CA2476896A1 - Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors - Google Patents

Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors Download PDF

Info

Publication number
CA2476896A1
CA2476896A1 CA002476896A CA2476896A CA2476896A1 CA 2476896 A1 CA2476896 A1 CA 2476896A1 CA 002476896 A CA002476896 A CA 002476896A CA 2476896 A CA2476896 A CA 2476896A CA 2476896 A1 CA2476896 A1 CA 2476896A1
Authority
CA
Canada
Prior art keywords
vertebrate
brain
disease
pro
inflammatory
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002476896A
Other languages
French (fr)
Inventor
Svetlana M. Ivanova
Kevin J. Tracey
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
North Shore Long Island Jewish Research Institute
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2476896A1 publication Critical patent/CA2476896A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/155Amidines (), e.g. guanidine (H2N—C(=NH)—NH2), isourea (N=C(OH)—NH2), isothiourea (—N=C(SH)—NH2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/341Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide not condensed with another ring, e.g. ranitidine, furosemide, bufetolol, muscarine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/10Inactivation or decontamination of a medicinal preparation prior to administration to an animal or a person
    • A61K41/17Inactivation or decontamination of a medicinal preparation prior to administration to an animal or a person by ultraviolet [UV] or infrared [IR] light, X-rays or gamma rays
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N1/00Electrotherapy; Circuits therefor
    • A61N1/18Applying electric currents by contact electrodes
    • A61N1/32Applying electric currents by contact electrodes alternating or intermittent currents
    • 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
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/12Keratolytics, e.g. wart or anti-corn preparations
    • 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
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/04Amoebicides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • 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
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

Methods for inhibiting pro-inflammatory cytokine release or inflammation in a vertebrate are provided. The methods comprise activating a brain muscarinic receptor of the vertebrate, or directly stimulating a vagus nerve pathway in the brain of the vertebrate. Also provided are methods for conditioning a vertebrate to inhibit the release of a pro-inflammatory cytokine or reduce inflammation in the vertebrate upon experiencing a sensory stimulus. The methods comprise (a) activating a muscarinic brain receptor or directly stimulating the vagus nerve pathway in the brain of the vertebrate and providing the sensory stimulus to the vertebrate within a time period sufficient to create an association between the stimulus and the activation of the brain muscarinic receptor; and (b) repeating step (a) at sufficient time intervals and duration to reinforce the association sufficiently for the inflammation to be reduced by the sensory stimulus alone.

Description

INHIBITION OF INFLAMMATORY CYTOKINE PRODUCTION BY
STIMULATION OF BRAIN MUSCARINIC RECEPTORS
RELATED APPLICATION
This application claims the benefit of U.S. Provisional Application No.
60/360,082, filed February 26, 2002. The entire teachings of the above application are incorporated herein by reference.
BACKGROUND OF THE INVENTION
Field of the Invention The present invention generally relates to methods of reducing inflammation.
More specifically, the invention relates to methods for reducing inflammation caused by proinflasnmatory cytol~ines or an inflammatory cytolcine cascade.
Vertebrates achieve internal homeostasis during infection or injury by balancing the activities of proinflammatory and anti-inflammatory pathways.
However, in many disease conditions, this internal homeostasis becomes out of balance. For example, endotoxin (lipopolysaccharide, LPS), produced by all Gram-negative bacteria, activates macrophages to release cytol~ines that are potentially lethal to the host (Tracey et cal., 1986; Dinarello, 1994; Wang, H., et al., 1999;
Nathan, 1987).
Inflammation and other deleterious conditions (such as septic shoclc caused by endotoxin exposure) are often induced by proinflammatory cytol~ines, such as tumor necrosis factor (TNF; also known as TNFa or cachectin), interleulcin (IL)-la, IL-1 Vii, IL-6, IL-8, IL-18, interferon-y, platelet-activating factor (PAF), macrophage migration inhibitory factor (MTF), and other compounds (Thompson, 1998).
Certain other compounds, for example, high mobility group protein 1 (HMG-B1), are induced during various conditions, such as sepsis, and can also serve as proinflanunatory cytol~ines (WO 00/47104). These proinflammatory cytol~ines are produced by several different cell types, most importantly immune cells (for example, monocytes, macrophages, and neutrophils), but also non-immune cells such as fibroblasts, osteoblasts, smooth muscle cells, epithelial cells, and neurons (Zhang and Tracey, 1998). Proinflammatory cytokines contribute to various disorders, notably sepsis, through their release during an inflammatory cytokine cascade.
lilflammatory cytokine cascades contribute to deleterious characteristics of numerous disorders. These deleterious characteristics include inflammation and apoptosis (Pull~l~i, 1997). Disorders where inflammatory cytol~ine cascades are involved at least in part, include, without limitation, diseases involving the gastrointestinal tract and associated tissues (such as appendicitis, peptic, gastric and duodenal ulcers, peritonitis, pancreatitis, ulcerative, pseudomembranous, acute and ischemic colitis, inflammatory bowel disease, diverticulitis, epiglottitis, achalasia, cholangitis, coeliac disease, cholecystitis, hepatitis, Crohn's disease, enteritis, and Whipple's disease); systemic or local inflammatory diseases and conditions (such as asthma, allergy, anaphylactic shock, immune complex disease, organ ischemia, reperfusion injury, organ necrosis, hay fever, sepsis, septicemia, endotoxic shoclc, cachexia, hyperpyrexia, eosinophilic granuloma, granulomatosis, and sarcoidosis);
diseases involving the urogenital system and associated tissues (such as septic abortion, epididymitis, vaginitis, prostatitis, and urethritis); diseases involving the respiratory system and associated tissues (such as bronchitis, emphysema, rhinitis, cystic fibrosis, adult respiratory distress syndrome, pneumonitis, pneumoultramicroscopic silicovolcanoconiosis, alveolitis, bronchiolitis, pharyngitis, pleurisy, and sinusitis); diseases arising from infection by various viruses (such as influenza, respiratory syncytial virus, HIV, hepatitis B virus, hepatitis C
virus, and herpes), bacteria (such as disseminated bacteremia, Dengue fever), fungi (such as candidiasis) and protozoal and multicellular parasites (such as malaria, filariasis, amebiasis, and hydatid cysts); dermatological diseases and conditions of the skin (such as burns, dermatitis, dermatomyositis, sunburn, urticaria warts, and wheals);
diseases involving the cardiovascular system and associated tissues (such as vasculitis, angiitis, endocarditis, arteritis, atherosclerosis, thrombophlebitis, pericarditis, myocarditis, myocardial ischemia, congestive heart failure, periameritis nodosa, and rheumatic fever); diseases involving the central or peripheral nervous system and associated tissues (such as Alzheimer's disease, meningitis, encephalitis, multiple sclerosis, cerebral infarction, cerebral embolism, Guillame-Barre syndrome, neuritis, neuralgia, spinal cord injury, paralysis, and uveitis); diseases of the bones, joints, muscles, and connective tissues (such as the various arthritis and amhralgias, osteomyelitis, fasciitis, Paget's disease, gout, periodontal disease, rheumatoid arthritis, and synovitis); other autoimmune and inflammatory disorders (such as myasthenia gravis, thyroiditis, systemic lupus erythematosus, Goodpasture's syndrome, Behcets's syndrome, allograft rejection, graft-versus-host disease, Type I
diabetes, Berger's disease, and Retier's syndrome); as well as various cancers, tumors and proliferative disorders (such as Hodgkins disease); and, in any case the inflammatory or immune host response to any primary disease (see, e.g., Gattorno et al., 2000; Yeh and Schuster, 1999;,McGuinness et al., 2000; Hsu et al., 1999;
Jander and Stoll, 2001; Kanai et al., 2001; Prystowsky and Rege, 1997; Kimmings et al., 2000; Hirano, T., 1999; Lee et al., 1995;Waserman et al., 2000; Watanabe et al., 1997; Katagiri, et al., 1997; Bumgardner, and Orosz, 1999; Dibbs, et al., 1999;
Blackwell and Christman, 1996; Blum and Miller, 199; Cameron, 2000; Fox, 2000;
Hommes and van Deventer, 2000; Gracie et al., 1999; Rayner et al. 2000).
Tumor necrosis factor is known to be a major pro-inflammatory cytokine mediator of various acute and chronic inflammatory diseases, e.g., gram negative bacterial sepsis, multi-system organ failure (MSOF), circulatory collapse and death.
The primary source of circulating TNF following a septic challenge is the liver.
Thus, rats subjected to two-thirds hepatectomy produce 64% less TNF after endotoxin, as compared to sham controls (Kumins et al., 1996).
Direct production of TNF by cardiac muscle also appears to play a major role in septic myocardial depression. Myocytes respond to stress by primary production of TNF, as well as by increasing TNF receptors (Irwin et al., 1999). TNF, either produced locally in the heam, or originating from other sources, causes myocyte apoptosis and thrombosis (Song et al., 2000). TNF has been implicated in various cardiac disorders including cardiac failure secondary to septic cardiomyopathy, bi-ventricular dysfunction, and pulmonary edema. TNF can also have a direct negative inotropic effect on cardiac function.
Vertebrates respond to inflammation caused by inflammatory cytolcine cascades in part through humoral mechanisms of the central nervous system (activation of the hypothalamus-pituitary adrenal [HPA] axis), by means of vagal nerve activation, and by means of peripheral anti-inflammatory cytokine production (e.g., IL-10 production). This response has been characterized in detail with respect to systemic humoral response mechanisms during inflammatory responses to endotoxin (Besedovsky et al., 1986; Woiciechowsky et al., 1998; Hu et al., 1991;
Lipton and Catania, 1997).
The vagus nerve is a critical cranial nerve in modulating whole body homeostasis, including, inter alia, inflammatory regulation through both afferent and efferent signaling. Vagus nerve fibers reach multiple internal organs, such as the trachea/bronchi, abdominal blood vessels, kidneys, small and large intestine, adrenals, liver, and heart. The paws of an animal have also been shown to receive vagus nerve innervation via nerve fibers traveling along the blood vessels, as well as nerve fibers in sweat glands, etc.
In one set of responses, afferent vagus nerve fibers are activated by endotoxin or cytokines, stimulating the release of humoral anti-inflammatory responses through glucocorticoid hormone release (Watlcins and Maier, 1999; Sternberg, 1997;
Scheinman et al, 1995). Cytolcines or endotoxin can stimulate the afferent vagus nerve, which in turn signals a number of critical brain nuclei, and leads to activation of the HPA anti-inflammatory responses and down-regulation of endotoxemia and cytol~inemia (Gaykema et al., 1995; Fleshner et al., 1998; Watlcins et al., 1995;
Romanovslcy et al., 1997). Similarly, direct efferent vagus nerve stimulation (VNS) in rats prevents shock secondary to an induced endotoxic challenge, by decreasing TNF synthesis in the liver (see U.S. Patent Application No. 09/855,446, the teachings of which are incorporated herein by reference). The efferent vagus nerve can also be stimulated to achieve immunosuppression by pharmacological means.
For example, the anti-inflammatory pharmacological agent CNI-1493, when administered peripherally, has the ability to cross the blood-brain barrier, and activate the efferent vagus nerve through a central mechanism of action, thus mediating peripheral immunosuppression, with anti-inflammatory effects (Borovikova et al., 2000). Intracerebroventricular administration of CIVI-1493 is also an effective anti-inflammatory treatment (Id.) The effect of direct stimulation of brain cholinergic agonists on inflammation was evaluated in Bhattacharya et al. (1991). In those studies, direct administration of high doses of muscarinic agonists caused augmentation of carrageenan-induced paw edema. Although low doses of the muscarine agonist carbachol caused attenuation of paw edema, the authors concluded that, overall, muscarinic agonist treatment of the brain caused augmentation of paw edema. There was also no suggestion in that paper that the muscarinic agonist could be useful in reducing inflammation.
Conditioning of the immune s, stem.
Conditioning is a method of training an animal by which a perceptible neutral stimulus is temporarily associated with a physiological stimulus so that the animal will ultimately respond to the neutral stimulus as if it were the physiological stimulus. Pavlov, for instance, trained dogs to respond with salivation to the ringing of a bell following prior experiments where the dogs were prescribed a food stimulus (associated with salivation) simultaneously with a ringing bell stimulus.
Elmer Green (1969) proposed that perception elicits mental and emotional responses, generating limbic, hypothalamic, and pituitary responses that bring about physiological changes. Ader and Cohen (1982) further extended the scope of conditioning to the immune system. They showed that rats could be conditioned to respond to a neutral stimulus, saccharin, with a decreased immune response after having been repeatedly and simultaneously exposed to cyclophosphamide, an immunosuppressive drug. The observed effects extended to both humoral immunity (i.e., antibody production) as well as to cellular immunity (i.e., graft vs.
host response)(Ader and Cohen, 1975; Cohen et al., 1979; Ader and Cohen, 1982; Ader and Cohen, 1992).
Human studies have also linked immune dysregulation with psychological disease (Cohen et al., 2001). Additionally, hypnosis (Wyler-Harper et al., 1994; Fox et al., 1999) and biofeedback (Peavey et al., 1985) has been found to be effective in modulating the immune response.
SUMMARY OF THE INVENTION
Accordingly, the inventors have succeeded in discovering that pro-inflammatory cytokine release in vertebrates, and the associated inflammatory responses, can be inhibited by activating brain muscarinic receptors. Further, the inventors have discovered that this anti-inflammatory response can be conditioned by repeated association of a sensory stimulus with activation of brain muscarinic receptors. These discoveries enable novel methods for inhibiting pro-inflamyatory cytokine release and inflammation.
Thus, in one aspect, the present invention is directed to methods of inhibiting release of a pro-inflannnatory cytokine in a vertebrate. The method comprises activating a brain muscarinic receptor in the vertebrate.
The present invention is also directed to methods of inhibiting release of a pro-inflarmnatory cytolcine in a vertebrate. The method comprises directly stimulating a vagus nerve pathway in the brain of the vertebrate.
In additional embodiments, the invention is directed to methods of treating an inflammatory disease in a vertebrate. The methods comprise activating a brain muscarinic receptor in the vertebrate.
The invention is additionally directed to methods of treating an inflammatory disease in a vertebrate. The methods comprise directly stimulating a vagus nerve pathway in the brain of the vertebrate.
In another aspect, the present invention is directed to methods of inhibiting apoptosis of a cardiac myocyte in a vertebrate at rislc for cardiac myocyte apoptosis.
The methods comprise activating a brain muscarinic receptor in the vertebrate.
The present invention is also directed to methods of inhibiting apoptosis of a cardiac myocyte in a vertebrate at risk for cardiac myocyte apoptosis. The methods comprise directly stimulating a vagus nerve pathway in the brain of the vertebrate.

