100 mL). The combined extracts were washed with water (50 mL) and were then dried over magnesium sulfate. After filtration, the extracts were stripped to give the product as a tan solid.
  • the solid was recrystallized from ethyl acetate (25 mL) and hexane (50 mL).
  • the product, 203-93 was isolated as an off white solid in a yield of 2.86 gm, 56.0%>.
  • the solid was isolated by filtration, washed with acetone. After drying, the crude product was recrystallized twice from n-butanol. A small amount of tetrachlorohydroquinone will co-crystallize under these conditions. It can be removed by stirring the solid in warm 5% sodium carbonate solution. The quinazolinone was isolated as a tan solid in a yield of 9.5 gm, 43%>.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Dermatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Neurology (AREA)
  • Cardiology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Transplantation (AREA)
  • Rheumatology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Other In-Based Heterocyclic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Quinoline Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The invention provides methods and compositions useful for modulating signaling through Toll-like receptors. The methods involve contacting a TLR-expressing cell with a small molecule having a core structure including at least two rings. Certain of the compounds are 4-primary amino quinolines. Many of the compounds and methods are useful specifically for inhibiting immune stimulation involving at least one of TLR9, TLR8, TLR7, and TLR3. The methods may have use in the treatment of autoimmunity, inflammation, allergy, asthma, graft rejection, graft versus host disease, infection, sepsis, cancer, and immunodeficiency.

