WO2022170396A1 - Agents and methods for therapy and prophylaxis - Google Patents

Agents and methods for therapy and prophylaxis Download PDF

Info

Publication number
WO2022170396A1
WO2022170396A1 PCT/AU2022/050089 AU2022050089W WO2022170396A1 WO 2022170396 A1 WO2022170396 A1 WO 2022170396A1 AU 2022050089 W AU2022050089 W AU 2022050089W WO 2022170396 A1 WO2022170396 A1 WO 2022170396A1
Authority
WO
WIPO (PCT)
Prior art keywords
virus
amino acid
tlr7
subject
betacoronavirus
Prior art date
Application number
PCT/AU2022/050089
Other languages
French (fr)
Inventor
Stavros SELEMIDIS
Doug A. BROOKS
John O'leary
Original Assignee
ViraLok Therapeutics Pty Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2021900345A external-priority patent/AU2021900345A0/en
Application filed by ViraLok Therapeutics Pty Ltd filed Critical ViraLok Therapeutics Pty Ltd
Publication of WO2022170396A1 publication Critical patent/WO2022170396A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This disclosure relates generally to the treatment and prophylaxis of acute inflammatory conditions. More particularly, the present disclosure relates to the use of selective TLR7 antagonists for treating or inhibiting the development of acute inflammatory conditions including, for example, acute inflammatory conditions associated with presence of pathogenic infections, multisystem inflammatory syndrome, systemic inflammatory response syndrome, acute respiratory distress syndrome, severe acute respiratory syndrome and/or symptoms arising from acute inflammatory conditions.
  • Inflammation is an adaptive process to harmful stimuli such as pathogens, damaged cells, or irritants. It is a protective attempt by an organism, which typically involves immune cells, blood vessels and molecular mediators, to remove the noxious stimuli and to initiate the healing process for damaged tissue.
  • inflammation which runs unchecked can also lead to a host of diseases, including cytokine release syndrome (CRS)/cytokine storm, systemic inflammation and multiple organ failure.
  • CRS cytokine release syndrome
  • cytokine storm systemic inflammation and multiple organ failure.
  • broad-based antivirals which can desirably: 1) treat new infection outbreaks irrespective of the infecting strain of virus; 2) have therapeutic utility after a viral outbreak has occurred and before vaccine strategies become available/or where vaccines are not effective; 3) have utility in patients that have already been infected by the pathogenic virus; and/or 4) can treat direct and indirect symptoms and conditions associated with viral infection.
  • the pathway of viral pathogenesis typically involves: 1) attachment of the virus to the cell surface and entry into endosomes that are a common conduit for viral transport, which facilitate viral replication and induction of a pathogenic cascade; 2) at the key regulatory point in this pathway is the toll-like receptor 7 (TLR7) protein, which is a gatekeeper that RNA viruses use to trigger a cytokine cascade and production of highly reactive and oxygen species (ROS), which ultimately drive inflammation, viral replication, cellular damage and limit effective host immunity.
  • TLR7 toll-like receptor 7
  • TLR7 is a pattern recognition receptor that recognizes viral RNA following endocytosis of the virus and initiates a powerful immune response characterized by Type I IFN production and pro-inflammatory cytokine production; including tumor necrosis factor-a (TNF-a) and interlukin-6 (IL-6) production. TLR7 recruits NOX2 to drive ROS production in endosomes and this establishes a microenvironment that potentiates aberrant inflammation and immune signaling. By altering the normal immune response, RNA viruses cause very significant pathology, which is highly ROS and inflammation-dependent. Critically, TLR7 is activated by an enzyme called furin, which is enriched in the Golgi apparatus, where it functions to cleave proteins into their active form(s).
  • furin an enzyme that is enriched in the Golgi apparatus, where it functions to cleave proteins into their active form(s).
  • furin is also utilized by a number of highly pathogenic viruses such as influenza, dengue, filoviruses including Ebola and Marburg virus, as well as SARS-CoV-2.
  • viruses such as influenza, dengue, filoviruses including Ebola and Marburg virus, as well as SARS-CoV-2.
  • the cleavage and activation of these viruses by furin or furin-like proteases underpins a key component of the viral infection and pathogenesis and this involves TLR7.
  • TLR7 protein is activated by a disulfide bond forming between cysteine residues at positions 98 (Cys 98 ) and 475 (Cys 475 ) of TLR7 and that this leads to toxic ROS production via NADPH oxidase (N0X2).
  • NADPH oxidase NADPH oxidase
  • TLR7 antagonism is able to inhibit morbidity, viral titer, furin synthesis, pattern recognition receptors, oxidative stressors, cytokine storm and clinical signs of inflammation in subjects with active RNA virus infections, including infections with highly pathogenic H1N1 influenza A virus (IAV), in which subjects had been infected for five days.
  • IAV highly pathogenic H1N1 influenza A virus
  • TLR7 antagonism can significantly reduce or stymie this release after it has developed.
  • selective TLR7 antagonists can be used to treat acute inflammatory conditions generally, particularly those caused or exacerbated at least in part by activation of TLR7, including ones associated with the presence of cytokine release syndrome or a cytokine storm.
  • the present findings also indicate that the treatment can be given and is effective even after the viral infection and or inflammatory condition has already established in the host.
  • these methods generally comprise, consist or consist essentially of administering to the subject an effective amount of a selective TLR7 antagonist.
  • the acute inflammatory condition is at least in part caused or exacerbated by, or is otherwise associated with, activation of TLR7 and optionally one or both of TLR8 and TLR9, and/or an increased level of one or more oxidative stressors (e.g., NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1; also known as p47phox) and ROS) and/or an increased level of furin.
  • oxidative stressors e.g., NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1; also known as p47phox) and ROS
  • the acute inflammatory condition is associated with presence of a pathogenic infection (e.g., a pathogenic infection in which the pathogen is an RNA virus), which is typically an active infection.
  • a pathogenic infection e.g., a pathogenic infection in which the pathogen is an RNA virus
  • the RNA virus may enter a host cell that it infects by receptor- mediated endocytosis or micropinocytosis. In specific embodiments, the RNA virus enters the endosome of a host cell that it infects.
  • the RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenza virus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus), Togaviridae (e.g., Chik
  • the RNA virus is an orthomyxovirus (e.g., an influenza virus such as influenza A, influenza B or influenza C) or a coronavirus (e.g., a coronavirus, including betacoronaviruses, capable of causing severe acute respiratory syndrome (SARS) such as MERS-CoV, SARS-CoV and SARS-CoV-2).
  • an influenza virus such as influenza A, influenza B or influenza C
  • a coronavirus e.g., a coronavirus, including betacoronaviruses, capable of causing severe acute respiratory syndrome (SARS) such as MERS-CoV, SARS-CoV and SARS-CoV-2).
  • SARS severe acute respiratory syndrome
  • the acute inflammatory condition is associated with presence of cytokine release syndrome (CRS) or a cytokine storm, which in representative examples comprises an elevation of at least 50% compared to basal state of one or more (e.g., 1, 2, 3, 4, 5, 6, 7 or 8) cytokines selected from IFN- Y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2.
  • the acute inflammatory condition is associated with a condition selected from multisystem inflammatory syndrome in children (MIS-C), systemic inflammatory response syndrome (SIRS), acute respiratory distress syndrome (ARDS), and severe acute respiratory syndrome (SARS).
  • MIMS multisystem inflammatory syndrome
  • SIRS systemic inflammatory response syndrome
  • ARDS acute respiratory distress syndrome
  • SARS severe acute respiratory syndrome
  • the treatment of conditions associated directly or indirectly with viral infection is also contemplated herein including the treatment of cardiovascular conditions as well as the reduction, loss, modification or distortion of olfactory sensation.
  • the selective TLR7 antagonist may be selected from nucleic acid antagonists (e.g., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide) and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines).
  • the selective TLR7 antagonist comprises a peptide corresponding to the amino acid sequence of TLR7, which antagonizes disulfide bond formation between C98 and C475 of human TLR7 or their corresponding positions in the TLR7s of other species.
  • selective antagonism is not a total or complete abrogation of TLR7 activity but rather selective inhibition to return the immune system to, or towards, homeostasis.
  • CRS or a cytokine storm, SARS, ARDS, SIRS or MIS-C in a subject These methods generally comprise, consist or consist essentially of administering to the subject an effective amount of a selective TLR7 antagonist.
  • the present disclosure provides selective TLR7 antagonists, suitably in the form of medicaments, for treating a condition selected from an acute inflammatory condition, CRS or a cytokine storm, SARS, ARDS, SIRS or MIS-C.
  • the methods suitably comprise administering the subject with a single dose of the selective TLR7 antagonist.
  • the single dose is administered only once during the course of treatment.
  • the selective TLR7 antagonist may be concurrently administered with an antigenic composition that stimulates or enhances the production of an immune response in the subject to a pathogen.
  • BALF bronchoalveolar lavage fluid
  • Figure 4 is H8i E imaging (4a and 4b) and graphic representations (4c) of lung inflammation in mice infected with PR8 virus (10 or 50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98L 4C are the results of the blinded analysis performed by 2 independent histologists.
  • BALF bronchoalveolar lavage fluid
  • mice were administered either PBS vehicle or C98i (2mg/kg).
  • 1-way ANOVA- Tukey Kramer *** PcO.OOl, **** PcO.OOOl.
  • BALF bronchoalveolar lavage fluid
  • Figure 10 is a graphic representation of the lung mRNA expression of furin
  • Figure 11 is a graphic representation of the lung mRNA expression of TLR7
  • BALF bronchoalveolar lavage fluid
  • BALF bronchoalveolar lavage fluid
  • Figure 14 is H8iE imaging of lung inflammation in mice infected with PR8 virus (50PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg) at Day 5 post infection.
  • Figure 16 is a graphic representation of the lung mRNA expression of furin
  • BALF bronchoalveolar lavage fluid
  • Figure 22 is a graphic representation of the bronchoalveolar lavage fluid (BALF) macrophage, neutrophil, lymphocyte and eosinophil recruitment in mice infected with RV1B virus (10 10 PFU/mouse). Some mice were not infected with virus.
  • BALF bronchoalveolar lavage fluid
  • mice were administered either PBS vehicle or C98i (2mg/kg).
  • Figure 23 is a graphic representation of the lung cytokine mRNA expression in mice infected with RV1B virus (10 10 PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg). All genes were normalized to the housekeeping gene RPS18 and then normalized as a fold-increase above the PBS control group. 1-way ANOVA- Tukey Kramer * P ⁇ 0.05; ** PcO.Ol.
  • the articles “a” and “an” are used herein to refer to one or to more than one (/.e., to at least one) of the grammatical object of the article.
  • a virus includes a single virus, as well as two or more viruses
  • an agent includes a single agent, as well as two or more agents
  • reference to “the disclosure” includes single and multiple aspects taught by the disclosure; and so forth.
  • the term "about”, as used herein when referring to a measurable value such as an amount, dose, time, temperature, activity, level, number, frequency, percentage, dimension, size, amount, weight, position, length and the like, is meant to encompass variations of ⁇ 15%, ⁇ 10%, ⁇ 5%, ⁇ 1%, ⁇ 0.5%, or even ⁇ 0.1% of the specified amount, dose, time, temperature, activity, level, number, frequency, percentage, dimension, size, amount, weight, position, length and the like.
  • active infection is used herein in its broadest sense and includes the invasion, establishment and/or multiplication of a virus in a host, which is typically associated with one or more pathological symptoms that may or may not be clinically apparent. Active infections include localized, subclinical or temporary infections. A local infection may persist and spread by extension to become an acute, subacute or chronic clinical infection or disease state. A local infection may also become systemic when a virus gains access to the lymphatic or vascular system. Typically, “active infection” refers to an infectious state in which a host's immune system is activated against an infectious agent.
  • acute inflammatory condition refers to a condition in which acute inflammation is present and represents a rapid, short-lived (minutes to days), relatively uniform response to acute injury characterized by accumulations of fluid, plasma proteins, and neutrophilic leukocytes.
  • removal of the stimulus halts the recruitment of monocytes (which become macrophages under appropriate activation) into the inflamed tissue, and existing macrophages exit the tissue via lymphatics.
  • injurious agents that cause acute inflammation include, but are not limited to, pathogens (e.g., bacteria, viruses, parasites), foreign bodies from exogenous (e.g., asbestos) or endogenous (e.g., urate crystals, immune complexes), sources, and physical (e.g., burns) or chemical (e.g., caustics) agents.
  • pathogens e.g., bacteria, viruses, parasites
  • foreign bodies from exogenous e.g., asbestos
  • endogenous e.g., urate crystals, immune complexes
  • sources e.g., burns
  • chemical agents e.g., caustics
  • vasodilation which results in a net increase in blood flow, is one of the earliest s physical responses to acute tissue injury
  • endothelial cells lining the venules contract widening the intracellular junctions to produce gaps, leading to increased vascular permeability, which permits leakage of plasma proteins and blood cells out of blood vessels
  • inflammation often is characterized by a strong infiltration of leukocytes at the site of inflammation, particularly neutrophils (polymorphonuclear cells).
  • ARDS acute respiratory distress syndrome
  • ARDS refers to a life- threatening lung condition that prevents enough oxygen from getting to the lungs and into the blood.
  • ARDS is also referred to as noncardiogenic pulmonary edema, increased-permeability pulmonary edema, stiff lung, shock lung, or acute lung injury.
  • ARDS can be caused by any major injury to the lung. Some common causes include, without limitation: breathing vomit into the lungs (aspiration), inhaling chemicals, lung transplant, pneumonia, septic shock (infection throughout the body), and trauma.
  • administering concurrently or “co administering” and the like refer to the administration of a single composition containing two or more actives, or the administration of each active as separate compositions and/or delivered by separate routes either contemporaneously or simultaneously or sequentially within a short enough period of time that the effective result is equivalent to that obtained when all such actives are administered as a single composition.
  • simultaneous is meant that the active agents are administered at substantially the same time, and desirably together in the same formulation.
  • temporary it is meant that the active agents are administered closely in time, e.g., one agent is administered within from about one minute to within about one day before or after another. Any contemporaneous time is useful.
  • the agents when not administered simultaneously, the agents will be administered within about one minute to within about eight hours and suitably within less than about one to about four hours. When administered contemporaneously, the agents are suitably administered at the same site on the subject.
  • the term "same site” includes the exact location but can be within about 0.5 to about 15 centimeters, preferably from within about 0.5 to about 5 centimeters.
  • the term "separately” as used herein means that the agents are administered at an interval, for example at an interval of about a day to several weeks or months.
  • the active agents may be administered in either order.
  • the term “sequentially” as used herein means that the agents are administered in sequence, for example at an interval or intervals of minutes, hours, days or weeks. If appropriate the active agents may be administered in a regular repeating cycle.
  • antagonist refers to a substance that prevents, blocks, inhibits, neutralizes, reduces, restores, or stimulates return to or towards restoration of, a biological activity or effect of another molecule, such as an enzyme or receptor.
  • selective antagonist refers to a compound with high selectivity for its target ⁇ e.g. for a TLR such as TLR7). Selectivity of a particular antagonist is defined as a ratio of the IC50 values of the particular antagonist for the target of interest versus another target.
  • an antagonist that is selective for target A will have an IC50 value for target A lower than that for target B (e.g., TLR8 and/or TLR9), representative examples of which include wherein the IC50 value for target A is at least 10 times lower, at least 100 times lower, at least 1000 times lower or at least 10,000 times lower than the IC50 value of the same antagonist for target B.
  • the antagonist may be a direct or indirect antagonist.
  • direct TLR7 antagonist refers to an antagonist that acts via contact (e.g., binding) with TLR7, i.e., the TLR7 antagonist binds to TLR7 and inhibits its activity and/or activation.
  • an "indirect TLR7 antagonist” may act without contacting TLR7 protein.
  • antisense RNA can be used to decrease expression of the TLR7 gene, or a small molecule can antagonize the effects of TLR7 via interactions with downstream signaling pathway members; these do not interact directly with the TLR7 protein.
  • an indirect antagonist in contrast to a direct antagonist, acts upstream or downstream from the TLR7 protein.
  • the term "antagonist”, including “selective antagonist” includes regulating or modulating the activity of TLR7 to maintain, return to, or stimulate a return towards, immune homeostasis. Hence, total or complete abrogation of TLR7 activity is not necessarily a requirement for TLR7 antagonism.
  • antigens refer to a compound, composition, or substance that may be specifically bound by the products of specific humoral or cellular immunity, such as an antibody molecule or T-cell receptor.
  • Antigens can be any type of molecule including, for example, haptens, simple intermediary metabolites, sugars (e.g., oligosaccharides), lipids, and hormones as well as macromolecules such as complex carbohydrates (e.g., polysaccharides), phospholipids, and proteins.
  • antigens include, but are not limited to, viral antigens, bacterial antigens, fungal antigens, protozoa and other parasitic antigens, tumor antigens, antigens involved in autoimmune disease, allergy and graft rejection, toxins, and other miscellaneous antigens.
  • a “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art, which can be generally sub-classified as follows:
  • Conservative amino acid substitution also includes groupings based on side chains.
  • a group of amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having aliphatic-hydroxyl side chains is serine and threonine; a group of amino acids having amide-containing side chains is asparagine and glutamine; a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan; a group of amino acids having basic side chains is lysine, arginine, and histidine; and a group of amino acids having sulfur-containing side chains is cysteine and methionine.
  • Amino acid substitutions falling within the scope of the present disclosure are, in general, accomplished by selecting substitutions that do not differ significantly in their effect on maintaining (a) the structure of the peptide backbone in the area of the substitution, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. After the substitutions are introduced, the variants are screened for biological activity.
  • nucleic acid sequence that displays substantial sequence identity to a reference nucleic acid sequence ⁇ e.g., at least about 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 97, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98
  • amino acid sequence refers to an amino acid sequence that displays substantial sequence similarity or identity to a reference amino acid sequence.
  • amino acid sequence will display at least about 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 97, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% or even up to 100% sequence similarity or identity to at least a portion of the reference amino acid sequence.
  • cytokine release syndrome refers to a form of systemic inflammatory response syndrome (SIRS) that can be triggered by a variety of factors such as infections and certain drugs. It refers to cytokine storm syndromes (CSS) and occurs when large numbers of white blood cells are activated and release inflammatory cytokines, which in turn activate yet more white blood cells. CRS is also an adverse effect of some monoclonal antibody medications, as well as adoptive T-cell therapies. When occurring as a result of a medication, it is also known as an infusion reaction.
  • the term cytokine storm is often used interchangeably with CRS but, despite the fact that they have similar clinical phenotype, their characteristics are different. When occurring as a result of a therapy, CRS symptoms may be delayed until days or weeks after treatment. Immediate-onset CRS is a cytokine storm, although severe cases of CRS have also been called cytokine storms.
  • cytokine storm refers to an excessively activated cytokine cascade or hypercytokinemia, i.e., an excessive or uncontrolled release of pro- inflammatory cytokines, which can be associated with a wide variety of infectious and noninfectious diseases or disorders.
  • Cytokine storm syndromes are associated with a group of disorders (such as, but not limited to, influenza, asthma, hantavirus pulmonary syndrome.
  • SIRS macrophage activation syndrome
  • SARS SARS
  • COVID-19 COVID-19
  • disseminated vascular coagulopathy DIC
  • the present disclosure does not depend on any particular one of these underlying causes, but is directed to preventing or delaying the onset of cytokine storm, or treating cytokine storm, arising from any underlying inflammatory cause.
  • the primary symptoms of a cytokine storm are high fever, swelling and redness, extreme fatigue and nausea.
  • the immune reaction can result in bleeding, clotting, internal organ injury, or shock, and may be fatal.
  • delaying progression of a disease or “decreasing the rate of progression of a disease” means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated.
  • an effective amount in the context of treating or preventing a condition is meant the administration of an amount of an agent or composition to an individual in need of such treatment or prophylaxis, either in a single dose or as part of a series, that is effective for the prevention of incurring a symptom, holding in check such symptoms, and/or treating existing symptoms, of that condition.
  • the effective amount will vary depending upon the health and physical condition of the individual to be treated, the taxonomic group of individual to be treated, the formulation of the composition, the assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials.
  • Non-limiting symptoms of coronavirus infections include acute febrile illness, malaise, fatigue, headache, flushing, diarrhea, nausea, vomiting, coughing including dry coughing, sore throat, runny nose, nasal congestion, and, in severe disease, symptoms of systemic inflammatory response syndrome including production of pro-inflammatory mediators, vascular leakage and organ failure.
  • An "effective amount” may also be measured by amelioration of indirect or direct symptoms such as reduction, loss, modification or distortion of olfactory sensation.
  • RNA transcript ⁇ e.g., mRNA, antisense RNA, siRNA, shRNA, miRNA, etc.
  • expression of a coding sequence results from transcription and translation of the coding sequence.
  • expression of a non-coding sequence results from the transcription of the non-coding sequence.
  • Immuno homeostasis refers to physiological equilibrium between “attack and defense” mechanism against pathogens, foreign or diseased tissues, and “tolerance” for self. This equilibrium is maintained by checks and balances involving many immune cell types and soluble mediators. Dysregulation of immune homeostasis compromises the body's defense and self/non-self-recognition leading to disease or autoimmunity.
  • an immune response refers to a response by the immune system of a subject.
  • an immune response may be to an antigen/immunogen that the subject's immune system recognizes as foreign (e.g., non-self-antigens) or self (e.g., self-antigens recognized as foreign).
  • Immune responses may be humoral, involving production of immunoglobulins or antibodies, or cellular, involving various types of B and T lymphocytes, dendritic cells, macrophages, antigen presenting cells and the like, or both. Immune responses may also involve the production or elaboration of various effector molecules such as cytokines.
  • immune response encompasses immunogenic responses that cause, activate, elicit, stimulate, or induce an immune response against a particular antigen (e.g., a pathogenic antigen) or organism (e.g., a pathogenic microorganism) in a subject, as well as immunosuppressive or tolerogenic immune responses that inhibit, suppress, diminish or eliminate an immune response, or render the immune system unresponsive, or delay the occurrence or onset of an immune response, to an allergen, or to a self-antigen or a cell, tissue or organ that expresses such an antigen.
  • a particular antigen e.g., a pathogenic antigen
  • organism e.g., a pathogenic microorganism
  • an immune response is one that includes an immunosuppressive or tolerogenic immune response that inhibits, suppresses, diminishes or eliminates a humoral and/or cellular immune response, including the production of autoantibodies, to a self-antigen or a cell, tissue or organ that expresses such an antigen.
  • the term "immunity” refers to protection from disease (e.g., preventing or attenuating (e.g., suppression) of a sign, symptom or condition of the disease) upon exposure to a microorganism (e.g., pathogen) capable of causing the disease.
  • Immunity can be innate (e.g., non-adaptive (e.g., non-acquired) immune responses that exist in the absence of a previous exposure to an antigen) and/or acquired/adaptive (e.g., immune responses that are mediated by B and T cells following a previous exposure to antigen (e.g., that exhibit increased specificity and reactivity to the antigen)).
  • immunogen refers to a molecule which stimulates a response from the adaptive immune system, which may include responses drawn from the group comprising an antibody response, a cytotoxic T cell response, a T helper response, and a T cell memory response.
  • An immunogen may stimulate an upregulation of the immune response with a resultant inflammatory response, or may result in down regulation or immunosuppression.
  • MIS-C multisystem inflammatory syndrome in children
  • MIS-C multisystem inflammatory syndrome in children
  • body parts including the heart, lungs, kidneys, brain, skin, eyes, or gastrointestinal organs.
  • MIS-C may have a fever and various symptoms, including abdominal (gut) pain, vomiting, diarrhea, neck pain, rash, bloodshot eyes, or feeling extra tired.
  • oxidative stressor refers to a molecule that generates oxidative stress in cells including ROS and ROS-generating enzymes such as NOX2 and NCF1.
  • patient refers to any subject, particularly a vertebrate subject, and even more particularly a mammalian subject, for whom therapy or prophylaxis is desired.
  • Suitable vertebrate animals that fall within the scope of the disclosure include, but are not restricted to, any member of the subphylum Chordata including primates (e.g., humans, monkeys and apes, and includes species of monkeys such from the genus Macaca (e.g., cynomolgus monkeys such as Macaca fascicularis, and/or rhesus monkeys ( Macaca mulatta )) and baboon ( Papio ursinus), as well as marmosets (species from the genus Callithrix), squirrel monkeys (species from the genus Saimiri ) and tamarins (species from the genus Saguinus), as well as species of apes such as chimpanzees ( Pan troglodytes)), rodents (e.g., mice rats, guinea pigs), lagomorphs (e.g., rabbits, hares), bovines (e.g.
  • pharmaceutically acceptable carrier a pharmaceutical vehicle comprised of a material that is not biologically or otherwise undesirable, i.e., the material may be administered to a subject along with the selected active agent without causing any or a substantial adverse reaction.
  • Carriers may include excipients and other additives such as diluents, fillers, detergents, coloring agents, wetting or emulsifying agents, pH buffering agents, preservatives and the like.
  • a "pharmacologically acceptable" salt, ester, amide, prodrug or derivative of a compound as provided herein is a salt, ester, amide, prodrug or derivative that this not biologically or otherwise undesirable.
  • composition or “pharmaceutical formulation” refers to a preparation which is in such a form as to permit the biological activity of the active ingredient(s) to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the composition or formulation would be administered.
  • Such formulations are typically sterile.
  • pro-inflammatory mediator means an immunoregulatory agent that favors inflammation.
  • agents include, cytokines such as chemokines, interleukins (IL), lymphokines, and tumor necrosis factor (TNF) as well as growth factors and oxidative stressors.
  • the pro-inflammatory mediator is a "pro-inflammatory cytokine”.
  • pro-inflammatory cytokines include IL-la, IL-Ib, IL-6, and TNF-a, which are largely responsible for early responses.
  • pro-inflammatory mediators include, but are not limited to, LIF, IFN-y, IFN- b, IFN-a, OSM, CNTF, TGF-.b, GM-CSF, TWEAK, IL-11, IL-12, IL-15, IL-17, IL-18, IL-19, IL-20, IL- 8, IL-16, IL-22, IL-23, IL-31 and IL-32 (Tato et al., 2008. Cell 132:900; Cell 132:500, Cell 132:324).
  • Pro-inflammatory mediators may act as endogenous pyrogens (IL-1, IL-6, IL-17, TNF- a), up-regulate the synthesis of secondary mediators and pro-inflammatory cytokines by both macrophages and mesenchymal cells (including fibroblasts, epithelial and endothelial cells), stimulate the production of acute phase proteins, or attract inflammatory cells.
  • pro-inflammatory cytokine relates to IFN-y, IFN-b, TNF-a, IL-Ib, IL-6, IL- 17A, CCL3 and CXCL2.
  • polypeptide As used herein, the terms “polypeptide”, “proteinaceous molecule”, “peptide” and “protein” are used interchangeably to refer to a polymer of amino acid residues and to variants and synthetic analogs of the same. Thus, these terms apply to amino acid polymers in which one or more amino acid residues is a synthetic non-naturally-occurring amino acid, such as a chemical analogue of a corresponding naturally-occurring amino acid, or a PEG group, as well as to naturally-occurring amino acid polymers. These terms do not exclude modifications, for example, glycosylations, acetylations, phosphorylations and the like. Soluble forms of the subject proteinaceous molecules are particularly desirable. Included within the definition are, for example, polypeptides containing one or more analogues of an amino acid including, for example, unnatural amino acids, polypeptides with substituted linkages and polypeptides with PEG groups and lipophilic moieties.
  • prevent refers to a decrease in the occurrence of disease symptoms in a patient. Prevention may be complete (no detectable symptoms) or partial, such that fewer symptoms are observed than would likely occur absent treatment. In some embodiments, prevent refers to inhibiting the development of a disease, disorder or condition, slowing the progression of a disease, disorder or condition or inhibiting progression thereof to a harmful or otherwise undesired state.
  • ROS reactive oxygen species
  • salts and “prodrugs” include any pharmaceutically acceptable salt, ester, hydrate or any other compound which, upon administration to the recipient, is capable of providing (directly or indirectly) a proteinaceous molecule of the invention, or an active metabolite or residue thereof.
  • pharmaceutically acceptable salts refers without limitation to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form ⁇ e.g. by reacting the free base group with a suitable organic acid).
  • Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate,
  • alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.
  • the pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
  • sequence identity refers to the extent that sequences are identical on a nucleotide-by-nucleotide basis or an amino acid-by-amino acid basis over a window of comparison.
  • a "percentage of sequence identity” is calculated by comparing two optimally aligned sequences over the window of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G, I) or the identical amino acid residue (e.g., Ala, Pro, Ser, Thr, Gly, Val, Leu, lie, Phe, Tyr, Trp, Lys, Arg, His, Asp, Glu, Asn, Gin, Cys and Met) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison (/.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity.
  • the identical nucleic acid base e.g., A, T, C, G, I
  • the identical amino acid residue e.g., Ala, Pro, Ser, Thr, Gly, Val, Leu, lie, Phe, Tyr, Trp, Lys, Arg, His
  • sequence identity will be understood to mean the “match percentage” calculated by an appropriate method.
  • sequence identity analysis may be carried out using the DNASIS computer program (Version 2.5 for windows; available from Hitachi Software engineering Co., Ltd., South San Francisco, California, USA) using standard defaults as used in the reference manual accompanying the software.
  • Similarity refers to the percentage number of amino acids that are identical or constitute conservative substitutions as defined in Tables A and B supra. Similarity may be determined using sequence comparison programs such as GAP (Deveraux et a/. 1984, Nucleic Acids Research 12: 387-395). In this way, sequences of a similar or substantially different length to those cited herein might be compared by insertion of gaps into the alignment, such gaps being determined, for example, by the comparison algorithm used by GAP.
  • references to describe sequence relationships between two or more polynucleotides or polypeptides include “reference sequence”, “comparison window”, “sequence identity”, “percentage of sequence identity” and “substantial identity”.
  • a “reference sequence” is at least 12 but frequently 15 to 18 and often at least 25 monomer units, inclusive of nucleotides and amino acid residues, in length.
  • two polynucleotides may each comprise (1) a sequence (/.e., only a portion of the complete polynucleotide sequence) that is similar between the two polynucleotides, and (2) a sequence that is divergent between the two polynucleotides
  • sequence comparisons between two (or more) polynucleotides are typically performed by comparing sequences of the two polynucleotides over a "comparison window" to identify and compare local regions of sequence similarity.
  • a “comparison window” refers to a conceptual segment of at least 6 contiguous positions, usually about 50 to about 100, more usually about 100 to about 150 in which a sequence is compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • the comparison window may comprise additions or deletions (/ ' .e., gaps) of about 20% or less as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • Optimal alignment of sequences for aligning a comparison window may be conducted by computerized implementations of algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package Release 7.0, Genetics Computer Group, 575 Science Drive Madison, WI, USA) or by inspection and the best alignment (/.e., resulting in the highest percentage homology over the comparison window) generated by any of the various methods selected.
  • GAP Garnier et al.
  • BESTFIT Pearson FASTA
  • FASTA Altschul et al.
  • TFASTA Pearson's Alignment of Altschul et al.
  • a detailed discussion of sequence analysis can be found in Unit 19.3 of Ausubel et al., "Current Protocols in Molecular Biology," John Wiley 8i Sons Inc, 1994-1998, Chapter 15.
  • single dose and “single dose treatment” are used interchangeably herein to refer to an effective amount of a drug that is to be taken at one time.
  • a single dose is administered only once during the course of treatment of a condition.
  • small molecule refers to a compound that has a molecular weight of less than 3 kilodaltons (kDa), and typically less than 1.5 kDa, and more preferably less than about 1 kDa.
  • Small molecules may be nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic (carbon-containing) or inorganic molecules.
  • extensive libraries of chemical and/or biological mixtures often fungal, bacterial, or algal extracts, may be screened with any of the assays of the invention to identify compounds that modulate a bioactivity.
  • a "small organic molecule” is an organic compound (or organic compound complexed with an inorganic compound ⁇ e.g., metal)) that has a molecular weight of less than 3 kDa, less than 1.5 kDa, or even less than about 1 kDa.
  • Hybridization generally depends on the ability of denatured DNA to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature which can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so.
  • stringency of hybridization reactions see Ausubel et a/., Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995).
  • "Stringent conditions” or “high stringency conditions”, as defined herein, can be identified by those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficol I/O.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C; or (3) overnight hybridization in a solution that employs 50% formamide, 5xSSC (0.75 M NaCI, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5xDenhardt's solution, sonicated salmon sperm DNA (50 pg/mL),
  • systemic inflammatory response syndrome refers to a clinical response arising from a non-specific insult with two or more of the following measureable clinical characteristics; a body temperature greater than 38° C or less than 36° C, a heart rate greater than 90 beats per minute, a respiratory rate greater than 20 per minute, a white blood cell count (total leukocytes) greater than 12,000 per mm 3 or less than 4,000 per mm 3 , or a band neutrophil percentage greater than 10%. From an immunological perspective, it may be seen as representing a systemic response to an infectious (e.g., pathogenic microbe) or non-infectious insult (e.g., major surgery) or systemic inflammation.
  • infectious e.g., pathogenic microbe
  • non-infectious insult e.g., major surgery
  • Confirmation of infection can be determined using any suitable procedure known in the art, illustrative examples of which include blood culture, nucleic acid detection (e.g., PCR, mass spectroscopy, immunological detection (e.g., ELISA), isolation of bacteria from infected cells, cell lysis and imaging techniques such as electron microscopy.
  • nucleic acid detection e.g., PCR
  • mass spectroscopy e.g., mass spectroscopy
  • immunological detection e.g., ELISA
  • isolation of bacteria from infected cells e.g., cell lysis
  • imaging techniques such as electron microscopy.
  • TLR7 refers to the product of the TLR7 gene, as well as homologs, orthologs, isoforms, precursor forms, processed (e.g., mature) forms, mutants, variants, derivatives, splice variants, alleles, and active fragments thereof.
  • Representative TLR7 amino acid sequences are presented in UniProt Accession Q9NYK1 and GenPept Accession NP_057646.
  • treatment refers to clinical intervention designed to alter the natural course of the individual or cell being treated during the course of clinical pathology. Desirable effects of treatment include decreasing the rate of disease progression, ameliorating or palliating the disease state, and remission or improved prognosis.
  • an individual is successfully "treated” if one or more symptoms associated with an acute inflammatory condition as described herein are mitigated or eliminated, including, but are not limited to, reducing exudation of fluids, including plasma proteins; and leukocytic migration into damaged or inflamed tissue, reducing classic signs of inflammation such as pain, heat, redness, swelling, and loss of function, reducing dilatation of arterioles, capillaries, and venules, and/or reducing vasculature permeability and blood flow, reducing pathogen infection, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, and/or prolonging survival of individuals.
  • Other conditions potentially ameliorated include reduced, abrogated, modified or distorted olfactory sensation, and stress on the cardiovascular system.
  • vaccine and "immunogenic composition” are used interchangeably herein to refer to a composition comprising at least one antigen which, upon inoculation into a subject, induces an immune response specific for that antigen or a cell or organism expressing the antigen and thereby confers protective immunity to the vaccinated subject against the antigen or a cell or organism expressing the antigen.
  • vaccine or "immunogenic composition” comprising, e.g., a live, attenuated virus, inoculation into a subject induces a complete or partial immunity to the pathogenic version of the virus, and/or alleviates the symptoms of disease caused by pathogenic versions of the virus.
  • the protective effects of a vaccine against a virus are normally achieved by inducing in the subject an immune response, either a cell-mediated or a humoral immune response, or a combination of both.
  • an immune response either a cell-mediated or a humoral immune response, or a combination of both.
  • abolished or reduced incidence of viral infection, amelioration of symptoms, or accelerated elimination of the viruses from infected subjects are indicative of the protective effects of the vaccine.
  • underscoring or italicizing the name of a gene shall indicate the gene, in contrast to its protein product, which is indicated by the name of the gene in the absence of any underscoring or italicizing.
  • TLR7 shall mean the TLR7 gene
  • TLR7 shall indicate the protein product or products generated from transcription and translation and/or alternative splicing of the "TLR7" gene.
  • TLR7 selective antagonism of TLR7 is useful for decreasing morbidity (e.g., weight loss), lowering viral titers, inhibiting expression of pattern recognition receptors (e.g., TLR7, TLR8 and TLR9), inhibiting expression of oxidative stressors (e.g., NOX2, NCF1 and related production of ROS), lowering the level of furin, and reducing the release of pro-inflammatory cytokines (e.g., IFN-y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2) in subjects with active RNA virus infections, including highly pathogenic H1N1 IAV infections.
  • pattern recognition receptors e.g., TLR7, TLR8 and TLR9
  • oxidative stressors e.g., NOX2, NCF1 and related production of ROS
  • pro-inflammatory cytokines e.g., IFN-y, IFN-b, TNF-
  • TLR7 antagonist administration of TLR7 antagonist at five days post infection with H1N1 IAV (which is well after release of pro-inflammatory cytokines into lung tissue induced by the virus) was effective in treating the acute inflammatory condition and associated infection. Strikingly, it was also found that a single dose of a selective TLR7 antagonist could ameliorate the symptoms of acute inflammation in subjects with active H1N1 IAV infection or active rhinovirus IB strain (RV1B) infection.
  • selective TLR7 antagonism was shown to be effective in reducing the level of oxidative stress mediators, as well as reducing the level of pro-inflammatory cytokines after their release into tissue, it is proposed that selective TLR7 antagonists can be used to treat acute inflammatory conditions generally, particularly those caused or exacerbated at least in part by activation of TLR7, including acute inflammatory conditions associated with the presence of cytokine release syndrome or a cytokine storm.
  • compositions and methods that take advantage of a selective TLR7 antagonist to reduce or inhibit the production of pro-inflammatory mediators, and to treat or hinder the development of acute inflammatory conditions in individuals that have acute inflammation, as described hereafter.
  • selective antagonism of TLR7 does not necessarily require complete abrogation of TLR7 activity; rather it encompasses modulation of TLR7 activity to effect any one or more of activities 1) to 5) listed above, and to maintain, return to, or stimulate a return towards, immune homeostasis.
  • Selective TLR7 antagonists with such properties are also referred to herein as "immune homeostasis-supporting agents”.
  • Selective TLR7 antagonists include and encompass any active agent that reduces the accumulation, function or stability of TLR7; or decrease expression of the TLR7 gene, and such inhibitors include without limitation, small molecules and macromolecules such as nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, polysaccharides, lipopolysaccharides, lipids or other organic (carbon containing) or inorganic molecules.
  • Selective TLR7 antagonists may be direct TLR7 antagonists or indirect TLR7 antagonists.
  • the selective TLR7 antagonist is an antagonistic nucleic acid molecule that functions to inhibit the transcription or translation of TLR7 transcripts.
  • Representative transcripts of this type include nucleotide sequences corresponding to any one the following sequences: (1) human TLR7 nucleotide sequences as set forth for example in GenBank Accession Nos. NM_016562; (2) nucleotide sequences that share at least 70, 71, 72, 73, 74, 75,
  • nucleotide sequences that hybridize under high stringency conditions to the sequences referred to in (1) (3) nucleotide sequences that encode human TLR7 amino acid sequences as set forth for example in GenPept Accession NP_057646 and UniProt Accession Q9NYK1; (5) nucleotide sequences that encode an amino acid sequence that shares at least 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83,
  • nucleotide sequences that encode an amino acid sequence that shares at least 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86,
  • Illustrative antagonist nucleic acid molecules include antisense molecules, ribozymes and triplex forming molecules, as well as shRNA molecules, siRNA molecules and external guide sequences.
  • Representative TLR7 inhibitory RNA molecules are available commercially from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA) and OriGene Technologies, Inc. (Rockville, MD, USA).
  • TLR7 antagonist oligonucleotides are disclosed for example by Uhlmann et al. in U.S. Publication No. 20160355822.
  • Illustrative oligonucleotides of this type are represented by formula (I):
  • Py is 5-substituted cytidine selected from the group consisting of 5-methyl-dC, 5- bromo-dC and 5-octadienyl-dC,
  • Pu is a 7-deaza purine derivative selected from the group consisting of 7-deaza- dG, 7-deaza-2'-0-methyl-G, inosine and 7-deaza-inosine,
  • Px is dA, 5-substituted deoxyuridine, or 5-iodo-uridine
  • XI is any nucleotide or no nucleotide.
  • each of Q and N is a nucleotide or nucleotide analog
  • x is an integer from 3 to 50
  • z is 0, 1 or 2
  • the polynucleotide does not comprise a CG dinucleotide.
  • the oligonucleotide comprises, consists or consists essentially of 5'-TICTCCTCCTTGAGIAII-3' [SEQ ID NO:l] or 5'-TICTTCTCCTTGAGIAII-3' [SEQ ID NO:2].
  • the present disclosure further contemplates proteinaceous molecules that selectively antagonize TLR7.
  • Exemplary proteinaceous molecules of this type include "decoy” (or "trap") peptides disclosed by Selemidis et at. in International Publication WO 2019/000045, which is incorporated herein by reference in its entirety.
  • proteinaceous molecules which are also referred to herein as "immune homeostasis-supporting agents", antagonize disulfide bond formation between C98 and C475 of human (or murine) TLR7 or their corresponding positions in the TLR7s of other species, and are generally from about 4 to about 190 amino acids in length and comprise, consist or consist essentially of an amino acid sequence with at least 70%, 75%, 80%, 85%.90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%, or 100%, sequence similarity or identity to up to 190 contiguous amino acids between amino acids 4 to 194 of TLR7 human (or murine), which includes a peptide comprising, consisting or consisting essentially of an amino acid sequence with at least 70% 70%, 75%, 80%, 85%.90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%, or 100%, sequence similarity or identity to up to
  • Zi and Z 2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
  • Xi is a hydrophobic amino acid (e.g., L, F or M, or modified forms thereof);
  • X2 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
  • X3 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid (e.g., A or P, or modified forms thereof); and X4 is small amino acid (e.g ., P, or modified forms thereof), a hydrophobic amino acid ( e.g ., L, or modified forms thereof) or a basic amino acid ⁇ e.g., K or R, or modified forms thereof).
  • X3 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid (e.g., A or P, or modified forms thereof)
  • X4 is small amino acid (e.g ., P, or modified forms thereof), a hydrophobic amino acid ( e.g ., L, or modified forms thereof) or a basic amino acid ⁇ e.g., K or R, or modified forms thereof).
  • a proteinaceous molecule represented by formula IV comprises, consists or consists essentially of the amino acid sequence DFRCNCVPIP [SEQ ID NO:3] corresponding to human TLR7 from D95 to P104, or DLRCNCVPVL [SEQ ID NO:4] corresponding to murine TLR7 from D95 to L104.
  • Additional amino acids or other substituents may be added to the N- or C- termini, if present, of the proteinaceous molecules.
  • the proteinaceous molecules may form part of a longer sequence with additional amino acids added to either or both of the N- and C- termini.
  • proteinaceous molecules with high levels of stability may be desired, for example, to increase the half-life of the proteinaceous molecule in a subject.
  • proteinaceous molecules comprise a stabilizing moiety or protecting moiety.
  • the stabilizing moiety or protecting moiety may be coupled at any point on the molecule.
  • Suitable stabilizing or protecting moieties include, but are not limited to, polyethylene glycol (PEG), a glycan or a capping moiety, including an acetyl group, pyroglutamate or an amino group.
  • PEG polyethylene glycol
  • glycan glycan
  • capping moiety including an acetyl group, pyroglutamate or an amino group.
  • the acetyl group and/or pyroglutamate are coupled to the N-terminal amino acid residue of the proteinaceous molecule.
  • the N-terminus of the proteinaceous molecule is an acetamide.
  • the amino group is coupled to the C-terminal amino acid residue of the proteinaceous molecule.
  • the proteinaceous molecule has a primary, secondary or tertiary amide, a hydrazide or a hydroxamide at the C-terminus; particularly a primary amide at the C-terminus.
  • the PEG is coupled to the N-terminal or C-terminal amino acid residue of the proteinaceous molecule or through the amino group of a lysine side-chain or other suitably modified side-chain that is already present or is artificially incorporated into the proteinaceous molecule.
  • proteinaceous molecules may inherently permeate membranes, membrane permeation may further be increased by the conjugation of a membrane permeating moiety to the proteinaceous molecule.
  • proteinaceous molecules according to formula IV may comprise a membrane permeating moiety.
  • the membrane permeating moiety may be coupled at any point on the proteinaceous molecule.
  • Suitable membrane permeating moieties include lipid moieties, cholesterol and proteins, such as cell penetrating peptides and polycationic peptides; especially lipid moieties.
  • Suitable cell penetrating peptides may include the peptides described in, for example, US 20090047272, US 20150266935 and US 20130136742. Accordingly, suitable cell penetrating peptides may include, but are not limited to, basic poly(Arg) and poly(Lys) peptides and basic poly(Arg) and poly(Lys) peptides containing non-natural analogues of Arg and Lys residues such as YGRKKRPQRRR (HIV TAT47-57; SEQ ID NO: 5), RRWRRWWRRWWRRWRR (W/R;
  • TRSSRAGLQFPVGRVHRLLRK (SEQ ID NO:39); hClock-(amino acids 35-47) (human Clock protein DNA-binding peptide), KRVSRNKSEKKRR (SEQ ID NO:40); MAP (model amphipathic peptide), KLALKLALKALKAALKLA (SEQ ID NO:41); K-FGF, AAVALLPAVLLALLAP (SEQ ID NO:42); Ku70- derived peptide, comprising a peptide selected from the group comprising VPMLKE (SEQ ID NO:43), VPMLK (SEQ ID NO:44), PMLKE (SEQ ID NO:45) or PMLK (SEQ ID NO:46); Prion, Mouse Prpe (amino acids 1-28), MANLGYWLLALFVTMWTDVGLCKKRPKP (SEQ ID NO:47); pVEC, LLIILRRRIRKQAHAHSK (SEQ ID NO:48); Pep-I, KETWW
  • the membrane permeating moiety is a lipid moiety, such as a Cio-C 2 o fatty acyl group, especially stearoyl (octadecanoyl; Cis), palmitoyl (hexadecanoyl; Ci 6 ) or myristoyl (tetradecanoyl; CM) .
  • a Cio-C 2 o fatty acyl group especially stearoyl (octadecanoyl; Cis), palmitoyl (hexadecanoyl; Ci 6 ) or myristoyl (tetradecanoyl; CM) .
  • the membrane permeating moiety may be coupled to the N- or C-terminal amino acid residue or through the amino group of a lysine side-chain that is already present or is artificially incorporated into the proteinaceous molecule or other suitably modified side-chain, especially the N-terminal amino acid residue of the proteinaceous molecule or through the amino group of a lysine side-chain.
  • the membrane permeating moiety is coupled through the amino group of the N-terminal amino acid residue.
  • cholestanol may be employed as the membrane permeating moiety to facilitate uptake of the proteinaceous molecule into cells.
  • cell uptake is facilitated by a NOX2-cholestanol-linker (PEG)-gp91ds-TAT construct and linking cholestanol and PEG to gp91ds-TAT facilities delivery of gp91ds-TAT to the endosome.
  • PEG NOX2-cholestanol-linker
  • TLR7 antagonists may be selected from those disclosed by Bou Karroum et al. ⁇ J Med Chem., 62(15): 7015-7031 (2019)).
  • Representative compounds of this type include imidazo[l,2-a]pyrazines, pyrazolo[l,5-a]quinoxalines, and imidazo[l,5-a]quinoxalines.
  • imidazo[l,2-a]pyrazine compounds are represented by formula (V): wherein:
  • R 1 is an acyl moiety that is suitably selected from: [0109]
  • the pyrazolo[l,5-a]quinoxaline compounds are represented by formula (VI): wherein:
  • R 1 is an acyl moiety that is suitably selected from:
  • R 2 and R 3 are independently H, or methyl.
  • imidazo[l,5-a]quinoxaline compounds are represented by formula (VII): wherein:
  • R 1 is an acyl moiety that is suitably selected from:
  • Preferred small molecule TLR7 antagonists of this type include pyrazolo[l,5- a]quinoxaline compounds according to formula (VI) wherein:
  • R1 is:
  • R 2 and R 3 are each H.
  • compositions or formulations comprising a selective TLR7 antagonist and a pharmaceutically acceptable carrier.
  • These compositions can be prepared by mixing the active ingredients ⁇ e.g., a small molecule, nucleic acid, or proteinaceous molecules) having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)).
  • Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arg
  • Exemplary pharmaceutically acceptable carriers herein further include interstitial drug dispersion agents such as soluble neutral-active hyaluronidase glycoproteins (sHASEGP), for example, human soluble PH-20 hyaluronidase glycoproteins, such as rHuPH20 (HYLENEX®, Baxter International, Inc.).
  • sHASEGP soluble neutral-active hyaluronidase glycoproteins
  • rHuPH20 HYLENEX®, Baxter International, Inc.
  • Certain exemplary sHASEGPs and methods of use, including rHuPH20 are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968.
  • a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
  • the active agents and optional pharmaceutically acceptable carriers are in the form of lyophilized formulations or aqueous solutions.
  • Exemplary lyophilized antibody formulations are described in U.S. Pat. No. 6,267,958.
  • Aqueous antibody formulations include those described in U.S. Pat. No. 6,171,586 and W02006/044908, the latter formulations including a histidine-acetate buffer.
  • compositions disclosed herein may also contain further active ingredients as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
  • Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (e.g., liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems e.g., liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsules.