In additional embodiments, the present invention is directed to methods of conditioning a vertebrate to inhibit the release of a pro-inflammatory cytol~ine upon experiencing a sensory stimulus. The methods comprise the following steps:
(a) activating a brain muscarinic receptor in the vertebrate and providing the sensory stimulus to the vertebrate within a time period sufficient to create an association between the stimulus and the activation of the brain muscarinic receptor;
and (b) repeating step (a) at sufficient time intervals and duration to reinforce the association sufficiently for the pro-inflammatory cytokine release to be inhibited by the sensory stimulus alone.
The invention is also directed to methods of conditioning a vertebrate to inhibit the release of a pro-inflammatory cytol~ine upon experiencing a sensory stimulus. The methods comprise the following steps:
(a) directly stimulating a vagus nerve pathway in the brain of the vertebrate and providing the sensory stimulus to the vertebrate within a time period sufficient to create an association between the stimulus and the stimulation of a vagus nerve pathway; and (b) repeating step (a) at sufficient time intervals and duration to reinforce the association sufficiently for the pro-inflammatory cytokine release to be inhibited by the sensory stimulus alone.
The invention is additionally directed to methods of conditioning a vertebrate to reduce inflammation in the vertebrate upon experiencing a sensory stimulus.
The methods comprise the following steps:
(a) activating a brain muscarinic receptor in the vertebrate and providing the sensory stimulus to the vertebrate within a time period sufficient to create an association between the stimulus and the activation of the brain muscarinic receptor;
and (b) repeating step (a) at sufficient time intervals and duration to reinforce the association sufficiently for the inflammation to be reduced by the sensory stimulus alone.

_g_ Additionally, the present invention is directed to methods of conditioning a vertebrate to reduce inflammation in the vertebrate upon experiencing a sensory stimulus. The methods comprise the following steps:
(a) directly stimulating a vagus nerve pathway in the brain of the vertebrate and providing the sensory stimulus to the vertebrate within a time period sufficient to create an association between the stimulus and the activation of the brain muscarinic receptor; and (b) repeating step (a) at sufficient time intervals and duration to reinforce the association sufficiently for the inflammation to be reduced by the sensory stimulus alone.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a graph summarizing the results of experiments showing that intracerebroventricular administration of CNI-1493 significantly inhibits LPS-induced release of TNF, and that atropine (ATR) reverses the effect.
Figure 2 is a graph summarizing the results of experiments showing that intracerebroventricular administration of nicotine or prozalc has no effect on LPS-induced release of TNF.
Figure 3 is a graph summarizing the results of experiments showing that intracerebroventricular administration of CNI-1493 significantly inhibits carageenan-induced paw edema, and that atropine (ATR) reverses the effect.
Figure 4 is a graph summarizing the results of experiments showing that intracerebroventricular administration of muscarine significantly inhibits carrageenan-induced paw edema in a dose-dependent manner.
Figure 5 is a graph summarizing the results of experiments showing that vagotomy abrogates the inhibitory effects of intracerebroventricular (i.c.v.) administration of muscarine on carrageenan-induced paw edema.
Figure 6 is a graph summarizing the results of experiments showing that intracerebroventricular administration of the M1 agonist McN-A-343 or the M4 agonist MT-3 significantly inhibits carrageenan-induced paw edema.

Figure 7 is a graph summarizing the results of experiments showing that intracerebroventricular (i.c.v.) administration of the M1 agoust McN-A-343 is significantly more potent in inhibiting carrageenan-induced paw edema as compared to intraperitoneal (i.p.) administration.
Figure 8 is a graph summarizing the results of experiments showing that conditioning animals by associating intraperitoneal CNI-1493 administration with bell ringing allowed the inhibition of LPS-induced TNF release by bell ringing without CNI-1493 administration.
Figure 9A is a graph summarizing the results of the effect of intracerebroventricular (i.c.v.) administration of no muscarine (control), or muscarine at 0.005 ~,g/kg body weight, 0.5 ~,g/kg body weight, 5.0 ~,g/kg body weight, or 50 ~,g/lcg body weight on LPS-induced TNF production (TNF
concentration (pg/ml)) in the serum of rats. R indicates the number of rats per test condition.
Figure 9B is a graph summarizing the results of the effect of intracerebroventricular (i.c.v.) administration of no muscarine (control), or muscarine at 0.005 ~g/kg body weight, 0.5 ~,g/kg body weight, 5.0 ~,g/lcg body weight, or 50 ~.g/kg body weight on LPS-induced TNF production (TNF
concentration (ng/g protein)) in the heart tissues of rats. R indicates the number of rats per test condition.
Figure 9C is a graph summarizing the results of the effect of intracerebroventricular (i.c.v.) administration of no muscarine (control), or muscarine at 0.005 ~g/kg body weight, 0.5 ~,g/kg body weight, 5.0 ~,g/lcg body weight, or 50 ~,g/lcg body weight on LPS-induced TNF production (TNF
concentration (ng/g protein)) in the spleens of rats. R indicates the number of rats per test condition.
Figure l0A is a graph summarizing the results of the effect of intravenous (i.v.) administration of no muscarine (control), or muscarine at 0.05 ~g/kg body weight, 0.5 ~.g/kg body weight, or 5.0 wg/lcg body weight on LPS-induced TNF
production (TNF concentration (pg/ml)) in the serum of rats. R indicates the number of rats per test condition.

Figure l OB is a graph summarizing the results of the effect of intravenous (i.v.) admiiustration of no muscarine (control), or muscarine at 0.05 ~,g/kg body weight, 0.5 ~,g/kg body weight, or 5.0 ~g/lcg body weight on LPS-induced TNF
production (TNF concentration (ng/g protein)) in the livers of rats. R
indicates the number of rats per test condition.
Figure l OC is a graph summarizing the results of the effect of intravenous (i.v.) administration of no muscarine (control), or muscarine at 0.05 ~,g/kg body weight, 0.5 ~.glkg body weight, or 5.0 ~,g/lcg body weight on LPS-induced TNF
production (TNF concentration (ng/g protein)) in the spleens of rats. R
indicates the number of rats per test condition.
Figure l OD is a graph summarizing the results of the effect of intravenous (i.v.) administration of no muscarine (control), or muscarine at 0.05 ~,g/kg body weight, 0.5 ~g/lcg body weight, or 5.0 wg/kg body weight on LPS-induced TNF
production (TNF concentration (ng/g protein)) in the heart tissues of rats. R
indicates the nmnber of rats per test condition.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is based on the discovery that activation of vertebrate brain muscarinic receptors causes an inhibition of the release of various pro-inflammatory cytolcines in the periphery, which in turn causes a reduction of peripheral inflammation. This reduction of peripheral inflammation can be achieved by muscarinic agonist treatment or by exposure to an external sensory stimulus after Pavlovian conditioning by prior repeated association of the stimulus with the muscarinic agonist treatment. The inhibition of pro-inflammatory cytolcine release and the reduction of peripheral inflammation is vagus nerve-dependent and can also be reduced by direct stimulation of the vagus nerve in the brain. These discoveries enable the treatment of various inflammatory conditions in novel ways.
As used herein, a cytol~ine is a soluble protein or peptide which is naturally produced by vertebrate cells and which act ih vivo as humoral regulators at micro- to picomolar concentrations. Cytokines can, either under normal or pathological conditions, modulate the functional activities of individual cells and tissues. A pro-inflammatory cytokine is a cytokine that is capable of causing any of the following physiological reactions associated with inflammation: vasodilatation, hyperemia, increased permeability of vessels with associated edema, accumulation of granulocytes and mononuclear phagocytes, or deposition of fibrin. In some cases, the pro-inflammatory cytokine can also cause apoptosis, such as iri chronic heart failure, where TNF bas been shown to stimulate cardiomyocyte apoptosis (Pulldci, 1997; Tsutsui et al., 2000). Nonlimiting examples of pro-inflammatory cytokines are tumor necrosis factor (TNF), interleukin (IL,)-1a, IL,-lei, IL-6, IL,-8, IL-18, interferon-'y, HMG-B1, platelet-activating factor (PAF), and macrophage migration inhibitory factor (MIF). In preferred embodiments of the invention, the pro-inflammatory cytokine that is inhibited by cholinergic agonist treatment is TNF, IL-1, IL-6, or IL-18, because these cytolcines are produced by macrophages and mediate deleterious conditions for many important disorders, for example, endotoxic shock, asthma, rheumatoid arthritis, inflammatory bile disease, heart failure, and allograft rejection. In most preferred embodiments, the pro-inflammatory cytokine is TNF.
Pro-inflammatory cytokines are to be distinguished from anti-inflammatory cytokines, such as IL-4, IL-10, and IL,-13, which tend to inhibit inflammation. In preferred embodiments, release of anti-inflammatory cytol~ines is not inhibited by cholinergic agonists.
In many instances, pro-inflammatory cytolcines are produced in an inflammatory cytolcine cascade, defined herein as an ira vivo release of at least one pro-inflammatory cytokine in a vertebrate, wherein the cytokine release affects a physiological condition of the vertebrate. Thus, an inflammatory cytokine cascade is inhibited in embodiments of the invention where pro-inflammatory cytokine release causes a deleterious physiological condition.
Nonlimiting examples of diseases characterized by the presence of deleterious physiological conditions at least partially mediated by pro-inflammatory cytokine release are appendicitis, peptic, gastric or duodenal ulcers, peritonitis, pancreatitis, ulcerative, pseudomembranous, acute or ischemic colitis, inflammatory bowel disease, diverticulitis, epiglottitis, achalasia, cholangitis, cholecystitis, hepatitis, Crohn's disease, enteritis, Whipple's disease, asthma, allergy, anaphylactic shock, immiule complex disease, organ ischemia, reperfusion injury, organ necrosis, hay fever, sepsis, septicemia, endotoxic shock, cachexia, hyperpyrexia, eosinophilic granuloma, granulomatosis, sarcoidosis, septic abortion, epididymitis, vaginitis, prostatitis, urethritis, bronchitis, emphysema, rhinitis, cystic fibrosis, pneumonitis, pneumoultramicroscopic silicovolcanoconiosis, alveolitis, bronchiolitis, pharyngitis, pleurisy, sinusitis, influenza, respiratory syncytial virus, herpes, disseminated bacteremia, Dengue fever, candidiasis, malaria, filariasis, amebiasis, hydatid cysts, burns, dermatitis, dermatomyositis, sunburn, urticaria, warts, wheals, vasculitis, angiitis, endocarditis, arteritis, atherosclerosis, thrombophlebitis, pericarditis, myocarditis, myocardial ischemia, periarteritis nodosa, rheumatic fever, Alzheimer's disease, coeliac disease, congestive heart failure, adult respiratory distress 'syndrome, meningitis, encephalitis, multiple sclerosis, cerebral infarction, cerebral embolism, Guillame-Bane syndrome, neuritis, neuralgia, spinal cord injury, paralysis, uveitis, arthritis, arthralgias, osteomyelitis, fasciitis, Paget's disease, gout, periodontal disease, rheumatoid arthritis, synovitis, myasthenia gravis, thyroiditis, systemic lupus erythematosus, Goodpasture's syndrome, Behcets's syndrome, allograft rejection, graft-versus-host disease, Type I diabetes, ankylosing spondylitis, Berger's disease, Retier's syndrome, and Hodgkins disease. Additional examples of conditions mediated by pro-inflammatory cytolcine release include shock, for example, hemorrhagic shock, chronic obstructive pulmonary disease (COPD) and psoriasis.
Any vertebrate cell that produces pro-inflammatory cytokines is useful for the practice of the invention. Nonlimiting examples are monocytes, macrophages, any cells resident in the liver that make, transport, or concentrate pro-inflammatory cytokines including Kupffer cells and biliary endothelial cells, neutrophils, epithelial cells, osteoblasts, fibroblasts, hepatocytes, muscle cells including smooth muscle cells and cardiac myocytes, and neurons. In preferred embodiments, the cell is a macrophage, Kupffer cell, monocyte, biliary endothelial cell, hepatocyte, or cardiac myocyte.