Description

SMALL MOLECULE TOLL-LIKE RECEPTOR (TLR) ANTAGONISTS
FIELD OF THE INVENTION The invention relates to generally to the field of immunology. More particularly, the invention relates to compositions and methods for altering immune function. More specifically, the invention relates to compositions and methods for affecting immune stimulation mediated through Toll-like receptor (TLR) molecules.
BACKGROUND OF THE INVENTION Stimulation of the immune system, which includes stimulation of either or both innate immunity and adaptive immunity, is a complex phenomenon that can result in either protective or adverse physiologic outcomes for the host. In recent years there has been increased interest in the mechanisms underlying innate immunity, which is believed to initiate and support adaptive immunity. This interest has been fueled in part by the recent discovery of a family of highly conserved pattern recognition receptor proteins known as Toll-like receptors (TLRs) believed to be involved in innate immunity as recep[tors for pathogen-associated molecular patterns (PAMPs). Compositions and methods useful for modulating innate immunity are therefore of great interest, as they may affect therapeutic approaches to conditions involving autoimmunity, inflammation, allergy, asthma, graft rejection, graft versus host disease (GvHD), infection, cancer, and immunodeficiency. Recently there have been a number of reports describing the immunostjmulatory effect of certain types of nucleic acid molecules, including CpG nucleic acids and double- stranded RNA. Of note, it was recently reported that Toll-like receptor 9 (TLR9) recognizes bacterial DNA and CpG DNA. Hemmi H et al. (2000) Nature 408:740-5; Bauer S et al. (2001) Proc Natl Acad Sci USA 98:9237-42. It was also recently reported thήt immune complexes containing IgG and nucleic acid can stimulate TLR9 and participate in B-cell activation in certain autoimmune diseases. Leadbetter EA et al. (2002) Nature 416:595-8. Chlroroquines have been recognized as useful not only as anti-malarial agents but also as anti-inflammatory agents. Although its mechanism of action is not well understood, chloroquine has been used effectively in the treatment of various autoimmune diseases, including rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). For a review, see Wallace DJ (1996) Lupus 5 Suppl 1 :S59-64. Recently Macfarlane and colleagues described a number of small molecule analogs and derivatives of chloroquine (4- aminoquinoline) and quinacrine (9-aminoacridine) which reportedly inhibit stimulation of the immune system. U.S. Pat. No. 6,221,882; U.S. Pat. No. 6,479,504; U.S. Pat. No. 6,521,637; PCT published application PCT/US00/16723 (WO 00/76982); and PCT published application PCT/US98/13820 (WO 99/01154). Macfarlane and colleagues report these sm$ll molecule inhibitors of the immune response, even when used at nanomolar concentrationjS, can block the action of immunostimulatory DNA. U.S. Pat. No. 6,221,882 Bl. Macfarlaώe and co workers studied a large number of compounds by varying substituents on a limited number of 4-aminoquinoline and 9-aminoacridine core structures related to chloroquine and quinacrine.
SUMMARY OF THE INVENTION The present invention is based, in part, on the discovery by the applicants that a number of small molecules, some previously known but distinct from those described by Macfarlane et al. and by Tobe et al., can alter TLR-mediated immunostimulatory signaling. Many of the compounds inhibit TLR signaling and are useful as inhibitors of immune stimulation. Compositions and methods described herein are useful for inhibiting immune stimulation in vitro and in vivo. Such compositions and methods thus will find use in a number of clinical applications, including as pharmaceutical agents and methods for treating conditions involving unwanted immune activity, including inflammatory and autoimmune disorders. The compositions of the invention can also be used in methods for the preparation of medicaments for use in the treatment of conditions involving unwanted imnlune activity, including a variety of inflammatory and autoimmune disorders. It was surprisingly discovered that molecules having similar substituents but different core structures compared to those related to chloroquine and quinacrine described by Macfarlane et al., are potent immunomodulatory small molecules. Without being bound to any theory or mechanism, it is believed that the small molecules described by tjhe present invention affect immune stimulation via interaction with a TLR. More particularly, it is believed that many of the small molecules described by the present invention inhibit immune stimulation via TLR antagonism. In particular, it is believed that many of the small molecules described by the present invention inhibit immune stimulation via TLR9 antagonism. The invention in certain aspects provides methods useful for altering TLR-mediated signaling. The methods of the invention are useful whenever it is desirable to alter TLR- mediated signaling in response to a suitable TLR ligand or TLR signaling agonist. For example, the methods can be used to treat any of a variety of conditions involving autoimmunity, inflammation, allergy, asthma, graft rejection, graft-versus-host disease (GvHD), infection, sepsis, cancer, and immunodeficiency. Generally, methods useful in the treatment of conditions involving autoimmunity, inflammation, allergy, asthma, graft rejection, and GvHD will employ small molecules that inhibit TLR-mediated signaling in response to a suitable TLR ligand or TLR signaling agonist. Generally, method$ useful in the treatment of conditions involving infection, cancer, and immunodeficiency will lemploy small molecules that augment TLR-mediated signaling in response to a suitable TLR ligand. In some instances the methods can be used to inhibit or promote TLR-mediated signaling in response to a TLR ligand or TLR signaling agonist. In some instances the methods can be used to inhibit TLR-mediated immunostimulatory signaling in response to a TLR ligand or TLR signaling agonist. In some instances the methods can be used to inhibit or promote TLR-mediated immunostimulation in a subject. In some instances the methods can be used to inhibit TLR-mediated immunostimulation in a subject. In some instances the methodss ' can be used to inhibit an immunostimulatory nucleic acid-associated response in a subject. The invention in certain aspects provides novel small molecule compositions useful for altering TLR-mediated signaling. The compositions of the invention are useful whenever it is desirable to alter TLR-mediated signaling in response to a suitable TLR ligand or TLR signaling agonist. For example, the small molecules can be used in methods td treat any of a variety of conditions involving autoimmunity, inflammation, allergy, asthma, graft rejection, GvHD, infection, sepsis, cancer, and immunodeficiency. Generally, methods μseful in the treatment of conditions involving autoimmunity, inflammation, allergy, asthma, graft rejection, and GvHD will employ small molecules that inhibit TLR-mediated signaling in response to a suitable TLR ligand or TLR signaling agonist. Generally, methods useful in the treatment of conditions involving infection, cancer, and immunodeficiency will employ small molecules that augment TLR-mediated signaling in response to a suitable TLR ligand. In some instances the molecules can be used in a method to inhibit or promote TLR-mediated signaling in response to a TLR ligand or TLR signaling agonist. In some instances the small molecules can be used in a method to inhibit TLR-mediated immunostimulatory signaling in response to a TLR ligand or TLR signaling agonist. In some instances the small molecules can be used in a method to inhibit or promote TLR-mediated immunostimulatioii in a subject. In some instances the small molecules can be used in a method to inhibit TLR-mediated immunostimulation in a subject. In some instances the small molecules can be uised to inhibit an immunostimulatory nucleic acid-associated response in a subject. As a feature of the present invention, the methods of the invention can be combined with administration of additional agents to achieve synergistic effect on TLR-m diated immunostimulation. More specifically, whereas the agents described herein have been discovered to affect TLRs directly and thus directly affect TLR-bearing cells, e.g., antigen- presenting cells (APCs), such agents can be used in conjunction with additional agents which affect non-APC immune cells, e.g., T lymphocytes (T cells). Such an approach effectively introduces an immunomodulatory intervention at two levels: innate immunity and acquired immunity. Since innate immunity is believed to initiate and support acquired immunity, the combination intervention is synergistic.
In one aspect of the invention, a method of affecting TLR-mediated signaling in response to a TLR ligand is provided. The method according to this aspect involves contacting a cell expressing a TLR with an effective amount of a compound of Formula I
Figure imgf000005_0001
Formula I
wherein 2 is a five- to seven-membered homocyclic or heterocyclic ring, wherein each of X, Y, and Z is independently chosen from a carbon atom (C), a nitrogen atom (N), an oxygen atom (O), and a sulfur atom (S), and wherein B2 optionally includes at least one atom selected from C, N, O, and S; wherein 1 and 2 are optionally bridged by B3 to form a five- to seven-membered ring (3), wherein B3 optionally includes at least one atom selected from C, N, O, and S, and wherein when A is carbon, (3) is not pyridine; wherein 2 optionally includes an unsaturated bond; wherein (3) optionally includes an unsaturated bond; wherejin R2, when present, is a hydrogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, optionally linked to Z via N, O, or S; wherein R3, R4, R5, R6, R7, and R8, when present, are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein A is an atom selected from C, N, O, and S; wherein each of A', A", and A'" independently is R9, -NR9R10, -OR9, or -CR9R10, wherein R9 is a hydrogen atom, hydroxyl, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, and wherein RIO is optionally absent or is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR.
In one aspect of the invention, a method of inhibiting TLR-mediated immunostimulatory signaling is provided. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula I, as provided above, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
In one aspect the invention provides a method of affecting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula I, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject. The invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula I, as provided above, to inhibit TLR-mediated immunostimulation in the subject.
In one aspect the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject. The method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula I, as provided above, to inhibit an immunostimulatory nucleic acid- associated response in the subject. In one embodiment the subject is otherwise free of symptoms calling for treatment with a compound of Formula I. In one embodiment according to any of the foregoing aspects of the invention, A is nitrogen and (3) is a five-membered ring. In one embodiment the compound is Formula II
Figure imgf000007_0001
Formula II
wherein A is chosen from C and N; and Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S. In various specific embodiments the compound is any one of compounds 455, 470, 564, 568, 593, 607, 612-614, 619-621, 636, 685, 875, 878, 890, 904, 918, 939, 944, 1039, 1050, 1132, 1241, and 1243 listed in Table 5 below. In one embodiment according to any of the foregoing aspects of the invention, A is nitrogen, (3) is a six-membered ring, and B3 is one atom selected from C, N, O, and S. In one embodiment A is nitrogen, 3 is a six-membered ring, and B3 is S. In one embodiment the compound is Formula III
Figure imgf000007_0002
Formula III wherein Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked viaN, O, or S, and wherein X is chosen from N, O, and S. In certain embodiments X is S. In various specific embodiments the compound is any one of compounds 53, 64, 125, 149, 313, 399, 529, 576, 693, 797, 840, and 842 listed in Table 6 below. In one embodiment according to any of the foregoing aspects of the invention, A is carbon, (3) is a six-membered ring, and B3 is C or S. In one embodiment the compound is Formula IV
Figure imgf000008_0001
Formula IV
wherein X is C or S and Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S. In various specific embodiments the compound is any one of compounds 43, 294, 340, 346, 348, 413, 491, 917, 1015, 1042, 1158, 1287, and 1337 listed in Table 7 below. In one embodiment according to any of the foregoing aspects of the invention, A is nitrogen, (3) is a seven-membered ring, and B3 includes two carbon atoms. In one embodiment the compound is Formula V
Figure imgf000008_0002
Formula V wherein X and Y are each independently C, N, O, or S; and Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S. In various specific embodiments the compound is any one of compounds 72, 74, 99, 109, 343, 488, 1013, and 1352 listed in Table 8 below. In one embodiment according to any of the foregoing aspects of the invention, 1 and 2 are unbridged by B3, A is carbon, and A' is -OR9. In one embodiment the compound is Formula VI
Figure imgf000009_0001
Formula VI
wherein Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and R9 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group. In some embodiments R9 and R4 are united to form a lactone. In various specific embodiments the compound is any one of compounds 65, 229, 239, 306, 386, 707, 793, 957, 970, 974, 1161, and 1308 listed in Table 9 below. In one embodiment according to any of the foregoing aspects of the invention, 1 and 2 are unbridged by B3, A is carbon, and A' is -NR9R10. In one embodiment the compound is Formula VII
Figure imgf000009_0002
Formula VII wherein Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked vi N, O, or S. In various specific embodiments the compound is any one of compounds 133, 267, 312, 457, and 510 listed in Table 10 below. In one embodiment according to any of the foregoing aspects of the invention, 1 and 2 are unbridged by B3, A is carbon, and A' is -CR9R10. In one embodiment the compound is Formula VIII
Figure imgf000010_0001
Formula VIIT
wherein Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S. In various specific embodiments the compound is any one of compounds 93, 108, 138, 144, 267, 270, 308, 381, 560, 778, 799, 822, 833, 884, 965, and 1187 listed in Table 11 below.
In one aspect the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand. The method according to this aspect of the inventioni involves contacting a cell expressing a TLR with an effective amount of a compound of Formula IX
Figure imgf000010_0002
Formula IX wherein R2 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein R10 is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR. In various specific embodiments the compound is any one of compounds 751, 858, 2000, and 2001 listed in Table 12 below.
In one aspect of the invention, a method of inhibiting TLR-mediated immunostimulatory signaling is provided. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula IX, as provided above, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
In one aspect the invention provides a method of affecting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula IX, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject.
The invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula IX, as provided above, to inhibit TLR-mediated immunostimulation in the subject.
In one aspect the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject. The method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula IX, as provided above, to inhibit an immunostimulatory nucleic acid- associated response in the subject. In one embodiment the subject is otherwise free of symptoms calling for treatment with a compound of Formula IX.
In one aspect the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula X
Figure imgf000012_0001
Formula X
wherein each of Rl, R2, R3, R4, R5, R6, R7, and R8 is independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S, to inhibit or promote TLR- mediated signaling in response to a ligand for the TLR. In various specific embodiments the compound is any one of compounds 431, 583, 586, 792, and 830 listed in Table 13 below.
In one aspect of the invention, a method of inhibiting TLR-mediated immunostimulatory signaling is provided. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula X, as provided above, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR. In one aspect the invention provides a method of affecting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula X, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject. The invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula X, as provided above, to inhibit TLR-mediated immunostimulation in the subject.
In one aspect the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject. The method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula X, as provided above, to inhibit an immunostimulatory nucleic acid- associated response in the subject. In one embodiment the subject is otherwise free of symptoms calling for treatment with a compound of Formula X.
In one aspect the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XI
Figure imgf000013_0001
Formula XI
wherein each of Rl, R2, R5, R6, R7, R8, Rl 1, and R12 is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and X is C, N, O, or S, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR. In some embodiments R2 includes a cycloalkyl, benzyl or phenyl group. In some embodiments X is N. In some embodiments Rl 1 and R12 are linked as a spiro group in a five- or six-membered ring. In various specific embodiments the compound is any one of compounds 891, 926, 1137, 1213, 1248, 1320, and 1322 listed in Table 14 below.
In one aspect of the invention, a method of inhibiting TLR-mediated immunostimulatory signaling is provided. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XI, as provided above, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
In one aspect the invention provides a method of affecting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XI, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject.
The invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XI, as provided above, to inhibit TLR-mediated immunostimulation in the subject.
In one aspect the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject. The method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula XI, as provided above, to inhibit an immunostimulatory nucleic acid- associated response in the subject. In one embodiment the subject is otherwise free of symptoms calling for treatment with a compound of Formula XI. In one aspect the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XII
Figure imgf000015_0001
Formula XII wherein Rl, R2, R3, R4, R5, R6, R7, R8, R9, RIO, and Rl 1 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR. In various specific embodiments the compound is any one of compounds 101, 600, and 609 listed in Table 15 below. In one aspect of the invention, a method of inhibiting TLR-mediated immunostimulatory signaling is provided. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XII, as provided above, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
In one aspect the invention provides a method of affecting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XII, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject. The invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XII, as provided above, to inhibit TLR-mediated immunostimulation in the subject.
In one aspect the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject. The method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula XII, as provided above, to inhibit an immunostimulatory nucleic acid-associated response in the subject. In one embodiment the subject is otherwise free of symptoms calling for treatment with a compound of Formula XII. In one aspect the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XIII
Figure imgf000016_0001
Formula XIII
wherein 2' is a five- to seven-membered heterocyclic ring, wherein each of X, Y, and Z is independently chosen from C, N, O, and S, and wherein B2' optionally includes at least one atom selected from C, N, O, and S; wherein 2' optionally includes an unsaturated bond; wherein R4, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein A" is hydrogen, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR. In various specific embodiments the compound is any one of compounds 990, 1003, 1091, 1142, 1185, 1212, 1217, 1224, 1244, and 1334 listed in Table 16 below.
In one aspect of the invention, a method of inhibiting TLR-mediated immunostimulatory signaling is provided. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XIII, as provided above, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
In one aspect the invention provides a method of affecting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XIII, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject.
The invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XIII, as provided above, to inhibit TLR-mediated immunostimulation in the subject. In one aspect the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject. The method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula XIII, as provided above, to inhibit an immunostirnulatory nucleic acid-associated response in the subject. In each of the foregoing aspects involving a compound of Formula XIII, in one embodiment A" is a substituted alkyl group selected from the group consisting of a cyclic amino group, an alkylamino group, a dialkylamino group, furyl, phenyl, thienyl, azabicyclooctyl, azabicycloheptyl, and any combination thereof. In one embodiment the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group. In one embodiment the subject is otherwise free of symptoms calling for treatment with a compound of Formula XIII.
In one aspect the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XIV or Formula XV
Figure imgf000018_0001
Formula XIV
Figure imgf000018_0002
Formula XV
wherein Rl, R2, R3, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR. In various specific embodiments the compound is any one of compounds 807, 1163, and 1367 listed in Table 17 below. In one aspect of the invention, a method of inhibiting TLR-mediated immunostimulatory signaling is provided. The method according to this aspect of the invention involves contacting a cell expressing a TLR with an effective amount of a compound of Formula XIV or Formula XV, as provided above, to inhibit TLR-rhediated immunostimulatory signaling in response to a ligand for the TLR.
In one aspect the invention provides a method of affecting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XIV or Forrhula XV, as provided above, to inhibit or promote TLR-mediated immunostimulation in the subject.
The invention in one aspect provides a method of inhibiting TLR-mediated immunostimulation in a subject. The method according to this aspect of the invention involves administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XIV or Formula XV, as provided above, to inhibit TLR-mediated immunostimulation in the subject. In one aspect the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject. The method according to this aspect of the invention involves administering to a subject in need of such treatment an effective amount of a compound of Formula XIV or Formula XV, as provided above, to inhibit an immunostimulatory nucleic acid-associated response in the subject. In one embodiment the subject is otherwise free of symptoms calling for treatment with a compound of Formula XIV or Formula XV. In each of the foregoing aspects of the invention, in some embodiments R2 is a substituted or unsubstituted phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group. In some embodiments R2 is a phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group substituted with one or more substituent groups selected from the group consisting of an alkyl group, an alkoxy group, an alkoxyalkyl group, an ester group, an alkylamino group, a dialkylamino group, a cyclic amino group, a halogen atom, and any combination thereof. In some embodiments R2 is a phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group substituted with one or more substituent groups selected from the group consisting of an alkylamino group, a dialkylamino group, and a cyclic amino group. In some embodiments the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group. In each of the foregoing aspects of the invention, in some embodiments R9 is a substituted alkyl group selected from the group consisting of a cyclic amino group, an alkylamino group, a dialkylamino group, furyl, phenyl, thienyl, azabicyclooctyl, azabicycloheptyl, and any combination thereof. In some embodiments the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group. In the foregoing aspects of the invention, in some embodiments each of R4 and R5 is hydrogen atom. In some embodiments each of R5 and R8 is a hydrogen atom. In some embodiments each of R4, R5, and R8 is a hydrogen atom. In some embodiments RIO is a hydrogen atom. In each of the foregoing aspects of the invention, in one embodiment the TLR is TLR9. In one embodiment the ligand for the TLR is an immunostimulatory nucleic acid. In one embodiment the immunostimulatory nucleic acid is a CpG nucleic acid. In each of the foregoing aspects of the invention, in one embodiment the TLR is
TLR8. In one embodiment the ligand for the TLR is a natural ligand for TLR8. In one embodiment the ligand for the TLR is resiquimod (R848). In each of the foregoing aspects of the invention, in one embodiment the TLR is TLR7. In one embodiment the ligand for the TLR is a natural ligand for TLR7. In one embodiment the ligand for the TLR is R848. In each of the foregoing aspects of the invention, in one embodiment the TLR is TLR3. In one embodiment the ligand for the TLR is a double stranded RNA. In one embodiment the ligand for the TLR is poly(LC). The present invention is based, in part, on the appreciation by the applicants of a potential connection between the antimalarial activity of known metabolites of chloroquine and the biological activity of related small molecules with respect to inhibition of TLR9. For chloroquine, the major metabolites are mono de-ethylated chloroquine and bis de ethylated chloroquine.
Figure imgf000021_0001
chloroquine mono de-ethyl (I) bis de-ethyl (II)
The half life of mono de-ethylchloroquine in humans is 1.5 times longer than that of chloroquine (649 hrs versus 432 hrs). de Vries PJ et al. (1994) Drug Invest 8:143-9. Data for bis de-ethylchloroquine is not available. In addition, the major metabolites of hydroxychloroquine are de-ethylhydroxychloroquine, mono de-ethylchloroquinej and bis de- ethylchloroquine. In the treatment of malaria the potency of chloroquine and mono de- ethylchloroquine against Plasmodium falciparum are about equal whereas bis de- ethylchloroquine is about half as active. Aderounmu AF (1984) Ann Trop Med Parasitol. 78(6):581-5. Also since the side chain is chiral, R and S chloroquine have differing activities with the (S)-(+)- mono de-ethylchloroquine being preferentially produced. Ansari AM et al. (1994) JPharm Sci. 83(7):1040-2. It was thus hypothesized by the applicants that both chloroquine and hydroxychloroquine derive a substantial portion of their efficacy from the long lived de-ethyl metabolite and that this relationship may carry over to the binding to TLR9. It turns out that chloroquine, hydroxychloroquine, mono de-ethylchloroqiliine, and bis de-ethylchloroquine have been examined by Macfarlane. The IC50 of these compounds by 7 1 his assay were: chloroquine, 1.1x10" M; hydroxychloroquine, 4.1x10" M; mono de- ethylchloroquine, 7.08X10'7M; and bis de-ethylchloroquine, 18.6xlO'7M. So by this data the loss of one ethyl group from chloroquine reduced the potency by about 7-fold and the loss of both ethyl groups reduced the potency by about 18-fold.
The present invention in certain aspects is related to compositions and methods involving certain 4-primary amino quinoline compounds which are useful for inhibiting TLR9 signaling and immune activation. In certain aspects the present invention is also related to compositions and methods involving certain quinazoline compounds, some of which are structurally related to the 4-primary amino quinoline compounds of the invention, which are also useful for inhibiting TLR9 signaling and immune activation. It was surprisingly discovered according to the invention that the 4-primary amino quinoline compounds and the quinazoline compounds of the invention are highly and similarly active as inhibitors of TLR9 signaling activity. It was also surprisingly discovered according to the instant invention that despite their high degree of structural and biological similarity, the quinazoline molecules exhibit a significantly improved toxicity profile in vivo compared with the quinoline compounds. In one aspect the invention provides novel substituted 4-primary amino quinoline compositions having a structural Formula XVI
Figure imgf000022_0001
Formula XVI wherein X is absent or is an aryl, alkyl, heterocyclic, or styryl group; Rt and R are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group, wherein Ri and R2 optionally are combined to form' a heterocycle; R3 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein Ri and R3 optionally are combined to form a heterocycle; Y is absent or is an oxygen atom, a sulfur atom, CRgR9, or NR10, where Rs, R9, and R10 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group; L is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; and Rt, R5, Re, and R are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein any pξiir of Rj, R5, R_, and R which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof. In one embodiment R5 and R6 are each independently a halogen atom or an alkoxy group. In one embodiment R5 and R_ are each independently a chlorine atom or a methoxy group. In one embodiment X is absent or is an aryl group; NRiR2 is a heterocyclic amine or is NRg(CH2)nNR9R10, wherein n is an integer from 2 to 6, inclusive, and R8, R , and R10 are each independently a hydrogen atom or an alkyl group; R3 is a hydrogen atom; Y is an aryl group or is NRπ where Rπ is a hydrogen atom or an aryl or alkyl group; L is absent or is a C2-C6 alkyl group; and t, R5, R_, and R7 are each independently a hydrogen atom, a halogen atom, or an alkoxy group. In one embodiment X is absent or is an aryl group; NRiR. is a substituted or unsubstituted piperazino or morpholino group or is
NRs(CH2)nNR9R10, wherein n is an integer from 2 to 6, inclusive, R8 is a hydrogen atom, and R9 and R10 are each independently an alkyl group; R3 is a hydrogen atom; Y is NH; L is a C2-C6 alkyl group; and Rj, R5, R_, and R are each independently a hydrogen atom, a halogen atom, or an alkoxy group. In one embodiment X is a phenyl group;
NRtR2 is
Figure imgf000023_0001
attached to a para position of the phenyl group X; Y is NH; L is -(CH2)n- where n is an integer between 2 and 6, inclusive; and each of R3, R4, R5, R and R is a hydrogen atom. In each of the foregoing embodiments,The composition is optionally in the form of a pharmaceutically acceptable hydrate or salt.
In one aspect the invention provides a method for inhibiting signaling by a TLR. The method according to this aspect of the invention involves contacting a cell expressing a functional TLR with an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII
Figure imgf000024_0001
Formula XVII wherein X is absent or is a nitrogen, oxygen, or sulfur atom or an SO or SO2 group; Ri is a hydrogen atom or a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group; R2 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein R] and R2 optionally are combined to form a heterocycle or carbocycle; Y is absent or is an oxygen atom, a sulfur atom, CR Rs, or NR , where R , R8, and R9 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group; L is absent or is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; and R3, t, R5, and R_ are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein any pair of R3, R4, R5, and R_ which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof, to inhibit signaling by the TLR.
In one aspect the invention provides a method for inhibiting signaling by a TLR. The method according to this aspect of the invention involves contacting an immune cell expressing a functional TLR with (a) an effective amount of a TLR signal agonist to stimulate signaling by the TLR in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to inhibit signaling by the TLR in response to the TLR signal agonist compared with the signaling by the TLR in response to the TLR signal agonist in absence of the substituted 4-primary amino quinoline composition. In one embodiment the TLR is TLR9 and the TLR signal agonist is a TLR9 signal agonist. The TLR signal agonist in one embodiment is CpG DNA, which can be an oligodeoxynucleotide (ODN). In one embodiment the TLR signal agonist is an immune complex. In one embodiment the TLR signal agonist is an immune complex that includes a nucleic acid. In one embodiment the TLR signal agonist is an immune complex that includes a nucleic acid that is self-DNA. In one embodiment the TLR signal agonist is an immune complex that includes a nucleic acid that is self-RNA. In one aspect the invention provides a method for inhibiting an immune response to an antigenic substance. The method according to this aspect of the invention involves contacting an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the substituted 4-primary amino quinoline composition. In one embodiment the immune response is an innate immune response. In one embodiment the immune response includes an adaptive immune response.
In one aspect of the invention, a method of treating an autoimmune disorder in a subject is provided. The method according to this aspect of the invention involves the step of administering to a subject having an autoimmune disorder an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to treat the autoimmune disorder. In one embodiment the autoimmune disorder is chosen from systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Sjogren's syndrome, polymyositis, vasculitis, Wegener's granulomatosis, sarcoidosis, ankylosing spondylitis, Reiter's syndrome, psoriatic arthritis, and Behcet's syndrome. In one particular embodiment the autoimmune disease is systemic lupus erythematosus. In one particular embodiment the autoimmune disease is rheumatoid arthritis. In one embodiment the subject is a human. In one embodiment the autoimmune disorder is an immune complex associated disease.
In one aspect the invention provides novel quinazoline compositions of the invention having a structural Formula XVIII
Figure imgf000026_0001
Foπ nula XVIII wherein X is absent or is an aryl, alkyl, heterocyclic or styryl group, provided that if X is a phenyl group, NR}R2 is part if a heterocycle or is a diamine; R! and R are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein Ri and R2 optionally are combined to form a heterocycle; Y is an oxygen atom, a sulfur atom, CR9R10, or NRπ, where R9, R10, and Rπ are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R9, R10, and Rπ optionally is combined with R3 or t to form a substituted or unsubstituted heterocycle; L is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; R3 and t are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R3 and t optionally are combined to form a heterocycle; and R5, Re, R7, and R8 are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein any pair of R5, Re, R , and R8 which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof. In one embodiment R and R7 are each independently a halogen atom or an alkoxy group. In one embodiment R_ and R are each independently a chlorine atom or a methoxy group. In one embodiment X is absent or is an aryl group; NRiR2 is a heterocyclic amine or NR (CH2)nNR10R11, wherein n is an integer from 2 to 6, inclusive, and R9, R10, and Rπ are each independently a hydrogen atom or ain alkyl group; Y is an aryl group or is NR12 where R12 is a hydrogen atom or an aryl or alkyl group; L is absent or is a C2-C6 alkyl group; R3 and Rt are each independently a hydrogen atom or an alkyl group, wherein R3 and R4 optionally are combined to form a heterocycle; and R5, R_, R , and R8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group. In one embodiment X is absent or is an aryl group; NRiR2 is a substituted or unsubstituted piperazino or morpholino group or is NR (CH2)nNR1oRιι, wherein n is an integer from 2 to 6, inclusive, R9 is a hydrogen atom, and R10 and Rπ are each independently an alkyl group; Y is NH; L is a C2-C6 alkyl group; R3 and R4 are each independently a hydrogen atom or an alkyl group, wherein R3 and Rt optionally are combined to form a heterocycle; and R5, R_, R , and R8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group. In one embodiment X is an aryl group; NRiR2 is substituted or unsubstituted piperazino or morpholino group; Y is NH; L is a C2-C6 alkyl group; R3 and t are each independently a methyl or ethyl group or R3 and t optionally are combined to form a morpholino group; and R5, R_, R , and R8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group. In one embodiment X is a phenyl group; NRiR2 is N-methylpiperazine; Y is NH; L is -CH2CH2-; R3 and t are each a methyl group; and R5, R , R , and R8 are each a hydrogen atom. In one embodiment X is a phenyl group; NRiR2 is N-methylpiperazine; Y is NH; L is -CH2CH2-; R3 and Rt are combined as a morpholino group; and R5, Re, R , and Rs are each a hydrogen atom. In one embodiment X is absent; NR^ is a substituted or unsubstituted piperazino or morpholino group; Y is NH; L is a C2-C6 alkyl group; R3 and t are each independently a methyl or ethyl group or R3 and t optionally are combined to form a morpholino group; and R5, R_, R , and R8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group. In one embodiment X is absent; NRtR is N-methylpiperazine; Y is NH; L is -CH2CH2-; R3 and t are each a methyl group; R_ and R are each a methoxy group; and R5 and R8 are each a hydrogen atom. In one embodiment X is absent; NRiR2 is N-phenylpiperazine; Y is NH; L is -CH2CH2-; R3 and Rt are each a methyl group; Re and R are each a methoxy group; and R5 and Rs are each a hydrogen atom. In one embodiment X is absent; NRjR2 is N-methylpiperazine; Y is NH; L is -CH2CH2; R3 and t are combined as a morpholino group; R and R are each a methoxy group; and R5 and R8 are each a hydrogen atom. In each of the foregoing embodiments, the composition is optionally in the form of a pharmaceutically acceptable hydrate or salt. In one aspect the invention provides a method for inhibiting signaling by a TLR. The method according to this aspect of the invention involves contacting a cell expressing a functional TLR with an effective amount of quinazoline composition having structural Formula XIX
Figure imgf000030_0001
Formula XIX wherein X is a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group, optionally attached to the quinazoline by a nitrogen, oxygen, or sulfur atom or by a SO or SO2 group; Y is absent or is an oxygen atom, a sulfur atom, CR R10, or NRπ, where R9, R10, and Rπ are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R , R10, and Rπ optionally is combined with R3 or Rt to form a heterocycle; L is absent or is a hydrogen atom, an alkyl or alkenyl group containing from 1 to 10 carbons, or an aryl group; R3 and R4 are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R3 and t optionally are combined to form a heterocycle; and R5, R_, R , and R8 are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein any pair of R5, Re, R , and Rs which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof, to inhibit signaling by the TLR.
In one aspect the invention provides a method for inhibiting signaling by a TLR. The method according to this aspect of the invention involves contacting an immune cell expressing a functional TLR with (a) an effective amount of a TLR signal agonist to stimulate signaling by the TLR in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having structural Formula XIX, as defined above, to inhibit signaling by the TLR in response to the TLR signal agonist compared with the signaling by the TLR in response to the TLR signal agonist in absence of the quinazoline composition. In one embodiment the TLR is TLR9 and the TLR signal agonist is a TLR9 signal agonist. The TLR signal agonist in one embodiment is CpG DNA, which can be an oligodeoxynucleotide (ODN). In one embodiment the TLR signal agonist is an immune complex that includes a nucleic acid.
In one aspect the invention provides a method for inhibiting an immune response to an antigenic substance. The method according to this aspect of the invention involves contacting an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having structural Formula XIX, as defined above, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the quinazoline composition. In one embodiment the immune response is an innate immune response. In one embodiment the immune response includes an adaptive immune response.
In one aspect of the invention, a method of treating an autoimmune disorder in a subject is provided. The method according to this aspect of the invention involves the step of administering to a subject having an autoimmune disorder an effective amount of a quinazoline composition having structural Formula XIX, as defined above, to treat the autoimmune disorder. In one embodiment the autoimmune disorder is chosen from systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Sjogren's syndrome, polymyositis, vasculitis, Wegener' s granulomatosis, sarcoidosis, ankylosing spondylitis,
Reiter's syndrome, psoriatic arthritis, and Behcet's syndrome. In one particular embodiment the autoimmune disease is systemic lupus erythematosus. In one particular embodiment the autoimmune disease is rheumatoid arthritis. In one embodiment the subject is a human. In one embodiment the autoimmune disorder is an immune complex associated disease, as described above. BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 is a graph depicting the concentration-dependent inhibition of TLR9 signaling in response to CpG ODN 2006 (5*-TCGTCGTTTTGTCGTTTTGTCGTT-3'; SEQ ID NO:l) by compounds 613 (harmine hydrochloride), 878 (3-hydroxymethyl-beta-carboline, and 470 (norharman), molecules representative of compounds of Formula II. FIG. 2 is a pair of graphs depicting results of an in vivo toxicity assay in mice for specific quinoline and quinazoline compounds. FIG. 2A shows change in mean body weight (grams) and FIG 2B shows mean body weight (grams) over 5 days for mice treated with dimethysulfoxide (DMSO) control (closed circles), compound CMZ 203-43 (also referred to herein as 203-43) administered at 1 mg and 4 mg (open and closed triangles, respectively), compound CMZ 203-34 at 1 mg and 4 mg (open and closed diamonds, respectively), and compound CMZ 203-49 at 1 mg and 4 mg (open and closed inverted triangles, respectively). N=3 for each treatment group. The group receiving CMZ 203-43 at 4 mg were sick and had to be sacrificed on day 3. FIG. 3 is a bar graph depicting total and differential white blood cell counts in mice measured five days after administration of a single intraperitoneal dose of specific quinoline and quinazoline compounds. N=3 for each treatment group. FIG. 4 is a bar graph depicting in vivo inhibition of IP- 10 induction (pg/ml) by CpG ODN 2006 following pretreatment with PBS control or a quinoline compound (203-43) or various indicated quinazoline compounds (203-34, 203-49, or 203-51). N=5 for each group. FIG. 5 is a bar graph depicting in vivo inhibition of TNF-α induction (pg/ml) by CpG
ODN 2006 following pretreatment with PBS control or a quinoline compound (203-43) or various indicated quinazoline compounds (203-34, 203-49, or 203-51). N=5 for each group.