  • the formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
  • the formulations may be administered systemically or locally.
  • Suitable routes may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • parenteral delivery including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • the pharmaceutical forms suitable for injectable use include sterile injectable solutions or dispersions and sterile powders for the preparation of sterile injectable solutions. Such forms should be stable under the conditions of manufacture and storage and may be preserved against reduction, oxidation and microbial contamination.
  • compositions of the present disclosure may further comprises at least one additional or ancillary therapeutic agent for treating the acute inflammatory condition and in the case of an acute inflammatory condition associated with the presence of a pathogenic infection, the ancillary therapeutic agent may be an antigenic composition for immunizing and eliciting an immune response to the pathogen.
  • the ancillary therapeutic agent comprises EXCOOl (an anti-sense RNA against connective tissue growth factor (CTGF)), AZX100 (a phosphopeptide analog of Heat Shock Protein 20 (HSP20)), PRM-151 (recombinant human serum amyloid P/Pentaxin 2), PXL01 (a synthetic peptide derived from human lactoferrin), DSC127 (an angiotensin analog), RXI-109 (a self-delivering RNAi compound that targets connective tissue growth factor (CTGF)), TCA (trichloroacetic acid), Botulium toxin type A, or a combination thereof.
  • EXCOOl an anti-sense RNA against connective tissue growth factor (CTGF)
  • AZX100 a phosphopeptide analog of Heat Shock Protein 20 (HSP20)
  • PRM-151 recombinant human serum amyloid P/Pentaxin 2
  • PXL01 a synthetic peptide derived from human lactoferrin
  • the ancillary therapeutic agent is an anti-inflammatory agent, representative examples of which include steroidal anti-inflammatory agents such as but not limited to compounds containing a 17-carbon 4-ring system, including sterols, various hormones (as anabolic steroids), and glycosides.
  • steroidal anti-inflammatory agents such as but not limited to compounds containing a 17-carbon 4-ring system, including sterols, various hormones (as anabolic steroids), and glycosides.
  • steroidal anti-inflammatory drugs include, without limitation, corticosteroids such as hydrocortisone, hydroxyltriamcinolone, alpha-methyl dexamethasone, dexamethasone-phosphate, beclomethasone dipropionates, clobetasol valerate, desonide, desoxymethasone, desoxycorticosterone acetate, dexamethasone, dichlorisone, diflucortolone valerate, fluadrenolone, fluclorolone acetonide, flumethasone pivalate, fluosinolone acetonide, fluocinonide, flucortine butylesters, fluocortolone, fluprednidene (fluprednylidene) acetate, flurandrenolone, halcinonide, hydrocortisone acetate, hydrocortisone butyrate, methylprednisolone, triamcinolone acetonide, cortic
  • the anti-inflammatory agent may be a nonsteroidal anti inflammatory agent, non-limiting examples of which include agents that are aspirin-like in their action, including, but not limited to, ibuprofen (ADVIL), naproxen sodium (ALEVE), and acetaminophen (TYLENOL).
  • ADVIL ibuprofen
  • ALEVE naproxen sodium
  • TYLENOL acetaminophen
  • non-steroidal anti-inflammatory agents include, without limitation, oxicams, such as piroxicam, isoxicam, tenoxicam, sudoxicam, and CP-14,304; disalcid, benorylate, trilisate, safapryn, solprin, diflunisal, and fendosal; acetic acid derivatives, such as diclofenac, fenclofenac, indomethacin, sulindac, tolmetin, isoxepac, furofenac, tiopinac, zidometacin, acematacin, fentiazac, zomepirac, clindanac, oxepinac, felbinac, and ketorolac; fenamates, such as mefenamic, meclofenamic, flufenamic, niflumic, and tolfenamic acids; propionic acid
  • the anti-inflammatory agent includes, without limitation, transforming growth factor-beta 3 (TGF-83), an anti-tumor necrosis factor-alpha (TNF-a) agent, an inhibitor or antagonist of IL-6 or IL-6 receptor, IL-1 receptor, IL-Ib, TNF, GM-CSF, IFN-y, JAK- STAT signaling, CCR2, CCR5, complement component C5, IRAK4 and M-CSF receptor, or a combination thereof.
  • TGF-83 transforming growth factor-beta 3
  • TNF-a anti-tumor necrosis factor-alpha
  • the ancillary therapeutic agent is an analgesic agent.
  • the analgesic agent relieves pain by elevating the pain threshold without disturbing consciousness or altering other sensory modalities.
  • the analgesic agent is a non-opioid analgesic, which include natural or synthetic substances that reduce pain but are not opioid analgesics.
  • non-opioid analgesics include, but are not limited to, etodolac, indomethacin, sulindac, tolmetin, nabumetone, piroxicam, acetaminophen, fenoprofen, flurbiprofen, ibuprofen, ketoprofen, naproxen, naproxen sodium, oxaprozin, aspirin, choline magnesium trisalicylate, diflunisal, meclofenamic acid, mefenamic acid, and phenylbutazone.
  • the analgesic is an opioid analgesic, illustrative examples of which include codeine, fentanyl, hydromorphone, levorphanol, meperidine, methadone, morphine, oxycodone, oxymorphone, propoxyphene, buprenorphine, butorphanol, dezocine, nalbuphine, and pentazocine.
  • the ancillary therapeutic agent is an antimicrobial agent, which includes without limitation compounds that kill or inhibit the growth of microorganisms such as viruses, bacteria, yeast, fungi, protozoa, etc. and thus include antibiotics, antifungals, anti protozoa Is, antimalarials, antituberculotics and antivirals.
  • antibiotics include quinolones (e.g., amifloxacin, cinoxacin, ciprofloxacin, enoxacin, fleroxacin, flumequine, lomefloxacin, nalidixic acid, norfloxacin, ofloxacin, levofloxacin, lomefloxacin, oxolinic acid, pefloxacin, rosoxacin, temafloxacin, tosufloxacin, sparfloxacin, clinafloxacin, gatifloxacin, moxifloxacin; gemifloxacin; and garenoxacin), tetracyclines, glycylcyclines and oxazolidinones (e.g., chlortetracycline, demeclocycline, doxycycline, lymecycline, methacycline, minocycline, oxytetracycline, tetracycline, tigecycl
  • antivirals include abacavir sulfate, acyclovir sodium, amantadine hydrochloride, amprenavir, cidofovir, delavirdine mesylate, didanosine, efavirenz, famciclovir, fomivirsen sodium, foscarnet sodium, ganciclovir, indinavir sulfate, lamivudine, lamivudine/zidovudine, nelfinavir mesylate, nevirapine, oseltamivir phosphate, ribavirin, rimantadine hydrochloride, ritonavir, saquinavir, saquinavir mesylate, stavudine, valacyclovir hydrochloride, valganciclovir, zalcitabine, zanamivir, and zidovudine.
  • antiprotozoals include atovaquone, chloroquine hydrochloride, chloroquine phosphate, metronidazole, metronidazole hydrochloride, and pentamidine isethionate.
  • Anthelmintics can be at least one selected from mebendazole, pyrantel pamoate, albendazole, ivermectin and thiabendazole.
  • Illustrative antifungals can be selected from amphotericin B, amphotericin B cholesteryl sulfate complex, amphotericin B lipid complex, amphotericin B liposomal, bifonazole, butoconazole, chlordantoin, chlorphenesin, ciclopirox olamine, clotrimazole, eberconazole, econazole, fluconazole, flucytosine, flutrimazole, griseofulvin microsize, griseofulvin ultramicrosize, itraconazole, isoconazole, itraconazole, ketoconazole, miconazole, nifuroxime, nystatin, terbinafine hydrochloride, tioconazole, terconazole and undecenoic acid.
  • Non-limiting examples of antimalarials include chloroquine hydrochloride, chloroquine phosphate, doxycycline, hydroxychloroquine sulfate, mefloquine hydrochloride, primaquine phosphate, pyrimethamine, and pyrimethamine with sulfadoxine.
  • Antituberculotics include but are not restricted to clofazimine, cycloserine, dapsone, ethambutol hydrochloride, isoniazid, pyrazinamide, rifabutin, rifampin, rifapentine, and streptomycin sulfate.
  • the ancillary agent is an antiviral, such as remdesivir, lopinavir, ritonavir, emtricitabine, tenofovir, ivermectin, faviparavir, imatininb and baricitinib, and combinations thereof (e.g. lopinavir/ritonavir, or emtricitabine/tenofovir).
  • antiviral such as remdesivir, lopinavir, ritonavir, emtricitabine, tenofovir, ivermectin, faviparavir, imatininb and baricitinib, and combinations thereof (e.g. lopinavir/ritonavir, or emtricitabine/tenofovir).
  • compounds which inhibit the cytokine release syndrome or cytokine storm include anti-coagulants and/or platelet aggregation inhibitors that address blood clots, compounds which chelate iron ions released from hemoglobin by viruses such as COVID-19, cytochrome P-450 (CYP450) inhibitors and/or NOX inhibitors, as ancillary therapeutic agents.
  • CYP450 cytochrome P-450
  • NOX inhibitors are disclosed in PCT/US2018/067674, and include AEBSF, Apocynin, DPI, GK-136901, ML171, Plumbagin, S17834, VAS2870, VAS3947, GKT-831, GKT771, GTL003 or amido thiadiazole derivatives thereof, as described in AU2015365465, EP20140198597; and WO2015/59659, Schisandrin B, as described in CN104147001 and CN20131179455), bi-aromatic and tri-aromatic compounds described in U.S. Publication No.
  • EP20080164857 PCT WO200954156, PCT W0200954150, EP20080164853, PCT W0200853390, U.S. Publication No. 20070896284, EP20070109555, PCT WO 200954148, EP20080164847, PCT WO200954155, and EP20080164849, quinazoline and quinoline derivatives disclosed in EP2886120, U.S. Publication No. 2014018384, U.S. Publication No. 20100407925,
  • Exemplary NOX inhibitors also include 2-phenylbenzo[d]isothiazol-3(2H)-one, 2- (4-methoxyphenyl)benzo[d]isothiazol-3(2H)-one, 2-(benzo[d][l,3]dioxol-5-yl)benzo[d]isothiazol- 3(2H)-one, 2-(2,4-dimethylphenyl)benzo[d]isothiazol-3(2H)-one, 2-(4- fluorophenyl)benzo[d]isothiazol-3(2H)-one, 2-(2, 4-dimethyl phenyl)-5-fl uorobenzo[d] isothiazol- 3(2H)-one, 5-fluoro-2-(4-fluorophenyl)benzo[d]isothiazol-3(2H)-one, 2-(2-chloro-6-methylphenyl)- 5-fluorobenzo[d] is
  • Non-limiting examples of CYP450 inhibitors include amiodarone, amlodipine, apigenin, aprepitant, bergamottin (grapefruit), buprenorphine, bupropion, caffeine, cafestol, cannabidiol, celecoxib, chloramphenicol, chlorphenamine, chlorpromazine, cimetidine, cinacalcet, ciprofloxacin, citalopram, clarithromycin, clemastine, clofibrate, clomipramine, clotrimazole, cobicistat, cocaine, curcumin (turmeric), cyclizine, delavirdine, desipramine, disulfiram, diltiazem, diphenhydramine, dithiocarbamate, domperidone, doxepin, doxorubicin, duloxetine, echinacea, entacapone, erythromycin, escitalopram, felba
  • Representative ACE-2 inhibitors include sulfhydryl-containing agents, such as alacepril, captopril (capoten), and zefnopril, dicarboxylate-containing agents, such as enalapril (vasotec), ramipril (altace), quinapril (accupril), perindopril (coversyl), lisinopril (listril), benazepril (lotensin), imidapril (tanatril), trandolapril (mavik), and cilazapril (inhibace), and phosphonate- containing agents, such as fosinopril (fositen/monopril).
  • sulfhydryl-containing agents such as alacepril, captopril (capoten), and zefnopril
  • dicarboxylate-containing agents such as enalapril (vasotec), ramipril (
  • Compounds which inhibit the cytokine storm include compounds that target fundamental immune pathways, such as the chemokine network and the cholinergic anti inflammatory pathway.
  • JAK inhibitors such as JAK 1 and JAK 2 inhibitors, can inhibit the cytokine storm, and in some cases, are also antiviral.
  • Representative JAK inhibitors include those disclosed in U.S. Pat. No.
  • 10,022,378, such as Jakafi, Tofacitinib, and Baricitinib, as well as LY3009104/INCB28050, Pacritinib/SB1518, VX-509, GLPG0634, INC424, R-348, CYT387, TG 10138, AEG 3482, and pharmaceutically acceptable salts and prodrugs thereof.
  • Still further examples include CEP-701 (Lestaurtinib), AZD1480, INC424, R-348, CYT387, TG 10138, AEG 3482, 7-iodo-N-(4-morpholinophenyl)thieno[3,2-d]pyrimidin-2-amine, 7-(4-aminophenyl)-N-(4- morpholinophenyl)thieno[3,2-d]pyrimidin-2-amine, N-(4-(2-(4-morpholinophenylamino)thieno[3,2- d]pyrimidin-7-yl) phenyl) acrylamide, 7-(3-aminophenyl)-N-(4-morpholinophenyl)thieno[3,2- d]pyrimidin-2-amine, N-(3-(2-(4-morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl) phenyl) acrylamide, N-(
  • HMGB1 antibodies and/or COX-2 inhibitors can be used, which downregulate the cytokine storm.
  • examples of such compounds include Actemra (Roche).
  • Celebrex (celecoxib), a COX-2 inhibitor, can be used.
  • IL-8 (CXCL8) inhibitors can also be used.
  • Chemokine receptor CCR2 antagonists such as PF-04178903 can reduce pulmonary immune pathology.
  • Selective TLR7 antagonists can also be co-administered with compounds that inhibit blood clot formation, such as blood thinners, or compounds that break up existing blood clots, such as tissue plasminogen activator (TPA), Integrilin (eptifibatide), abciximab (ReoPro) or tirofiban (Aggrastat).
  • Representative platelet aggregation inhibitors include glycoprotein IIB/IIIA inhibitors, phosphodiesterase inhibitors, adenosine reuptake inhibitors, and adenosine diphosphate (ADP) receptor inhibitors. These can optionally be administered in combination with an anticoagulant.
  • Illustrative anti-coagulants include coumarins (vitamin K antagonists), heparin and derivatives thereof, including unfractionated heparin (UFH), low molecular weight heparin (LMWH), and ultra-low-molecular weight heparin (ULMWH), synthetic pentasaccharide inhibitors of factor Xa, including Fondaparinux, Idraparinux, and Idrabiotaparinux, directly acting oral anticoagulants (DAOCs), such as dabigatran, rivaroxaban, apixaban, edoxaban and betrixaban, and antithrombin protein therapeutics/thrombin inhibitors, such as bivalent drugs hirudin, lepirudin, and bivalirudin and monovalent argatroban.
  • DAOCs directly acting oral anticoagulants
  • antithrombin protein therapeutics/thrombin inhibitors such as bivalent drugs hirudin, lepirudin, and bivalirudin and monovalent argatroban.
  • ancillary therapeutic agents include, without limitation, rose hip oil, vitamin E, 5-fluorouracil, bleomycin, onion extract, pentoxifylline, prolyl-4-hydroxylase, verapamil, tacrolimus, tamoxifen, tretinoin, colchicine, a calcium antagonist, tranilst, zinc, and a combination thereof.
  • the selective TLR7 antagonists of the present disclosure are used in conjunction with an intervention, such as a ventilator.
  • the present disclosure encompasses co-administration of a selective TLR7 antagonist in concert with an ancillary therapeutic agent or intervention, as described for example above and elsewhere herein.
  • a selective TLR7 antagonist in concert with an ancillary therapeutic agent or intervention, as described for example above and elsewhere herein.
  • the dosages of the actives in the combination may on their own comprise an effective amount and the additional agent(s) may further augment the therapeutic benefit to the patient.
  • the selective TLR7 antagonist and the additional agent(s) may together comprise an effective amount for treating an acute inflammatory condition.
  • effective amounts may be defined in the context of particular treatment regimens, including, e.g., timing and number of administrations, modes of administrations, formulations, etc.
  • the selective TLR7 antagonist and optionally the ancillary therapeutic agent are administered on a routine schedule.
  • the ancillary therapeutic agent may be administered as symptoms arise.
  • a "routine schedule" as used herein, refers to a predetermined designated period of time.
  • the routine schedule may encompass periods of time which are identical, or which differ in length, as long as the schedule is predetermined.
  • the routine schedule may involve administration of the selective TLR7 antagonist on a daily basis, every two days, every three days, every four days, every five days, every six days, a weekly basis, a monthly basis or any set number of days or weeks there between, every two months, three months, four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, twelve months, etc.
  • the predetermined routine schedule may involve concurrent administration of the selective TLR7 antagonist and the ancillary therapeutic agent on a daily basis for the first week, followed by a monthly basis for several months, and then every three months after that. Any particular combination would be covered by the routine schedule as long as it is determined ahead of time that the appropriate schedule involves administration on a certain day.
  • the selective TLR7 antagonist is administered in the form of a single dose and suitably only once during the course of treatment.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture (e.g., the concentration of an active agent, which achieves a half-maximal inhibition in TLR7 function or activity). Such information can be used to determine useful doses more accurately in humans.
  • Dosing is dependent on severity and responsiveness of the inflammatory disease state to be treated, with the course of treatment lasting from several days to several months, or until a cure is achieved or a diminution of the disease state is achieved.
  • Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient. Persons of ordinary skill can easily determine optimum dosages, dosing methodologies and repetition rates. Optimum dosages may vary depending on the relative potency of individual selective TLR7 antagonists and can generally be estimated based on ECso s found to be effective in in vitro and in vivo animal models. In general, dosage is from 0.01 pg to 100 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly. Persons of ordinary skill in the art can easily estimate repetition rates for dosing based on measured residence times and concentrations of the drug in bodily fluids or tissues.
  • Toxicity and therapeutic efficacy of selective TLR7 antagonists can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50.
  • Compounds that exhibit large therapeutic indices are preferred.
  • the data obtained from these cell culture assays, and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See for example Fingl et ai., 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 pi).
  • compositions of the present disclosure may be provided in a kit.
  • the kit may comprise additional components to assist in performing the methods of the present disclosure such as, for example, administration device(s), buffer(s), and/or diluent(s).
  • the kits may include containers for housing the various components and instructions for using the kit components in the methods of the present disclosure.
  • selective antagonism of TLR7 in subjects with active RNA virus infections is useful for correcting the host immune response with consequential improved immunity, including enhanced production of antibody, to the virus.
  • the present disclosure contemplates co-administration of antigenic compositions, including immunogenic compositions and vaccines, with a selective TLR7 antagonist to a subject with an acute inflammatory condition associated with RNA virus infection.
  • Representative antigenic compositions encompassed by the present disclosure include but are not limited to killed or attenuated live viral vaccines, live-vectored vaccines, subunit vaccines, virus-like particle vaccines, and DNA or RNA vaccines. See Roth et al., New Technology For Improved Vaccine Safety And Efficacy", Veterinary Clinics North America: Food Animal Practice 17(3): 585-597 (2011).
  • the RNA virus to which the antigenic composition elicits an immune response may be a positive sense RNA virus, negative sense RNA virus, or a double-stranded RNA virus.
  • RNA viruses include Picornaviruses (e.g., hepatitis A virus, enteroviruses such as poliovirus, enterovirus 71, 70, 69, and 68, Coxsackieviruses, echoviruses, foot and mouth disease virus, and rhinoviruses), Caliciviruses (e.g., hepatitis E virus, noroviruses such as Norwalk virus, feline calicivirus), Arteriviruses (e.g., equine arteritis virus), Togaviruses (e.g., Sindbis virus, the equine encephalitis viruses, chikungunya virus, rubella virus, Ross River virus, bovine diarrhea virus, hog cholera virus, Semliki forest virus), Flaviviruses (e.g., dengue virus, West Nile virus, yellow fever virus, Japanese encephalitis virus, St.
  • enteroviruses such as poliovirus, enterovirus 71, 70,
  • Coronaviruses e.g., human coronaviruses, including betacoronavi ruses such OC43 and HKU1 of the A lineage, SARS-CoV and SARS-CoV-2 of the B lineage and MERS-CoV of the C lineage, swine gastroenteritis virus
  • Rhabdoviruses e.g., rabies virus, Australian bat lyssavirus, vesicular stomatitis viruses
  • Filoviruses e.g., Marburg virus, Ebola virus
  • Paramyxoviruses e.g., measles virus, canine distemper virus, mumps virus, parainfluenza viruses, respiratory syncytial virus, Newcastle disease virus, rinderpest virus, Nipah virus, Hendra virus
  • Orthomyxoviruses e.g., human influenza viruses, including human influenza virus types A, B and C, avian influenza viruses, equine influenza viruses
  • Bunyaviruses e.g., hantavirus, LaCrosse virus, Rift Valley fever virus
  • Arenaviruses e.g., Lassa virus, Machupo virus
  • Reoviruses e.g., human and animal reoviruses, such as rotaviruses, bluetongue virus
  • Birnaviruses e.g., infectious bursal virus, fish pancreatic necrosis virus
  • Retroviruses
  • Adenoviruses e.g., human, canine, bovine, and porcine adenoviruses
  • Herpesviruses e.g., herpes simplex viruses, varicella-zoster virus, infectious bovine rhinotracheitis virus, cytomegalovirus, human herpesvirus 6, human herpesvirus 7, human herpesvirus 8, Epstein-Barr virus
  • Poxviruses e.g., vaccinia, fowlpoxviruses, raccoon poxvirus, skunkpox virus, monkeypoxvirus, cowpox virus, buffalopox virus, musculum contagiosum virus. 6.
  • TLR7 antagonists optionally in combination with at least one ancillary therapeutic agent, for treating a subject with an acute inflammatory condition, particularly one caused or exacerbated at least in part by activation of TLR7, and optionally one or both of TLR8 and TLR9, and/or an increased level of one or more oxidative stressors ⁇ e.g., NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1; also known as p47phox) and ROS) and/or an increased level of furin.
  • NOX2 NADPH oxidase type 2
  • NCF1 neutrophil cytosol factor 1
  • ROS neutrophil cytosol factor 1
  • the acute inflammatory condition may be associated with presence of a pathogenic infection (e.g., a pathogenic infection in which the pathogen is an RNA virus), which is typically an active infection.
  • a pathogenic infection e.g., a pathogenic infection in which the pathogen is an RNA virus
  • the RNA virus may enter a host cell that it infects by receptor- mediated endocytosis or micropinocytosis. In specific embodiments, the RNA virus enters the endosome of a host cell that it infects.
  • the RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenzavirus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus), Togaviridae (e.g., Chik
  • the RNA virus is an orthomyxovirus (e.g., an influenza virus such as influenza A, influenza B or influenza C) or a coronavirus (e.g., a coronavirus, including betacoronaviruses, capable of causing severe acute respiratory syndrome (SARS) such as MERS-CoV, SARS-CoV and SARS-CoV-2).
  • an influenza virus such as influenza A, influenza B or influenza C
  • a coronavirus e.g., a coronavirus, including betacoronaviruses, capable of causing severe acute respiratory syndrome (SARS) such as MERS-CoV, SARS-CoV and SARS-CoV-2).
  • SARS severe acute respiratory syndrome
  • the acute inflammatory condition is associated with presence of cytokine release syndrome (CRS) or a cytokine storm, which in representative examples comprises an elevation of at least 50% compared to basal state of one or more (e.g., 1, 2, 3, 4, 5, 6, 7 or 8) cytokines selected from IFN- Y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2.
  • the acute inflammatory condition is associated with a condition selected from multisystem inflammatory syndrome in children (MIS-C), systemic inflammatory response syndrome (SIRS), acute respiratory distress syndrome (ARDS), and severe acute respiratory syndrome (SARS).
  • MIS-C multisystem inflammatory syndrome in children
  • SIRS systemic inflammatory response syndrome
  • ARDS acute respiratory distress syndrome
  • SARS severe acute respiratory syndrome
  • the acute inflammatory condition is associated with presence of CRS.
  • the subject has one or more symptoms selected from fever, fatigue, loss of appetite, muscle and joint pain, nausea, vomiting, diarrhea, rashes, fast breathing, rapid heartbeat, low blood pressure, seizures, headache, confusion, delirium, hallucinations, tremor, and loss of coordination, or any combination thereof.
  • the acute inflammatory condition may be associated with presence of a cytokine storm in which the subject in illustrative examples has one or more symptoms selected from high fever, swelling and redness, extreme fatigue, nausea, bleeding, clotting, internal organ injury, and shock, or any combination thereof.
  • the acute inflammatory condition is associated with presence of MIS-C, wherein the subject in non-limiting examples has one or more symptoms selected from fever, vomiting, diarrhea, stomach pain, skin rash, red eyes, redness or swelling of the lips and tongue, feeling unusually tired, redness or swelling of the hands or feet, severe stomach pain, cardiac symptoms, including chest pain, palpitations and shortness of breath, bluish lips or face, mental confusion, inability to wake up or stay awake, abdominal pain with vomiting and diarrhea, skin rash and swelling of extremities, faintness and low blood pressure that is new, or any combination thereof.
  • the subject in non-limiting examples has one or more symptoms selected from fever, vomiting, diarrhea, stomach pain, skin rash, red eyes, redness or swelling of the lips and tongue, feeling unusually tired, redness or swelling of the hands or feet, severe stomach pain, cardiac symptoms, including chest pain, palpitations and shortness of breath, bluish lips or face, mental confusion, inability to wake up or stay awake, abdominal pain with vomiting and diarrhea, skin rash and swelling of extremities,
  • the acute inflammatory condition is associated with SIRS.
  • the subject may have one or more symptoms associated with a particular stage of SIRS, representative ones of which are as follows:
  • Stage 1 is a local reaction at the site of injury that aims at containing the injury and limit spread.
  • Immune effector cells at the site release cytokines that in turn stimulate the reticuloendothelial system promoting wound repair through local inflammation.
  • the leakage of cells and protein-rich fluid in extravascular space causes swelling (tumor) and increased heat (calor).
  • Inflammatory mediators impact the local somatosensory nerves causing pain (dolor) and loss of function (functio laesa). That loss of function also allows the part of the body to repair instead of persistent use.
  • Stage 2 is an early CARS in an attempt to maintain immunological balance.
  • Stage 3 is when the scale tips over towards pro-inflammatory SIRS resulting in progressive endothelial dysfunction, coagulopathy, and activation of the coagulation pathway. It results in end-organ micro thrombosis, and a progressive increase in capillary permeability, eventually resulting in loss of circulatory integrity.
  • Stage 4 is characterized by CARS taking over SIRS, resulting in a state of relative immunosuppression.
  • the individual therefore, becomes susceptible to secondary or nosocomial infections, thus perpetuating the sepsis cascade.
  • Stage 5 manifests in MODS with persistent dysregulation of both SIRS and CARS response.
  • the acute inflammatory condition is associated with ARDS.
  • Representative symptoms of ARDS include one or more of mild, moderate or severe hypoxemia as determined by Partial Pressure of arterial oxygen/Fraction of inspired oxygen (Pa0 2 /Fi0 2 ) or positive end-expiratory pressure (PEEP), bilateral opacities, respiratory failure, shortness of breath, labored breathing, cough, fever, increased heart rate, low blood pressure, confusion, extreme tiredness, rapid breathing, organ failure, chest pain, bluish coloring of nails or lips, an change in the level of one or more inflammatory markers, and need for mechanical ventilation, or any combination thereof.
  • the acute inflammatory condition is associated with SARS and in non-limiting examples the subject has one or more symptoms selected from acute febrile illness, malaise, fatigue, headache, flushing, diarrhea, nausea, vomiting, coughing including dry coughing, sore throat, runny nose, nasal congestion, production of pro-inflammatory mediators, vascular leakage and organ failure, or any combination thereof.
  • a method of treating an acute inflammatory condition in a subject comprising administering to the subject an effective amount of a selective TLR7 antagonist.
  • oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
  • RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
  • RNA virus is an enveloped virus.
  • RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
  • RNA virus enters the endosome of a host cell that it infects.
  • RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenza virus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus), Togavirid
  • Orthomyxoviridae e.g.
  • RNA virus Yellow fever virus, Hepacivirus C
  • Hantaviridae e.g., Hantavirus
  • influenza virus is selected from influenza A, influenza B and influenza C.
  • RNA virus is a coronavirus.
  • betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
  • betacoronavirus is a lineage B betacoronavirus.
  • RNA virus is a picornavirus.
  • emergency warning signs of MIS-C can include: severe stomach pain, cardiac symptoms, including chest pain, palpitations and shortness of breath, bluish lips or face, mental confusion, inability to wake up or stay awake, abdominal pain with vomiting and diarrhea, skin rash and swelling of extremities, faintness and low blood pressure that is new.
  • the selective TLR7 antagonist is selected from nucleic acid antagonists (e.g., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide) and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines).
  • nucleic acid antagonists e.g., antisense molecules, RNAi and external guide sequences
  • proteinaceous antagonists e.g., decoy peptide
  • small molecule antagonists e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines.
  • the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7.
  • the proteinaceous molecule is represented by Formula I:
  • Zi and Z 2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
  • Xi is a hydrophobic amino acid ⁇ e.g., L, F or M, or modified forms thereof);
  • X2 is a hydrophobic amino acid ⁇ e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
  • X3 is a hydrophobic amino acid ⁇ e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid ⁇ e.g., A or P, or modified forms thereof); and
  • X 4 is small amino acid ⁇ e.g., P, or modified forms thereof), a hydrophobic amino acid ⁇ e.g., L, or modified forms thereof) or a basic amino acid ⁇ e.g., K or R, or modified forms thereof).
  • a method of treating cytokine release syndrome (CRS) or a cytokine storm in a subject comprising administering to the subject an effective amount of a TLR7 antagonist.
  • CRS cytokine release syndrome
  • oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
  • RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
  • RNA virus is an enveloped virus.
  • RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
  • RNA virus enters the endosome of a host cell that it infects.
  • RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenza virus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian
  • Orthomyxoviridae e.g., Influenza virus
  • Coronaviridae e.g.,
  • RNA virus is an orthomyxovirus.
  • influenza virus is selected from influenza A, influenza B and influenza C.
  • RNA virus is a coronavirus.
  • coronavirus is capable of causing severe acute respiratory syndrome (SARS).
  • SARS severe acute respiratory syndrome
  • betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
  • RNA virus is a picornavirus.
  • 71 The method of any one of embodiment 45 to 70, wherein the subject has CRS and has one or more symptoms selected from fever, fatigue, loss of appetite, muscle and joint pain, nausea, vomiting, diarrhea, rashes, fast breathing, rapid heartbeat, low blood pressure, seizures, headache, confusion, delirium, hallucinations, tremor, and loss of coordination.
  • 72 The method of any one of embodiment 45 to 70, wherein the subject has a cytokine storm and has one or more symptoms selected from high fever, swelling and redness, extreme fatigue, nausea, bleeding, clotting, internal organ injury, and shock, or any combination thereof.
  • emergency warning signs of multisystem inflammatory syndrome can include: severe stomach pain, cardiac symptoms, including chest pain, palpitations and shortness of breath, bluish lips or face, mental confusion, inability to wake up or stay awake, abdominal pain with vomiting and diarrhea, skin rash and swelling of extremities, faintness and low blood pressure that is new.
  • the selective TLR7 antagonist is selected from nucleic acid antagonists ⁇ e.g., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists ⁇ e.g., decoy peptide) and small molecule antagonists ⁇ e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines).
  • the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7.
  • Zi and Z 2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
  • Xi is a hydrophobic amino acid ⁇ e.g., L, F or M, or modified forms thereof);
  • X2 is a hydrophobic amino acid ⁇ e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
  • X3 is a hydrophobic amino acid ⁇ e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid ⁇ e.g., A or P, or modified forms thereof); and
  • X 4 is small amino acid ⁇ e.g., P, or modified forms thereof), a hydrophobic amino acid ⁇ e.g., L, or modified forms thereof) or a basic amino acid ⁇ e.g., K or R, or modified forms thereof).
  • a method of treating SARS in a subject comprising administering to the subject an effective amount of a TLR7 antagonist.
  • oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
  • RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
  • RNA virus is an enveloped virus.
  • RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
  • RNA virus enters the endosome of a host cell that it infects.
  • RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenzavirus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus),
  • Orthomyxoviridae e.g., Influenzavirus
  • RNA virus is an orthomyxovirus.
  • influenza virus is selected from influenza A, influenza B and influenza C.
  • RNA virus is a coronavirus.
  • betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
  • betacoronavirus is a lineage B betacoronavirus.
  • betacoronavirus is a lineage C betacoronavirus.
  • emergency warning signs of multisystem inflammatory syndrome can include: severe stomach pain, cardiac symptoms, including chest pain, palpitations and shortness of breath, bluish lips or face, mental confusion, inability to wake up or stay awake, abdominal pain with vomiting and diarrhea, skin rash and swelling of extremities, faintness and low blood pressure that is new.
  • the selective TLR7 antagonist is selected from nucleic acid antagonists (e.g., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide) and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines).
  • nucleic acid antagonists e.g., antisense molecules, RNAi and external guide sequences
  • proteinaceous antagonists e.g., decoy peptide
  • small molecule antagonists e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines.
  • the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7.
  • Zi and Z 2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
  • Xi is a hydrophobic amino acid (e.g., L, F or M, or modified forms thereof);
  • X2 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
  • X 3 is a hydrophobic amino acid (e.g ., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid ⁇ e.g., A or P, or modified forms thereof); and
  • X 4 is small amino acid ⁇ e.g., P, or modified forms thereof), a hydrophobic amino acid ⁇ e.g., L, or modified forms thereof) or a basic amino acid ⁇ e.g., K or R, or modified forms thereof).
  • a method of treating ARDS in a subject comprising administering to the subject an effective amount of a TLR7 antagonist.
  • oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
  • RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
  • RNA virus is an enveloped virus.
  • RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
  • RNA virus enters the endosome of a host cell that it infects.
  • RNA virus is selected from the following families: Orthomyxoviridae ⁇ e.g., Influenzavirus), Coronaviridae ⁇ e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus ⁇ e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae ⁇ e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae ⁇ e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae ⁇ e.g., Human metapneumovirus), Rhabdoviridae ⁇ e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus),
  • RNA virus is an orthomyxovirus.
  • influenza virus is selected from influenza A, influenza B and influenza C.
  • RNA virus is a coronavirus.
  • betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
  • betacoronavirus is a lineage B betacoronavirus.
  • betacoronavirus is a lineage C betacoronavirus.
  • RNA virus is a picornavirus.
  • any one of embodiments 124 to 150 wherein the subject has one or more symptoms selected from mild, moderate or severe hypoxemia as determined by Partial Pressure of arterial oxygen/Fraction of inspired oxygen (PaCh/FiCh) or positive end-expiratory pressure (PEEP), bilateral opacities, respiratory failure, shortness of breath, labored breathing, cough, fever, increased heart rate, low blood pressure, confusion, extreme tiredness, rapid breathing, organ failure, chest pain, bluish coloring of nails or lips, an change in the level of one or more inflammatory markers, or need for mechanical ventilation.
  • PaCh/FiCh Partial Pressure of arterial oxygen/Fraction of inspired oxygen
  • PEEP positive end-expiratory pressure
  • the selective TLR7 antagonist is selected from nucleic acid antagonists (e.g., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide), and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines).
  • nucleic acid antagonists e.g., antisense molecules, RNAi and external guide sequences
  • proteinaceous antagonists e.g., decoy peptide
  • small molecule antagonists e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines.
  • the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7.
  • Zi and Z 2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
  • Xi is a hydrophobic amino acid ⁇ e.g., L, F or M, or modified forms thereof);
  • X2 is a hydrophobic amino acid ⁇ e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
  • X3 is a hydrophobic amino acid ⁇ e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid ⁇ e.g., A or P, or modified forms thereof); and
  • X4 is small amino acid ⁇ e.g., P, or modified forms thereof), a hydrophobic amino acid ⁇ e.g., L, or modified forms thereof) or a basic amino acid ⁇ e.g., K or R, or modified forms thereof).
  • a method of treating SIRS in a subject comprising administering to the subject an effective amount of a selective TLR7 antagonist.
  • oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
  • RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
  • RNA virus is an enveloped virus.
  • RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
  • RNA virus enters the endosome of a host cell that it infects.
  • RNA virus is selected from the following families: Orthomyxoviridae ⁇ e.g., Influenzavirus), Coronaviridae ⁇ e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus ⁇ e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae ⁇ e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae ⁇ e.g., Human parainfluenza virus,
  • Orthomyxoviridae ⁇ e.g., Influenzavirus
  • Coronaviridae ⁇ e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus ⁇ e.g., SARS-CoV and SARS-CoV-2)
  • Picornaviridae ⁇ e.g., Rhinovirus, Poliovirus, Enterovirus, Co
  • Rhabdoviridae e.g ., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus
  • Filoviridae Marburg virus, Ebola virus
  • Togaviridae e.g.
  • RNA virus is an orthomyxovirus.
  • influenza virus is selected from influenza A, influenza B and influenza C.
  • RNA virus is a coronavirus.
  • betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
  • betacoronavirus is a lineage B betacoronavirus.
  • betacoronavirus is a lineage C betacoronavirus.
  • RNA virus is a picornavirus.
  • the selective TLR7 antagonist is selected from nucleic acid antagonists (e.g., aptamers, ribozymes and triplex forming molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide) and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxa lines).
  • nucleic acid antagonists e.g., aptamers, ribozymes and triplex forming molecules, RNAi and external guide sequences
  • proteinaceous antagonists e.g., decoy peptide
  • small molecule antagonists e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxa lines.
  • the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7.
  • Zi and Z 2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
  • Xi is a hydrophobic amino acid (e.g., L, F or M, or modified forms thereof);
  • X2 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
  • X3 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid (e.g., A or P, or modified forms thereof); and
  • X4 is small amino acid (e.g., P, or modified forms thereof), a hydrophobic amino acid (e.g., L, or modified forms thereof) or a basic amino acid (e.g., K or R, or modified forms thereof).
  • a method of treating a MIS-C in a subject comprising administering to the subject an effective amount of a selective TLR7 antagonist.
  • oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
  • RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
  • RNA virus is an enveloped virus.
  • RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
  • RNA virus enters the endosome of a host cell that it infects.
  • RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenzavirus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus),
  • Orthomyxoviridae e.g., Influenzavirus
  • RNA virus is an orthomyxovirus.
  • influenza virus is selected from influenza A, influenza B and influenza C.
  • RNA virus is a coronavirus.
  • betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
  • betacoronavirus is a lineage B betacoronavirus.
  • betacoronavirus is a lineage C betacoronavirus.
  • RNA virus is a picornavirus.
  • emergency warning signs of multisystem inflammatory syndrome can include: severe stomach pain, cardiac symptoms, including chest pain, palpitations and shortness of breath, bluish lips or face, mental confusion, inability to wake up or stay awake, abdominal pain with vomiting and diarrhea, skin rash and swelling of extremities, faintness and low blood pressure that is new, or any combination thereof.
  • the selective TLR7 antagonist is selected from nucleic acid antagonists (e.g ., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide) and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines).
  • nucleic acid antagonists e.g ., antisense molecules, RNAi and external guide sequences
  • proteinaceous antagonists e.g., decoy peptide
  • small molecule antagonists e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines.
  • the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7.
  • Zi and Z 2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
  • Xi is a hydrophobic amino acid (e.g., L, F or M, or modified forms thereof);
  • X2 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
  • X3 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid (e.g., A or P, or modified forms thereof); and
  • X4 is small amino acid (e.g., P, or modified forms thereof), a hydrophobic amino acid (e.g., L, or modified forms thereof) or a basic amino acid (e.g., K or R, or modified forms thereof).
  • ancillary agent is selected from is an anti-inflammatory agent, an analgesic agent, an antimicrobial agent, an agent that inhibits CRS or cytokine storm, an anti-coagulant, a platelet aggregation inhibitor, an agent that chelates iron ions released from hemoglobin by viruses, a cytochrome P-450 (CYP450) inhibitor and a NOX inhibitor.
  • the ancillary agent is selected from is an anti-inflammatory agent, an analgesic agent, an antimicrobial agent, an agent that inhibits CRS or cytokine storm, an anti-coagulant, a platelet aggregation inhibitor, an agent that chelates iron ions released from hemoglobin by viruses, a cytochrome P-450 (CYP450) inhibitor and a NOX inhibitor.
  • CYP450 cytochrome P-450
  • a selective TLR7 antagonist for use in treating an acute inflammatory condition is provided.
  • a selective TLR7 antagonist for use in treating CRS or a cytokine storm is provided.
  • a selective TLR7 antagonist for use in treating MIS-C is provided.
  • a selective TLR7 antagonist for use in treating SIRS is provided.
  • a selective TLR7 antagonist for use in treating SARS is a selective TLR7 antagonist for use in treating SARS.
  • a selective TLR7 antagonist and at least one ancillary therapeutic agent for use in treating an acute inflammatory condition are provided.
  • a selective TLR7 antagonist and an antigenic composition for use in stimulating or enhancing the production of an immune response in a subject to a pathogen.
  • mice were infected with highly pathogenic influenza A virus (IAV) at either 10 or 50PFUs/mouse at a point termed here as Day 0.
  • IAV influenza A virus
  • Mice bodyweights were monitored daily from initial point of infection with IAV.
  • mice that were treated with C98i 0.2mg/kg/day
  • mice that were treated with C98i lost significantly less of their bodyweight due to IAV ( Figure 1).
  • Mice that remained uninfected that were either treated with PBS or C98i (0.2/mg/kg/day) maintained a steady bodyweight over the 7-day period ( Figure 1).
  • mice that were treated with C98i had significantly less bronchoalveolar lavage fluid (BALF) inflammation (Figure 2); and significantly less lung viral mRNA levels (Figure 3).
  • BALF bronchoalveolar lavage fluid
  • mice were infected with highly pathogenic influenza A virus (IAV) at 50 PFUs/mouse at a point termed here as Day 0 and at Day 4 post infection were either treated with a single dose of PBS vehicle or a single dose of C98i (2mg/kg/day intranasally).
  • IAV influenza A virus
  • Mice bodyweights were monitored daily from initial point of infection with IAV.
  • Mice that were treated with PBS vehicle displayed a significant loss of weight by Day 7 post IAV infection ( Figure 6a).
  • mice that were treated with C98i 2.0mg/kg/day
  • mice infected with IAV displayed a significant elevation in airway macrophages, neutrophils, lymphocytes and eosinophils ( Figure 7).
  • mice that were treated with C98i (2mg/kg/day) had significantly less macrophage, neutrophil and lymphocyte airway recruitment and a strong trend in eosinophil reductions ( Figure 7).
  • mice infected with IAV displayed a significant elevation in peribronchial inflammation and alveolitis which was significantly inhibited by C98i treatment ( Figure 8a-c).
  • Mice groups that were not infected with IAV displayed no alteration in alveolitis and peribronchial inflammatory cells following a single dose of C98i treatment ( Figure 8a- c).
  • C98i treatment was also examined to determine whether it could suppress the pattern recognition receptors TLR7, TLR8 and TLR9 expression and expression of oxidative genes NOX2 and p47phox to IAV infection.
  • IAV infection resulted in a significant elevation in lung mRNA expression of TLR7, TLR8, TLR9, p47phox expression in mice treated with PBS vehicle ( Figure 11).
  • mice that were treated with C98i at Day 4 post IAV infection displayed a significantly reduced TLR7, TLR9, NOX2 and p47phox response in the lungs ( Figure 11).
  • Uninfected mice treated with C98i displayed no significant alteration in TLR7, TLR8, TLR9, NOX2 or p47phox expression ( Figure 11).
  • mice were infected with highly pathogenic influenza A virus (IAV) at 50 PFUs/mouse at a point termed here as Day 0, and at Day 5 post infection were either treated with a single dose of PBS vehicle (intranasally) or a single dose of C98i (2mg/kg/day intranasally).
  • IAV influenza A virus
  • mice bodyweights were monitored daily from initial point of infection with IAV. Mice that were treated with PBS vehicle displayed a significant loss of weight by Day 7 post IAV infection ( Figure 12A). In contrast, mice that were treated with C98i (2mg/kg/day) had an ⁇ 70% improvement in the bodyweight loss due to IAV ( Figure 12).
  • BALF bronchoalveolar lavage fluid
  • mice that were treated with C98i at Day 5 post infection displayed a significantly blunted cytokine response in the lungs ( Figure 15), as compared to mice treated with the PBS vehicle ( Figure 15).
  • C98i treatment modifies furin, ACE2, angiotensin ATA1 and AT2 receptor expression following IAV infection.
  • C98i treatment of mice infected with IAV had a reduced level of lung furin mRNA expression ( Figure 16), as compared to mice treated with PBS.
  • Mice treated with C98i had a significantly higher level of ACE2 and ATI receptor expression ( Figure 16).
  • mice were infected with a high inoculum (10 10 PFUs/mouse) of rhinovirus IB strain (RVIB) at a point termed here as Day 0 and at 4h post infection were either treated with a single dose of PBS vehicle or a single dose of C98i (2mg/kg/day intranasally).
  • RVIB rhinovirus IB strain
  • influenza A virus Puerto Rican-8 (PR8) strain was provided by School of Medicine, Deakin University, Victoria and The Peter Doherty Institute for Infection and Immunity (Melbourne Australia).
  • the virus was provided in PBS (Cat # D8537, Sigma-Aldrich, USA) and stored at -80 °C until used. On the day of use, virus is thawed quickly and incubated at 37 °C prior to infection. Animal ethics
  • C57BL/6 mice will be obtained from Animal Research Centre (Perth, Australia)
  • RAF RMIT Animal Facility
  • mice Eight-to-twelve-week-old C57BL6 male mice were anaesthetized and infected intranasally with the mouse-adapted IAV strain at doses: PR8 (50 PFUs/mouse) or PBS (mock infection).
  • PR8 50 PFUs/mouse
  • PBS PBS
  • mice were randomly treated with vehicle or treated with a single dose of C98i (2.0mg/kg/day via intranasal delivery) at either 4 or 5 days post IAV infection. Mice were then culled at Day 6 or 7 post infection for the comprehensive analysis including: morbidity (body weight), inflammation, lung pathology and components of the innate and adaptive immune response (see below for details).
  • mice were anaesthetized and infected intranasally with the mouse-adapted IAV strain at doses:
  • mice were randomly treated with vehicle or treated with a dose of C98i (0.2mg/kg/day via intranasal delivery) daily for 3 days beginning at Day 2 post infection. Mice were then culled at Day 6 or 7 post infection for the comprehensive analysis including: morbidity (body weight), inflammation, lung pathology and components of the innate and adaptive immune response (see below for details).
  • mice were monitored daily for signs of illness such as loss of body weight, lethargy, ruffled fur, hunched appearance, panting, loss of appetite, isolation from the group and loss of grooming. Weights were recorded daily.
  • mice were killed by an intraperitoneal (i.p.) injection of ketamine/xylazine (20 mg/kg) mixture. An incision was made from the lower jaw to the top of the rib cage, where the salivary glands were separated to expose the surface of the trachea. The layer of smooth muscle on the trachea was removed, allowing a small incision to be made near the top of the trachea. A sheathed 21-Gauge needle was inserted to the lumen and 300-400 pi of PBS was lavaged repeatedly (four times).
  • the total number of cells in the BALF were stained with ethidium bromide (cat # 15585011, Invitrogen, USA) and acridine orange (cat # A3568, Invitrogen, USA) and viable cells were counted with a hemocytometer.
  • ethidium bromide cat # 15585011, Invitrogen, USA
  • acridine orange cat # A3568, Invitrogen, USA
  • the red blood cells were lysed with lysis buffer (ACK lysis buffer; NH4CI 155 mM, NaHC03 12 mM, EDTA 0.1 mM), and the WBC stained with respective fluorescent-labelled anti-mouse antibodies for flow cytometric analysis of: Leukocytes (CD45; 30-F11); Dendritic cells (MHCII; M5/114.15.2 )(CDllc; N418 )(CDllb; Ml-70 )(CD103; 2E7); T cells (CD3 (145-2C11), CD8 (53-6.7), CD4 (RM4-5)), activation markers for T cells (CD69; H1.2F3), B cells (B220; RA3-6B2), Macrophages (F4/80; BM8) and Neutrophils (Ly6G; 1A8). All antibodies are from Biolegend, USA; BD Pharmigen, USA; BD Biosciences, USA or eBioscience,
  • the following gating strategy was used to quantify the overall numbers of macrophages (CD45+CDllb+F4/80+), neutrophils (CDllb+Ly6g + MHC Class II) and monocytes (CD45+CDllb+Ly6C+Ly6G-). Resident (MHCII+CDllc+CD8+CDllb+) and migratory (MHCII+CD11C+B220-CD103+/ MHCII+CDllc+B220-CDllb+) dendritic cell populations in the lungs and mediastinal lymph nodes were determined as a measure of cross-presentation to antigen-cognate na ' ive CD8+ T cells.
  • CD8 + T cells their activation was assessed by measuring cell surface activation markers CD69 + , as well as cytokine staining (TNF-a and IFN-g). Tetramer staining of virus-specific CD8+ T cells was performed using the peptides D b NP 366 (ASNENMETM) and D b PA 22 4 (SSLENFRAYV) that were synthesized at Biomolecular Resource Facility, Australian National University, Australia.
  • CD8+PA 224 +/ CD8+NP 66 + virus-specific CD8+ T cells
  • the left lung was dissected from mice and immersed in neutral buffered formalin (10%) for 24-48 hours. After fixation, the lung tissue was processed, embedded in paraffin wax, and longitudinal 4pm sections cut and stained with hematoxylin and eosin (H8iE). Slides were scanned by light microscopy and uploaded to Aperio microscope scanner (Leica Biosystems, Nussloch, Germany). Histology was performed by the Department of Histology (Monash University, Clayton, Australia) and analyzed blindly by two independent assessors by team at RMIT, UniSA and TCD. Five random fields from each lung section were analyzed for alveolitis, which is inflammation within the alveolar space.
  • Peribronchiolar inflammation was characterized by the infiltration of inflammatory cells into the alveolar wall around the bronchioles. The degree of inflammatory cellular infiltrate was taken by observing the density of cells throughout the entire lung section. A score of 0 is indicative of healthy lungs (i.e. no damage); 1-very mild damage; 2- mild damage; 3-moderate damage, 4-severe damage and 5-extremely severe histological changes.
  • Lungs were harvested from terminally anesthetized mice and tissue crushed into fine powder and total RNA extracted using a RNeasy mini kit (cat # 74106, QIAGEN, Germany). Synthesis of cDNA was performed using the High-Capacity cDNA RT kit (cat # 4368813, Applied Biosystems, USA) using 1.0-3.0 pg total RNA. Quantitative polymerase chain reaction was carried out using the TaqMan Fast advanced Master Mix (cat # 4444965, Applied Biosystems, USA) or PowerUp SYBR Green PCR Master Mix (cat # A25777, Applied Biosystems, USA) and analyzed on the Quant Studio 7 Flex Real-Time PCR system (Applied Biosystems).
  • PCR primers for the following genes - pro-inflammatory cytokines IFN-b, IL-Ib, TNF-a, IL-6, IL-17 and IFN-y
  • chemokines CCL2, CCL5, CXCL2, CXCL10
  • TLRs TLR2, TLR4, TLR7, TLR9
  • Oxidative and antioxidant genes NOX2, NOX1, NOX4, CYBA, NCF1, NCF2, SOD1, SOD2, SOD3, GPX1, CAT, PRDX5, NOS1, NOS2, NOS3
  • renin angiotensin system ACE1, ACE2, AGTRla, AGTR2
  • glycolytic genes HEX1, HEX2, SLC2A1, SLC2A4
  • endosomal genes EAA1, APPL1, RAB5A, RAB7, SORT1, SDC1 and serine protease (FURIN) was included in the Assay-on-Demand Gene Expression Assay Mix (Applied Biosystems, USA).
  • PCR program run settings were: 50 ° C for 2 min, followed by 95 ° C for 1 hr, then 95 ° C for 15 s + 60 ° C for 60 s + plate read (40 cycles).
  • program settings were: 50 ° C for 2 minutes, 95 ° C for 2 minutes, 95 ° C for 1 second, 60 ° C for 20 seconds + plate read (40 cycles).
  • Quantitative values were obtained from the threshold cycle (Ct) number.
  • Target gene expression was normalized against glyceraldehyde 3-phosphate dehydrogenase (GAPDH; cat # 4352339E, Applied Biosystems, USA) mRNA expression for each sample and data were expressed relative to the naive control group.
  • GPDH glyceraldehyde 3-phosphate dehydrogenase
  • cytokines IFN-b, IL-Ib, TNF-a, IL-6, and IFN-g
  • chemokines CCL2, CCL5, CXCL2, CXCL10
  • the cytokine titers in samples were determined by plotting the optical densities using a 4-parameter fit for the standard curve. If any other of the genes measured by QPCR (as performed according to the method detailed above) were modified by IAV infection and C98i treatment, the protein analysis will occur via ELISA.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Virology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Pulmonology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Toxicology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Disclosed are selective TLR7 antagonists for use in treating or inhibiting the development of acute inflammatory conditions including, for example, acute inflammatory conditions associated with presence of pathogenic infections, multisystem inflammatory syndrome, systemic inflammatory response syndrome, acute respiratory distress syndrome, severe acute respiratory syndrome and/or symptoms arising from acute inflammatory conditions.