As used herein, a cholinergic agonist is a compound that binds to cholinergic receptors on cells. The skilled artisan can determine whether any particular compound is a cholinergic agonist by any of several well known methods.
When referring to the effect of the cholinergic agonist on release of pro-inflammatory cytolcines or an inflammatory cytokine cascade, or the effect of vagus nerve stimulation on an inflammatory cytokine cascade, the use of the terms "inhibit" or "decrease" encompasses at least a small but measurable reduction in pro-inflarmnatory cytolcine release. In preferred embodiments, the release of the pro-inflammatory cytolcine is inhibited by at least 20% over non-treated controls; in more preferred embodiments, the inhibition is at least 50%; in still more preferred embodiments, the inhibition is at least 70%, and in the most preferred embodiments, the inhibition is at least 80%. Such reductions in pro-inflammatory cytokine release are capable of reducing the deleterious effects of an inflammatory cytolcine cascade.
Accordingly, in some embodiments, the present invention is directed to methods of inhibiting the release of a pro-inflammatory cytokine in a vertebrate.
The methods comprise activating a brain muscarinic receptor in the vertebrate.
In preferred embodiments, the pro-inflammatory cytokine is tumor necrosis factor (TNF), interleukin (IL)-1(3, IL-6, IL-18, HMG-B1, MIP-la, MIP-1(3, MIF, interferon-'y, or PAF. In more preferred embodiments, the pro-inflammatory cytokine is selected from the group consisting of tumor necrosis factor (TNF), interleulcin (IL)-1 (3, IL-6, IL-18, and HMG-Bl. In the most preferred embodiments, the pro-inflammatory cytokine is TNF.
These methods are useful for preventing the release of pro-inflammatory cytolcines in any vertebrate. In preferred embodiments, the vertebrate is a mammal.
In particularly preferred embodiments, the vertebrate is a human. The vertebrate is preferably a patient suffering from, or at risk for, a condition mediated by an inflammatory cytokine cascade. As used herein, a patient can be any vertebrate individual from a species that has a vagus nerve. Preferably, the condition is appendicitis, peptic, gastric and duodenal ulcers, peritonitis, pancreatitis, ulcerative, pseudomembranous, acute and ischemic colitis, inflammatory bowel disease, diverticulitis, epiglottitis, achalasia, cholangitis, cholecystitis, hepatitis, Crohn's disease, enteritis, Whipple's disease, asthma, allergy, anaphylactic shock, immune complex disease, organ ischemia, reperfusion injury, organ necrosis, hay fever, sepsis, septicemia, endotoxic shock, cachexia, hyperpyrexia, eosinophilic granuloma, granulomatosis, sarcoidosis, septic abortion, epididymitis, vaginitis, prostatitis, urethritis, bronchitis, emphysema, rhinitis, cystic fibrosis, pneumonitis, pneumoultramicroscopic silicovolcanoconiosis, alveolitis, bronchiolitis, pharyngitis, pleurisy, sinusitis, influenza, respiratory syncytial virus infection, herpes infection, HIV infection, hepatitis B virus infection,.hepatitis C virus infection, disseminated bacteremia, Dengue fever, candidiasis, malaria, filariasis, amebiasis, hydatid cysts, burns, dermatitis, dermatomyositis, sunburn, urticaria, waits, wheals, vasculitis, angiitis, endocarditis, arteritis, atherosclerosis, thrombophlebitis, pericarditis, myocarditis, myocardial ischemia, periarteritis nodosa, rheumatic fever, Alzheimer's disease, coeliac disease, congestive heart failure, adult respiratory distress syndrome, meningitis, encephalitis, multiple sclerosis, cerebral infarction, cerebral embolism, Guillame-Barre syndrome, neuritis, neuralgia, spinal cord injury, paralysis, uveitis, arthritis, arthralgias, osteomyelitis, fasciitis, Paget's disease, gout, periodontal disease, rheumatoid arthritis, synovitis, myasthenia gravis, thyroiditis, systemic lupus erythematosus, Goodpasture's syndrome, Behcets's syndrome, allograft rejection, graft-versus-host disease, Type I diabetes, anlcylosing spondylitis, Berger's disease, Retier's syndrome, and Hodgkins disease. More preferably, the condition is appendicitis, peptic, gastric and duodenal ulcers, peritonitis, pancreatitis, ulcerative, pseudomembranous, acute and ischemic colitis, inflammatory bowel disease, hepatitis, Crohn's disease, asthma, allergy, anaphylactic shoclc, organ ischemia, reperfusion injury, organ necrosis, hay fever, sepsis, septicemia, endotoxic shock, cachexia, septic abortion, disseminated bacteremia, burns, Alzheimer's disease, coeliac disease, congestive heart failure, adult respiratory distress syndrome, cerebral infarction, cerebral embolism, spinal cord injury, multiple sclerosis, paralysis, allograft rejection and graft-versus-host disease. In most preferred embodiments, the condition is endotoxic shock.
These methods can be used to prevent release of pro-inflammatory cytol~ines in the brain or any peripheral organ served by the vagus nerve. Preferred examples include the liver, which makes pro-inflammatory cytokines involved in systemic inflammatory cascades such as endotoxic shock. Another preferred peripheral organ is the heart, since it is known that cardiac myocytes release pro-inflammatory cytokines implicated in myocyte apoptosis and thrombosis.
The preferred brain muscarinic receptors to be activated in these methods are the M1, M2, and M4 receptors, since these receptors cause the strongest effect in inhibiting release of pro-inflammatory cytokines. See Example 2. Thus, in embodiments that utilize a muscarinic agonist to activate the muscarinic receptor, one that activates the M1, M2, and/or M4 receptors are particularly preferred.
Nonlimiting examples of preferred muscarinic agonists useful for these methods include muscarine, McN-A-343, and MT-3. In one embodiment, the muscarinic agonist is not N,N'-bis(3,5-diacetylphenyl) decanediamide tetrakis (amidinohydrazone) tetrahydrochloride (CNI-1493). In another embodiment, the muscarinic agonist is not a CNI-1493 compound. As used herein, "a CNI-1493 compound" means an aromatic guanylhydrazone ("Ghy", more properly termed amidinohydrazone, i.e., NHZ(CNH(-NH=) compound having the formula:
x, ~ ~ / x,, x ~ ~ ~x~

wherein XZ =GhyCH-, GhyCCH3- or H-; Xl, X'1 and X'Z independently=GhyCH- or GhyCCH3-; Z=-NH(CO)NH-, -(C6Hd)-, -(CSNH3)- or -A-(CH2)ri A-, n=2-10, which is unsubstituted, mono- or di-C-methyl substituted, or a mono or di- unsaturated derivative thereof; and A, independently,= -NH(CO)-, -NH(CO)NH-, -NH- or -O-and salts thereof. GhyCH- = NHZ(CNH)-NH-N=CH-, and GhyCCH3- _ NHZ(CNH)-NH-N=CCH3-. A preferred embodiment includes those compounds wherein A is a single functionality. Also included are compounds having the same formula wherein Xl and XZ =H; X'1 and X'2 independently=GhyCH- or GhyCCH3 -;

Z=-A-(CHz)n A-, n=3-8; and A=-NH(CO)- or -NH(CO)NH-, and salts thereof. Also included are compounds wherein Xl and Xz =H; X'1 and X'z independently=GhyCH-or GhyCCH3 - and Z=O-(CHz)Z-O-.
Further examples of CNT-1493 compounds include: compounds of the above formula wherein: Xz =GhyCH-, GhyCCH3- or H-; Xl, X'1 and X'z =GhyCH- or GhyCCH3-; and Z=-O-(CHZ)n -O-, n=2-10 and salts thereof; and the related compounds wherein, when XZ is other than H, XZ is meta or para to Xl and wherein X'Z is meta or para to X'1. A compound having the above formula wherein: XZ
=GhyCH, GhyCCH3 or H; Xl, X'1 and X'2, =GhyCH- or GhyCCH3-; and Z=-NH-(C=O)-NH- and salts thereof; and the related genus wherein, when XZ is other than H, XZ is meta or para to XI and wherein X'2 is meta or para to X'1.
A "CNI-1493 compound" also means an aromatic guanylhydrazone compound having the formula:
X~1 ~X1 iCH2)m1 A
A (CH2)ms Z2 X3 (CH2)m2 A
~/ X.2 wherein, X,, XZ and X3, independently=GhyCH- or GhyCCH3-; X'1, XZ and X'3, independently=H, GhyCH- or GhyCCH3-; Z=(C6H3), when ml, m2, m3 0 or Z N, when, independently, ml, mz, m3=2-6; and A=-NH(CO)-, -NH(CO)NH-, -NH- or -O-and salts thereof. Further examples of CNI-1493 include the genus wherein when any of X'1, XZ and X'3 are other than H, then the corresponding substituent of the group consisting of Xl, Xz and X3 is meta or para to X'1, XZ and X'3, respectively; the genus wherein, ml, m2, m3 0 and A=-NH(CO)-; and the genus wherein ml, m2, m3=2-6 and A=-NH(CO)NH-. Examples of CNI-1493 and methods for mal~ing such compounds are described in U.S. Patent No. 5,854,289 (the teachings of which are incorporared herein by reference). In a preferred embodiment, the CNI-1493 compound is N,N'-bis(3,5-diacetylphenyl) decanediamide tetrakis (amidinohydrazone) tetrahydrochloride (also known as CNI-1493), which can be made by combining N,N'-bis(3,5-diacetylphenyl)decanediamide (0.65 g), aminoguanidine hydrochloride (0.691 g), and aminoguanidine dihydrochloride (0.01 g) and heating in 91% ethanol (5.5 mL) for 18 hr, followed by cooling and filtration.
The synthesis results in a compound having a melting point of 323°C-324°C. The composition can be formulated in a physiologically acceptable carrier.
Activation of brain muscarinic receptors can thus be achieved by treatment with a muscarinic agonist. As used herein, a muscarinic agonist is an agonist that can bind to a muscarine receptor. In an embodiment, the muscarinic agonist can bind to other receptor types) in addition to the muscarine receptor, for example, another cholinergic receptor. An example of such a muscarinic agonist is acetylcholine. In another embodiment, the muscarinic agonist binds muscarine receptors) with greater affinity than other cholinergic receptors, e.g., nicotinic receptors (e.g., with at least 10% greater affinity, 20% greater affinity 50%
greater affinity, 75% greater affinity 90% greater affinity or 95% greater affinity).
In one embodiment the muscarinic agonist is selective for an M1, M2, or M4 receptor.
As used herein, an agonist that is "selective" for an M1, M2, or M4 receptor is an agonist that binds to an M1, M2, and/or M4 receptor with greater affinity than it binds to one, two, or more other receptors, for example, one or more other muscarinic receptors (e.g., M3 or MS muscarinic receptors), or one or more other cholinergic receptors. In an embodiment, the agonist binds with at least 10%
greater affinity, 20% greater affinity 50% greater affinity, 75% greater affinity 90%
greater affinity or 95% greater affinity than it binds to receptors other than an M1, M2, and/or M4 receptor. Binding affinities can be determined as described herein or using other receptor binding assays known to one of shill in the art. In one embodiment, the brain muscarinic receptor is activated with a sufficient amount of muscarinic agonist or at a sufficient level to inhibit release of a pro-inflammatory cytokine from a vertebrate cell.
The muscarinic agonist can be administered to the brain muscaxinic receptors by intracerebroventricular injection. Alternatively, the muscarinic agonist can be administered orally, paxenterally, intranasally, vaginally, rectally, lingually, sublingually, bucally, intrabuccaly, or transdermally to the patient, provided the muscaxinic agonist can cross the blood-brain barner.
The route of administration of the muscarinic agonist can depend on the condition to be treated. For example, intravenous inj ection may be preferred for treatment of a systemic disorder such as septic shock, and oral administration may be preferred to treat a gastrointestinal disorder such as a gastric ulcer. The route of administration and the dosage of the cholinergic agonist to be administered can be determined by the skilled artisan without undue experimentation in conjunction with standard dose-response studies. Relevant circumstances to be considered in making those determinations include the condition or conditions to be treated, the choice of composition to be administered, the age, weight, and response of the individual patient, and the severity of the patient's symptoms.
Muscarinic agonist compositions useful for the present invention can be administered parenterally such as, for example, by intravenous, intramuscular, intrathecal, or subcutaneous injection. Parenteral administration can be accomplished by incorporating the muscarinic agonist compositions of the present invention into a solution or suspension. Such solutions or suspensions may also include sterile diluents such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol, or other synthetic solvents.
Parenteral formulations may also include antibacterial agents such as, for example, benzyl alcohol, or methyl parabens, antioxidants such as, for example, ascorbic acid or sodium bisulfate and chelating agents such as EDTA. Buffers such as acetates, citrates, or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose may also be added. The parenteral preparation can be enclosed in aanpules, disposable syringes, or multiple dose vials made of glass or plastic.
Rectal administration includes administering the pharmaceutical compositions into the rectum or large intestine. This can be accomplished using suppositories or enemas. Suppository formulations can be made by methods known in the art. For example, suppository formulations can be prepared by heating glycerin to about 120° C, dissolving the cholinergic agonist in the glycerin, mixing the heated glycerin after which purifaed water may be added, and pouring the hot mixture into a suppository mold.
Transdermal administration includes percutaneous absorption of the cholinergic agonist through the skin. Transdermal formulations include patches, ointments, creams, gels, salves, and the like.
The present invention includes nasally administering to the vertebrate a therapeutically effective amount of the muscarinic agonist. As used herein, nasal administration includes administering the cholinergic agonist to the mucous membranes of the nasal passage or nasal cavity of the patient. As used herein, pharmaceutical compositions for nasal administration of a cholinergic agonist include therapeutically effective amounts of the agonist prepared by well-known methods to be administered, for example, as a nasal spray, nasal drop, suspension, gel, ointment, cream, or powder. Administration of the cholinergic agonist may also take place using a nasal tampon, or nasal sponge.
Accordingly, muscarinic agonist compositions designed for oral, lingual, sublingual, buccal and intrabuccal administration can be made without undue experimentation by means well known in the art, for example, with an inert diluent or with an edible carrier. The compositions may be enclosed in gelatin capsules or compressed into tablets. For the puapose of oral therapeutic administration, the pharmaceutical compositions of the present invention may be incorporated with excipients and used in the form of tablets, troches, capsules, elixirs, suspensions, syrups, wafers, chewing gums, and the like.
Tablets, pills, capsules, troches, and the like may also contain binders, recipients, disintegrating agent, lubricants, sweetening agents, and flavoring agents.
Some examples of binders include microcrystalline cellulose, gum tragacanth, or gelatin. Examples of excipients include starch or lactose. Some examples of disintegrating agents include alginic acid, com starch, and the like. Examples of lubricants include magnesium stearate or potassium stearate. An example of a glidant is colloidal silicon dioxide. Some examples of sweetening agents include sucrose, saccharin, and the like. Examples of flavoring agents include peppermint, methyl salicylate, orange flavoring, and the like. Materials used in preparing these various compositions should be pharmaceutically pure and nontoxic in the amounts used.
As previously discussed, the effect of activation of a brain muscarinic receptor on inhibiting the release of pro-inflammatory cytokines in the periphery is established herein to be dependent on an intact vagus nerve. Without being limited to any particular mechanism, the inventors believe that brain muscarinic receptor activation stimulates the vagus nerve pathway, and this stimulation causes the inhibition of pro-inflammatory cytolcine release. This stimulation of the brain vagus nerve pathway is "upstream" in the vagus nerve pathway from the previously established effect of stimulation of peripheral vagus nerves on inhibiting pro-inflammatory cytokine release (Borovilcova et al., 2000a; see also IJ.S.
Patent Application 09/855,446). Based on the determination that an intact vagus pathway is required for the inhibition of pro-inflammatory cytolcine release effected by brain muscarinic agonist activation, as established herein, it is clear that pro-inflammatory cytol~ines can be inhibited by directly stimulating a vagus nerve pathway in the brain. In one embodiment, the vagus nerve pathway is stimulated at a sufficient level to inhibit release of a pro-inflammatory cytol~ine from a vertebrate cell.
Accordingly, some embodiments of the present invention are directed to methods of inhibiting release of a pro-inflammatory cytokine in a vertebrate.
The methods comprise directly stimulating the vagus nerve pathway in the brain of the vertebrate. In these methods the vagus nerve pathway can be stimulated by any known method. Nonlimiting examples include mechanical means such as a needle, ultrasound, or vibration; pharmacological or chemical stimulation, any electromagnetic radiation such as infrared, visible or ultraviolet light;
heat, or any other energy source. In preferred embodiments, the vagus nerve is stimulated electrically, for example, with a commercial deep brain stimulator, such as the Medtronic SOLETRA device, which is currently in use for the treatment of Parlcinson's disease, etc. In preferred embodiments, the vagus nerve pathway is stimulated electrically.
These methods have the same effect on inhibiting the production of pro-inflammatory cytokines as the previously described methods of activating brain muscarinic receptors, i.e., would inhibit the same pro-inflammatory cytolcines, would reduce inflammation in patients with the same inflammatory conditions, and would inhibit the release of pro-inflammatory cytokines from the brain or any peripheral organ or cell served by vagus nerve pathways, for example, the liver or cardiac myocytes.
As previously discussed, activation of brain muscarinic receptors inhibit the release of pro-inflarninatory cytokines. By inhibiting the release of pro-inflammatory cytokines, inflammation can be reduced in diseases that are characterized by inflammation mediated by a pro-inflammatory cytolcine cascade.
Accordingly, the present invention is directed to methods of treating an inflammatory disease in a vertebrate. The methods comprise activating a brain muscarinic receptor in the vertebrate. The methods are useful for treating any disease in any vertebrate, including humans, that is at least partially mediated by a pro-inflammatory cytokine cascade, including systemic inflammatory diseases.
Examples of such diseases have been previously provided. Even though the signal that inhibits the release of pro-inflammatory cytokines is apparently carried by the vagus nerve, these methods are effective in inhibiting systemic inflammatory diseases because the vagus nerve innervates the liver, which is a primary source of pro-inflammatory cytokines in systemic disease.