DETAILED DESCRIPTION OF THE INVENTION The present invention provides novel compositions and methods for inhibiting immune responses, including immune responses involved in clinical disorders characterized as autoimmune disorders and immune complex associated diseases. In addition to the novel compositions disclosed herein, which include 4-primary amino quinolines and structurally similar quinazolines, the invention also provides methods for use of previously known compositions within these and other classes of compounds in inhibiting immune responses, including immune responses involved in clinical disorders characterized as autoimmune disorders and immune complex associated diseases. The present invention is based in part on discoveries made by the applicants arising from screening a library of small molecule compounds in an in vitro assay of TLR9 signaling in response to immunostimulatory CpG oligodeoxynucleotides (ODN). It was observed that certain of the small molecules in the library were effective in altering TLR9 signaling in response to CpG ODN. Based on the initial screening results, additional compounds were selected for additional screening. Importantly, the small molecules so identified were distinct from those described by Macfarlane and colleagues. It has now been discovered according to the present invention that certain small molecules characterized by certain minimum features are able to modulate TLR signaling, e.g., in the presence of or in response to a PAMP or other TLR ligand. Some of the small molecules are potent inhibitors of TLR signaling and thus can be used to inhibit TLR- mediated immunostimulation. The minimal features for many, but not all, of the small molecules can be summarized as follows: a core structure including at least two rings, which can be fused or unfused, wherein at least one of the rings contains a nitrogen atom and/or has a side chain that contains a nitrogen atom. Certain of the molecules are effective even without presence of a nitrogen atom. It has been observed that fused three-ring structures such as quinacrine are typically an order of magnitude more potent as inhibitors of TLR-mediated immunostimulation than are fused two-ring structures such as chloroquine. For example, quinacrine is reported to have an EC50 of about 8 nM, where EC50 is the concentration required for half-maximal inhibition of CpG-ODN effect on thymidine uptake by WEHI 231 B-cells in the presence of anti-surface IgM. U.S. Pat. No. 6,479,504 Bl . It has now also been discovered according to the present invention that fused two-ring core structures to which are attached a nitrogen- containing ring substituent can be as potent as fused three-ring structures such as quinacrine. In particular, such compounds can exhibit EC50 in the low nanomolar range. In certain embodiments the nitrogen-containing ring substituent is a C2-C6 alkyl group terminated by a tertiary amine, such as methylethylamine, diethyl amine, dimethyl amine, or a ring containing at least one nitrogen. Without meaning to be bound to any particular theory or mechanism, it is the belief of the instant inventors that both the ring-containing core structure and the side chain substituents contribute to affinity and efficacy. It is believed that molecules with high affinity core structure and low affinity substituents can be as effective as molecules with low affinity core structure and high affinity substituents. Over many years there have been observations that certain common classes of drugs used for purposes other than affecting the immune system in fact have side effects that include immune suppression. For example, the phenothiazine chlorpromazine has been reported to inhibit messenger RNA expression for interleukin 2 (IL-2), tumor necrosis factor alpha (TNF-α), and gamma interferon (IFN-γ). Schleuning MJ et al. (1989) Eur J Immunol 19: 1491-6. In a separate study, chlorpromazine was reported to depress contact hypersensitivity to dinitrochlorobenzene in guinea-pigs. Descotes J et al. (1982) J Neuroimmunol 2:21-5. Yet another study reported that acute, high dose administration of the tricyclic antidepressant drug imipramine inhibits lipopolysaccharide (LPS)-induced increases in TNF-α but has little or no effect on LPS-induced interleukin 1 beta (IL- 1 β) or interleukin 10 (IL-10). Dredge K et al. (1999) Int J Immunopharm 21:663-73. Histamine is a widely recognized mediator of immediate hypersensitivity and inflammation, which are principally mast cell- and basophil-mediated immune phenomena. The pleiotropic effects of histamine are mediated through three types of membrane- associated histamine receptor, histamine HI receptor (H1R), histamine H2 receptor (H2R), and histamine H3 receptor (H3R). Pharmacologic inhibitors for these receptors are known and include pyrilamine and tripelennamine (H1R); cimetidine, famotidine, and ranitidine (H2R); and thioperamide and clobenpropit (H3R). Recently it was reported that signaling through H1R augments T-cell and B-cell antigen receptor-mediated responses. Banu Y et al. (1999) JExp Med 189:673-82. T cells and B cells derived from HlR-deficient (H1R) mice were reported to have normal responses to LPS. Banu et al. also reported that administration of the H2R-specific antagonist famotidine further depressed T-cell and B-cell antigen receptor-mediated responses in H1R"A mice. Jutel and coworkers recently reported that in their study of T cells, the Thl subset of CD4+ T cells preferentially express H1R, while Th2 cells preferentially express H2R. They also reported that histamine enhances Thl -type responses by triggering H1R, and that H1R7" mice have suppressed levels of the Thl cytokine IFN-γ and increased levels of the Th2 cytokines IL-4 and IL-13. Jutel M et al. (2001) Nature 413:420-5. In contrast, with respect to dendritic cells (DC), Mazzoni and others have reported that histamine inhibits IL-12 production and stimulates IL-10 secretion in LPS-stimulated monocyte-derived DC, resulting in a shift from a Thl- to a Th2-polarized immune response. Mazzoni A et al. (2001) JClin Invest 108:1865; Caron G et al. (2001) J Immunol 166:6000-6. Recently Mazzoni et al. also reported their observation that histamine, acting through H2R, inhibits release of type I IFN and TNF-α by plasmacytoid DC (pDC), the principal producers of IFN-α, in response to exposure to CpG ODN or live influenza virus. Mazzoni A et al. (2003) J Immunol 170:2269-73. Finally, it was recently reported that histamine, acting through H2R on monocyte/macrophages, suppresses NADPH oxidase, a key enzyme in oxygen radical formation, resulting in protection of natural killer (NK) cells and T cells against oxygen radical-induced dysfunction and apoptosis. Hellstrand K (2002) Semin Oncol 29(3 Suppl 7):35-40.
Definitions Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. As used herein, the term "adaptive immune response" refers to any type of antigen- specific immune response. Adaptive immune responses, which are also known in the art as specific immune responses, involve lymphocytes are also characterized by immunological memory, whereby response to a second or subsequent exposure to antigen is more vigorous than the response to a first exposure to the antigen. The term adaptive immune response encompasses both humoral (antibody) immunity and cell-mediated (cellular) immunity. As used herein, "allergy" refers to acquired hypersensitivity to a substance (allergen). Allergic conditions include eczema, allergic rhinitis or coryza, hay fever, asthma, urticaria (hives) and food allergies, and other atopic conditions. As used herein, the term "antigenic substance" refers to any substance that induces an adaptive (specific) immune response. An antigen typically is any substance that can be specifically bound by a T-cell antigen receptor, antibody, or B-cell antigen receptor. Antigenic substances include, without limitation, peptides, proteins, carbohydrates, lipids, phospholipids, nucleic acids, autacoids, and hormones. Antigenic substances further specifically include antigens that are classified as allergens, cancer antigens, and microbial antigens. As used herein, "asthma" refers to a disorder of the respiratory system characterized by inflammation, narrowing of the airways and increased reactivity of the airways to inhaled agents. Asthma is frequently, although not exclusively associated with atopic or allergic symptoms. For example, asthma can be precipitated by exposure to an allergen, exposure to cold air, respiratory infection, and exertion. As used herein, the terms "autoimmune disease" and, equivalently, "autoimmune disorder" and " autoimmunity", refer to immunologically mediated acute or chronic injury to a tissue or organ derived from the host. The terms encompass both cellular and antibody- mediated autoimmune phenomena, as well as organ-specific and organ-nonspecific autoimmunity. Autoimmune diseases include insulin-dependent diabetes mellitus, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, atherosclerosis, and inflammatory bowel disease. Autoimmune diseases also include, without limitation, ankylosing spondylitis, autoimmune hemolytic anemia, Behcet's syndrome, Goodpasture's syndrome, Graves' disease, Guillain-Barre syndrome, Hashimoto's thyroiditis, idiopathic thrombocytopenia, myasthenia gravis, pernicious anemia, polyarteritis nodosa, polymyositis/dermatomyositis, primary biliary sclerosis, psoriasis, sarcoidosis, sclerosing cholangitis, Sjogren's syndrome, systemic sclerosis (scleroderma and CREST syndrome), Takayasu's arteritis, temporal arteritis, and Wegener's granulomatosis. Autoimmune diseases also include certain immune complex-associated diseases. As used herein, the terms "cancer" and, equivalently, "tumor" refer to a condition in which abnormally replicating cells of host origin are present in a detectable amount in a subject. The cancer can be a malignant or non-malignant cancer. Cancers or tumors include but are not limited to biliary tract cancer; brain cancer; breast cancer; cervical cancer; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric (stomach) cancer; intraepithelial neoplasms; leukemias; lymphomas; liver cancer; lung cancer (e.g., small cell and non-small cell); melanoma; neuroblastomas; oral cancer; ovarian cancer; pancreatic cancer; prostate cancer; rectal cancer; renal (kidney) cancer; sarcomas; skin cancer; testicular cancer; thyroid cancer; as well as other carcinomas and sarcomas. Cancers can be primary or metastatic. As used herein, the term "CpG DNA" refers to an immunostimulatory nucleic acid which contains a cytosine-guanine (CG) dinucleotide, the C residue of which is unmethylated. The effects of CpG nucleic acids on immune modulation have been described extensively in U.S. patents such as U.S. Pat. Nos. 6,194,388; 6,207,646; 6,239,116; and 6,218,371, and published international patent applications, such as WO98/37919, WO98/40100, WO98/52581, and WO99/56755. The entire contents of each of these patents and published patent applications is hereby incorporated by reference. The entire immunostimulatory nucleic acid can be unmethylated or portions may be unmethylated but at least the C of the 5'-CG-3' must be unmethylated. CpG DNA includes both naturally occurring immunostimulatory nucleic acids, as found in bacterial DNA and plasmids, as well as synthetic oligodeoxynucleotides (ODN). In one embodiment the CpG DNA is a CpG ODN that has a base sequence provided by 5'-TCGTCGTTTTGTCGTTTTGTCGTT-3' (ODN 2006; SEQ ID NO: 1). CpG ODN have been further classified by structure and function into at least the following three classes or types, all of which are intended to be encompassed within the term CpG DNA as used herein: B-class CpG ODN such as ODN 2006 include the originally described immunostimulatory CpG ODN and characteristically activate B cells and NK cells but do not induce or only weakly induce expression of type I interferon (e.g., IFN-α). A- class CpG ODN, described in published PCT international application WO 01/22990, incorporate a CpG motif, include a chimeric phosphodiester/phosphorothioate backbone, and characteristically activate NK cells and induce plasmacytoid dendritic cells to express large amounts of IFN-α but do not activate or only weakly activate B cells. An example of an A- class CpG ODN is 5'-G*G*G_G_G_A_C_G_A_T_C_G_T_C_G*G*G*G*G*G-3' (ODN 2216, SEQ ID NO:2), wherein "*" represents phosphorothioate and "_" represents phosphodiester. C-class CpG ODN incorporate a CpG, include a wholly phosphorothioate backbone, include a GC-rich palindromic or nearly-palindromic region, and are capable of both activating B cells and inducing expression of IFN-α. C-class CpG ODN have been described, for example, in published U.S. patent application 2003/0148976. An example of a C-class CpG ODN is 5'-TCGTCGTTTTCGGCGCGCGCCG-3' (ODN 2395; SEQ ID NO:3). For a review of the various classes of CpG ODN, see also Vollmer J et al. (2004) Eur J Immunol 34:251-62. As used herein, "cytokine" refers to any of a number of soluble proteins or glycoproteins that act on immune cells through specific receptors to affect the state of activation and function of the immune cells. Cytokines include interferons, interleukins, tumor necrosis factor, transforming growth factor beta, colony-stimulating factors (CSFs), chemokines, as well as others. Various cytokines affect innate immumty, acquired immunity, or both. Cytokines specifically include, without limitation, IFN-α, IFN-β, IFN-γ, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12, IL-13, IL-18, TNF-α, TGF-β, granulocyte colony-stimulating factor (G-CSF), and granulocyte-macrophage colony-stimulating factor (GM-CSF). Chemokines specifically include, without limitation, IL-8, IP- 10, 1-TAC, RANTES, MlP-lα, MlP-lβ, Gro-α, Gro-β, Gro-γ, MCP-1, MCP-2, and MCP-3. Most mature CD4+ T helper cells can be categorized into one of two cytokine- associated, cross-regulatory subsets or phenotypes: Thl or Th2. Thl cells are associated with IL-2, IL-3, IFN, GM-CSF and high levels of TNF-α. Th2 cells are associated with IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-13, GM-CSF and low levels of TNF-α. The Thl subset promotes both cell-mediated immunity and humoral immunity that is characterized by immunoglobulin class switching to IgG2ain mice. Thl responses can also be associated with delayed-type hypersensitivity and autoimmune disease. The Th2 subset induces primarily humoral immunity and induces immunoglobulin class switching to IgE and IgGl in mice. The antibody isotypes associated with Thl responses generally have good neutralizing and opsonizing capabilities, whereas those associated with Th2 responses are associated more with allergic responses. Several factors have been shown to influence commitment to Thl or Th2 profiles. The best characterized regulators are cytokines. IL-12 and IFN-γ are positive Thl and negative Th2 regulators. IL-12 promotes IFN-γ production, and IFN- γ provides positive feedback for IL-12. IL-4 and IL-10 appear to be required for the establishment of the Th2 cytokine profile and to down-regulate Thl cytokine production; the effects of IL-4 are in some cases dominant over those of IL-12. IL-13 was shown to inhibit expression of inflammatory cytokines, including IL-12 and TNF-α by LPS-induced monocytes, in a way similar to IL-4. As used herein, "effective amount" refers to any amount that is necessary or sufficient for achieving or promoting a desired outcome. In some instances an effective amount is a therapeutically effective amount. A therapeutically effective amount is any amount that is necessary or sufficient for promoting or achieving a desired biological response in a subject. The effective amount for any particular application can vary depending on such factors as the disease or condition being treated, the particular agent being administered, the size of the subject, or the severity of the disease or condition. One of ordinary skill in the art can empirically determine the effective amount of a particular agent without necessitating undue experimentation. As used herein, "graft rejection" refers to immunologically mediated hyperacute, acute, or chronic injury to a tissue or organ derived from a source other than the host. The term thus encompasses both cellular and antibody-mediated rejection, as well as rejection of both allografts and xenografts. As used herein, the term "immune cell" refers to a cell belonging to the immune system. Immune cells include T lymphocytes (T cells), B lymphocytes (B cells), natural killer (NK) cells, granulocytes, neutrophils, macrophages, monocytes, dendritic cells, and specialized forms of any of the foregoing, e.g., plasmacytoid dendritic cells, plasma cells, NKT, T helper, and cytotoxic T lymphocytes (CTL). As used herein, the term "immune complex" refers to any conjugate including an antibody and an antigen specifically bound by the antibody. In one embodiment the antigen is an autoantigen. As used herein, the term "immune complex comprising a nucleic acid" refers to any conjugate including an antibody and a nucleic acid-containing antigen specifically bound by the antibody. The nucleic acid-containing antigen can include chromatin, ribosomes, small nuclear proteins, histones, nucleosomes, DNA, RNA, or any combination thereof. The antibody can but need not necessarily bind specifically to a nucleic acid component of the nucleic acid-containing antigen. As used herein, the term "immune complex-associated disease" refers to any disease characterized by the production and/or tissue deposition of immune complexes, including, but not limited to systemic lupus erythematosus (SLE) and related connective tissue diseases, rheumatoid arthritis, hepatitis C- and hepatitis B-related immune complex disease (e.g., cryoglobulinemia), Behcet's syndrome, autoimmune glomerulonephritides, and vasculopathy associated with the presence of LDL/anti-LDL immune complexes. As used herein, "immunodeficiency" refers to a disease or disorder in which the subject's immune system is not functioning in normal capacity or in which it would be useful to boost a subject's immune response for example to eliminate a tumor or cancer (e.g., tumors of the brain, lung (e.g., small cell and non-small cell), ovary, breast, prostate, colon, as well as other carcinomas and sarcomas) or an infection in a subject. The immunodeficiency can be acquired or it can be congenital. As used herein, "immunostimulatory nucleic acid-associated response in a subject" refers to a measurable response in a subject associated with administration to the subject of an immunostimulatory nucleic acid. Such responses include, without limitation, elaboration of cytokines, chemokines, growth factors, or immunoglobulin; expression of immune cell surface activation markers; Thl/Th2 skewing; and clinical disease activity. As used herein, the terms "infection" and, equivalently, "infectious disease" refer to a condition in which an infectious organism or agent is present in a detectable amount in the blood or in a normally sterile tissue or normally sterile compartment of a subject. Infectious organisms and agents include viruses, bacteria, fungi, and parasites. The terms encompass both acute and chronic infections, as well as sepsis. As used herein, the term "innate immune response" refers to any type of immune response to certain pathogen-associated molecular patterns (PAMPs). Innate immunity, which is also known in the art as natural or native immunity, involves principally neutrophils, granulocytes, mononuclear phagocytes, dendritic cells, NKT cells, and NK cells. Innate immune responses can include, without limitation, type I interferon production (e.g., IFN-α), neutrophil activation, macrophage activation, phagocytosis, opsonization, complement activation, and any combination thereof. As used herein, the term "self-DNA" refers to any DNA derived from the genome of a host subject. In one embodiment self-DNA includes complementary DNA (cDNA) derived from a host subject. Self-DNA includes intact and degraded DNA. As used herein, the term "self-RNA" refers to any RNA derived from the genome of a host subject. In one embodiment self-RNA is a messenger RNA (mRNA) derived from a host subject. In one embodiment self-RNA includes ribosomal RNA (rRNA) derived from a host subject. Self-RNA includes intact and degraded RNA. As used herein, the term "subject" refers to a vertebrate animal. In one embodiment the subject is a mammal. In one embodiment the subject is a human. In other embodiments the subject is a non-human vertebrate animal, including, without limitation, non-human primates, laboratory animals, livestock, domesticated animals, and non-domesticated animals. As used herein, "subject having or at risk of developing TLR-mediated immunostimulation" refers to a subject exposed to or at risk of exposure to a PAMP or other TLR ligand. As used herein, the terms "Toll-like receptor" and, equivalently, "TLR" refer to any member of a family of at least ten highly conserved mammalian pattern recognition receptor proteins (TLRl - TLR10) which recognize pathogen-associated molecular patterns (PAMPs) and act as key signaling elements in innate immunity. TLR polypeptides share a characteristic structure that includes an extracellular (extracytoplasmic) domain that has leucine-rich repeats, a transmembrane domain, and an intracellular (cytoplasmic) domain that is involved in TLR signaling. TLRs include but are not limited to human TLRs. Nucleic acid and amino acid sequences for all ten currently known human TLRs are available from public databases such as GenBank. Similarly, nucleic acid and amino acid sequences for various TLRs from numerous non-human species are also available from public databases including GenBank. For example, nucleic acid and amino acid sequences for human TLR9 (hTLR9) can be found as GenBank accession numbers AF245704 (coding region spanning nucleotides 145-3243) and AAF78037, respectively. Nucleic acid and amino acid sequences for murine TLR9 (mTLR9) can be found as GenBank accession numbers AF348140 (coding region spanning nucleotides 40-3138) and AAK29625, respectively. The deduced human TLR9 protein contains 1,032 amino acids and shares an overall amino acid identity of 75.5% with mouse TLR9. Like other TLR proteins, human TLR9 contains extracellular leucine-rich repeats (LRRs) and a cytoplasmic Toll interleukin- 1R (TIR) domain. It also has a signal peptide (residues 1-25) and a transmembrane domain (residues 819-836). Nucleic acid and amino acid sequences for human TLR8 (hTLR8) can be found as GenBank accession numbers AF245703 (coding region spanning nucleotides 49-3174) and AAF78036, respectively. Nucleic acid and amino acid sequences for murine TLR8 (mTLR8) can be found as GenBank accession numbers AY035890 (coding region spanning nucleotides 59-3157) and AAK62677, respectively. Nucleic acid and amino acid sequences for human TLR7 (hTLR7) can be found as GenBank accession numbers AF240467 (coding region spanning nucleotides 135-3285) and AAF60188, respectively. Nucleic acid and amino acid sequences for murine TLR7 (mTLR7) can be found as GenBank accession numbers AY035889 (coding region spanning nucleotides 49-3201) and AAK62676, respectively. Nucleic acid and amino acid sequences for human TLR3 (hTLR3) can be found as GenBank accession numbers NM_003265 (coding region spanning nucleotides 102-2816) and NP_003256, respectively. Nucleic acid and amino acid sequences for murine TLR3 (hTLR3) can be found as GenBank accession numbers AF355152 (coding region spanning nucleotides 44-2761 ) and AAK26117, respectively. While hTLRl is ubiquitously expressed, hTLR2, hTLR4 and hTLR5 are present in monocytes, polymorphonuclear phagocytes, and dendritic cells. Muzio M et al. (2000) J Leukoc Biol 67:450-6. Recent publications reported that hTLRl, hTLR6, hTLR7, hTLR9 and hTLRl 0 are present in human B cells. Human TLR7 and hTLR9 are present in plasmacytoid dendritic cells (pDCs), while myeloid dendritic cells express hTLR7 and hTLR8 but not hTLR9. Human TLR8, however, appears not to be expressed in pDCs. As members of the pro-inflammatory interleukin- 1 receptor (IL-1R) family, TLRs share homologies in their cytoplasmic domains called Toll/IL-IR homology (TIR) domains. PCT published applications PCT/US98/08979 and PCT/US01/16766. Intracellular signaling mechanisms mediated by TLRs appear generally similar, with MyD88 and tumor necrosis factor receptor-associated factor 6 (TRAF6) believed to have critical roles. Wesche H et al.
(1997) Immunity 7:837-47; Medzhitov R et al. (1998) Mol Cell 2:253-8; Adachi O et al.
(1998) Immunity 9:143-50; Kawai T et al. (1999) Immunity 11:115-22); Cao Z et al. (1996) Nature 383:443-6; Lomaga MA et al. (1999) Genes Dev 13:1015-24. Signal transduction between MyD88 and TRAF6 is known to involve members of the serine-threonine kinase IL- 1 receptor-associated kinase (IRAK) family, including at least IRAK-1 and IRAK-2. Muzio M et al. (1997) Science 278:1612-5. Briefly, MyD88 is believed to act as an adapter molecule between the TIR domain of a TLR or IL-1R and IRAK (which includes at least any one of IRAK- 1, IRAK-2, IRAK-4, and IRAK-M). MyD88 includes a C-terminal Toll homology domain and an N-terminal death domain. The Toll homology domain of MyD88 binds the TIR domain of TLR or IL- 1R, and the death domain of MyD88 binds the death domain of the serine kinase IRAK. IRAK interacts with TRAF6, which acts as an entryway into at least two pathways, one leading to activation of the transcription factor NF-κB and another leading to activation of Jun and Fos, members of the activator protein- 1 (AP-1) transcription factor family. Activation of NF-κB involves the activation of TAK-1, a member of the MAP 3 kinase (MAPK) family, and IκB kinases. The IκB kinases phosphorylate IκB, leading to its degradation and the translocation of NF-κB to the nucleus. Activation of Jun and Fos is believed to involve MAP kinase kinases (MAPKKs) and MAP kinases ERK, p38, and JNK/SAPK. Both NF-κB and AP-1 are involved in controlling the transcription of a number of key immune response genes, including genes for various cytokines and costimulatory molecules. See Aderem A et al. (2000) Nature 406:782-7; Hacker H et al. (1999) EMBO J 18:6973-82. As used herein, the terms "TLR ligand" and, equivalently, "ligand for a TLR" and "TLR signaling agonist", refer to a molecule, other than a small molecule according to Formula I-XIX described herein or a 4-primary amino quinoline or quinazoline molecule according to the invention, that interacts, directly or indirectly, with a TLR through a TLR domain other than a TIR domain and induces TLR-mediated signaling. In one embodiment a TLR ligand is a natural ligand, i.e., a TLR ligand that is found in nature. In one embodiment a TLR ligand refers to a molecule other than a natural ligand of a TLR, e.g., a molecule prepared by human activity. In one embodiment the TLR is TLR9 and the TLR signal agonist is a CpG nucleic acid. Ligands for many but not all of the TLRs have been described. For instance, it has been reported that TLR2 signals in response to peptidoglycan and lipopeptides. Yoshimura A et al. (1999) J Immunol 163:1-5; Brightbill HD et al. (1999) Science 285:732-6; Aliprantis AO et al. (1999) Science 285:736-9; Takeuchi O et al. (1999) Immunity 11:443-51; Underhill DM et al. (1999) Nature 401 :811-5. TLR4 has been reported to signal in response to lipopolysaccharide (LPS). Hoshino K et al. (1999) J Immunol 162:3749-52; Poltorak A et al. (1998) Science 282:2085-8; Medzhitov R et al. (1997) Nature 388:394-7. Bacterial flagellin has been reported to be a natural ligand for TLR5. Hayashi F et al. (2001) Nature 410:1099- 1103. TLR6, in conjunction with TLR2, has been reported to signal in response to proteoglycan. Ozinsky A et al. (2000) Proc Natl Acad Sci USA 97:13766-71; Takeuchi O et al. (2001) Int Immunol 13:933-40. Recently it was reported that TLR9 is a receptor for CpG DNA. Hernmi H et al. (2000) Nature 408:740-5; Bauer S et al. (2001) Proc Natl Acad Sci USA 98:9237-42. CpG DNA, which includes bacterial DNA and synthetic DNA with CG dinucleotides in which cytosine is unmethylated, is described in greater detail elsewhere herein. Marshak-Rothstein and colleagues also recently reported their finding that TLR9 signaling can occur in certain autoimmune diseases in response to immune complexes containing IgG and chromatin. Leadbetter EA et al. (2002) Nature 416:595-8. Thus, in a broader sense it appears that TLR9 can signal in response to self or non-self nucleic acid, either DNA or RNA, when the nucleic acid is presented in a suitable context, e.g., as part of an immune complex. Recently it was reported that certain imidazoquinoline compounds having antiviral activity are ligands of TLR7 and TLR8. Hemmi H et al. (2002) Nat Immunol 3:196-200; Jurk M et al. (2002) Nat Immunol 3:499. Imidazoquinolines are potent synthetic activators of immune cells with antiviral and antitumor properties. Using macrophages from wildtype and MyD88-deficient mice, Hemmi et al. recently reported that two imidazoquinolines, imiquimod and resiquimod (R848), induce tumor necrosis factor (TNF) and interleukin- 12 (IL-12) and activate NF-κB only in wildtype cells, 'consistent with activation through a TLR. Hemmi H et al. (2002) Nat Immunol 3 : 196-200. Macrophages from mice deficient in TLR7 but not other TLRs produced no detectable cytokines in response to these imidazoquinolines. In addition, the imidazoquinolines induced dose-dependent proliferation of splenic B cells and the activation of intracellular signaling cascades in cells from wildtype but not TLR7 -/- mice. Luciferase analysis established that expression of human TLR7, but not TLR2 or TLR4, in human embryonic kidney cells results in NF-κB activation in response to resiquimod. The findings of Hemmi et al. thus sμggested that these imidazoquinoline compounds are non-natural ligands of TLR7 that jean induce signaling through TLR7. Recently it was reported that R848 is also a ligand for human TLR8. Jurk M et al.
(2002) Nat Immunol 3 :499. > It was recently reported that ligands of TLR3 include poly( C) and double-stranded RΝA (dsRΝA). For purposes of this invention, poly(LC) and double-stranded R A (dsRΝA) are classified as oligonucleotide molecules. By stimulating kidney cells expressing one of a range of TLRs with poly(I:C), Alexopoulou et al. reported that only cells expressing TLR3 respond by activating ΝF-κB. Alexopoulou L et al. (2001) Nature 413: 732-8. Alexopoulou et al. also reported that wildtype cells stimulated with poly(I:C) activate NF-κB and produce inflammatory cytokines IL-6, IL-12, and TNF-α, whereas the corresponding responses of TLR3 cells were significantly impaired. In contrast, TLR3'7" cells responded equivalently to wildtype cells in response to lipopolysaccharide, peptidoglycan, and CpG dinucleotides. Analysis of MyD88_/" cells indicated that this adaptor protein is involved in dsRNA-induced production of cytokines and proliferative responses, although activation of NF-κB and MAP kinases are not affected, indicating distinct pathways for these cellular responses. Alexopoulou et al. proposed that TLR3 may have a role in host defense against viruses. As used herein, a "cell expressing a TLR" refers to any cell which expresses, either naturally or artificially, a functional TLR. A functional TLR is a full-length TLR protein or a fragment thereof capable of inducing a signal in response to interaction with its ligand. Generally the functional TLR will include at least a TLR ligand-binding fragment of the extracellular domain of the full-length TLR and at least a fragment of a TIR domain capable of interacting with another Toll homology domain-containing polypeptide, e.g., lv_yD88. In various embodiments the functional TLR is a full-length TLR selected from TLRl , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, and TLR10. In certain embodiments the functional TLR is naturally expressed by a cell. In one embodiment the cell naturally expresses functional TLR and is an isolated cell from human multiple myeloma cell line RPMI 8226 (ATCC CCL-155). This cell line was established from the peripheral blood of a 61 year old man at the time of diagnosis of multiple myeloma (IgG lambda type). Matsuoka Y et al. (1967) Proc Soc Exp Biol Med 125:1246-50. RPMI 8226 was previously reported as responsive to CpG nucleic acids as evidenced by the induction of IL-6 protein and IL-12p40 mRNA. Takeshita F et al. (2000) Eur J Immunol 30:108-16; Takeshita F et al. (2000) Eur J Immunol 30:1967-76. Takeshita et al. used the cell line solely to study promoter constructs in order to identify transcription factor binding sites important for CpG nucleic acid signaling. It is now known that RPMI 8226 cells secrete a number of other chemokines and cytokines including IL-8, IL-10 and IP- 10 in response to immunostimulatory nucleic acids. Because this cell line expresses TLR9, through which immunostimulatory nucleic acids such as for example CpG nucleic acids mediate their effects, it is a suitable cell line for use in the methods of the invention relating to CpG nucleic acids as reference and test compounds, as well as to other TLR9 ligands. Similar to peripheral blood mononuclear cells (PBMCs), the RPMI 8226 cell line has been observed to upregulate its cell surface expression of markers such as CD71, CD86 and HLA-DR in response to CpG nucleic acid exposure. This has been observed by flow cytometric analysis of the cell line. Accordingly, the methods provided herein can be structured to use appropriately selected cell surface marker expression as a readout, in addition to or in place of chemokine or cytokine production or other readouts described elsewhere herein. The RPMI 8226 cell line has also been found to respond to certain small molecules including imidazoquinoline compounds. For example, incubation of RPMI 8226 cells with the imidazoquinoline compound R848 (resiquimod) induces IL-8, IL-10, and IP-10 production. It has recently been reported that R848 mediates its immunostimulatory effects through TLR7 and TLR8. The ability of RPMI 8226 to respond to R848 suggests that the RPMI 8226 cell line also expresses TLR7, as previously reported for normal human B cells. The RPMI cell line can be used in unmodified form or in a modified form. In one embodiment, the RPMI 8226 cell is transfected with a reporter construct. Preferably, the cell is stably transfected with the reporter construct. The reporter construct generally includes a promoter, a coding sequence and a polyadenylation signal. The coding sequence can include a reporter sequence selected from the group consisting of an enzyme (e.g., luciferase, alkaline phosphatase, beta-galactosidase, chloramphenicol acetyltransferase (CAT), secreted alkaline phosphatase, etc.), a bioluminescence marker (e.g., green fluorescent protein (GFP, U.S. Pat. No. 5,491,084), etc.), a surface-expressed molecule (e.g., CD25), a secreted molecule (e.g., IL-8, IL-12 p40, TNF-α, etc.), and other detectable protein products known to those of skill in the art. Preferably, the coding sequence encodes a protein having a level or an activity tha is quantifiable. In certain embodiments the functional TLR is artificially expressed (including over- expressed) by a cell, for example by introduction into the "cell of an expression vector bearing a coding sequence for the functional TLR wherein the coding sequence is operably linked to a gene expression sequence. As used herein, a coding sequence and the gene expression sequence are said to be operably linked when they are covalently linked in such a way as to place the expression or transcription and/or translation of the coding sequence under the influence or control of the gene expression sequence. Two DNA sequences are said to be operably linked if induction of a promoter in the 5' gene expression sequence results in the transcription of the coding sequence and if the nature of the linkage between the two DNA sequences does not (1) result in the introduction of a frame-shift mutation, (2) interfere with the ability of the promoter region to direct the transcription of the coding sequence, or (3) interfere with the ability of the corresponding RNA transcript to be translated into a protein. Thus, a gene expression sequence would be operably linked to a coding sequence if the gene expression sequence were capable of effecting transcription of that coding sequence such that the resulting transcript is translated into the desired protein or polypeptide. In some embodiments a coding sequence refers to a nucleic acid sequence coding for a functional TLR. In some embodiments a coding sequence refers to a nucleic acid sequence coding for a reporter. A cell that artificially expresses a functional TLR can be a cell that does not express the functional TLR but for the TLR expression vector. For example, human 293 fibroblasts (ATCC CRL-1573) do not express TLR3, TLR7, TLR8, or TLR9. As described in the examples below, such cells can be transiently or stably transfected with suitable expression vector (or vectors) so as to yield cells that do express TLR3, TLR7, TLR8, TLR9, or any combination thereof. Alternatively, a cell that artificially expresses a functional TLR can be a cell that expresses the functional TLR at a significantly higher level with the TLR expression vector than it does without the TLR expression vector. For use in the methods of the instant invention, a cell that artificially expresses a functional TLR is preferably a stably transfected cell that expresses the functionήl TLR. Such a cell can also be stably transfected with a suitable reporter construct. As used herein, "TLR-mediated signaling" refers to any portion of the intracellular signal transduction pathway involving interaction of a TLR with a suitable TLR ligand and engagement by the TLR of MyD88 or any aspect downstream of MyD88 engagement, described above in relation to the structure and function of the TIR domain of a TLR. As used herein, "TLR-mediated immunostimulatory signaling" refers to TLR- mediated signaling that results in a measurable immunostimulatory response. This term applies to such signaling both in vitro and in vivo. As used herein, "TLR-mediated immunostimulation in a subject" refers to TLR- mediated immunostimulatory signaling as it applies in vivo. As used herein, the term "treat" as used in reference to a disorder, disease, or condition means to intervene in such disorder, disease, or condition so as to prevent or slow the development of, to prevent, slow or halt the progression of, or to eliminate the disorder, disease, or condition.
Immunostimulatory Nucleic Acids As used herein, "immunostimulatory nucleic acid" refers to a nucleic acid molecule that, when contacted with a cell of the immune system, induces the cell of the immune system to become activated. Immune cell activation can be determined using any suitable means known to one of skill in the art, including but not limited to measurement of stimulated growth, proliferation, differentiation, migration, transcription or expression of gene products that are expressed or secreted in association with immune activation, activity of intracellular signaling pathways, and the like. Examples of markers of immune cell activation include, without limitation, secretion of cytokines (e.g., IL-6), secretion of immunoglobulin (e.g., IgG), expression of cell surface antigens (e.g., CD86), induction of cytolytic activity, and induction and nuclear translocation of transcription factors (e.g., NF-κB). A "nucleic acid" as used herein with respect to the methods of the invention, shall refer to any polymer of two or more individual nucleoside or nucleotide units. Nucleic acids can be single- or double-stranded. Typically individual nucleoside or nucleotide units will include any one or combination of deoxyribonucleosides, ribonucleosides, deoxyribonucleotides, and ribonucleotides. The individual nucleotide or nucleoside units of the nucleic acid can be naturally occurring or not naturally occurring. For example, the individual nucleotide units can include deoxyadenosine, deoxycytidine, deoxyguanosine, thymidine, and uracil. In addition to naturally occurring 2'-deoxy and 2'-hydroxyl forms, individual nucleosides also include synthetic nucleosides having modified base moieties and/or modified sugar moieties, e.g., as described in Uhlmann E et al. (1990) Chem Rev 90:543-84. The linkages between individual nucleotide or nucleoside units can be naturally occurring or not naturally occurring. For example, the linkages can be phosphodiester, phosphorothioate, phosphorodithioate, phosphoramidate, as well as peptide linkages and other covalent linkages, known in the art, suitable for joining adjacent nucleoside or nucleotide units. Immunostimulatory nucleic acids typically range in size from 3-4 units to a few tens of units, e.g., 18-40 units, although longer nucleic acids are also contemplated by the invention. In some embodiments the nucleic acids are oligonucleotides made up of 2 to about 100 nucleotides, and more typically 4 to about 40 nucleotides. Oligonucleotides composed exclusively of deoxyribonucleotides are termed oligodeoriboxynucleotides or, equivalently, oligodeoxynucleotides (ODN). An immunostimulatory nucleic acid includes any of a number of different types of immunostimulatory nucleic acids, including specifically immunostimulatory CpG nucleic acids (CpG nucleic acids), including but not limited to types A, B, and C; and immunostimulatory non-CpG nucleic acids, including without limitation methylated CpG nucleic acids, T-rich nucleic acids, and poly-G nucleic acids. Certain of these various classes of immunostimulatory nucleic acids can coexist in a given nucleic acid molecule. As used herein, the terms "CpG nucleic acid" and, equivalently, "CpG ODN" refer to an immunostimulatory nucleic acid which contains a cytosine-guanine (CG) dinucleotide, the C residue of which is unmethylated. The effects of CpG nucleic acids on immune modulation have been described extensively in U.S. patents such as U.S. Pat. Nos. 6,194,388; 6,207,646; 6,218,371; and 6,239,116, and published international patent applications, such as WO 98/37919, WO 98/40100, WO 98/52581, and WO 99/56755. The entire contents of each of these patents and published patent applications is hereby incorporated by reference. The entire immunostimulatory nucleic acid can be unmethylated or portions may be unmethylated but at least the C of the 5'-CG-3' must be unmethylated. The CpG nucleic acid sequences of the invention include those broadly described above as well as disclosed in U.S. Pat. Nos. 6,207,646 B 1 and 6,239, 116 B 1. In one embodiment the CpG nucleic acid has a base sequence provided by 5'-TCGTCGTTTTGTCGTTTTGTCGTT-3' (ODN 2006; SEQ ID NO:l). CpG nucleic acids have been further classified by structure and function into at least the following three types, all of which are intended to be encompassed within the methods of the instant invention: Type B CpG nucleic acids such as ODN 2006 include the earliest described CpG nucleic acids and characteristically activate B cells but do not induce or only weakly induce expression of IFN-α. Type A CpG nucleic acids, described in published international application PCT/US00/26527 (WO 01/22990), incorporate a CpG motif, include a hybrid phosphodiester/phosphorothioate backbone, and characteristically induce plasmacytoid dendritic cells to express large amounts of IFN-α but do not activate or only weakly activate B cells. Type C oligonucleotides incorporate a CpG, include a chimeric backbone, include a GC-rich palindromic or nearly-palindromic region, and are capable of both activating B cells and inducing expression of IFN-α. These have been described, for example, in published U.S. patent application 2003/0148976. In other embodiments of the invention, a non-CpG nucleic acid is used. A non-CpG nucleic acid is an immunostimulatory nucleic acid which either does not have a CpG motif in its sequence, or has a CpG motif which contains a methylated C residue. In some instances, the non-CpG nucleic acid may still be immunostimulatory by virtue of its having other immunostimulatory motifs such as those described herein and known in the art. In one embodiment the non-CpG nucleic acid is a methylated CpG nucleic acid. In som|e instances the non-CpG nucleic acid is still immunostimulatory despite methylation of the C of the CpG motif, even without having another non-CpG immunostimulatory motif described herein and known in the art. In one embodiment the non-CpG nucleic acid is a methylated CpG nucleic acid having a base sequence provided by 5'-TZGTZGTTTTGTZGTTTTGTZGTT-3' (ODN 2117; SEQ ID NO:4, wherein Z represents 5-methylcytidine). Non-CpG nucleic acids include T-rich immunostimulatory nucleic acids. The T-rich immunostimulatory nucleic acids include those disclosed in published PCT patent application PCT/USOO/26383, the entire contents of which are incorporated herein by reference. In some embodiments, T-rich nucleic acids 24 bases in length are used. A T-rich nucleic acid is a nucleic acid which includes at least one poly T sequence and/or which has a nucleotide composition of greater than 25% T nucleotide residues. A nucleic acid having a poly-T sequence includes at least four Ts in a row, such as 5'-TTTT-3'. In some embodiments the T- rich nucleic acid includes more than one poly T sequence. In important embodiments, the T- rich nucleic acid may have 2, 3, 4, or more poly T sequences, such as ODN 2006. Non-CpG nucleic acids also include poly-G immunostimulatory nucleic acids. A variety of references describe the immunostimulatory properties of poly-G nucleic acids. Pisetsky DS et al. (1993) Mol Biol Reports 18:217-221; Krieger M et al. (1994) Ann Rev Biochem 63:601-637; Macaya RF et al. (1993) Proc Natl Acad Sci USA 90:3745-3749; Wyatt JR et al. (1994) Proc Nat! Acad Sci USA 91:1356-1360; Rando and Hogan, 1998, In Applied Antisense Oligonucleotide Technology, Krieg and Stein, eds., pp. 335-352; Kimura Y et al. (1994) J Biochem (Tokyo) 116:991-994. The immunostimulatory nucleic acids of the invention can also be those which do not possess CpG, methylated CpG, T-rich, or poly-G motifs. Exemplary immunostimulatory nucleic acid sequences further include but are not limited to those immunostimulatory sequences described and listed in published PCT patent application WO 01/22972.
In one aspect the invention provides novel compounds that fall within Formula VI and Formula VII as disclosed herein. These compounds include various diarylmethane TLR9 antagonists denoted herein as CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91. Syntheses for each of these compounds are provided in Examples 12-17, respectively. Similar to other compounds of Formula VI and Formula VII, these specific compounds were tested in vitro and found to inhibit TLR9 signaling. See Example 18. Thus compounds CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91 are believed to be useful in the methods of the invention. More specifically, in one aspect the invention provides a method of affecting TLR-mediated signaling in response to a TLR ligand. The method includes the step of contacting a cell expressing a TLR with an effective amount of any one or combination of compounds CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR. In one aspect the the invention provides a method of inhibiting TLR-mediated signaling in response to a TLR ligand. The method includes the step of contacting a cell expressing a TLR with an effective amount of any one or combination of compounds CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR. In one aspect the invention provides a method of affecting TLR-mediated immunostimulation in a subject. The method includes the step of administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of any one or combination of compounds CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91, to inhibit or promote TLR- mediated immunostimulation in the subject. In one aspect the invention provides a method of inhibiting TLR-mediated immunostimulation in a subject. The method includes the step of administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of any one or combination of compounds CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91, to inhibit TLR-mediated immunostimulation in the subject. In one aspect the invention provides a method of inhibiting an immunostimulatory nucleic acid-associated response in a subject. The method according to this aspect includes the step of administering to a subject in need of such treatment an effective amount of any one or combination of compounds CMZ 203-84, CMZ 203-85, CMZ 203-88, CMZ 203-88-1, CMZ 203-89, and CMZ 203-91, to inhibit an immunostimulatory nucleic acid-associated response in the subject. In one aspect the invention provides novel substituted 4-primary amino quinoline compositions. As described further below, these compositions and other substituted 4-primary amino quinoline compositions have been discovered to be useful in methods for inhibiting an immune response, both in vitro and in vivo, including methods for treating immune complex associated diseases and autoimmune disorders. Due to their similarity to certain known antimalarial agents, it is also believed that the novel substituted 4-primary amino quinoline compositions of the invention will also be useful for prevention and treatment of malaria, as well as for treatment of other diseases which have been described to be responsive to treatment with chloroquines. The novel substituted 4-primary amino quinoline compositions of the invention have a structural Formula XVI
Figure imgf000052_0001
Formula XVI wherein X is absent or is an aryl, alkyl, heterocyclic, or styryl group; R! and R2 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group, wherein R\ and R optionally are combined to form a heterocycle; R3 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein R\ and R3 optionally are combined to form a heterocycle or a carbocycle; Y is absent or is an oxygen atom, a sulfur atom, CRgR , or NRio, where R8, R9, and R10 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group; L is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; and Rt, R5, Re, and R7 are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein any pair of Rt, R5, Re, and R which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof. In one embodiment R5 and Rg are each independently a halogen atom or an alkoxy group. In one embodiment R5 and Rg are each independently a chlorine atom or a methoxy group. In one embodiment X is absent or is an aryl group; NR]R2 is a heterocyclic amine or is NR8(CH2)nNR Rιo, wherein n is an integer from 2 to 6, inclusive, and Rg, R , and R10 are each independently a hydrogen atom or an alkyl group; R3 is a hydrogen atom; Y is an aryl group or is NRπ where Rπ is a hydrogen atom or an aryl or alkyl group; L is absent or is a C -C6 alkyl group; and Rt, R5, Rg, and R are each independently a hydrogen atom, a halogen atom, or an alkoxy group. In one embodiment X is absent or is an aryl group; NRtR2 is a substituted or unsubstituted piperazino or morpholino group or is