Description

TITLE
"AGENTS AND METHODS FOR THERAPY AND PROPHYLAXIS"
RELATED APPLICATIONS
[0001] This application claims priority to Australian Provisional Application No. 2021900345 entitled "Agents and methods for therapy and prophylaxis" filed 12 February 2021 and Australian Provisional Application No. 2021903239 entitled "Agents and methods for therapy and prophylaxis" filed 8 October 2021 the contents of which are incorporated herein by reference in their entirety.
FIELD
[0002] This disclosure relates generally to the treatment and prophylaxis of acute inflammatory conditions. More particularly, the present disclosure relates to the use of selective TLR7 antagonists for treating or inhibiting the development of acute inflammatory conditions including, for example, acute inflammatory conditions associated with presence of pathogenic infections, multisystem inflammatory syndrome, systemic inflammatory response syndrome, acute respiratory distress syndrome, severe acute respiratory syndrome and/or symptoms arising from acute inflammatory conditions.
BACKGROUND
[0003] Inflammation is an adaptive process to harmful stimuli such as pathogens, damaged cells, or irritants. It is a protective attempt by an organism, which typically involves immune cells, blood vessels and molecular mediators, to remove the noxious stimuli and to initiate the healing process for damaged tissue. However, inflammation which runs unchecked can also lead to a host of diseases, including cytokine release syndrome (CRS)/cytokine storm, systemic inflammation and multiple organ failure.
[0004] Viral infections that can give rise to such acute inflammatory diseases underlie some of the most devastating diseases worldwide, notable examples of which include pandemic and epidemic seasonal influenza and the current COVID-19 pandemic. Annual epidemics result in ~1 billion cases of influenza, with about 3 to 5 million cases of severe illness, and up to 650,000 deaths. The COVID-19 pandemic has already caused >401 million infections and has killed over 5.8 million people (Feb 2022). The economic burden of COVID-19 is estimated in the trillions of dollars. These two viruses alone have caused significant burden to our society as well as hospitalizing and killing large numbers of patients, but there are also numerous other RNA viruses that cause very significant pathogenesis in humans. There is a constant emergence of new viral strains that are not adequately neutralized by vaccines and the current treatment strategies for treating influenza are limited and not very effective once the virus is established. With the ever-present threat of epidemics and pandemics, for which global health systems are chronically underprepared, there is a desperate need for novel therapeutic approaches.
[0005] It is imperative, therefore, that broad-based antivirals are developed, which can desirably: 1) treat new infection outbreaks irrespective of the infecting strain of virus; 2) have therapeutic utility after a viral outbreak has occurred and before vaccine strategies become available/or where vaccines are not effective; 3) have utility in patients that have already been infected by the pathogenic virus; and/or 4) can treat direct and indirect symptoms and conditions associated with viral infection.
[0006] The pathway of viral pathogenesis typically involves: 1) attachment of the virus to the cell surface and entry into endosomes that are a common conduit for viral transport, which facilitate viral replication and induction of a pathogenic cascade; 2) at the key regulatory point in this pathway is the toll-like receptor 7 (TLR7) protein, which is a gatekeeper that RNA viruses use to trigger a cytokine cascade and production of highly reactive and oxygen species (ROS), which ultimately drive inflammation, viral replication, cellular damage and limit effective host immunity.
[0007] TLR7 is a pattern recognition receptor that recognizes viral RNA following endocytosis of the virus and initiates a powerful immune response characterized by Type I IFN production and pro-inflammatory cytokine production; including tumor necrosis factor-a (TNF-a) and interlukin-6 (IL-6) production. TLR7 recruits NOX2 to drive ROS production in endosomes and this establishes a microenvironment that potentiates aberrant inflammation and immune signaling. By altering the normal immune response, RNA viruses cause very significant pathology, which is highly ROS and inflammation-dependent. Critically, TLR7 is activated by an enzyme called furin, which is enriched in the Golgi apparatus, where it functions to cleave proteins into their active form(s). In addition, furin is also utilized by a number of highly pathogenic viruses such as influenza, dengue, filoviruses including Ebola and Marburg virus, as well as SARS-CoV-2. The cleavage and activation of these viruses by furin or furin-like proteases underpins a key component of the viral infection and pathogenesis and this involves TLR7.
SUMMARY
[0008] In work leading up to the present disclosure, it was found that TLR7 protein is activated by a disulfide bond forming between cysteine residues at positions 98 (Cys98) and 475 (Cys475) of TLR7 and that this leads to toxic ROS production via NADPH oxidase (N0X2). Based on this finding, the present inventors developed a selective TLR7 antagonist peptide that can correct TLR7 signaling and shut down excessive ROS production, to thereby suppress viral activity, increase antiviral antibody production and inhibit the process of RNA virus pathogenesis (see, International Publication WO 2019/000045).
[0009] The present inventors have now strikingly found that selective TLR7 antagonism is able to inhibit morbidity, viral titer, furin synthesis, pattern recognition receptors, oxidative stressors, cytokine storm and clinical signs of inflammation in subjects with active RNA virus infections, including infections with highly pathogenic H1N1 influenza A virus (IAV), in which subjects had been infected for five days. Surprisingly, they have also found that treatment of subjects with a single dose of selective TLR7 antagonist was sufficient to ameliorate the symptoms of acute inflammation in infected subjects. Based on the known release of pro-inflammatory cytokines into lung tissue at an early stage of H1N1 IAV infection (e.g., day 1-3 post infection) and the present finding that TLR7 antagonism can significantly reduce or stymie this release after it has developed, it is proposed that selective TLR7 antagonists can be used to treat acute inflammatory conditions generally, particularly those caused or exacerbated at least in part by activation of TLR7, including ones associated with the presence of cytokine release syndrome or a cytokine storm. Notably, the present findings also indicate that the treatment can be given and is effective even after the viral infection and or inflammatory condition has already established in the host. [0010] Accordingly, disclosed herein in one aspect are methods for treating an acute inflammatory condition in a subject. These methods generally comprise, consist or consist essentially of administering to the subject an effective amount of a selective TLR7 antagonist. Suitably, the acute inflammatory condition is at least in part caused or exacerbated by, or is otherwise associated with, activation of TLR7 and optionally one or both of TLR8 and TLR9, and/or an increased level of one or more oxidative stressors (e.g., NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1; also known as p47phox) and ROS) and/or an increased level of furin. In specific embodiments, the acute inflammatory condition is associated with presence of a pathogenic infection (e.g., a pathogenic infection in which the pathogen is an RNA virus), which is typically an active infection. The RNA virus may enter a host cell that it infects by receptor- mediated endocytosis or micropinocytosis. In specific embodiments, the RNA virus enters the endosome of a host cell that it infects. In representative examples, the RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenza virus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus), Togaviridae (e.g., Chikungunya virus, Ross River virus, Semliki Forest virus), Flaviviridae (e.g., Zika virus, Dengue virus, Japanese encephalitis virus, West Nile virus, Yellow fever virus, Hepacivirus C) and Hantaviridae (e.g., Hantavirus). In certain embodiments, the RNA virus is an orthomyxovirus (e.g., an influenza virus such as influenza A, influenza B or influenza C) or a coronavirus (e.g., a coronavirus, including betacoronaviruses, capable of causing severe acute respiratory syndrome (SARS) such as MERS-CoV, SARS-CoV and SARS-CoV-2). Suitably, the acute inflammatory condition is associated with presence of cytokine release syndrome (CRS) or a cytokine storm, which in representative examples comprises an elevation of at least 50% compared to basal state of one or more (e.g., 1, 2, 3, 4, 5, 6, 7 or 8) cytokines selected from IFN- Y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2. In some embodiments, the acute inflammatory condition is associated with a condition selected from multisystem inflammatory syndrome in children (MIS-C), systemic inflammatory response syndrome (SIRS), acute respiratory distress syndrome (ARDS), and severe acute respiratory syndrome (SARS). The treatment of conditions associated directly or indirectly with viral infection is also contemplated herein including the treatment of cardiovascular conditions as well as the reduction, loss, modification or distortion of olfactory sensation. The selective TLR7 antagonist may be selected from nucleic acid antagonists (e.g., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide) and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines). In specific embodiments, the selective TLR7 antagonist comprises a peptide corresponding to the amino acid sequence of TLR7, which antagonizes disulfide bond formation between C98 and C475 of human TLR7 or their corresponding positions in the TLR7s of other species. In some embodiments, "selective antagonism" is not a total or complete abrogation of TLR7 activity but rather selective inhibition to return the immune system to, or towards, homeostasis.
[0011] In related aspects of the present disclosure, methods are provided for treating
CRS or a cytokine storm, SARS, ARDS, SIRS or MIS-C in a subject. These methods generally comprise, consist or consist essentially of administering to the subject an effective amount of a selective TLR7 antagonist.
[0012] In other related aspects, the present disclosure provides selective TLR7 antagonists, suitably in the form of medicaments, for treating a condition selected from an acute inflammatory condition, CRS or a cytokine storm, SARS, ARDS, SIRS or MIS-C.
[0013] In any of the aspects disclosed herein, the methods suitably comprise administering the subject with a single dose of the selective TLR7 antagonist. In specific embodiments, the single dose is administered only once during the course of treatment.
[0014] In any of the aspects disclosed herein relating to pathogenic infections, the selective TLR7 antagonist may be concurrently administered with an antigenic composition that stimulates or enhances the production of an immune response in the subject to a pathogen.
BRIEF DESCRIPTION OF THE DRAWINGS
[0015] Some figures contain color representations or entities. Color photographs are available from the Patentee upon request or from an appropriate Patent Office. A fee may be imposed if obtained from a Patent Office.
[0016] Figure 1 is a graphic representation showing mice bodyweight change over the 7-day infection period with PR8 virus (10 or 50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98L 2-way ANOVA- Tukey Kramer * P<0.05; *** P O.OOl; **** P O.OOOl. PBS n=12, C98i n=12, PR8 n=33, PR8 + C98i n=35.
[0017] Figure 2 is a graphic representation of the bronchoalveolar lavage fluid (BALF) inflammation in mice infected with PR8 virus (10 or 50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98L 1-way ANOVA- Tukey Kramer ** P O.Ol**** PcO.OOOl. PBS n=12, C98i n=12, PR8 n=33, PR8 + C98i n=35.
[0018] Figure 3 is a graphic representation of the viral mRNA levels in lungs from mice infected with PR8 virus (10 or 50PFU/mouse). Mice were administered either PBS vehicle or C98i. Unpaired t-test * P<0.05 PR8 n=22, PR8 + C98i n=21.
[0019] Figure 4 is H8i E imaging (4a and 4b) and graphic representations (4c) of lung inflammation in mice infected with PR8 virus (10 or 50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98L 4C are the results of the blinded analysis performed by 2 independent histologists.
[0020] Figure 5 is a graphic representation of organ weights in mice infected with PR8 virus (10 or 50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98i. 1-way ANOVA- Tukey Kramer *P<0.05, **** PcO.OOOl, ns- not significant. PBS n=12, C98i n=12, PR8 n=33, PR8 + C98i n=35.
[0021] Figure 6 is A) a graphic representation showing mice bodyweight change over the 7-day infection period with PR8 virus (50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98i (2mg/kg) at Day 4 post infection. 2-way ANOVA- Tukey Kramer * Pc0.05; **** PcO.OOOl. PBS n=6, C98i n=16, PR8 n=14, PR8 + C98i n=14. B) Graphic representation of the bronchoalveolar lavage fluid (BALF) inflammation in mice infected with PR8 virus (50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98i (2mg/kg). 1-way ANOVA- Tukey Kramer *** PcO.OOl, **** PcO.OOOl. PBS n=6, C98i n=16, PR8 n=14, PR8 + C98i n=14. C). Graphic representation of the viral mRNA levels in lungs from mice infected with PR8 virus (50PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg). Unpaired t-test * P<0.05. ** P O.Ol PBS n=6, C98i n=16, PR8 n=14, PR8 + C98i n=14. D) Graphic representation of lung weights in mice infected with PR8 virus (50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98i (2mg/kg). 1-way ANOVA- Tukey Kramer **P<0.01, **** P O.OOOl, PBS n=6, C98i n = 16, PR8 n = 14, PR8 + C98i n = 14.
[0022] Figure 7 is a graphic representation of the bronchoalveolar lavage fluid (BALF) macrophage, neutrophil, lymphocyte and eosinophil recruitment in mice infected with PR8 virus (50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98i (2mg/kg) at Day 4 post infection. 1-way ANOVA- Tukey Kramer * P<0.05; ** PcO.Ol, *** PcO.OOl. PBS n=6, C98i n=16, PR8 n=14, PR8 + C98i n = 14.
[0023] Figure 8 is H8iE imaging (a and b) and graphic representations (c) of lung inflammation in mice infected with PR8 virus (50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98L 4C are the results of the blinded analysis performed by 2 independent histologists. Unpaired t-test ** PcO.Ol, *** PcO.OOl. PBS n=6, C98i n = 16, PR8 n=14, PR8 + C98i n=14.
[0024] Figure 9 is a graphic representation of the lung cytokine mRNA expression in mice infected with PR8 virus (50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98i (2mg/kg) at Day 4 post infection. All genes were normalized to the housekeeping gene RPS18 and then normalized as a fold-increase above the PBS control group. Unpaired t-test * Pc0.05,** PcO.Ol, PBS n=6, C98i n=16, PR8 n=14, PR8 + C98i n=14.
[0025] Figure 10 is a graphic representation of the lung mRNA expression of furin,
ACE2, ATI and AT2 receptors in mice infected with PR8 virus (50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98i (2mg/kg) at Day 4 post infection. All genes were normalized to the housekeeping gene RPS18 and then normalized as a fold-increase above the PBS control group. Unpaired t-test * Pc0.05,** PcO.Ol, ns=not significant. PBS n=6, C98i n=16, PR8 n=14, PR8 + C98i n=14.
[0026] Figure 11 is a graphic representation of the lung mRNA expression of TLR7,
TLR8, TLR9, N0X2 AND P47PHOX in mice infected with PR8 virus (50PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98i (2mg/kg) at Day 4 post infection. All genes were normalized to the housekeeping gene RPS18 and then normalized as a fold-increase above the PBS control group. Unpaired t-test * Pc0.05,** PcO.Ol, ns=not significant. PBS n=6, C98i n=16, PR8 n=14, PR8 + C98i n=14.
[0027] Figure 12 is A) a graphic representation showing mice bodyweight change over the 7-day infection period with PR8 virus (50PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg) at Day 5 post infection. 2-way ANOVA- Tukey Kramer * Pc0.05; **** PcO.OOOl. PR8 + PBS n=6, PR8 + C98i n=6. B) a graphic representation of the bronchoalveolar lavage fluid (BALF) inflammation in mice infected with PR8 virus (50PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg). Unpaired t-test * Pc0.05,** PcO.Ol. PR8 + PBS n=6, PR8 + C98i n=6. C). a graphic representation of the viral mRNA levels in lungs from mice infected with PR8 virus (50PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg). Unpaired t-test * Pc0.05,** PcO.Ol. PR8 + PBS n=6, PR8 + C98i n=6. D) a graphic representation of lung weight in mice infected with PR8 virus (50PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg). Unpaired t-test * P<0.05, ** P O.Ol. PR8 +
PBS n=6, PR8 + C98i n=6.
[0028] Figure 13 is a graphic representation of the bronchoalveolar lavage fluid (BALF) macrophage, neutrophil, lymphocyte and eosinophil recruitment in mice infected with PR8 virus (50PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg) at Day 5 post infection. Unpaired t-test * P<0.05, ** PcO.Ol. PR8 + PBS n=6, PR8 + C98i n=6.
[0029] Figure 14 is H8iE imaging of lung inflammation in mice infected with PR8 virus (50PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg) at Day 5 post infection.
[0030] Figure 15 is a graphic representation of the lung cytokine mRNA expression in mice infected with PR8 virus (50PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg) at Day 5 post infection. All genes were normalized to the housekeeping gene RPS18 and then normalized as a fold-increase above the PBS control group. Unpaired t-test * P<0.05,** PcO.Ol. *** PcO.OOl. ns=not significant. PR8 + PBS n=6, PR8 + C98i n=6.
[0031] Figure 16 is a graphic representation of the lung mRNA expression of furin,
ACE2, ATI and AT2 receptors in mice infected with PR8 virus (50PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg) at Day 5 post infection. All genes were normalized to the housekeeping gene RPS18 and then normalized as a fold-increase above the PBS control group. Unpaired t-test * Pc0.05,** PcO.Ol, ns=not significant. PR8 + PBS n=6, PR8 +
C98i n=6.
[0032] Figure 17 is a graphic representation of the lung mRNA expression of TLR7, TLR8, TLR9, N0X2 and P47PHOX in mice infected with PR8 virus (50PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg) at Day 5 post infection. All genes were normalized to the housekeeping gene RPS18 and then normalized as a fold-increase above the PBS control group. Unpaired t-test * Pc0.05,** PcO.Ol, ns=not significant. PR8 + PBS n=6, PR8 +
C98i n=6.
[0033] Figure 18 is a graphic representation showing mice bodyweight over the infection period with RV1B virus (1010 PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98i. PBS n=12, C98i n=12, RV1B n=16, RV1B + C98i n=16.
[0034] Figure 19 is a graphic representation of the bronchoalveolar lavage fluid (BALF) inflammation in mice infected with RV1B virus (1010 PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98L 1-way ANOVA- Tukey Kramer * PcO.05. PBS n=12, C98i n=12, RV1B n = 16, RV1B + C98i n=16.
[0035] Figure 20 is a graphic representation of the viral mRNA levels in lungs from mice infected with RV1B virus (1010 PFU/mouse). Mice were administered either PBS vehicle or C98i. Unpaired t-test * Pc0.05 RV1B n=16, RV1B + C98i n=16.
[0036] Figure 21 is a graphic representation of lung weights in mice infected with RV1B virus (1010 PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98i. 1-way ANOVA- Tukey Kramer *P<0.05, ** PcO.Ol. PBS n=12, C98i n=12, RV1B n=16, RV1B + C98i n=16. [0037] Figure 22 is a graphic representation of the bronchoalveolar lavage fluid (BALF) macrophage, neutrophil, lymphocyte and eosinophil recruitment in mice infected with RV1B virus (1010 PFU/mouse). Some mice were not infected with virus. Mice were administered either PBS vehicle or C98i (2mg/kg). 1-way ANOVA- Tukey Kramer * P<0.05; ** P O.Ol. PBS n=12, C98i n = 12, RV1B n=16, RV1B + C98i n = 16.
[0038] Figure 23 is a graphic representation of the lung cytokine mRNA expression in mice infected with RV1B virus (1010 PFU/mouse). Mice were administered either PBS vehicle or C98i (2mg/kg). All genes were normalized to the housekeeping gene RPS18 and then normalized as a fold-increase above the PBS control group. 1-way ANOVA- Tukey Kramer * P<0.05; ** PcO.Ol.
*** PcO.OOl. PBS n = 12, C98i n = 12, RV1B n=16, RV1B + C98i n=16.
DETAILED DESCRIPTION OF THE INVENTION
1. Definitions
[0039] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by those of ordinary skill in the art to which the present disclosure belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, preferred methods and materials are described. For the purposes of the present disclosure, the following terms are defined below.
[0040] The articles "a" and "an" are used herein to refer to one or to more than one (/.e., to at least one) of the grammatical object of the article. By way of example, reference to "a virus" includes a single virus, as well as two or more viruses; reference to "an agent" includes a single agent, as well as two or more agents; reference to "the disclosure" includes single and multiple aspects taught by the disclosure; and so forth.
[0041] Further, the term "about", as used herein when referring to a measurable value such as an amount, dose, time, temperature, activity, level, number, frequency, percentage, dimension, size, amount, weight, position, length and the like, is meant to encompass variations of ± 15%, ± 10%, ± 5%, ± 1%, ± 0.5%, or even ± 0.1% of the specified amount, dose, time, temperature, activity, level, number, frequency, percentage, dimension, size, amount, weight, position, length and the like.
[0042] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the disclosure. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges is also encompassed within the disclosure, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either both of those included limits are also included in the disclosure.
[0043] The term "active infection" is used herein in its broadest sense and includes the invasion, establishment and/or multiplication of a virus in a host, which is typically associated with one or more pathological symptoms that may or may not be clinically apparent. Active infections include localized, subclinical or temporary infections. A local infection may persist and spread by extension to become an acute, subacute or chronic clinical infection or disease state. A local infection may also become systemic when a virus gains access to the lymphatic or vascular system. Typically, "active infection" refers to an infectious state in which a host's immune system is activated against an infectious agent.
[0044] The term "acute inflammatory condition" as used herein refers to a condition in which acute inflammation is present and represents a rapid, short-lived (minutes to days), relatively uniform response to acute injury characterized by accumulations of fluid, plasma proteins, and neutrophilic leukocytes. In acute inflammation, removal of the stimulus halts the recruitment of monocytes (which become macrophages under appropriate activation) into the inflamed tissue, and existing macrophages exit the tissue via lymphatics. Examples of injurious agents that cause acute inflammation include, but are not limited to, pathogens (e.g., bacteria, viruses, parasites), foreign bodies from exogenous (e.g., asbestos) or endogenous (e.g., urate crystals, immune complexes), sources, and physical (e.g., burns) or chemical (e.g., caustics) agents. Generally, the physiologic changes accompanying acute inflammation encompass four main features: (1) vasodilation, which results in a net increase in blood flow, is one of the earliest s physical responses to acute tissue injury; (2) in response to inflammatory stimuli, endothelial cells lining the venules contract, widening the intracellular junctions to produce gaps, leading to increased vascular permeability, which permits leakage of plasma proteins and blood cells out of blood vessels; (3) inflammation often is characterized by a strong infiltration of leukocytes at the site of inflammation, particularly neutrophils (polymorphonuclear cells). These cells promote tissue damage by releasing toxic substances at the vascular wall or in uninjured tissue; and (4) fever, produced by pyrogens released from leukocytes in response to specific stimuli. Other conditions include a reduction in olfactory sensation and adverse effects on the cardiovascular system.
[0045] As used herein, the term "acute respiratory distress syndrome" refers to a life- threatening lung condition that prevents enough oxygen from getting to the lungs and into the blood. ARDS is also referred to as noncardiogenic pulmonary edema, increased-permeability pulmonary edema, stiff lung, shock lung, or acute lung injury. ARDS can be caused by any major injury to the lung. Some common causes include, without limitation: breathing vomit into the lungs (aspiration), inhaling chemicals, lung transplant, pneumonia, septic shock (infection throughout the body), and trauma.
[0046] The terms "administration concurrently" or "administering concurrently" or "co administering" and the like refer to the administration of a single composition containing two or more actives, or the administration of each active as separate compositions and/or delivered by separate routes either contemporaneously or simultaneously or sequentially within a short enough period of time that the effective result is equivalent to that obtained when all such actives are administered as a single composition. By "simultaneously" is meant that the active agents are administered at substantially the same time, and desirably together in the same formulation. By "contemporaneously" it is meant that the active agents are administered closely in time, e.g., one agent is administered within from about one minute to within about one day before or after another. Any contemporaneous time is useful. However, it will often be the case that when not administered simultaneously, the agents will be administered within about one minute to within about eight hours and suitably within less than about one to about four hours. When administered contemporaneously, the agents are suitably administered at the same site on the subject. The term "same site" includes the exact location but can be within about 0.5 to about 15 centimeters, preferably from within about 0.5 to about 5 centimeters. The term "separately" as used herein means that the agents are administered at an interval, for example at an interval of about a day to several weeks or months. The active agents may be administered in either order. The term "sequentially" as used herein means that the agents are administered in sequence, for example at an interval or intervals of minutes, hours, days or weeks. If appropriate the active agents may be administered in a regular repeating cycle.
[0047] As used herein, "and/or" refers to and encompasses any and all possible combinations of one or more of the associated listed items, as well as the lack of combinations when interpreted in the alternative (or).
[0048] The term "antagonist" is used interchangeably herein with the term "inhibitor" to refer to a substance that prevents, blocks, inhibits, neutralizes, reduces, restores, or stimulates return to or towards restoration of, a biological activity or effect of another molecule, such as an enzyme or receptor. The term "selective antagonist" refers to a compound with high selectivity for its target {e.g. for a TLR such as TLR7). Selectivity of a particular antagonist is defined as a ratio of the IC50 values of the particular antagonist for the target of interest versus another target. For example, an antagonist that is selective for target A (e.g., TLR7) will have an IC50 value for target A lower than that for target B (e.g., TLR8 and/or TLR9), representative examples of which include wherein the IC50 value for target A is at least 10 times lower, at least 100 times lower, at least 1000 times lower or at least 10,000 times lower than the IC50 value of the same antagonist for target B. The antagonist may be a direct or indirect antagonist. The term "direct TLR7 antagonist", as used herein, refers to an antagonist that acts via contact (e.g., binding) with TLR7, i.e., the TLR7 antagonist binds to TLR7 and inhibits its activity and/or activation. In contrast, an "indirect TLR7 antagonist" may act without contacting TLR7 protein. For example, antisense RNA can be used to decrease expression of the TLR7 gene, or a small molecule can antagonize the effects of TLR7 via interactions with downstream signaling pathway members; these do not interact directly with the TLR7 protein. Thus, an indirect antagonist, in contrast to a direct antagonist, acts upstream or downstream from the TLR7 protein. The term "antagonist", including "selective antagonist", includes regulating or modulating the activity of TLR7 to maintain, return to, or stimulate a return towards, immune homeostasis. Hence, total or complete abrogation of TLR7 activity is not necessarily a requirement for TLR7 antagonism.
[0049] As used herein, the term "antigen" and its grammatically equivalent expressions (e.g., "antigenic") refer to a compound, composition, or substance that may be specifically bound by the products of specific humoral or cellular immunity, such as an antibody molecule or T-cell receptor. Antigens can be any type of molecule including, for example, haptens, simple intermediary metabolites, sugars (e.g., oligosaccharides), lipids, and hormones as well as macromolecules such as complex carbohydrates (e.g., polysaccharides), phospholipids, and proteins. Common categories of antigens include, but are not limited to, viral antigens, bacterial antigens, fungal antigens, protozoa and other parasitic antigens, tumor antigens, antigens involved in autoimmune disease, allergy and graft rejection, toxins, and other miscellaneous antigens.
[0050] Throughout this specification, unless the context requires otherwise, the words "comprise," "comprises" and "comprising" will be understood to imply the inclusion of a stated step or element or group of steps or elements but not the exclusion of any other step or element or group of steps or elements. Thus, use of the term "comprising" and the like indicates that the listed elements are required or mandatory, but that other elements are optional and may or may not be present. By "consisting of" is meant including, and limited to, whatever follows the phrase "consisting of". Thus, the phrase "consisting of" indicates that the listed elements are required or mandatory, and that no other elements may be present. By "consisting essentially of" is meant including any elements listed after the phrase and limited to other elements that do not interfere with or contribute to the activity or action specified in the disclosure for the listed elements. Thus, the phrase "consisting essentially of" indicates that the listed elements are required or mandatory, but that other elements are optional and may or may not be present depending upon whether or not they affect the activity or action of the listed elements.
[0051] A "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art, which can be generally sub-classified as follows:
TABLE A
AMINO ACID SUB-CLASSIFICATION
Figure imgf000011_0001
[0052] Conservative amino acid substitution also includes groupings based on side chains. For example, a group of amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having aliphatic-hydroxyl side chains is serine and threonine; a group of amino acids having amide-containing side chains is asparagine and glutamine; a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan; a group of amino acids having basic side chains is lysine, arginine, and histidine; and a group of amino acids having sulfur-containing side chains is cysteine and methionine. For example, it is reasonable to expect that replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, a threonine with a serine, or a similar replacement of an amino acid with a structurally related amino acid will not have a major effect on the properties of the resulting variant polypeptide. Whether an amino acid change results in a functional polypeptide can readily be determined by assaying its activity. Conservative substitutions are shown in TABLE B under the heading of exemplary and preferred substitutions. Amino acid substitutions falling within the scope of the present disclosure, are, in general, accomplished by selecting substitutions that do not differ significantly in their effect on maintaining (a) the structure of the peptide backbone in the area of the substitution, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. After the substitutions are introduced, the variants are screened for biological activity.
TABLE B
EXEMPLARY AND PREFERRED AMINO ACID SUBSTITUTIONS
Figure imgf000012_0001
[0053] As used herein, the term "contiguous" in the context of a sequence of subunits such as a nucleic acid or amino acid sequence means that the sequence is a single sequence, uninterrupted by any intervening sequence or sequences. [0054] The terms "corresponds to" and "corresponding to" and their grammatical equivalents as applied to nucleic acid sequences refer to a nucleic acid sequence that displays substantial sequence identity to a reference nucleic acid sequence {e.g., at least about 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 97, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% or even up to 100% sequence identity to all or a portion of the reference nucleic acid sequence). In the context of amino acid sequence, the terms "corresponds to" and "corresponding to" and their grammatical equivalents refer to an amino acid sequence that displays substantial sequence similarity or identity to a reference amino acid sequence. In general the amino acid sequence will display at least about 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 97, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% or even up to 100% sequence similarity or identity to at least a portion of the reference amino acid sequence. [0055] The term "cytokine release syndrome" or "CRS" refers to a form of systemic inflammatory response syndrome (SIRS) that can be triggered by a variety of factors such as infections and certain drugs. It refers to cytokine storm syndromes (CSS) and occurs when large numbers of white blood cells are activated and release inflammatory cytokines, which in turn activate yet more white blood cells. CRS is also an adverse effect of some monoclonal antibody medications, as well as adoptive T-cell therapies. When occurring as a result of a medication, it is also known as an infusion reaction. The term cytokine storm is often used interchangeably with CRS but, despite the fact that they have similar clinical phenotype, their characteristics are different. When occurring as a result of a therapy, CRS symptoms may be delayed until days or weeks after treatment. Immediate-onset CRS is a cytokine storm, although severe cases of CRS have also been called cytokine storms.
[0056] As used herein, the term "cytokine storm" refers to an excessively activated cytokine cascade or hypercytokinemia, i.e., an excessive or uncontrolled release of pro- inflammatory cytokines, which can be associated with a wide variety of infectious and noninfectious diseases or disorders. Cytokine storm syndromes are associated with a group of disorders (such as, but not limited to, influenza, asthma, hantavirus pulmonary syndrome. SIRS, macrophage activation syndrome (MAS), SARS, COVID-19 and disseminated vascular coagulopathy (DIC)), representing a variety of inflammatory causes; the present disclosure does not depend on any particular one of these underlying causes, but is directed to preventing or delaying the onset of cytokine storm, or treating cytokine storm, arising from any underlying inflammatory cause. Typically, the primary symptoms of a cytokine storm are high fever, swelling and redness, extreme fatigue and nausea. In some cases, the immune reaction can result in bleeding, clotting, internal organ injury, or shock, and may be fatal.
[0057] As used herein, "delaying progression of a disease" or "decreasing the rate of progression of a disease" means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated.
[0058] By "effective amount", in the context of treating or preventing a condition is meant the administration of an amount of an agent or composition to an individual in need of such treatment or prophylaxis, either in a single dose or as part of a series, that is effective for the prevention of incurring a symptom, holding in check such symptoms, and/or treating existing symptoms, of that condition. The effective amount will vary depending upon the health and physical condition of the individual to be treated, the taxonomic group of individual to be treated, the formulation of the composition, the assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials. Non-limiting symptoms of coronavirus infections, for example, include acute febrile illness, malaise, fatigue, headache, flushing, diarrhea, nausea, vomiting, coughing including dry coughing, sore throat, runny nose, nasal congestion, and, in severe disease, symptoms of systemic inflammatory response syndrome including production of pro-inflammatory mediators, vascular leakage and organ failure. An "effective amount" may also be measured by amelioration of indirect or direct symptoms such as reduction, loss, modification or distortion of olfactory sensation. [0059] The term "expression" with respect to a gene sequence refers to transcription of the gene to produce a RNA transcript {e.g., mRNA, antisense RNA, siRNA, shRNA, miRNA, etc.) and, as appropriate, translation of a resulting mRNA transcript to a protein. Thus, as will be clear from the context, expression of a coding sequence results from transcription and translation of the coding sequence. Conversely, expression of a non-coding sequence results from the transcription of the non-coding sequence.
[0060] "Immune homeostasis", "immune system homeostasis" and the like refer to physiological equilibrium between "attack and defense" mechanism against pathogens, foreign or diseased tissues, and "tolerance" for self. This equilibrium is maintained by checks and balances involving many immune cell types and soluble mediators. Dysregulation of immune homeostasis compromises the body's defense and self/non-self-recognition leading to disease or autoimmunity.
[0061] An "immune response" as used herein, refers to a response by the immune system of a subject. For example, an immune response may be to an antigen/immunogen that the subject's immune system recognizes as foreign (e.g., non-self-antigens) or self (e.g., self-antigens recognized as foreign). Immune responses may be humoral, involving production of immunoglobulins or antibodies, or cellular, involving various types of B and T lymphocytes, dendritic cells, macrophages, antigen presenting cells and the like, or both. Immune responses may also involve the production or elaboration of various effector molecules such as cytokines. The term "immune response" encompasses immunogenic responses that cause, activate, elicit, stimulate, or induce an immune response against a particular antigen (e.g., a pathogenic antigen) or organism (e.g., a pathogenic microorganism) in a subject, as well as immunosuppressive or tolerogenic immune responses that inhibit, suppress, diminish or eliminate an immune response, or render the immune system unresponsive, or delay the occurrence or onset of an immune response, to an allergen, or to a self-antigen or a cell, tissue or organ that expresses such an antigen. In specific embodiments, an immune response is one that includes an immunosuppressive or tolerogenic immune response that inhibits, suppresses, diminishes or eliminates a humoral and/or cellular immune response, including the production of autoantibodies, to a self-antigen or a cell, tissue or organ that expresses such an antigen.