As previously discussed, the same effect as achieved by activating a muscarinic receptor is also achieved by directly stimulating a vagus nerve pathway in the brain. Thus, the invention is also directed to methods of treating an inflammatory disease in a vertebrate, the methods comprising directly stimulating a vagus nerve pathway in the brain of the vertebrate. As previously discussed, the vagus nerve pathway can be stimulated by any means known in the art, and is useful for treating any inflammatory disease in any vertebrate (including humans) that is at least partially mediated by an inflammatory cytokine cascade.
Since the vagus nerve serves the heart, and since cytolcine release is at least partially responsible for myocyte apoptosis in several inflammatory diseases, it is also contemplated that apoptosis of cardiac myocytes can be inhibited in vertebrates, including humans, at risk for cardiac myocyte apoptosis by methods comprising activating a brain muscarinic receptor in the vertebrate. Preferred muscarinic receptors are M1, M2, and M4 receptors. Inflammatory diseases that could be treated by these methods include vasculitis, angiitis, endocarditis, pericarditis, myocarditis, myocardial ischemia, periarteritis nodosa, rheumatic fever, congestive heart failure, adult respiratory distress syndrome, fasciitis, or graft-versus-host disease. As with previously described methods, the brain muscarinic receptor can be activated by administering a muscarinic agonist to the vertebrate, either directly to the brain of the vertebrate, enterically or parenterally. Preferred muscarinic agonists are muscarine, McN-A-343 and MT-3.
Similarly, apoptosis in cardiac myocytes can be inhibited by directly stimulating a vagus nerve pathway in the brain of the vertebrate, for example, electrically.
It has also been discovered that vertebrates can be conditioned to inhibit the release of a pro-inflammatory cytokine by associating the activation of brain muscarinic receptors with a sensory stimulus. Thus, in some embodiments, the invention is directed to methods of conditioning a vertebrate to inhibit the release of a pro-inflammatory cytokine upon experiencing a sensory stimulus. These methods comprise the following steps:

(a) activating a brain muscarinic receptor in the vertebrate and providing the sensory stimulus to the vertebrate within a time period sufficient to create an association between the stimulus and the activation of the brain muscarinic receptor;
and (b) repeating step (a) at sufficient time intervals and duration to reinforce the association sufficiently for the pro-inflammatory cytokine release to be inhibited by the sensory stimulus alone.
These methods are particularly useful for treating chronic inflammatory conditions, such as arthritic conditions, where the methods allow a patient to reduce the need for anti-inflammatory medication. Thus, potential side effects of anti-inflammatory medication, such as gastrointestinal, kidney, heart, or liver effects, can be reduced.
These methods can be used to reduce the release of any of the pro-inflammatory cytokines as with the methods previously discussed, including tumor necrosis factor (TNF), interleukin (IL)-1(3, IL-6, IL-18, HMG-B1, MIP-la, MIP-1~3, M1F, interferon-'y, and PAF. In particular, pro-inflammatory cytolcine release is inhibited in any organ, tissue, or cell subject to influence by vagus nerve stimulation, including the liver and cardiac myocytes. They are useful for any vertebrate having a vagus nerve, including all mammals. They are particularly useful for vertebrates (including humans) suffering from, or at risk for, a condition mediated by an inflammatory cytokine cascade. Examples of such conditions have been previously discussed.
hi the conditioning step of these methods (step (a)), the brain muscarinic receptor can be activated by any means previously discussed. It is believed that the association between the stimulus and the brain muscarinic receptor activation is most effectively created if the stimulus and activation is as close together temporally as possible, preferably within one minute. The time interval between repetitions of the stimulus-activation procedures should also be short enough to optimize the reinforcement of the association. A preferred time interval is twice daily:
The duration of the conditioning should also be sufficient to provide optimum reinforcement of the association. A preferred duration is at least one week.

Optimum time intervals and durations can be determined by the spilled artisan without undue experimentation by standard methods known in the art.
The sensory stimulus can be from any of the five senses. Nonlimiting examples of suitable sensory stimuli are sounds such as a bell ring, a buzzer, and a musical passage; a touch such as a pin stick, a feather touch, and an electric shock; a taste, or the ingestion of a particular chemical, such as a sweet taste, a sour taste, a salty taste, and saccharine ingestion; a visual image such as a still picture, a playing card, or a short video presentation.
As with previously described methods, the conditioning to inhibit pro-inflammatory cytolcine release with a sensory stimulus can utilize stimulation of a vagus nerve pathway in the vertebrate brain rather than activation of brain muscarinic receptors.
Additionally, since inhibiting pro-inflammatory cytolcine release also effects a reduction in inflammation, as discussed above, the conditioning methods described above are useful for reducing inflammation in the treated vertebrate. Thus, the present invention is directed to methods of conditioning a vertebrate to reduce inflammation in the vertebrate upon experiencing a sensory stimulus. The methods comprise the following steps:
(a) activating a brain muscarinic receptor in the vertebrate, or directly stimulating a vagus nerve pathway in the brain, and providing the sensory stimulus to the vertebrate within a time period sufficient to create an association between the stimulus and the activation of the brain muscarinic receptor; and (b) repeating step (a) at sufficient time intervals and duration to reinforce the association sufficiently for the inflammation to be reduced by the sensory stimulus alone.
Preferred embodiments of the invention are described in the following examples.

Example 1 This example describes experiments establishing that CNI-1493 binds to brain muscarinic receptors, that intracerebroventricular (i.c.v.) injections of CNI
suppresses carrageenan-induced hindpaw edema and release of TNF into the blood, that these effects are reversed by atropine, and that neither nicotine nor prozalc i.c.v.
injections inhibits TNF production.
Methods Method of determining CNI-1493 receptor binding-. CNI-1493 was tested at a single concentration (10 ~,M) in a panel of receptor binding assays by NovaScreen Biosciences Corporation (Hanover, MD). Values were expressed as the percent inhibition of specific binding, and represented the average of duplicate tubes.
Method of stereotactic intracerebroventricular injections. A rat model of intracerebroventricular (i.c.v.) injections was established in order to be able to directly deliver pharmacological agents into the brain of rats. This was necessary in order to separate drug effects on peripheral inflammation that occurred through central versus peripheral mechanisms. Lewis rats were anaesthetized with urethane (1 glkg, i.p.) and xylazine (15 mg/rat, i.m. (intramuscular)). Rats were then placed in a stereotactic head frame (Stoelting, Wood Dale, IL, USA). The incisor bar was adjusted until the plane defined by the lambda and bregma was parallel to the base plate. For i.c.v. injections the needle of a Hamilton syringe (25 ~,1) was positioned stereotactically above the lateral ventricle (0.2 mm and 1.5 mm posterior to bregma, 3.2 mm below the dura.) Solutions of the drugs tested were prepared in sterile endotoxin-free water, at the specified concentrations, and a 10-~,l injection/rat was administered over 2 min, 1 h prior to either carrageenan injection, or to LPS.
The tested drugs, in either the carrageenan and/or LPS experiments, were:
saline control; fluoxetine hydrochloride, (also known as Prozak) (0.01 mg/100 g);
muscarine (50 ~,g/rat, 5 ~,g/rat, 0.5 ~g/rat, 0.05 ~,g/rat, 0.005 ~,g/rat); 4-(N-[3-chlorophenyl]carbamoyloxy)-2-butynyltrimethylammonium chloride (also known as McN-A-343) (5 ~.g/rat); Muscarinic Toxin-3, (also known as MT-3) from Dendroaspis an ug sticeps snake venom (0.37 ~,g/rat); nicotine (10 fig/ rat);

(1 ~,g/kg, 50 ~,g/rat); atropine (1 ~,g/kg, 5 ~,glrat); CNT-1493 plus atropine (1 ~,g/kg of each of the drugs; 50 ~,g/rat, 5 ~g/rat respectively); naloxone hydrochloride (2 ~,g/rat), CNI-1493 plus naloxone (50 ~,g/rat + 5 wg/rat respectively); and morplune (20 ~g/rat).
Method of carrageenan-induced hindpaw edema. Paw edema was induced in anaesthetized rats by injection of 1% solution of 1-carrageenan (100 ~,1) into the plantar surface of the left hindpaw. The right hindpaw was injected with the same volume of saline alone (as control). The thickness of the carrageenan-treated and saline-treated hindpaw was measured using a caliper at 3 h post carrageenan, and the difference between paw thickness calculated as an index of inflammation (paw swelling).
Method of LPS infections and TNF determination. LPS (15 mg/kg, i.v.) was injected in the tail vein 1 h after drug injection. Blood was obtained 2 h post LPS
injection by paraorbital bleeding. Serum TNF concentrations were determined by an L929 bioactivity assay.
Method of assessing TNF by the L929 bioactivit.~~ L929 cells were suspended in Dulbecco's minimal Eagle's medium (DMEM; GibcoBRL) supplemented with fetal bovine serum (10%; Hyclone) and penicillin/ streptomycin (0.5%; Sigma Chemical Co.), and plated at 2 x 104 cells per well in 96-well flat-bottomed microtiter plates. After 24 h, media were respirated and replaced with medium containing cycloheximide (10 ~,g/ml; Sigma Chemical Co.) and the samples to be assayed/ TNF standards. Plates were incubated overnight, at which time cell viability as a function of TNF concentration was assessed by the MTT assay.
Absorbance values were converted to units per milliliter by comparison with a standard curve for rat TNF.