Figure imgf000054_0001
wherein n is an integer from 2 to 6, inclusive, R8 is a hydrogen atom, and R and R10 are each independently an alkyl group; R3 is a hydrogen atom; Y is NH; L is a C2-Cg alkyl group; and Rt, R5, R_, and R7 are each independently a hydrogen atom, a halogen atom, or an alkoxy group. In one embodiment X is a phenyl group;
Figure imgf000054_0002
NRtR2 is attached to a para position of the phenyl group X; Y is NH; L is -(CH2)n- where n is an integer between 2 and 6, inclusive; and each of R3, Rt, R , R<5, and R7 is a hydrogen atom. In each of the foregoing embodiments, the composition is optionally in the form of a pharmaceutically acceptable hydrate or salt. Representative, non-limiting examples of substituted 4-primary amino quinoline compositions of Formula XVI of the invention are compounds 101-104, 106-109, 111, 113- 116, and 118-119, presented in Table 1.
Table 1. Substituted 4-Primary Amino Quinoline Compositions of the Invention
Figure imgf000054_0003
Figure imgf000055_0001
It has been discovered according to the invention that 4-primary amino quinoline compositions, similar to 4-secondary and 4-tertiary amino quinoline compounds, can be used to inhibit TLR9 signaling.
In one aspect the invention provides a method for inhibiting signaling by a TLR. The method according to this aspect of the invention involves contacting a cell expressing a functional TLR with an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII
Figure imgf000056_0001
Formula XVII wherein X is absent or is a nitrogen, oxygen, or sulfur atom or an SO or SO2 group; Ri is a hydrogen atom or a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group; R2 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein R] and R2 optionally are combined to form a heterocycle or carbocycle; Y is absent or is an oxygen atom, a sulfur atom, CR Rs, or NR , where R7, R8, and R9 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group; L is absent or is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; and R3, Rt, Rs, and Re are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein any pair of R3, Rt, R5, and R$ which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof, to inhibit signaling by the TLR. The substituted 4-primary amino quinoline composition in this and all other aspects of the invention involving the use of a substituted 4-primary amino quinoline composition can be in the form a hydrate or pharmaceutically acceptable salt. The method according to this aspect of the invention can be performed in vitro or it can be performed in vivo. In addition, the cell expressing the functional TLR can, but need not necessarily, be an immune cell. For vector that directs expression of the TLR by the cell. In one embodiment the TLR is TLR9 and the method is thus a method for inhibiting intracellular signaling by TLR9. In addition to compounds 101-104, 106-109, 111, 113-116, and 118-119, presented in Table 1, the following additional representative and non-limiting substituted 4-primary amino quinoline compounds, compounds 105, 110, 117, and 120-133, presented in Table 2 with reference to Formula XVII, can be used in the method according to this and all other aspects of the invention directed to methods involving use of substituted 4-primary amino quinoline compounds. Additional examples are presented in the Examples below. Table 2. Additional Substituted 4-Primary Amino Quinoline Compositions for Use in Methods of the Invention
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
In one aspect the invention provides a method for inhibiting signaling by a TLR. The method according to this aspect of the invention involves contacting an immune cell expressing a functional TLR with (a) an effective amount of a TLR signal agonist to stimulate signaling by the TLR in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to inhibit signaling by the TLR in response to the TLR signal agonist compared with the signaling by the TLR in response to the TLR signal agonist in absence of the substituted 4-primary amino quinoline composition. The substituted 4-primary amino quinoline composition in this aspect of the invention can be in the form a hydrate or pharmaceutically acceptable salt. The method according to this aspect of the invention can be performed in vitro or it can be performed in vivo. Also according to this aspect of the invention, in one embodiment the TLR is TLR9 and the TLR signal agonist is a TLR9 signal agonist, and the method in one embodiment is thus a method of inhibiting intracellular signaling by TLR9 in response to a TLR9 signal agonist. The TLR signal agonist in one embodiment is CpG DNA, for example a CpG ODN such as ODN 2006. The CpG ODN can belong to any class of CpG ODN, including A-class (e.g., ODN 2216), B-class (e.g., ODN 2006), or C-class (e.g., ODN 2395). In one embodiment the TLR signal agonist is an immune complex that includes a nucleic acid. Immune complexes that include a nucleic acid are known by those of skill in the art to include immunoglobulin complexed with nucleic acids that are either naked or, more commonly, that are associated with proteins or other non-nucleic acid components. The nucleic acids that are associated with proteins or other non-nucleic acid components can include, for example, chromatin, ribosomes, small nuclear proteins, ribonuclear proteins (RNP), histones, nucleosomal protein-DNA complexes, and nucleosomes. Examples of clinically important antibodies specific for nucleic acids and for nucleic acid-containing material include antinuclear antibodies (ANA), anti-dsDNA, anti-ssDNA, anti-Sm, anti-RNP, anti-Ro (SS-A), anti-La (SS-B), and antihistone antibodies. Immune complexes that include a nucleic acid include, without limitation, immunoglobulin complexed with DNA, including specifically double-stranded DNA, and immunoglobulin complexed with nucleosomal material, both characteristic of systemic lupus erythematosus, and immunoglobulin complexed with RNA. In one aspect the invention provides a method for inhibiting an immune response to an antigenic substance. The method according to this aspect of the invention involves contacting an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the substituted 4-primary amino quinoline composition. The substituted 4-primary amino quinoline composition in this aspect of the invention can be in the form a hydrate or pharmaceutically acceptable salt. The method according to this aspect of the invention can be performed in vitro or it can be performed in vivo. In one embodiment the immune response is an innate immune response. In one embodiment the immune response includes an adaptive immune response. ; In one embodiment the method involves contacting, in a subject, an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response in the subject to the antigenic substance in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to inhibit an immune response in the subject to the antigenic substance compared with the immune response to the antigenic substance in absence of the substituted 4-primary amino quinoline composition. In one embodiment the step of contacting an immune cell expressing a functional
Toll-like receptor with an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a substituted 4-primary amino quinoline composition involves the active step of administering an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a substituted 4-primary amino quinoline composition. The antigenic substance can be administered using any effective route or means for administering the antigenic substance to effect the contacting. By way of example, the administering can be by local or systemic injection, inhalation, oral ingestion, topical administration, mucosal administration, or any combination thereof. In one embodiment the step of contacting an immune cell expressing a functional Toll-like receptor with an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a substituted 4-primary amino quinoline composition is passive. For example, in one embodiment the immune cell is already in contact or has already been in contact with an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance, at the time of or prior to the step of contacting the immune cell expressing a functional Toll-like receptor with an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the substituted 4-primary amino quinoline composition. The antigenic substance can be an allergen. An allergen is a substance that can induce an allergic or asthmatic response in a susceptible subject. The list of allergens is enormous and can include pollens, insect venoms, animal dander, dust, fungal spores and drugs (e.g., penicillin). Examples of natural animal and plant allergens include proteins specific to the following genera: Canis (Ca is familiaris); Dermatophagoides (e.g., Dermatophagoides farinae); Felis (Felis domesticus); Ambrosia {Ambrosia artemiisfolia); Lolium (e.g., Lolium perenne or Lolium multiflorum); Cryptomeria (Cryptomeriajaponicd); Alternaria (Alternaria alternata); Alder; Alnus (Alnus gultinosa); Betula (Betula verrucosd); Quercus (Quercus alba); Olea (Olea europa); Artemisia (Artemisia vulgaris); Plantago (e.g., Plantago lanceolata); Parietaria (e.g., Parietaria officinalis or Par ietaria judaica); Blattella (e.g., Blattella germanica); Apis (e.g., Apis multiforum); Cupressus (e.g., Cupressus sempervirens, Cupressus arizonica, and Cupressus macrocarpd); Juniperus (e.g., Juniperus sabinoides, Juniperus virginiana, Juniperus communis, and Juniperus ashel); Thuya (e.g., Thuya orientalis); Chamaecyparis (e.g., Chamaecyparis obtusa); Periplaneta (e.g., Periplaneta americana); Agropyron (e.g., Agropyron repens); Secale (e.g., Secale cereale); Triticum (e.g., Triticum aestivum); Dactylis (e.g., Dactylis glomerata); Festuca (e.g., Festuca elatior); Poa (e.g., Poa pratensis or Poa compressa); Avena (e.g., Avena sativa); Holcus (e.g., Holcus lanatus); Anthoxanthum (e.g., Anthoxanthum odoratum); Arrhenatherum (e.g., Arrhenatherum elatius); Agrostis (e.g., Agrostis alba); Phleum (e.g., Phleum pratense); Phalaris (e.g., Phalaris arundinacea); Paspalum (e.g., Paspalum notatum); Sorghum (e.g., Sorghum halepensis); and Bromus (e.g., Bromus inermis). The term "allergy" refers to acquired hypersensitivity to a substance (allergen). An "allergic reaction" is the response of an immune system to an allergen in a subject allergic to the allergen. Allergic conditions include eczema, allergic rhinitis or coryza, hay fever, bronchial asthma, urticaria (hives) and food allergies, and other atopic conditions. The antigenic substance can be an antigen that is or is derived from an infectious microbial agent, including a bacterium, a virus, a fungus, or a parasite. Examples of infectious bacteria include: Helicobacter pyloris, Borrelia burgdorferi, Legionella pneumophilia, Mycobacteria sps (such as. M. tuberculosis, M. avium, M. intracellulare, M. kansasii, and M. gordonae), Staphylococcus aureus, Neisseria gonorrhoeae, Neisseria meningitidis, Listeria monocytogenes, Streptococcus pyogenes (Group A Streptococcus), Streptococcus agalactiae (Group B Streptococcus), Streptococcus (viridans group), Streptococcus faecalis, Streptococcus bovis, Streptococcus (anaerobic sps.), Streptococcus pneumoniae, pathogenic Campylobacter sp., Enterococcus sp., Haemophilus influenzae, Bacillus anthracis, Corynebacterium diphtheriae, Corynebacterium sp., Erysipelothrix rhusiopathiae, Clostridium perfringens, Clostridium tetani, Enterobacter aerogenes, Klebsiella pneumoniae, Pasturella multocida, Bacteroides sp., Fusobacterium nucleatum, Streptobacillus moniliformis, Treponema pallidum, Treponema pertenue, Leptospira, and Actinomyces israelii. Examples of infectious virus include: Retro viridae (including but not limited to human immunodeficiency virus (HIV)); Picornaviridae (for example, polio viruses, hepatitis A virus; enteroviruses, human coxsackie viruses, rhinoviruses, echoviruses); Calciviridae (such as strains that cause gastroenteritis); Togaviridae (for example, equine encephalitis viruses, rubella viruses); Flaviviridae (for example, dengue viruses, encephalitis viruses, yellow fever viruses); Coronaviridae (for example, coronaviruses); Rhabdoviridae (for example, vesicular stomatitis viruses, rabies viruses); Filoviridae (for example, ebola viruses); Paramyxoviridae (for example, parainfluenza viruses, mumps virus, measles virus, respiratory syncytial virus); Orthomyxoviridae (for example, influenza viruses); Bunyaviridae (for example, Hantaan viruses, bunya viruses, phleboviruses, and Nairo viruses); Arena viridae (hemorrhagic fever viruses); Reoviridae (e.g., reoviruses, orbiviurses, and rotaviruses); Birnaviridae; Hepadnaviridae (Hepatitis B virus); Parvoviridae (parvoviruses); Papovaviridae (papilloma viruses, polyoma viruses); Adenoviridae (most adeno viruses); Herpesviridae (herpes simplex virus (HSV) 1 and HSV-2, varicella zoster virus, cytomegalovirus (CMV), he es viruses); Poxviridae (variola viruses, vaccinia viruses, pox viruses); and Iridoviridae (such as African swine fever virus); and unclassified viruses (for example, the etiological agents of spongiform encephalopathies, the agent of delta hepatitis (thought to be a defective satellite of hepatitis B virus), the agents of non-A, non-B hepatitis (class l=internally transmitted; class 2=parenterally transmitted (i.e., Hepatitis C); Norwalk and related viruses, and astro viruses). Examples of infectious fungi include, but are not limited to, Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis, and Candida albicans. The antigenic substance can be a cancer antigen. A cancer antigen as used herein is a compound, such as a peptide or protein, associated with a tumor or cancer cell surface and which is capable of provoking an immune response when expressed on the surface of an antigen-presenting cell in the context of a major histocompatibility complex (MHC) molecule. Cancer antigens can be prepared from cancer cells either by preparing crude extracts of cancer cells, for example, as described in Cohen PA et al. (1994) Cancer Res 54: 1055-8, by partially purifying the antigens, by recombinant technology, or by de novo synthesis of known antigens. Cancer antigens include but are not limited to antigens that are recombinantly expressed, an immunogenic portion thereof, or a whole tumor or cancer cell. Such antigens can be isolated or prepared recombinantly or by any other means known in the art. The terms "cancer antigen" and "tumor antigen" are used interchangeably and refer to antigens which are differentially expressed by cancer cells and can thereby be exploited in order to target cancer cells. Cancer antigens are antigens which can potentially stimulate apparently tumor-specific immune responses. Some of these antigens are encoded, although not necessarily expressed, by normal cells. These antigens can be characterized as those which are normally silent (i.e., not expressed) in normal cells, those that are expressed only at certain stages of differentiation and those that are temporally expressed such as embryonic and fetal antigens. Other cancer antigens are encoded by mutant cellular genes, such as oncogenes (e.g., activated ras oncogene), suppressor genes (e.g., mutant p53), fusion proteins resulting from internal deletions or chromosomal translocations. Still other cancer antigens can be encoded by viral genes such as those carried on RNA and DNA tumor viruses. Examples of tumor antigens include MAGE, MART-1/Melan-A, gplOO, Dipeptidyl peptidase IV (DPPIV), adenosine deaminase-binding protein (ADAbp), cyclophilin b, Colorectal associated antigen (CRC)~C017-1A/GA733, Carcinoembryonic Antigen (CEA) and its immunogenic epitopes CAP-1 and CAP-2, etv6, amll, Prostate Specific Antigen (PSA) and its immunogenic epitopes PSA-1, PSA-2, and PSA-3, prostate-specific membrane antigen (PSMA), T-cell receptor/CD3-zeta chain, MAGE-family of tumor antigens (e.g., MAGE-A1 , MAGE-A2, MAGE-A3, MAGE-A4, MAGE-A5, MAGE-A6, MAGE-A7, MAGE-A8, MAGE-A9, MAGE-A10, MAGE-A11, MAGE-A12, MAGE-Xp2 (MAGE-B2), MAGE-Xp3 (MAGE-B3), MAGE-Xp4 (MAGE-B4), MAGE-C1, MAGE-C2, MAGE-C3, MAGE-C4, MAGE-C5), GAGE-family of tumor antigens (e.g., GAGE-1, GAGE-2, GAGE-3, GAGE-4, GAGE-5, GAGE-6, GAGE-7, GAGE-8, GAGE-9), B AGE, RAGE, LAGE- 1 , NAG, GnT-V, MUM-1, CDK4, tyrosinase, p53, MUC family, HER2/neu, p21ras, RCAS1, α-fetoprotein, E- cadherin, α-catenin, β-catenin and γ-catenin, pl20ctn, gpl00Pmel117, PRAME, NY-ESO-1, cdc27, adenomatous polyposis coli protein (APC), fodrin, Connexin 37, Ig-idiotype, pi 5, gp75, GM2 and GD2 gangliosides, viral products such as human papillomavirus proteins, Smad family of tumor antigens, Imp- 1 , P 1 A, EBV-encoded nuclear antigen (EBNA)- 1 , brain glycogen phosphorylase, SSX-1, SSX-2 (HOM-MEL-40), SSX-1, SSX-4, SSX-5, SCP-1 and CT-7, and c-erbB-2. Cancers or tumors and tumor antigens associated with such tumors (but not exclusively), include acute lymphoblastic leukemia (etv6; amll; cyclophilin b), B cell lymphoma (Ig-idiotype), glioma (E-cadherin; α-catenin; β-catenin; γ-catenin; pl20ctn), bladder cancer (p21ras), biliary cancer (p21ras), breast cancer (MUC family; HER2/neu; c- erbB-2), cervical carcinoma (p53; p21ras), colon carcinoma (p21ras; HER2/neu; c-erbB-2; MUC family), colorectal cancer (Colorectal associated antigen (CRC)~C017-1A/GA733; APC), choriocarcinoma (CEA), epithelial cell cancer (cyclophilin b), gastric cancer (HER2/neu; c-erbB-2; ga733 glycoprotein), hepatocellular cancer (α-fetoprotein), Hodgkins lymphoma (lmp-1; EBNA-1), lung cancer (CEA; MAGE-3; NY-ESO-1), lymphoid cell-derived leukemia (cyclophilin b), melanoma (pi 5 protein, gp75, onco fetal antigen, GM2 and GD2 gangliosides), myeloma (MUC family; p21ras), non-small cell lung carcinoma (HER2/neu; c-erbB-2), nasopharyngeal cancer (lmp-1; EBNA-1), ovarian cancer (MUC family; HER2/neu; c-erbB-2), prostate cancer (Prostate Specific Antigen (PSA) and its immunogenic epitopes PSA-1, PSA-2, and PSA-3; prostate-specific membrane antigen (PSMA); HER2/neu; c-erbB-2), pancreatic cancer (p21ras; MUC family; HER2/neu; c-erbB- 2; ga733 glycoprotein), renal cancer (HER2/neu; c-erbB-2), squamous cell cancers of cervix and esophagus (viral products such as human papillomavirus proteins), testicular cancer (NY- ESO-1), T-cell leukemia (HTLV-1 epitopes), and melanoma (Melan-A/MART-1; cdc27; MAGE-3; p21ras; gpl00Pmel117).
In one aspect of the invention, a method of treating an autoimmune disorder in a subject is provided. The method according to this aspect of the invention involves the step of administering to a subject having an autoimmune disorder an effective amount of a substituted 4-primary amino quinoline composition having structural Formula XVII, as defined above, to treat the autoimmune disorder. The substituted 4-primary amino quinoline composition in this aspect of the invention can be in the form a hydrate or pharmaceutically acceptable salt. In one embodiment the autoimmune disorder is chosen from systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Sjogren's syndrome, polymyositis, vasculitis, Wegener' s granulomatosis, sarcoidosis, ankylosing spondylitis, Reiter's syndrome, psoriatic arthritis, and Behcet's syndrome. In one particular embodiment the autoimmune disease is systemic lupus erythematosus. In one particular embodiment the autoimmune disease is rheumatoid arthritis. In one embodiment the subject is a human. The method can also be applied to animal models of any of the autoimmune disorders listed above. In one embodiment the autoimmune disorder is an immune complex associated disease. Immune complex associated diseases specifically include, without limitation, systemic lupus erythematosus, rheumatoid arthritis, polyarteritis nodosa, poststreptococcal glomerulonephritis, cryoglobulinemia, and acute and chronic serum sickness. The substituted 4-primary amino quinoline composition can be administered to the subject by any suitable route of administration, including, without limitation, oral and parenteral. Parenteral routes of administration include, without limitation, intravenous, intramuscular, intraperitoneal, subcutaneous, intranasal, intrapulmonary, transdermal, topical, and mucosal. Parenteral routes of administration also include direct injection into a specific tissue or other site of injection, including, for example, lymphoid tissue and a site of inflammation. It has been discovered according to the invention that quinzoline compounds structurally similar to quinolines of the invention are unexpectedly useful in methods for inhibiting an immune response, both in vitro and in vivo, including methods for treating immune complex associated diseases and autoimmune disorders. The quinazoline compounds and their methods of use according to the invention were unexpectedly found to retain much or all of the immunoinhibitory features of corresponding quinoline compounds and their methods of use, but with the additional feature of having reduced toxicity, at least in vivo.
In one aspect the invention provides novel quinazoline compositions. As described further below, these compositions and other quinazoline compositions have been discovered to be useful in methods for inhibiting an immune response, both in vitro and in vivo, including methods for treating immune complex associated diseases and autoimmune disorders. Due to their similarity to certain known antimalarial agents, it is also believed that the novel quinazoline compositions of the invention will also be useful for prevention and treatment of malaria, as well as for treatment of other diseases which have been described to be responsive to treatment with chloroquines. The novel quinazoline compositions of the invention have a structural Formula XVIII
Figure imgf000068_0001
wherein X is absent or is an aryl, alkyl, heterocyclic or styryl group, provided that if X is a phenyl group, NRjR. is part if a heterocycle or is a diamine; R\ and R2 are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R\ and R2 optionally are combined to form a heterocycle; Y is an oxygen atom, a sulfur atom, CR9R10, or NRπ, where R , R10, and Rπ are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R , R10, and Rπ optionally is combined with R3 or Rf to form a substituted or unsubstituted heterocycle; L is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; R3 and t are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R3 and t optionally are combined to form a heterocycle; and R5, Rg, R , and R8 are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein any pair of R5, Rg, R7, and R8 which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof. In one embodiment Rg and R are each independently a halogen atom or an alkoxy group. In one embodiment Rg and R are each independently a chlorine atom or a methoxy group. In one embodiment X is absent or is an aryl group; NRiR2 is a heterocyclic amine or NR (CH2)nNR10Rπ, wherein n is an integer from 2 to 6, inclusive, and R , R10, and Rπ are each independently a hydrogen atom or an alkyl group; Y is an aryl group or is NR12 where R12 is a hydrogen atom or an aryl or alkyl group; L is absent or is a C2-Cg alkyl group; R3 and t are each independently a hydrogen atom or an alkyl group, wherein R3 and Rt optionally are combined to form a heterocycle; and R5, Rg, R7, and Rg are each independently a hydrogen atom, a halogen atom, or an alkoxy group. In one embodiment X is absent or is an aryl group; NR!R2 is a substituted or unsubstituted piperazino or morpholino group or is NR9(CH2)nNR1oR11, wherein n is an integer from 2 to 6, inclusive, R9 is a hydrogen atom, and R10 and Rπ are each independently an alkyl group; Y is NH; L is a C2-Cg alkyl group; R3 and t are each independently a hydrogen atom or an alkyl group, wherein R3 and Rt optionally are combined to form a heterocycle; and R5, Rg, R , and R8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group. In one embodiment X is an aryl group; NRjR2 is substituted or unsubstituted piperazino or morpholino group; Y is NH; L is a C2-Cg alkyl group; R3 and Rt are each independently a methyl or ethyl group or R3 and R^t optionally are combined to form a morpholino group; and R5, Rg, R7, and R8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group. In one embodiment X is a phenyl group; NRtR2 is N-methylpiperazine; Y is NH; L is -CH2CH2-; R3 and f are each a methyl group; and R5, Rg, R7, and R8 are each a hydrogen atom. In one embodiment X is a phenyl group; NRiR2 is N-methylpiperazine; Y is NH; L is -CH2CH2-; R3 and R4 are combined as a morpholino group; and R5, Rg, R7, and R8 are each a hydrogen atom. In one embodiment X is absent; NRiR. is a substituted or unsubstituted piperazino or morpholino group; Y is NH; L is a C2-Cg alkyl group; R3 and Rt are each independently a methyl or ethyl group or R3 and t optionally are combined to form a morpholino group; and R5, Rg, R , and R8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group. In one embodiment X is absent; NRjR2 is N-methylpiperazine; Y is NH; L is -CH2CH2-; R3 and t are each a methyl group; Rg and R are each a methoxy group; and R5 and Rs are each a hydrogen atom. In one embodiment X is absent; NRtR2 is N-phenylpiperazine; Y is NH; L is -CH2CH2-; R3 and Rf are each a methyl group; Rg and R are each a methoxy group; and R5 and Rs are each a hydrogen atom. In one embodiment X is absent; NRiR2 is N-methylpiperazine; Y is NH; L is -CH2CH2; R3 and f are combined as a morpholino group; Rg and R are each a methoxy group; and R5 and R8 are each a hydrogen atom. In each of the foregoing embodiments, the composition is optionally in the form of a pharmaceutically acceptable hydrate or salt. Representative, non-limiting examples of quinazoline compositions of Formula XVIII of the invention are compounds 201-214, presented in Table 3.
Table 3. Quinazoline Compositions of the Invention
Figure imgf000071_0001
Figure imgf000072_0001
It has been discovered according to the invention that certain quinazoline compositions can be used to inhibit TLR9 signaling. In one aspect the invention provides additional novel quinazoline compounds useful in the methods of the invention. Such compounds are referred to herein as CMZ 203-34, CMZ 203-44, CMZ 203-45, CMZ 203-49, CMZ 203-51, CMZ 203-76, CMZ 203-78, CMZ 203-87, CMZ 203-93, and CMZ 203-95. Syntheses for each of these novel compounds are provided in Examples 19-28, respectively. Certain of these specific compounds have already been tested in vitro and found to inhibit TLR9 signaling. See Example 29. Thus compounds CMZ 203-34, CMZ 203-44, CMZ 203-45, CMZ 203-49, CMZ 203-51, CMZ 203-76, CMZ 203-78, CMZ 203-87, CMZ 203-93, and CMZ 203-95 are believed to be useful in the methods of the invention.
In one aspect the invention provides a method for inhibiting signaling by a TLR. The method according to this aspect of the invention involves contacting a cell expressing a functional TLR with an effective amount of quinazoline composition having structural Formula XIX
Figure imgf000073_0001
Formula XIX wherein X is a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group, optionally attached to the quinazoline by a nitrogen, oxygen, or sulfur atom or by a SO or SO2 group; Y is absent or is an oxygen atom, a sulfur atom, CR R10, or NRπ, where R , R10, and Rπ are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R9, R10, and Rπ optionally is combined with R3 or t to form a heterocycle; L is absent or is a hydrogen atom, an alkyl or alkenyl group containing from 1 to 10 carbons, or an aryl group; R3 and R4 are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R3 and R4 optionally are combined to form a heterocycle; and Rs, Rg, R , and R8 are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein any pair of R5, Rg, R7, and Rs which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof, to inhibit signaling by the TLR. The method according to this aspect of the invention can be performed in vitro or it can be performed in vivo. In addition, the cell expressing the functional TLR can, but need not necessarily, be an immune cell. For example, the cell expressing the functional TLR can be a cell transfected with an expression vector that directs expression of the TLR by the cell. In one embodiment the TLR is TLR9 and the method is thus a method for inhibiting intracellular signaling by TLR9. In addition to compounds 201-214, presented in Table 3, the following additional representative and non-limiting quinazoline compounds, compounds 215-229, presented in Table 4 with reference to Formula XIX, can be used in the method according to this and all other aspects of the invention directed to methods involving use of substituted 4-primary amino quinoline compounds. Additional examples are presented in the Examples below.
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
In one aspect the invention provides a method for inhibiting signaling by a TLR. The method according to this aspect of the invention involves contacting an immune cell expressing a functional TLR with (a) an effective amount of a TLR signal agonist to stimulate signaling by the TLR in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having structural Formula XIX, as defined above, to inhibit signaling by the TLR in response to the TLR signal agonist compared with the signaling by the TLR in response to the TLR signal agonist in absence of the quinazoline composition. The quinazoline composition in this aspect of the invention can be in the form a hydrate or pharmaceutically acceptable salt. The method according to this aspect of the invention can be performed in vitro or it can be performed in vivo. Also according to this aspect of the invention, in one embodiment the TLR is TLR9 and the TLR signal agonist is a TLR9 signal agonist, and the method in one embodiment is thus a method of inhibiting intracellular signaling by TLR9 in response to a TLR9 signal agonist. The TLR signal agonist in one embodiment is CpG DNA, for example a CpG ODN such as ODN 2006. The CpG ODN can belong to any class of CpG ODN, including A-class (e.g., ODN 2216), B-class (e.g., ODN 2006), or C-class (e.g., ODN 2395). In one embodiment the TLR signal agonist is an immune complex that includes a nucleic acid, as described above.
In one aspect the invention provides a method for inhibiting an immune response to an antigenic substance. The method according to this aspect of the invention involves contacting an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having structural Formula XIX, as defined above, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the quinazoline composition. The quinazoline composition in this aspect of the invention can be in the form a hydrate or pharmaceutically acceptable salt. The method according to this aspect of the invention can be performed in vitro or it can be performed in vivo. In one embodiment the immune response is an innate immune response. In one embodiment the immune response includes an adaptive immune response. In one embodiment the step of contacting an immune cell expressing a functional Toll-like receptor with an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a quinazoline composition involves the active step of administering an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a quinazoline composition. The antigenic substance can be administered using any effective route or means for administering the antigenic substance to effect the contacting. By way of example, the administering can be by local or systemic injection, inhalation, oral ingestion, topical administration, mucosal administration, or any combination thereof. In one embodiment the method involves contacting, in a subject, an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response in the subject to the antigenic substance in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having structural Formula XVII, as defined above, to inhibit an immune response in the subject to the antigenic substance compared with the immune response to the antigenic substance in absence of the quinazoline composition. In one embodiment the step of contacting an immune cell expressing a functional
Toll-like receptor with an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a quinazoline composition is passive. For example, in one embodiment the immune cell is already in contact or has already been in contact with an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance, at the time of or prior to the step of contacting the immune cell expressing a functional Toll-like receptor with an effective amount of a quinazoline composition having structural Formula XVII, as defined above, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the quinazoline composition. The antigenic substance can be an allergen, as described above. The antigenic substance can be an antigen that is or is derived from an infectious microbial agent, including a bacterium, a virus, a fungus, or a parasite, as described above. The antigenic substance can be a cancer antigen, as described above.
In one aspect of the invention, a method of treating an autoimmune disorder in a subject is provided. The method according to this aspect of the invention involves the step of administering to a subject having an autoimmune disorder an effective amount of a quinazoline composition having structural Formula XIX, as defined above, to treat the autoimmune disorder. The quinazoline composition in this aspect of the invention can be in the form a hydrate or pharmaceutically acceptable salt. In one embodiment the autoimmune disorder is chosen from systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Sjδgren's syndrome, polymyositis, vasculitis, Wegener's granulomatosis, sarcoidosis, ankylosing spondylitis, Reiter's syndrome, psoriatic arthritis, and Behcet's syndrome. In one particular embodiment the autoimmune disease is systemic lupus erythematosus. In one particular embodiment the autoimmune disease is rheumatoid arthritis. In one embodiment the subject is a human. The method can also be applied to animal models of any of the autoimmune disorders listed above. In one embodiment the autoimmune disorder is an immune complex associated disease, as described above. The quinazoline composition can be administered to the subject by any suitable route of administration, including, without limitation, oral and parenteral. Parenteral routes of administration are as described above with respect to substituted 4-primary amino quinolines.
Assays for Effectiveness The methods of the invention can be assessed using any of a number of possible readout systems based upon a TLR/IL-1R signal transduction pathway. In some embodiments, the readout for the method is based on the use of native genes or, alternatively, transfected or otherwise artificially introduced reporter gene constructs which are responsive to the TLR/IL-1R signal transduction pathway involving MyD88, TRAP, p38, and/or ERK. Hacker H et al. (1999) E BO J 18:6973-82. These pathways activate kinases including KB kinase complex and c-Jun N-terminal kinases. Thus reporter genes and reporter gene constructs particularly useful for the assays include, e.g., a reporter gene operatively linked to a promoter sensitive to NF-κB. Examples of such promoters include, without limitation, those for NF-KB, IL-lβ, IL-6, IL-8, IL-12 p40, IP-10, CD80, CD86, and TNF-α. The reporter gene operatively linked to the TLR-sensitive promoter can include, without limitation, an enzyme (e.g., luciferase, alkaline phosphatase, β-galactosidase, chloramphenicol acetyltransferase (CAT), etc.), a bioluminescence marker (e.g., green- fluorescent protein (GFP, e.g., U.S. Pat. No. 5,491,084), blue fluorescent protein (BFP, e.g., U.S. Pat. No. 6,486,382), etc.), a surface-expressed molecule (e.g., CD25, CD80, CD86), and a secreted molecule (e.g., IL-1, IL-6, IL-8, IL-12 p40, TNF-α). In certain embodiments the reporter is selected from IL-8, TNF-α, NF-κB-luciferase (NF-κB-luc; Hacker H et al. (1999) EMBOJ 18:6973-82), IL-12 p40-luc (Murphy TL et al. (1995) Mol Cell Biol 15:5258-67), and TNF-luc (Hacker H et al. (1999) EMBO J 18:6973-82). In assays relying on enzyme activity readout, substrate can be supplied as part of the assay, and detection can involve measurement of chemiluminescence, fluorescence, color development, incorporation of radioactive label, drug resistance, or other marker of enzyme activity. For assays relying on surface expression of a molecule, detection can be accomplished using flow cytometry (FACS) analysis or functional assays. Secreted molecules can be assayed using enzyme- linked immunosorbent assay (ELISA) or bioassays. Many of these and other suitable readout systems are well known in the art and are commercially available.
Reporter Constructs A cell expressing a functional TLR and useful for the methods of the invention has, in some embodiments, an expression vector including an isolated nucleic acid which encodes a reporter construct useful for detecting TLR signaling. The expression vector including an isolated nucleic acid which encodes a reporter construct useful for detecting TLR signaling can include a reporter gene under control of a promoter response element (enhancer element). In some embodiments the promoter response element is associated with a minimal promoter responsive to a transcription factor believed by the applicant to be activated as a consequence of TLR signaling. Examples of such minimal promoters include, without limitation, promoters for the following genes: AP-1, NF-κB, ATF2, IRF3, and IRF7. These minimal promoters contain corresponding promoter response elements sensitive to AP-1, NF-κB, ATF2, IRF3, and IRF7, respectively. In other embodiments the expression vector including an isolated nucleic acid which encodes a reporter construct useful for detecting TLR signaling can include a gene under control of a promoter response element selected from response elements sensitive to IL-6, IL-8, IL-12 p40 subunit, a type I IFN, RANTES, TNF, IP-10, 1-TAC, and interferon-stimulated response element (ISRE). The promoter response element generally will be present in multiple copies, e.g., as tandem repeats. For example, in one reporter construct, coding sequence for luciferase is under control of an upstream 6X tandem repeat of NF-κB response element. In another example, an ISRE-luciferase reporter construct useful in the invention is available from Sfratagene (catalog no. 219092) and includes a 5x ISRE tandem repeat joined to a TATA box upstream of a luciferase reporter gene. As discussed further elsewhere herein, the reporter itself can be any gene product suitable for detection by methods recognized in the art. Such methods for detection can include, for example, measurement of spontaneous or stimulated light emission, enzyme activity, expression of a soluble molecule, expression of a cell surface molecule, etc. Readouts typically involve usual elements of Toll/IL-IR signaling, e.g., MyD88, TRAF, and IRAK molecules, although in the case of TLR3 the role of MyD88 is less clear than for other TLR family members. As demonstrated herein such responses include the induction of a gene under control of a specific promoter such as a NF-κB promoter, increases in particular cytokine levels, increases in particular chemokine levels, etc. The gene under the control of the NF-κB promoter can be a gene which naturally includes an NF-κB promoter or it can be a gene in a construct in which an NF-κB promoter has been inserted. Genes and constructs which include the NF-κB promoter include but are not limited to IL-8, IL-12 p40, NF-κB-luc, IL-12 p40-luc, and TNF-luc. Increases in cytokine levels can result from increased production, increased stability, increased secretion, or any combination of the forgoing, of the cytokine in response to the TLR-mediated signaling. Cytokines generally include, without limitation, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-10, IL-11, IL-12, IL-13, IL-15, IL-18, IFN-α, IFN-β, IFN-γ, TNF-α, GM-CSF, G-CSF, M-CSF. Thl cytokines include but are not limited to IL-2, IFN-γ, and IL-12. Th2 cytokines include but are not limited to IL-4, IL-5, and IL-10. Increases in chemokine levels can result from increased production, increased Λability, increased secretion, or any combination of the forgoing, of the chemokine in response to the TLR-mediated signaling. Chemokines of particular significance in the invention include but are not limited to CCL5 (RANTES), CXCL9 (Mig), CXCL10 (IP- 10), and CXCL11 (I-TAC), IL-8, and MCP-1. Administration to a Subject Some aspects of the invention involve administering an effective amount of a composition to a subject to achieve a specific outcome. The small molecule compositions useful according to the methods of the present invention thus can be formulated in any manner suitable for pharmaceutical use. The formulations of the invention are administered in pharmaceutically acceptable solutions, which may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic ingredients. For use in therapy, an effective amount of the compound can be administered to a subject by any mode allowing the compound to be taken up by the appropriate target cells. "Administering" the pharmaceutical composition of the present invention can be accomplished by any means known to the skilled artisan. Specific routes of administration include but are not limited to oral, transdermal (e.g., via a patch), parenteral injection (subcutaneous, intradermal, intramuscular, intravenous, intraperitoneal, intrathecal, etc.), or mucosal (intranasal, intratracheal, inhalation, intrarectal, intravaginal, etc.). An injection can be in a bolus or a continuous infusion. For example the pharmaceutical compositions according to the invention are often administered by intravenous, intramuscular, or other parenteral means, or by biolistic "gene- gun" application to the epidermis. They can also be administered by intranasal application, inhalation, topically, orally, or as implants, and even rectal or vaginal use is possible. Suitable liquid or solid pharmaceutical preparation forms are, for example, aqueous or saline solutions for injection or inhalation, microencapsulated, encochleated, coated onto microscopic gold particles, contained in liposomes, nebulized, aerosols, pellets for implantation into the skin, or dried onto a sharp object to be scratched into the skin. The pharmaceutical compositions also include granules, powders, tablets, coated tablets, (micro)capsules, suppositories, syrups, emulsions, suspensions, creams, drops or preparations with protracted release of active compounds, in whose preparation excipients and additives and/or auxiliaries such as disintegrants, binders, coating agents, swelling agents, lubricants, flavorings, sweeteners or solubilizers are customarily used as described above. The pharmaceutical compositions are suitable for use in a variety of drug delivery systems. For a brief review of present methods for drug delivery, see Langer R (1990) Science 249:1527-33, which is incorporated herein by reference. The concentration of compounds included in compositions used in the methods of the invention can range from about 1 nM to about 100 μM. Effective doses are believed to range from about 10 picomole/kg to about 100 micromole/kg. The pharmaceutical compositions are preferably prepared and administered in dose units. Liquid dose units are vials or ampoules for injection or other parenteral administration. Solid dose units are tablets, capsules, powders, and suppositories. For treatment of a patient, depending on activity of the compound, manner of administration, purpose of the administration (i.e., prophylactic or therapeutic), nature and severity of the disorder, age and body weight of the patient, different doses may be necessary. The administration of a given dose can be carried out both by single administration in the form of an individual dose unit or else several smaller dose units. Repeated and multiple administration of doses at specific intervals of days, weeks, or months apart are also contemplated by the invention. The compositions can be administered per se (neat) or in the form of a pharmaceutically acceptable salt. When used in medicine the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts can conveniently be used to prepare pharmaceutically acceptable salts thereof. Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic. Also, such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group. Suitable buffering agents include: acetic acid and a salt (1-2% w/v); citric acid and a salt (1-3% w/v); boric acid and a salt (0.5-2.5% w/v); and phosphoric acid and a salt (0.8-2% w/v). Suitable preservatives include benzalkonium chloride (0.003-0.03% w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004-0.02% w/v). The pharmaceutical compositions of the invention optionally include an active ingredient and optionally a pharmaceutically-acceptable carrier. The term "pharmaceutically- acceptable carrier" means one or more compatible solid or liquid fillers, dilutants or encapsulating substances which are suitable for administration to a human or other vertebrate animal. The term "carrier" denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The components of the pharmaceutical compositions also are capable of being comingled with the compounds of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficiency. Compositions suitable for parenteral administration conveniently include sterile aqueous preparations, which can be isotonic with the blood of the recipient. Among the acceptable vehicles and solvents are water, Ringer's solution, phosphate buffered saline, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed mineral or non-mineral oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. Carrier formulations suitable for subcutaneous, intramuscular, intraperitoneal, intravenous, etc. administrations can be found in Remingto 's Pharmaceutical Sciences, Mack Publishing Company, Easton, PA. The compounds useful in the invention can be delivered in mixtures of more than two such compounds. A mixture can further include one or more adjuvants in addition to the combination of compounds. A variety of administration routes is available. The particular mode selected will depend, of course, upon the particular compound selected, the age land general health status of the subject, the particular condition being treated, and the dosage required for therapeutic efficacy. The methods of this invention, generally speaking, can be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of response without causing clinically unacceptable adverse effects. Preferred modes of administration are discussed above. The compositions can conveniently be presented in unit dosage form and can be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the compounds into association with a carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the compounds into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product. Other delivery systems can include time-release, delayed release or sustained release delivery systems. Such systems can avoid repeated administrations of the compounds, increasing convenience to the subject and the physician. Many types of release delivery systems are available and known to those of ordinary skill in the art. They include polymer base systems such as poly(lactide-glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides. Microcapsules of the foregoing polymers containing drugs are described in, for example, U.S. Pat. No. 5,075,109. Delivery systems also include non-polymer systems that are: lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di-and tri-glycerides; hydrogel release systems; silastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially fused implants; and the like. Specific examples include, but are not limited to: (a) erosional systems in which an agent of the invention is contained in a form within a matrix such as those described in U.S. Pat. Nos. 4,452,775, 4,675,189, and 5,736,152, and (b) diffusional systems in which an active component permeates at a controlled rate from a polymer such as described in U.S. Pat. Nos. 3,854,480, 5,133,974 and 5,407,686. In addition, pump-based hardware delivery systems can be used, some of which are adapted for implantation. The invention will be further described in the following examples, which do not limit the scope of the invention described in the claims.
EXAMPLES Example 1 In Vitro Screening of a Library of Small Molecules for Inhibitors of Human TLR9 Candidate small molecules for initial screening were obtained from a commercially available library of 880 off-patent small molecules selected for structural diversity and proven bioavailability in humans (Prestwick Chemical Library). Human embryonic kidney HEK293 cells stably transfected with a human TLR9 (hTLR9) expression vector were incubated overnight in the presence of 50 nM CpG ODN 2006 (i.e., the EC50 concentration of ODN 2006) and selected small molecule candidate compounds at different concentrations ranging from 5x10" M to 5x10" M. TLR9 activity was assayed in terms of induction of a 6x NF-κB-luciferase reporter construct cotransfected in the cells. Results were measured as fold induction over baseline luciferase activity measured in the absence of ODN 2006 and compared to fold induction over baseline in the presence of the EC50 concentration of ODN 2006 alone. From this initial screening a number of small molecules were identified as lead compounds and were categorized by structure and activity. Additional screening was performed in like manner using additional small molecules selected from another library or specifically prepared for this purpose. Results of some of these assays are presented in FIG. l and in Tables 5-16. The method is described in greater detail as follows. The cells for each individual cell clone (hTLR3-NFκB-293, hTLR7-NFκB-293, hTLR8-NFκB-293 and hTLR9-NFκB-293) were counted and seeded one day before the stimulation at 1.5x104 cells per well in 96-well cell culture plates. To become adherent the cells were incubated after seeding overnight at 37°C in 5% CO2 humidified air. The screen for each compound was performed at the same time for all of the receptors TLR3, TLR7, TLR8 and TLR9. Up to fifteen test compounds, each assayed in duplicate at each of three different final concentrations (0.5 μg/ml, 5 μg/ml, and 50 μg/ml), were used for 16h cell stimulation on a single multiwell culture plate. A 4X final concentration master plate was created to prepare the different test compounds before they were added to the cells. Controls were added as duplicates individually for each cell clone on the cell culture plate. Positive controls were used as follows (final concentrations): 50 μg/ml poly(I:C) for hTLR3-NFκB-293; 8 μM resiquimod (R848) for hTLR7-NFκB-293; 30 μM R848 for hTLR8-NFκB-293; and 2.5 μM CpG-ODN 2006 for hTLR9-NFκB-293. Media alone was used as negative control. After addition of controls and small molecules, the cell clones were additionally stimulated with the EC50 concentration of the appropriate receptor-specific ligand. To calculate the baseline response to the EC50 concentration of the appropriate receptor-specific ligand, wells H3 and H6 did not receive receptor-specific ligand. The mean value of wells H2 and H5 (each with cells and EC50 concentration of the appropriate receptor- specific ligand) divided by the mean of wells H3 and H6 (each with cells alone) yielded the baseline response (i.e., fold induction of luciferase activity) to EC50 concentration of the appropriate receptor-specific ligand. After 16h stimulation the supernatant was removed and the cells treated with lysis buffer and stored at -80°C before measuring. An agonist screen was performed in parallel with the antagonist screen just described.
The procedure was comparable to the antagonist screen except there was no addition of EC50 concentration of receptor-specific ligand. The outcome of this screen reflected agonistic and toxicity effects.
Graphical ranking system For each screen result a graphical documentation was performed. A numerical score, describing the graphical results, was determined for each compound. For positive compounds either for antagonists or agonists or synergists the screen has been repeated at least twice. Antagonists The baseline result (measured with addition of the EC.o concentration of the receptor- specific ligand) for each cell culture plate was measured as described above. Antagonistic effects were measured by down regulation compared to the baseline. Antagonists were scored as follows: 13: compounds for which there was clear down regulation at 0.5 μg/ml, 5 μg/ml, and 50 μg/ml; 12: compounds for which there was clear down regulation at 5 μg/ml and 50 μg/ml; 11 : compounds for which there was clear down regulation only at 50 μg/ml; — : compounds for which there was no clear down regulation even at 50 μg/ml.
Agonists To detect agonist activity the cells were treated in the above described manner but without adding the EC50 concentration of receptor-specific ligand after treatment of the cells with small molecules. Usually the baseline in the agonist screen is taken to be 1 because these cells have not been additionally stimulated with the EC50 concentration of the receptor- specific ligand. Agonist activity was present when there was above-baseline activity. Agonists were scored as follows:
For compounds with highest agonist activity at 0.5 μg/ml,
-1 : compounds for which there was a greater than 5-fold induction compared to baseline; -2: compounds for which there was a 2- to 5-fold induction compared to baseline;
-3: compounds for which there was a less than 2-fold induction compared to baseline. For compounds with highest agonist activity at 5 μg/ml, 0.25: compounds for which there was a greater than 5-fold induction compared to baseline; 0.5: compounds for which there was a 2- to 5-fold induction compared to baseline; 0.75: compounds for which there was a less than 2-fold induction compared to baseline.
For compounds with highest agonist activity at 50 μg/ml, 1 : compounds for which there was a greater than 5-fold induction compared to baseline; 2: compounds for which there was a 2- to 5-fold induction compared to baseline; 3: compounds for which there was a less than 2-fold induction compared to baseline.
Toxicity Measuring of "toxicity" of the small molecules was performed by analysing below- baseline activity within the agonist screen. "Toxicity" can have different explanations - either differences between the cell clones, the compound are effecting the signal transduction pathway or the TLR within the cell clone, or the compounds do reέilly have a toxic effect on the cells. Each compound was screened for "toxicity" on each of the four cell lines. Usually the baseline in the agonist screen is taken to be 1 because these cells have not been additionally stimulated with the EC50 concentration of the receptor-specific ligand. Apparent toxicity was scored as follows:
T3: compounds for which there was clear down regulation at 0.5 μg/ml, 5 μg/ml, and 50 μg/ml;
T2: compounds for which there was clear down regulation at 5 μg/ml and 50 μg/ml; TI : compounds for which there was clear down regulation only at 50 μg/ml.
Example 2 In Vitro Screening of a Library of Small Molecules for Inhibitors of Human TLR8 Candidate small molecules for initial screening were identified in an assay similar to that in Example 1 except HEK293 cells were stably transfected with a human TLR8 (hTLR8) expression vector and incubated overnight in the presence of 500 nM R848 (EC50 of R848 for TLR8) and selected small molecule candidate compounds at different concentrations ranging from 5x10"7 M to 5xl0"5 M. TLR8 activity was assayed in terms of induction of a 6x NF-κB- luciferase reporter construct cotransfected in the cells. Results were expressed as fold induction over baseline luciferase activity measured in the absence of R848. From this initial screening a number of small molecules were identified as lead compounds and were categorized by structure and activity. Additional screening was performed in like manner using additional small molecules selected from another library or specifically prepared for this purpose. Results of some of these assays are presented in Tables 5-16. Scoring is reported as above but as applied to TLR8.
Example 3 In Vitro Screening of a Library of Small Molecules for Inhibitors of Human TLRl Candidate small molecules for initial screening were identified in an assay similar to that in Example 1 except HEK293 cells were stably transfected with a human TLR7 (hTLR7) expression vector and incubated overnight in the presence of 2 μM R848 (EC50 of R848 for TLR7) and selected small molecule candidate compounds at different concentrations ranging 7 ^ from 5x10" M to 5x10" M. TLR7 activity was assayed in terms of induction of a 6x NF-κB- luciferase reporter construct cotransfected in the cells. Results were expressed as fold induction over baseline luciferase activity measured in the absence of R848. From this initial screening a number of small molecules were identified as lead compounds and were categorized by structure and activity. Additional screening was performed in like manner using additional small molecules selected from another library or specifically prepared for this purpose. Results of some of these assays are presented in Tables 5-16. Scoring is reported as above but as applied to TLR7.
Example 4 In Vitro Screening of a Library of Small Molecules for Inhibitors of Human TLRS Candidate small molecules for initial screening were identified in an assay similar to that in Example 1 except HEK293 cells were stably transfected with a human TLR3 (hTLR3) expression vector and incubated overnight in the presence of 2.5 μg/ml poly(I:C) (EC50 of poly(LC) for TLR3) and selected small molecule candidate compounds at different concentrations ranging from 5x10"7 M to 5xl0"5 M. TLR3 activity was assayed in terms of induction of a 6x NF-κB -luciferase reporter construct cotransfected in the cells. Results were expressed as fold induction over baseline luciferase activity measured in the absence of poly(I:C). From this initial screening a number of small molecules were identified as lead compounds and were categorized by structure and activity. Additional screening was performed in like manner using additional small molecules selected from another library or specifically prepared for this purpose. Results of some of these assays are presented in Tables 5-16. Scoring is reported as above but as applied to TLR3.
In the data presented in Tables 5-17 below, it should be recognized that at least certain compounds can conform to more than a single Formula. Table 5. Exemplary Compounds of Formula II and Their Inhibitory Effect on Select TLRs
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Table 6. Exemplary Compounds of Formula III and Their Inhibitory Effect on Select TLRs ID structure chemical name hTLR3 hTLR7 hTLR8 hTLR9 149 rrhioproperasine 13 αimesylate
53 [Triflupromazine 1 1 11 11 13 hydrochloride
576 Αcetopromazine 11 12 rnaleate salt
Figure imgf000093_0001
64 Chlorpromazine 11 11 11 12 riydrochloride
693 Promazine 12 12 riydrochloride
399 Prochlorperazine 12 12 11 12 dimaleate
Figure imgf000093_0002
529 Mesoridazine besylate 11 o 125 Perphenazine 11 11
840 Ethopropazine 11 11 ydrochloride
797 Methotrimeprazine 11 11 rnaleat salt
Figure imgf000094_0002
Table 7. Exemplary Compounds of Formula IV and Their Inhibitory Effect on Select TLRs
Figure imgf000094_0001
Figure imgf000095_0001
Table 8. Exemplary Compounds of Formula V and Their Inhibitory Effect on Select TLRs
Figure imgf000095_0002
Figure imgf000096_0003
Table 9. Exemplary Compounds of Formula VI and Their Inhibitory Effect on Select TLRs ID structure chemical name hTLR3 hTLR7 hTLR8 hTLR9 239 rphenadrine 13 riydrochloride
Figure imgf000096_0001
229 Nefopam hydrochloride 12
707 iphenylpyraline 11 12 hydrochloride
793 ploperastine 11 1 1 12 hydrochloride
Figure imgf000096_0002
Figure imgf000097_0001
Table 10. Exemplary Compounds of Formula VII and Their Inhibitory Effect on Select TLRs
Figure imgf000098_0001
Table 11. Exemplary Compounds of Formula VIII and Their Inhibitory Effect on Select TLRs
Figure imgf000098_0002
Figure imgf000099_0001
Figure imgf000100_0001
Table 12. Exemplary Compounds of Formula IX and Their Inhibitory Effect on Select TLRs
Figure imgf000100_0002
Table 13. Exemplary Compounds of Formula X and Their Inhibitory Effect on Select TLRs ID structure chemical name hTLR3 hTLR7 hTLR8 hTLR9 431 oralyne chloride 13 ηydrate
792 Propidium iodide 12 12
583 Papaverine 11 12 riydrochloride
830 -:thaverine 11 11 11 lydrochloride
586 Berberine chloride 1 1 12
Figure imgf000101_0001
Table 14. Exemplary Compounds of Formula XI and Their Inhibitory Effect on Select TLRs
Figure imgf000101_0002
Figure imgf000102_0001
Table 15. Exemplary Compounds of Formula XII and Their Inhibitory Effect on Select TLRs
Figure imgf000102_0002
Table 16. Exemplary Compounds of Formula XIII and Their Inhibitory Effect on Select TLRs
Figure imgf000102_0003
Figure imgf000103_0001
Table 17. Exemplary Compounds of Formula XIV and Their Inhibitory Effect on Select TLRs
Figure imgf000104_0001
Example 5 In Vivo Screening of Selected Small Molecules Experimental mice are administered known amounts of candidate small molecules and a source of PAMP or other suitable TLR ligand, e.g., CpG nucleic acid. Negative control mice receive the source of PAMP or other suitable TLR ligand, e.g., CpG nucleic acid, alone. After an appropriate period, blood samples are obtained from control and experimental mice and evaluated for serum concentration of cytokine using a suitable method, e.g., enzyme- linked immunosorbent assay (ELISA). Alternatively or in addition, peripheral blood mononuclear cells (PBMC) are isolated from both groups of animals and assessed for expression of activation marker using a suitable technique such as fluorescence activated cell sorting (FACS). Control and experimental results are compared in pairwise fashion. Reduced expression of activation marker or reduced concentration of cytokine in an experimental animal compared to a negative control animal indicates the small molecule inhibits TLR-mediated signaling in response to a ligand for the TLR. Increased expression of activation marker or increased concentration of cytokine in an experimental animal compared to a negative control animal indicates the small molecule promotes TLR-mediated signaling in response to a ligand for the TLR. Example 6 In Vivo Screening of Selected Small Molecules Experimental mice are administered candidate small molecules. Negative control mice are administered carrier alone. After administration or small molecule or carrier alone, PBMC are isolated and then exposed in vitro to CpG nucleic acid under conditions in which the PBMC, in the absence of small molecule, are stimulated to express an activation marker such as CD86 or secrete a product such as cytokine (e.g., IFN-α, IL-6, TNF-α) or chemokine (e.g., IP- 10). The expression of the activation marker or the secretion of the product is quantified using FACS, ELISA, or other suitable method, and comparison is made between results obtained with and without the small molecule. Reduced expression of activation marker or reduced concentration of cytokine in an experimental animal compared to a negative control animal indicates the small molecule inhibits TLR-mediated signaling in response to a ligand for the TLR. Increased expression of activation marker or increased concentration of cytokine in an experimental animal compared to a negative control animal indicates the small molecule promotes TLR-mediated signaling in response to a ligand for the TLR.
Example 7 Human TLR9 Antagonism by 4-Primary Amino Quinolines Stably co-transfecting the human TLR9 receptor (hTLR9) and an NF-κB promoter- driven luciferase gene into HEK-293 cells created the cell line hTLR9-NFkB-293. hTLR9- NFkB-293 cells were counted and seeded at 1.5xl04 cells per well in 96-well cell culture plates one day before assaying and cultured at 37°C / 5% CO2. The day of the assay, antagonist was added at 50 μM, 5.0 μM or 0.5 μM. The cells were then stimulated with the EC50 dose of the TLR9 agonist, CpG-ODN 2006, and cultured for 16 h in a humidified incubator at 37°C. The culture supernatant was removed and the cells were treated with lysis buffer and stored at -80°C before measuring luciferase activity. The luciferase readout was measured according to manufacturer's instructions using a luciferase assay system available from Promega, USA. Results are summarized in Table 18. The antagonist dose listed in Table 18 is the minimum effective dose (μM) for blockade of the TLR9 agonist. As evident from the results presented in Table 18, the minimum effective dose (μM) for blockade of the TLR9 agonist was 0.5 μM or 5 μM for all of the compounds tested, as measured by this three-point assay.
Table 18. Human TLR9 Antagonism by 4-Amino-Quinolines.
Figure imgf000106_0001
Figure imgf000107_0001
Example 8 Human TLR9 Antagonism by Quinazolines hTLR9-NFkB-293 cells were counted and seeded at 1.5xl04 cells per well in 96-well cell culture plates one day before assaying and cultured at 37°C / 5% CO2, as described in Example 7. The day of the assay, antagonist was added at varying concentrations starting at 50 μM with a 1/5 to 1/6 dilution for 7 steps. The cells were then stimulated with the EC50 dose of the TLR9 agonist, CpG-ODN 2006, and cultured for 16 h in a humidified incubator at 37°C. The culture supernatant was removed and the cells were treated with lysis buffer and stored at -80°C before measuring luciferase activity as described in Example 7. Sigmoidal antagonism curves were generated and the inhibitor concentration at which 50% of the agonist response was blocked (IC50) calculated. Results shown in Table 19 under the heading hTLR9 are the IC50 dose (μM) for blockade of a CpG ODN-generated signal in hTLR9-NFkB-293 cells. Alternatively, TLR9 ligand antagonism in human peripheral blood mononuclear cells (PBMC) was monitored. Peripheral blood buffy coat preparations from healthy male and female human donors were obtained from the Blood Bank of the University of Diisseldorf (Germany) and from these, PBMC were purified by centrifugation over Ficoll-Hypaque (Sigma). The purified PBMC were resuspended in RPMI 1640 culture medium supplemented with 5% (v/v) heat-inactivated human AB serum (BioWhittaker, Belgium) or 10% (v/v) heat-inactivated fetal calf serum (FCS), 1.5 mM L-glutamine, 100 U/ml penicillin and 100 μg/ml streptomycin (all from Sigma). Fresh PBMC at a concentration of 3x107ml to 5x107ml were added to 96-well round-bottomed plates (150 μl/well). After cell plating, antagonist was added at varying concentrations starting at 50 μM with a 1/5 to 1/6 dilution for 7 steps. The cells were then stimulated with the TLR9 agonist, CpG-ODN 2006, and cultured for 16 h in a humidified incubator at 37°C. Culture supernatants were collected and, if not used immediately, frozen at -20°C until required. Interleukin 6 (IL-6) in the supernatants was quantitatively assessed using commercially available ELISA Kits (IL6, Diaclone, USA). Sigmoidal antagonism curves were generated and IC50 calculated. Results shown in Table 19 under the heading IL-6 are the IC50 dose (μM) for blockade of CpG ODN-generated IL-6 production by human PBMC.
Table 19. Antagonism of TLR9 Signal Agonist CpG ODN 2006 by Quinazolines
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0002
ND: No Data.
Example 9 Reduced In Vivo Toxicity of Quinazoline Compounds Compared with a Structurally Similar Quinoline Female BALB/c mice (n = 3 per group) were given a single intraperitoneal bolus injection of CMZ 203-43 (1.0 or 4.0 mg), CMZ 203-34 (1.0 or 4.0 mg) or CMZ 203-49 (1.0 or 4.0 mg) in a volume of 0.2 ml. Compound CMZ 203-43 has the structural formula
Figure imgf000110_0001
CMZ 203-43 Other mice received 10 % dimethyl sulfoxide (DMSO) in an identical manner. Animals were weighed immediately prior to injection (day 1) and then daily until day 5. On day 5, blood was collected by cardiac puncture and analyzed for hematological and biochemical parameters. Animals were monitored daily for morbidity and mortality. Results are shown in
FIG. 2 and FIG. 3. FIG. 2A shows change in body weight (relative to body weight prior to first injection)
± standard error of the means for different groups. FIG. 2B shows mean body weight ± standard error of the means for different groups. Mice receiving 1.0 mg CMZ 203-43 had significant weight loss, while mice receiving quinazoline at either dose did not. All of the mice receiving 4.0 mg CMZ 203-43 group were dead on day 4. Necropsy of mice in the 4.0 mg CMZ 203-43 group revealed bowel obstruction. FIG. 3 shows white blood cell (WBC) differential as mean percentage of total white cells ± standard error of the means for different groups. Mice treated with 1.0 mg CMZ 203- 43 had a significant neutrophilia while mice receiving quinazoline at either dose did not.
Data for mice treated with 4.0 mg CMZ 203-43 was not available because all these mice were dead before day 5.
Example 10 In Vivo Inhibition of IP- 10 by 4-Primary Amino Quinoline and by Quinazolines BALB/c mice (n=5) were pretreated with intraperitoneal injection of 40 μg quinoline 203-43, quinazoline 203-34, 203-49, or 203-51, or with 100 μl of sterile phosphate buffered saline (PBS) control. One hour after the injection, mice were injected subcutaneously with 100 μg CpG ODN 2006. Animals were bled at 3 hr post injection with CpG ODN 2006 and IP- 10 levels in plasma measured by IP- 10-specific enzyme-linked immunosorbent assay (ELISA). Results are presented in FIG. 4. Serum IP- 10 concentration was significantly reduced in mice pretreated with 203-43 (p=0.00013 versus PBS control) or 203-34 (p=0.00044 versus PBS control). - I l l - Example 11 In Vivo Inhibition ofTNF-a by 4-Primary Amino Quinoline and by Quinazolines Groups of BALB/c mice treated as in Example 10 were also analyzed for TNF-α. Animals were bled at 1 hr post injection with CpG ODN 2006 and TNF-α levels in plasma measured by TNF-α-specific ELISA. Results are presented in FIG. 5. Serum TNF-α concentration was significantly reduced in mice pretreated with 203-43 (p=0.0023 versus PBS control) or 203-34 (p=0.0012 versus PBS control). Mice treated with 203-49 showed a trend toward reduced serum TNF-α concentration.
Example 12 Synthesis of CMZ 203-84
Figure imgf000112_0001
200.21 100.17 268.35 A mixture of 4-fluorobenzophenone (16 gm, 0.08 moles) and N-methylpiperazine (14 gm, 0.14 moles) was stirred and refiuxed for 20 hours in water containing sodium carbonate (12.7 gm, 0.12 moles). After cooling, the mixture was extracted with ethyl acetate (200 mL). The organic phase was washed with water (100 mL) and was then extracted with 10% hydrochloric acid solution (3 X 50 mL). The combined acid extracts were washed with ethyl acetate (100 mL) and were then made basic by the addition of 40% sodium hydroxide solution. The solid aminobenzophenone was extracted into ethyl acetate (200 mL) and the extracts were washed with brine. The ethyl acetate was evaporated under vacuum to give the product as a white solid in a yield of 7.0 gm, 33%.
Figure imgf000113_0001
268.35 270.37 CMZ 203-84
Example 13 Synthesis of CMZ 203-85 A mixture of the benzophenone (15 gm, 0.056 moles) and powdered sodium hydroxide (15 gm) in ethanol (150 mL) was stirred and heated to reflux. The heat was removed and zinc dust (15 gm) was added in portions at a rate that kept the mixture at reflux. After the addition was complete, the mixture was heated at reflux for one hour. The reaction mixture was allowed to cool and was then filtered to remove zinc. The zinc was washed with ethanol (20 mL) and the combined filtrates were added to water (500 mL). The crystalline white product was isolated by filtration, washed with water and dried. The carbinol was obtained in a yield of 12.1 gm, 80%.
270.37 353.50 CMZ 203-85
A solution of the carbinol (1.13 gm, 4.18 X 10" moles) in NMP (10 mL) was stirred under nitrogen as a sodium hydride dispersion (0.336 gm of 60%, 8.36 X 10"3 moles) was added. This mixture was stirred at 65°C until hydrogen evolution stopped. Then 2- (dimethylamino)ethyl chloride hydrochloride (0.602 gm, 4.18 X 10"3 moles) was added and stirring at 65 °C was continued for one hour. The addition of identical portions of sodium hydride dispersion and 2-(dimethylamino)ethyl chloride hydrochloride was done twice more at one hour intervals. After the last addition, the mixture was stirred and heated for two hours and then cooled. The mixture was poured into 10% sodium hydroxide solution (200 mL) and this suspension was extracted with methylene chloride (2 X 100 mL). The combined extracts were washed with 10% sodium hydroxide solution (100 mL) and were then stripped under vacuum to give the crude product contaminated with some NMP as well as a small amount of starting material. The product was purified by chromatography on silica using 20% methanol in methylene chloride to elute the impurities. The product was then eluted with 20% methanol in methylene chloride containing 1% diethylamine. The yield of CMZ 203-85 was 0.540 gm, 37% as a light brown oil. Example 14 Synthesis of CMZ 203-88
Figure imgf000115_0001
268.35 295.38 CMZ 203-88
The benzophenone (4.65 gm, 0.0173 moles) and hydroxylamine hydrochloride (1.89 gm, 0.027 moles) were combined in ethanol (25 mL). To this mixture was added 40% sodium hydroxide solution (8.7 gm, 0.087) moles. The resulting mixture was stirred at reflux until TLC (silica, 10%) methanol in methylene chloride) showed that the reaction had gone to completion (about 30 minutes). After cooling, a solution of sodium bicarbonate (20 gm) in water (300 mL) was added and the precipitated solid was isolated by filtration. After washing with water and drying, there was obtained 4.85 gm (95%) of CMZ 203-88 as a white solid.
Example 15 Synthesis of CMZ 203-88-1
Figure imgf000116_0001
295.38 266.38 CMZ 203-88-1
A solution of the oxime (1.0 gm, 3.39 X 10" moles) in ethanol (20 mL) was stirred at reflux in the presence of 10% palladium on carbon (200 mg). To this mixture was added 90%) formic acid (0.35 gm, 6.77 X 10" moles) dropwise. Upon complete addition of the formic acid, the reaction mixture was heated at reflux for 30 minutes. The mixture was cooled and filtered free of the catalyst and the filtrates were stripped under vacuum. The residual oil quickly solidified to provide CMZ 203-88-1 in quantitative yield.
Example 16 Synthesis of CMZ 203-89
Figure imgf000117_0001
270.37 352.52 CMZ 203-89
A solution of the carbinol (1.4 gm, 5.0 X 10"3 moles) in DMF (10 mL) and methylene chloride (20 mL) was stirred as thionyl chloride (0.6 gm, 5.0 X 10"3 moles) was slowly added. After stirring at room temperature for 30 minutes, N,N-dimethylethylenediamine (0.88 gm, 1.0 X 10'3 moles) was added dropwise to the hazy solution. This mixture was stirred at room temperature for 30 minutes and was then poured into 5% sodium hydroxide solution (400 mL). This mixture was extracted with methylene chloride (2 X 100 mL) and the combined extracts were washed with 5% sodium hydroxide solution (200 mL). After drying over magnesium sulfate the extracts were filtered and stripped under vacuum to give an oil. The product was purified by chromatography on silica using 20%) methanol in methylene chloride to elute the impurities. The product was then eluted with 20% methanol in methylene chloride containing 1% diethylamine. The yield of CMZ 203-89 was 0.588 gm, 33.4% as a light brown oil. Example 17 Synthesis of CMZ 203-91
Figure imgf000118_0001
295.38 377.53 CMZ 203-91
A solution of the carbinol (0.6 gm, 2.03 X 10"3 moles) in DMF (5 mL) was stirred as thionyl chloride (0.242 gm, 2.03 X 10' moles) was slowly added. After stirring at room temperature for 30 minutes, N-methylpiperazine (0.400 gm, 4.0 X 10"3 moles) was added dropwise to the solution. This mixture was stirred at room temperature for 30 minutes and was then poured into 5% sodium hydroxide solution (200 mL). This suspension was extracted with methylene chloride (2 X 100 mL) and the combined extracts were washed with water (100 mL). The extracts were stripped under vacuum to give an oil. The product was purified by chromatography on silica using 20% methanol in methylene chloride to elute the impurities. The product was then eluted with 20% methanol in methylene chloride containing 1% diethylamine. The yield of CMZ 203-91 was 0.58 gm, 77% as an oil which crystallized upon standing. Example 18 Human TLR9 Antagonism by Novel Diarylmethane Compounds of the Invention hTLR9-NFkB-293 cells were counted and seeded at 1.5xl04 cells per well in 96-well cell culture plates one day before assaying and cultured at 37°C / 5% CO2, as described in Example 7. The day of the assay, antagonist was added at varying concentrations starting at 50 μM with a 1/5 to 1/6 dilution for 7 steps. The cells were then stimulated with the EC50 dose of the TLR9 agonist, CpG-ODN 2006, and cultured for 16 h in a humidified incubator at 37°C. The culture supernatant was removed and the cells were treated with lysis buffer and stored at -80°C before measuring luciferase activity as described in Example 7. Sigmoidal antagonism curves were generated and the inhibitor concentration at which 50% of the agonist response was blocked (IC50) calculated. Results shown in Table 20 under the heading hTLR9 are the IC50 dose (μM) for blockade of a CpG ODN-generated signal in hTLR9-NFkB-293 cells.
Table 20. Human TLR9 Antagonism by Novel Diarylmethane Compounds of the Invention
Figure imgf000119_0001
Example 19 Synthesis of CMZ 203-34
Step l.
Figure imgf000120_0001
A mixture of 4-fluorobenzaldehyde (10 gm, 0.08 moles) and N-methylpiperazine (14 gm, 0.14 moles) in water (80 mL) containing sodium carbonate (12.7 gm, 0.12 moles) was stirred and refluxed for 21 hours. After cooling, the mixture was poured into a separatory funnel containing water (200 mL) and the oily product was extracted into methylene chloride (3 X 50 mL). The combined extracts were washed with water and were then stripped under vacuum. The product, which was isolated as an oil, solidified into a tan solid upon cooling. The solid product was triturated in hexane and isolated by filtration. After drying, there was obtained 14.9 gm (91%) of N-(4-formylphenyl)-N'-methylpiperazine as an off white solid.
Step 2.
Figure imgf000120_0002
136.15 204.27 320.39
Anthranilamide (9.94 gm, 0.073 moles) and N-(4-formylphenyl)-N'-methylpiperazine (14.9 gm, 0.073 moles) were combined in NMP (100 mL). This mixture was stirred and warmed until dissolution was complete. To the warm solution was added / toluenesulfonic acid monohydrate (2.0 gm) after which the solution was heated at 100 °C for 30 minutes. Acetic acid (50 mL) was added and the mixture was stirred and heated at 100 °C for an additional 30 minutes. Chloranil (17.9 gm, 0.073 moles) was added in portions over a period of approximately two minutes. The dark solution was heated at 100 °C for 10 minutes. The solution was cooled to room temperature and was diluted with water (500 mL) and tert- butylmethyl ether (TBME, 500 mL). The mixture was stirred and made basic by the addition of solid sodium carbonate. After stirring for 15 minutes the precipitated product was isolated by filtration. The crude product was re-suspended in warm water, stirred for 5 minutes and collected by filtration. After washing with water followed by TBME, the solid was air dried. The dried quinazolinone was recrystallized from n-butanol to give 11.1 gm (50%) of product as colorless needles.
Step 3.
Figure imgf000121_0001
320.39 338.83 734.94 CMZ203-34 The quinazolinone (2.4 gm, 7.5X10"3 moles) was added in portions with stirring to phosphorous oxychloride (10 mL). This mixture was warmed to 80°C to give a red solution from which solid began to precipitate. The mixture was stirred at 80°C for 15 minutes and was then heated briefly to reflux. Upon cooling the slurry was slowly added to a cold, stirred solution of 10% sodium carbonate (650 mL). After stirring for 15 minutes, the solid chloroquinazoline was extracted into chloroform (2 X 150 mL). The combined extracts were washed with brine and then dried over magnesium sulfate. The solution was filtered to remove the drying agent and the filtrates were stripped under vacuum to give a yellow solid. N-methylpyrrolidinone (NMP, 20 mL) was added and the solid was dissolved by warming. N,N-dimethylethylenediamine (1.32 gm, 0.015 moles) was added and the solution was then warmed at 100°C for 30 minutes. After cooling, the NMP solution was diluted with water (200 mL) and concentrated ammonium hydroxide (50 mL). The product, which had precipitated, was extracted into methylene chloride (2 X 100 mL). The extracts were combined, dried over magnesium sulfate and then, after filtration, were stripped to give the product as a light brown oil. This was purified by chromatography on silica using 10% methanol in methylene chloride as eluent. In this manner 600 mg (1.54 X 10"3 moles) of pure quinazoline was isolated as an oil. This was dissolved in t-butylmethyl ether (TBME, 50 mL) and was stirred as a solution of toluenesulfonic acid monohydrate (584 mg, 3.1 X 10"3 moles) dissolved in TBME (50 mL) was added. The solid bis-tosylate salt was isolated by filtration, washed with TBME and dried to give 1.0 gm of the product as a yellow solid.
Example 20 Synthesis of CMZ 203-44
Figure imgf000122_0001
240.69 88.15 390.46 CMZ 203-44
N,N-dimethylethylenediamine (3.5 gm, 0.04 moles) and 2-phenyl-4- chloroquinazoline (2.4 gm, 0.01 moles) were combined in n-butanol (50 mL) and were brought to reflux. Once reflux had been achieved, TLC (silica, 10% methanol in dichloromethane) showed that the reaction was complete. The solution was cooled and stripped. The residue was dissolved in t-butylmethyl ether (TBME) and the solution was washed with water. After drying over magnesium sulfate the solution was filtered and stripped to give 1.37 gm (0.0047 moles, 47%) of the crude product as an oil. This was dissolved in ethanol (25 mL) and was treated with a solution of concentrated sulfuric acid (0.46 gm, 0.0047 moles) dissolved in ethanol (5 mL). The crystalline sulfate salt began to separate within a few seconds and after 30 minutes at room temperature was isolated by filtration. The salt was washed with ethanol and dried. The yield was 1.77 gm, (96% from crude).
Example 21 Synthesis of CMZ 203-45
Figure imgf000123_0001
240.69 89.14 391.44 CMZ 203-45
W A slurry of 60% sodium hydride (0.80 gm, 0.02 moles) in NMP (50 mL) was stirred under nitrogen as N,N-dimethylethanolamine (1.96 gm, 0.022 moles, 2.2 mL) was slowly added via syringe. After the amine had been added the solution was stirred at 30°C for 10 minutes after which 2-phenyl-4-chloroquinazoline (2.4 gm, 0.01 moles) was added in a single portion. Stirring was continued at 40°C for 30 minutes after which TLC (silica, 10%
5 methanol in methylene chloride) showed that the reaction had gone to completion. The solution was cooled and poured into water (250 mL). The product was extracted into methylene chloride (3 X 50 mL). After drying over magnesium sulfate the solution was filtered and stripped to give the crude product as an oil. This was dissolved in ethanol (20 mL) and was treated with a solution of concentrated sulfuric acid (0.85 gm, 0.0087 moles)
0 dissolved in ethanol (10 mL). The crystalline sulfate salt separated slowly and after 60 minutes at room temperature was isolated by filtration. The salt was washed with ethanol and dried. The yield was 1.37 gm, (40% based on the sulfuric acid). Example 22 Synthesis of CMZ 203-49
Figure imgf000124_0001
320.39 489.59 CMZ 203-49
The quinazolinone (3.2 gm, 0.01 moles) was added in portions with stirring to phosphorous oxychloride (10 mL). This mixture was refluxed for 30 minutes. The orange
10 slurry was cooled and TBME (100 mL) was added. After stirring for 10 minutes the chloroquinazoline was isolated by filtration and added to a solution of the sodium salt of N,N-dimethylaminoethanol in NMP (50 mL). The solution of the sodium salt of N,N- dimethylaminoethanol in NMP was prepared as follows. A slurry of 60% sodium hydride (2.1 gm, 0.05 moles) in NMP (50 mL) was stirred under nitrogen as N,N-
15 dimethylethanolamine (4.9 gm, 0.055 moles) was slowly added via syringe. After the amine had been added the solution was stirred at 30°C for 10 minutes. This mixture was stirred at 100°C for 30 minutes. The cooled solution was added to methylene chloride (150 mL) and this solution was extracted with water (3 X 150 mL). The methylene chloride solution was stripped and the residue was dissolved in ethanol (50 mL). To this solution was added
W concentrated sulfuric acid (.8 gm, .008 moles) dissolved in ethanol (5 mL). The solid salt which separated was isolated by filtration, washed with ethanol and TBME and dried. The yield was 1.3 gm, 23%. Example 23 Synthesis of CMZ 203-51
Step 1.
Figure imgf000125_0001
136.15 182.22 298.33
A solution of anthranilamide (6.8 gm, 0.05 moles) and biphenylcarboxaldehyde (9.1 gm, 0.05 moles) in DMF (100 mL) was stirred as p-toluenesulfonic acid monohydrate (1.0 gm) was added. A yellow solution formed which quickly changed to a thick slurry as the dihydroquinazoline precipitated. The slurry was heated to 100°C which caused most of the solid to dissolve. To this hot mixture was added chloranil (12.3 gm, 0.05 moles) in portions over a 2 minute period. A dark solution formed from which the product began to crystallize. The mixture was heated to boiling to provide a dark solution. Upon cooling, the quinazolinone crystallized as colorless needles. The product was isolated by filtration, washed well with DMF and acetone, and then dried. The yield was 11.9 gm, 80%.
Figure imgf000125_0002
298.33 371.25 CMZ 203-51 Phosphorous oxychloride (12 mL) and the biphenylquinazoline (2.98 gm, 0.01 moles) were stirred together and refluxed for one hour. The excess phosphorous oxychloride was removed by distillation and the oily yellow residue was placed under aspirator vacuum to remove traces of phosphorous oxychloride. The resulting yellow solid was triturated in hexane which was removed by decantation. To the yellow solid was added n-butanol (50 mL) and N,N-dimethylethylenediamine (6 mL). This solution was stirred and refluxed for 30 minutes. The butanol solution was cooled and poured into a separatory funnel containing TBME (200 mL) and 10% aqueous hydrochloric acid (200 mL). The funnel was vigorously shaken after which the layers were allowed to separate. The aqueous layer was isolated and the flask in which it was collected was scratched with a glass rod. The product began to immediately separate as a pale yellow solid. After 30 minutes at room temperature the product was isolated by filtration and was washed with a small amount of cold water. After drying there was obtained 3.64 gm (82%) of product as a pale yellow solid.
Example 24 Synthesis of CMZ 203-76
Step l.
Figure imgf000126_0001
259.09 88.15 310.78
A solution of 2,4-dichloro-6,7-dimethoxyquinazoline (5.2 gm, 0.02 moles) and N,N- dimethylethylenediamine (1.76 gm, 0.02 moles) in methylene chloride (65 mL) was stirred at room temperature for 2 hours. To the slurry that had formed another portion of N,N- dimethylethylenediamine (1.76 gm, 0.02 moles) was added forming a clear solution. This solution was stirred at room temperature overnight. The solution was poured into 10% sodium carbonate solution (200 mL) causing the product to separate as a solid. The solid was isolated by filtration and then washed with water followed by methylene chloride. This solid was recrystallized from 2-propanol to give 2.6 gm (42%) of the purified product as a white crystalline solid.
Step 2.
Figure imgf000127_0001
310.76 100.17 570.03 CMZ 203-76 A mixture of the chloroquinazoline (930 mg, 0.003 moles) and N-methylpiperazine
(5 mL) was heated at 100°C for 3 hours. The solution was cooled and stripped under vacuum. n-Butanol (10 mL) was added and the solution was again stripped under vacuum to remove residual N-methyl piperazine. The residue was dissolved in ethanol (20 mL) and a solution of sulfuric acid (0.59 gm, 0.006 moles) in ethanol (5 mL) was added. The mixture was refluxed for one minute and then cooled. The product was isolated by filtration, washed with ethanol followed by TBME and then dried. The yield was 0.85 gm, (50%).
Example 25 Synthesis of CMZ 203-78
Figure imgf000127_0002
310.78 162.24 436.56 CMZ 203-78 A mixture of the chloroquinazoline (1.55 gm, 0.005 moles) and N-phenylpiperazine (1.6 gm, 0.01 moles) in n-butanol (5 mL) was heated at 100°C for 2 hours. The resulting slurry was diluted with n-Butanol (15 mL) and the mixture was heated to boiling to give a clear solution. Upon cooling a solid separated which was isolated by filtration. The solid was stirred in warm water for 15 minutes to remove a small amount of N-phenyl-piperazine. The product was isolated by filtration, washed with warm water and dried to provide 1.0 gm (46%) of the product as a white solid.
Example 26 Synthesis of CMZ 203-87
Figure imgf000128_0001
338.83 130.19 432.56 CMZ 203-87
A mixture of the 4-chloroquinazoline (2.74 gm, 8.0X10" moles) and N-aminoethyl- morpholine (2.10 gm, 1.6X10" moles) in ethanol (25 mL) was refluxed for 2 hours. The reaction mixture was cooled and poured into water (200 mL) containing ammonia (50 mL of 28%) and the precipitated product was extracted into methylene chloride (3 X 100 mL). The combined extracts were washed with water (200 mL) and then dried over magnesium sulfate. After filtering to remove the drying agent the solution was stripped under vacuum to give the crude product as a tan solid. This solid was dissolved in warm methylene chloride (10 mL) and the solution was diluted with warm hexane (20 mL). Upon cooling, the product crystallized as a tan solid. This was isolated by filtration, the solid washed with 30% methylene chloride in hexane and dried. The yield was 1.2 gm, (35%). Example 27 Synthesis of CMZ 203-93
Step 1.
Figure imgf000129_0001
136.15 107.11 223.23
To a solution of anthranilamide (13.6 gm, 0.10 moles) and 3-pyridinecarboxaldehyde (10.7 gm, 0.10 moles) in NMP (100 mL) and acetic acid (50 mL) was added p-toluenesulfonic acid monohydrate (2.0 gm). This solution was stirred at 50°C for 30 minutes at which point chloranil (24.6 gm, 0.10 moles) was added in portions through a funnel during one minute. The dark solution was stirred and cooled to room temperature which caused the quinazolinone to crystallize. The solid was isolated by filtration, washed with acetone/NMP (1 :1) followed by acetone. After drying, the crude product was recrystallized from n-butanol (200 mL) and NMP (90 mL). The quinazolinone was isolated as tan fibrous crystals in a yield of 10.6 gm, 47.5%.
Step 2.
Figure imgf000129_0002
223.23 241.67 335.40 CMZ 203-93 A portion of the quinazolinone (4.46 gm, 0.02 moles) was refluxed in phosphorous oxychloride (20 mL) for one hour. After cooling, the solution was carefully poured into cold 20%) sodium carbonate solution (500 mL). The solid chloroquinazoline was isolated by filtration, washed with water and dissolved in methylene chloride (400 mL). The solution was dried over magnesium sulfate, filtered and stripped to give the chloroquinazoline as a pale yellow solid in a yield of 3.7 gm, 76.5%. The 4-chloroquinazoline (3.7 gm, 0.015 moles) was refluxed in ethanol with N-2-aminoethylmorpholine (3.99 gm, 0.031 moles) for one hour. Once cooled, the solution was stripped and the residue was dissolved in water (400 mL). The solution was made basic by the addition of sodium carbonate and the product was extracted into methylene chloride (2 X 100 mL). The combined extracts were washed with water (50 mL) and were then dried over magnesium sulfate. After filtration, the extracts were stripped to give the product as a tan solid. The solid was recrystallized from ethyl acetate (25 mL) and hexane (50 mL). The product, 203-93 was isolated as an off white solid in a yield of 2.86 gm, 56.0%>.
Example 28 Synthesis of CMZ 203-95
Step 1.
136.15 107.11 223.23
To a solution of anthranilamide (13.6 gm, 0.10 moles) and 2-pyridinecarboxaldehyde (10.7 gm, 0.10 moles) in methanol (250 mL) and acetic acid (25 mL) was added p-toluenesulfonic acid monohydrate (2.0 gm). This solution was stirred at 50°C for 30 minutes at which point chloranil (24.6 gm, 0.10 moles) was added in portions through a funnel during one minute. The funnel was washed with NMP (25 mL). This mixture was heated to reflux for 5 minutes. The dark solution was stirred and cooled to room temperature which caused the quinazolinone to crystallize. The solid was isolated by filtration, washed with acetone. After drying, the crude product was recrystallized twice from n-butanol. A small amount of tetrachlorohydroquinone will co-crystallize under these conditions. It can be removed by stirring the solid in warm 5% sodium carbonate solution. The quinazolinone was isolated as a tan solid in a yield of 9.5 gm, 43%>.
Step 2.
Figure imgf000131_0001
CMZ 203-95
A portion of the quinazolinone (4.46 gm, 0.02 moles) was refluxed in phosphorous oxychloride (20 mL) for 30 minutes. After cooling, the solution was carefully poured into cold water (500 mL). This solution was neutralized by the addition of 40% sodium hydroxide solution which caused the chloroquinazoline to separate as a white solid. The solid chloroquinazoline was isolated by filtration, washed with water and dissolved in methylene chloride (300 mL). The solution was dried over magnesium sulfate, filtered and stripped to give the chloroquinazoline as a pale yellow solid in a yield of 2.75gm, 57%). The 4-chloroquinazoline (2.75 gm, 0.0114 moles) was refluxed in ethanol with N-2- aminoethylmorpholine (2.97 gm, 0.0228 moles) for one 30 minutes. Once cooled, the solution was poured into 10% sodium hydroxide solution (200 mL) and the product was extracted into methylene chloride (3 X 100 mL). The combined extracts were washed with 10% sodium hydroxide (150 mL) and were then dried over magnesium sulfate. After filtration, the extracts were stripped to give the product as a tan solid. The solid was recrystallized from n-butanol (10 mL) and hexane (20 mL). The product, 203-95 was isolated as an off white solid in a yield of 1.32 gm, 34.5%). EQUIVALENTS The foregoing written specification is considered to be sufficient to enable one skilled in the art to practice the invention. The present invention is not to be limited in scope by examples provided, since the examples are intended as a single illustration of one aspect of the invention and other functionally equivalent embodiments are within the scope of the invention. Various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims. The advantages and objects of the invention are not necessarily encompassed by each embodiment of the invention. All references, patents and patent publications that are recited in this application are incorporated in their entirety herein by reference.
We claim:

Claims

CLAIMS 1. A method of affecting TLR-mediated signaling in response to a TLR ligand, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula I
Figure imgf000133_0001
wherein 2 is a five- to seven-membered homocyclic or heterocyclic ring, wherein each of X, Y, and Z is independently chosen from C, N, O, and S, and wherein B2 optionally includes at least one atom selected from C, N, O, and S; wherein 1 and 2 are optionally bridged by B3 to form a five- to seven-membered ring (3), wherein B3 optionally includes at least one atom selected from C, N, O, and S, and wherein when A is carbon, (3) is not pyridine; wherein 2 optionally includes an unsaturated bond; wherein (3) optionally includes an unsaturated bond; wherein R2, when present, is a hydrogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, optionally linked to Z via N, O, or S; wherein R3, R4, R5, R6, R7, and R8, when present, are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein A is an atom selected from C, N, O, and S; wherein each of A', A", and A'" independently is R9, -NR9R10, -OR9, or -CR9R10, wherein R9 is a hydrogen atom, hydroxyl, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, and wherein RIO is optionally absent or is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR.
2. A method of inhibiting TLR-mediated immunostimulatory signaling, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula I
Figure imgf000134_0001
Formula I
wherein 2 is a five- to seven-membered homocyclic or heterocyclic ring, wherein each of X, Y, and Z is independently chosen from C, N, O, and S, and wherein B2 optionally includes at least one atom selected from C, N, O, and S; wherein 1 and 2 are optionally bridged by B3 to form a five- to seven-membered ring (3), wherein B3 optionally includes at least one atom selected from C, N, O, and S, and wherein when A is carbon, (3) is not pyridine; wherein 2 optionally includes an unsaturated bond; wherein (3) optionally includes an unsaturated bond; wherein R2, when present, is a hydrogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, optionally linked to Z viaN, O, or S; wherein R3, R4, R5, R6, R7, and R8, when present, are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein A is an atom selected from C, N, O, and S; wherein each of A', A", and A'" independently is R9, -NR9R10, -OR9, or -CR9R10, wherein R9 is a hydrogen atom, hydroxyl, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, and wherein RIO is optionally absent or is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
3. A method of affecting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula I
Figure imgf000135_0001
Formula I
wherein 2 is a five- to seven-membered homocyclic or heterocyclic ring, wherein each of X, Y, and Z is independently chosen from C, N, O, and S, and wherein B2 optionally includes at least one atom selected from C, N, O, and S; wherein 1 and 2 are optionally bridged by B 3 to form a five- to seven-membered ring (3), wherein B3 optionally includes at least one atom selected from C, N, O, and S, and wherein when A is carbon, (3) is not pyridine; wherein 2 optionally includes an unsaturated bond; wherein (3) optionally includes an unsaturated bond; wherein R2, when present, is a hydrogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, optionally linked to Z via N, O, or S; wherein R3, R4, R5, R6, R7, and R8, when present, are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein A is an atom selected from C, N, O, and S; wherein each of A', A", and A'" independently is R9, -NR9R10, -OR9, or -CR9R10, wherein R9 is a hydrogen atom, hydroxyl, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, and wherein RIO is optionally absent or is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated immunostimulation in the subject.
4. A method of inhibiting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula I
Figure imgf000136_0001
wherein 2 is a five- to seven-membered homocyclic or heterocyclic ring, wherein each of X, Y, and Z is independently chosen from C, N, O, and S, and wherein B2 optionally includes at least one atom selected from C, N, O, and S; wherein 1 and 2 are optionally bridged by B3 to form a five- to seven-membered ring (3), wherein B3 optionally includes at least one atom selected from C, N, O, and S, and wherein when A is carbon, (3) is not pyridine; wherein 2 optionally includes an unsaturated bond; wherein (3) optionally includes an unsaturated bond; wherein R2, when present, is a hydrogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, optionally linked to Z viaN, O, or S; wherein R3, R4, R5, R6, R7, and R8, when present, are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein A is an atom selected from C, N, O, and S; wherein each of A', A", and A'" independently is R9, -NR9R10, -OR9, or -CR9R10, wherein R9 is a hydrogen atom, hydroxyl, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, and wherein RIO is optionally absent or is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit TLR-mediated immunostimulation in the subject.
5. A method of inhibiting an immunostimulatory nucleic acid-associated response in a subject, comprising administering to a subject in need of such treatment an effective amount of a compound of Formula I
Figure imgf000137_0001
Formula I
wherein 2 is a five- to seven-membered homocyclic or heterocyclic ring, wherein each of X, Y, and Z is independently chosen from C, N, O, and S, and wherein B2 optionally includes at least one atom selected from C, N, O, and S; wherein 1 and 2 are optionally bridged by B3 to form a five- to seven-membered ring (3), wherein B3 optionally includes at least one atom selected from C, N, O, and S, and wherein when A is carbon, (3) is not pyridine; wherein 2 optionally includes an unsaturated bond; wherein (3) optionally includes an unsaturated bond; wherein R2, when present, is a hydrogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, optionally linked to Z via N, O, or S; wherein R3, R4, R5, R6, R7, and R8, when present, are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein A is an atom selected from C, N, O, and S; wherein each of A', A", and A'" independently is R9, -NR9R10, -OR9, or -CR9R10, wherein R9 is a hydrogen atom, hydroxyl, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, and wherein RIO is optionally absent or is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit an immunostimulatory nucleic acid-associated response in the subject.
6. The method according to any one of claims 3-5, wherein the subject is otherwise free of symptoms calling for treatment with a compound of Formula I.
7. The method according to any one of claims 1-5, wherein R2 is a substituted or unsubstituted phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group.
8. The method according to any one of claims 1 -5, wherein R2 is a phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group substituted with one or more substituent groups selected from the group consisting of an alkyl group, an alkoxy group, an alkoxyalkyl group, an ester group, an alkylamino group, a dialkylamino group, a cyclic amino group, a halogen atom, and any combination thereof.
9. The method according to any one of claims 1-5, wherein R2 is a phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group substituted with one or more substituent groups selected from the group consisting of an alkylamino group, a dialkylamino group, and a cyclic amino group.
10. The method according to claim 9, wherein the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group.
11. The method according to any one of claims 1 -5, wherein R9 is a substituted alkyl group selected from the group consisting of a cyclic amino group, an alkylamino group, a dialkylamino group, furyl, phenyl, thienyl, azabicyclooctyl, azabicycloheptyl, and any combination thereof.
12. The method according to claim 11, wherein the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group.
13. The method according to any one of claims 1-5, wherein each of R4 and R5 is hydrogen atom.
14. The method according to any one of claims 1-5, wherein each of R4, R5, and R8 is a hydrogen atom.
15. The method according to any one of claims 1-5, wherein RIO is a hydrogen atom.
16. The method according to any one of claims 1-5, wherein A is nitrogen and (3) is a five-membered ring.
17. The method according to any one of claims 1-5, wherein the compound is Formula II
Figure imgf000139_0001
F ormula [I
wherein Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S.
18. The method according to any one of claims 1-5, wherein A is nitrogen, (3) is a six- membered ring, and B3 is one atom selected from C, N, O, and S.
19. The method according to any one of claims 1-5, wherein A is nitrogen, 3 is a six- membered ring, and B3 is S.
20. The method according to any one of claims 1-5, wherein the compound is Formula III
Figure imgf000140_0001
Formula III
wherein Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S, and wherein X is chosen from N, O, and S.
21. The method according to any one of claims 1-5, wherein A is carbon, (3) is a six- membered ring, and B3 is S.
22. The method according to any one of claims 1-5, wherein the compound is Formula IV
Figure imgf000140_0002
Formula IV
wherein X is C or S and Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S.
23. The method according to any one of claims 1-5, wherein A is nitrogen, (3) is a seven- membered ring, and B3 comprises two carbon atoms.
24. The method according to any one of claims 1-5, wherein the compound is Formula V
Figure imgf000141_0001
Formula V
wherein X and Y are each independently C, N, O, or S; and Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S.
25. The method according to any one of claims 1-5, wherein 1 and 2 are unbridged by B3, A is carbon, and A' is -OR9.
26. The method according to any one of claims 1-5, wherein the compound is Formula VI
Figure imgf000141_0002
Formula VI
wherein Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and R9 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group.
27. The method according to any one of claims 1-5, wherein 1 and 2 are unbridged by B3, A is carbon, and A' is -NR9R10.
28. The method according to any one of claims 1-5, wherein the compound is Formula VII
Figure imgf000142_0001
Formula VII
wherein Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S.
29. The method according to any one of claims 1 -5, wherein 1 and 2 are unbridged by B3, A is carbon, and A' is -CR9R10.
30. The method according to any one of claims 1-5, wherein the compound is Formula VIII
Figure imgf000142_0002
Formula VIII
wherein Rl is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S.
31. A method of affecting TLR-mediated signaling in response to a TLR ligand, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula IX
Figure imgf000143_0001
Formula IX
wherein R2 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR.
32. A method of inhibiting TLR-mediated immunostimulatory signaling, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula IX
Figure imgf000143_0002
Formula IX
wherein R2 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
33. A method of affecting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula IX
Figure imgf000144_0001
Formula IX
wherein R2 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated immunostimulation in the subject.
34. A method of inhibiting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula IX
Figure imgf000145_0001
Formula IX
wherein R2 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit TLR-mediated immunostimulation in the subject.
35. A method of inhibiting an immunostimulatory nucleic acid-associated response in a subject, comprising administering to a subject in need of such treatment an effective amount of a compound of Formula IX
Figure imgf000145_0002
Formula IX wherein R2 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit an immunostimulatory nucleic acid-associated response in the subject.
36. The method according to any one of claims 33-35, wherein the subject is otherwise free of symptoms calling for treatment with a compound of Formula IX.
37. The method according to any one of claims 31-35, wherein R2 is a substituted or unsubstituted phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group.
38. The method according to any one of claims 31-35, wherein R2 is a phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group substituted with one or more substituent groups selected from the group consisting of an alkyl group, an alkoxy group, an alkoxyalkyl group, an ester group, an alkylamino group, a dialkylamino group, a cyclic amino group, a halogen atom, and any combination thereof.
39. The method according to any one of claims 31-35, wherein R2 is a phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group substituted with one or more substituent groups selected from the group consisting of an alkylamino group, a dialkylamino group, and a cyclic amino group.
40. The method according to claim 39, wherein the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group.
41. The method according to any one of claims 31-35, wherein R9 is a substituted alkyl group selected from the group consisting of a cyclic amino group, an alkylamino group, a dialkylamino group, furyl, phenyl, thienyl, azabicyclooctyl, azabicycloheptyl, and any combination thereof.
42. The method according to claim 41 , wherein the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group.
43. The method according to any one of claims 31-35, wherein each of R5 and R8 is a hydrogen atom.
44. The method according to any one of claims 31-35, wherein R10 is a hydrogen atom.
45. A method of affecting TLR-mediated signaling in response to a TLR ligand, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula X
Figure imgf000147_0001
Formula X
wherein R2 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked viaN, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR.
46. A method of inhibiting TLR-mediated immunostimulatory signaling, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula X
Figure imgf000148_0001
Formula X
wherein R2 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
47. A method of affecting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula X
Figure imgf000149_0001
Formula X
wherein R2 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked viaN, O, or S; wherein R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated immunostimulation in the subject.
48. A method of inhibiting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula X
Figure imgf000149_0002
Formula X
wherein R2 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked viaN, O, or S; wherein R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit TLR-mediated immunostimulation in the subject.
49. A method of inhibiting an immunostimulatory nucleic acid-associated response in a subject, comprising administering to a subject in need of such treatment an effective amount of a compound of Formula X
Figure imgf000150_0001
wherein R2 is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit an immunostimulatory nucleic acid-associated response in the subject.
50. The method according to any one of claims 47-49, wherein the subject is otherwise free of symptoms calling for treatment with a compound of Formula X.
51. The method according to any one of claims 45-49, wherein R2 is a substituted or unsubstituted phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group.
52. The method according to any one of claims 45-49, wherein R2 is a phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group substituted with one or more substituent groups selected from the group consisting of an alkyl group, an alkoxy group, an alkoxyalkyl group, an ester group, an alkylamino group, a dialkylamino group, a cyclic amino group, a halogen atom, and any combination thereof.
53. The method according to any one of claims 45-49, wherein R2 is a phenyl, naphthyl, phenanthryl, styryl, azabicyclooctane, or azabicycloheptane group substituted with one or more substituent groups selected from the group consisting of an alkylamino group, a dialkylamino group, and a cyclic amino group.
54. The method according to claim 39, wherein the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group.
55. A method of affecting TLR-mediated signaling in response to a TLR ligand, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula XI
Figure imgf000151_0001
Formula XI wherein each of Rl, R2, R5, R6, R7, R8, Rl 1, and R12 is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and X is C, N, O, or S, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR.
56. A method of inhibiting TLR-mediated immunostimulatory signaling, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula XI
Figure imgf000152_0001
Formula XI
wherein each of Rl, R2, R5, R6, R7, R8, Rl 1, and R12 is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and X is C, N, O, or S, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
57. A method of affecting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XI
Figure imgf000152_0002
Formula XI wherein each of Rl, R2, R5, R6, R7, R8, Rl 1, and R12 is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and X is C, N, O, or S, to inhibit or promote TLR-mediated immunostimulation in the subject.
58. A method of inhibiting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XI
10
Figure imgf000153_0001
Formula XI wherein each of Rl, R2, R5, R6, R7, R8, Rl 1, and R12 is a hydrogen atom, a halogen atom, a 75 substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and X is C, N, O, or S, to inhibit TLR-mediated immunostimulation in the subject.
59. A method of inhibiting an immunostimulatory nucleic acid-associated response in a 20 subject, comprising administering to a subject in need of such treatment an effective amount of a compound of Formula XI
Figure imgf000154_0001
Formula XI
wherein each of Rl, R2, R5, R6, R7, R8, Rl 1, and R12 is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and X is C, N, O, or S, to inhibit an immunostimulatory nucleic acid-associated response in the subject.
60. The method according to any one of claims 57-59, wherein the subject is otherwise free of symptoms calling for treatment with a compound of Formula XI.
61. The method according to any one of claims 55-59, wherein Rl is a substituted alkyl group selected from the group consisting of a cyclic amino group, an alkylamino group, a dialkylamino group, furyl, phenyl, thienyl, azabicyclooctyl, azabicycloheptyl, and any combination thereof.
62. The method according to claim 61, wherein the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group.
63. A method of affecting TLR-mediated signaling in response to a TLR ligand, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula XII
Figure imgf000155_0001
Formula XII wherein each of Rl-Rl 1 is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR.
64. A method of inhibiting TLR-mediated immunostimulatory signaling, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula XII
10
Figure imgf000155_0002
Formula XII wherein each of Rl-Rl 1 is a hydrogen atom, a halogen atom, a substituted or unsubstituted 75 lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
65. A method of affecting TLR-mediated immunostimulation in a subject, comprising 20 administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XII
Figure imgf000156_0001
Formula XII
wherein each of Rl-Rl 1 is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S, to inhibit or promote TLR-mediated immunostimulation in the subject.
66. A method of inhibiting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XII
Figure imgf000156_0002
Formula XII
wherein each of Rl-Rl 1 is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S, to inhibit TLR-mediated immunostimulation in the subject.
67. A method of inhibiting an immunostimulatory nucleic acid-associated response in a subject, comprising administering to a subject in need of such treatment an effective amount of a compound of Formula XII
Figure imgf000157_0001
Formula XII
wherein each of Rl-Rl 1 is a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S, to inhibit an immunostimulatory nucleic acid-associated response in the subject.
68. The method according to any one of claims 65-67, wherein the subject is otherwise free of symptoms calling for treatment with a compound of Formula XII.
69. A method of affecting TLR-mediated signaling in response to a TLR ligand, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula XIII
Figure imgf000157_0002
Formula XIII wherein 2' is a five- to seven-membered homocyclic or heterocyclic ring, wherein each of X, Y, and Z is independently chosen from C, N, O, and S, and wherein B2' optionally includes at least one atom selected from C, N, O, and S; wherein 2' optionally comprises an unsaturated bond; wherein R4, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein A" is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR.
70. A method of inhibiting TLR-mediated immunostimulatory signaling, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula XIII
Figure imgf000158_0001
Formula XIII
wherein 2' is a five- to seven-membered homocyclic or heterocyclic ring, wherein each of X, Y, and Z is independently chosen from C, N, O, and S, and wherein B2' optionally includes at least one atom selected from C, N, O, and S; wherein 2' optionally comprises an unsaturated bond; wherein R4, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein A" is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
71. A method of affecting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XIII
Figure imgf000159_0001
Formula XIII
wherein 2' is a five- to seven-membered homocyclic or heterocyclic ring, wherein each of X,
Y, and Z is independently chosen from C, N, O, and S, and wherein B2' optionally includes at least one atom selected from C, N, O, and S; wherein 2' optionally comprises an unsaturated bond; wherein R4, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked viaN, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein A" is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated immunostimulation in the subject.
72. A method of inhibiting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XIII
Figure imgf000160_0001
Formula XIII
wherein 2' is a five- to seven-membered homocyclic or heterocyclic ring, wherein each of X,
Y, and Z is independently chosen from C, N, O, and S, and wherein B2' optionally includes at least one atom selected from C, N, O, and S; wherein 2' optionally comprises an unsaturated bond; wherein R4, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; wherein RIO is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein A" is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit TLR-mediated immunostimulation in the subject.
13>. A method of inhibiting an immunostimulatory nucleic acid-associated response in a subject, comprising administering to a subject in need of such treatment an effective amount of a compound of Formula XIII
Figure imgf000161_0001
Formula XIII
wherein 2' is a five- to seven-membered homocyclic or heterocyclic ring, wherein each of X, Y, and Z is independently chosen from C, N, O, and S, and wherein B2' optionally includes at least one atom selected from C, N, O, and S; wherein 2' optionally comprises an unsaturated bond; wherein R4, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; wherein R10 is a hydrogen atom, lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group; and wherein A" is a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit an immunostimulatory nucleic acid-associated response in the subject.
74. The method according to any one of claims 71-73, wherein the subject is otherwise free of symptoms calling for treatment with a compound of Formula XIII.
75. The method according to any one of claims 69-73, wherein R9 is a substituted alkyl group selected from the group consisting of a cyclic amino group, an alkylamino group, a dialkylamino group, furyl, phenyl, thienyl, azabicyclooctyl, azabicycloheptyl, and any combination thereof.
76. The method according to claim 75, wherein the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group.
77. The method according to any one of claims 69-73, wherein A" is a substituted alkyl group selected from the group consisting of a cyclic amino group, an alkylamino group, a dialkylamino group, furyl, phenyl, thienyl, azabicyclooctyl, azabicycloheptyl, and any combination thereof.
78. The method according to claim 77, wherein the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group.
79. The method according to any one of claims 69-73, wherein each of R5 and R8 is hydrogen atom.
80. The method according to any one of claims 69-73, wherein R10 is a hydrogen atom.
81. A method of affecting TLR-mediated signaling in response to a TLR ligand, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula XIV or Formula XV
Figure imgf000162_0001
Formula XIV
Figure imgf000163_0001
Formula XV
wherein Rl, R2, R3, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated signaling in response to a ligand for the TLR.
82. A method of inhibiting TLR-mediated immunostimulatory signaling, comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula XIV or Formula XV
Figure imgf000163_0002
Formula XIV
Figure imgf000163_0003
Formula XV wherein Rl, R2, R3, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit TLR-mediated immunostimulatory signaling in response to a ligand for the TLR.
83. A method of affecting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XIV or Formula XV
Figure imgf000164_0001
Formula XIV
Figure imgf000164_0002
Formula XV
wherein Rl, R2, R3, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit or promote TLR-mediated immunostimulation in the subject.
84. A method of inhibiting TLR-mediated immunostimulation in a subject, comprising administering to a subject having or at risk of developing TLR-mediated immunostimulation an effective amount of a compound of Formula XIV or Formula XV
Figure imgf000165_0001
Formula XIV
Figure imgf000165_0002
Formula XV
wherein Rl, R2, R3, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit TLR-mediated immunostimulation in the subject.
85. A method of inhibiting an immunostimulatory nucleic acid-associated response in a subject, comprising administering to a subject in need of such treatment an effective amount of a compound of Formula XIV or Formula XV
Figure imgf000166_0001
Formula XIV
Figure imgf000166_0002
Formula XV
wherein Rl, R2, R3, R5, R6, R7, and R8 are each independently a hydrogen atom, a halogen atom, a substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, each optionally linked via N, O, or S; and wherein R9 is a hydrogen atom, substituted or unsubstituted lower alkyl, aryl, aralkyl, heterocyclic, or alkylheterocyclic group, to inhibit an immunostimulatory nucleic acid-associated response in the subject.
86. The method according to any one of claims 83-85, wherein the subject is otherwise free of symptoms calling for treatment with a compound of Formula XIV or Formula XV.
87. The method according to any one of claims 81-85, wherein R9 is a substituted alkyl group selected from the group consisting of a cyclic amino group, an alkylamino group, a dialkylamino group, furyl, phenyl, thienyl, azabicyclooctyl, azabicycloheptyl, and any combination thereof.
88. The method according to claim 87, wherein the cyclic amino group is a piperazino group, a piperidino group, a pyrrolidino group, an imidazolyl group, a pyridyl group, or a morpholino group.
89. A substituted 4-primary amino quinoline composition having a structural formula
Figure imgf000167_0001
Formula XVI wherein X is absent or is an aryl, alkyl, heterocyclic, or styryl group; R] and R2 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group, wherein R\ and R optionally are combined to form a heterocycle; R3 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein R\ and R3 optionally are combined to form a heterocycle; Y is absent or is an oxygen atom, a sulfur atom, CR8R9, or NR10, where R8, R9, and Rio are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group; L is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; and Rt, R5, Re, and R are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein any pair of R^ R5, Re, and R7 which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof.
90. The substituted 4-primary amino quinoline composition of claim 89, wherein R and Re are each independently a halogen atom or an alkoxy group.
91. The substituted 4-primary amino quinoline composition of claim 89, wherein R5 and R are each independently a chlorine atom or a methoxy group.
92. The substituted 4-primary amino quinoline composition of claim 89, wherein X is absent or is an aryl group; NR]R2 is a heterocyclic amine or is NR8(CH2)nNR9R10, wherein n is an integer from 2 to 6, inclusive, and R8, R9, and R10 are each independently a hydrogen atom or an alkyl group; R3 is a hydrogen atom; Y is an aryl group or is NRπ where Rπ is a hydrogen atom or an aryl or alkyl group; L is absent or is a C2-C6 alkyl group; and R;, Rs, Re, and R are each independently a hydrogen atom, a halogen atom, or an alkoxy group, and pharmaceutically acceptable hydrates and salts thereof.
93. The substituted 4-primary amino quinoline composition of claim 89, wherein: X is absent or is an aryl group; NRjR2 is a substituted or unsubstituted piperazino or morpholino group or is NR8(CH2)nNR9R10, wherein n is an integer from 2 to 6, inclusive, R8 is a hydrogen atom, and R9 and Rι0 are each independently an alkyl group; R3 is a hydrogen atom; Y is NH; L is a C2-C6 alkyl group; and R , R5, R_, and R7 are each independently a hydrogen atom, a halogen atom, or an alkoxy group, and pharmaceutically acceptable hydrates and salts thereof.
94. The substituted 4-primary amino quinoline composition of claim 89, wherein X is a phenyl group;
Figure imgf000169_0001
attached to a para position of the phenyl group X; Y is NH; L is -(CH2)n- where n is an integer between 2 and 6, inclusive; and each of R3, R , R5, Re, and R7 is a hydrogen atom, and pharmaceutically acceptable hydrates and salts thereof.
95. A method of inhibiting signaling by a Toll-like receptor (TLR), comprising contacting a cell expressing a functional TLR with an effective amount of a substituted 4-primary amino quinoline composition having a structural formula
Figure imgf000169_0002
Formula XVII wherein X is absent or is a nitrogen, oxygen, or sulfur atom or an SO or SO2 group; R] is a hydrogen atom or a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group; R2 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonarnido group, wherein Ri and R optionally are combined to form a heterocycle or carbocycle; Y is absent or is an oxygen atom, a sulfur atom, CR7R8, or NR9, where R , R8, and R9 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group; L is absent or is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; and R3, RA, R5, and Re are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein any pair of R3, R4, R5, and R_ which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof, to inhibit signaling by the TLR.
96. The method of claim 95, wherein the substituted 4-primary amino quinoline composition is a substituted 4-primary amino quinoline composition of any one of claims 89- 94.
97. The method of claim 95, wherein the TLR is Toll-like receptor 9 (TLR9).
98. A method of inhibiting signaling by a Toll-like receptor (TLR), comprising contacting an immune cell expressing a functional TLR with (a) an effective amount of a TLR signal agonist to stimulate signaling by the TLR in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having a structural formula
Figure imgf000170_0001
Formula XVII wherein X is absent or is a nitrogen, oxygen, or sulfur atom or an SO or SO2 group; Ri is a hydrogen atom or a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group; R2 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein Ri and R2 optionally are combined to form a heterocycle or carbocycle; Y is absent or is an oxygen atom, a sulfur atom, CR7R8, or NR9, where R7, R8, and R9 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group; L is absent or is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; and R3, Rt, R5, and Re are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein any pair of R3, Rt, R5, and Re which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof, to inhibit signaling by the TLR in response to the TLR signal agonist compared with the signaling by the TLR in response to the TLR signal agonist in absence of the substituted 4-primary amino quinoline composition.
99. The method of claim 98, wherein the substituted 4-primary amino quinoline composition is a substituted 4-primary amino quinoline composition of any one of claims 89- 94.
100. The method of claim 98, wherein the TLR signal agonist is CpG DNA.
101. The method of claim 98, wherein the TLR signal agonist is an immune complex comprising a nucleic acid.
102. The method of claim 98, wherein the TLR is Toll-like receptor 9 (TLR9).
103. A method of inhibiting an immune response to an antigenic substance, comprising contacting an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a substituted 4-primary amino quinoline composition, and (b) an effective amount of a substituted 4-primary amino quinoline composition having a structural formula
Figure imgf000172_0001
Formula XVII wherein X is absent or is a nitrogen, oxygen, or sulfur atom or an SO or SO2 group; R\ is a hydrogen atom or a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group; R2 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein Ri and R2 optionally are combined to form a heterocycle or carbocycle; Y is absent or is an oxygen atom, a sulfur atom, CR R8, or NR9, where R , R8, and R9 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group; L is absent or is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; and R3, R<t, R5, and R. are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein any pair of R3, R4, R5, and Re which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the substituted 4-primary amino quinoline composition.
104. The method of claim 103, wherein the substituted 4-primary amino quinoline composition is a substituted 4-primary amino quinoline composition of any one of claims 89- 94.
105. The method of claim 103, wherein the contacting takes place in vivo.
106. The method of claim 103, wherein the immune response comprises an adaptive immune response.
107. The method of claim 103, wherein the immune response is an innate immune response.
108. A method of treating an autoimmune disorder in a subject, comprising administering to a subject having an autoimmune disorder an effective amount of a substituted 4-primary amino quinoline composition having a structural formula
Figure imgf000173_0001
Formula XVII wherein X is absent or is a nitrogen, oxygen, or sulfur atom or an SO or SO2 group; R\ is a hydrogen atom or a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group; R2 is a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein Ri and R optionally are combined to form a heterocycle or carbocycle; Y is absent or is an oxygen atom, a sulfur atom, CR R8, or NR , where R7, R8, and R9 are each independently a hydrogen atom or a substituted or unsubstituted alkyl, alkenyl, or aryl group; L is absent or is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; and R3, Rt, R5, and R are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein any pair of R3, R4, R5, and R which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof, to treat the autoimmune disorder.
109. The method of claim 108, wherein the substituted 4-primary amino quinoline composition is a substituted 4-primary amino quinoline composition of any one of claims 89- 94.
110. The method of claim 108, wherein the autoimmune disorder is chosen from systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Sjogren's syndrome, polymyositis, vasculitis, Wegener' s granulomatosis, sarcoidosis, ankylosing spondylitis, Reiter's syndrome, psoriatic arthritis, Behcet's syndrome.
111. The method of claim 108, wherein the autoimmune disorder is an immune complex associated disease.
112. The method of claim 108, wherein the subject is a human.
113. A quinazoline composition having a structural formula
Figure imgf000175_0001
wherein X is absent or is an aryl, alkyl, heterocyclic or styryl group, provided that if X is a phenyl group, NRjR is part if a heterocycle or is a diamine; Ri and R2 are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R\ and R2 optionally are combined to form a heterocycle; Y is an oxygen atom, a sulfur atom, CR90, or NRn, where R , Rio, and Rn are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R , Rio, and R\ \ optionally is combined with R3 or R4 to form a substituted or unsubstituted heterocycle; L is an alkyl or alkenyl group containing from 1 to 10 carbons or is an aryl group; R3 and Rt are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R3 and optionally are combined to form a heterocycle; and R5, Re, Rη, and R8 are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein any pair of R5, R^, R7, and Rg which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof.
114. The quinazoline composition of claim 113, wherein Re and R are each independently a halogen atom or an alkoxy group.
115. The quinazoline composition of claim 113, wherein R_ and R7 are each independently a chlorine atom or a methoxy group.
116. The quinazoline composition of claim 113, wherein X is absent or is an aryl group; NRιR2 is a heterocyclic amine or NR9(CH2)nNRι0Rn, wherein n is an integer from 2 to 6, inclusive, and R , Rι0, and Rn are each independently a hydrogen atom or an alkyl group; Y is an aryl group or is NRj2 where Rι2 is a hydrogen atom or an aryl or alkyl group; L is absent or is a C2-C6 alkyl group; R3 and t are each independently a hydrogen atom or an alkyl group, wherein R3 and Rt optionally are combined to form a heterocycle; and R5, Re, Rη, and R8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group, and pharmaceutically acceptable hydrates and salts thereof.
117. The quinazoline composition of claim 116, wherein X is absent or is an aryl group; NR]R2 is a substituted or unsubstituted piperazino or morpholino group or is NR9(CH2)nNRιoRn, wherein n is an integer from 2 to 6, inclusive, R9 is a hydrogen atom, and Rio and Rn are each independently an alkyl group; Y is NH; L is a C2-C6 alkyl group; R3 and Rt are each independently a hydrogen atom or an alkyl group, wherein R and Rt optionally are combined to form a heterocycle; and R5, Re, R7, and R8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group, and pharmaceutically acceptable hydrates and salts thereof.
118. The quinazoline composition of claim 117, wherein X is an aryl group; NRιR2 is substituted or unsubstituted piperazino or morpholino group; Y is NH; L is a C2-C6 alkyl group; R3 and R4 are each independently a methyl or ethyl group or R3 and R4 optionally are combined to form a morpholino group; and R5, R , R , and R8 are each independently a hydrogen atom, a halogen atom, or an alkoxy group, and pharmaceutically acceptable hydrates and salts thereof.
119. The quinazoline composition of claim 118, wherein X is a phenyl group; NRιR2 is N-methylpiperazine; Y is NH; L is -CH2CH2-; R3 and Rt are each a methyl group; and R5, Re, R7, and R8 are each a hydrogen atom, and pharmaceutically acceptable hydrates and salts thereof.
120. The quinazoline composition of claim 118, wherein X is a phenyl group; NR1R2 is N-methylpiperazine; Y is NH; L is -CH2CH2-; R3 and Rt are combined as a morpholino group; and R5, R_, R , and Rg are each a hydrogen atom, and pharmaceutically acceptable hydrates and salts thereof.
121. The quinazoline composition of claim 117, wherein X is absent; NR]R2 is a substituted or unsubstituted piperazino or morpholino group; Y is NH; L is a C2-C6 alkyl group; R3 and Rt are each independently a methyl or ethyl group or R3 and t optionally are combined to form a morpholino group; and Rs, Re, R7, and Rg are each independently a hydrogen atom, a halogen atom, or an alkoxy group, and pharmaceutically acceptable hydrates and salts thereof.
122. The quinazoline composition of claim 121, wherein X is absent; NRιR2 is N-methylpiperazine; Y is NH; L is -CH2CH2-; R and R4 are each a methyl group; Re and R are each a methoxy group; and R5 and Rg are each a hydrogen atom, and pharmaceutically acceptable hydrates and salts thereof.
123. The quinazoline composition of claim 121, wherein X is absent; NRιR2 is N-phenylpiperazine; Y is NH; L is -CH2CH2; R3 and Rt are each a methyl group; Re and R7 are each a methoxy group; and R5 and Rg are each a hydrogen atom, and pharmaceutically acceptable hydrates and salts thereof.
124. The quinazoline composition of claim 121, wherein X is absent; NR1R2 is N-methylpiperazine; Y is NH; L is -CH2CH2; R3 and t are combined as a morpholino group; Re and R7 are each a methoxy group; and R5 and Rg are each a hydrogen atom, and pharmaceutically acceptable hydrates and salts thereof.
125. A method of inhibiting signaling by a Toll-like receptor (TLR), comprising contacting a cell expressing a functional TLR with an effective amount of quinazoline composition having a structural formula
Figure imgf000179_0001
Formula XIX wherein X is a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group, optionally attached to the quinazoline by a nitrogen, oxygen, or sulfur atom or by a SO or SO2 group; Y is absent or is an oxygen atom, a sulfur atom, CR90, or NRπ, where R9, Rio, and Rn are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R , Rio, and Ri 1 optionally is combined with R3 or t to form a heterocycle; L is absent or is a hydrogen atom, an alkyl or alkenyl group containing from 1 to 10 carbons, or an aryl group; R3 and R» are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R3 and t optionally are combined to form a heterocycle; and R5, Re, Rη, and Rg are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein any pair of R5, Re, R7, and R8 which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof, to inhibit signaling by the TLR.
126. The method of claim 125, wherein the quinazoline composition is a quinazoline composition of any one of claims 113-124.
127. The method of claim 125, wherein the TLR is Toll-like receptor 9 (TLR9).
128. A method of inhibiting signaling by a Toll-like receptor (TLR), comprising contacting an immune cell expressing a functional TLR with (a) an effective amount of a TLR signal agonist to stimulate signaling by the TLR in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having a structural formula
Figure imgf000180_0001
Formula XIX wherein X is a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group, optionally attached to the quinazoline by a nitrogen, oxygen, or sulfur atom or by a SO or SO2 group; Y is absent or is an oxygen atom, a sulfur atom, CR90, or NRn, where R9, R10, and Rn are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R9, Rio, and Rn optionally is combined with R3 or t to form a heterocycle; L is absent or is a hydrogen atom, an alkyl or alkenyl group containing from 1 to 10 carbons, or an aryl group; R3 and t are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R3 and Rt optionally are combined to form a heterocycle; and R5, R_, R7, and R8 are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein any pair of R5, R6, R7, and R8 which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof, to inhibit signaling by the TLR in response to the TLR signal agonist compared with the signaling by the TLR in response to the TLR signal agonist in absence of the quinazoline composition.
129. The method of claim 128, wherein the quinazoline composition is a quinazoline composition of any one of claims 113-124.
130. The method of claim 128, wherein the TLR signal agonist is CpG DNA.
131. The method of claim 128, wherein the TLR signal agonist is an immune complex comprising a nucleic acid.
132. The method of claim 128, wherein the TLR is Toll-like receptor 9 (TLR9).
133. A method of inhibiting an immune response to an antigenic substance, comprising contacting an immune cell expressing a functional Toll-like receptor with (a) an effective amount of an antigenic substance to stimulate an immune response to the antigenic substance in absence of a quinazoline composition, and (b) an effective amount of a quinazoline composition having a structural formula
Figure imgf000181_0001
Formula XIX wherein X is a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group, optionally attached to the quinazoline by a nitrogen, oxygen, or sulfur atom or by a SO or SO2 group; Y is absent or is an oxygen atom, a sulfur atom, CR90, or NRi i, where R9, R]0, and Ri i are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R , Rio, and Rn optionally is combined with R3 or t to form a heterocycle; L is absent or is a hydrogen atom, an alkyl or alkenyl group containing from 1 to 10 carbons, or an aryl group; R and R4 are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R3 and * optionally are combined to form a heterocycle; and R5, R_, R7, and Rg are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein any pair of R5, R_, R7, and Rg which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof, to inhibit an immune response to the antigenic substance compared with the immune response to the antigenic substance in absence of the quinazoline composition.
134. The method of claim 133, wherein the quinazoline composition is a quinazoline composition of any one of claims 113-124.
135. The method of claim 133, wherein the contacting takes place in vivo.
136. The method of claim 133, wherein the immune response comprises an adaptive immune response.
137. The method of claim 133, wherein the immune response is an innate immune response.
138. A method of treating an autoimmune disorder in a subject, comprising administering to a subject having an autoimmune disorder an effective amount of a quinazoline composition having a structural formula
Figure imgf000183_0001
Formula XIX wherein X is a substituted or unsubstituted aryl, alkyl, heterocyclic or styryl group, optionally attached to the quinazoline by a nitrogen, oxygen, or sulfur atom or by a SO or SO2 group; Y is absent or is an oxygen atom, a sulfur atom, CR9Rιo, or NRπ, where R9, Rι0, and Ri 1 are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein any one of R9, Rio, and Rn optionally is combined with R3 or R4 to form a heterocycle; L is absent or is a hydrogen atom, an alkyl or alkenyl group containing from 1 to 10 carbons, or an aryl group; R3 and R4 are each independently a hydrogen atom or an alkyl, alkenyl, or aryl group, wherein R3 and Rt optionally are combined to form a heterocycle; and R5, Re, R7, and R8 are each independently a hydrogen atom, a halogen atom, or an alkyl, alkenyl, aryl, heterocyclic, nitro, cyano, carboxy, ester, ketone, amino, amido, hydroxy, alkoxy, mercapto, thio, sulfoxide, sulfone, or sulfonamido group, wherein any pair of R5, Re, Rη, and Rg which are adjacent one another optionally are combined to form a heterocycle or a carbocycle, and pharmaceutically acceptable hydrates and salts thereof, to treat the autoimmune disorder.
139. The method of claim 138, wherein the quinazoline composition is a quinazoline composition of any one of claims 113-124.
140. The method of claim 138, wherein the autoimmune disorder is chosen from systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Sjogren's syndrome, polymyositis, vasculitis, Wegener' s granulomatosis, sarcoidosis, ankylosing spondylitis, Reiter's syndrome, psoriatic arthritis, and Behcet's syndrome.
141. The method of claim 138, wherein the autoimmune disorder is an immune complex associated disease.
142. The method of claim 138, wherein the subj ect is a human.
PCT/US2004/019714 2003-06-20 2004-06-18 Small molecule toll-like receptor (tlr) antagonists WO2005007672A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
EA200600069A EA200600069A1 (en) 2003-06-20 2004-06-18 LOW-MOLECULAR ANTAGONISTS OF TOLL-SIMILAR RECEPTORS (TLR)
AU2004257149A AU2004257149A1 (en) 2003-06-20 2004-06-18 Small molecule toll-like receptor (TLR) antagonists
CA002528774A CA2528774A1 (en) 2003-06-20 2004-06-18 Small molecule toll-like receptor (tlr) antagonists
BRPI0411514-7A BRPI0411514A (en) 2003-06-20 2004-06-18 small molecule toll-like receptor antagonists
JP2006517471A JP2007524615A (en) 2003-06-20 2004-06-18 Low molecular weight Toll-like receptor (TLR) antagonist
CN200480017064.7A CN1809357B (en) 2003-06-20 2004-06-18 Small molecule Toll-like receptor (TLR) antagonists
EP04776820A EP1635846A4 (en) 2003-06-20 2004-06-18 Small molecule toll-like receptor (tlr) antagonists
MXPA05013922A MXPA05013922A (en) 2003-06-20 2004-06-18 Small molecule toll-like receptor (tlr) antagonists.
IL172580A IL172580A0 (en) 2003-06-20 2005-12-14 Small molecule toll-like receptor (tlr) antagonists

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US48058803P 2003-06-20 2003-06-20
US60/480,588 2003-06-20
US55600704P 2004-03-23 2004-03-23
US60/556,007 2004-03-23

Publications (3)

Publication Number Publication Date
WO2005007672A2 true WO2005007672A2 (en) 2005-01-27
WO2005007672A3 WO2005007672A3 (en) 2005-09-15
WO2005007672A8 WO2005007672A8 (en) 2007-11-01

Family

ID=34083269

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/019714 WO2005007672A2 (en) 2003-06-20 2004-06-18 Small molecule toll-like receptor (tlr) antagonists

Country Status (11)

Country Link
US (2) US7410975B2 (en)
EP (1) EP1635846A4 (en)
JP (1) JP2007524615A (en)
KR (1) KR20060016817A (en)
AU (1) AU2004257149A1 (en)
BR (1) BRPI0411514A (en)
CA (1) CA2528774A1 (en)
EA (1) EA200600069A1 (en)
IL (1) IL172580A0 (en)
MX (1) MXPA05013922A (en)
WO (1) WO2005007672A2 (en)

Cited By (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006035066A2 (en) * 2004-09-29 2006-04-06 Institut National De La Sante Et De La Recherche Medicale (Inserm) Toll-like receptor 3, its signalling associated molecule trif and their use in the prevention and treatment of host inflammation response induced by a rna virus
EP1655027A1 (en) * 2004-11-04 2006-05-10 Biofrontera Pharmaceuticals GmbH 9-(N-methyl-piperidyliden-4)-thioxanthene for treatment of pulmonary hypertension and migraine
WO2007038720A2 (en) * 2005-09-27 2007-04-05 Coley Pharmaceutical Gmbh Modulation of tlr-mediated immune responses using adaptor oligonucleotides
WO2006113703A3 (en) * 2005-04-18 2007-06-28 Ptc Therapeutics Inc Carboline derivatives useful in the treatment of cancer
WO2007078335A2 (en) * 2005-12-21 2007-07-12 Decode Genetics, Ehf. Biaryl nitrogen heterocycle inhibitors of lta4h for treating inflammation
WO2007127624A1 (en) * 2006-04-26 2007-11-08 The Uab Research Foundation Reducing cancer cell invasion using an inhibitor of toll like receptor signaling
EP1858326A1 (en) * 2005-02-22 2007-11-28 The Regents of the University of California Methods of treating gastrointestinal inflammation
WO2007047396A3 (en) * 2005-10-12 2007-12-21 Idera Pharmaceuticals Inc Immune regulatory oligonucleotide (iro) compounds to modulate toll-like receptor based immune response
GB2447791A (en) * 2007-03-23 2008-09-24 Univ Dundee Pharmaceutical formulations and compounds for use in alleviating conditions related to Down's syndrome
WO2008152471A1 (en) * 2007-06-12 2008-12-18 Coley Pharmaceutical Group, Inc. Quinazoline derivative useful as toll-like receptor antagonist
WO2008155468A1 (en) * 2007-06-20 2008-12-24 Marikki Laiho Activators and therapeutic applications thereof
JP2009504763A (en) * 2005-08-19 2009-02-05 アレイ バイオファーマ、インコーポレイテッド 8-Substituted Benzazepines as Toll-like Receptor Modulators
WO2009047791A2 (en) * 2007-09-24 2009-04-16 Reliance Life Sciences Pvt. Ltd. Toll like receptor (tlr) signaling antagonist
US7601840B2 (en) 2004-03-15 2009-10-13 Ptc Therapeutics, Inc. Carboline derivatives useful in the inhibition of angiogenesis
WO2010009845A1 (en) * 2008-07-24 2010-01-28 Bayer Schering Pharma Aktiengesellschaft Sulfone substituted quinazoline derivatives as immune-modulators for the treatment of inflammatory and allergic diseases
WO2010024717A1 (en) * 2008-08-22 2010-03-04 Алла Xem, Ллс Ligand with a broad spectrum of pharmacological activity, a pharmaceutical composition, a medicinal agent and a method of treatment
EP2203057A1 (en) * 2007-09-20 2010-07-07 University Of Rochester Method and compositions for treatment or prevention of inflammatory conditions
US20100190761A1 (en) * 2007-06-20 2010-07-29 Anthony Ogawa Diphenyl substituted alkanes
WO2010002835A3 (en) * 2008-07-03 2011-04-21 Osteogenex Inc. Vinpocetine and eburnamonine derivatives for promoting bone growth
US8076353B2 (en) 2004-03-15 2011-12-13 Ptc Therapeutics, Inc. Inhibition of VEGF translation
US8076352B2 (en) 2004-03-15 2011-12-13 Ptc Therapeutics, Inc. Administration of carboline derivatives useful in the treatment of cancer and other diseases
WO2012156498A1 (en) * 2011-05-18 2012-11-22 Janssen R&D Ireland Quinazoline derivatives for the treatment of viral infections and further diseases
US8377898B2 (en) 2006-10-12 2013-02-19 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
US8383598B2 (en) 2005-10-12 2013-02-26 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
US8399423B2 (en) 2005-10-12 2013-03-19 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
US8404669B2 (en) 2007-03-16 2013-03-26 University Of Florida Research Foundation, Inc. Kinase modulating compounds and uses thereof for treatment of cancer
US8426375B2 (en) 2005-10-12 2013-04-23 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
US8440672B2 (en) 2006-09-01 2013-05-14 Merck Sharp & Dohme Corp. Diphenyl substituted alkanes
US8575184B2 (en) 2009-09-03 2013-11-05 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
US8592383B2 (en) 2011-06-17 2013-11-26 Merck Sharp & Dohme Corp. Cycloalkyl-fused tetrahydroquinolines as CRTH2 receptor modulators
US8686002B2 (en) 2005-08-21 2014-04-01 AbbVie Deutschland GmbH & Co. KG Heterocyclic compounds and their use as binding partners for 5-HT5 receptors
US8697662B2 (en) 2009-05-27 2014-04-15 Ptc Therapeutics, Inc. Methods for treating Kaposi sarcoma
US8703726B2 (en) 2009-05-27 2014-04-22 Ptc Therapeutics, Inc. Methods for treating prostate conditions
EP2769723A1 (en) * 2013-02-22 2014-08-27 Ruprecht-Karls-Universität Heidelberg Compounds for use in inhibiting HIV capsid assembly
US8853177B2 (en) 2008-10-06 2014-10-07 Idera Pharmaceuticals, Inc. Use of inhibitors of toll-like receptors in the prevention and treatment of hypercholesterolemia and hyperlipidemia and diseases related thereto
WO2015057659A1 (en) * 2013-10-14 2015-04-23 Eisai R&D Management Co., Ltd. Selectively substituted quinoline compounds
WO2015088045A1 (en) * 2013-12-13 2015-06-18 Takeda Pharmaceutical Company Limited Pyrrolo[3,2-c]pyridine derivatives as tlr inhibitors
US9351964B2 (en) 2009-05-27 2016-05-31 Ptc Therapeutics, Inc. Methods for treating cancer and non-neoplastic conditions
EP3072891A1 (en) 2015-03-24 2016-09-28 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. New Toll-Like Receptor 9 Antagonists
US9663486B2 (en) 2013-10-14 2017-05-30 Eisai R&D Management Co., Ltd. Selectively substituted quinoline compounds
WO2017163264A1 (en) * 2016-03-21 2017-09-28 Council Of Scientific & Industrial Research Blocking toll-like receptor 9 signaling with small molecule antagonist
WO2018146171A1 (en) 2017-02-09 2018-08-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for treating mast cell activation diseases
WO2019020732A1 (en) 2017-07-27 2019-01-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for determining whether a patient suffering from rhabdomyolysis achieves a response with a tlr9 antagonist
US10253003B2 (en) 2012-11-16 2019-04-09 Janssen Sciences Ireland Uc Heterocyclic substituted 2-amino quinazoline derivatives for the treatment of viral infections
US10259793B2 (en) 2013-02-21 2019-04-16 Janssen Sciences Ireland Uc 2-aminopyrimidine derivatives for the treatment of viral infections
US10259814B2 (en) 2012-10-10 2019-04-16 Janssen Sciences Ireland Uc Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
US10266543B2 (en) 2013-03-29 2019-04-23 Janssen Sciences Ireland Uc Macrocyclic deaza-purinones for the treatment of viral infections
US10272085B2 (en) 2011-04-08 2019-04-30 Janssen Sciences Ireland Uc Pyrimidine derivatives for the treatment of viral infections
US10280167B2 (en) 2011-11-09 2019-05-07 Janssen Sciences Ireland Uc Purine derivatives for the treatment of viral infections
US10280180B2 (en) 2012-07-13 2019-05-07 Janssen Sciences Ireland Uc Macrocyclic purines for the treatment of viral infections
US10316043B2 (en) 2013-07-30 2019-06-11 Janssen Sciences Ireland Unlimited Company Thieno[3,2-d]pyrimidines derivatives for the treatment of viral infections
US10377738B2 (en) 2013-05-24 2019-08-13 Janssen Sciences Ireland Unlimited Company Pyridone derivatives for the treatment of viral infections and further diseases
US10385054B2 (en) 2013-06-27 2019-08-20 Janssen Sciences Ireland Unlimited Company Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
WO2019197682A1 (en) 2018-04-14 2019-10-17 4Sc Ag Pharmaceutical combination products comprising a histone deacetylase (hdac) inhibitor and a tlr7 agonist and/or tlr8 agonist for the treatment of cancer
WO2019207066A1 (en) 2018-04-26 2019-10-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for the treatment of sjögren's syndrome
US10947231B2 (en) 2009-05-27 2021-03-16 Ptc Therapeutics, Inc. Processes for the preparation of substituted tetrahydro beta-carbolines
US10968184B2 (en) 2016-09-29 2021-04-06 Janssen Sciences Ireland Unlimited Company Pyrimidine prodrugs for the treatment of viral infections and further diseases
US11053256B2 (en) 2016-07-01 2021-07-06 Janssen Sciences Ireland Unlimited Company Dihydropyranopyrimidines for the treatment of viral infections
WO2022060805A1 (en) * 2020-09-15 2022-03-24 The Johns Hopkins University Compounds which inhibit rna polymerase
US11458126B2 (en) 2017-08-01 2022-10-04 Ptc Therapeutics, Inc. DHODH inhibitor for use in treating hematologic cancers
EP4119145A1 (en) * 2021-07-15 2023-01-18 Dompe' Farmaceutici S.P.A. Compounds for the treatment of covid-19
US11597704B2 (en) 2018-03-01 2023-03-07 Janssen Sciences Ireland Unlimited Company 2,4-diaminoquinazoline derivatives and medical uses thereof

Families Citing this family (166)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6429199B1 (en) * 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
US6239116B1 (en) * 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US20030026782A1 (en) * 1995-02-07 2003-02-06 Arthur M. Krieg Immunomodulatory oligonucleotides
US7935675B1 (en) * 1994-07-15 2011-05-03 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
EP0855184A1 (en) * 1997-01-23 1998-07-29 Grayson B. Dr. Lipford Pharmaceutical composition comprising a polynucleotide and an antigen especially for vaccination
US6406705B1 (en) 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
ATE356630T1 (en) * 1998-04-03 2007-04-15 Univ Iowa Res Found METHOD AND PRODUCTS FOR STIMULATING THE IMMUNE SYSTEM USING IMMUNOTHERAPEUTIC OLIGONUCLEOTIDES AND CYTOKINE
EP1674574A1 (en) * 1998-05-14 2006-06-28 Coley Pharmaceutical GmbH Methods for Regulating Hematopoiesis using CpG-Oligonucleotides
SI1077722T1 (en) 1998-05-22 2007-02-28 Ottawa Health Research Inst Methods and products for inducing mucosal immunity
US20030022854A1 (en) 1998-06-25 2003-01-30 Dow Steven W. Vaccines using nucleic acid-lipid complexes
US7776343B1 (en) * 1999-02-17 2010-08-17 Csl Limited Immunogenic complexes and methods relating thereto
EE200200158A (en) * 1999-09-25 2003-06-16 University Of Iowa Research Foundation Immunostimulatory nucleic acids
US6949520B1 (en) * 1999-09-27 2005-09-27 Coley Pharmaceutical Group, Inc. Methods related to immunostimulatory nucleic acid-induced interferon
EP1239876B1 (en) * 1999-11-19 2008-07-30 Csl Limited Hcv vaccine compositions
US7585847B2 (en) * 2000-02-03 2009-09-08 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids for the treatment of asthma and allergy
US20040131628A1 (en) * 2000-03-08 2004-07-08 Bratzler Robert L. Nucleic acids for the treatment of disorders associated with microorganisms
EP1296714B1 (en) * 2000-06-22 2009-08-26 University Of Iowa Research Foundation Combination of CpG and antibodies directed against CD19,CD20, CD22 or CD40 for the treatment or prevention of cancer.
ATE510850T1 (en) * 2000-09-15 2011-06-15 Coley Pharm Gmbh METHOD FOR HIGH-THROUGHPUT SCREENING OF CPG-BASED IMMUNAGONISTS AND ANTAGONISTS
DE60229422D1 (en) * 2001-08-17 2008-11-27 Coley Pharm Gmbh COMBINATION MOTIF IMMUNOSTIMULATING OLIGONUCLEOTIDES WITH IMPROVED EFFECT
AU2002360278A1 (en) * 2001-10-12 2003-11-11 Coley Pharmaceutical Gmbh Methods and products for enhancing immune responses using imidazoquinoline compounds
EP1499187B1 (en) 2002-04-04 2015-06-17 Zoetis Belgium S.A. Immunostimulatory g,u-containing oligoribonucleotides
US7576066B2 (en) * 2002-07-03 2009-08-18 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US7605138B2 (en) 2002-07-03 2009-10-20 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US7807803B2 (en) * 2002-07-03 2010-10-05 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US20040053880A1 (en) 2002-07-03 2004-03-18 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US7569553B2 (en) 2002-07-03 2009-08-04 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
AR040996A1 (en) 2002-08-19 2005-04-27 Coley Pharm Group Inc IMMUNE STIMULATING NUCLEIC ACIDS
EA008777B1 (en) * 2002-10-29 2007-08-31 Коли Фармасьютикал Груп, Лтд. Use of cpg oligonucleotides in the treatment of hepatitis c virus infection
CA2502015A1 (en) 2002-12-11 2004-06-24 Coley Pharmaceutical Group, Inc. 5' cpg nucleic acids and methods of use
WO2005007672A2 (en) * 2003-06-20 2005-01-27 Coley Pharmaceutical Gmbh Small molecule toll-like receptor (tlr) antagonists
EP1666059A4 (en) * 2003-08-11 2008-08-27 Univ Osaka Res Found Novel vaccine containing adjuvant capable of inducing mucosal immunity
JP2007502293A (en) 2003-08-12 2007-02-08 スリーエム イノベイティブ プロパティズ カンパニー Hydroxylamine-substituted imidazo-containing compounds
RU2006105101A (en) 2003-08-27 2007-10-10 3М Инновейтив Пропертиз Компани (US) Aryloxy and arylalkylene-substituted substituted imidazoquinolines
CA2537763A1 (en) * 2003-09-05 2005-03-17 3M Innovative Properties Company Treatment for cd5+ b cell lymphoma
JP4989225B2 (en) * 2003-09-25 2012-08-01 コーリー ファーマシューティカル グループ,インコーポレイテッド Nucleic acid lipophilic conjugate
AU2004278014B2 (en) 2003-10-03 2011-04-28 3M Innovative Properties Company Alkoxy substituted imidazoquinolines
US7544697B2 (en) 2003-10-03 2009-06-09 Coley Pharmaceutical Group, Inc. Pyrazolopyridines and analogs thereof
OA13278A (en) 2003-10-30 2007-01-31 Coley Pharm Gmbh C-Class oligonucleotide analogs with enhanced immunostimulatory potency.
US20050239733A1 (en) * 2003-10-31 2005-10-27 Coley Pharmaceutical Gmbh Sequence requirements for inhibitory oligonucleotides
US20050100983A1 (en) * 2003-11-06 2005-05-12 Coley Pharmaceutical Gmbh Cell-free methods for identifying compounds that affect toll-like receptor 9 (TLR9) signaling
JP2007511527A (en) 2003-11-14 2007-05-10 スリーエム イノベイティブ プロパティズ カンパニー Oxime-substituted imidazo ring compounds
US8598192B2 (en) 2003-11-14 2013-12-03 3M Innovative Properties Company Hydroxylamine substituted imidazoquinolines
WO2005051317A2 (en) 2003-11-25 2005-06-09 3M Innovative Properties Company Substituted imidazo ring systems and methods
WO2005066170A1 (en) 2003-12-29 2005-07-21 3M Innovative Properties Company Arylalkenyl and arylalkynyl substituted imidazoquinolines
CA2551399A1 (en) 2003-12-30 2005-07-21 3M Innovative Properties Company Imidazoquinolinyl, imidazopyridinyl, and imidazonaphthyridinyl sulfonamides
TW200533750A (en) * 2004-02-19 2005-10-16 Coley Pharm Group Inc Immunostimulatory viral RNA oligonucleotides
US8697873B2 (en) 2004-03-24 2014-04-15 3M Innovative Properties Company Amide substituted imidazopyridines, imidazoquinolines, and imidazonaphthyridines
TWI392678B (en) * 2004-03-26 2013-04-11 Dainippon Sumitomo Pharma Co 9-substituted-8-oxoadenine compound
CA2567789A1 (en) * 2004-06-08 2006-08-03 Coley Pharmaceutical Gmbh Abasic oligonucleotide as carrier platform for antigen and immunostimulatory agonist and antagonist
US8017779B2 (en) 2004-06-15 2011-09-13 3M Innovative Properties Company Nitrogen containing heterocyclyl substituted imidazoquinolines and imidazonaphthyridines
US7897609B2 (en) 2004-06-18 2011-03-01 3M Innovative Properties Company Aryl substituted imidazonaphthyridines
US8026366B2 (en) 2004-06-18 2011-09-27 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
US7915281B2 (en) 2004-06-18 2011-03-29 3M Innovative Properties Company Isoxazole, dihydroisoxazole, and oxadiazole substituted imidazo ring compounds and method
KR100958505B1 (en) * 2004-07-18 2010-05-17 씨에스엘 리미티드 Immuno stimulating complex and oligonucleotide formulations for inducing enhanced interferon-gamma responses
MY159370A (en) * 2004-10-20 2016-12-30 Coley Pharm Group Inc Semi-soft-class immunostimulatory oligonucleotides
AU2005306533B2 (en) * 2004-11-17 2012-05-31 Arbutus Biopharma Corporation siRNA silencing of apolipoprotein B
US20120189643A1 (en) * 2004-11-30 2012-07-26 Carton Jill M Toll Like Receptor 3 Antagonists, Methods and Uses
AR051836A1 (en) * 2004-11-30 2007-02-14 Centocor Inc RECEIVER ANTAGONISTS 3 SIMIL TOLL METHODS AND USES
PT1830876E (en) * 2004-12-30 2015-07-13 Meda Ab Use of imiquimod for the treatment of cutaneous metastases derived from a breast cancer tumor
JP5543068B2 (en) 2004-12-30 2014-07-09 スリーエム イノベイティブ プロパティズ カンパニー Chiral fused [1,2] imidazo [4,5-c] cyclic compound
JP5313502B2 (en) 2004-12-30 2013-10-09 スリーエム イノベイティブ プロパティズ カンパニー Substituted chiral condensed [1,2] imidazo [4,5-c] cyclic compounds
JP2008530022A (en) 2005-02-04 2008-08-07 コーリー ファーマシューティカル グループ,インコーポレイテッド Aqueous gel formulation containing immune response modifier
AU2006213746A1 (en) 2005-02-11 2006-08-17 Coley Pharmaceutical Group, Inc. Oxime and hydroxylamine substituted imidazo(4,5-c) ring compounds and methods
AU2006232375A1 (en) 2005-04-01 2006-10-12 Coley Pharmaceutical Group, Inc. 1-substituted pyrazolo (3,4-c) ring compounds as modulators of cytokine biosynthesis for the treatment of viral infections and neoplastic diseases
WO2006107853A2 (en) 2005-04-01 2006-10-12 Coley Pharmaceutical Group, Inc. Pyrazolopyridine-1,4-diamines and analogs thereof
EP1874325A2 (en) * 2005-04-08 2008-01-09 Coley Pharmaceutical Group, Inc. Methods for treating infectious disease exacerbated asthma
WO2006116458A2 (en) * 2005-04-26 2006-11-02 Coley Pharmaceutical Gmbh Modified oligoribonucleotide analogs with enhances immunostimulatory activity
AU2006269555A1 (en) * 2005-07-07 2007-01-18 Coley Pharmaceutical Group, Inc. Anti-CTLA-4 antibody and CpG-motif-containing synthetic oligodeoxynucleotide combination therapy for cancer treatment
US20070054873A1 (en) * 2005-08-26 2007-03-08 Protiva Biotherapeutics, Inc. Glucocorticoid modulation of nucleic acid-mediated immune stimulation
CN101321868A (en) * 2005-09-16 2008-12-10 科利制药公司 Modulation of immunostimulatory properties of short interfering ribonucleic acid (siRNA) by nucleotide modification
AU2006308765B2 (en) 2005-11-02 2013-09-05 Arbutus Biopharma Corporation Modified siRNA molecules and uses thereof
ES2536103T3 (en) 2005-11-25 2015-05-20 Zoetis Belgium S.A. Immunostimulatory oligonucleotides
JPWO2007123186A1 (en) * 2006-04-20 2009-09-03 武田薬品工業株式会社 Medicine
US8957035B2 (en) * 2006-05-15 2015-02-17 University Of Kentucky Research Foundation Toll like receptor (TLR) stimulation for ocular angiogenesis and macular degeneration
US7915399B2 (en) * 2006-06-09 2011-03-29 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
JP2009542645A (en) * 2006-07-05 2009-12-03 アストラゼネカ・アクチエボラーグ 8-oxoadenine derivatives acting as modulators of TLR7
WO2008008432A2 (en) 2006-07-12 2008-01-17 Coley Pharmaceutical Group, Inc. Substituted chiral fused( 1,2) imidazo (4,5-c) ring compounds and methods
AU2007284036A1 (en) * 2006-08-16 2008-02-21 Protiva Biotherapeutics, Inc. Nucleic acid modulation of Toll-like receptor-mediated immune stimulation
NZ575437A (en) 2006-09-27 2012-02-24 Coley Pharm Gmbh Cpg oligonucleotide analogs containing hydrophobic t analogs with enhanced immunostimulatory activity
US20090142362A1 (en) * 2006-11-06 2009-06-04 Avant Immunotherapeutics, Inc. Peptide-based vaccine compositions to endogenous cholesteryl ester transfer protein (CETP)
TW200831105A (en) * 2006-12-14 2008-08-01 Astrazeneca Ab Novel compounds
US7629135B2 (en) * 2006-12-22 2009-12-08 The Board Of Trustees Of The University Of Illinois Toll-like receptor agonists and antagonists and methods of use thereof
US8067413B2 (en) * 2007-03-19 2011-11-29 Astrazeneca Ab 9-substituted-8-oxo-adenine compounds as toll-like receptor (TLR7 ) modulators
WO2008114006A1 (en) * 2007-03-19 2008-09-25 Astrazeneca Ab 9-substituted-8-oxo-adenine compounds as toll-like receptor (tlr7) modulators
WO2008114819A1 (en) * 2007-03-20 2008-09-25 Dainippon Sumitomo Pharma Co., Ltd. Novel adenine compound
US8436178B2 (en) * 2007-05-08 2013-05-07 Astrazeneca Ab Imidazoquinolines with immuno-modulating properties
RU2010112771A (en) * 2007-10-09 2011-11-20 Коули Фармасьютикал ГмбХ (DE) IMMUNITY MULATING ANALOGUES OF OLIGONUCLEOTIDES CONTAINING MODIFIED SUGAR GROUPS
US20100331545A1 (en) 2007-10-24 2010-12-30 Nippon Chemiphar Co., Ltd. Regulator for signaling toll-like receptor, which comprises cathepsin inhibitor as active ingredient
WO2009059050A2 (en) * 2007-10-30 2009-05-07 The Regents Of The University Of Colorado Tlr modulators and methods for using the same
US20110251229A1 (en) * 2007-10-30 2011-10-13 The Regents Of The University Of Colorado (+)-opioids and methods of use
PE20091236A1 (en) * 2007-11-22 2009-09-16 Astrazeneca Ab PYRIMIDINE DERIVATIVES AS IMMUNOMODULATORS OF TLR7
PE20091156A1 (en) 2007-12-17 2009-09-03 Astrazeneca Ab SALTS OF (3 - {[[3- (6-AMINO-2-BUTOXY-8-OXO-7,8-DIHIDRO-9H-PURIN-9-IL) PROPYL] (3-MORFOLIN-4-ILPROPIL) AMINO] METHYL} PHENYL) METHYL ACETATE
WO2009105260A2 (en) * 2008-02-21 2009-08-27 University Of Kentucky Ultra-small rnas as toll-like receptor-3 antagonists
US8772243B2 (en) * 2008-08-20 2014-07-08 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods for predicting the response to anti-cancer treatment with an agonist of TLR7 or an agonist of TLR8
AU2009308763B2 (en) 2008-10-31 2017-03-09 Janssen Biotech, Inc. Toll-like receptor 3 antagonists
US8460659B2 (en) 2008-10-31 2013-06-11 Janssen Biotech, Inc. Toll-like receptor 3 antagonists for the treatment of metabolic and cardiovascular diseases
US20100136095A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems for modulating inflammation
US20100136096A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems for modulating inflammation
US20100137843A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Delivery devices for modulating inflammation
US20100136094A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems for modulating inflammation
US20100135984A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Anti-inflammatory compositions and methods
US20100135983A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Anti-inflammatory compositions and methods
US20100136097A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems for modulating inflammation
JP5645840B2 (en) 2008-12-02 2014-12-24 株式会社Wave Life Sciences Japan Method for synthesizing phosphorous atom-modified nucleic acid
US20100137246A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Anti-inflammatory compositions and methods
US8883174B2 (en) 2009-03-25 2014-11-11 The Board Of Regents, The University Of Texas System Compositions for stimulation of mammalian innate immune resistance to pathogens
US8367316B2 (en) * 2009-05-01 2013-02-05 Alfagene Bioscience, Inc. Human gastrointestinal stem cell-derived primary intestinal epithelial cell system and methods of use thereof
GB0908772D0 (en) * 2009-05-21 2009-07-01 Astrazeneca Ab New salts 756
SG175796A1 (en) 2009-05-21 2011-12-29 Astrazeneca Ab Novel pyrimidine derivatives and their use in the treatment of cancer and further diseases
CN102596204B (en) 2009-07-06 2016-11-23 波涛生命科学有限公司 New nucleic acid prodrugs and using method thereof
WO2011011594A2 (en) * 2009-07-23 2011-01-27 Shriners Hospitals For Children Intracellular toll-like receptors pathways and axonal degeneration
EP2483407A2 (en) 2009-09-30 2012-08-08 President and Fellows of Harvard College Methods for modulation of autophagy through the modulation of autophagy-enhancing gene products
EP2507237A1 (en) * 2009-12-03 2012-10-10 Dainippon Sumitomo Pharma Co., Ltd. Imidazoquinolines which act via toll - like receptors (tlr)
JP2013147428A (en) * 2010-04-27 2013-08-01 Dainippon Sumitomo Pharma Co Ltd Novel 2-heteroaryl monocyclic pyrimidine derivative
JP2013166700A (en) * 2010-06-02 2013-08-29 Dainippon Sumitomo Pharma Co Ltd Novel 4, 5-fused pyrimidine derivative
AU2011281192A1 (en) 2010-07-19 2013-02-28 Yeda Research And Development Co. Ltd. Peptides based on the transmembrane domain of a Toll-like receptor (TLR) for treatment of TLR-mediated diseases
WO2012024433A2 (en) * 2010-08-17 2012-02-23 Translational Genomics Research Institute Compounds that inhibit tau phosphorylation
EP2620428B1 (en) 2010-09-24 2019-05-22 Wave Life Sciences Ltd. Asymmetric auxiliary group
JP5978225B2 (en) 2010-12-16 2016-08-24 大日本住友製薬株式会社 Imidazo [4,5-c] quinolin-1-yl derivatives useful for therapy
EP2651943B1 (en) 2010-12-17 2017-03-22 Sumitomo Dainippon Pharma Co., Ltd. Purine derivatives
KR20130142164A (en) * 2011-01-11 2013-12-27 우니베르시태트 바젤 Combination of syrosingopine and mitochondrial inhibitors for the treatment of cancer and immunosuppression
JP6128529B2 (en) 2011-07-19 2017-05-17 ウェイブ ライフ サイエンシズ リミテッドWave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
IN2014KN00948A (en) 2011-10-04 2015-08-21 Janus Biotherapeutics Inc
ES2639941T3 (en) * 2011-12-19 2017-10-30 Becton Dickinson And Company White blood cell labeling with phosphorothioate oligonucleotides
WO2013116590A1 (en) * 2012-02-01 2013-08-08 George Miller Inhibition of pattern recognition receptors in pancreatic cancer treatment using tlr inhibitors
AU2013261267B2 (en) 2012-05-18 2017-06-29 Sumitomo Pharma Co., Ltd. Carboxylic acid compounds
AU2013287630B2 (en) * 2012-07-13 2017-05-25 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant
PL2872485T3 (en) 2012-07-13 2021-05-31 Wave Life Sciences Ltd. Asymmetric auxiliary group
WO2014012081A2 (en) 2012-07-13 2014-01-16 Ontorii, Inc. Chiral control
JPWO2014034719A1 (en) * 2012-08-29 2016-08-08 興和株式会社 Quinoline derivative having TLR inhibitory action
US9868955B2 (en) 2012-09-29 2018-01-16 Dynavax Technologies Corporation Human toll-like receptor inhibitors and methods of use thereof
US9228184B2 (en) 2012-09-29 2016-01-05 Dynavax Technologies Corporation Human toll-like receptor inhibitors and methods of use thereof
JP6434487B2 (en) 2013-03-15 2018-12-05 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー Desethylhydroxychloroquine for the treatment of diseases with inflammation
WO2015108047A1 (en) 2014-01-15 2015-07-23 株式会社新日本科学 Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
JPWO2015108048A1 (en) 2014-01-15 2017-03-23 株式会社新日本科学 Chiral nucleic acid adjuvant and antitumor agent having antitumor activity
EP3095460A4 (en) 2014-01-15 2017-08-23 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
US10160969B2 (en) 2014-01-16 2018-12-25 Wave Life Sciences Ltd. Chiral design
WO2016029077A1 (en) * 2014-08-22 2016-02-25 Janus Biotherapeutics, Inc. Novel n2, n4, n7, 6-tetrasubstituted pteridine-2,4,7-triamine and 2, 4, 6, 7-tetrasubstituted pteridine compounds and methods of synthesis and use thereof
WO2016044839A2 (en) 2014-09-19 2016-03-24 The Board Of Regents Of The University Of Texas System Compositions and methods for treating viral infections through stimulated innate immunity in combination with antiviral compounds
US9932350B2 (en) 2014-09-30 2018-04-03 Merck Sharp & Dohme Corp. Inhibitors of IRAK4 activity
WO2016053769A1 (en) * 2014-09-30 2016-04-07 Merck Sharp & Dohme Corp. Inhibitors of irak4 activity
EP3200787B1 (en) 2014-09-30 2019-09-04 Merck Sharp & Dohme Corp. Inhibitors of irak4 activity
US9943516B2 (en) 2014-09-30 2018-04-17 Merck Sharp & Dohme Corp. Inhibitors of IRAK4 activity
US10087148B2 (en) * 2014-12-05 2018-10-02 The United States Of America As Represented By The Secretary, Department Of Health And Human Services Office Of Technology Transfer, National Institute Of Health Quinazolines as biogenic amine transport modulators
KR20170117023A (en) 2014-12-05 2017-10-20 서브라마니암 아난탄 Heterocyclic compounds as biogenic amine transport modulators
US10329258B2 (en) 2015-04-30 2019-06-25 University Of Washington CGAS in systemic lupus erythematosus (SLE)
JP6750177B2 (en) * 2015-12-11 2020-09-02 ロート製薬株式会社 Anthranilamide derivative and therapeutic agent for diseases involving TLR3 containing the same
WO2017173334A1 (en) 2016-04-01 2017-10-05 Checkmate Pharmaceuticals, Inc. Fc receptor-mediated drug delivery
US10071079B2 (en) 2016-06-29 2018-09-11 Bristol-Myers Squibb Company [1,2,4]triazolo[1,5-a]pyridinyl substituted indole compounds
CN109715617B (en) 2016-07-30 2022-04-19 百时美施贵宝公司 Dimethoxyphenyl-substituted indole compounds as inhibitors of TLR7, TLR8 or TLR9
US10660877B2 (en) 2016-09-09 2020-05-26 Bristol-Myers Squibb Company Pyridyl substituted indole compounds
WO2018067302A2 (en) * 2016-09-19 2018-04-12 North Western University Therapeutic effects of cellular delivery of small molecules and macromolecules with liposomal spherical nucleic acids
JP7266576B2 (en) 2017-08-04 2023-04-28 ブリストル-マイヤーズ スクイブ カンパニー [1,2,4]triazolo[4,3-a]pyridinyl-substituted indole compounds
WO2019028302A1 (en) 2017-08-04 2019-02-07 Bristol-Myers Squibb Company Substituted indole compounds useful as inhibitors of tlr7/8/9
CA3080102A1 (en) 2017-11-08 2019-05-16 Council Of Scientific & Industrial Research Purine based compounds as toll-like receptor 9 antagonist
JP7265554B2 (en) 2017-11-14 2023-04-26 ブリストル-マイヤーズ スクイブ カンパニー substituted indole compounds
DK3728252T3 (en) 2017-12-18 2023-11-13 Bristol Myers Squibb Co 4-AZAINDOLE COMPOUNDS
EA202091483A1 (en) 2017-12-19 2020-10-28 Бристол-Маерс Сквибб Компани AMIDE-SUBSTITUTED INDOLIC COMPOUNDS SUITABLE AS TLR INHIBITORS
EP3728225B1 (en) 2017-12-19 2022-11-09 Bristol-Myers Squibb Company Substituted indole compounds useful as tlr inhibitors
KR20200101956A (en) 2017-12-19 2020-08-28 브리스톨-마이어스 스큅 컴퍼니 6-azaindole compound
SG11202005733QA (en) 2017-12-20 2020-07-29 Bristol Myers Squibb Co Diazaindole compounds
CA3085942A1 (en) 2017-12-20 2019-06-27 Bristol-Myers Squibb Company Aryl and heteroaryl substituted indole compounds
AU2018390610A1 (en) 2017-12-20 2020-08-06 Bristol-Myers Squibb Company Amino indole compounds useful as TLR inhibitors
JP2022544412A (en) 2019-08-14 2022-10-18 キュアバック アーゲー RNA combinations and compositions with reduced immunostimulatory properties
CN115368300B (en) * 2022-10-27 2023-03-24 北京拓领博泰生物科技有限公司 Compound for TLR8 inhibitor and preparation method and application thereof

Family Cites Families (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4034075A (en) * 1972-07-03 1977-07-05 Allen & Hanburys Limited Quinaldic acid derivatives
CH612432A5 (en) * 1975-05-12 1979-07-31 Sandoz Ag
US4716168A (en) * 1985-01-08 1987-12-29 Norwich Eaton Pharmaceuticals, Inc. Imidazo(4,5-f)quinolines useful as immunomodulating agents
US4766211A (en) * 1985-09-06 1988-08-23 Ciba-Geigy Corporation Chromogenic quinazolines
JP2657760B2 (en) * 1992-07-15 1997-09-24 小野薬品工業株式会社 4-aminoquinazoline derivatives and pharmaceuticals containing them
FR2711670B1 (en) * 1993-10-22 1996-01-12 Pasteur Institut Nucleotide vector, composition containing it and vaccine for immunization against hepatitis.
US6727230B1 (en) * 1994-03-25 2004-04-27 Coley Pharmaceutical Group, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
WO1995026204A1 (en) 1994-03-25 1995-10-05 Isis Pharmaceuticals, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
US6207646B1 (en) * 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US20030050263A1 (en) * 1994-07-15 2003-03-13 The University Of Iowa Research Foundation Methods and products for treating HIV infection
US6194388B1 (en) * 1994-07-15 2001-02-27 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
US7935675B1 (en) * 1994-07-15 2011-05-03 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6429199B1 (en) * 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US20030026782A1 (en) * 1995-02-07 2003-02-06 Arthur M. Krieg Immunomodulatory oligonucleotides
WO1998029397A1 (en) * 1996-12-27 1998-07-09 Yoshitomi Pharmaceutical Industries, Ltd. Fused pyrimidine compounds and medicinal use thereof
EP0855184A1 (en) * 1997-01-23 1998-07-29 Grayson B. Dr. Lipford Pharmaceutical composition comprising a polynucleotide and an antigen especially for vaccination
CA2281838A1 (en) * 1997-02-28 1998-09-03 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated cpg dinucleotide in the treatment of lps-associated disorders
US6406705B1 (en) * 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
DE69838294T2 (en) * 1997-05-20 2009-08-13 Ottawa Health Research Institute, Ottawa Process for the preparation of nucleic acid constructs
US5939421A (en) * 1997-07-01 1999-08-17 Signal Pharmaceuticals, Inc. Quinazoline analogs and related compounds and methods for treating inflammatory conditions
US6221882B1 (en) * 1997-07-03 2001-04-24 University Of Iowa Research Foundation Methods for inhibiting immunostimulatory DNA associated responses
US6310070B1 (en) * 1997-07-15 2001-10-30 Japan Energy Corporation Purine derivatives and medicinal use thereof
DE19756388A1 (en) * 1997-12-18 1999-06-24 Hoechst Marion Roussel De Gmbh New 2-aryl-4-amino-6,7-di:methoxy-quinazoline derivatives useful as guanylate cyclase activators for treating cardiovascular diseases, etc.
ATE356630T1 (en) * 1998-04-03 2007-04-15 Univ Iowa Res Found METHOD AND PRODUCTS FOR STIMULATING THE IMMUNE SYSTEM USING IMMUNOTHERAPEUTIC OLIGONUCLEOTIDES AND CYTOKINE
EP1674574A1 (en) 1998-05-14 2006-06-28 Coley Pharmaceutical GmbH Methods for Regulating Hematopoiesis using CpG-Oligonucleotides
SI1077722T1 (en) 1998-05-22 2007-02-28 Ottawa Health Research Inst Methods and products for inducing mucosal immunity
AU5648599A (en) * 1998-09-11 2000-04-03 Kyorin Pharmaceutical Co. Ltd. Phosphonic ester derivatives and process for producing the same
WO2000076982A1 (en) 1999-06-16 2000-12-21 University Of Iowa Research Foundation Antagonism of immunostimulatory cpg-oligonucleotides by 4-aminoquinolines and other weak bases
EE200200158A (en) * 1999-09-25 2003-06-16 University Of Iowa Research Foundation Immunostimulatory nucleic acids
US6949520B1 (en) * 1999-09-27 2005-09-27 Coley Pharmaceutical Group, Inc. Methods related to immunostimulatory nucleic acid-induced interferon
CA2396871A1 (en) 2000-01-20 2001-12-20 Ottawa Health Research Institute Immunostimulatory nucleic acids for inducing a th2 immune response
US7585847B2 (en) * 2000-02-03 2009-09-08 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids for the treatment of asthma and allergy
US20020156033A1 (en) * 2000-03-03 2002-10-24 Bratzler Robert L. Immunostimulatory nucleic acids and cancer medicament combination therapy for the treatment of cancer
US20040131628A1 (en) 2000-03-08 2004-07-08 Bratzler Robert L. Nucleic acids for the treatment of disorders associated with microorganisms
EP1296714B1 (en) * 2000-06-22 2009-08-26 University Of Iowa Research Foundation Combination of CpG and antibodies directed against CD19,CD20, CD22 or CD40 for the treatment or prevention of cancer.
US20020165178A1 (en) 2000-06-28 2002-11-07 Christian Schetter Immunostimulatory nucleic acids for the treatment of anemia, thrombocytopenia, and neutropenia
US20020198165A1 (en) 2000-08-01 2002-12-26 Bratzler Robert L. Nucleic acids for the prevention and treatment of gastric ulcers
US20020091097A1 (en) * 2000-09-07 2002-07-11 Bratzler Robert L. Nucleic acids for the prevention and treatment of sexually transmitted diseases
CA2422367C (en) * 2000-09-15 2010-05-18 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors
ATE510850T1 (en) * 2000-09-15 2011-06-15 Coley Pharm Gmbh METHOD FOR HIGH-THROUGHPUT SCREENING OF CPG-BASED IMMUNAGONISTS AND ANTAGONISTS
AU2001293817A1 (en) * 2000-09-20 2002-04-02 Merck Patent Gmbh 4-amino-quinazolines
AU2001297693A1 (en) 2000-12-08 2002-09-12 Coley Pharmaceutical Gmbh Cpg-like nucleic acids and methods of use thereof
WO2002053141A2 (en) * 2000-12-14 2002-07-11 Coley Pharmaceutical Group, Inc. Inhibition of angiogenesis by nucleic acids
WO2002062767A1 (en) * 2001-02-07 2002-08-15 Sumitomo Pharmaceuticals Company, Limited Novel quinazoline derivatives
US20030050268A1 (en) * 2001-03-29 2003-03-13 Krieg Arthur M. Immunostimulatory nucleic acid for treatment of non-allergic inflammatory diseases
CN1642982A (en) * 2001-07-26 2005-07-20 唐诚公司 Agents that activate or inhibit Toll-like receptor 9
AU2002318944A1 (en) * 2001-08-01 2003-02-17 Coley Pharmaceutical Gmbh Methods and compositions relating to plasmacytoid dendritic cells
DE60229422D1 (en) * 2001-08-17 2008-11-27 Coley Pharm Gmbh COMBINATION MOTIF IMMUNOSTIMULATING OLIGONUCLEOTIDES WITH IMPROVED EFFECT
IL160837A0 (en) 2001-10-05 2004-08-31 Coley Pharm Gmbh Toll-like receptor 3 signaling agonists and antagonists
AU2002360278A1 (en) * 2001-10-12 2003-11-11 Coley Pharmaceutical Gmbh Methods and products for enhancing immune responses using imidazoquinoline compounds
EP1499187B1 (en) * 2002-04-04 2015-06-17 Zoetis Belgium S.A. Immunostimulatory g,u-containing oligoribonucleotides
US20040009949A1 (en) * 2002-06-05 2004-01-15 Coley Pharmaceutical Group, Inc. Method for treating autoimmune or inflammatory diseases with combinations of inhibitory oligonucleotides and small molecule antagonists of immunostimulatory CpG nucleic acids
US7807803B2 (en) * 2002-07-03 2010-10-05 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US7605138B2 (en) 2002-07-03 2009-10-20 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US7576066B2 (en) * 2002-07-03 2009-08-18 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US20040053880A1 (en) * 2002-07-03 2004-03-18 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US7569553B2 (en) * 2002-07-03 2009-08-04 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
AR040996A1 (en) * 2002-08-19 2005-04-27 Coley Pharm Group Inc IMMUNE STIMULATING NUCLEIC ACIDS
JP3896933B2 (en) * 2002-09-18 2007-03-22 セイコーエプソン株式会社 Electro-optical device and electronic apparatus
EA008777B1 (en) * 2002-10-29 2007-08-31 Коли Фармасьютикал Груп, Лтд. Use of cpg oligonucleotides in the treatment of hepatitis c virus infection
CA2502015A1 (en) 2002-12-11 2004-06-24 Coley Pharmaceutical Group, Inc. 5' cpg nucleic acids and methods of use
CL2004000409A1 (en) * 2003-03-03 2005-01-07 Vertex Pharma COMPOUNDS DERIVED FROM 2- (REPLACED CILO) -1- (AMINO OR REPLACED OXI) -CHINAZOLINE, INHIBITORS OF IONIC SODIUM AND CALCIUM VOLTAGE DEPENDENTS; PHARMACEUTICAL COMPOSITION; AND USE OF THE COMPOUND IN THE TREATMENT OF ACUTE PAIN, CHRONIC, NEU
WO2004087203A2 (en) * 2003-04-02 2004-10-14 Coley Pharmaceutical Group, Ltd. Immunostimulatory nucleic acid oil-in-water formulations for topical application
WO2005007672A2 (en) 2003-06-20 2005-01-27 Coley Pharmaceutical Gmbh Small molecule toll-like receptor (tlr) antagonists
JP4989225B2 (en) * 2003-09-25 2012-08-01 コーリー ファーマシューティカル グループ,インコーポレイテッド Nucleic acid lipophilic conjugate
US20050100983A1 (en) * 2003-11-06 2005-05-12 Coley Pharmaceutical Gmbh Cell-free methods for identifying compounds that affect toll-like receptor 9 (TLR9) signaling
WO2005111057A2 (en) * 2004-04-02 2005-11-24 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids for inducing il-10 responses
CA2567789A1 (en) * 2004-06-08 2006-08-03 Coley Pharmaceutical Gmbh Abasic oligonucleotide as carrier platform for antigen and immunostimulatory agonist and antagonist
EP1776105A2 (en) * 2004-07-18 2007-04-25 Coley Pharmaceutical Group, Ltd Methods and compositions for inducing innate immune responses
AU2005289426A1 (en) * 2004-09-27 2006-04-06 Amgen Inc. Substituted heterocyclic compounds and methods of use
MY159370A (en) * 2004-10-20 2016-12-30 Coley Pharm Group Inc Semi-soft-class immunostimulatory oligonucleotides
JP2008531018A (en) * 2005-02-24 2008-08-14 コーリー ファーマシューティカル グループ,インコーポレイテッド Immunostimulatory oligonucleotide
US7601355B2 (en) * 2005-06-01 2009-10-13 Northwestern University Compositions and methods for altering immune function
EA013468B1 (en) * 2005-09-16 2010-04-30 Коли Фармасьютикал Гмбх Immunostimulatory single-stranded ribonucleic acid with phosphodiester backbone
CN101321868A (en) * 2005-09-16 2008-12-10 科利制药公司 Modulation of immunostimulatory properties of short interfering ribonucleic acid (siRNA) by nucleotide modification
ES2536103T3 (en) * 2005-11-25 2015-05-20 Zoetis Belgium S.A. Immunostimulatory oligonucleotides
CA2642152C (en) * 2006-02-15 2016-11-01 Coley Pharmaceutical Gmbh Compositions and methods for oligonucleotide formulations
US20090142362A1 (en) * 2006-11-06 2009-06-04 Avant Immunotherapeutics, Inc. Peptide-based vaccine compositions to endogenous cholesteryl ester transfer protein (CETP)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1635846A4 *

Cited By (129)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8367694B2 (en) 2004-03-15 2013-02-05 Ptc Therapeutics, Inc. Carboline derivatives useful in the inhibition of angiogenesis
US8076352B2 (en) 2004-03-15 2011-12-13 Ptc Therapeutics, Inc. Administration of carboline derivatives useful in the treatment of cancer and other diseases
US8076353B2 (en) 2004-03-15 2011-12-13 Ptc Therapeutics, Inc. Inhibition of VEGF translation
US8372860B2 (en) 2004-03-15 2013-02-12 Ptc Therapeutics, Inc. Carboline derivatives useful in the inhibition of angiogenesis
US7767689B2 (en) 2004-03-15 2010-08-03 Ptc Therapeutics, Inc. Carboline derivatives useful in the treatment of cancer
US7601840B2 (en) 2004-03-15 2009-10-13 Ptc Therapeutics, Inc. Carboline derivatives useful in the inhibition of angiogenesis
WO2006035066A2 (en) * 2004-09-29 2006-04-06 Institut National De La Sante Et De La Recherche Medicale (Inserm) Toll-like receptor 3, its signalling associated molecule trif and their use in the prevention and treatment of host inflammation response induced by a rna virus
WO2006035066A3 (en) * 2004-09-29 2006-09-21 Inst Nat Sante Rech Med Toll-like receptor 3, its signalling associated molecule trif and their use in the prevention and treatment of host inflammation response induced by a rna virus
EP1655027A1 (en) * 2004-11-04 2006-05-10 Biofrontera Pharmaceuticals GmbH 9-(N-methyl-piperidyliden-4)-thioxanthene for treatment of pulmonary hypertension and migraine
US7560436B2 (en) 2005-02-22 2009-07-14 The Regents Of The University Of California Methods of treating gastrointestinal inflammation
EP1858326A1 (en) * 2005-02-22 2007-11-28 The Regents of the University of California Methods of treating gastrointestinal inflammation
EP1858326A4 (en) * 2005-02-22 2008-10-15 Univ California Methods of treating gastrointestinal inflammation
WO2006113703A3 (en) * 2005-04-18 2007-06-28 Ptc Therapeutics Inc Carboline derivatives useful in the treatment of cancer
EP2298304A3 (en) * 2005-04-18 2011-10-12 PTC Therapeutics, Inc. Carboline derivatives useful in the treatment of cancer
US8304407B2 (en) 2005-08-19 2012-11-06 Array Biopharma Inc. 8-substituted benzoazepines as toll-like receptor modulators
KR101442846B1 (en) * 2005-08-19 2014-10-01 어레이 바이오파마 인크. 8-substituted benzoazepines as toll-like receptor modulators
JP2009504763A (en) * 2005-08-19 2009-02-05 アレイ バイオファーマ、インコーポレイテッド 8-Substituted Benzazepines as Toll-like Receptor Modulators
US8686002B2 (en) 2005-08-21 2014-04-01 AbbVie Deutschland GmbH & Co. KG Heterocyclic compounds and their use as binding partners for 5-HT5 receptors
WO2007038720A2 (en) * 2005-09-27 2007-04-05 Coley Pharmaceutical Gmbh Modulation of tlr-mediated immune responses using adaptor oligonucleotides
WO2007038720A3 (en) * 2005-09-27 2007-06-21 Coley Pharm Gmbh Modulation of tlr-mediated immune responses using adaptor oligonucleotides
US8426375B2 (en) 2005-10-12 2013-04-23 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
US9206430B2 (en) 2005-10-12 2015-12-08 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
WO2007047396A3 (en) * 2005-10-12 2007-12-21 Idera Pharmaceuticals Inc Immune regulatory oligonucleotide (iro) compounds to modulate toll-like receptor based immune response
AU2006304205C1 (en) * 2005-10-12 2012-11-15 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
US8357665B2 (en) 2005-10-12 2013-01-22 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
US8399423B2 (en) 2005-10-12 2013-03-19 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
US8383598B2 (en) 2005-10-12 2013-02-26 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
AU2006304205B2 (en) * 2005-10-12 2011-09-08 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
WO2007078335A2 (en) * 2005-12-21 2007-07-12 Decode Genetics, Ehf. Biaryl nitrogen heterocycle inhibitors of lta4h for treating inflammation
WO2007078335A3 (en) * 2005-12-21 2007-11-29 Decode Genetics Inc Biaryl nitrogen heterocycle inhibitors of lta4h for treating inflammation
US7750012B2 (en) 2005-12-21 2010-07-06 Decode Genetics Ehf Biaryl nitrogen-heterocycle inhibitors of LTA4H for treating inflammation
WO2007127624A1 (en) * 2006-04-26 2007-11-08 The Uab Research Foundation Reducing cancer cell invasion using an inhibitor of toll like receptor signaling
US8440672B2 (en) 2006-09-01 2013-05-14 Merck Sharp & Dohme Corp. Diphenyl substituted alkanes
US8377898B2 (en) 2006-10-12 2013-02-19 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (IRO) compounds to modulate toll-like receptor based immune response
US8404669B2 (en) 2007-03-16 2013-03-26 University Of Florida Research Foundation, Inc. Kinase modulating compounds and uses thereof for treatment of cancer
GB2447791A (en) * 2007-03-23 2008-09-24 Univ Dundee Pharmaceutical formulations and compounds for use in alleviating conditions related to Down's syndrome
WO2008152471A1 (en) * 2007-06-12 2008-12-18 Coley Pharmaceutical Group, Inc. Quinazoline derivative useful as toll-like receptor antagonist
WO2008155468A1 (en) * 2007-06-20 2008-12-24 Marikki Laiho Activators and therapeutic applications thereof
US8680107B2 (en) 2007-06-20 2014-03-25 The Johns Hopkins University Activators and therapeutic applications thereof
WO2008155441A1 (en) * 2007-06-20 2008-12-24 Marikki Laiho Activators and therapeutic applications thereof
US20100190761A1 (en) * 2007-06-20 2010-07-29 Anthony Ogawa Diphenyl substituted alkanes
US9241932B2 (en) 2007-09-20 2016-01-26 University Of Rochester Method and compositions for treatment or prevention of inflammatory conditions
EP2203057A1 (en) * 2007-09-20 2010-07-07 University Of Rochester Method and compositions for treatment or prevention of inflammatory conditions
EP2203057A4 (en) * 2007-09-20 2010-09-29 Univ Rochester Method and compositions for treatment or prevention of inflammatory conditions
US9682070B2 (en) 2007-09-20 2017-06-20 University Of Rochester Method and compositions for treatment or prevention of inflammatory conditions
WO2009047791A3 (en) * 2007-09-24 2009-08-06 Reliance Life Sciences Pvt Ltd Toll like receptor (tlr) signaling antagonist
WO2009047791A2 (en) * 2007-09-24 2009-04-16 Reliance Life Sciences Pvt. Ltd. Toll like receptor (tlr) signaling antagonist
WO2010002835A3 (en) * 2008-07-03 2011-04-21 Osteogenex Inc. Vinpocetine and eburnamonine derivatives for promoting bone growth
US8198292B2 (en) 2008-07-03 2012-06-12 Osteogenex, Inc. Vinpocetine and eburnamonine derivatives for promoting bone growth, treating renal damage and cancer, and devices thereof
EP2149565A1 (en) * 2008-07-24 2010-02-03 Bayer Schering Pharma AG Sulfoximine substituted chinazoline derivatives as immune modulators for the treatment of inflammatory and allergic diseases
WO2010009845A1 (en) * 2008-07-24 2010-01-28 Bayer Schering Pharma Aktiengesellschaft Sulfone substituted quinazoline derivatives as immune-modulators for the treatment of inflammatory and allergic diseases
US8841311B2 (en) 2008-07-24 2014-09-23 Bayer Intellectual Property Gmbh Sulphone-substituted quinazoline derivatives as immuno-modulators, their preparation and use as medicaments
WO2010024717A1 (en) * 2008-08-22 2010-03-04 Алла Xem, Ллс Ligand with a broad spectrum of pharmacological activity, a pharmaceutical composition, a medicinal agent and a method of treatment
EA018788B1 (en) * 2008-08-22 2013-10-30 Андрей Александрович ИВАЩЕНКО Ligand with a broad spectrum of pharmacological activity and use thereof
EP3087988A2 (en) 2008-10-06 2016-11-02 Idera Pharmaceuticals, Inc. Use of inhibitors of toll-like receptors in the prevention and treatment of hypercholesterolemia and hyperlipidemia and diseases related thereto
US8853177B2 (en) 2008-10-06 2014-10-07 Idera Pharmaceuticals, Inc. Use of inhibitors of toll-like receptors in the prevention and treatment of hypercholesterolemia and hyperlipidemia and diseases related thereto
US9351964B2 (en) 2009-05-27 2016-05-31 Ptc Therapeutics, Inc. Methods for treating cancer and non-neoplastic conditions
US11613538B2 (en) 2009-05-27 2023-03-28 Ptc Therapeutics, Inc. Method of inhibiting or reducing a viral infection
US8703726B2 (en) 2009-05-27 2014-04-22 Ptc Therapeutics, Inc. Methods for treating prostate conditions
US8697662B2 (en) 2009-05-27 2014-04-15 Ptc Therapeutics, Inc. Methods for treating Kaposi sarcoma
US10947231B2 (en) 2009-05-27 2021-03-16 Ptc Therapeutics, Inc. Processes for the preparation of substituted tetrahydro beta-carbolines
US10214511B2 (en) 2009-09-03 2019-02-26 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
US9822096B2 (en) 2009-09-03 2017-11-21 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
US8575184B2 (en) 2009-09-03 2013-11-05 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
US10676460B2 (en) 2009-09-03 2020-06-09 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
US9458114B2 (en) 2009-09-03 2016-10-04 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
US11008306B2 (en) 2009-09-03 2021-05-18 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
US10780089B2 (en) 2011-04-08 2020-09-22 Janssen Sciences Ireland Uc Pyrimidine derivatives for the treatment of viral infections
US11541050B2 (en) 2011-04-08 2023-01-03 Janssen Sciences Ireland Uc Pyrimidine derivatives for the treatment of viral infections
US10420767B2 (en) 2011-04-08 2019-09-24 Janssen Sciences Ireland Uc Pyrimidine derivatives for the treatment of viral infections
US10272085B2 (en) 2011-04-08 2019-04-30 Janssen Sciences Ireland Uc Pyrimidine derivatives for the treatment of viral infections
AU2012258220B2 (en) * 2011-05-18 2017-01-19 Janssen Sciences Ireland Uc Quinazoline derivatives for the treatment of viral infections and further diseases
WO2012156498A1 (en) * 2011-05-18 2012-11-22 Janssen R&D Ireland Quinazoline derivatives for the treatment of viral infections and further diseases
EA028254B1 (en) * 2011-05-18 2017-10-31 Янссен Сайенсиз Айрлэнд Юси Quinazoline derivatives for the treatment of viral infections and further diseases
US8916575B2 (en) 2011-05-18 2014-12-23 Janssen R&D Ireland Quinazoline derivatives for the treatment of viral infections and further diseases
US8592383B2 (en) 2011-06-17 2013-11-26 Merck Sharp & Dohme Corp. Cycloalkyl-fused tetrahydroquinolines as CRTH2 receptor modulators
US11104678B2 (en) 2011-11-09 2021-08-31 Janssen Sciences Ireland Unlimited Company Purine derivatives for the treatment of viral infections
US10280167B2 (en) 2011-11-09 2019-05-07 Janssen Sciences Ireland Uc Purine derivatives for the treatment of viral infections
US10280180B2 (en) 2012-07-13 2019-05-07 Janssen Sciences Ireland Uc Macrocyclic purines for the treatment of viral infections
US10822349B2 (en) 2012-07-13 2020-11-03 Janssen Sciences Ireland Unlimited Company Macrocyclic purines for the treatment of viral infections
US11220504B2 (en) 2012-10-10 2022-01-11 Janssen Sciences Ireland Unlimited Company Pyrrolo[3,2-d] pyrimidine derivatives for the treatment of viral infections and other diseases
US10259814B2 (en) 2012-10-10 2019-04-16 Janssen Sciences Ireland Uc Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
US10723707B2 (en) 2012-11-16 2020-07-28 Janssen Sciences Ireland Unlimited Company Heterocyclic substituted 2-amino quinazoline derivatives for the treatment of viral infections
US10253003B2 (en) 2012-11-16 2019-04-09 Janssen Sciences Ireland Uc Heterocyclic substituted 2-amino quinazoline derivatives for the treatment of viral infections
US10647684B2 (en) 2013-02-21 2020-05-12 Janssen Sciences Ireland Unlimited Company 2-aminopyrimidine derivatives for the treatment of viral infections
US10259793B2 (en) 2013-02-21 2019-04-16 Janssen Sciences Ireland Uc 2-aminopyrimidine derivatives for the treatment of viral infections
WO2014128206A1 (en) * 2013-02-22 2014-08-28 Ruprecht-Karls-Universität Heidelberg Compounds for use in inhibiting hiv capsid assembly
EP2769723A1 (en) * 2013-02-22 2014-08-27 Ruprecht-Karls-Universität Heidelberg Compounds for use in inhibiting HIV capsid assembly
US11702426B2 (en) 2013-03-29 2023-07-18 Janssen Sciences Ireland Unlimited Company Macrocyclic deaza-purinones for the treatment of viral infections
US10829494B2 (en) 2013-03-29 2020-11-10 Janssen Sciences Ireland Unlimited Company Macrocyclic deaza-purinones for the treatment of viral infections
US10266543B2 (en) 2013-03-29 2019-04-23 Janssen Sciences Ireland Uc Macrocyclic deaza-purinones for the treatment of viral infections
US10865193B2 (en) 2013-05-24 2020-12-15 Janssen Sciences Ireland Unlimited Company Pyridone derivatives for the treatment of viral infections and further diseases
US10377738B2 (en) 2013-05-24 2019-08-13 Janssen Sciences Ireland Unlimited Company Pyridone derivatives for the treatment of viral infections and further diseases
US10781216B2 (en) 2013-06-27 2020-09-22 Janssen Sciences Ireland Unlimited Company Pyrrolo [3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
US10385054B2 (en) 2013-06-27 2019-08-20 Janssen Sciences Ireland Unlimited Company Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
US10822347B2 (en) 2013-07-30 2020-11-03 Janssen Sciences Ireland Unlimited Company Thieno[3,2-d]pyrimidines derivatives for the treatment of viral infections
US10316043B2 (en) 2013-07-30 2019-06-11 Janssen Sciences Ireland Unlimited Company Thieno[3,2-d]pyrimidines derivatives for the treatment of viral infections
KR101823488B1 (en) 2013-10-14 2018-01-30 에자이 알앤드디 매니지먼트 가부시키가이샤 Selectively substituted quinoline compounds
EP3995495A1 (en) * 2013-10-14 2022-05-11 Eisai R&D Management Co., Ltd. Selectively substituted quinoline compounds
US9428495B2 (en) 2013-10-14 2016-08-30 Eisai R&D Management Co., Ltd. Selectively substituted quinoline compounds
EP3626717A1 (en) * 2013-10-14 2020-03-25 Eisai R&D Management Co., Ltd. Selectively substituted quinoline compounds
US9663486B2 (en) 2013-10-14 2017-05-30 Eisai R&D Management Co., Ltd. Selectively substituted quinoline compounds
WO2015057659A1 (en) * 2013-10-14 2015-04-23 Eisai R&D Management Co., Ltd. Selectively substituted quinoline compounds
TWI624467B (en) * 2013-10-14 2018-05-21 衛材R&D企管股份有限公司 Selectively substituted quinoline compounds
AU2018264036B2 (en) * 2013-10-14 2020-07-16 Eisai R&D Management Co., Ltd. Selectively substituted quinoline compounds
RU2679622C2 (en) * 2013-10-14 2019-02-12 Эйсай Ар Энд Ди Менеджмент Ко., Лтд. Selectively substituted quinoline compounds
TWI702218B (en) * 2013-10-14 2020-08-21 日商衛材R&D企管股份有限公司 Selectively substituted quinoline compounds
USRE47193E1 (en) 2013-10-14 2019-01-08 Eisai R&D Management Co., Ltd. Selectively substituted quinoline compounds
AU2014334554B2 (en) * 2013-10-14 2018-12-06 Eisai R&D Management Co., Ltd. Selectively substituted quinoline compounds
US10087174B2 (en) 2013-10-14 2018-10-02 Eisai R&D Management Co., Ltd. Selectively substituted quinoline compounds
CN105992766A (en) * 2013-12-13 2016-10-05 武田药品工业株式会社 Pyrrolo[3,2-c]pyridine derivatives as tlr inhibitors
US9643967B2 (en) 2013-12-13 2017-05-09 Takeda Pharmaceutical Company Limited Pyrrolo[3,2-c]pyridine derivatives as TLR inhibitors
WO2015088045A1 (en) * 2013-12-13 2015-06-18 Takeda Pharmaceutical Company Limited Pyrrolo[3,2-c]pyridine derivatives as tlr inhibitors
EP3072891A1 (en) 2015-03-24 2016-09-28 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. New Toll-Like Receptor 9 Antagonists
WO2016151085A1 (en) 2015-03-24 2016-09-29 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. New toll-like receptor 9 antagonists
US11104677B2 (en) 2015-03-24 2021-08-31 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Toll-like receptor 9 antagonists
US10662177B2 (en) 2016-03-21 2020-05-26 Council Of Scientific & Industrial Research Blocking toll-like receptor 9 signaling with small molecule antagonist
AU2017238758B2 (en) * 2016-03-21 2021-01-21 Council Of Scientific & Industrial Research Blocking toll-like receptor 9 signaling with small molecule antagonist
WO2017163264A1 (en) * 2016-03-21 2017-09-28 Council Of Scientific & Industrial Research Blocking toll-like receptor 9 signaling with small molecule antagonist
US11053256B2 (en) 2016-07-01 2021-07-06 Janssen Sciences Ireland Unlimited Company Dihydropyranopyrimidines for the treatment of viral infections
US10968184B2 (en) 2016-09-29 2021-04-06 Janssen Sciences Ireland Unlimited Company Pyrimidine prodrugs for the treatment of viral infections and further diseases
WO2018146171A1 (en) 2017-02-09 2018-08-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for treating mast cell activation diseases
WO2019020732A1 (en) 2017-07-27 2019-01-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for determining whether a patient suffering from rhabdomyolysis achieves a response with a tlr9 antagonist
US11458126B2 (en) 2017-08-01 2022-10-04 Ptc Therapeutics, Inc. DHODH inhibitor for use in treating hematologic cancers
US11597704B2 (en) 2018-03-01 2023-03-07 Janssen Sciences Ireland Unlimited Company 2,4-diaminoquinazoline derivatives and medical uses thereof
WO2019197682A1 (en) 2018-04-14 2019-10-17 4Sc Ag Pharmaceutical combination products comprising a histone deacetylase (hdac) inhibitor and a tlr7 agonist and/or tlr8 agonist for the treatment of cancer
WO2019207066A1 (en) 2018-04-26 2019-10-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for the treatment of sjögren's syndrome
WO2022060805A1 (en) * 2020-09-15 2022-03-24 The Johns Hopkins University Compounds which inhibit rna polymerase
EP4119145A1 (en) * 2021-07-15 2023-01-18 Dompe' Farmaceutici S.P.A. Compounds for the treatment of covid-19

Also Published As

Publication number Publication date
EP1635846A4 (en) 2009-01-28
AU2004257149A1 (en) 2005-01-27
WO2005007672A8 (en) 2007-11-01
EA200600069A1 (en) 2006-08-25
CA2528774A1 (en) 2005-01-27
KR20060016817A (en) 2006-02-22
US20070232622A1 (en) 2007-10-04
BRPI0411514A (en) 2006-08-01
MXPA05013922A (en) 2006-02-24
EP1635846A2 (en) 2006-03-22
IL172580A0 (en) 2006-04-10
US20050119273A1 (en) 2005-06-02
US7410975B2 (en) 2008-08-12
WO2005007672A3 (en) 2005-09-15
JP2007524615A (en) 2007-08-30

Similar Documents

Publication Publication Date Title
US7410975B2 (en) Small molecule toll-like receptor (TLR) antagonists
US9637489B2 (en) Immune system modulators
US20100160314A1 (en) Small Molecule Inhibitors of Toll-Like Receptor 9
AU2012318694B2 (en) Novel imidazole quinoline-based immune system modulators
AU2012262021B2 (en) Novel immune system modulators
JP2017525711A (en) Novel N2, N4, N7,6-tetrasubstituted pteridine-2,4,7-triamine and 2,4,6,7-tetrasubstituted pteridine compounds and methods for their synthesis and use
US20090169472A1 (en) Methods and compositions for treating immune disorders
ZA200510028B (en) Small molicule toll-like receptor (TLR) antagonists
KR20080048067A (en) Immunostimulatory single-stranded ribonucleic acid with phosphodiester backbone
BRPI0814260B1 (en) composition comprising a single ribbon polymer, method comprising contacting a cell with such polymer and use of such polymer

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

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

Ref document number: 2528774

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 200510028

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 12005502256

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 172580

Country of ref document: IL

Ref document number: 2006517471

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2004776820

Country of ref document: EP

Ref document number: 544207

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: PA/a/2005/013922

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 20048170647

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 1020057024410

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2004257149

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1200600082

Country of ref document: VN

WWE Wipo information: entry into national phase

Ref document number: 153/KOLNP/2006

Country of ref document: IN

Ref document number: 200600069

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 2004257149

Country of ref document: AU

Date of ref document: 20040618

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 1020057024410

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2004776820

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0411514

Country of ref document: BR