[0062] As used herein, the term "immunity" refers to protection from disease (e.g., preventing or attenuating (e.g., suppression) of a sign, symptom or condition of the disease) upon exposure to a microorganism (e.g., pathogen) capable of causing the disease. Immunity can be innate (e.g., non-adaptive (e.g., non-acquired) immune responses that exist in the absence of a previous exposure to an antigen) and/or acquired/adaptive (e.g., immune responses that are mediated by B and T cells following a previous exposure to antigen (e.g., that exhibit increased specificity and reactivity to the antigen)).
[0063] As used herein, the term "immunogen" refers to a molecule which stimulates a response from the adaptive immune system, which may include responses drawn from the group comprising an antibody response, a cytotoxic T cell response, a T helper response, and a T cell memory response. An immunogen may stimulate an upregulation of the immune response with a resultant inflammatory response, or may result in down regulation or immunosuppression.
[0064] As used herein, the term "multisystem inflammatory syndrome in children" or "MIS-C" is a rare life-threatening illness where different body parts can become inflamed, including the heart, lungs, kidneys, brain, skin, eyes, or gastrointestinal organs. Children with MIS-C may have a fever and various symptoms, including abdominal (gut) pain, vomiting, diarrhea, neck pain, rash, bloodshot eyes, or feeling extra tired.
[0065] The term "oxidative stressor", as used herein, refers to a molecule that generates oxidative stress in cells including ROS and ROS-generating enzymes such as NOX2 and NCF1.
[0066] The terms "patient", "subject", "host" or "individual" used interchangeably herein, refer to any subject, particularly a vertebrate subject, and even more particularly a mammalian subject, for whom therapy or prophylaxis is desired. Suitable vertebrate animals that fall within the scope of the disclosure include, but are not restricted to, any member of the subphylum Chordata including primates (e.g., humans, monkeys and apes, and includes species of monkeys such from the genus Macaca (e.g., cynomolgus monkeys such as Macaca fascicularis, and/or rhesus monkeys ( Macaca mulatta )) and baboon ( Papio ursinus), as well as marmosets (species from the genus Callithrix), squirrel monkeys (species from the genus Saimiri ) and tamarins (species from the genus Saguinus), as well as species of apes such as chimpanzees ( Pan troglodytes)), rodents (e.g., mice rats, guinea pigs), lagomorphs (e.g., rabbits, hares), bovines (e.g., cattle), ovines (e.g., sheep), caprines (e.g., goats), porcines (e.g., pigs), equines (e.g., horses), canines (e.g., dogs), felines (e.g., cats), avians (e.g., chickens, turkeys, ducks, geese, companion birds such as canaries, budgerigars etc.), marine mammals (e.g., dolphins, whales), reptiles (snakes, frogs, lizards etc.), and fish. In specific embodiments, the subject is a primate such as a human.
[0067] By "pharmaceutically acceptable carrier" is meant a pharmaceutical vehicle comprised of a material that is not biologically or otherwise undesirable, i.e., the material may be administered to a subject along with the selected active agent without causing any or a substantial adverse reaction. Carriers may include excipients and other additives such as diluents, fillers, detergents, coloring agents, wetting or emulsifying agents, pH buffering agents, preservatives and the like.
[0068] Similarly, a "pharmacologically acceptable" salt, ester, amide, prodrug or derivative of a compound as provided herein is a salt, ester, amide, prodrug or derivative that this not biologically or otherwise undesirable.
[0069] The term "pharmaceutical composition" or "pharmaceutical formulation" refers to a preparation which is in such a form as to permit the biological activity of the active ingredient(s) to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the composition or formulation would be administered. Such formulations are typically sterile.
[0070] The term "pro-inflammatory mediator" means an immunoregulatory agent that favors inflammation. Such agents include, cytokines such as chemokines, interleukins (IL), lymphokines, and tumor necrosis factor (TNF) as well as growth factors and oxidative stressors. In specific embodiments, the pro-inflammatory mediator is a "pro-inflammatory cytokine". Typically, pro-inflammatory cytokines include IL-la, IL-Ib, IL-6, and TNF-a, which are largely responsible for early responses. Other pro-inflammatory mediators include, but are not limited to, LIF, IFN-y, IFN- b, IFN-a, OSM, CNTF, TGF-.b, GM-CSF, TWEAK, IL-11, IL-12, IL-15, IL-17, IL-18, IL-19, IL-20, IL- 8, IL-16, IL-22, IL-23, IL-31 and IL-32 (Tato et al., 2008. Cell 132:900; Cell 132:500, Cell 132:324). Pro-inflammatory mediators may act as endogenous pyrogens (IL-1, IL-6, IL-17, TNF- a), up-regulate the synthesis of secondary mediators and pro-inflammatory cytokines by both macrophages and mesenchymal cells (including fibroblasts, epithelial and endothelial cells), stimulate the production of acute phase proteins, or attract inflammatory cells. In specific embodiments, the term "pro-inflammatory cytokine" relates to IFN-y, IFN-b, TNF-a, IL-Ib, IL-6, IL- 17A, CCL3 and CXCL2.
[0071] As used herein, the terms "polypeptide", "proteinaceous molecule", "peptide" and "protein" are used interchangeably to refer to a polymer of amino acid residues and to variants and synthetic analogs of the same. Thus, these terms apply to amino acid polymers in which one or more amino acid residues is a synthetic non-naturally-occurring amino acid, such as a chemical analogue of a corresponding naturally-occurring amino acid, or a PEG group, as well as to naturally-occurring amino acid polymers. These terms do not exclude modifications, for example, glycosylations, acetylations, phosphorylations and the like. Soluble forms of the subject proteinaceous molecules are particularly desirable. Included within the definition are, for example, polypeptides containing one or more analogues of an amino acid including, for example, unnatural amino acids, polypeptides with substituted linkages and polypeptides with PEG groups and lipophilic moieties.
[0072] The terms "prevent", "prevention", "preventing" and the like refer to a decrease in the occurrence of disease symptoms in a patient. Prevention may be complete (no detectable symptoms) or partial, such that fewer symptoms are observed than would likely occur absent treatment. In some embodiments, prevent refers to inhibiting the development of a disease, disorder or condition, slowing the progression of a disease, disorder or condition or inhibiting progression thereof to a harmful or otherwise undesired state.
[0073] As used herein, the term "reactive oxygen species" or "ROS" refers to chemically reactive molecules containing oxygen. Examples include peroxides, superoxide, hydroxyl radical, singlet oxygen, and alpha-oxygen.
[0074] As used herein, the terms "salts" and "prodrugs" include any pharmaceutically acceptable salt, ester, hydrate or any other compound which, upon administration to the recipient, is capable of providing (directly or indirectly) a proteinaceous molecule of the invention, or an active metabolite or residue thereof. The term "pharmaceutically acceptable salts" refers without limitation to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form {e.g. by reacting the free base group with a suitable organic acid). Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate and valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. The pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
Lists of suitable salts are found in, for example, Remington (1985) Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 17th edition; Stahl and Wermuth (2002)
Pharmaceutical Salts: Properties, Selection, and Use, Wiley-VCH; and Berge et a/. (1977 ) Journal of Pharmaceutical Science, 66: 1-19, each of which is incorporated herein by reference in its entirety.
[0075] The term "sequence identity" as used herein refers to the extent that sequences are identical on a nucleotide-by-nucleotide basis or an amino acid-by-amino acid basis over a window of comparison. Thus, a "percentage of sequence identity" is calculated by comparing two optimally aligned sequences over the window of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G, I) or the identical amino acid residue (e.g., Ala, Pro, Ser, Thr, Gly, Val, Leu, lie, Phe, Tyr, Trp, Lys, Arg, His, Asp, Glu, Asn, Gin, Cys and Met) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison (/.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity. For the purposes of the present invention, "sequence identity" will be understood to mean the "match percentage" calculated by an appropriate method. For example, sequence identity analysis may be carried out using the DNASIS computer program (Version 2.5 for windows; available from Hitachi Software engineering Co., Ltd., South San Francisco, California, USA) using standard defaults as used in the reference manual accompanying the software.
[0076] "Similarity" refers to the percentage number of amino acids that are identical or constitute conservative substitutions as defined in Tables A and B supra. Similarity may be determined using sequence comparison programs such as GAP (Deveraux et a/. 1984, Nucleic Acids Research 12: 387-395). In this way, sequences of a similar or substantially different length to those cited herein might be compared by insertion of gaps into the alignment, such gaps being determined, for example, by the comparison algorithm used by GAP.
[0077] Terms used to describe sequence relationships between two or more polynucleotides or polypeptides include "reference sequence", "comparison window", "sequence identity", "percentage of sequence identity" and "substantial identity". A "reference sequence" is at least 12 but frequently 15 to 18 and often at least 25 monomer units, inclusive of nucleotides and amino acid residues, in length. Because two polynucleotides may each comprise (1) a sequence (/.e., only a portion of the complete polynucleotide sequence) that is similar between the two polynucleotides, and (2) a sequence that is divergent between the two polynucleotides, sequence comparisons between two (or more) polynucleotides are typically performed by comparing sequences of the two polynucleotides over a "comparison window" to identify and compare local regions of sequence similarity. A "comparison window" refers to a conceptual segment of at least 6 contiguous positions, usually about 50 to about 100, more usually about 100 to about 150 in which a sequence is compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. The comparison window may comprise additions or deletions (/'.e., gaps) of about 20% or less as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. Optimal alignment of sequences for aligning a comparison window may be conducted by computerized implementations of algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package Release 7.0, Genetics Computer Group, 575 Science Drive Madison, WI, USA) or by inspection and the best alignment (/.e., resulting in the highest percentage homology over the comparison window) generated by any of the various methods selected. Reference also may be made to the BLAST family of programs as for example disclosed by Altschul et al., 1997, Nucl. Acids Res. 25:3389. A detailed discussion of sequence analysis can be found in Unit 19.3 of Ausubel et al., "Current Protocols in Molecular Biology," John Wiley 8i Sons Inc, 1994-1998, Chapter 15.
[0078] As used herein, the terms "single dose" and "single dose treatment" are used interchangeably herein to refer to an effective amount of a drug that is to be taken at one time. In specific embodiments, a single dose is administered only once during the course of treatment of a condition.
[0079] As used herein a "small molecule" refers to a compound that has a molecular weight of less than 3 kilodaltons (kDa), and typically less than 1.5 kDa, and more preferably less than about 1 kDa. Small molecules may be nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic (carbon-containing) or inorganic molecules. As those skilled in the art will appreciate, based on the present disclosure, extensive libraries of chemical and/or biological mixtures, often fungal, bacterial, or algal extracts, may be screened with any of the assays of the invention to identify compounds that modulate a bioactivity. A "small organic molecule" is an organic compound (or organic compound complexed with an inorganic compound {e.g., metal)) that has a molecular weight of less than 3 kDa, less than 1.5 kDa, or even less than about 1 kDa.
[0080] "Stringency" of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization generally depends on the ability of denatured DNA to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature which can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency of hybridization reactions, see Ausubel et a/., Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995).
[0081] "Stringent conditions" or "high stringency conditions", as defined herein, can be identified by those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficol I/O.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C; or (3) overnight hybridization in a solution that employs 50% formamide, 5xSSC (0.75 M NaCI, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5xDenhardt's solution, sonicated salmon sperm DNA (50 pg/mL), 0.1% SDS, and 10% dextran sulfate at 42° C, with a 10 minute wash at 42° C in 0.2xSSC (sodium chloride/sodium citrate) followed by a 10 minute high-stringency wash consisting of O.lxSSC containing EDTA at 55° C.
[0082] As used herein, the term "systemic inflammatory response syndrome" or "SIRS" refers to a clinical response arising from a non-specific insult with two or more of the following measureable clinical characteristics; a body temperature greater than 38° C or less than 36° C, a heart rate greater than 90 beats per minute, a respiratory rate greater than 20 per minute, a white blood cell count (total leukocytes) greater than 12,000 per mm3 or less than 4,000 per mm3, or a band neutrophil percentage greater than 10%. From an immunological perspective, it may be seen as representing a systemic response to an infectious (e.g., pathogenic microbe) or non-infectious insult (e.g., major surgery) or systemic inflammation. Confirmation of infection can be determined using any suitable procedure known in the art, illustrative examples of which include blood culture, nucleic acid detection (e.g., PCR, mass spectroscopy, immunological detection (e.g., ELISA), isolation of bacteria from infected cells, cell lysis and imaging techniques such as electron microscopy.
[0083] The term "TLR7" as used herein refers to the product of the TLR7 gene, as well as homologs, orthologs, isoforms, precursor forms, processed (e.g., mature) forms, mutants, variants, derivatives, splice variants, alleles, and active fragments thereof. Representative TLR7 amino acid sequences are presented in UniProt Accession Q9NYK1 and GenPept Accession NP_057646.
[0084] As used herein, the term "treatment" and its grammatical equivalents refer to clinical intervention designed to alter the natural course of the individual or cell being treated during the course of clinical pathology. Desirable effects of treatment include decreasing the rate of disease progression, ameliorating or palliating the disease state, and remission or improved prognosis. For example, an individual is successfully "treated" if one or more symptoms associated with an acute inflammatory condition as described herein are mitigated or eliminated, including, but are not limited to, reducing exudation of fluids, including plasma proteins; and leukocytic migration into damaged or inflamed tissue, reducing classic signs of inflammation such as pain, heat, redness, swelling, and loss of function, reducing dilatation of arterioles, capillaries, and venules, and/or reducing vasculature permeability and blood flow, reducing pathogen infection, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, and/or prolonging survival of individuals. Other conditions potentially ameliorated include reduced, abrogated, modified or distorted olfactory sensation, and stress on the cardiovascular system.
[0085] The terms "vaccine" and "immunogenic composition" are used interchangeably herein to refer to a composition comprising at least one antigen which, upon inoculation into a subject, induces an immune response specific for that antigen or a cell or organism expressing the antigen and thereby confers protective immunity to the vaccinated subject against the antigen or a cell or organism expressing the antigen. With respect to a "vaccine" or "immunogenic composition" comprising, e.g., a live, attenuated virus, inoculation into a subject induces a complete or partial immunity to the pathogenic version of the virus, and/or alleviates the symptoms of disease caused by pathogenic versions of the virus. The protective effects of a vaccine against a virus are normally achieved by inducing in the subject an immune response, either a cell-mediated or a humoral immune response, or a combination of both. Generally speaking, abolished or reduced incidence of viral infection, amelioration of symptoms, or accelerated elimination of the viruses from infected subjects are indicative of the protective effects of the vaccine.
[0086] As used herein, underscoring or italicizing the name of a gene shall indicate the gene, in contrast to its protein product, which is indicated by the name of the gene in the absence of any underscoring or italicizing. For example, "TLR7" shall mean the TLR7 gene, whereas "TLR7" shall indicate the protein product or products generated from transcription and translation and/or alternative splicing of the "TLR7" gene.
[0087] Each embodiment described herein is to be applied mutatis mutandis to each and every embodiment unless specifically stated otherwise.
2. Abbreviations
[0088] The following abbreviations are used throughout the specification:
Figure imgf000020_0001
Figure imgf000021_0001
WT Wild type
3. Agents and methods for treating acute inflammatory conditions
[0089] The present disclosure is predicated in part on the determination that selective antagonism of TLR7 is useful for decreasing morbidity (e.g., weight loss), lowering viral titers, inhibiting expression of pattern recognition receptors (e.g., TLR7, TLR8 and TLR9), inhibiting expression of oxidative stressors (e.g., NOX2, NCF1 and related production of ROS), lowering the level of furin, and reducing the release of pro-inflammatory cytokines (e.g., IFN-y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2) in subjects with active RNA virus infections, including highly pathogenic H1N1 IAV infections. Of note, it was found that administration of TLR7 antagonist at five days post infection with H1N1 IAV (which is well after release of pro-inflammatory cytokines into lung tissue induced by the virus) was effective in treating the acute inflammatory condition and associated infection. Strikingly, it was also found that a single dose of a selective TLR7 antagonist could ameliorate the symptoms of acute inflammation in subjects with active H1N1 IAV infection or active rhinovirus IB strain (RV1B) infection.
[0090] Based on these findings, it is proposed that selective antagonism of TLR7 in subjects with active RNA virus infections is useful for effecting any one or more of the following activities:
1) Correcting aberrant immune signaling from TLR7, leading to reduced inflammation;
2) Reducing endosomal NOX2 mediated ROS production;
3) Reducing viral load and restricting capacity for viral replication;
4) Reducing tissue pathogenesis and morbidity in the host; and
5) Correcting the host immune response with alterations to cytokine production and improved immunity, including enhanced production of antibody, to the virus.
[0091] Since selective TLR7 antagonism was shown to be effective in reducing the level of oxidative stress mediators, as well as reducing the level of pro-inflammatory cytokines after their release into tissue, it is proposed that selective TLR7 antagonists can be used to treat acute inflammatory conditions generally, particularly those caused or exacerbated at least in part by activation of TLR7, including acute inflammatory conditions associated with the presence of cytokine release syndrome or a cytokine storm.
[0092] Thus, in accordance with the present disclosure, compositions and methods are provided that take advantage of a selective TLR7 antagonist to reduce or inhibit the production of pro-inflammatory mediators, and to treat or hinder the development of acute inflammatory conditions in individuals that have acute inflammation, as described hereafter. As noted above, "selective antagonism" of TLR7 does not necessarily require complete abrogation of TLR7 activity; rather it encompasses modulation of TLR7 activity to effect any one or more of activities 1) to 5) listed above, and to maintain, return to, or stimulate a return towards, immune homeostasis. Selective TLR7 antagonists with such properties are also referred to herein as "immune homeostasis-supporting agents". 3.1 Selective TLR7 antagonists
[0093] Selective TLR7 antagonists include and encompass any active agent that reduces the accumulation, function or stability of TLR7; or decrease expression of the TLR7 gene, and such inhibitors include without limitation, small molecules and macromolecules such as nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, polysaccharides, lipopolysaccharides, lipids or other organic (carbon containing) or inorganic molecules. Selective TLR7 antagonists may be direct TLR7 antagonists or indirect TLR7 antagonists.
[0094] In some embodiments, the selective TLR7 antagonist is an antagonistic nucleic acid molecule that functions to inhibit the transcription or translation of TLR7 transcripts. Representative transcripts of this type include nucleotide sequences corresponding to any one the following sequences: (1) human TLR7 nucleotide sequences as set forth for example in GenBank Accession Nos. NM_016562; (2) nucleotide sequences that share at least 70, 71, 72, 73, 74, 75,
76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% sequence identity with any one of the sequences referred to in (1); (3) nucleotide sequences that hybridize under high stringency conditions to the sequences referred to in (1); (4) nucleotide sequences that encode human TLR7 amino acid sequences as set forth for example in GenPept Accession NP_057646 and UniProt Accession Q9NYK1; (5) nucleotide sequences that encode an amino acid sequence that shares at least 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83,
84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% sequence similarity with any one of the sequences referred to in (4); and (6) nucleotide sequences that encode an amino acid sequence that shares at least 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86,
87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% sequence identity with any one of the sequences referred to in (4).
[0095] Illustrative antagonist nucleic acid molecules include antisense molecules, ribozymes and triplex forming molecules, as well as shRNA molecules, siRNA molecules and external guide sequences. Representative TLR7 inhibitory RNA molecules are available commercially from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA) and OriGene Technologies, Inc. (Rockville, MD, USA).
[0096] Selective TLR7 antagonist oligonucleotides are disclosed for example by Uhlmann et al. in U.S. Publication No. 20160355822. Illustrative oligonucleotides of this type are represented by formula (I):
XiAATGGPyPuGGGPxAGPy (I) wherein:
Py is 5-substituted cytidine selected from the group consisting of 5-methyl-dC, 5- bromo-dC and 5-octadienyl-dC,
Pu is a 7-deaza purine derivative selected from the group consisting of 7-deaza- dG, 7-deaza-2'-0-methyl-G, inosine and 7-deaza-inosine,
Px is dA, 5-substituted deoxyuridine, or 5-iodo-uridine, and
XI is any nucleotide or no nucleotide.
[0097] Alternative selective TLR7 antagonist oligonucleotides are disclosed by Guiducci et al. in U.S. Publication No. 20150252363, which are represented by formula (II) or formula (III): 5'-QzTICNx-3 (II)
5'-QzTTCNx-3 (III) wherein: each of Q and N is a nucleotide or nucleotide analog, x is an integer from 3 to 50, z is 0, 1 or 2, and wherein the polynucleotide does not comprise a CG dinucleotide.
[0098] In exemplary embodiments, the oligonucleotide comprises, consists or consists essentially of 5'-TICTCCTCCTTGAGIAII-3' [SEQ ID NO:l] or 5'-TICTTCTCCTTGAGIAII-3' [SEQ ID NO:2].
[0099] The present disclosure further contemplates proteinaceous molecules that selectively antagonize TLR7. Exemplary proteinaceous molecules of this type include "decoy" (or "trap") peptides disclosed by Selemidis et at. in International Publication WO 2019/000045, which is incorporated herein by reference in its entirety. These proteinaceous molecules, which are also referred to herein as "immune homeostasis-supporting agents", antagonize disulfide bond formation between C98 and C475 of human (or murine) TLR7 or their corresponding positions in the TLR7s of other species, and are generally from about 4 to about 190 amino acids in length and comprise, consist or consist essentially of an amino acid sequence with at least 70%, 75%, 80%, 85%.90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%, or 100%, sequence similarity or identity to up to 190 contiguous amino acids between amino acids 4 to 194 of TLR7 human (or murine), which includes a peptide comprising, consisting or consisting essentially of an amino acid sequence with at least 70% 70%, 75%, 80%, 85%.90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%, or 100%, sequence similarity or identity to up to 100 contiguous amino acids between amino acids 48 to 148 of TLR7 and which also includes a peptide comprising, consisting or consisting essentially of an amino acid sequence with at least 70%, 75%, 80%, 85%.90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%, or 100%, sequence similarity or identity to up to 40 contiguous amino acids between amino acid 78 to 118 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7 (C98). In specific embodiments, these proteinaceous molecules are represented by formula IV:
Z1DX1RCNCX2PX3X4Z2 (IV) wherein:
Zi and Z2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
Xi is a hydrophobic amino acid (e.g., L, F or M, or modified forms thereof);
X2 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
X3 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid (e.g., A or P, or modified forms thereof); and X4 is small amino acid ( e.g ., P, or modified forms thereof), a hydrophobic amino acid ( e.g ., L, or modified forms thereof) or a basic amino acid {e.g., K or R, or modified forms thereof).
[00100] In representative examples of this type, a proteinaceous molecule represented by formula IV comprises, consists or consists essentially of the amino acid sequence DFRCNCVPIP [SEQ ID NO:3] corresponding to human TLR7 from D95 to P104, or DLRCNCVPVL [SEQ ID NO:4] corresponding to murine TLR7 from D95 to L104.
[0100] Additional amino acids or other substituents may be added to the N- or C- termini, if present, of the proteinaceous molecules. For example, the proteinaceous molecules may form part of a longer sequence with additional amino acids added to either or both of the N- and C- termini.
[0101] For particular uses and methods of the present disclosure, proteinaceous molecules with high levels of stability may be desired, for example, to increase the half-life of the proteinaceous molecule in a subject. Thus, in some embodiments, proteinaceous molecules comprise a stabilizing moiety or protecting moiety. The stabilizing moiety or protecting moiety may be coupled at any point on the molecule. Suitable stabilizing or protecting moieties include, but are not limited to, polyethylene glycol (PEG), a glycan or a capping moiety, including an acetyl group, pyroglutamate or an amino group. In specific embodiments, the acetyl group and/or pyroglutamate are coupled to the N-terminal amino acid residue of the proteinaceous molecule. In particular embodiments, the N-terminus of the proteinaceous molecule is an acetamide. In some embodiments, the amino group is coupled to the C-terminal amino acid residue of the proteinaceous molecule. In particular embodiments, the proteinaceous molecule has a primary, secondary or tertiary amide, a hydrazide or a hydroxamide at the C-terminus; particularly a primary amide at the C-terminus. In some embodiments, the PEG is coupled to the N-terminal or C-terminal amino acid residue of the proteinaceous molecule or through the amino group of a lysine side-chain or other suitably modified side-chain that is already present or is artificially incorporated into the proteinaceous molecule.
[0102] Although the proteinaceous molecules may inherently permeate membranes, membrane permeation may further be increased by the conjugation of a membrane permeating moiety to the proteinaceous molecule. Accordingly, in some embodiments, proteinaceous molecules according to formula IV may comprise a membrane permeating moiety. The membrane permeating moiety may be coupled at any point on the proteinaceous molecule. Suitable membrane permeating moieties include lipid moieties, cholesterol and proteins, such as cell penetrating peptides and polycationic peptides; especially lipid moieties.
[0103] Suitable cell penetrating peptides may include the peptides described in, for example, US 20090047272, US 20150266935 and US 20130136742. Accordingly, suitable cell penetrating peptides may include, but are not limited to, basic poly(Arg) and poly(Lys) peptides and basic poly(Arg) and poly(Lys) peptides containing non-natural analogues of Arg and Lys residues such as YGRKKRPQRRR (HIV TAT47-57; SEQ ID NO: 5), RRWRRWWRRWWRRWRR (W/R;
SEQ ID NO:6), CWKi8 (AlkCWKi8; SEQ ID NO:7), Ki8WCCWKi8 (Di-CWKi8; SEQ ID NO:8),
WTLN SAGYLLG KI N LKALAALAKKI L (Transportan; SEQ ID NO:9), GLFEALEELWEAK (DipaLytic; SEQ ID NO: 10), KieGGCRGDMFGCAKieRGD (KieRGD; SEQ ID NO: 11), KieGGCMFGCGG (PI; SEQ ID NO: 12), KielCRRARGDNPDDRCT (P2; SEQ ID NO: 13), KKWKMRRNQFWVKVQRbAK (B) bA (P3; SEQ ID NO: 14), VAYISRGGVSTYYSDTVKGRFTRQKYNKRA (P3a; SEQ ID NO: 15), IGRIDPANGKTKYAPKFQDKATRSNYYGNSPS (P9.3; SEQ ID NO: 16), KETWWETWWTEWSQPKKKRKV (Pep-1; SEQ ID NO: 17), PLAEIDGIELTY (Plae; SEQ ID NO: 18), KieGGPLAEIDGIELGA (Kplae; SEQ ID NO: 19), KieGGPLAEIDGIELCA (cKplae; SEQ ID NO: 20), GALFLGFLGGAAGSTMGAWSQPKSKRKV (MGP; SEQ ID NO:21), WEAK(LAKA)2-LAKH(LAKA)2LKAC (HA2; SEQ ID NO:22), (LARL)6NHCH3 (LARL46; SEQ ID NO:23), KLLKLLLKLWLLKLLL (Hel-11-7; SEQ ID NO:24), (KKKK)2GGC (KK; SEQ ID NO:25), (KWKK)2GCC (KWK; SEQ ID NO:26), (RWRR)2GGC (RWR; SEQ ID NO:27), PKKKRKV (SV40 NLS7; SEQ ID NO:28), PEVKKKRKPEYP (NLS12; SEQ ID NO:29), TPPKKKRKVEDP (NLS12a; SEQ ID NO:30), GGGGPKKKRKVGG (SV40 NLS13; SEQ ID NO:31), GGGFSTSLRARKA (AV NLS13; SEQ ID NO:32), CKKKKKKSEDEYPYVPN (AV RME NLS17; SEQ ID NO:33), CKKKKKKKSEDEYPYVPNFSTSLRARKA (AV FP NLS28; SEQ ID NO:34), LVRKKRKTEEESPLKDKDAKKSKQE (SV40 N1 NLS24; SEQ ID NO:35), and K9K2K4K8GGK5 (Loligomer; SEQ ID NO:36); HSV-1 tegument protein VP22; HSV-1 tegument protein VP22r fused with nuclear export signal (NES); mutant B-subunit of Escherichia coli enterotoxin EtxB (H57S); detoxified exotoxin A (ETA); the protein transduction domain of the HIV-1 Tat protein, GRKKRRQRRRPPQ (SEQ ID NO:37); the Drosophila melanogaster Antennapedia domain Antp (amino acids 43-58), RQIKIWFQNRRMKWKK (SEQ ID NO:38); Buforin II,
TRSSRAGLQFPVGRVHRLLRK (SEQ ID NO:39); hClock-(amino acids 35-47) (human Clock protein DNA-binding peptide), KRVSRNKSEKKRR (SEQ ID NO:40); MAP (model amphipathic peptide), KLALKLALKALKAALKLA (SEQ ID NO:41); K-FGF, AAVALLPAVLLALLAP (SEQ ID NO:42); Ku70- derived peptide, comprising a peptide selected from the group comprising VPMLKE (SEQ ID NO:43), VPMLK (SEQ ID NO:44), PMLKE (SEQ ID NO:45) or PMLK (SEQ ID NO:46); Prion, Mouse Prpe (amino acids 1-28), MANLGYWLLALFVTMWTDVGLCKKRPKP (SEQ ID NO:47); pVEC, LLIILRRRIRKQAHAHSK (SEQ ID NO:48); Pep-I, KETWWETWWTEWSQPKKKRKV (SEQ ID NO:49); SynBI, RGGRLSYSRRRFSTSTGR (SEQ ID NO:50); Transportan, G WTLN SAGYLLG KI N LKALAALAKKI L (SEQ ID NO:51); Transportan-10, AGYLLG KIN LKALAALAKKI L (SEQ ID NO:52); CADY, Ac- GLWRALWRLLRSLWRLLWRA-cysteamide (SEQ ID NO: 53); Pep-7, SDLWEMMMVSLACQY (SEQ ID NO:54); HN-1, TSPLNIHNGQKL (SEQ ID NO:55); VT5, D P KG D PKG VTVTVTVTVT GKGDPKPD (SEQ ID NO:56); or pISL, RVIRVWFQNKRCKDKK (SEQ ID NO: 57).
[0104] In some embodiments, the membrane permeating moiety is a lipid moiety, such as a Cio-C2o fatty acyl group, especially stearoyl (octadecanoyl; Cis), palmitoyl (hexadecanoyl; Ci6) or myristoyl (tetradecanoyl; CM) . The membrane permeating moiety may be coupled to the N- or C-terminal amino acid residue or through the amino group of a lysine side-chain that is already present or is artificially incorporated into the proteinaceous molecule or other suitably modified side-chain, especially the N-terminal amino acid residue of the proteinaceous molecule or through the amino group of a lysine side-chain. In particular embodiments, the membrane permeating moiety is coupled through the amino group of the N-terminal amino acid residue.
[0105] Alternatively, cholestanol may be employed as the membrane permeating moiety to facilitate uptake of the proteinaceous molecule into cells. In specific embodiments of this type, cell uptake is facilitated by a NOX2-cholestanol-linker (PEG)-gp91ds-TAT construct and linking cholestanol and PEG to gp91ds-TAT facilities delivery of gp91ds-TAT to the endosome.
[0106] The present disclosure also contemplates small molecule agents that antagonize the function, including activation of TLR7. [0107] Non-limiting examples of small molecule TLR7 antagonists may be selected from those disclosed by Bou Karroum et al. {J Med Chem., 62(15): 7015-7031 (2019)). Representative compounds of this type include imidazo[l,2-a]pyrazines, pyrazolo[l,5-a]quinoxalines, and imidazo[l,5-a]quinoxalines.
[0108] In specific embodiments, the imidazo[l,2-a]pyrazine compounds are represented by formula (V):
Figure imgf000027_0001
wherein:
R1 is an acyl moiety that is suitably selected from:
Figure imgf000027_0002
[0109] In certain embodiments, the pyrazolo[l,5-a]quinoxaline compounds are represented by formula (VI):
Figure imgf000027_0003
wherein:
R1 is an acyl moiety that is suitably selected from:
Figure imgf000028_0004
R2 and R3 are independently H, or methyl.
[0110] In specific embodiments, the imidazo[l,5-a]quinoxaline compounds are represented by formula (VII):
Figure imgf000028_0001
wherein:
R1 is an acyl moiety that is suitably selected from:
Figure imgf000028_0002
[0111] Preferred small molecule TLR7 antagonists of this type include pyrazolo[l,5- a]quinoxaline compounds according to formula (VI) wherein:
R1 is:
Figure imgf000028_0003
R2 and R3 are each H.
4. Pharmaceutical compositions
[0112] Also provided herein are pharmaceutical compositions or formulations comprising a selective TLR7 antagonist and a pharmaceutically acceptable carrier. These compositions can be prepared by mixing the active ingredients {e.g., a small molecule, nucleic acid, or proteinaceous molecules) having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)). Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes {e.g., Zn-protein complexes); and/or non-ionic surfactants such as polyethylene glycol (PEG). Exemplary pharmaceutically acceptable carriers herein further include interstitial drug dispersion agents such as soluble neutral-active hyaluronidase glycoproteins (sHASEGP), for example, human soluble PH-20 hyaluronidase glycoproteins, such as rHuPH20 (HYLENEX®, Baxter International, Inc.). Certain exemplary sHASEGPs and methods of use, including rHuPH20, are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968. In one aspect, a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
[0113] In some embodiments, especially relating to proteinaceous compounds (e.g., TLR7 decoy peptides), the active agents and optional pharmaceutically acceptable carriers are in the form of lyophilized formulations or aqueous solutions. Exemplary lyophilized antibody formulations are described in U.S. Pat. No. 6,267,958. Aqueous antibody formulations include those described in U.S. Pat. No. 6,171,586 and W02006/044908, the latter formulations including a histidine-acetate buffer.
[0114] The compositions disclosed herein may also contain further active ingredients as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. Such active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
[0115] Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (e.g., liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
[0116] Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsules. The formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
[0117] Depending on the specific conditions being treated, the formulations may be administered systemically or locally. Suitable routes may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections. Techniques for formulation and administration may be found in "Remington's Pharmaceutical Sciences", Mack Publishing Co., Easton, Pa., latest edition.
[0118] The pharmaceutical forms suitable for injectable use include sterile injectable solutions or dispersions and sterile powders for the preparation of sterile injectable solutions. Such forms should be stable under the conditions of manufacture and storage and may be preserved against reduction, oxidation and microbial contamination.
5. Ancillary therapeutic agents
[0119] The pharmaceutical compositions of the present disclosure may further comprises at least one additional or ancillary therapeutic agent for treating the acute inflammatory condition and in the case of an acute inflammatory condition associated with the presence of a pathogenic infection, the ancillary therapeutic agent may be an antigenic composition for immunizing and eliciting an immune response to the pathogen.
[0120] According to some such embodiments, the ancillary therapeutic agent comprises EXCOOl (an anti-sense RNA against connective tissue growth factor (CTGF)), AZX100 (a phosphopeptide analog of Heat Shock Protein 20 (HSP20)), PRM-151 (recombinant human serum amyloid P/Pentaxin 2), PXL01 (a synthetic peptide derived from human lactoferrin), DSC127 (an angiotensin analog), RXI-109 (a self-delivering RNAi compound that targets connective tissue growth factor (CTGF)), TCA (trichloroacetic acid), Botulium toxin type A, or a combination thereof.