_27_ Results When tested with an in vitro panel of receptor binding assays, CNI-1493 at ~,M inhibited receptor binding by greater than 50% for seven different receptors, respectively alpha 1 adrenergic (89.7%), muscarinic (60.6%), serotonin (75.6%), 5 Type N calcium channel (84.2%), voltage-insensitive potassium channel (60.2%), voltage-sensitive potassium channel (73.0%), and vasoactive intestinal peptide (58.5%).
CNI-1493 at 10 ~,M inhibited receptor binding by less than 50% (considered by NovaScreen to be indicative of marginal or no activity) at the following receptors:
10 beta adrenergic, dopamine, glutamate (NMDA agoust site), H1 histamine, Type L
calcium channel, chloride channel, site 1 sodium, site 2 sodium, NKl neurolcinin, vasopressin l, leukotriene D4 and LTD4, thromboxane A2, and epidermal growth factor.
The above-described studies provided a list of receptors to be tested for determination as to whether their alternative pharmacological activation by other drugs would separately cause peripheral immunosuppressive activity, and whether tlus activity would be further dependent on the efferent vagus nerve. To achieve this purpose, we established an animal model of paw edema and an animal model of endotoxic shock, where the effects of the various drugs were tested by their stereotactic intracerebroventricular delivery into the brain.
In one set of experiments, rats were injected by i.c.v. means with either saline (n=1), CNI-1493 (5 ~,g/rat, n=3),, CNI-1493 plus atropine (5 ~,g/rat each), or atropine (5 ~,g/rat). LPS (15 mg/kg, i.v.) was given 1 h later. Blood was collected 2 h post LPS administration. Serum TNF was determined by the L929 assay.
The results of these experiments are summarized in Figure 1.
W tracerebroventicularly administered CNI-1493 inhibited LPS-induced serum TNF
levels by more than 80%. Atropine reversed the inhibitory effect of CNI-1493 to the TNF level of atropine alone.
These results indicate that i.c.v. CNI-1493 can suppress peripheral inflammation, and that this effect is reversed by co-administration of i.c.v.
atropine.
Since atropine is an antagonist at muscarinic receptors, these results thus indicate that the immunosuppressive effects of CNI-1493 are mediated via muscarinic receptors in the brain.
In a second set of experiments, rats were injected by i.c.v. means with either saline (n=4), nicotine (10 ~,g/rat, n=3), or prozak (0.01 mg/100g, n=3). LPS
(15 mg/kg, i.v.) was given 1 h later. Blood was collected 2 h post LPS
administration.
Serum TNF was determined by the L929 assay.
The results are summarized in Figure 2. Neither nicotine nor prozal~ had any effect in reducing LPS-induced serum TNF levels. These results indicate that neither nicotine nor prozak show central effects on peripheral immmiosuppression.
In a third set of experiments, rats were injected by i.c.v. means with either saline (n=4), CNI-1493 (5 ~g/rat, n=3), CNI-1493 plus atropine (5 ~,g/rat each), or atropine (5 ~,g/rat). Carrageenan was given to the animals 1 h later, and paw edema was determined 3 h post carrageenan.
The results of these experiments are summarized in Figure 3. As with LPS
induced serum TNF levels, intracerebroventricular administration of CNI-1493 significantly inhibits carageenan-induced paw edema, and atropine (ATR) reverses the effect.
These results indicate again, by a different method, that i.c.v. CNI-1493 suppresses peripheral inflammation, and that this effect is reversed by co-administration of i.c.v. atropine. Since atropine is an antagoust at muscarinic receptors, these results thus indicate that the immunosuppressive effects of CNI-1493 are mediated via muscarinic receptors in the brain.
In another set of experiments, rats were injected by i.c.v. means with either saline, or muscarine (from left to right on the bar graph- 5 ~,g/rat, 0.5 ~,g/rat, 0.05 ~,g/rat, 0.005 ~,g/rat, n=4 animals/group). Carrageenan was given to the animals 1 h later, and paw edema was determined 3 h post carrageenan.
Figure 4 summarizes the results of these experiments.
Intracerebroventricular administration of muscarine significantly inhibits carrageenan-induced paw edema in a dose-dependent manner. These results further establish that i.c.v. muscarine produces peripheral suppression of inflammation.

In other experiments, rats were subj ected to bilateral cervical vagotomy (VGX) or alternatively to bilateral vagus nerve isolation.
Intracerebroventriculax injections were then performed (26-66 min. later) in each of the four groups of either saline (SAL, n=2 animals/group), or muscarine (MUS, 0.5 ~,g/rat, n=4 animals/group). Carrageenan was given to the animals 1 h post the i.c.v. drug injections, and paw edema was determined 3 h post carrageenan. P=0.015 SAL v.
MUS. P=0.039 MUS v. MUS-VGX.
Figure 5 summarizes the results of these experiments. Vagotomy clearly abrogates the inhibitory effects of intracerebroventricular (i.c.v.) administration~of muscarine on carrageenan-induced paw edema. Thus, vagotomy abrogates the peripheral immunosuppressive effects of centrally administered muscaxine, establishing that activation of muscarinic receptors in the brain carries a peripheral immunosuppressive signal through the vagus nerve.
Example 2 This example provides experimental results establishing the preferred muscarinic receptor subtypes useful for the present invention.
Methods Method of determining muscarinic receptor subtxpe. CNI-1493 was tested at a single concentration (10 ~,M) in a panel of muscarinic receptor binding assays by NovaScreen Biosciences Corporation (Hanover, MD). Values were expressed as the percent inhibition of specific binding, and represented the average of duplicate tubes.
Other methods are as described in Example 1.
Results Table 1 summarizes the results of testing of CNI-1493 for inhibiting binding to a panel of muscarinic receptors as indicated.

Receptor Percent inhibition Muscarinic, M1 83%
Muscarinic, Ml (Human recombinant) 72%
Muscarinic, M2 85%
Muscarinic, M2 (Human recombinant) 58%
Muscarinic, M3 9%
Muscarinic, M3 (Human recombinant) 40%

Muscarinic, M4 (Human recombinant) 57%

Muscarinic, MS (Human recombinant)43%

Values of less than 50% are considered by NovaScreen to show marginal or no activity.
This results indicate that M1, M2, and M4 are the primary muscarinic receptors that bind to CNI-1493.
In another set of experiments, animals were injected by i.c.v. as described in Example 1 with either saline, the M1 agonist McN-A-343 (5 ~,g/rat, n=5), or the M4-agonist MT-3 (0.37 ~,g/rat, n=4). Carrageenan was given to the animals 1 h later as described in Example 1, and paw edema was determined 3 h post carrageenan administration.
The results of these experiments are provided in Figure 6.
Intracerebroventricular administration of the 1VI1 agonist McN-A-343 or the M4 agonist MT-3 significantly inhibits carrageenan-induced paw edema. These results further establish that central activation of M1 and M4 receptors plays a role in suppressing peripheral immune processes.
In other experiments, animals were injected i.c.v. with either saline, or the Ml agonist McN-A-343 at 5 ~g/rat (n=5). Alternatively, McN-A-343 was given peripherally at a much higher concentration (5 mg/kg, i.p., n=2). Carrageenan was given to the animals 1 h post i.c.v. or i.p. drug administration, and paw edema was determined 3 h post carrageenan.

Results of these experiments are summarized in Figure 7.
Intracerebroventricular (i.c.v.) administration of the M1 agonist McN-A-343 has a comparable effect on inhibition of carrageenan-induced paw edema as a higher dose administered intraperitoneally (i.p.). These results indicate that the significantly higher i.p. concentration of an M1 agonist that is needed to achieve peripheral immunosuppression is attributable to a small degree of blood brain barner penetration of this compound. Thus, it is likely that the small amount of centrally penetrated compound that is responsible for the observed immunosuppressive effects of the drug.
Example 3 This Example provides experimental results that indicate that mammals can be conditioned to mount an anti-inflammatory response through a sensory stimulus that has been associated with activation of brain muscarinic receptors.
Methods Mice were grouped into four groups (n=4 animals/group). The conditioning training for Groups 2-4 consisted of morning and afternoon sessions. Mice in group 2 were together taken to a room, where each mouse was injected with CNI-1493 (2.5 mg/kg, i.p.). Simultaneously with the injection, each mouse was subjected to seconds of bell ringing. Group 4 mice, similar to Group 2 mice, were subj ected to control conditioning, whereby nuce were injected with saline, instead of CNI-1493.
Group 3 mice, like Group 2 mice, were subjected to saline injections but not bell ringing. This protocol was performed over a 10 day period, on days 1-4 and 8-10.
On day 11, Group 1 mice were injected with CNI-1493 (2.5 mg/kg, i.p.). Also on day 11, 30 min after the Group 1 mice injections were performed, animals in all groups were injected with LPS (5 mg/kg, i.p.). After 2 hours, the mice were euthaiuzed via COZ inhalation, and blood was withdrawn. Serum TNF was determined by the L929 assay.

Results The results of this experiment are summarized in Figure 8. The mean LPS-induced TNF release was reduced by about 60% in animals conditioned by associating repeated intraperitoneal CNI-1493 administration with bell ringing vs.
animals exposed to bell ringing and intraperitoneal saline injections (Group 2 vs.
Group 4; p=0.22) On the basis of these experiments, immunosuppression mediated via stimulation of the efferent vagus nerve can be expected to be achieved by conditioned exposure to a neutral stimulus (i.e., bell) following conditioning training with a neutral stimulus and a drug known to activate brain muscarinic receptors (here, CIVI-1493).
Example 4 This Example provides experimental results that indicate that intracerebroventricular administration of muscarine into rats causes a dose-dependent decrease in serum, spleen, and heart TNF concentrations.
Methods Methods of stereotactic intracerebroventricular injection of muscarine into rats and LPS injections were as described in Example 1. TNF levels in serum and tissues were determined using an enzyme-inked immunosorbent assay (ELISA) according to the manufacturere's instructions (R & D Systems (Minneapolis, Minnesota)).
Results Rats were injected by i.c.v. means with either saline (control) or muscarine (0.005 wglkg body weight, 0.5 wg/kg~~body weight, 5.0 ~,g/kg body weight, or ~,g/kg body weight). LPS was administered 1 hour later. Two hours after LPS
administration the rats were sacrificed and blood, heart tissue, and spleen tissue were isolated from the rats. The results of these experiments are summarized in Figures 9A-9C. As shown in Figures 9A-9C, i.c.v, administration of muscarine inhibited LPS-induced serum, heart, and spleen (peripheral) TNF levels. These results demonstrate that peripheral TNF production can be inhibited by the activation of central muscarinic receptors.
Example 5 This Example provides experimental results that indicate that intravenous administration of muscarine into rats has no effect on rat spleen, liver, and heart TNF concentrations.
Methods Methods of LPS injections were as described in Example 1. Determination of serum and tissue TNF levels were as described in Example 4. Muscarine (or control saline) was intravenously injected into rats at concentrations of 0.05 ~,g/kg body weight, 0.5 ~g/kg body weight, or S.0 ~,g/kg body weight.
Results Rats were injected by i.v. means with either saline (control) or muscarine (0.05 ~,g/kg body weight, 0.5 ~,g/kg body weight, or 5.0 ~,g/kg body weight).
LPS
was administered 1 hour later. Two hours after LPS administration the rats were sacrificed and blood, liver tissue, heart tissue, and spleen tissue were isolated from the rats and assayed for TNF concentrations. The results of these experiments are summarized in Figures l0A-l OD. As shown in Figures l0A-l OD, intravenous administration of muscarine had no effect on LPS-induced serum, liver, heart, and spleen TNF levels.
Muscarine is a quarternary salt, and as such it does not readily cross the blood brain barrier. The above results demonstrate that the activation of peripheral muscarinic receptors has no effect on LPS induced TNF production.
In view of the above, it will be seen that the several advantages of the invention are achieved and other advantages attained.
As various changes could be made in the above methods and compositions without departing from the scope of the invention, it is intended that all matter contained in the above description and shown in the accompanying drawings shall be interpreted as illustrative and not in a limiting sense.
All references cited in this specification are incorporated herein by reference.
The discussion of the references herein is intended merely to summarize the assertions made by the authors and no admission is made that any reference constitutes prior art. Applicants reserve the right to challenge the accuracy and pertinence of the cited references.
While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those spilled in the art that various changes in fonn and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

References cited Antonica, A., et al., J. Auton. Nerv. Syst., 48:187-97, 1994.
Bhattacharya S.K., et al., Res. Exp. Med. 191:65-76, 1991.
Besedovslcy, H., et al., Science, 233:652-54, 1986.
Blackwell, T.S., and Christman, J.W., Br. J. Anaestla., 77: 110-17, 1996.
Blum, A. and Miller, H., Am. Heart J., 135:181-86, 1998.
Borovikova, L.V., et al., Nature, 405: 458-62, 2000a.
Borovilcova, L.V., et al., Auton. Neurosci., 20:141-47, 2000b.
Bumgardner, G.L., and Orosz, C.G., Senain. LiverDis., 19: 189-204, 1999.
Carteron, N.L., Mol. Med. Today, 6:315-23, 2000.
Dibbs, Z., et al., Proc. Assoc. Am. Physicians, 111:423-28, 1999.
Dinarello, C.A., FASEB J., 8:1314-25, 1994.
Fleshner, M., et al., J. Neuroinznaunol., 86:134-41, 1998.
Fox, D.A., Arch. Intern. Med., 28:437-444, 2000.
Gattorno, M., et al., J. Rheumatol., 27:2251-2255, 2000.
Gaykema, R.P., et al., Endocrinology, 136:4717-4720, 1995.
Gracie, J.A., et al., J. Clip. Invest., 104:1393-1401, 1999.
Green, E., Psycophysiology, 6, 371-377, 1969.
Gregory, S.H. and Wing, E.J., Immunology Today, 19:507-10, 1998.
Guslandi, M., Br. J. Clin. Pharmacol., 48:481-84, 1999.
Hirano, T., J. Surg. Res., 81:224-29, 1999.
Homrnes, D.W. and van Deventer, S.J., Curr. Opin. Clin. Nutr. Metab.
Care, 3:191-95, 2000.
Hsu, H.Y., et al., J. Pediatr. Gastroenterol., 29:540-45, 1999.
Hu, X.X., et al., J. Neuroimnaunol., 31:35-42, 1991.
Dander, S. and Stoll, G., J. Neuroimmunol., 114:253-58, 2001.
Kanai, T. et al., Digestion, 63 Suppl. 1:37-42, 2001.
Katagiri, M., et al., J. Clin, Gastroenterol., 25 Suppl. 1: 5211-14, 1997.
Kimmings, A.N., et al., Eur. J. Surg., 166:700-OS, 2000.
Kumins, N.H., et al., SHOCK, 5:385-88, 1996.
Lee, H.G., et al., Clin. Exp. Immunol., 100:139-44, 1995.

Lipton, J.M. and Catania, A., Immunol. Today, 18:140-45, 1997.
Madretsma, G.S., et al., Immunopha~macology, 35:47-51, 1996.
McGuinness, P.H., et al., Gut, 46:260-69, 2000.
Nathan, C.F., J. Clin. Invest., 79:319-26, 1987.
Pulkl~i, K.J., Ann. Med., 29:339-43, 1997.
Prystowslcy, J.B. and Rege, R.V., J. Sung. Res., 71;123-26 1997.
Rayner, S.A. et al., Clira. Exp. Inamunol., 122:109-16, 2000.
Romanovslcy, A.A., et al., Am. J. Physiol., 273:8407-13, 1997.
Sandborn, W.J., et al., Ann. Intel n. Med, 126:364-71, 1997.
Sato, E., et al., Am. J. Physiol., 274:L970-79, 1998.
Sato, K.Z., et al., Neu~osci. Lett., 266:17-20, 1999.
Scheinman, R.L, et al., Science, 270:283-86, 1995.
Sher, M.E., et al., Inflamrn. Bowel Dis., 5:73-78, 1999.
Sternberg, E.M., J. Clin. Invest., 100:2641-47, 1997.
Thompson, A., Ed. The Cytokine Handbook, 3'''~ ed., Academic Press, 1998.
Tracey, K.J. et al., Nature, 330:662-64, 1987.
Tracey, K.J. et al., ScieyZCe, 234:470-74, 1986.
Tsutsui, H., et al., Imrraunol. Rev., 174:192-209, 2000.
van Dijk, A.P., et al., Eu~. J. Clin. Invest., 28:664-71, 1998.
Wang, H., et al., Science, 285:248-51, 1999.
Waserman, S., et al., Can. Respir. J., 7:229-37, 2000.
Watanabe, H. et al., J. Reconstr. Microsuyg., 13:193-97, 1997.
Wathey, J.C., et al., Biophys. J., 27:145-64, 1979.
Watkins, L.R. and Maier, S.F., P~oc. Natl. Acad. Sci. U. .S. A., 96:7710-13, 1999.
Watkins L.R., et al., Neurosci. Lett. 183:27-31, 1995.
Whaley, K., et al., Nature, 293:580-83, 1981.
Woiciechowsky, C., et al., Nat. Med., 4: 808-13, 1998.
Yeh, S.S., and Schuster, M.W., Am. J. Clin. Nutn., 70, 183-97, 1999.
Zhang and Tracey, in The Cytokine Handbook, 3''~ ed., Ed. Thompson, Academic Press, 515-47, 1998.

PCT patent publication WO 00/47104.

Claims (21)

What is claimed is:
1. A method of inhibiting release of a pro-inflammatory cytokine in a vertebrate, at risk for or having a condition mediated by an inflammatory cytokine cascade, the method comprising activating a brain muscarinic receptor in the vertebrate.
2. The method of Claim 1, wherein the pro-inflammatory cytokine is selected from the group consisting of tumor necrosis factor (TNF), interleukin (IL)-1.alpha., IL,-1.beta., IL-6, IL-18, HMG-B1, MIP-1.alpha., MIP-1.beta., MIF, interferon-.gamma., and PAF.
3. The method of Claim 1, wherein the pro-inflammatory cytokine is TNF.
4. The method of Claim 1, wherein the vertebrate is a human.
5. The method of Claim 1, wherein the condition is selected from the group consisting of appendicitis, peptic ulcers, gastric ulcers, duodenal ulcers, peritonitis, pancreatitis, inflammatory bowel disease, diverticulitis, epiglottitis, achalasia, cholangitis, cholecystitis, hepatitis, enteritis, Whipple's disease, asthma, allergy, anaphylactic shock, immune complex disease, organ ischemia, reperfusion injury, organ necrosis, hay fever, sepsis, septicemia, endotoxic shock, cachexia, hyperpyrexia, eosinophilic granuloma, granulomatosis, sarcoidosis, septic abortion, epididymitis, vaginitis, prostatitis, urethritis, bronchitis, emphysema, rhinitis, cystic fibrosis, pneumonitis, pneumoultramicroscopic silicovolcanoconiosis, alveolitis, bronchiolitis, pharyngitis, pleurisy, sinusitis, influenza, respiratory syncytial virus infection, herpes infection, HIV infection, hepatitis B virus infection, hepatitis C
virus infection, disseminated bacteremia, Dengue fever, candidiasis, malaria, filariasis, amebiasis, hydatid cysts, burns, dermatitis, dermatomyositis, sunburn, urticaria, warts, wheals, vasculitis, angiitis, endocarditis, arteritis, atherosclerosis, thrombophlebitis, pericarditis, myocarditis, myocardial ischemia, periarteritis nodosa, rheumatic fever, coeliac disease, congestive heart failure, adult respiratory distress syndrome, meningitis, encephalitis, multiple sclerosis, cerebral infarction, cerebral embolism, Guillame-Barre syndrome, neuritis, neuralgia, spinal cord injury, paralysis, uveitis, arthritis, arthralgias, osteomyelitis, fasciitis, Paget's disease, gout, periodontal disease, synovitis, myasthenia gravis, thyroiditis, systemic lupus erythematosus, Goodpasture's syndrome, Behcets's syndrome, allograft rejection, graft-versus-host'disease, Type T diabetes, ankylosing spondylitis, Berger's disease, Retier's syndrome, and Hodgkins disease.
6. ~The method of Claim 5, wherein the inflammatory bowel disease is selected from the group consisting of ulcerative colitis, pseudomembranous colitis, acute colitis, ischemic colitis, and Crohn's disease.
7. ~The method of Claim 5, wherein the arthritis is rheumatoid arthritis.
8. ~The method of Claim 1, wherein the condition is selected from the group consisting of allograft rejection, arthritis, asthma, lupus, adult respiratory distress syndrome, pancreatitis, peritonitis, burns, Behcet's disease, graft versus host disease, inflammatory bowel disease, multiple sclerosis, organ ischemia, reperfusion injury, myocardial ischemia, and cachexia.
9. ~The method of Claim 1, wherein the condition is shock, chronic obstructive pulmonary disease, or psoriasis.
10. The method of Claim 1, wherein the condition is sepsis.
11. The method of Claim 1, wherein the brain muscarinic receptor is selected from the group consisting of an M1, an M2, and an M4 receptor.
12. The method of Claim 1, wherein the brain muscarinic receptor is activated by administering a muscarinic agonist to the vertebrate.
13. The method of Claim 12, wherein the muscarinic agonist is administered directly to the brain of the vertebrate.
14. The method of Claim 12, wherein the muscarinic agonist can cross the blood-brain barrier of the vertebrate, and wherein the agonist is administered enterically or parentally, or is injected into the bloodstream of the vertebrate.
15. The method of Claim 12, wherein the muscarinic agonist is selected from the group consisting of muscarine, McN-A-343, and MT-3.
16. A method of inhibiting release of a pro-inflammatory cytokine in a vertebrate at risk for or having a condition mediated by an inflammatory cytokine cascade, the method comprising directly stimulating a vagus nerve pathway in the brain of the vertebrate.
17. The method of Claim 16, wherein the vagus nerve pathway is stimulated electrically.
18. A method of inhibiting apoptosis of a cardiac myocyte in a vertebrate at risk for cardiac myocyte apoptosis, the method comprising activating a brain muscarinic receptor in the vertebrate.
19. A method of inhibiting apoptosis of a cardiac myocyte in a vertebrate at risk for cardiac myocyte apoptosis, the method comprising directly stimulating a vagus nerve pathway in the brain of the vertebrate.
20. A method of conditioning a vertebrate to inhibit the release of a pro-inflammatory cytokine upon experiencing a sensory stimulus, the method comprising (a) activating a brain muscarinic receptor in the vertebrate and providing the sensory stimulus to the vertebrate within a time period sufficient to create an association between the stimulus and the activation of the brain muscarinic receptor; and (b) repeating step (a) at sufficient time intervals and duration to reinforce the association sufficiently for the pro-inflammatory cytokine release to be inhibited by the sensory stimulus alone.
21. A method of conditioning a vertebrate to inhibit the release of a pro-inflammatory cytokine upon experiencing a sensory stimulus, the method comprising (a) directly stimulating a vagus nerve pathway in the brain of the vertebrate and providing the sensory stimulus to the vertebrate within a time period sufficient to create an association between the stimulus and the stimulation of a vagus nerve pathway; and (b) repeating step (a) at sufficient time intervals and duration to reinforce the association sufficiently for the pro-inflammatory cytokine release to be inhibited by the sensory stimulus alone.
CA002476896A 2002-02-26 2003-02-26 Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors Abandoned CA2476896A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US36008202P 2002-02-26 2002-02-26
US60/360,082 2002-02-26
PCT/US2003/005873 WO2003072135A2 (en) 2002-02-26 2003-02-26 Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors

Publications (1)

Publication Number Publication Date
CA2476896A1 true CA2476896A1 (en) 2003-09-04

Family

ID=27766183

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002476896A Abandoned CA2476896A1 (en) 2002-02-26 2003-02-26 Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors

Country Status (6)

Country Link
US (2) US20040048795A1 (en)
EP (1) EP1487494A2 (en)
JP (1) JP2005522457A (en)
AU (1) AU2003217747A1 (en)
CA (1) CA2476896A1 (en)
WO (1) WO2003072135A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014113893A1 (en) * 2013-01-28 2014-07-31 University Of Manitoba Use of galantamine and related compounds for treatment of inflammatory bowel diseases