[0121] In other embodiments, the ancillary therapeutic agent is an anti-inflammatory agent, representative examples of which include steroidal anti-inflammatory agents such as but not limited to compounds containing a 17-carbon 4-ring system, including sterols, various hormones (as anabolic steroids), and glycosides. Representative examples of steroidal anti-inflammatory drugs include, without limitation, corticosteroids such as hydrocortisone, hydroxyltriamcinolone, alpha-methyl dexamethasone, dexamethasone-phosphate, beclomethasone dipropionates, clobetasol valerate, desonide, desoxymethasone, desoxycorticosterone acetate, dexamethasone, dichlorisone, diflucortolone valerate, fluadrenolone, fluclorolone acetonide, flumethasone pivalate, fluosinolone acetonide, fluocinonide, flucortine butylesters, fluocortolone, fluprednidene (fluprednylidene) acetate, flurandrenolone, halcinonide, hydrocortisone acetate, hydrocortisone butyrate, methylprednisolone, triamcinolone acetonide, cortisone, cortodoxone, flucetonide, fludrocortisone, difluorosone diacetate, fluradrenolone, fludrocortisone, diflorosone diacetate, fluradrenolone acetonide, medrysone, amcinafel, amcinafide, betamethasone and the balance of its esters, chloroprednisone, chlorprednisone acetate, clocortelone, clescinolone, dichlorisone, diflurprednate, flucloronide, flunisolide, fluoromethalone, fluperolone, fluprednisolone, hydrocortisone valerate, hydrocortisone cyclopentylpropionate, hydrocortamate, meprednisone, paramethasone, prednisolone, prednisone, beclomethasone dipropionate, triamcinolone, and mixtures thereof.
[0122] Alternatively, the anti-inflammatory agent may be a nonsteroidal anti inflammatory agent, non-limiting examples of which include agents that are aspirin-like in their action, including, but not limited to, ibuprofen (ADVIL), naproxen sodium (ALEVE), and acetaminophen (TYLENOL). Additional examples of non-steroidal anti-inflammatory agents that are usable in the context of the described invention include, without limitation, oxicams, such as piroxicam, isoxicam, tenoxicam, sudoxicam, and CP-14,304; disalcid, benorylate, trilisate, safapryn, solprin, diflunisal, and fendosal; acetic acid derivatives, such as diclofenac, fenclofenac, indomethacin, sulindac, tolmetin, isoxepac, furofenac, tiopinac, zidometacin, acematacin, fentiazac, zomepirac, clindanac, oxepinac, felbinac, and ketorolac; fenamates, such as mefenamic, meclofenamic, flufenamic, niflumic, and tolfenamic acids; propionic acid derivatives, such as benoxaprofen, flurbiprofen, ketoprofen, fenoprofen, fenbufen, indopropfen, pirprofen, carprofen, oxaprozin, pranoprofen, miroprofen, tioxaprofen, suprofen, alminoprofen, and tiaprofenic; pyrazoles, such as phenylbutazone, oxyphenbutazone, feprazone, azapropazone, and trimethazone. Mixtures of these non-steroidal anti-inflammatory agents also may be employed, as well as the dermatologically acceptable salts and esters of these agents. For example, etofenamate, a flufenamic acid derivative, is particularly useful for topical application.
[0123] In other embodiments, the anti-inflammatory agent includes, without limitation, transforming growth factor-beta 3 (TGF-83), an anti-tumor necrosis factor-alpha (TNF-a) agent, an inhibitor or antagonist of IL-6 or IL-6 receptor, IL-1 receptor, IL-Ib, TNF, GM-CSF, IFN-y, JAK- STAT signaling, CCR2, CCR5, complement component C5, IRAK4 and M-CSF receptor, or a combination thereof.
[0124] In still other embodiments, the ancillary therapeutic agent is an analgesic agent. In representative examples of this type, the analgesic agent relieves pain by elevating the pain threshold without disturbing consciousness or altering other sensory modalities. According to some such embodiments, the analgesic agent is a non-opioid analgesic, which include natural or synthetic substances that reduce pain but are not opioid analgesics. Examples of non-opioid analgesics include, but are not limited to, etodolac, indomethacin, sulindac, tolmetin, nabumetone, piroxicam, acetaminophen, fenoprofen, flurbiprofen, ibuprofen, ketoprofen, naproxen, naproxen sodium, oxaprozin, aspirin, choline magnesium trisalicylate, diflunisal, meclofenamic acid, mefenamic acid, and phenylbutazone. According to some other embodiments, the analgesic is an opioid analgesic, illustrative examples of which include codeine, fentanyl, hydromorphone, levorphanol, meperidine, methadone, morphine, oxycodone, oxymorphone, propoxyphene, buprenorphine, butorphanol, dezocine, nalbuphine, and pentazocine.
[0125] In some embodiments, the ancillary therapeutic agent is an antimicrobial agent, which includes without limitation compounds that kill or inhibit the growth of microorganisms such as viruses, bacteria, yeast, fungi, protozoa, etc. and thus include antibiotics, antifungals, anti protozoa Is, antimalarials, antituberculotics and antivirals. Illustrative antibiotics include quinolones (e.g., amifloxacin, cinoxacin, ciprofloxacin, enoxacin, fleroxacin, flumequine, lomefloxacin, nalidixic acid, norfloxacin, ofloxacin, levofloxacin, lomefloxacin, oxolinic acid, pefloxacin, rosoxacin, temafloxacin, tosufloxacin, sparfloxacin, clinafloxacin, gatifloxacin, moxifloxacin; gemifloxacin; and garenoxacin), tetracyclines, glycylcyclines and oxazolidinones (e.g., chlortetracycline, demeclocycline, doxycycline, lymecycline, methacycline, minocycline, oxytetracycline, tetracycline, tigecycline; linezolide, eperozolid), glycopeptides, aminoglycosides (e.g., amikacin, arbekacin, butirosin, dibekacin, fortimicins, gentamicin, kanamycin, meomycin, netilmicin, ribostamycin, sisomicin, spectinomycin, streptomycin, tobramycin), b-lactams (e.g., imipenem, meropenem, biapenem, cefaclor, cefadroxil, cefamandole, cefatrizine, cefazedone, cefazolin, cefixime, cefmenoxime, cefodizime, cefonicid, cefoperazone, ceforanide, cefotaxime, cefotiam, cefpimizole, cefpodoxime, cefsulodin, ceftazidime, cefteram, ceftezole, ceftibuten, ceftizoxime, ceftriaxone, cefuroxime, cefuzonam, cephaacetrile, cephalexin, cephaloglycin, cephaloridine, cephalothin, cephapirin, cephradine, cefinetazole, cefoxitin, cefotetan, azthreonam, carumonam, flomoxef, moxalactam, amidinocillin, amoxicillin, ampicillin, azlocillin, carbenicillin, benzyl penici Min, carfecillin, cloxacillin, d icloxaci Min, methicillin, mezlocillin, nafcillin, oxacillin, penicillin G, piperacillin, sulbenicillin, temocillin, ticarcillin, cefditoren, SC004, KY-020, cefdinir, ceftibuten, FK-312, S-1090, CP-0467, BK-218, FK-037, DQ-2556, FK-518, cefozopran, ME1228, KP-736, CP-6232, Ro 09-1227, OPC-20000, LY206763), rifamycins, macrolides (e.g., azithromycin, clarithromycin, erythromycin, oleandomycin, rokitamycin, rosaramicin, roxithromycin, troleandomycin), ketolides (e.g., telithromycin, cethromycin), coumermycins, lincosamides (e.g., clindamycin, lincomycin) and chloramphenicol. Representative antivirals include abacavir sulfate, acyclovir sodium, amantadine hydrochloride, amprenavir, cidofovir, delavirdine mesylate, didanosine, efavirenz, famciclovir, fomivirsen sodium, foscarnet sodium, ganciclovir, indinavir sulfate, lamivudine, lamivudine/zidovudine, nelfinavir mesylate, nevirapine, oseltamivir phosphate, ribavirin, rimantadine hydrochloride, ritonavir, saquinavir, saquinavir mesylate, stavudine, valacyclovir hydrochloride, valganciclovir, zalcitabine, zanamivir, and zidovudine. Non-limiting examples antiprotozoals include atovaquone, chloroquine hydrochloride, chloroquine phosphate, metronidazole, metronidazole hydrochloride, and pentamidine isethionate. Anthelmintics can be at least one selected from mebendazole, pyrantel pamoate, albendazole, ivermectin and thiabendazole. Illustrative antifungals can be selected from amphotericin B, amphotericin B cholesteryl sulfate complex, amphotericin B lipid complex, amphotericin B liposomal, bifonazole, butoconazole, chlordantoin, chlorphenesin, ciclopirox olamine, clotrimazole, eberconazole, econazole, fluconazole, flucytosine, flutrimazole, griseofulvin microsize, griseofulvin ultramicrosize, itraconazole, isoconazole, itraconazole, ketoconazole, miconazole, nifuroxime, nystatin, terbinafine hydrochloride, tioconazole, terconazole and undecenoic acid. Non-limiting examples of antimalarials include chloroquine hydrochloride, chloroquine phosphate, doxycycline, hydroxychloroquine sulfate, mefloquine hydrochloride, primaquine phosphate, pyrimethamine, and pyrimethamine with sulfadoxine. Antituberculotics include but are not restricted to clofazimine, cycloserine, dapsone, ethambutol hydrochloride, isoniazid, pyrazinamide, rifabutin, rifampin, rifapentine, and streptomycin sulfate. In specific embodiments, the ancillary agent is an antiviral, such as remdesivir, lopinavir, ritonavir, emtricitabine, tenofovir, ivermectin, faviparavir, imatininb and baricitinib, and combinations thereof (e.g. lopinavir/ritonavir, or emtricitabine/tenofovir).
[0126] Alternatively, or in addition, one can administer compounds which inhibit the cytokine release syndrome or cytokine storm, anti-coagulants and/or platelet aggregation inhibitors that address blood clots, compounds which chelate iron ions released from hemoglobin by viruses such as COVID-19, cytochrome P-450 (CYP450) inhibitors and/or NOX inhibitors, as ancillary therapeutic agents.
[0127] Representative NOX inhibitors are disclosed in PCT/US2018/067674, and include AEBSF, Apocynin, DPI, GK-136901, ML171, Plumbagin, S17834, VAS2870, VAS3947, GKT-831, GKT771, GTL003 or amido thiadiazole derivatives thereof, as described in AU2015365465, EP20140198597; and WO2015/59659, Schisandrin B, as described in CN104147001 and CN20131179455), bi-aromatic and tri-aromatic compounds described in U.S. Publication No. 2015045387, GB 20110016017, and W0201200725, methoxyflavone derivatives described in JP 2015227329, JP 20140097875, and JP 20150093939, peptides, such as NOX2ds-tat and PR-39, as described in U.S. Publication No. 2015368301, TN 2015000295, U.S. Publication No.
201514689803, U.S. Publication No. 201462013916, PCT WO 201450063, and EP 20130150187, piperazine derivatives described in U.S. Publication No. 2014194422, U.S. Pat. No. 9,428,478, U.S. Publication No. 201214123877, U.S. Publication No. 201161496161, and PCT WO 2012US41988, pyrazole derivatives disclosed in KR101280198, KR20110025151, and KR20090082518, pyrazoline dione derivatives disclosed in HK1171748, PCT WO201054329, and EP 20090171466, pyrazolo piperidine derivatives disclosed in KR20130010109, KR20130002317, EP20100153927, PCT W0201150667, EP20100153929, and PCT W02011IB50668, pyrazolo pyridine derivatives described in KR20170026643, HK1158948, HK1141734, HK1159096, HK1159092,
EP20080164857, PCT WO200954156, PCT W0200954150, EP20080164853, PCT W0200853390, U.S. Publication No. 20070896284, EP20070109555, PCT WO 200954148, EP20080164847, PCT WO200954155, and EP20080164849, quinazoline and quinoline derivatives disclosed in EP2886120, U.S. Publication No. 2014018384, U.S. Publication No. 20100407925,
EP20110836947, GB20110004600, and PCT WO 201250586, tetrahydroindole derivatives disclosed in U.S. Publication No. 2010120749, U.S. Pat. No. 8,288,432, U.S. Publication No. 20080532567, EP20070109561, U.S. Publication No. 20070908414, and PCT WO 200853704, tetrahydroisoquinoline derivatives disclosed in U.S. Publication No. 2016083351, U.S. Publication No. 201414888390, U.S. Publication No. 201361818726, and PCT WO 201436402, Scopoletin, described in TW201325588 and TW20110147671, and 2,5-disubstituted benzoxazole and benzothiazole derivatives disclosed in TW201713650 and PCT WO 201554662. Representative NOX inhibitors also include those disclosed in PCT WO2011062864.
[0128] Exemplary NOX inhibitors also include 2-phenylbenzo[d]isothiazol-3(2H)-one, 2- (4-methoxyphenyl)benzo[d]isothiazol-3(2H)-one, 2-(benzo[d][l,3]dioxol-5-yl)benzo[d]isothiazol- 3(2H)-one, 2-(2,4-dimethylphenyl)benzo[d]isothiazol-3(2H)-one, 2-(4- fluorophenyl)benzo[d]isothiazol-3(2H)-one, 2-(2, 4-dimethyl phenyl)-5-fl uorobenzo[d] isothiazol- 3(2H)-one, 5-fluoro-2-(4-fluorophenyl)benzo[d]isothiazol-3(2H)-one, 2-(2-chloro-6-methylphenyl)- 5-fluorobenzo[d]isothiazol-3(2H)-one, 5-fluoro-2-phenylbenzo[d]isothiazol-3(2H)-one, 2- (benzo[d][l,3]dioxol-5-yl)-5-fluorobenzo[d]isothiazol-3(2H)-one, methyl 4-(3- oxobenzo[d]isothiazol-2(3H)-yl) benzoate, methyl 4-(5-fluoro-3-oxobenzo[d]isothiazol-2(3H)- yl)benzoate, ethyl 4-(3-oxobenzo[d]isothiazol-2(3H)-yl)benzoate, tert-butyl 4-(3- oxobenzo[d]isothiazol-2(3H)-yl) benzoate, methyl 2-methoxy-4-(3-oxobenzo[d]isothiazol-2(3H)- yl)benzoate, methyl 3-chloro-4-(3-oxobenzo[d]isothiazol-2(3H)-yl)benzoate, 4-(3- oxobenzo[d]isothiazol-2(3H)-yl)benzonitrile, methyl 2-(3-oxobenzo[d]isothiazol-2(3H)-yl) benzoate, 2-(4-acetylphenyl)benzo[d]isothiazol-3(2H)-one, 2-(4-nitrophenyl)benzo[d]isothiazol-3(2H)-one, 2-(4-hydroxyphenyl)benzo[d]isothiazol-3(2H)-one, methyl 6-(3-oxobenzo[d]isothiazol-2(3H)- yl)nicotinate, 6-(3-oxobenzo[d]isothiazol-2(3H)-yl)nicoti nonitrile, 2-(4-
(hydroxymethyl)phenyl)benzo[d]isothiazol-3(2H)-one, 2-benzylbenzo[d]isothiazol-3(2H)-one, N- methyl-4-(3-oxobenzo[d]isothiazol-2(3H)-yl)benzamide, 2-(4-hydroxyphenyl)benzo[d]isothiazol- 3(2H)-one, 2-(2,4-dimethylphenyl)- 1-methyl- lH-indazol-3(2H)-one, 2-(4-fluorophenyl)-l-methyl- lH-indazol-3 (2H)-one, 2-(2,4-dimethylphenyl)-lH-indazo-3(2H)-one, l-methyl-2-phenyl-lH- indazol-3 (2H)-one, 2-(l,3,4-thiadiazol-2-yl)benzo[d]isothiazol-3(2H)-one, 2-(5-phenyl-l,3,4- thiadiazol-2-yl)benzo[d]isothiazol-3(2H)-one, 2-(5-(ethylthio)-l,3,4-thiadiazol-2- yl)benzo[d]isothiazol-3(2H)-one, 2-(5-(methylthio)-l,3,4-thiadiazol-2-yl)benzo[d]isothiazol-3(2H)- one, 5-fluoro-2-(l,3,4-thiadiazol-2-yl)benzo[d]isothiazol-3(2H)-one, 2-(5-(tert-butyl)-l,3,4- thiadiazol-2-yl)benzo[d]isothiazol-3(2H)-one, 2-(5-(4-bromophenyl)-l,3,4-thiadiazol-2- yl)benzo[d]isothiazol-3(2H)-one 2-(4-methylthiazol-2-yl)benzo[d]isothiazol-3(2H)-one, 2-(4,5- dimethylthiazol-2-yl)benzo[d]isothiazol-3(2H)-one, 2-(benzo[d][l,3]dioxol-5-yl)-4,5- difluorobenzo[d][l,2]selenazol-3(2H)-one- , 2-(benzo[d][l,3]dioxol-5-yl)-5- fluorobenzo[d][l,2]selenazol-3(2H)-one, 2-(2,3-dihydrobenzo[b][l,4]dioxin-6-yl)-5- fluorobenzo[d][l,2]selenazol-3(- 2H)-2-(4-( 1, 3-dioxolan-2-yl) phenyl) benzo[d][l, 2]selenazol- 3(2H)-one, 2-(benzo[d][l,3]dioxol-5-yl)-6, 7-dimethoxybenzo[d][l,2]selenazol-3(2H)-one, methyl 4-(3-oxobenzo[d][l,2]selenazol-2(3H)-yl) benzoate, methyl 4-(3-oxoisothiazolo[5,4-b]pyridin- 2(3H)-yl)benzoate, and ethyl 4-(3-oxoisothiazol-2(3H)-yl)benzoate, and pharmaceutically acceptable salts and prodrugs thereof.
[0129] Non-limiting examples of CYP450 inhibitors include amiodarone, amlodipine, apigenin, aprepitant, bergamottin (grapefruit), buprenorphine, bupropion, caffeine, cafestol, cannabidiol, celecoxib, chloramphenicol, chlorphenamine, chlorpromazine, cimetidine, cinacalcet, ciprofloxacin, citalopram, clarithromycin, clemastine, clofibrate, clomipramine, clotrimazole, cobicistat, cocaine, curcumin (turmeric), cyclizine, delavirdine, desipramine, disulfiram, diltiazem, diphenhydramine, dithiocarbamate, domperidone, doxepin, doxorubicin, duloxetine, echinacea, entacapone, erythromycin, escitalopram, felbamate, fenofibrate, flavonoids (grapefruit), fluoroquinolones (e.g., ciprofloxacin), fluoxetine, fluvoxamine, fluconazole, fluvastatin, gabapentin, gemfibrozil, gestodene, halofantrine, haloperidol, hydroxyzine, imatinib, indomethacin, indinavir, interferon, isoniazid, itraconazole, JWH-018, ketoconazole, letrozole, lovastatin, levomepromazine, memantine, methylphenidate, metoclopramide, methadone, methimazole, methoxsalen, metyrapone, mibefradil, miconazole, midodrine, mifepristone, milk thistle, moclobemide, modafinil, montelukast, moclobemide, naringenin (grapefruit), nefazodone, nelfinavir, niacin, niacinamide, nicotine, nicotinamide, nilutamide, norfloxacin, orphenadrine, paroxetine, perphenazine, pilocarpine, piperine, phenylbutazone, probenecid, promethazine, proton pump inhibitors (e.g., lansoprazole, omeprazole, pantoprazole, rabeprazole), quercetin, quinidine, ranitidine, risperidone, ritonavir, saquinavir, selegiline, sertraline, star fruit, St. John's wort, sulconazole, sulfamethoxazole, sulfaphenazole, telithromycin, teniposide, terbinafine, thiazolidinediones, thioridazine, ticlopidine, tioconazole, thiotepa, trimethoprim, topiramate, tranylcypromine, tripelennamine, valerian, valproic acid, verapamil, voriconazole, zafirlukast, and zuclopenthixol.
[0130] Representative ACE-2 inhibitors include sulfhydryl-containing agents, such as alacepril, captopril (capoten), and zefnopril, dicarboxylate-containing agents, such as enalapril (vasotec), ramipril (altace), quinapril (accupril), perindopril (coversyl), lisinopril (listril), benazepril (lotensin), imidapril (tanatril), trandolapril (mavik), and cilazapril (inhibace), and phosphonate- containing agents, such as fosinopril (fositen/monopril).
[0131] Compounds which inhibit the cytokine storm include compounds that target fundamental immune pathways, such as the chemokine network and the cholinergic anti inflammatory pathway. For example, JAK inhibitors, such as JAK 1 and JAK 2 inhibitors, can inhibit the cytokine storm, and in some cases, are also antiviral. Representative JAK inhibitors include those disclosed in U.S. Pat. No. 10,022,378, such as Jakafi, Tofacitinib, and Baricitinib, as well as LY3009104/INCB28050, Pacritinib/SB1518, VX-509, GLPG0634, INC424, R-348, CYT387, TG 10138, AEG 3482, and pharmaceutically acceptable salts and prodrugs thereof. Still further examples include CEP-701 (Lestaurtinib), AZD1480, INC424, R-348, CYT387, TG 10138, AEG 3482, 7-iodo-N-(4-morpholinophenyl)thieno[3,2-d]pyrimidin-2-amine, 7-(4-aminophenyl)-N-(4- morpholinophenyl)thieno[3,2-d]pyrimidin-2-amine, N-(4-(2-(4-morpholinophenylamino)thieno[3,2- d]pyrimidin-7-yl) phenyl) acrylamide, 7-(3-aminophenyl)-N-(4-morpholinophenyl)thieno[3,2- d]pyrimidin-2-amine, N-(3-(2-(4-morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl) phenyl) acrylamide, N-(4-morpholinophenyl)thieno[3,2-d]pyrimidin-2-amine, methyl 2-(4- morpholinophenylamino)thieno[3,2-d]pyrimidine-7-carboxylate, N-(4-morpholinophenyl)-5H- pyrrolo[3,2-d]pyrimidin-2-amine, 7-(4-amino-3-methoxyphenyl)-N-(4- morpholinophenyl)thieno[3,2-d]pyrimidin-- 2-amine, 4-(2-(4-morpholinophenylamino)thieno[3,2- d]pyrimidin-7-yl)benzene- -sulfonamide, N,N-dimethyl-3-(2-(4- morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl)benzenesulfonamide, l-ethyl-3-(2-methoxy- 4-(2-(4-morpholinophenylamino)thieno[3,2-d]pyrimidin- -7-yl) phenyl) urea, N-(4-(2-(4- morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl)phenyl)methanesulfonamide, 2-methoxy-4- (2-(4-morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl)pheno- -I, 2-cyano-N-(3-(2-(4- morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl)phenyl)acetamide, N-(cyanomethyl)-2-(4- morpholinophenylamino)thieno[3,2-d]pyrimidine-7-carboxamide, N-(3-(2-(4- morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl)phen- yl)methanesulfonamide, l-ethyl-3-(4- (2-(4-morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl)-2-(- trifluoromethoxy) phenyl) urea, N- (3-nitrophenyl)-7-phenylthieno[3,2-d]pyrimidin-2-amine, 7-iodo-N-(3-nitrophenyl)thieno[3,2- d]pyrimidin-2-amine, Nl-(7-(2-ethylphenyl)thieno[3,2-d]pyrimidin-2-yl) benzene- 1,3-diamine, N- tert-butyl-3-(2-(4-morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl)benzenesulfonamide, Nl- (7-iodothieno[3,2-d]pyrimidin-2-yl) benzene- 1,3-diamine, 7-(4-amino-3-(trifluoromethoxy)phenyl)- N-(4-morpholinophenyl)thieno[3,2-d- ]pyrimidin-2-amine, 7-(2-ethylphenyl)-N-(4- morpholinophenyl)thieno[3,2-d]pyrimidin-2-amine, N-(3-(2-(4-morpholinophenylamino)thieno[3,2- d]pyrimidin-7-yl)phenyl)acetamide, N-(cyanomethyl)-N-(3-(2-(4- morpholinophenylamino)thieno[3,2-d]pyrim- idin-7-yl) phenyl) methanesulfonamide, N- (cyanomethyl)-N-(4-(2-(4-morpholinophenylamino)thieno[3,2-d]pyrimidin-7- - yl) phenyl) methanesulfonamide, N-(3-(5-methyl-2-(4-morpholinophenylamino)-5H-pyrrolo[3,2- d]pyrimidin-7-y- I) phenyl) methanesulfonamide, 4-(5-methyl-2-(4-morpholinophenylamino)-5H- pyrrolo[3,2-d]pyrimidin-7-yl)benzenesulfonamide, N-(4-(5-methyl-2-(4-morpholinophenylamino)- 5H-pyrrolo[3,2-d]pyrimidin-7-y- -1) phenyl) methanesulfonamide, 7-iodo-N-(4-morpholinophenyl)- 5H-pyrrolo[3, 2-d]pyri mid in-2-a mine, 7-(2-isopropylphenyl)-N-(4-morpholinophenyl)thieno[3,2- d]pyrimidin-2-amin- e, 7-bromo-N-(4-morpholinophenyl)thieno[3,2-d]pyrimidin-2-amine, N7-(2- isopropylphenyl)-N2-(4-morpholinophenyl)thieno[3,2-d] pyrimidine-2,7- -diamine, N7-(4- isopropylphenyl)-N2-(4-morpholinophenyl)thieno[3,2-d]pyrim- id ine-2, 7-diamine, 7-(5-amino-2- methylphenyl)-N-(4-morpholinophenyl)thieno[3,2-d]pyrimidin-2- -amine, N-(cyanomethyl)-4-(2- (4-morpholinophenylamino)thieno[3,2-d]pyrimid- in-7-yl)benzamide, 7-iodo-N-(3- morpholinophenyl)thieno[3,2-d]pyrimidin-2-amine, 7-(4-amino-3-nitrophenyl)-N-(4- morpholinophenyl)thieno[3,2-d]pyrimidin-2-- amine, 7-(2-methoxypyridin-3-yl)-N-(4- morpholinophenyl)thieno[3,2-d]pyrimi- din-2-amine, (3-(7-iodothieno[3,2-d]pyrimidin-2- ylamino) phenyl) methanol, N-tert-butyl-3-(2-(3-morpholinophenylamino)thieno[3,2-d]pyrimidin-7- yl)benzenesulfonamide, N-tert-butyl-3-(2-(3-(hydroxymethyl)phenylamino)thieno[3,2-d]pyrimidin- 7-- yl)benzenesulfonamide, N-(4-morpholinophenyl)-7-(4-nitrophenylthio)-5H-pyrrolo[3,2- d]pyrimidin-2- -amine, N-tert-butyl-3-(2-(3,4,5-trimethoxyphenylamino)thieno[3,2-d]pyrimi- -din- 7-yl)benzenesulfonamide, 7-(4-amino-3-nitrophenyl)-N-(3,4-dimethoxyphenyl)thieno[3,2- d]pyrimidin-2- -amine, N-(3,4-dimethoxyphenyl)-7-(2-methoxypyridin-3-yl)thieno[3,2-d]pyri- midin-2-amine, N-tert-butyl-3-(2-(3,4-dimethoxyphenylamino)thieno[3,2-d]pyrimidin-7-yl)b- enzenesulfonamide, 7-(2-aminopyrimidin-5-yl)-N-(3,4-dimethoxyphenyl)thieno[3,2-d]pyrimidin-2- -amine, N-(3,4-dimethoxyphenyl)-7-(2,6-dimethoxypyridin-3-yl)thieno[3,2-d]- -pyrimidine- amine, N-(3,4-dimethoxyphenyl)-7-(2,4-dimethoxypyrimidin-5-yl)thieno[3,2-d]pyrim- -idin-2- amine, 7-iodo-N-(4-(morpholinomethyl)phenyl)thieno[3,2-d]pyrimidin-2-amine, N-tert-butyl-3-(2- (4-(morpholinomethyl)phenylamino)thieno[3,2-d]pyrimidin- -7-yl)benzenesulfonamide, 2-cyano-N- (4-methyl-3-(2-(4-morpholinophenylamino)thieno[3,2-d]pyrimidin-- 7-yl)phenyl)acetamide, ethyl
3-(2-(4-morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl) benzoate, 7-bromo-N-(4-(2- (pyrrolidin-l-yl)ethoxy)phenyl)thieno[3,2-d]pyrimidin-2-a- mine, N-(3-(2-(4-(2-(pyrrolidin-l- yl)ethoxy)phenylamino)thieno[3,2-d]pyrim- idin-7-yl)phenyl)acetamide, N-(cyanomethyl)-3-(2-(4- morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl- )benzamide, N-tert-butyl-3-(2-(4- morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl)be- nzamide, N-tert-butyl-3-(2-(4-(l- ethylpiperidin-4-yloxy)phenylamino)thieno- -[3,2-d]pyrimidin-7-yl)benzenesulfonamide, tert-butyl-
4-(2-(4-(morpholinomethyl)phenylamino)thieno[3,2-d]pyrimidin-7- -yl)-lH-pyrazole-l- carboxylate, 7-bromo-N-(4-((4-ethylpiperazin-l-yl)methyl)phenyl)thieno[3,2-d]pyrimidin- -2- amine, N-tert-butyl-3-(2-(4-((4-ethylpiperazin-l-yl)methyl)phenylamino)- -thieno[3,2-d]pyrimidin- 7-yl)benzenesulfonamide, N-(4-((4-ethylpiperazin-l-yl)methyl)phenyl)-7-(lH-pyrazol-4- yl)thieno[3,2- -d]pyrimidin-2-amine, N-(cyanomethyl)-3-(2-(4-
(morpholinomethyl)phenylamino)thieno[3,2-d]pyrimi- -din-7-yl)benzamide, N-tert-butyl-3-(2-(4- (2-(pyrrolidin-l-yl)ethoxy)phenylamino)thieno[3,2-d]- -pyrimidin-7-yl)benzenesulfonamide, tert- butyl pyrrolidin-l-yl)ethoxy)phenylamino)thieno[3,2-d]pyrimidin-7-yl)benzylcarb- -a mate, 3-(2-(4- (2-(pyrrolidin-l-yl)ethoxy)phenylamino)thieno[3,2-d]pyrimi- din-7-yl)benzenesulfonamide, 7-(3- chloro-4-fluorophenyl)-N-(4-(2-(pyrrolidin-l-yl)ethoxy)phenyl)thieno- -[3,2-d]pyrimidin-2-amine, tert-butyl 4-(2-(4-(l-ethylpiperidin-4-yloxy)phenylamino)thieno[3,2-d]pyrimidin-7-yl- -)-lH- pyrazole-l-carboxylate, 7(benzo[d][l,3]dioxol-5-yl)-N-(4-(morpholinomethyl)phenyl)thieno[3,2- d]py- ri midi n-2-a mine, tert-butyl 5-(2-(4-(morpholinomethyl)phenylamino)thieno[3,2-d]pyrimidin- 7-yl)-lH-ind- ole-l-carboxylate, 7-(2-aminopyrimidin-5-yl)-N-(4- (morpholinomethyl)phenyl)thieno[3,2-d]pyri- midin-2-amine, tert-butyl 4-(2-(-4- (morpholinomethyl)phenylamino)thieno[3,2-d]pyrimidin-7-yl)-5,6-d- i-hydropyridine-l(2H)- carboxylate, tert-butyl morpholi nomethyl) phenyla mino)thieno[3, 2-d]pyri midi n-7- yl)benzylcarbamate, N-(3-(2-(4-(morpholinomethyl)phenylamino)thieno[3,2-d]pyrimidin-7- yl)phen- -yl)acetamide, N-(4-(2-(4-(morpholinomethyl)phenylamino)thieno[3,2-d]pyrimidin-7- yl)phen- -yl)acetamide, N-(3-(2-(4-(morpholinomethyl)phenylamino)thieno[3,2-d]pyrimidin-7- yl)phen- -yl)methanesulfonamide, 7-(4-(4-methylpiperazin-l-yl)phenyl)-N-(4- (morpholinomethyl)phenyl)thieno- -[3,2-d]pyrimidin-2-amine, N-(2-methoxy-4-(2-(4- (morpholinomethyl)phenylamino)thieno[3,2-d]pyrimidin- -7-yl)phenyl)acetamide, 7-bromo-N- (3,4,5-trimethoxyphenyl)thieno[3,2-d]pyrimidin-2-amine, (3-(2-(3,4,5- trimethoxyphenylamino)thieno[3,2-d]pyrimidin-7-yl) phenyl) met- -hanol, (4-(2-(3,4,5- trimethoxyphenylamino)thieno[3,2-d]pyrimidin-7-yl)phe- n-yl) methanol, (3-(2-(4- morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl)phenyl)methano- -1, (4-(2-(4- morpholinophenylamino)thieno[3,2-d]pyrimidin-7-yl)phenyl)meth- anol, N-(pyrrolidin-l- yl)ethoxy)phenylamino)thieno[3,2-d]pyrimidin-7-yl)be- nzyl)methanesulfonamide, tert-butyl morpholinomethyl)phenylamino)thieno[3,2-d]pyrimidin-7-yl)benzylcarbamate, N-(4- (morpholinomethyl)phenyl)-7-(3-(piperazin-l-yl)phenyl)thieno[3,2-d]p- yrimidin-2-amine, 7-(6-(2- morpholinoethylamino)pyridin-3-yl)-N-(3,4,5-trimethoxyphenyl)thie- no[3,2-d]pyrimidin-2-amine, 7-(2-ethylphenyl)-N-(4-(2-(pyrrolidin-l-yl)ethoxy)phenyl)thieno[3,2-d]pyr- imidin-2-amine, 7-(4- (a mi nomethyl) phenyl) -N-(4-(morpholinomethyl)phenyl)thieno[3,2-d]pyri- mid in-2-a mine, N-(4-(l- ethyl pi peridin-4-yloxy) phenyl) -7-(lH-pyrazol-4-yl)thieno[3,2-d]py- rimidin-2-amine, N-(2,4- dimethoxyphenyl)-7-phenylthieno[3,2-d]pyrimidin-2-amine, 7-bromo-N-(3,4- dimethoxyphenyl)thieno[3,2-d]pyrimidin-2-amine, N-(3,4-dimethoxyphenyl)-7-phenylthieno[3,2- d]pyrimidin-2-amine, and pharmaceutically acceptable salts and prodrugs thereof.
[0132] Alternatively, or in addition, HMGB1 antibodies and/or COX-2 inhibitors can be used, which downregulate the cytokine storm. Examples of such compounds include Actemra (Roche). Celebrex (celecoxib), a COX-2 inhibitor, can be used. IL-8 (CXCL8) inhibitors can also be used.
[0133] Chemokine receptor CCR2 antagonists, such as PF-04178903 can reduce pulmonary immune pathology.
[0134] Selective TLR7 antagonists can also be co-administered with compounds that inhibit blood clot formation, such as blood thinners, or compounds that break up existing blood clots, such as tissue plasminogen activator (TPA), Integrilin (eptifibatide), abciximab (ReoPro) or tirofiban (Aggrastat). Representative platelet aggregation inhibitors include glycoprotein IIB/IIIA inhibitors, phosphodiesterase inhibitors, adenosine reuptake inhibitors, and adenosine diphosphate (ADP) receptor inhibitors. These can optionally be administered in combination with an anticoagulant. Illustrative anti-coagulants include coumarins (vitamin K antagonists), heparin and derivatives thereof, including unfractionated heparin (UFH), low molecular weight heparin (LMWH), and ultra-low-molecular weight heparin (ULMWH), synthetic pentasaccharide inhibitors of factor Xa, including Fondaparinux, Idraparinux, and Idrabiotaparinux, directly acting oral anticoagulants (DAOCs), such as dabigatran, rivaroxaban, apixaban, edoxaban and betrixaban, and antithrombin protein therapeutics/thrombin inhibitors, such as bivalent drugs hirudin, lepirudin, and bivalirudin and monovalent argatroban.
[0135] Other examples of ancillary therapeutic agents include, without limitation, rose hip oil, vitamin E, 5-fluorouracil, bleomycin, onion extract, pentoxifylline, prolyl-4-hydroxylase, verapamil, tacrolimus, tamoxifen, tretinoin, colchicine, a calcium antagonist, tranilst, zinc, and a combination thereof.
[0136] In other embodiments, the selective TLR7 antagonists of the present disclosure are used in conjunction with an intervention, such as a ventilator.
[0137] Accordingly, the present disclosure encompasses co-administration of a selective TLR7 antagonist in concert with an ancillary therapeutic agent or intervention, as described for example above and elsewhere herein. It will be understood that, in embodiments comprising administration of the selective TLR7 antagonist with other agents, the dosages of the actives in the combination may on their own comprise an effective amount and the additional agent(s) may further augment the therapeutic benefit to the patient. Alternatively, the selective TLR7 antagonist and the additional agent(s) may together comprise an effective amount for treating an acute inflammatory condition. It will also be understood that effective amounts may be defined in the context of particular treatment regimens, including, e.g., timing and number of administrations, modes of administrations, formulations, etc. In some embodiments, the selective TLR7 antagonist and optionally the ancillary therapeutic agent are administered on a routine schedule. Alternatively, the ancillary therapeutic agent may be administered as symptoms arise. A "routine schedule" as used herein, refers to a predetermined designated period of time. The routine schedule may encompass periods of time which are identical, or which differ in length, as long as the schedule is predetermined. For instance, the routine schedule may involve administration of the selective TLR7 antagonist on a daily basis, every two days, every three days, every four days, every five days, every six days, a weekly basis, a monthly basis or any set number of days or weeks there between, every two months, three months, four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, twelve months, etc. Alternatively, the predetermined routine schedule may involve concurrent administration of the selective TLR7 antagonist and the ancillary therapeutic agent on a daily basis for the first week, followed by a monthly basis for several months, and then every three months after that. Any particular combination would be covered by the routine schedule as long as it is determined ahead of time that the appropriate schedule involves administration on a certain day. In specific embodiments, the selective TLR7 antagonist is administered in the form of a single dose and suitably only once during the course of treatment.
[0138] For any compound used in the treatment methods of the disclosure, the therapeutically effective dose can be estimated initially from cell culture assays. For example, a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture (e.g., the concentration of an active agent, which achieves a half-maximal inhibition in TLR7 function or activity). Such information can be used to determine useful doses more accurately in humans.
[0139] Dosing is dependent on severity and responsiveness of the inflammatory disease state to be treated, with the course of treatment lasting from several days to several months, or until a cure is achieved or a diminution of the disease state is achieved. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient. Persons of ordinary skill can easily determine optimum dosages, dosing methodologies and repetition rates. Optimum dosages may vary depending on the relative potency of individual selective TLR7 antagonists and can generally be estimated based on ECsos found to be effective in in vitro and in vivo animal models. In general, dosage is from 0.01 pg to 100 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly. Persons of ordinary skill in the art can easily estimate repetition rates for dosing based on measured residence times and concentrations of the drug in bodily fluids or tissues.
[0140] Toxicity and therapeutic efficacy of selective TLR7 antagonists can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds that exhibit large therapeutic indices are preferred. The data obtained from these cell culture assays, and animal studies can be used in formulating a range of dosage for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See for example Fingl et ai., 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 pi).
[0141] Pharmaceutical compositions of the present disclosure may be provided in a kit. The kit may comprise additional components to assist in performing the methods of the present disclosure such as, for example, administration device(s), buffer(s), and/or diluent(s). The kits may include containers for housing the various components and instructions for using the kit components in the methods of the present disclosure.
[0142] As disclosed herein, selective antagonism of TLR7 in subjects with active RNA virus infections is useful for correcting the host immune response with consequential improved immunity, including enhanced production of antibody, to the virus. Accordingly, the present disclosure contemplates co-administration of antigenic compositions, including immunogenic compositions and vaccines, with a selective TLR7 antagonist to a subject with an acute inflammatory condition associated with RNA virus infection.
[0143] Representative antigenic compositions encompassed by the present disclosure include but are not limited to killed or attenuated live viral vaccines, live-vectored vaccines, subunit vaccines, virus-like particle vaccines, and DNA or RNA vaccines. See Roth et al., New Technology For Improved Vaccine Safety And Efficacy", Veterinary Clinics North America: Food Animal Practice 17(3): 585-597 (2011). The RNA virus to which the antigenic composition elicits an immune response may be a positive sense RNA virus, negative sense RNA virus, or a double-stranded RNA virus. Representative RNA viruses include Picornaviruses (e.g., hepatitis A virus, enteroviruses such as poliovirus, enterovirus 71, 70, 69, and 68, Coxsackieviruses, echoviruses, foot and mouth disease virus, and rhinoviruses), Caliciviruses (e.g., hepatitis E virus, noroviruses such as Norwalk virus, feline calicivirus), Arteriviruses (e.g., equine arteritis virus), Togaviruses (e.g., sindbis virus, the equine encephalitis viruses, chikungunya virus, rubella virus, Ross River virus, bovine diarrhea virus, hog cholera virus, Semliki forest virus), Flaviviruses (e.g., dengue virus, West Nile virus, yellow fever virus, Japanese encephalitis virus, St. Louis encephalitis virus, tick-borne encephalitis virus, bovine viral diarrhea virus, classical swine fever virus), Coronaviruses (e.g., human coronaviruses, including betacoronavi ruses such OC43 and HKU1 of the A lineage, SARS-CoV and SARS-CoV-2 of the B lineage and MERS-CoV of the C lineage, swine gastroenteritis virus), Rhabdoviruses (e.g., rabies virus, Australian bat lyssavirus, vesicular stomatitis viruses),
Filoviruses (e.g., Marburg virus, Ebola virus), Paramyxoviruses (e.g., measles virus, canine distemper virus, mumps virus, parainfluenza viruses, respiratory syncytial virus, Newcastle disease virus, rinderpest virus, Nipah virus, Hendra virus), Orthomyxoviruses (e.g., human influenza viruses, including human influenza virus types A, B and C, avian influenza viruses, equine influenza viruses), Bunyaviruses (e.g., hantavirus, LaCrosse virus, Rift Valley fever virus), Arenaviruses (e.g., Lassa virus, Machupo virus), Reoviruses (e.g., human and animal reoviruses, such as rotaviruses, bluetongue virus), Birnaviruses (e.g., infectious bursal virus, fish pancreatic necrosis virus), Retroviruses (e.g., HIV 1, HIV 2, HTLV-1, HTLV-2, bovine leukemia virus, feline immunodeficiency virus, feline sarcoma virus, mouse mammary tumor virus), Hepadnaviruses (e.g., hepatitis B virus), Parvoviruses (e.g., B19 virus, canine parvovirus, feline panleukopenia virus), Papovaviruses (e.g., human papillomaviruses, SV40, bovine papillomaviruses),
Adenoviruses (e.g., human, canine, bovine, and porcine adenoviruses), Herpesviruses (e.g., herpes simplex viruses, varicella-zoster virus, infectious bovine rhinotracheitis virus, cytomegalovirus, human herpesvirus 6, human herpesvirus 7, human herpesvirus 8, Epstein-Barr virus), Poxviruses (e.g., vaccinia, fowlpoxviruses, raccoon poxvirus, skunkpox virus, monkeypoxvirus, cowpox virus, buffalopox virus, musculum contagiosum virus). 6. Therapeutic uses
[0144] The present disclosure also extends to the use of TLR7 antagonists, optionally in combination with at least one ancillary therapeutic agent, for treating a subject with an acute inflammatory condition, particularly one caused or exacerbated at least in part by activation of TLR7, and optionally one or both of TLR8 and TLR9, and/or an increased level of one or more oxidative stressors {e.g., NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1; also known as p47phox) and ROS) and/or an increased level of furin.
[0145] The acute inflammatory condition may be associated with presence of a pathogenic infection (e.g., a pathogenic infection in which the pathogen is an RNA virus), which is typically an active infection. The RNA virus may enter a host cell that it infects by receptor- mediated endocytosis or micropinocytosis. In specific embodiments, the RNA virus enters the endosome of a host cell that it infects. In representative examples, the RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenzavirus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus), Togaviridae (e.g., Chikungunya virus, Ross River virus, Semliki Forest virus), Flaviviridae (e.g., Zika virus, Dengue virus, Japanese encephalitis virus, West Nile virus, Yellow fever virus, Hepacivirus C) and Hantaviridae (e.g., Hantavirus). In certain embodiments, the RNA virus is an orthomyxovirus (e.g., an influenza virus such as influenza A, influenza B or influenza C) or a coronavirus (e.g., a coronavirus, including betacoronaviruses, capable of causing severe acute respiratory syndrome (SARS) such as MERS-CoV, SARS-CoV and SARS-CoV-2). Suitably, the acute inflammatory condition is associated with presence of cytokine release syndrome (CRS) or a cytokine storm, which in representative examples comprises an elevation of at least 50% compared to basal state of one or more (e.g., 1, 2, 3, 4, 5, 6, 7 or 8) cytokines selected from IFN- Y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2. In some embodiments, the acute inflammatory condition is associated with a condition selected from multisystem inflammatory syndrome in children (MIS-C), systemic inflammatory response syndrome (SIRS), acute respiratory distress syndrome (ARDS), and severe acute respiratory syndrome (SARS). The treatment of conditions associated directly or indirectly with viral infection is also contemplated herein including the treatment of cardiovascular conditions as well as the reduction, loss, modification or distortion of olfactory sensation.
[0146] In some embodiments, the acute inflammatory condition is associated with presence of CRS. In representative examples of this type, the subject has one or more symptoms selected from fever, fatigue, loss of appetite, muscle and joint pain, nausea, vomiting, diarrhea, rashes, fast breathing, rapid heartbeat, low blood pressure, seizures, headache, confusion, delirium, hallucinations, tremor, and loss of coordination, or any combination thereof.
[0147] The acute inflammatory condition may be associated with presence of a cytokine storm in which the subject in illustrative examples has one or more symptoms selected from high fever, swelling and redness, extreme fatigue, nausea, bleeding, clotting, internal organ injury, and shock, or any combination thereof. [0148] In some embodiments, the acute inflammatory condition is associated with presence of MIS-C, wherein the subject in non-limiting examples has one or more symptoms selected from fever, vomiting, diarrhea, stomach pain, skin rash, red eyes, redness or swelling of the lips and tongue, feeling unusually tired, redness or swelling of the hands or feet, severe stomach pain, cardiac symptoms, including chest pain, palpitations and shortness of breath, bluish lips or face, mental confusion, inability to wake up or stay awake, abdominal pain with vomiting and diarrhea, skin rash and swelling of extremities, faintness and low blood pressure that is new, or any combination thereof.
[0149] In other embodiments, the acute inflammatory condition is associated with SIRS. The subject may have one or more symptoms associated with a particular stage of SIRS, representative ones of which are as follows:
[0150] Stage 1 is a local reaction at the site of injury that aims at containing the injury and limit spread. Immune effector cells at the site release cytokines that in turn stimulate the reticuloendothelial system promoting wound repair through local inflammation. There is local vasodilatation induced by nitric oxide and prostacyclin (rubor) and disruption of the endothelial tight junction to allow margination and transfer of leucocytes into tissue space. The leakage of cells and protein-rich fluid in extravascular space causes swelling (tumor) and increased heat (calor). Inflammatory mediators impact the local somatosensory nerves causing pain (dolor) and loss of function (functio laesa). That loss of function also allows the part of the body to repair instead of persistent use.
[0151] Stage 2 is an early CARS in an attempt to maintain immunological balance.
There is a stimulation of growth factors and recruitment of macrophages and platelets as the level of pro-inflammatory mediators decreases to maintain homeostasis.
[0152] Stage 3 is when the scale tips over towards pro-inflammatory SIRS resulting in progressive endothelial dysfunction, coagulopathy, and activation of the coagulation pathway. It results in end-organ micro thrombosis, and a progressive increase in capillary permeability, eventually resulting in loss of circulatory integrity.
[0153] Stage 4 is characterized by CARS taking over SIRS, resulting in a state of relative immunosuppression. The individual, therefore, becomes susceptible to secondary or nosocomial infections, thus perpetuating the sepsis cascade.
[0154] Stage 5 manifests in MODS with persistent dysregulation of both SIRS and CARS response.
[0155] In other embodiments, the acute inflammatory condition is associated with ARDS. Representative symptoms of ARDS include one or more of mild, moderate or severe hypoxemia as determined by Partial Pressure of arterial oxygen/Fraction of inspired oxygen (Pa02/Fi02) or positive end-expiratory pressure (PEEP), bilateral opacities, respiratory failure, shortness of breath, labored breathing, cough, fever, increased heart rate, low blood pressure, confusion, extreme tiredness, rapid breathing, organ failure, chest pain, bluish coloring of nails or lips, an change in the level of one or more inflammatory markers, and need for mechanical ventilation, or any combination thereof.
[0156] In still other embodiments, the acute inflammatory condition is associated with SARS and in non-limiting examples the subject has one or more symptoms selected from acute febrile illness, malaise, fatigue, headache, flushing, diarrhea, nausea, vomiting, coughing including dry coughing, sore throat, runny nose, nasal congestion, production of pro-inflammatory mediators, vascular leakage and organ failure, or any combination thereof.
[0157] In order that the disclosure may be readily understood and put into practical effect, particular preferred embodiments will now be described by way of the following non-limiting examples.
7. Embodiments of the disclosure
[0158] The present disclosure includes and encompasses without limitation the following embodiments:
1. A method of treating an acute inflammatory condition in a subject, the method comprising administering to the subject an effective amount of a selective TLR7 antagonist.
2. The method of embodiment 1, wherein the acute inflammatory condition is caused or exacerbated by, or is otherwise associated with, activation of TLR7 and optionally one or both of TLR8 and TLR9.
3. The method of embodiment 1 or embodiment 2, wherein the acute inflammatory condition is caused or exacerbated by, or is otherwise associated with, an increased level of one or more oxidative stress mediators.
4. The method of embodiment 3, wherein the oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
5. The method of any one of embodiments 1 to 4, wherein the acute inflammatory condition is caused or exacerbated by, or is otherwise associated with, an increased level of furin.
6. The method of any one of embodiments 2 to 5, wherein the activation of TLR7 and optionally one or both of TLR8 and TLR9 is associated with presence of a pathogenic infection.
7. The method of embodiment 6, wherein the pathogen is an RNA virus.
8. The method of embodiment 7, wherein the RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
9. The method of embodiment 7 or embodiment 8, wherein the RNA virus is an enveloped virus.
10. The method of any one of embodiments 7 to 9, wherein the RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
11. The method of any one of embodiments 7 to 10, wherein the RNA virus enters the endosome of a host cell that it infects.
12. The method of any one of embodiments 7 to 11, wherein the RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenza virus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus), Togaviridae (e.g., Chikungunya virus, Ross River virus, Semliki Forest virus), Flaviviridae (e.g., Zika virus, Dengue virus, Japanese encephalitis virus, West
Nile virus, Yellow fever virus, Hepacivirus C) and Hantaviridae (e.g., Hantavirus). 13. The method of any one of embodiments 7 to 12, wherein the RNA virus is an orthomyxovirus.
14. The method of embodiment 13, wherein the orthomyxovirus is an influenza virus.
15. The method of embodiment 14, wherein the influenza virus is selected from influenza A, influenza B and influenza C.
16. The method of any one of embodiments 7 to 12, wherein the RNA virus is a coronavirus.
17. The method of embodiment 16, wherein the coronavirus is capable of causing severe acute respiratory syndrome (SARS).
18. The method of embodiment 16 or embodiment 17, wherein the coronavirus is a beta coronavirus.
19. The method of embodiment 18, wherein the betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
20. The method of embodiment 19, wherein the betacoronavirus is a lineage B betacoronavirus.
21. The method of embodiment 20, wherein the lineage B betacoronavirus is selected from SARS-CoV and SARS-CoV-2.
22. The method of embodiment 19, wherein the betacoronavirus is a lineage C betacoronavirus.
23. The method of embodiment 22, wherein the lineage C betacoronavirus is MERS-CoV.
24. The method of any one of embodiments 7 to 12, wherein the RNA virus is a picornavirus.
25. The method of embodiment 24, wherein the picornavirus is a rhinovirus.
26. The method of any one of embodiments 1 to 25, wherein the acute inflammatory condition is associated with presence of cytokine release syndrome (CRS) or a cytokine storm.
27. The method of embodiment 26, wherein the CRS or cytokine storm comprises an elevation of at least 50% compared to basal state of one or more cytokines selected from IFN-y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2.
28. The method of embodiment 26 or embodiment 27, wherein the subject has CRS and has one or more symptoms selected from fever, fatigue, loss of appetite, muscle and joint pain, nausea, vomiting, diarrhea, rashes, fast breathing, rapid heartbeat, low blood pressure, seizures, headache, confusion, delirium, hallucinations, tremor, and loss of coordination.
29. The method of embodiment 26 or embodiment 27, wherein the subject has a cytokine storm and has one or more symptoms selected from high fever, swelling and redness, extreme fatigue, nausea, bleeding, clotting, internal organ injury, and shock, or any combination thereof.
30. The method of any one of embodiments 1 to 29, wherein the acute inflammatory condition is associated with a multisystem inflammatory syndrome in children (MIS-C).
31. The method of embodiment 30, wherein the subject has one or more symptoms selected from fever, vomiting, diarrhea, stomach pain, skin rash, red eyes, redness or swelling of the lips and tongue, feeling unusually tired, redness or swelling of the hands or feet, emergency warning signs of MIS-C can include: severe stomach pain, cardiac symptoms, including chest pain, palpitations and shortness of breath, bluish lips or face, mental confusion, inability to wake up or stay awake, abdominal pain with vomiting and diarrhea, skin rash and swelling of extremities, faintness and low blood pressure that is new.
32. The method of any one of embodiments 1 to 31, wherein the acute inflammatory condition is associated with a systemic inflammatory response syndrome (SIRS).
33. The method of embodiment 32, wherein the subject has Stage 1 SIRS (local reaction).
34. The method of embodiment 32, wherein the subject has Stage 2 SIRS (early compensatory anti-inflammatory response syndrome (CARS) in an attempt to maintain immunological balance).
35. The method of embodiment 32, wherein the subject has Stage 3 SIRS (pro- inflammatory SIRS resulting in progressive endothelial dysfunction, coagulopathy, and activation of the coagulation pathway).
36. The method of embodiment 32, wherein the subject has Stage 4 SIRS (characterized by CARS taking over SIRS, resulting in a state of relative immunosuppression. The individual, therefore, becomes susceptible to secondary or nosocomial infections, thus perpetuating the sepsis cascade).
37. The method of embodiment 32, wherein the subject has Stage 5 SIRS (manifests in Multiple organ dysfunction syndrome (MODS) with persistent dysregulation of both SIRS and CARS response).
38. The method of any one of embodiments 1 to 31, wherein the acute inflammatory condition is associated with acute respiratory distress syndrome (ARDS).
39. The method of embodiment 38, wherein the subject has one or more symptoms selected from mild, moderate or severe hypoxemia as determined by Partial Pressure of arterial oxygen/Fraction of inspired oxygen (PaCh/FiCh) or positive end-expiratory pressure (PEEP), bilateral opacities, respiratory failure, shortness of breath, labored breathing, cough, fever, increased heart rate, low blood pressure, confusion, extreme tiredness, rapid breathing, organ failure, chest pain, bluish coloring of nails or lips, an change in the level of one or more inflammatory markers, or need for mechanical ventilation.
40. The method of any one of embodiments 1 to 31, wherein the acute inflammatory condition is associated with severe acute respiratory syndrome (SARS).
41. The method of embodiment 40, wherein the subject has one or more symptoms selected from acute febrile illness, malaise, fatigue, headache, flushing, diarrhea, nausea, vomiting, coughing including dry coughing, sore throat, runny nose, nasal congestion, production of pro-inflammatory mediators, vascular leakage and organ failure.
42. The method of any one of embodiments 1 to 41, wherein the selective TLR7 antagonist is selected from nucleic acid antagonists (e.g., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide) and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines).
43. The method of any one of embodiments 1 to 42, wherein the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7. 44. The method of embodiment 43, wherein the proteinaceous molecule is represented by Formula I:
Z1DX1RCNCX2PX3X4Z2 (I) wherein:
Zi and Z2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
Xi is a hydrophobic amino acid {e.g., L, F or M, or modified forms thereof);
X2 is a hydrophobic amino acid {e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
X3 is a hydrophobic amino acid {e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid {e.g., A or P, or modified forms thereof); and
X4 is small amino acid {e.g., P, or modified forms thereof), a hydrophobic amino acid {e.g., L, or modified forms thereof) or a basic amino acid {e.g., K or R, or modified forms thereof).
45. A method of treating cytokine release syndrome (CRS) or a cytokine storm in a subject, the method comprising administering to the subject an effective amount of a TLR7 antagonist.
46. The method of embodiment 45, wherein the CRS or cytokine storm is caused or exacerbated by, or is otherwise associated with, activation of TLR7 and optionally one or both of TLR8 and TLR9.
47. The method of embodiment 45 or embodiment 46, wherein the CRS or cytokine storm is caused or exacerbated by, or is otherwise associated with, an increased level of one or more oxidative stress mediators.
48. The method of embodiment 47, wherein the oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
49. The method of any one of embodiments 45 to 48, wherein the CRS or cytokine storm is caused or exacerbated by, or is otherwise associated with, an increased level of furin.
50. The method of any one of embodiments 46 to 49, wherein the activation of TLR7 and optionally one or both of TLR8 and TLR9 is associated with presence of a pathogenic infection.
51. The method of embodiment 50, wherein the pathogen is an RNA virus.
52. The method of embodiment 51, wherein the RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
53. The method of embodiment 46 or embodiment 47, wherein the RNA virus is an enveloped virus.
54. The method of any one of embodiments 51 to 53, wherein the RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
55. The method of any one of embodiments 51 to 54, wherein the RNA virus enters the endosome of a host cell that it infects. 56. The method of any one of embodiments 51 to 55, wherein the RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenza virus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus), Togaviridae (e.g., Chikungunya virus, Ross River virus, Semliki Forest virus), Flaviviridae (e.g., Zika virus, Dengue virus, Japanese encephalitis virus, West Nile virus, Yellow fever virus, Hepacivirus C) and Hantaviridae (e.g., Hantavirus).
57. The method of any one of embodiments 51 to 56, wherein the RNA virus is an orthomyxovirus.
58. The method of embodiment 57, wherein the orthomyxovirus is an influenza virus.
59. The method of embodiment 58, wherein the influenza virus is selected from influenza A, influenza B and influenza C.
60. The method of any one of embodiments 51 to 56, wherein the RNA virus is a coronavirus.
61. The method of embodiment 60, wherein the coronavirus is capable of causing severe acute respiratory syndrome (SARS).
62. The method of embodiment 60 or embodiment 61, wherein the coronavirus is a beta coronavirus.
63. The method of embodiment 62, wherein the betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
64. The method of embodiment 63, wherein the betacoronavirus is a lineage B betacoronavirus.
65. The method of embodiment 64, wherein the lineage B betacoronavirus is selected from SARS-CoV and SARS-CoV-2.
66. The method of embodiment 63, wherein the betacoronavirus is a lineage C betacoronavirus.
67. The method of embodiment 66, wherein the lineage C betacoronavirus is MERS-CoV.
68. The method of any one of embodiments 51 to 56, wherein the RNA virus is a picornavirus.
69. The method of embodiment 68, wherein the picornavirus is a rhinovirus.
70. The method of any one of embodiments 45 to 69, wherein the CRS or cytokine storm comprises an elevation of at least 50% compared to basal state of one or more cytokines selected from IFN-Y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2.
71. The method of any one of embodiment 45 to 70, wherein the subject has CRS and has one or more symptoms selected from fever, fatigue, loss of appetite, muscle and joint pain, nausea, vomiting, diarrhea, rashes, fast breathing, rapid heartbeat, low blood pressure, seizures, headache, confusion, delirium, hallucinations, tremor, and loss of coordination. 72. The method of any one of embodiment 45 to 70, wherein the subject has a cytokine storm and has one or more symptoms selected from high fever, swelling and redness, extreme fatigue, nausea, bleeding, clotting, internal organ injury, and shock, or any combination thereof.
73. The method of any one of embodiments 45 to 72, wherein the CRS or cytokine storm is associated with a MIS-C.
74. The method of embodiment 73, wherein the subject has one or more symptoms selected from fever, vomiting, diarrhea, stomach pain, skin rash, red eyes, redness or swelling of the lips and tongue, feeling unusually tired, redness or swelling of the hands or feet, emergency warning signs of multisystem inflammatory syndrome can include: severe stomach pain, cardiac symptoms, including chest pain, palpitations and shortness of breath, bluish lips or face, mental confusion, inability to wake up or stay awake, abdominal pain with vomiting and diarrhea, skin rash and swelling of extremities, faintness and low blood pressure that is new.
75. The method of any one of embodiments 45 to 72, wherein the CRS or cytokine storm is associated with a SIRS.
76. The method of embodiment 75, wherein the subject has Stage 1 SIRS (local reaction).
77. The method of embodiment 75, wherein the subject has Stage 2 SIRS (early CARS in an attempt to maintain immunological balance).
78. The method of embodiment 75, wherein the subject has Stage 3 SIRS (pro- inflammatory SIRS resulting in progressive endothelial dysfunction, coagulopathy, and activation of the coagulation pathway).
79. The method of embodiment 75, wherein the subject has Stage 4 SIRS (characterized by CARS taking over SIRS, resulting in a state of relative immunosuppression. The individual, therefore, becomes susceptible to secondary or nosocomial infections, thus perpetuating the sepsis cascade).
80. The method of embodiment 75, wherein the subject has Stage 5 SIRS (manifests in MODS with persistent dysregulation of both SIRS and CARS response).
81. The method of any one of embodiments 45 to 72, wherein the CRS or cytokine storm is associated with ARDS.
82. The method of embodiment 81, wherein the subject has one or more symptoms selected from mild, moderate or severe hypoxemia as determined by Partial Pressure of arterial oxygen/Fraction of inspired oxygen (PaCh/FiCh) or positive end-expiratory pressure (PEEP), bilateral opacities, respiratory failure, shortness of breath, labored breathing, cough, fever, increased heart rate, low blood pressure, confusion, extreme tiredness, rapid breathing, organ failure, chest pain, bluish coloring of nails or lips, an change in the level of one or more inflammatory markers, or need for mechanical ventilation.
83. The method of any one of embodiments 45 to 72, wherein the CRS or cytokine storm is associated with SARS.
84. The method of embodiment 83, wherein the subject has one or more symptoms selected from acute febrile illness, malaise, fatigue, headache, flushing, diarrhea, nausea, vomiting, coughing including dry coughing, sore throat, runny nose, nasal congestion, production of pro-inflammatory mediators, vascular leakage and organ failure. 85. The method of any one of embodiments 45 to 84, wherein the selective TLR7 antagonist is selected from nucleic acid antagonists {e.g., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists {e.g., decoy peptide) and small molecule antagonists {e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines).
86. The method of any one of embodiments 45 to 85, wherein the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7.
87. The method of embodiment 86, wherein the proteinaceous molecule is represented by Formula I:
Z1DX1RCNCX2PX3X4Z2 (I) wherein:
Zi and Z2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
Xi is a hydrophobic amino acid {e.g., L, F or M, or modified forms thereof);
X2 is a hydrophobic amino acid {e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
X3 is a hydrophobic amino acid {e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid {e.g., A or P, or modified forms thereof); and
X4 is small amino acid {e.g., P, or modified forms thereof), a hydrophobic amino acid {e.g., L, or modified forms thereof) or a basic amino acid {e.g., K or R, or modified forms thereof).
88. A method of treating SARS in a subject, the method comprising administering to the subject an effective amount of a TLR7 antagonist.
89. The method of embodiment 88, wherein the SARS is caused or exacerbated by, or is otherwise associated with, activation of TLR7 and optionally one or both of TLR8 and TLR9.
90. The method of embodiment 88 or embodiment 89, wherein the SARS is caused or exacerbated by, or is otherwise associated with, an increased level of one or more oxidative stress mediators.
91. The method of embodiment 90, wherein the oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
92. The method of any one of embodiments 88 to 91, wherein the SARS is caused or exacerbated by, or is otherwise associated with, an increased level of furin.
93. The method of any one of embodiments 89 to 92, wherein the activation of TLR7 and optionally one or both of TLR8 and TLR9 is associated with presence of a pathogenic infection.
94. The method of embodiment 93, wherein the pathogen is an RNA virus.
95. The method of embodiment 94, wherein the RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
96. The method of embodiment 94 or embodiment 95, wherein the RNA virus is an enveloped virus. 97. The method of any one of embodiments 94 to 96, wherein the RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
98. The method of any one of embodiments 94 to 97, wherein the RNA virus enters the endosome of a host cell that it infects.
99. The method of any one of embodiments 94 to 98, wherein the RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenzavirus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus), Togaviridae (e.g., Chikungunya virus, Ross River virus, Semliki Forest virus), Flaviviridae (e.g., Zika virus, Dengue virus, Japanese encephalitis virus, West Nile virus, Yellow fever virus, Hepacivirus C) and Hantaviridae (e.g., Hantavirus).
100. The method of any one of embodiments 94 to 99, wherein the RNA virus is an orthomyxovirus.
101. The method of embodiment 100, wherein the orthomyxovirus is an influenza virus.
102. The method of embodiment 101, wherein the influenza virus is selected from influenza A, influenza B and influenza C.
103. The method of any one of embodiments 94 to 99, wherein the RNA virus is a coronavirus.
104. The method of embodiment 103, wherein the coronavirus is a betacoronavirus.
105. The method of embodiment 104, wherein the betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
106. The method of embodiment 105, wherein the betacoronavirus is a lineage B betacoronavirus.
107. The method of embodiment 106, wherein the lineage B betacoronavirus is selected from SARS-CoV and SARS-CoV-2.
108. The method of embodiment 105, wherein the betacoronavirus is a lineage C betacoronavirus.
109. The method of embodiment 108, wherein the lineage C betacoronavirus is MERS-CoV.
110. The method of any one of embodiments 88 to 109, wherein the subject has one or more symptoms selected from acute febrile illness, malaise, fatigue, headache, flushing, diarrhea, nausea, vomiting, coughing including dry coughing, sore throat, runny nose, nasal congestion, production of pro-inflammatory mediators, vascular leakage and organ failure, or any combination thereof.
111. The method of any one of embodiments 88 to 110, wherein the SARS is associated with presence of CRS or a cytokine storm.
112. The method of embodiment 111, wherein the CRS or cytokine storm comprises an elevation of at least 50% compared to basal state of one or more cytokines selected from IFN-y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2. 113. The method of any one of embodiments 88 to 112, wherein the SARS is associated with a MISC-C.
114. The method of embodiment 113, wherein the subject has one or more symptoms selected from fever, vomiting, diarrhea, stomach pain, skin rash, red eyes, redness or swelling of the lips and tongue, feeling unusually tired, redness or swelling of the hands or feet, emergency warning signs of multisystem inflammatory syndrome can include: severe stomach pain, cardiac symptoms, including chest pain, palpitations and shortness of breath, bluish lips or face, mental confusion, inability to wake up or stay awake, abdominal pain with vomiting and diarrhea, skin rash and swelling of extremities, faintness and low blood pressure that is new.
115. The method of any one of embodiments 88 to 114, wherein the SARS is associated with a SIRS.
116. The method of embodiment 115, wherein the subject has Stage 1 SIRS (local reaction).
117. The method of embodiment 115, wherein the subject has Stage 2 SIRS (early CARS in an attempt to maintain immunological balance).
118. The method of embodiment 115, wherein the subject has Stage 3 SIRS (pro- inflammatory SIRS resulting in progressive endothelial dysfunction, coagulopathy, and activation of the coagulation pathway).
119. The method of embodiment 115, wherein the subject has Stage 4 SIRS (characterized by CARS taking over SIRS, resulting in a state of relative immunosuppression. The individual, therefore, becomes susceptible to secondary or nosocomial infections, thus perpetuating the sepsis cascade).
120. The method of embodiment 115, wherein the subject has Stage 5 SIRS (manifests in MODS with persistent dysregulation of both SIRS and CARS response).
121. The method of any one of embodiments 88 to 120, wherein the selective TLR7 antagonist is selected from nucleic acid antagonists (e.g., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide) and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines).
122. The method of any one of embodiments 88 to 121, wherein the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7.
123. The method of embodiment 109, wherein the proteinaceous molecule is represented by Formula I:
Z1DX1RCNCX2PX3X4Z2 (I) wherein:
Zi and Z2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
Xi is a hydrophobic amino acid (e.g., L, F or M, or modified forms thereof);
X2 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof); X3 is a hydrophobic amino acid ( e.g ., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid {e.g., A or P, or modified forms thereof); and
X4 is small amino acid {e.g., P, or modified forms thereof), a hydrophobic amino acid {e.g., L, or modified forms thereof) or a basic amino acid {e.g., K or R, or modified forms thereof).
124. A method of treating ARDS in a subject, the method comprising administering to the subject an effective amount of a TLR7 antagonist.
125. The method of embodiment 124, wherein the ARDS is caused or exacerbated by, or is otherwise associated with, activation of TLR7 and optionally one or both of TLR8 and TLR9.
126. The method of embodiment 124 or embodiment 125, wherein the ARDS is caused or exacerbated by, or is otherwise associated with, an increased level of one or more oxidative stress mediators.
127. The method of embodiment 126, wherein the oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
128. The method of any one of embodiments 126 to 127, wherein the ARDS is caused or exacerbated by, or is otherwise associated with, an increased level of furin.
129. The method of any one of embodiments 125 to 128, wherein the activation of TLR7 and optionally one or both of TLR8 and TLR9 is associated with presence of a pathogenic infection.
130. The method of embodiment 129, wherein the pathogen is an RNA virus.
131. The method of embodiment 130, wherein the RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
132. The method of embodiment 130 or embodiment 131, wherein the RNA virus is an enveloped virus.
133. The method of any one of embodiments 130 to 132, wherein the RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
134. The method of any one of embodiments 130 to 133, wherein the RNA virus enters the endosome of a host cell that it infects.
135. The method of any one of embodiments 130 to 134, wherein the RNA virus is selected from the following families: Orthomyxoviridae {e.g., Influenzavirus), Coronaviridae {e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus {e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae {e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae {e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae {e.g., Human metapneumovirus), Rhabdoviridae {e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus), Togaviridae {e.g., Chikungunya virus, Ross River virus, Semliki Forest virus), Flaviviridae {e.g., Zika virus, Dengue virus, Japanese encephalitis virus, West Nile virus, Yellow fever virus, Hepacivirus C) and Hantaviridae {e.g., Hantavirus).
136. The method of any one of embodiments 130 to 135, wherein the RNA virus is an orthomyxovirus.
137. The method of embodiment 136, wherein the orthomyxovirus is an influenza virus. 138. The method of embodiment 137, wherein the influenza virus is selected from influenza A, influenza B and influenza C.
139. The method of any one of embodiments 130 to 135, wherein the RNA virus is a coronavirus.
140. The method of embodiment 139, wherein the coronavirus is capable of causing SARS.
141. The method of embodiment 139 or embodiment 140, wherein the coronavirus is a beta coronavirus.
142. The method of embodiment 141, wherein the betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
143. The method of embodiment 142, wherein the betacoronavirus is a lineage B betacoronavirus.
144. The method of embodiment 143, wherein the lineage B betacoronavirus is selected from SARS-CoV and SARS-CoV-2.
145. The method of embodiment 142, wherein the betacoronavirus is a lineage C betacoronavirus.
146. The method of embodiment 145, wherein the lineage C betacoronavirus is MERS-CoV.
147. The method of any one of embodiments 130 to 135, wherein the RNA virus is a picornavirus.
148. The method of embodiment 147, wherein the picornavirus is a rhinovirus.
149. The method of any one of embodiments 124 to 148, wherein the ARDS is associated with presence of CRS or a cytokine storm.
150. The method of embodiment 149, wherein the CRS or cytokine storm comprises an elevation of at least 50% compared to basal state of one or more cytokines selected from IFN-y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2.
151. The method of any one of embodiments 124 to 150, wherein the subject has one or more symptoms selected from mild, moderate or severe hypoxemia as determined by Partial Pressure of arterial oxygen/Fraction of inspired oxygen (PaCh/FiCh) or positive end-expiratory pressure (PEEP), bilateral opacities, respiratory failure, shortness of breath, labored breathing, cough, fever, increased heart rate, low blood pressure, confusion, extreme tiredness, rapid breathing, organ failure, chest pain, bluish coloring of nails or lips, an change in the level of one or more inflammatory markers, or need for mechanical ventilation.
152. The method of any one of embodiments 124 to 151, wherein the selective TLR7 antagonist is selected from nucleic acid antagonists (e.g., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide), and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines).
153. The method of any one of embodiments 124 to 152, wherein the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7.
154. The method of embodiment 153, wherein the proteinaceous molecule is represented by Formula I: Z1DX1RCNCX2PX3X4Z2 (I) wherein:
Zi and Z2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
Xi is a hydrophobic amino acid {e.g., L, F or M, or modified forms thereof);
X2 is a hydrophobic amino acid {e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
X3 is a hydrophobic amino acid {e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid {e.g., A or P, or modified forms thereof); and
X4 is small amino acid {e.g., P, or modified forms thereof), a hydrophobic amino acid {e.g., L, or modified forms thereof) or a basic amino acid {e.g., K or R, or modified forms thereof).
155. A method of treating SIRS in a subject, the method comprising administering to the subject an effective amount of a selective TLR7 antagonist.
156. The method of embodiment 155, wherein the SIRS is caused or exacerbated by, or is otherwise associated with, activation of TLR7 and optionally one or both of TLR8 and TLR9.
157. The method of embodiment 155 or embodiment 156, wherein the SIRS is caused or exacerbated by, or is otherwise associated with, an increased level of one or more oxidative stress mediators.
158. The method of embodiment 157, wherein the oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
159. The method of any one of embodiments 155 to 158, wherein the SIRS is caused or exacerbated by, or is otherwise associated with, an increased level of furin.
160. The method of any one of embodiments 155 to 159, wherein the activation of TLR7 and optionally one or both of TLR8 and TLR9 is associated with presence of a pathogenic infection.
161. The method of embodiment 160, wherein the pathogen is an RNA virus.
162. The method of embodiment 161, wherein the RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
163. The method of embodiment 161 or embodiment 162, wherein the RNA virus is an enveloped virus.
164. The method of any one of embodiments 161 to 163, wherein the RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
165. The method of any one of embodiments 161 to 164, wherein the RNA virus enters the endosome of a host cell that it infects.
166. The method of any one of embodiments 161 to 165, wherein the RNA virus is selected from the following families: Orthomyxoviridae {e.g., Influenzavirus), Coronaviridae {e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus {e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae {e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae {e.g., Human parainfluenza virus,
Nipah henipavirus, Hendra henipavirus), Pneumoviridae {e.g., Human metapneumovirus), Rhabdoviridae ( e.g ., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus), Togaviridae (e.g., Chikungunya virus, Ross River virus, Semliki Forest virus), Flaviviridae (e.g., Zika virus, Dengue virus, Japanese encephalitis virus, West Nile virus, Yellow fever virus, Hepacivirus C) and Hantaviridae (e.g., Hantavirus).
167. The method of any one of embodiments 161 to 166 wherein the RNA virus is an orthomyxovirus.
168. The method of embodiment 167, wherein the orthomyxovirus is an influenza virus.
169. The method of embodiment 168, wherein the influenza virus is selected from influenza A, influenza B and influenza C.
170. The method of any one of embodiments 161 to 166, wherein the RNA virus is a coronavirus.
171. The method of embodiment 170, wherein the coronavirus is capable of causing SARS.
172. The method of embodiment 170 or embodiment 171, wherein the coronavirus is a beta coronavirus.
173. The method of embodiment 172, wherein the betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
174. The method of embodiment 152, wherein the betacoronavirus is a lineage B betacoronavirus.
175. The method of embodiment 153, wherein the lineage B betacoronavirus is selected from SARS-CoV and SARS-CoV-2.
176. The method of embodiment 152, wherein the betacoronavirus is a lineage C betacoronavirus.
177. The method of embodiment 155, wherein the lineage C betacoronavirus is MERS-CoV.
178. The method of any one of embodiments 161 to 166, wherein the RNA virus is a picornavirus.
179. The method of embodiment 178, wherein the picornavirus is a rhinovirus.
180. The method of any one of embodiments 155 to 179, wherein the SIRS is associated with presence of CRS or a cytokine storm.
181. The method of embodiment 180, wherein the CRS or cytokine storm comprises an elevation of at least 50% compared to basal state of one or more cytokines selected from IFN-y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2.
182. The method of any one of embodiments 155 to 181, wherein the subject has Stage 1 SIRS (local reaction).
183. The method of any one of embodiments 155 to 181, wherein the subject has Stage 2 SIRS (early CARS in an attempt to maintain immunological balance).
184. The method of any one of embodiments 155 to 181, wherein the subject has Stage 3 SIRS (pro-inflammatory SIRS resulting in progressive endothelial dysfunction, coagulopathy, and activation of the coagulation pathway).
185. The method of any one of embodiments 155 to 181, wherein the subject has Stage 4 SIRS (characterized by CARS taking over SIRS, resulting in a state of relative immunosuppression. The individual, therefore, becomes susceptible to secondary or nosocomial infections, thus perpetuating the sepsis cascade).
186. The method of any one of embodiments 155 to 181, wherein the subject has Stage 5 SIRS (manifests in MODS with persistent dysregulation of both SIRS and CARS response).
187. The method of any one of embodiments 155 to 186, wherein the selective TLR7 antagonist is selected from nucleic acid antagonists (e.g., aptamers, ribozymes and triplex forming molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide) and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxa lines).
188. The method of any one of embodiments 155 to 187, wherein the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7.
189. The method of embodiment 188, wherein the proteinaceous molecule is represented by Formula I:
Z1DX1RCNCX2PX3X4Z2 (I) wherein:
Zi and Z2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
Xi is a hydrophobic amino acid (e.g., L, F or M, or modified forms thereof);
X2 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
X3 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid (e.g., A or P, or modified forms thereof); and
X4 is small amino acid (e.g., P, or modified forms thereof), a hydrophobic amino acid (e.g., L, or modified forms thereof) or a basic amino acid (e.g., K or R, or modified forms thereof).
190. A method of treating a MIS-C in a subject, the method comprising administering to the subject an effective amount of a selective TLR7 antagonist.
191. The method of embodiment 190, wherein the MIS-C is caused or exacerbated by, or is otherwise associated with, activation of TLR7 and optionally one or both of TLR8 and TLR9.
192. The method of embodiment 190 or embodiment 191, wherein the MIS-C is caused or exacerbated by, or is otherwise associated with, an increased level of one or more oxidative stress mediators.
193. The method of embodiment 192, wherein the oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
194. The method of any one of embodiments 190 to 193, wherein the MIS-C is caused or exacerbated by, or is otherwise associated with, an increased level of furin.
195. The method of any one of embodiments 190 to 194, wherein the activation of TLR7 and optionally one or both of TLR8 and TLR9 is associated with presence of a pathogenic infection.
196. The method of embodiment 195, wherein the pathogen is an RNA virus. 197. The method of embodiment 196, wherein the RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
198. The method of embodiment 196 or embodiment 197, wherein the RNA virus is an enveloped virus.
199. The method of any one of embodiments 196 to 198, wherein the RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
200. The method of any one of embodiments 196 to 199, wherein the RNA virus enters the endosome of a host cell that it infects.
201. The method of any one of embodiments 196 to 200, wherein the RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenzavirus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus), Togaviridae (e.g., Chikungunya virus, Ross River virus, Semliki Forest virus), Flaviviridae (e.g., Zika virus, Dengue virus, Japanese encephalitis virus, West Nile virus, Yellow fever virus, Hepacivirus C) and Hantaviridae (e.g., Hantavirus).
202. The method of any one of embodiments 196 to 201 wherein the RNA virus is an orthomyxovirus.
203. The method of embodiment 202, wherein the orthomyxovirus is an influenza virus.
204. The method of embodiment 203, wherein the influenza virus is selected from influenza A, influenza B and influenza C.
205. The method of any one of embodiments 196 to 201, wherein the RNA virus is a coronavirus.
206. The method of embodiment 205, wherein the coronavirus is capable of causing SARS.
207. The method of embodiment 205 or embodiment 206, wherein the coronavirus is a beta coronavirus.
208. The method of embodiment 207, wherein the betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
209. The method of embodiment 208, wherein the betacoronavirus is a lineage B betacoronavirus.
210. The method of embodiment 209, wherein the lineage B betacoronavirus is selected from SARS-CoV and SARS-CoV-2.
211. The method of embodiment 208, wherein the betacoronavirus is a lineage C betacoronavirus.
212. The method of embodiment 211, wherein the lineage C betacoronavirus is MERS-CoV.
213. The method of any one of embodiments 196 to 201, wherein the RNA virus is a picornavirus.
214. The method of embodiment 213, wherein the picornavirus is a rhinovirus. 215. The method of any one of embodiments 190 to 214, wherein the MIS-C is associated with presence of CRS or a cytokine storm.
216. The method of embodiment 215, wherein the CRS or cytokine storm comprises an elevation of at least 50% compared to basal state of one or more cytokines selected from IFN-y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2.
217. The method of any one of embodiments 190 to 216, wherein the subject has one or more symptoms selected from fever, vomiting, diarrhea, stomach pain, skin rash, red eyes, redness or swelling of the lips and tongue, feeling unusually tired, redness or swelling of the hands or feet, emergency warning signs of multisystem inflammatory syndrome can include: severe stomach pain, cardiac symptoms, including chest pain, palpitations and shortness of breath, bluish lips or face, mental confusion, inability to wake up or stay awake, abdominal pain with vomiting and diarrhea, skin rash and swelling of extremities, faintness and low blood pressure that is new, or any combination thereof.
218. The method of any one of embodiments 190 to 217, wherein the selective TLR7 antagonist is selected from nucleic acid antagonists ( e.g ., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide) and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxalines).
219. The method of any one of embodiments 109 to 218, wherein the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7.
220. The method of embodiment 219, wherein the proteinaceous molecule is represented by Formula I:
Z1DX1RCNCX2PX3X4Z2 (I) wherein:
Zi and Z2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety;
Xi is a hydrophobic amino acid (e.g., L, F or M, or modified forms thereof);
X2 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
X3 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid (e.g., A or P, or modified forms thereof); and
X4 is small amino acid (e.g., P, or modified forms thereof), a hydrophobic amino acid (e.g., L, or modified forms thereof) or a basic amino acid (e.g., K or R, or modified forms thereof).
221. The method of any one of embodiments 1 to 220, wherein the selective TLR7 antagonist is concurrently administered with at least one ancillary therapeutic agent for treating the acute inflammatory condition.
222. The method of embodiment 221, wherein the ancillary agent is selected from is an anti-inflammatory agent, an analgesic agent, an antimicrobial agent, an agent that inhibits CRS or cytokine storm, an anti-coagulant, a platelet aggregation inhibitor, an agent that chelates iron ions released from hemoglobin by viruses, a cytochrome P-450 (CYP450) inhibitor and a NOX inhibitor.
223. The method of embodiment 221, wherein the ancillary agent an antigenic composition that stimulates or enhances the production of an immune response in the subject to a pathogen.
224. The method of any one of embodiments 1 to 223, wherein the subject is administered a single dose of the selective TLR7 antagonist during the course of the treatment.
225. A selective TLR7 antagonist for use in treating an acute inflammatory condition.
226. A selective TLR7 antagonist for use in treating CRS or a cytokine storm.
227. A selective TLR7 antagonist for use in treating MIS-C.
228. A selective TLR7 antagonist for use in treating SIRS.
229. A selective TLR7 antagonist for use in treating ARDS.
230. A selective TLR7 antagonist for use in treating SARS.
231. A selective TLR7 antagonist and at least one ancillary therapeutic agent for use in treating an acute inflammatory condition.
232. A selective TLR7 antagonist and an antigenic composition for use in stimulating or enhancing the production of an immune response in a subject to a pathogen.
EXAMPLES
EXAMPLE 1
TLR7 ANTAGONIST TREATMENT REDUCES MORBIDITY, INFLAMMATION AND VIRAL BURDEN FOLLOWING POST HIGHLY PATHOGENIC INFLUENZA A VIRUS INFECTION IN MICE
[0159] To address the potential therapeutic effect of C98i, mice were infected with highly pathogenic influenza A virus (IAV) at either 10 or 50PFUs/mouse at a point termed here as Day 0. Mice bodyweights were monitored daily from initial point of infection with IAV. Mice that were treated with drug vehicle (i.e. PBS) from Day 2 post-infection and at Day 3 and Day 4 post infection, displayed a substantial loss in bodyweight over a 7 day period (Figure 1). In contrast, mice that were treated with C98i (0.2mg/kg/day) lost significantly less of their bodyweight due to IAV (Figure 1). Mice that remained uninfected that were either treated with PBS or C98i (0.2/mg/kg/day) maintained a steady bodyweight over the 7-day period (Figure 1).
[0160] Compared to PBS treated mice, mice that were treated with C98i (0.2/mg/kg/day) had significantly less bronchoalveolar lavage fluid (BALF) inflammation (Figure 2); and significantly less lung viral mRNA levels (Figure 3).
[0161] The present inventors next examined whether C98i treatment suppressed lung inflammation to IAV infection. Mice infected with IAV displayed a significant elevation in peribronchial inflammation and alveolitis which was significantly inhibited by C98i treatment (Figure 4a-c). C98i treatment also significantly reduced the increase in lung weight that was caused by IAV infection (Figure 5). There were no significant alterations in heart, kidney, spleen or liver weight by C98i treatment (Figure 5). EXAMPLE 2
A SINGLE DOSE OF TLR7 ANTAGONIST EITHER4 OR5 DAYS POST IAV INFECTION INHIBITS MORBIDITY,
VIRAL TITER, CYTOKINE STORM AND INFLAMMATION
[0162] To examine the therapeutic effect of C98i, mice were infected with highly pathogenic influenza A virus (IAV) at 50 PFUs/mouse at a point termed here as Day 0 and at Day 4 post infection were either treated with a single dose of PBS vehicle or a single dose of C98i (2mg/kg/day intranasally). Mice bodyweights were monitored daily from initial point of infection with IAV. Mice that were treated with PBS vehicle displayed a significant loss of weight by Day 7 post IAV infection (Figure 6a). In contrast, mice that were treated with C98i (2.0mg/kg/day) lost significantly less of their bodyweight due to IAV (Figure 6a). There was an ~60% improvement in weight loss in mice treated with C98i.
[0163] Compared to PBS treated mice, mice that were treated with C98i (2mg/kg/day) had significantly less bronchoalveolar lavage fluid (BALF) inflammation (Figure 6b); significantly less lung viral mRNA levels (Figure 6c) and significantly reduced lung weight (Figure 6d). Mice groups that were not infected with IAV displayed no alteration in bodyweight, BALF inflammation, and lung weight following a single dose of C98i treatment (Figure 6a-c).
[0164] A study was then carried out to investigate whether C98i treatment suppressed macrophage, neutrophil, lymphocyte and eosinophil inflammation to IAV infection. Mice infected with IAV that were treated only with PBS vehicle displayed a significant elevation in airway macrophages, neutrophils, lymphocytes and eosinophils (Figure 7). However, compared to PBS treated mice, mice that were treated with C98i (2mg/kg/day) had significantly less macrophage, neutrophil and lymphocyte airway recruitment and a strong trend in eosinophil reductions (Figure 7).
[0165] The present inventors also examined whether C98i treatment suppressed lung inflammation to IAV infection and it was found that mice infected with IAV displayed a significant elevation in peribronchial inflammation and alveolitis which was significantly inhibited by C98i treatment (Figure 8a-c). Mice groups that were not infected with IAV displayed no alteration in alveolitis and peribronchial inflammatory cells following a single dose of C98i treatment (Figure 8a- c).
[0166] An investigation was also conducted to determine whether C98i treatment suppressed the lung cytokine storm to IAV infection. Notably, IAV infection resulted in a significant elevation in lung mRNA expression of interleukin lb (IL-lb), interferon-b (IFN-b), tumour necrosis factor-a (TNF-a), CXCL2, CCL3, IFN-gamma (IFN-y), interleukin-6 (IL-6) and IL-17A in mice that were treated with PBS vehicle (Figure 9). However, mice that were treated with C98i at Day 4 post infection displayed a significantly blunted cytokine response in the lungs (Figure 9). Uninfected mice treated with C98i displayed no significant alteration in lung cytokine levels (Figure 9).
[0167] A study was also undertaken to examine whether C98i treatment modifies furin, ACE2, angiotensin ATA1 and AT2 receptor expression following IAV infection. C98i treatment of mice infected with IAV had a reduced level of lung furin mRNA expression (Figure 10) compared to mice treated with PBS. ACE2, ATI and AT2 receptor expression decreased with IAV infection in mice that were treated with PBS vehicle (Figure 10). In contrast, mice treated with C98i had a significantly higher level of ACE2 and ATI receptor expression (Figure 10). Uninfected mice treated with C98i displayed no significant alteration in furin, ACE2, ATI or AT2 expression (Figure 10). [0168] C98i treatment was also examined to determine whether it could suppress the pattern recognition receptors TLR7, TLR8 and TLR9 expression and expression of oxidative genes NOX2 and p47phox to IAV infection. IAV infection resulted in a significant elevation in lung mRNA expression of TLR7, TLR8, TLR9, p47phox expression in mice treated with PBS vehicle (Figure 11). However, mice that were treated with C98i at Day 4 post IAV infection displayed a significantly reduced TLR7, TLR9, NOX2 and p47phox response in the lungs (Figure 11). Uninfected mice treated with C98i displayed no significant alteration in TLR7, TLR8, TLR9, NOX2 or p47phox expression (Figure 11).
[0169] To further examine the therapeutic effect of C98i, mice were infected with highly pathogenic influenza A virus (IAV) at 50 PFUs/mouse at a point termed here as Day 0, and at Day 5 post infection were either treated with a single dose of PBS vehicle (intranasally) or a single dose of C98i (2mg/kg/day intranasally).
[0170] Mice bodyweights were monitored daily from initial point of infection with IAV. Mice that were treated with PBS vehicle displayed a significant loss of weight by Day 7 post IAV infection (Figure 12A). In contrast, mice that were treated with C98i (2mg/kg/day) had an ~70% improvement in the bodyweight loss due to IAV (Figure 12).
[0171] Compared to PBS treated mice, mice that were treated with C98i (2mg/kg/day) had significantly less bronchoalveolar lavage fluid (BALF) inflammation (Figure 12B), lung viral mRNA levels (Figure 12C) and lung weight (Figure 12D).
[0172] It was examined whether C98i treatment suppressed macrophage, neutrophil, lymphocyte and eosinophil inflammation to IAV infection. Mice infected with IAV that were treated only with PBS vehicle displayed a significant elevation in airway macrophages, neutrophils, lymphocytes and eosinophils (Figure 13). However, compared to PBS treated mice, mice that were treated with C98i (2mg/kg/day) had significantly less macrophage, neutrophil and lymphocyte airway recruitment and a strong trend in eosinophil reductions (Figure 13). It was examined whether C98i treatment suppressed lung inflammation to IAV infection. Mice infected with IAV displayed a significant elevation in peribronchial inflammation and alveolitis which was significantly inhibited by C98i treatment (Figure 14).
[0173] Next, the present inventors examined whether C98i treatment suppressed the lung cytokine storm to IAV infection. Mice that were treated with C98i at Day 5 post infection displayed a significantly blunted cytokine response in the lungs (Figure 15), as compared to mice treated with the PBS vehicle (Figure 15).
[0174] A study was also carried out to determine whether C98i treatment modifies furin, ACE2, angiotensin ATA1 and AT2 receptor expression following IAV infection. C98i treatment of mice infected with IAV had a reduced level of lung furin mRNA expression (Figure 16), as compared to mice treated with PBS. Mice treated with C98i had a significantly higher level of ACE2 and ATI receptor expression (Figure 16).
[0175] An investigation was also undertaken to assess whether C98i treatment suppressed the pattern recognition receptors TLR7, TLR8 and TLR9 expression and expression of oxidative genes NOX2 and p47phox to IAV infection. Mice that were treated with C98i at Day 5 post IAV infection displayed a lower level of TLR7, TLR9 and NOX2 in the lungs (Figure 17) compared to mice treated with PBS vehicle (Figure 17). EXAMPLE 3
A SINGLE DOSE OF TLR7 ANTAGONIST 4HR POST RVIB INFECTION INHIBITS PRO-INFLAMMATORY
CYTOKINE PRODUCTION AND INFLAMMATION
[0176] To examine the therapeutic effect of C98i, mice were infected with a high inoculum (1010 PFUs/mouse) of rhinovirus IB strain (RVIB) at a point termed here as Day 0 and at 4h post infection were either treated with a single dose of PBS vehicle or a single dose of C98i (2mg/kg/day intranasally). Mice bodyweights were taken from initial point of infection with RVIB and when mice were culled at Day 1 post infection. For all study groups, mice did not gain or lose weight over the 24h period (Figure 18).
[0177] A study was then carried out to investigate whether C98i treatment suppressed BALF inflammation to RVIB infection. Compared to PBS treated mice, mice that were infected with RVIB displayed a significant BALF inflammatory response (Figure 19). Mice that were treated with C98i (2mg/kg/day) had significantly less BALF inflammation (Figure 19).
[0178] A study was then performed to investigate whether C98i (2mg/kg/day) treatment modified the viral mRNA load in the lungs. Compared to PBS treated mice, mice that were treated with C98i had a small but significant increase in lung viral mRNA levels (Figure 20).
[0179] A study was then carried out to investigate whether C98i treatment prevented the increase in lung weight to the RVIB infection. RVIB infection resulted in a significant increase in lung weight in the mice (Figure 4). Compared to PBS treated mice, mice that were treated with C98i (2mg/kg/day) displayed no significant increase in lung weight (Figure 21).
[0180] A study was then carried out to investigate whether C98i treatment (2mg/kg/day) suppressed macrophage, neutrophil, lymphocyte and eosinophil inflammation to RVIB infection. Mice infected with RVIB that were treated only with PBS vehicle displayed a significant elevation in airway macrophages and neutrophils (Figure 5). However, compared to PBS treated mice, mice that were treated with C98i had significantly less neutrophils and a strong trend in eosinophil reductions (Figure 22).
[0181] An investigation was also conducted to determine whether C98i treatment (2mg/kg/day) suppressed lung pro-inflammatory cytokine expression. Infected mice that were treated with C98i displayed a significantly blunted interleukin lb (IL-lb), tumor necrosis factor-a (TNF-a), CXCL2, CCL3 and interleukin-6 (IL-6) expression. Uninfected mice treated with C98i also displayed a significant decreased expression in the lung of IL-lb, TNF-a, CCL, CXLC2 and IL-6 mRNA levels (Figure 23).
Materials and methods
Viruses
[0182] The influenza A virus, Puerto Rican-8 (PR8) strain was provided by School of Medicine, Deakin University, Victoria and The Peter Doherty Institute for Infection and Immunity (Melbourne Australia). The virus was provided in PBS (Cat # D8537, Sigma-Aldrich, USA) and stored at -80 °C until used. On the day of use, virus is thawed quickly and incubated at 37 °C prior to infection. Animal ethics
[0183] The mouse experiments described in this manuscript will be approved by the Animal Experimentation Ethics Committee of RMIT University and conducted in compliance with the guidelines of the National Health and Medical Research Council (NHMRC) of Australia on animal experimentation.
Mouse Housing
[0184] C57BL/6 mice will be obtained from Animal Research Centre (Perth, Australia)
14 days from placing a request to the RMIT Animal Facility (RAF). All animals will be housed in a PC2 facility with ad libitum access to filtered air, water, and standard rodent chow (4.8% fat and 0.02% cholesterol). Animals will be housed in groups of 2-6 under RAF standard animal conditions with sterilized IVC cages, bedding and environment enrichment (e.g. tissues, cardboard rolls). All pre-protocol mice will be monitored daily by RAF staff and allowed to acclimatize for at least 1 day.
Mouse Infection: In vivo influenza virus infection models and treatment regimens
[0185] Eight-to-twelve-week-old C57BL6 male mice were anaesthetized and infected intranasally with the mouse-adapted IAV strain at doses: PR8 (50 PFUs/mouse) or PBS (mock infection). For these therapeutic studies, mice were randomly treated with vehicle or treated with a single dose of C98i (2.0mg/kg/day via intranasal delivery) at either 4 or 5 days post IAV infection. Mice were then culled at Day 6 or 7 post infection for the comprehensive analysis including: morbidity (body weight), inflammation, lung pathology and components of the innate and adaptive immune response (see below for details).
[0186] In a separate series of experiments, eight-to-twelve-week-old C57BL6 male mice were anaesthetized and infected intranasally with the mouse-adapted IAV strain at doses:
PR8 (10 or 50 PFUs/mouse) or PBS (mock infection). For these therapeutic studies, mice were randomly treated with vehicle or treated with a dose of C98i (0.2mg/kg/day via intranasal delivery) daily for 3 days beginning at Day 2 post infection. Mice were then culled at Day 6 or 7 post infection for the comprehensive analysis including: morbidity (body weight), inflammation, lung pathology and components of the innate and adaptive immune response (see below for details).
Monitoring Mice
[0187] Mice were monitored daily for signs of illness such as loss of body weight, lethargy, ruffled fur, hunched appearance, panting, loss of appetite, isolation from the group and loss of grooming. Weights were recorded daily.
Airway Bronchoalveolar Lavaae Fluid (BALF) Inflammation
[0188] Mice were killed by an intraperitoneal (i.p.) injection of ketamine/xylazine (20 mg/kg) mixture. An incision was made from the lower jaw to the top of the rib cage, where the salivary glands were separated to expose the surface of the trachea. The layer of smooth muscle on the trachea was removed, allowing a small incision to be made near the top of the trachea. A sheathed 21-Gauge needle was inserted to the lumen and 300-400 pi of PBS was lavaged repeatedly (four times). The total number of cells in the BALF were stained with ethidium bromide (cat # 15585011, Invitrogen, USA) and acridine orange (cat # A3568, Invitrogen, USA) and viable cells were counted with a hemocytometer. Differential Cell Analysis
[0189] This analysis was prepared from BALF (5 x 104 cells) that were centrifuged at 112.9 x g for 5 min on the Cytospin 3 (Shandon, UK). Following this, slides were fixed in 100% propanol for 1 min and allowed to dry overnight. Finally, samples were stained with Rapid I Aqueous Red Stain™ (cat # RS1-1L, Australian BioStain, Australia) and Rapid II Blue Stain™ (cat # RS11-L, Australian BioStain, Australia) for 10 min, then submerged in 70% ethanol and absolute ethanol twice before being placed into histolene for 5 min (two times). Samples were then mounted in DPX neutral mounting medium (cat # AJA3197-500ML, ThermoFisher Scientific, USA) and coverslips were firmly placed on top. 500 cells per sample from random fields were differentiated into macrophages, neutrophils, eosinophils and lymphocytes by standard morphological criteria. Data were represented as total cell numbers that were calculated by the respective cell type multiplied by the total live cell numbers and as a percentage of the cell population.
Innate and Adaptive Immune Response: Flow Cvtometrv and Tetramer Staining
[0190] Whole lung and mediastinal lymph nodes were removed and then finely minced with scissors and enzymatically digested using liberase (cat # 5401127001, Sigma-Aldrich, USA). Single cell suspensions were prepared from homogenized tissue straining through a 40 mM mesh strainer (cat # 542040, Greiner Bio-One, Austria). The red blood cells were lysed with lysis buffer (ACK lysis buffer; NH4CI 155 mM, NaHC03 12 mM, EDTA 0.1 mM), and the WBC stained with respective fluorescent-labelled anti-mouse antibodies for flow cytometric analysis of: Leukocytes (CD45; 30-F11); Dendritic cells (MHCII; M5/114.15.2 )(CDllc; N418 )(CDllb; Ml-70 )(CD103; 2E7); T cells (CD3 (145-2C11), CD8 (53-6.7), CD4 (RM4-5)), activation markers for T cells (CD69; H1.2F3), B cells (B220; RA3-6B2), Macrophages (F4/80; BM8) and Neutrophils (Ly6G; 1A8). All antibodies are from Biolegend, USA; BD Pharmigen, USA; BD Biosciences, USA or eBioscience,
USA.
[0191] The following gating strategy was used to quantify the overall numbers of macrophages (CD45+CDllb+F4/80+), neutrophils (CDllb+Ly6g + MHC Class II) and monocytes (CD45+CDllb+Ly6C+Ly6G-). Resident (MHCII+CDllc+CD8+CDllb+) and migratory (MHCII+CD11C+B220-CD103+/ MHCII+CDllc+B220-CDllb+) dendritic cell populations in the lungs and mediastinal lymph nodes were determined as a measure of cross-presentation to antigen-cognate na'ive CD8+ T cells. Additionally, the subpopulations of CD4+ and CD8+ T cells were assessed. For CD8+ T cells, their activation was assessed by measuring cell surface activation markers CD69+, as well as cytokine staining (TNF-a and IFN-g). Tetramer staining of virus-specific CD8+ T cells was performed using the peptides DbNP366 (ASNENMETM) and DbPA224 (SSLENFRAYV) that were synthesized at Biomolecular Resource Facility, Australian National University, Australia.
In addition to this, trafficking of virus-specific CD8+ T cells (CD8+PA224+/ CD8+NP 66+) in the mediastinal lymph nodes and lung tissue was measured. Within each antibody cocktail mixture, cells were incubated with CD16/32 (2.4G2) to block Fc-mediated adherence of the antibodies. Cell viability was determined by LIVE/DEAD Fixable Violet Dead Cell Stain Kit (cat # L34955,
Invitrogen, USA). Countbright counting beads (cat # C36950, Invitrogen, USA), were added to each sample and a minimum of 10000 beads were collected to determine the cell number on the LSRFortessa X-20 with the FACs DIVA software (Becton Dickinson, USA). Absolute numbers were calculated according to the countbright counting beads manufacturers guidelines. Data were analyzed using FlowJo software (Tree Star, Inc.). H&E Staining: Examining the Histopatholoaical Changes in Luna Inflammation
[0192] The left lung was dissected from mice and immersed in neutral buffered formalin (10%) for 24-48 hours. After fixation, the lung tissue was processed, embedded in paraffin wax, and longitudinal 4pm sections cut and stained with hematoxylin and eosin (H8iE). Slides were scanned by light microscopy and uploaded to Aperio microscope scanner (Leica Biosystems, Nussloch, Germany). Histology was performed by the Department of Histology (Monash University, Clayton, Australia) and analyzed blindly by two independent assessors by team at RMIT, UniSA and TCD. Five random fields from each lung section were analyzed for alveolitis, which is inflammation within the alveolar space. This is determined by the regularity and branching of the alveoli and the density of cells within the alveolar spaces. Peribronchiolar inflammation was characterized by the infiltration of inflammatory cells into the alveolar wall around the bronchioles. The degree of inflammatory cellular infiltrate was taken by observing the density of cells throughout the entire lung section. A score of 0 is indicative of healthy lungs (i.e. no damage); 1-very mild damage; 2- mild damage; 3-moderate damage, 4-severe damage and 5-extremely severe histological changes.
Quantification of Cvtokine and Oxidative Storm Gene expression of mRNA bv OPCR
[0193] Lungs were harvested from terminally anesthetized mice and tissue crushed into fine powder and total RNA extracted using a RNeasy mini kit (cat # 74106, QIAGEN, Germany). Synthesis of cDNA was performed using the High-Capacity cDNA RT kit (cat # 4368813, Applied Biosystems, USA) using 1.0-3.0 pg total RNA. Quantitative polymerase chain reaction was carried out using the TaqMan Fast advanced Master Mix (cat # 4444965, Applied Biosystems, USA) or PowerUp SYBR Green PCR Master Mix (cat # A25777, Applied Biosystems, USA) and analyzed on the Quant Studio 7 Flex Real-Time PCR system (Applied Biosystems). The PCR primers for the following genes - pro-inflammatory cytokines (IFN-b, IL-Ib, TNF-a, IL-6, IL-17 and IFN-y); chemokines (CCL2, CCL5, CXCL2, CXCL10); TLRs (TLR2, TLR4, TLR7, TLR9); Oxidative and antioxidant genes (NOX2, NOX1, NOX4, CYBA, NCF1, NCF2, SOD1, SOD2, SOD3, GPX1, CAT, PRDX5, NOS1, NOS2, NOS3); renin angiotensin system (ACE1, ACE2, AGTRla, AGTR2); glycolytic genes (HEX1, HEX2, SLC2A1, SLC2A4); endosomal genes (EEA1, APPL1, RAB5A, RAB7, SORT1, SDC1) and serine protease (FURIN) was included in the Assay-on-Demand Gene Expression Assay Mix (Applied Biosystems, USA). Additionally, a custom designed forward and reverse primer of the segment 3 polymerase (PA) of IAV was used to measure viral mRNA levels (Note this is in addition to the 'Plaque assay'). The PCR program run settings were: 50°C for 2 min, followed by 95°C for 1 hr, then 95°C for 15 s + 60°C for 60 s + plate read (40 cycles). For fast advanced master mix, the program settings were: 50°C for 2 minutes, 95°C for 2 minutes, 95°C for 1 second, 60°C for 20 seconds + plate read (40 cycles). Quantitative values were obtained from the threshold cycle (Ct) number. Target gene expression was normalized against glyceraldehyde 3-phosphate dehydrogenase (GAPDH; cat # 4352339E, Applied Biosystems, USA) mRNA expression for each sample and data were expressed relative to the naive control group.
Cvtokine and Oxidative Storm - Luna analysis: ELISA and Multiplex Immunoassay
[0194] Protein levels of cytokines (IFN-b, IL-Ib, TNF-a, IL-6, and IFN-g) and chemokines (CCL2, CCL5, CXCL2, CXCL10) secreted into the BALF and plasma were measured using ELISAs and performed using commercially available kits according to the manufacturer's instructions (R8iD systems, USA). The cytokine titers in samples were determined by plotting the optical densities using a 4-parameter fit for the standard curve. If any other of the genes measured by QPCR (as performed according to the method detailed above) were modified by IAV infection and C98i treatment, the protein analysis will occur via ELISA.
Statistical Analysis [0195] All statistical tests were performed using GraphPad Prism (GraphPad Software
Version 7.0, San Diego CA, USA). P < 0.05 was taken to indicate significance. Data were represented a the mean - standard error of the mean ( SEM ). Cytokine mRNA expression and antibody titers were analyzed using one - way ANOVA followed by Tukey's post hoc test for multiple comparisons. All tests were performed by GraphPad Prism 7.0b ( San Diego , Calif. USA ) and statistical significance was taken at P < 0.05.
[0196] The disclosure of every patent, patent application, and publication cited herein is hereby incorporated herein by reference in its entirety.
[0197] The citation of any reference herein should not be construed as an admission that such reference is available as "Prior Art" to the instant application.
[0198] Throughout the specification the aim has been to describe the preferred embodiments of the invention without limiting the invention to any one embodiment or specific collection of features. Those of skill in the art will therefore appreciate that, in light of the instant disclosure, various modifications and changes can be made in the particular embodiments exemplified without departing from the scope of the present invention. All such modifications and changes are intended to be included within the scope of the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A method of treating an acute inflammatory condition in a subject, the method comprising administering to the subject an effective amount of a selective TLR7 antagonist.
2. The method of claim 1, wherein the acute inflammatory condition is caused or exacerbated by, or is otherwise associated with, activation of TLR7 and optionally one or both of TLR8 and TLR9.
3. The method of claim 1 or claim 2, wherein the acute inflammatory condition is caused or exacerbated by, or is otherwise associated with, an increased level of one or more oxidative stress mediators.
4. The method of claim 3, wherein the oxidative stress mediators are selected from NADPH oxidase type 2 (NOX2) and neutrophil cytosol factor 1 (NCF1).
5. The method of any one of claims 1 to 4, wherein the acute inflammatory condition is caused or exacerbated by, or is otherwise associated with, an increased level of furin.
6. The method of any one of claims 2 to 5, wherein the activation of TLR7 and optionally one or both of TLR8 and TLR9 is associated with presence of a pathogenic infection.
7. The method of claim 6, wherein the pathogen is an RNA virus.
8. The method of claim 7, wherein the RNA virus is selected from flaviviruses, alphaviruses, togaviruses, coronaviruses, orthomyxoviruses, paramyxoviruses, rhabdoviruses, bunyaviruses, filoviruses, retroviruses, picornaviruses and caliciviruses.
9. The method of claim 7 or claim 8, wherein the RNA virus is an enveloped virus.
10. The method of any one of claims 7 to 9, wherein the RNA virus enters a host cell that it infects by receptor-mediated endocytosis or micropinocytosis.
11. The method of any one of claims 7 to 10, wherein the RNA virus enters the endosome of a host cell that it infects.
12. The method of any one of claims 7 to 11, wherein the RNA virus is selected from the following families: Orthomyxoviridae (e.g., Influenza virus), Coronaviridae (e.g., Middle East respiratory syndrome coronavirus (MERS-CoV), Severe acute respiratory syndrome coronavirus (e.g., SARS-CoV and SARS-CoV-2)), Picornaviridae (e.g., Rhinovirus, Poliovirus, Enterovirus, Coxsackievirus and Hepacivirus A), Paramyxoviridae (e.g., Human parainfluenza virus, Nipah henipavirus, Hendra henipavirus), Pneumoviridae (e.g., Human metapneumovirus), Rhabdoviridae (e.g., Rabies virus, Australian bat lyssavirus and Vesicular stomatitis virus), Filoviridae (Marburg virus, Ebola virus), Togaviridae (e.g., Chikungunya virus, Ross River virus, Semliki Forest virus), Flaviviridae (e.g., Zika virus, Dengue virus, Japanese encephalitis virus, West Nile virus, Yellow fever virus, Hepacivirus C) and Hantaviridae (e.g., Hantavirus).
13. The method of any one of claims 7 to 12, wherein the RNA virus is an orthomyxovirus.
14. The method of claim 13, wherein the orthomyxovirus is an influenza virus.
15. The method of claim 14, wherein the influenza virus is selected from influenza A, influenza B and influenza C.
16. The method of any one of claims 7 to 12, wherein the RNA virus is a coronavirus.
17. The method of claim 16, wherein the coronavirus is capable of causing severe acute respiratory syndrome (SARS).
18. The method of claim 16 or claim 17, wherein the coronavirus is a beta coronavirus.
19. The method of claim 18, wherein the betacoronavirus is selected from a lineage betacoronavirus A, a lineage B betacoronavirus, a lineage C betacoronavirus and a lineage D betacoronavirus.
20. The method of claim 19, wherein the betacoronavirus is a lineage B betacoronavirus.
21. The method of claim 20, wherein the lineage B betacoronavirus is selected from SARS-CoV and SARS-CoV-2.
22. The method of claim 19, wherein the betacoronavirus is a lineage C betacoronavirus.
23. The method of claim 22, wherein the lineage C betacoronavirus is MERS-CoV.
24. The method of any one of claims 7 to 12, wherein the RNA virus is a picornavirus.
25. The method of claim 24, wherein the picornavirus is a rhinovirus.
26. The method of any one of claims 1 to 25, wherein the acute inflammatory condition is associated with presence of cytokine release syndrome (CRS) or a cytokine storm.
27. The method of claim 26, wherein the CRS or cytokine storm comprises an elevation of at least 50% compared to basal state of one or more cytokines selected from IFN-Y, IFN-b, TNF-a, IL-Ib, IL-6, IL-17A, CCL3 and CXCL2.
28. The method of claim 26 or claim 27, wherein the subject has CRS and has one or more symptoms selected from fever, fatigue, loss of appetite, muscle and joint pain, nausea, vomiting, diarrhea, rashes, fast breathing, rapid heartbeat, low blood pressure, seizures, headache, confusion, delirium, hallucinations, tremor, and loss of coordination.
29. The method of claim 26 or claim 27, wherein the subject has a cytokine storm and has one or more symptoms selected from high fever, swelling and redness, extreme fatigue, nausea, bleeding, clotting, internal organ injury, and shock, or any combination thereof.
30. The method of any one of claims 1 to 29, wherein the acute inflammatory condition is associated with a multisystem inflammatory syndrome in children (MIS-C).
31. The method of claim 30, wherein the subject has one or more symptoms selected from fever, vomiting, diarrhea, stomach pain, skin rash, red eyes, redness or swelling of the lips and tongue, feeling unusually tired, redness or swelling of the hands or feet, emergency warning signs of MIS-C can include: severe stomach pain, cardiac symptoms, including chest pain, palpitations and shortness of breath, bluish lips or face, mental confusion, inability to wake up or stay awake, abdominal pain with vomiting and diarrhea, skin rash and swelling of extremities, faintness and low blood pressure that is new.
32. The method of any one of claims 1 to 31, wherein the acute inflammatory condition is associated with a systemic inflammatory response syndrome (SIRS).
33. The method of claim 32, wherein the subject has Stage 1 SIRS (local reaction).
34. The method of claim 32, wherein the subject has Stage 2 SIRS (early compensatory anti-inflammatory response syndrome (CARS) in an attempt to maintain immunological balance).
35. The method of claim 32, wherein the subject has Stage 3 SIRS (pro- inflammatory SIRS resulting in progressive endothelial dysfunction, coagulopathy, and activation of the coagulation pathway).
36. The method of claim 32, wherein the subject has Stage 4 SIRS (characterized by CARS taking over SIRS, resulting in a state of relative immunosuppression. The individual, therefore, becomes susceptible to secondary or nosocomial infections, thus perpetuating the sepsis cascade).
37. The method of claim 32, wherein the subject has Stage 5 SIRS (manifests in Multiple organ dysfunction syndrome (MODS) with persistent dysregulation of both SIRS and CARS response).
38. The method of any one of claims 1 to 31, wherein the acute inflammatory condition is associated with acute respiratory distress syndrome (ARDS).
39. The method of claim 38, wherein the subject has one or more symptoms selected from mild, moderate or severe hypoxemia as determined by Partial Pressure of arterial oxygen/Fraction of inspired oxygen (PaCh/FiC^) or positive end-expiratory pressure (PEEP), bilateral opacities, respiratory failure, shortness of breath, labored breathing, cough, fever, increased heart rate, low blood pressure, confusion, extreme tiredness, rapid breathing, organ failure, chest pain, bluish coloring of nails or lips, an change in the level of one or more inflammatory markers, or need for mechanical ventilation.
40. The method of any one of claims 1 to 31, wherein the acute inflammatory condition is associated with severe acute respiratory syndrome (SARS).
41. The method of claim 40, wherein the subject has one or more symptoms selected from acute febrile illness, malaise, fatigue, headache, flushing, diarrhea, nausea, vomiting, coughing including dry coughing, sore throat, runny nose, nasal congestion, production of pro-inflammatory mediators, vascular leakage and organ failure.
42. The method of any one of claims 1 to 41, wherein the selective TLR7 antagonist is selected from nucleic acid antagonists (e.g., antisense molecules, RNAi and external guide sequences), proteinaceous antagonists (e.g., decoy peptide) and small molecule antagonists (e.g., imidazo[l,2-a]pyrazines, imidazo[l,5-a]quinoxalines, and pyrazolo[l,5-a]quinoxa lines).
43. The method of any one of claims 1 to 42, wherein the selective TLR7 antagonist is a proteinaceous molecule comprising, consisting or consisting essentially of an amino acid sequence corresponding to the sequence defined by residues 95 to 104 of TLR7 with the proviso that the peptide comprises a cysteine residue at the equivalent of position 98 of TLR7.
44. The method of claim 43, wherein the proteinaceous molecule is represented by Formula I: Z1DX1RCNCX2PX3X4Z2 (I) wherein:
Zi and Z2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integer residues in between), and a protecting moiety; Xi is a hydrophobic amino acid (e.g., L, F or M, or modified forms thereof);
X2 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof);
X3 is a hydrophobic amino acid (e.g., an aliphatic amino acid such as V or I, or modified forms thereof) or a small amino acid (e.g., A or P, or modified forms thereof); and
X4 is small amino acid (e.g., P, or modified forms thereof), a hydrophobic amino acid (e.g., L, or modified forms thereof) or a basic amino acid (e.g., K or R, or modified forms thereof).
45. The method of any one of claims 1 to 44, wherein the selective TLR7 antagonist is concurrently administered with at least one ancillary therapeutic agent for treating the acute inflammatory condition.
46. The method of claim 45, wherein the ancillary agent is selected from is an anti inflammatory agent, an analgesic agent, an antimicrobial agent, an agent that inhibits CRS or cytokine storm, an anti-coagulant, a platelet aggregation inhibitor, an agent that chelates iron ions released from hemoglobin by viruses, a cytochrome P-450 (CYP450) inhibitor and a NOX inhibitor.
47. The method of claim 45, wherein the ancillary agent an antigenic composition that stimulates or enhances the production of an immune response in the subject to a pathogen.
48. The method of any one of claims 1 to 47, wherein the subject is administered a single dose of the selective TLR7 antagonist during the course of the treatment.
49. A selective TLR7 antagonist for use in treating an acute inflammatory condition.
50. A selective TLR7 antagonist for use in treating CRS or a cytokine storm.
51. A selective TLR7 antagonist for use in treating MIS-C.
52. A selective TLR7 antagonist for use in treating SIRS.
53. A selective TLR7 antagonist for use in treating ARDS.
54. A selective TLR7 antagonist for use in treating SARS.
55. A selective TLR7 antagonist and at least one ancillary therapeutic agent for use in treating an acute inflammatory condition.
56. A selective TLR7 antagonist and an antigenic composition for use in stimulating or enhancing the production of an immune response in a subject to a pathogen.
PCT/AU2022/050089 2021-02-12 2022-02-11 Agents and methods for therapy and prophylaxis WO2022170396A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
AU2021900345 2021-02-12
AU2021900345A AU2021900345A0 (en) 2021-02-12 Agents and methods for therapy and prophylaxis
AU2021903239 2021-10-08
AU2021903239A AU2021903239A0 (en) 2021-10-08 Agents and methods for therapy and prophylaxis

Publications (1)

Publication Number Publication Date
WO2022170396A1 true WO2022170396A1 (en) 2022-08-18

Family

ID=82838874

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2022/050089 WO2022170396A1 (en) 2021-02-12 2022-02-11 Agents and methods for therapy and prophylaxis

Country Status (1)

Country Link
WO (1) WO2022170396A1 (en)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010080509A1 (en) * 2008-12-19 2010-07-15 Philadelphia Health And Education Corporation Compositions and methods for diminishing viral infection and inflammation associated with viral infection
US20140179780A1 (en) * 2011-04-29 2014-06-26 Korea Research Institute Of Bioscience And Biotechnology Pharmaceutical composition containing oleanolic acid acetate as an active ingredient for preventing or treating tlr- or il-6-mediated diseases
WO2016134422A1 (en) * 2015-02-26 2016-09-01 Prince Henry's Institute Of Medical Research Trading As The Hudson Institute Of Medical Research A method of treatment
US20170335325A1 (en) * 2012-11-29 2017-11-23 Sbi Biotech Co., Ltd. Inhibitory oligonucleotide and use thereof
WO2018157107A1 (en) * 2017-02-27 2018-08-30 National Jewish Health Methods and compositions for treating and preventing respiratory related diseases and conditions with xylitol-headgroup lipid analogs
WO2019000045A1 (en) * 2017-06-30 2019-01-03 The Provost, Fellows, Foundation Scholars, And The Other Members Of Board, Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth, Near Dublin A method of treatment
WO2021213946A1 (en) * 2020-04-19 2021-10-28 Englmeier Ludwig Prophylaxis and treatment of coronavirus infection
WO2021231941A1 (en) * 2020-05-14 2021-11-18 Merck Healthcare Kgaa Tlr7/8 antagonists for the treatment of coronavirus infections

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010080509A1 (en) * 2008-12-19 2010-07-15 Philadelphia Health And Education Corporation Compositions and methods for diminishing viral infection and inflammation associated with viral infection
US20140179780A1 (en) * 2011-04-29 2014-06-26 Korea Research Institute Of Bioscience And Biotechnology Pharmaceutical composition containing oleanolic acid acetate as an active ingredient for preventing or treating tlr- or il-6-mediated diseases
US20170335325A1 (en) * 2012-11-29 2017-11-23 Sbi Biotech Co., Ltd. Inhibitory oligonucleotide and use thereof
WO2016134422A1 (en) * 2015-02-26 2016-09-01 Prince Henry's Institute Of Medical Research Trading As The Hudson Institute Of Medical Research A method of treatment
WO2018157107A1 (en) * 2017-02-27 2018-08-30 National Jewish Health Methods and compositions for treating and preventing respiratory related diseases and conditions with xylitol-headgroup lipid analogs
WO2019000045A1 (en) * 2017-06-30 2019-01-03 The Provost, Fellows, Foundation Scholars, And The Other Members Of Board, Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth, Near Dublin A method of treatment
WO2021213946A1 (en) * 2020-04-19 2021-10-28 Englmeier Ludwig Prophylaxis and treatment of coronavirus infection
WO2021231941A1 (en) * 2020-05-14 2021-11-18 Merck Healthcare Kgaa Tlr7/8 antagonists for the treatment of coronavirus infections

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
RAPPE JULIE C.F., FINSTERBUSCH KATJA, CROTTA STEFANIA, MACK MATTHIAS, PRIESTNALL SIMON L., WACK ANDREAS: "A TLR7 antagonist restricts interferon-dependent and -independent immunopathology in a mouse model of severe influenza", JOURNAL OF EXPERIMENTAL MEDICINE, ROCKEFELLER UNIVERSITY PRESS, US, vol. 218, no. 11, 1 November 2021 (2021-11-01), US , pages 1 - 15, XP055963638, ISSN: 0022-1007, DOI: 10.1084/jem.20201631 *
TIAN JING; JIAO XUE; WANG XIAONAN; GENG JIN; WANG REIZHE; LIU NING; GAO XINHUA; GRIFFIN NOREEN; SHAN FENGPING: "Novel effect of methionine enkephalin against influenza A virus infection through inhibiting TLR7-MyD88-TRAF6-NF-κB p65 signaling pathway", INTERNATIONAL IMMUNOPHARMACOLOGY, ELSEVIER, AMSTERDAM, NL, vol. 55, 22 December 2017 (2017-12-22), NL , pages 38 - 48, XP085352907, ISSN: 1567-5769, DOI: 10.1016/j.intimp.2017.12.001 *
WENJI PIAO, KARI ANN SHIREY, LISA W RU, WENDY LAI, HENRYK SZMACINSKI , GREG A SNYDER , ERIC J SUNDBERG , JOSEPH R LAKOWICZ , STEFA: "A Decoy Peptide that Disrupts TIRAP Recruitment to TLRs Is Protective in a Murine Model of Influenza", CELL REPORTS, vol. 11, 30 June 2015 (2015-06-30), pages 1941 - 1952, XP055963630 *

Similar Documents

Publication Publication Date Title
US11298403B2 (en) Therapeutic agent for inflammatory bowel disease
MX2011003625A (en) Use of inhibitors of toll-like receptors in the prevention and treatment of hypercholesterolemia and hyperlipidemia and diseases related thereto.
US20190309021A1 (en) Composition Comprising a Peptide and an Inhibitor of Viral Neuraminidase
WO2017201199A9 (en) Compositions and methods for the treatment of prader-willi syndrome
AU2018241248A1 (en) Treatment of respiratory infection with a TLR2 agonist
US10413586B2 (en) Antiviral agent comprising recombinant mistletoe lectins
US20210401751A1 (en) Compositions for treating viral infections and methods for same
ES2710748T3 (en) Compounds to treat the blocking of remyelination in diseases associated with the expression of the HERV-W envelope protein
AU2016317574A1 (en) Compositions and methods for the treatment of neurodamage
IL237119A (en) Heterocyclyl carboxamides for treating viral diseases
WO2022170396A1 (en) Agents and methods for therapy and prophylaxis
US11197912B2 (en) Prevention and treatment of viral infection and viral infection-induced organ failure
JP2020531586A (en) Methods and compositions for antiviral applications of synthetic lysine analogs and mimetics
CA2600714A1 (en) Methods of treating or preventing a disease, disorder or condition associated with a viral infection
WO2021237067A1 (en) Interferon- inducing oligonucleotide duplexes and methods of use
JP2009057318A (en) S-nitroso group-containing albumin, method for producing the same and anticancer agent
EA012342B1 (en) Treatment or prevention of hemorrhagic viral infections with immunomodulator compounds
EP3678702A1 (en) Treatment of neurodegenerative diseases
US9138466B2 (en) Factor H for transplantation
US20240115693A1 (en) Sars-cov-2 antigen nanoparticles and uses there of
US20230277650A1 (en) Use of a birnavirus for the treatment of a disease caused by varicella zoster virus (vzv)
KR20100005063A (en) Compositions comprising a statin and caffeine for the treatment of viral infection
WO2012105467A1 (en) Oligonucleic acid composition and antiallergic agent
WO2022265697A1 (en) Compositions for treating viral infections and methods for same
WO2022112358A2 (en) Compounds for treating enveloped virus infections

Legal Events

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

Ref document number: 22752009

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22752009

Country of ref document: EP

Kind code of ref document: A1