Families Citing this family (100)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8914114B2 (en) 2000-05-23 2014-12-16 The Feinstein Institute For Medical Research Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US6907295B2 (en) 2001-08-31 2005-06-14 Biocontrol Medical Ltd. Electrode assembly for nerve control
US6684105B2 (en) 2001-08-31 2004-01-27 Biocontrol Medical, Ltd. Treatment of disorders by unidirectional nerve stimulation
US7734355B2 (en) * 2001-08-31 2010-06-08 Bio Control Medical (B.C.M.) Ltd. Treatment of disorders by unidirectional nerve stimulation
US8571653B2 (en) * 2001-08-31 2013-10-29 Bio Control Medical (B.C.M.) Ltd. Nerve stimulation techniques
US7885709B2 (en) * 2001-08-31 2011-02-08 Bio Control Medical (B.C.M.) Ltd. Nerve stimulation for treating disorders
US7904176B2 (en) 2006-09-07 2011-03-08 Bio Control Medical (B.C.M.) Ltd. Techniques for reducing pain associated with nerve stimulation
US7778711B2 (en) * 2001-08-31 2010-08-17 Bio Control Medical (B.C.M.) Ltd. Reduction of heart rate variability by parasympathetic stimulation
US7974693B2 (en) 2001-08-31 2011-07-05 Bio Control Medical (B.C.M.) Ltd. Techniques for applying, configuring, and coordinating nerve fiber stimulation
US7778703B2 (en) * 2001-08-31 2010-08-17 Bio Control Medical (B.C.M.) Ltd. Selective nerve fiber stimulation for treating heart conditions
EP1487494A2 (en) * 2002-02-26 2004-12-22 North Shore-Long Island Jewish Research Institute Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors
US9592409B2 (en) 2002-04-30 2017-03-14 The Regents Of The University Of California Methods for modifying electrical currents in neuronal circuits
US7283861B2 (en) * 2002-04-30 2007-10-16 Alexander Bystritsky Methods for modifying electrical currents in neuronal circuits
US7561922B2 (en) * 2004-12-22 2009-07-14 Biocontrol Medical Ltd. Construction of electrode assembly for nerve control
US7321793B2 (en) * 2003-06-13 2008-01-22 Biocontrol Medical Ltd. Vagal stimulation for atrial fibrillation therapy
WO2004110550A2 (en) * 2003-06-13 2004-12-23 Biocontrol Medical Ltd. Vagal stimulation for anti-embolic therapy
US7844346B2 (en) * 2002-05-23 2010-11-30 Biocontrol Medical Ltd. Electrode assembly for nerve control
US8204591B2 (en) * 2002-05-23 2012-06-19 Bio Control Medical (B.C.M.) Ltd. Techniques for prevention of atrial fibrillation
US8880192B2 (en) 2012-04-02 2014-11-04 Bio Control Medical (B.C.M.) Ltd. Electrode cuffs
US7627384B2 (en) * 2004-11-15 2009-12-01 Bio Control Medical (B.C.M.) Ltd. Techniques for nerve stimulation
US8060197B2 (en) 2003-05-23 2011-11-15 Bio Control Medical (B.C.M.) Ltd. Parasympathetic stimulation for termination of non-sinus atrial tachycardia
US7738952B2 (en) * 2003-06-09 2010-06-15 Palo Alto Investors Treatment of conditions through modulation of the autonomic nervous system
EP1648558A4 (en) * 2003-06-13 2015-05-27 Biocontrol Medical B C M Ltd Applications of vagal stimulation
US7657312B2 (en) 2003-11-03 2010-02-02 Cardiac Pacemakers, Inc. Multi-site ventricular pacing therapy with parasympathetic stimulation
US7869881B2 (en) * 2003-12-24 2011-01-11 Cardiac Pacemakers, Inc. Baroreflex stimulator with integrated pressure sensor
US8126560B2 (en) * 2003-12-24 2012-02-28 Cardiac Pacemakers, Inc. Stimulation lead for stimulating the baroreceptors in the pulmonary artery
US8024050B2 (en) 2003-12-24 2011-09-20 Cardiac Pacemakers, Inc. Lead for stimulating the baroreceptors in the pulmonary artery
JP2007530586A (en) * 2004-03-25 2007-11-01 ザ ファインスタイン インスティテュート フォー メディカル リサーチ Nervous hemostasis
US10912712B2 (en) 2004-03-25 2021-02-09 The Feinstein Institutes For Medical Research Treatment of bleeding by non-invasive stimulation
CA2998199A1 (en) * 2004-06-01 2005-12-15 Kwalata Trading Limited Methods for use with stem cells involving culturing on a surface with antibodies
US7244765B2 (en) 2004-06-25 2007-07-17 Cytokine Pharmasciences, Inc Guanylhydrazone salts, compositions, processes of making and methods of using
US7291647B2 (en) * 2004-08-17 2007-11-06 Cytokine Pharmasciences, Inc. Guanylhydrazone compounds, compositions, methods of making and using
US20060253100A1 (en) 2004-10-22 2006-11-09 Medtronic, Inc. Systems and Methods to Treat Pain Locally
AU2005323463B2 (en) 2004-12-27 2009-11-19 The Feinstein Institutes For Medical Research Treating inflammatory disorders by electrical vagus nerve stimulation
US11207518B2 (en) 2004-12-27 2021-12-28 The Feinstein Institutes For Medical Research Treating inflammatory disorders by stimulation of the cholinergic anti-inflammatory pathway
US8609082B2 (en) 2005-01-25 2013-12-17 Bio Control Medical Ltd. Administering bone marrow progenitor cells or myoblasts followed by application of an electrical current for cardiac repair, increasing blood supply or enhancing angiogenesis
DE102005025906A1 (en) * 2005-06-06 2006-12-07 Dorian Bevec Use of a guanylhydrazone compound in the manufacture of medicament for the treatment and/or prevention of e.g. acute and chronic hepatitis, liver cirrhosis, liver cell carcinoma, myocarditis, pharyngitis, pneumonia and pericarditis
US20070191904A1 (en) * 2006-02-14 2007-08-16 Imad Libbus Expandable stimulation electrode with integrated pressure sensor and methods related thereto
TW200734462A (en) 2006-03-08 2007-09-16 In Motion Invest Ltd Regulating stem cells
US20130165824A1 (en) * 2006-05-17 2013-06-27 The Research Foundation Of The State University Of New York Method and system for physical stimulation of tissue
AU2007265631B2 (en) * 2006-06-23 2012-11-08 The Feinstein Institute For Medical Research Inhibitors of A-Beta and synuclein aggregation
US8170668B2 (en) 2006-07-14 2012-05-01 Cardiac Pacemakers, Inc. Baroreflex sensitivity monitoring and trending for tachyarrhythmia detection and therapy
WO2009029614A1 (en) 2007-08-27 2009-03-05 The Feinstein Institute For Medical Research Devices and methods for inhibiting granulocyte activation by neural stimulation
US9662490B2 (en) 2008-03-31 2017-05-30 The Feinstein Institute For Medical Research Methods and systems for reducing inflammation by neuromodulation and administration of an anti-inflammatory drug
WO2009146030A1 (en) 2008-03-31 2009-12-03 The Feinstein Institute For Medical Research Methods and systems for reducing inflammation by neuromodulation of t-cell activity
USRE48948E1 (en) 2008-04-18 2022-03-01 Warsaw Orthopedic, Inc. Clonidine compounds in a biodegradable polymer
US20090275997A1 (en) * 2008-05-01 2009-11-05 Michael Allen Faltys Vagus nerve stimulation electrodes and methods of use
CN102123706B (en) * 2008-06-17 2013-08-14 米纳斯吉拉斯联合大学 Use of paf receptor for treating infections caused by flaviviridae
CA2737522A1 (en) * 2008-09-19 2010-04-08 Cytokine Pharmasciences, Inc. Guanylhydrazones for treatment of postoperative intestinal inflammation
CN102215909B (en) * 2008-11-18 2014-09-10 赛博恩特医疗器械公司 Devices and methods for optimizing electrode placement for anti-inflamatory stimulation
US20100239632A1 (en) 2009-03-23 2010-09-23 Warsaw Orthopedic, Inc. Drug depots for treatment of pain and inflammation in sinus and nasal cavities or cardiac tissue
US9211410B2 (en) 2009-05-01 2015-12-15 Setpoint Medical Corporation Extremely low duty-cycle activation of the cholinergic anti-inflammatory pathway to treat chronic inflammation
US8612002B2 (en) * 2009-12-23 2013-12-17 Setpoint Medical Corporation Neural stimulation devices and systems for treatment of chronic inflammation
US8996116B2 (en) 2009-10-30 2015-03-31 Setpoint Medical Corporation Modulation of the cholinergic anti-inflammatory pathway to treat pain or addiction
AU2010258792B2 (en) 2009-06-09 2015-07-02 Setpoint Medical Corporation Nerve cuff with pocket for leadless stimulator
US9833621B2 (en) 2011-09-23 2017-12-05 Setpoint Medical Corporation Modulation of sirtuins by vagus nerve stimulation
WO2014169145A1 (en) 2013-04-10 2014-10-16 Setpoint Medical Corporation Closed-loop vagus nerve stimulation
US8788045B2 (en) 2010-06-08 2014-07-22 Bluewind Medical Ltd. Tibial nerve stimulation
US9186504B2 (en) 2010-11-15 2015-11-17 Rainbow Medical Ltd Sleep apnea treatment
US9457186B2 (en) 2010-11-15 2016-10-04 Bluewind Medical Ltd. Bilateral feedback
CN103619405B (en) 2011-05-09 2015-11-25 赛博恩特医疗器械公司 The individual pulse being used for the treatment of the cholinergic anti-inflammatory pathway of chronic inflammatory disease activates
US8466159B2 (en) 2011-10-21 2013-06-18 Abbvie Inc. Methods for treating HCV
TW201600087A (en) 2011-10-21 2016-01-01 艾伯維有限公司 Methods for treating HCV
US8492386B2 (en) 2011-10-21 2013-07-23 Abbvie Inc. Methods for treating HCV
DK2583677T1 (en) 2011-10-21 2015-01-19 Abbvie Inc Methods for treatment of HCV comprising at least two direct-acting antiviral agents ribavirin, interferon but not
BR112014008036A2 (en) * 2011-10-28 2017-04-11 Ampio Pharmaceuticals Inc rhinitis treatment
US20150018728A1 (en) 2012-01-26 2015-01-15 Bluewind Medical Ltd. Wireless neurostimulators
US9572983B2 (en) 2012-03-26 2017-02-21 Setpoint Medical Corporation Devices and methods for modulation of bone erosion
WO2014087337A1 (en) 2012-12-06 2014-06-12 Bluewind Medical Ltd. Delivery of implantable neurostimulators
US9061133B2 (en) 2012-12-27 2015-06-23 Brainsonix Corporation Focused ultrasonic transducer navigation system
US10512794B2 (en) 2016-12-16 2019-12-24 Brainsonix Corporation Stereotactic frame
US10974078B2 (en) 2012-12-27 2021-04-13 Brainsonix Corporation Treating degenerative dementia with low intensity focused ultrasound pulsation (LIFUP) device
US9370660B2 (en) 2013-03-29 2016-06-21 Rainbow Medical Ltd. Independently-controlled bidirectional nerve stimulation
US11311725B2 (en) 2014-10-24 2022-04-26 Setpoint Medical Corporation Systems and methods for stimulating and/or monitoring loci in the brain to treat inflammation and to enhance vagus nerve stimulation
US10004896B2 (en) 2015-01-21 2018-06-26 Bluewind Medical Ltd. Anchors and implant devices
US9597521B2 (en) 2015-01-21 2017-03-21 Bluewind Medical Ltd. Transmitting coils for neurostimulation
US9764146B2 (en) 2015-01-21 2017-09-19 Bluewind Medical Ltd. Extracorporeal implant controllers
US11406833B2 (en) 2015-02-03 2022-08-09 Setpoint Medical Corporation Apparatus and method for reminding, prompting, or alerting a patient with an implanted stimulator
US9782589B2 (en) 2015-06-10 2017-10-10 Bluewind Medical Ltd. Implantable electrostimulator for improving blood flow
US10105540B2 (en) 2015-11-09 2018-10-23 Bluewind Medical Ltd. Optimization of application of current
US9713707B2 (en) 2015-11-12 2017-07-25 Bluewind Medical Ltd. Inhibition of implant migration
US10596367B2 (en) 2016-01-13 2020-03-24 Setpoint Medical Corporation Systems and methods for establishing a nerve block
EP3405255A4 (en) 2016-01-20 2019-10-16 Setpoint Medical Corporation Implantable microstimulators and inductive charging systems
US10695569B2 (en) 2016-01-20 2020-06-30 Setpoint Medical Corporation Control of vagal stimulation
US11471681B2 (en) 2016-01-20 2022-10-18 Setpoint Medical Corporation Batteryless implantable microstimulators
US10583304B2 (en) 2016-01-25 2020-03-10 Setpoint Medical Corporation Implantable neurostimulator having power control and thermal regulation and methods of use
JP7129703B2 (en) 2016-04-28 2022-09-02 エモリー ユニバーシティー Alkyne-Containing Nucleotide and Nucleoside Therapeutic Compositions and Uses Associated Therewith
US10124178B2 (en) 2016-11-23 2018-11-13 Bluewind Medical Ltd. Implant and delivery tool therefor
US20180353764A1 (en) 2017-06-13 2018-12-13 Bluewind Medical Ltd. Antenna configuration
US11173307B2 (en) 2017-08-14 2021-11-16 Setpoint Medical Corporation Vagus nerve stimulation pre-screening test
US11179412B2 (en) * 2017-12-04 2021-11-23 University of Pittsburgh—of the Commonwealth System of Higher Education Methods of treating conditions involving elevated inflammatory response
US11026905B2 (en) 2018-04-19 2021-06-08 Tvardi Therapeutics, Inc. STAT3 inhibitors
PL3773537T3 (en) 2018-04-19 2022-05-23 Tvardi Therapeutics, Inc. Stat3 inhibitors
US11660443B2 (en) 2018-04-20 2023-05-30 The Feinstein Institutes For Medical Research Methods and apparatuses for reducing bleeding via electrical trigeminal nerve stimulation
US11260229B2 (en) 2018-09-25 2022-03-01 The Feinstein Institutes For Medical Research Methods and apparatuses for reducing bleeding via coordinated trigeminal and vagal nerve stimulation
JP2023520273A (en) 2020-01-24 2023-05-17 トゥヴァルディ セラピューティクス,インク. THERAPEUTIC COMPOUNDS, FORMULATIONS AND USES THEREOF
US11759661B2 (en) 2020-05-20 2023-09-19 Brainsonix Corporation Ultrasonic transducer treatment device
US11938324B2 (en) 2020-05-21 2024-03-26 The Feinstein Institutes For Medical Research Systems and methods for vagus nerve stimulation
WO2021246399A1 (en) * 2020-06-01 2021-12-09 学校法人 慶應義塾 Therapeutic drug for disease caused by intestinal immune disorder
US11400299B1 (en) 2021-09-14 2022-08-02 Rainbow Medical Ltd. Flexible antenna for stimulator

Family Cites Families (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3363623A (en) * 1965-07-28 1968-01-16 Charles F. Atwell Hand-held double-acting nerve reflex massager
FR2315274A1 (en) * 1975-06-27 1977-01-21 Parcor NEW DERIVATIVES OF THIENO (2,3-C) PYRIDINE, THEIR PREPARATION AND THEIR APPLICATIONS
US5025807A (en) * 1983-09-14 1991-06-25 Jacob Zabara Neurocybernetic prosthesis
US4573481A (en) * 1984-06-25 1986-03-04 Huntington Institute Of Applied Research Implantable electrode array
US4929734A (en) * 1987-03-31 1990-05-29 Warner-Lambert Company Tetrahydropyridine oxime compounds
US4935234A (en) * 1987-06-11 1990-06-19 Dana-Farber Cancer Institute Method of reducing tissue damage at an inflammatory site using a monoclonal antibody
US4840793A (en) * 1987-06-11 1989-06-20 Dana-Farber Cancer Institute Method of reducing tissue damage at an inflammatory site using a monoclonal antibody
US5019648A (en) * 1987-07-06 1991-05-28 Dana-Farber Cancer Institute Monoclonal antibody specific for the adhesion function domain of a phagocyte cell surface protein
DE3736664A1 (en) * 1987-10-29 1989-05-11 Boehringer Ingelheim Kg TETRAHYDRO-FURO- AND -THIENO (2,3-C) PYRIDINE, THEIR USE AS A MEDICAMENT AND METHOD FOR THE PRODUCTION THEREOF
US5106853A (en) * 1989-05-15 1992-04-21 Merck Sharp & Dohme, Ltd. Oxadiazole and its salts, their use in treating dementia
US5186170A (en) * 1989-11-13 1993-02-16 Cyberonics, Inc. Simultaneous radio frequency and magnetic field microprocessor reset circuit
US5179950A (en) * 1989-11-13 1993-01-19 Cyberonics, Inc. Implanted apparatus having micro processor controlled current and voltage sources with reduced voltage levels when not providing stimulation
US5496938A (en) * 1990-06-11 1996-03-05 Nexstar Pharmaceuticals, Inc. Nucleic acid ligands to HIV-RT and HIV-1 rev
US5705337A (en) * 1990-06-11 1998-01-06 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: chemi-SELEX
US5726017A (en) * 1990-06-11 1998-03-10 Nexstar Pharmaceuticals, Inc. High affinity HIV-1 gag nucleic acid ligands
US5503978A (en) * 1990-06-11 1996-04-02 University Research Corporation Method for identification of high affinity DNA ligands of HIV-1 reverse transcriptase
US5637459A (en) * 1990-06-11 1997-06-10 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: chimeric selex
US6168778B1 (en) * 1990-06-11 2001-01-02 Nexstar Pharmaceuticals, Inc. Vascular endothelial growth factor (VEGF) Nucleic Acid Ligand Complexes
US5712375A (en) * 1990-06-11 1998-01-27 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: tissue selex
US5111815A (en) * 1990-10-15 1992-05-12 Cardiac Pacemakers, Inc. Method and apparatus for cardioverter/pacer utilizing neurosensing
US5188104A (en) * 1991-02-01 1993-02-23 Cyberonics, Inc. Treatment of eating disorders by nerve stimulation
US5299569A (en) * 1991-05-03 1994-04-05 Cyberonics, Inc. Treatment of neuropsychiatric disorders by nerve stimulation
US5215086A (en) * 1991-05-03 1993-06-01 Cyberonics, Inc. Therapeutic treatment of migraine symptoms by stimulation
US6028186A (en) * 1991-06-10 2000-02-22 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands of cytokines
US5205285A (en) * 1991-06-14 1993-04-27 Cyberonics, Inc. Voice suppression of vagal stimulation
US5222494A (en) * 1991-07-31 1993-06-29 Cyberonics, Inc. Implantable tissue stimulator output stabilization system
US5175166A (en) * 1991-08-27 1992-12-29 The University Of Toledo Muscarinic agonists
US5215089A (en) * 1991-10-21 1993-06-01 Cyberonics, Inc. Electrode assembly for nerve stimulation
US5304206A (en) * 1991-11-18 1994-04-19 Cyberonics, Inc. Activation techniques for implantable medical device
US5203326A (en) * 1991-12-18 1993-04-20 Telectronics Pacing Systems, Inc. Antiarrhythmia pacer using antiarrhythmia pacing and autonomic nerve stimulation therapy
JP3034953B2 (en) * 1992-08-31 2000-04-17 ユニバーシティー オブ フロリダ リサーチ ファンデーション,インコーポレーテッド Anabaseine derivatives useful for the treatment of degenerative diseases of the nervous system
JP3269125B2 (en) * 1994-01-28 2002-03-25 東レ株式会社 Atopic dermatitis drug
US6017891A (en) * 1994-05-06 2000-01-25 Baxter Aktiengesellschaft Stable preparation for the treatment of blood coagulation disorders
US6405732B1 (en) * 1994-06-24 2002-06-18 Curon Medical, Inc. Method to treat gastric reflux via the detection and ablation of gastro-esophageal nerves and receptors
KR100366331B1 (en) * 1994-08-24 2003-04-10 아스트라제네카 악티에볼라그 Spiro-azabicyclic Compounds Useful for Treatment
US5604231A (en) * 1995-01-06 1997-02-18 Smith; Carr J. Pharmaceutical compositions for prevention and treatment of ulcerative colitis
US5707400A (en) * 1995-09-19 1998-01-13 Cyberonics, Inc. Treating refractory hypertension by nerve stimulation
WO1997014473A1 (en) * 1995-10-18 1997-04-24 Novartis Ag Thermopile powered transdermal drug delivery device
US5611350A (en) * 1996-02-08 1997-03-18 John; Michael S. Method and apparatus for facilitating recovery of patients in deep coma
US5913876A (en) * 1996-02-20 1999-06-22 Cardiothoracic Systems, Inc. Method and apparatus for using vagus nerve stimulation in surgery
DK0796623T3 (en) * 1996-03-20 2005-08-01 Baxter Ag Pharmaceutical composition for the treatment of blood clotting disorders
US5618818A (en) * 1996-03-20 1997-04-08 The University Of Toledo Muscarinic agonist compounds
US5690681A (en) * 1996-03-29 1997-11-25 Purdue Research Foundation Method and apparatus using vagal stimulation for control of ventricular rate during atrial fibrillation
US5726179A (en) * 1996-04-01 1998-03-10 The University Of Toledo Muscarinic agonists
US6449507B1 (en) * 1996-04-30 2002-09-10 Medtronic, Inc. Method and system for nerve stimulation prior to and during a medical procedure
US6532388B1 (en) * 1996-04-30 2003-03-11 Medtronic, Inc. Method and system for endotracheal/esophageal stimulation prior to and during a medical procedure
US6006134A (en) * 1998-04-30 1999-12-21 Medtronic, Inc. Method and device for electronically controlling the beating of a heart using venous electrical stimulation of nerve fibers
US6904318B2 (en) * 2000-09-26 2005-06-07 Medtronic, Inc. Method and system for monitoring and controlling systemic and pulmonary circulation during a medical procedure
USRE38705E1 (en) * 1996-04-30 2005-02-22 Medtronic, Inc. Method and device for electronically controlling the beating of a heart using venous electrical stimulation of nerve fibers
US6735471B2 (en) * 1996-04-30 2004-05-11 Medtronic, Inc. Method and system for endotracheal/esophageal stimulation prior to and during a medical procedure
US6628987B1 (en) * 2000-09-26 2003-09-30 Medtronic, Inc. Method and system for sensing cardiac contractions during vagal stimulation-induced cardiopalegia
US7225019B2 (en) * 1996-04-30 2007-05-29 Medtronic, Inc. Method and system for nerve stimulation and cardiac sensing prior to and during a medical procedure
AU3304997A (en) * 1996-05-31 1998-01-05 Southern Illinois University Methods of modulating aspects of brain neural plasticity by vagus nerve stimulation
US5718912A (en) * 1996-10-28 1998-02-17 Merck & Co., Inc. Muscarine agonists
CA2271693C (en) * 1996-11-15 2009-01-20 The Picower Institute For Medical Research Guanylhydrazones useful for treating diseases associated with t cell activation
US6479523B1 (en) * 1997-08-26 2002-11-12 Emory University Pharmacologic drug combination in vagal-induced asystole
US6528529B1 (en) * 1998-03-31 2003-03-04 Acadia Pharmaceuticals Inc. Compounds with activity on muscarinic receptors
US6018682A (en) * 1998-04-30 2000-01-25 Medtronic, Inc. Implantable seizure warning system
US7209787B2 (en) * 1998-08-05 2007-04-24 Bioneuronics Corporation Apparatus and method for closed-loop intracranial stimulation for optimal control of neurological disease
US7599736B2 (en) * 2001-07-23 2009-10-06 Dilorenzo Biomedical, Llc Method and apparatus for neuromodulation and physiologic modulation for the treatment of metabolic and neuropsychiatric disease
US6208902B1 (en) * 1998-10-26 2001-03-27 Birinder Bob Boveja Apparatus and method for adjunct (add-on) therapy for pain syndromes utilizing an implantable lead and an external stimulator
US6564102B1 (en) * 1998-10-26 2003-05-13 Birinder R. Boveja Apparatus and method for adjunct (add-on) treatment of coma and traumatic brain injury with neuromodulation using an external stimulator
US6356788B2 (en) * 1998-10-26 2002-03-12 Birinder Bob Boveja Apparatus and method for adjunct (add-on) therapy for depression, migraine, neuropsychiatric disorders, partial complex epilepsy, generalized epilepsy and involuntary movement disorders utilizing an external stimulator
US6366814B1 (en) * 1998-10-26 2002-04-02 Birinder R. Boveja External stimulator for adjunct (add-on) treatment for neurological, neuropsychiatric, and urological disorders
US6205359B1 (en) * 1998-10-26 2001-03-20 Birinder Bob Boveja Apparatus and method for adjunct (add-on) therapy of partial complex epilepsy, generalized epilepsy and involuntary movement disorders utilizing an external stimulator
US20050137644A1 (en) * 1998-10-26 2005-06-23 Boveja Birinder R. Method and system for vagal blocking and/or vagal stimulation to provide therapy for obesity and other gastrointestinal disorders
US5994330A (en) * 1998-11-09 1999-11-30 El Khoury; Georges F. Topical application of muscarinic agents such as neostigmine for treatment of acne and other inflammatory conditions
FR2786770B1 (en) * 1998-12-04 2001-01-19 Synthelabo NONANE 1,4-DIAZABICYCLO [3.2.2.] DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
US6341236B1 (en) * 1999-04-30 2002-01-22 Ivan Osorio Vagal nerve stimulation techniques for treatment of epileptic seizures
US6587719B1 (en) * 1999-07-01 2003-07-01 Cyberonics, Inc. Treatment of obesity by bilateral vagus nerve stimulation
US6171795B1 (en) * 1999-07-29 2001-01-09 Nexstar Pharmaceuticals, Inc. Nucleic acid ligands to CD40ligand
US20020026141A1 (en) * 1999-11-04 2002-02-28 Medtronic, Inc. System for pancreatic stimulation and glucose measurement
US6885888B2 (en) * 2000-01-20 2005-04-26 The Cleveland Clinic Foundation Electrical stimulation of the sympathetic nerve chain
US20060085046A1 (en) * 2000-01-20 2006-04-20 Ali Rezai Methods of treating medical conditions by transvascular neuromodulation of the autonomic nervous system
US6356787B1 (en) * 2000-02-24 2002-03-12 Electro Core Techniques, Llc Method of treating facial blushing by electrical stimulation of the sympathetic nerve chain
US6826428B1 (en) * 2000-04-11 2004-11-30 The Board Of Regents Of The University Of Texas System Gastrointestinal electrical stimulation
US6610713B2 (en) * 2000-05-23 2003-08-26 North Shore - Long Island Jewish Research Institute Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US8914114B2 (en) * 2000-05-23 2014-12-16 The Feinstein Institute For Medical Research Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US6511500B1 (en) * 2000-06-06 2003-01-28 Marc Mounir Rahme Use of autonomic nervous system neurotransmitters inhibition and atrial parasympathetic fibers ablation for the treatment of atrial arrhythmias and to preserve drug effects
WO2002016357A2 (en) * 2000-08-18 2002-02-28 Pharmacia & Upjohn Company Quinuclidine-substituted aryl moieties for treatment of disease (nicotinic acetylcholine receptor ligands)
US20020077675A1 (en) * 2000-09-26 2002-06-20 Transneuronix, Inc. Minimally invasive surgery placement of stimulation leads in mediastinal structures
US6487446B1 (en) * 2000-09-26 2002-11-26 Medtronic, Inc. Method and system for spinal cord stimulation prior to and during a medical procedure
US7011638B2 (en) * 2000-11-14 2006-03-14 Science Medicus, Inc. Device and procedure to treat cardiac atrial arrhythmias
US6609025B2 (en) * 2001-01-02 2003-08-19 Cyberonics, Inc. Treatment of obesity by bilateral sub-diaphragmatic nerve stimulation
US6735475B1 (en) * 2001-01-30 2004-05-11 Advanced Bionics Corporation Fully implantable miniature neurostimulator for stimulation as a therapy for headache and/or facial pain
US7167751B1 (en) * 2001-03-01 2007-01-23 Advanced Bionics Corporation Method of using a fully implantable miniature neurostimulator for vagus nerve stimulation
US7369897B2 (en) * 2001-04-19 2008-05-06 Neuro And Cardiac Technologies, Llc Method and system of remotely controlling electrical pulses provided to nerve tissue(s) by an implanted stimulator system for neuromodulation therapies
US6684105B2 (en) * 2001-08-31 2004-01-27 Biocontrol Medical, Ltd. Treatment of disorders by unidirectional nerve stimulation
US7054686B2 (en) * 2001-08-30 2006-05-30 Biophan Technologies, Inc. Pulsewidth electrical stimulation
US7778703B2 (en) * 2001-08-31 2010-08-17 Bio Control Medical (B.C.M.) Ltd. Selective nerve fiber stimulation for treating heart conditions
US7885709B2 (en) * 2001-08-31 2011-02-08 Bio Control Medical (B.C.M.) Ltd. Nerve stimulation for treating disorders
US6721603B2 (en) * 2002-01-25 2004-04-13 Cyberonics, Inc. Nerve stimulation as a treatment for pain
EP1487494A2 (en) * 2002-02-26 2004-12-22 North Shore-Long Island Jewish Research Institute Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors
WO2006007048A2 (en) * 2004-05-04 2006-01-19 The Cleveland Clinic Foundation Methods of treating medical conditions by neuromodulation of the sympathetic nervous system

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014113893A1 (en) * 2013-01-28 2014-07-31 University Of Manitoba Use of galantamine and related compounds for treatment of inflammatory bowel diseases

Also Published As

Publication number Publication date
US20080140138A1 (en) 2008-06-12
US20040048795A1 (en) 2004-03-11
WO2003072135A3 (en) 2004-07-22
EP1487494A2 (en) 2004-12-22
WO2003072135A2 (en) 2003-09-04
JP2005522457A (en) 2005-07-28
AU2003217747A1 (en) 2003-09-09

Similar Documents

Publication Publication Date Title
CA2476896A1 (en) Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors
US10561846B2 (en) Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US6610713B2 (en) Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
AU2001261629A1 (en) Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US11241429B2 (en) Method of treating metabolic disorders and depression with dopamine receptor agonists
EP1581223B1 (en) Inhibition of inflamation using alpha 7 receptor-binding cholinergic agonists
US6919366B2 (en) Benzimidazole derivatives as modulators of IgE
JP2002516276A (en) Benzimidazole derivatives as modulators of IgE
US20080003280A1 (en) Combination cough treatment compounds and method of treating common coughs
SK12172000A3 (en) Pharmaceutical combinations containing tramadol
AU2007202036A1 (en) Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors
SK8572003A3 (en) Histamine receptor antagonists
WO2022246930A1 (en) Application of ligand small molecule targeting pdcd4 in preparation of antidepressant drugs
JPH1160483A (en) Tnf production inhibitor

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued