Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS AND COMPOSITIONS FOR TREATMENT OF SARS-COV-2 INFECTION
Document Type and Number:
WIPO Patent Application WO/2021/198511
Kind Code:
A1
Abstract:
Inventors collect and analyze the blood of patients to determine their cytokine profile in the serum and the profile/activation status of circulating leukocytes by flow cytometry and measure inflammasomes activation using the FAM-FLICA probe that labels activated caspase-1 (that processes pro-IL-1β in mature IL-1β). Moreover, inventors describes that a use of a specific NLRP3 inhibitor (MCC950) allows them to determine that the activation observed in the blood samples is NLRP3 dependent. Accordingly, the invention relates to NLRP3 inhibitors for use in the treatment of SARS-CoV-2 infection and SARS-CoV-2 related disorders in a subject in need thereof.

Inventors:
BOYER LAURENT (FR)
AUBERGER PATRICK (FR)
IVANOV STOYAN (FR)
COURJON JOHAN (FR)
POMARES CHRISTELLE (FR)
GIORDANENGO VALÉRIE (FR)
VISVIKIS ORANE (FR)
LOUBATIER CÉLINE (FR)
VITALE SÉBASTIEN (FR)
DUFIES OCÉNANE (FR)
TORRE CÉDRIC (FR)
DOYE ANNE (FR)
MUNRO PATRICK (FR)
LOTTE ROMAIN (FR)
JACQUEL ARNAUD (FR)
ROBERT ALEX (FR)
Application Number:
PCT/EP2021/058799
Publication Date:
October 07, 2021
Filing Date:
April 02, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INST NAT SANTE RECH MED (FR)
UNIV COTE D'AZUR (FR)
CENTRE HOSPITALIER UNIV DE NICE (FR)
HOPITAL DE CANNES (FR)
International Classes:
A61K31/18; A61K31/00; A61K31/4184; A61K31/4706; A61K31/64; A61K31/713; A61K45/06; A61P31/14; G01N33/50
Domestic Patent References:
WO2017129897A12017-08-03
WO2013007763A12013-01-17
WO2016012322A12016-01-28
WO2017031161A12017-02-23
WO2017017469A12017-02-02
WO2017184746A12017-10-26
WO2019025467A12019-02-07
WO2019034693A12019-02-21
WO1993011161A11993-06-10
WO2006030220A12006-03-23
WO2006003388A22006-01-12
WO2004007504A12004-01-22
WO2006072831A12006-07-13
WO2007023382A22007-03-01
WO2007072153A22007-06-28
WO2009086204A22009-07-09
WO2010071846A22010-06-24
WO2011044264A22011-04-14
WO2011044535A22011-04-14
WO2011156640A22011-12-15
WO2011156646A22011-12-15
WO2011156775A22011-12-15
WO2011156780A22011-12-15
WO2011156786A22011-12-15
WO2013026914A12013-02-28
WO2012078605A12012-06-14
WO2012150543A12012-11-08
WO2011075592A12011-06-23
WO1993016189A11993-08-19
WO2012028556A12012-03-08
Foreign References:
CN110575436A2019-12-17
CN112168966A2021-01-05
EP0404097A21990-12-27
US4816567A1989-03-28
US6982321B22006-01-03
US7087409B22006-08-08
US6075181A2000-06-13
US6150584A2000-11-21
EP0368684A11990-05-16
US0006566A1849-07-03
US0000135A1837-03-03
US6566131B12003-05-20
US6365354B12002-04-02
US6410323B12002-06-25
US6107091A2000-08-22
US6046321A2000-04-04
US5981732A1999-11-09
Other References:
LOH DORIS: "COVID-19, PNEUMONIA & INFLAMMASOMES - THE MELATONIN CONNECTION 0SHARES", 14 March 2020 (2020-03-14), XP055724831, Retrieved from the Internet [retrieved on 20200825]
ANONYMOUS: "ICTRP Search Portal", 19 February 2020 (2020-02-19), XP055724814, Retrieved from the Internet [retrieved on 20200825]
WEN ZHANG ET AL: "The use of anti-inflammatory drugs in the treatment of people with severe coronavirus disease 2019 (COVID-19): The Perspectives of clinical immunologists from China", CLINICAL IMMUNOLOGY, vol. 214, 25 March 2020 (2020-03-25), US, pages 108393, XP055724895, ISSN: 1521-6616, DOI: 10.1016/j.clim.2020.108393
ADNAN SHAH: "Novel Coronavirus-Induced NLRP3 Inflammasome Activation: A Potential Drug Target in the Treatment of COVID-19", FRONTIERS IN IMMUNOLOGY, vol. 11, 19 May 2020 (2020-05-19), pages 1021, XP055724935, DOI: 10.3389/fimmu.2020.01021
"cas", Database accession no. 210826-40-7
ZETSCHE ET AL.: "Cpfl is a Single RNA-guided Endonuclease of a Class 2 CRISPR-Cas System", CELL, vol. 163, 2015, pages 1 - 13
YI ET AL., BIOCHEMICAL PHARMACOLOGY, vol. 80, 2010, pages 683 - 689
LESPINE A: "Lipid-like properties and pharmacology of the anthelmintic macrocyclic lactones", EXPERT OPIN DRUG METAB TOXICOL., vol. 9, no. 12, December 2013 (2013-12-01), pages 1581 - 95
HASMIMOTO ET AL.: "Ivermectin inactivates the kinase PAK-1 and blocks the PAK-1 dependent growth of human ovarian cancer and NF2 tumor cell lines", DRUG DISCOV. THERAPEUTICS, vol. 3, no. 6, 2009, pages 243 - 246
CAS, no. 2016806-55-4
Attorney, Agent or Firm:
INSERM TRANSFERT (FR)
Download PDF:
Claims:
CLAIMS:

1. A NOD-like receptor family pyrin domain containing 3 (NLRP3) inhibitor for use in the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and SARS-CoV-2 related disorders in a subject in need thereof.

2. The NLRP3 inhibitor for use according to claim 1, wherein the inhibitor is siRNA.

3. The NLRP3 inhibitor for use according to claim 1, wherein the inhibitor is a small molecule.

4. The NLRP3 inhibitor for use according to claim 3, wherein the small molecule is selected from the group consisting of but not limited to MCC950; colchicine, a sufonylurea drug and its derivatives thereof, glyburide precursors and its derivatives that lack the cyclohexylurea moiety, or functionally equivalent precursors or derivatives that contain the sulfonyl and benamido groups; 5-chloro-2-methoxy-N-[2-(4-sulfamoylphenyl)- ethyl]- benzamide and 1 - [(4-methylbenzene)sulfonyl] - 1 H- 1 ,3 -benzodiazol-2- amine.

5. The NLRP3 inhibitor for use according to claims 1 to 4, wherein the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is selected from the group consisting of but not limited to: COVID-19; cytokine storm syndrome (CSS); Respiratory distress syndrome (RDS).

6. i)A NLRP3 inhibitor and ii) a classical treatment used as a combined preparation for treating SARS-CoV-2 infection in a subject.

7. i)A NLRP3 inhibitor and ii) an anti-parasitic treatment used as a combined preparation for treating SARS-CoV-2 infection in a subject.

8. The combined preparation according to claim 7, wherein the NLRP3 inhibitor is MCC950 and the anti-parasitic treatment is chloroquine.

9. i)A NLRP3 inhibitor and ii) a neutralizing monoclonal anti-IL-Ib antibody used as a combined preparation for treating SARS-CoV-2 infection in a subject 10. i)A NLRP3 inhibitor and ii) a recombinant human IL-1B receptor antagonist used as a combined preparation for treating SARS-CoV-2 infection in a subject.

11. A pharmaceutical composition comprising a NLRP3 inhibitor for treating SARS-CoV- 2 infection. 12. A method of screening a drug suitable for the treating SARS-CoV-2 infection comprising i) providing a test compound and ii) determining the ability of said test compound to activate or inhibit the expression or activity ofNLRP3.

Description:
METHODS AND COMPOSITIONS FOR TREATMENT OF SARS-CoV-2

INFECTION

FIELD OF THE INVENTION:

The invention is in the field of infection disorders. More particularly, the invention relates to methods and compositions for treatment of SARS-CoV-2.

BACKGROUND OF THE INVENTION:

Why some patients with no medical history develop a serious form of COVID-19 while most are paucisymptomatic? Considering that we have not herd immunity against SARS-CoV- 2, part of the answer is possibly that patients developing a serious form are not able to mount a protective antiviral innate immune response or have an excessive and detrimental immune response.

It is thus critical to investigate the innate immune response to SARS-CoV-2 in order to decipher the underlying mechanisms of the immune response to this novel pathogen.

SARS-CoV-2 is a novel human coronavirus which emerged in December 2019 in Wuhan, China 1. The virus is responsible for a contagious respiratory illness named COVID- 19 (COronaVIrus Disease- 19) that can evolve in a life-threatening Severe Acute Respiratory Syndrome (SARS) in some cases 2 . However, some patients infected by SARS-CoV-2 suffer from mild COVID-19 conditions, reporting only slight cough and low-grade fever, and even cases of asymptomatic carriers have been reported 2 . As for most viral infections, it is very likely that the outcome of the infection is mainly governed by the interplay between virus and host antiviral immunity. Innate immunity is the first line of defense against pathogen invasion in naive patients. It plays an essential role in restricting viral replication and activating adaptive immunity during the first stages of infection. Innate immune defects have been involved in susceptibility to infection while activating mutations can cause auto-inflammatory diseases 3 . Both innate and adaptive immunity works as a continuum that starts by an efficient detection of the pathogen by the innate immune system 4 . The innate immune detection system of viruses relies on Pattern Recognition Receptors (PRRs). PRRs are conserved proteins able to sense Pathogen-Associated- Molecular-Patterns (PAMPS) that are specific to microbes 5 . Viral nucleic acids as well as viral proteins have been shown to be interplay with PRR 6 . Among the PPR involved in virus detection, Toll like receptors (TLRs), Nod like receptors (NLRs), RIG- I-like receptors (RLRs) as well as the cGAS-STING pathway are critical for the antiviral response. Indeed TLRs stimulation triggers the activation of the NF-kB transcription factor as well as Interferon Responsive Factors (IRFs), while inflammasomes control the maturation of the Interleukine-ΐb (IL-Ib) and IL-18 cytokines and the activation of the RLR and STING pathway regulate type I Interferons (IFNs) response 7 . Activation of the STING pathway results in nuclear translocation of IRF3 and subsequent production of IFN-b. IFN-b plays a crucial role in restricting viral replication during the very first steps of virus invasion through autocrine and paracrine activation of Interferon-Stimulated Genes (ISGs) 8 9 . In particular, IFN-b was shown to inhibit SARSCoV replication 10 .

It has been shown that several RNA viruses have evolved strategies to counteract activation of the cGAS-STING pathway and evade host innate immunity to favor virus spread. Particularly, SARS-CoV and other coronaviruses encode papain-like proteases that disrupt STING signaling u 12 . Genome sequencing studies have shown that SARS-CoV-2 shares high similarity with SARS-CoV, the etiological agent of a previous S ARS outbreak which took place in China in 2002-2003 13 . Thus, by analogy, SARS-CoV-2 papain-like protease may be involved in inhibition of STING and the subsequent IFN-b production. In addition, the ~30 kb genome of SARS-CoV-2 contains multiple ORFs encoding non- structural and structural proteins that could activate or interfere with innate immune signaling, but to date data on the contribution of SARS-CoV-2 proteins to innate immunity are lacking 14 .

Recent studies suggest that the death of COVID-19 patients with no medical history can be attributed to a cytokine storm that is similar to what is observed during sepsis with excessive plasma IL-6 and IL-Ib levels 15 .

In this context it is critical to determine the monitor the status of patients and propose therapies that could dampen the detrimental inflammation while preserving the efficiency of the anti-viral response efficient.

SUMMARY OF THE INVENTION:

The invention relates to a NOD-like receptor family, pyrin domain containing 3 (NLRP3) inhibitor for use in the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and SAR-CoV-2 related disorders in a subject in need thereof. In particular, the invention is claimed by the claims.

DETAILED DESCRIPTION OF THE INVENTION: Inventors collect and analyze the blood of COVID-19 patients to measure inflammasomes activation using the FAM-FLICA probe that labels activated caspase-1 (that processes pro-IL-Ib in mature IL-Ib).

Moreover, inventors describes that a use of a specific NLRP3 inhibitor (MCC950 or Colchicine) allows them to determine if the activation observed in the blood samples is NLRP3 dependent.

Accordingly, in a first aspect the invention relates to a NOD-like receptor family, pyrin domain containing 3 (NLRP3) inhibitor for use in the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) disorders and SARS-CoV-2 related disorders in a subject in need thereof.

In a particular embodiment, the invention relates to a method for treating SARS-CoV-2 infection and SARS-CoV-2 related disorders in a subject in need thereof comprising a step of administering said subject with a therapeutically effective amount of a NLRP3 inhibitor.

As used herein, the terms “treating” or “treatment” refer to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of subject at risk of contracting the disease or suspected to have contracted the disease as well as subject who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse. The treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment. By "therapeutic regimen" is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy. A therapeutic regimen may include an induction regimen and a maintenance regimen. The phrase "induction regimen" or "induction period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease. The general goal of an induction regimen is to provide a high level of drug to a subject during the initial period of a treatment regimen. An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both. The phrase "maintenance regimen" or "maintenance period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a subject during treatment of an illness, e.g., to keep the subject in remission for long periods of time (months or years). A maintenance regimen may employ continuous therapy (e.g., administering a drug at a regular intervals, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., pain, disease manifestation, etc.]).

As used herein, the term “SARS-CoV-2” refers to severe acute respiratory syndrome coronavirus 2 known by the provisional name 2019 novel coronavirus (2019-nCoV) is the cause of the respiratory coronavirus disease 2019 (COVID-19). Taxonomically, it is a strain of the Severe acute respiratory syndrome-related coronavirus (SARSr-CoV), a positive-sense single- stranded RNA virus. It is contagious in humans, and the World Health Organization (WHO) has designated the ongoing pandemic of COVID-19 a Public Health Emergency of International Concern. SARS-CoV-2 virion is approximately 50-200 nanometres in diameter. Like other coronaviruses, SARS-CoV-2 has four structural proteins, known as the S (spike), E (envelope), M (membrane), and N (nucleocapsid) proteins; the N protein holds the RNA genome, and the S, E, and M proteins together create the viral envelope. The spike protein, which has been imaged at the atomic level using cryogenic electron microscopy is the protein responsible for allowing the virus to attach to the membrane of a host cell.

As used herein, “SARS-CoV-2 infection” refers to the transmission of this virus from an animal and/or human to another animal and/or human primarily via respiratory droplets from coughs and sneezes within a range of about 2 meters. Indirect contact via contaminated surfaces is another possible cause of infection.

As used herein, “SARS-CoV-2 related disorders” refers to all diseases and/or complications linked to SAR-CoV-2 infection. In a particular embodiment, the SARS-CoV-2 related disorder is selected from the group consisting of but not limited to: Covid-19; cytokine storm syndrome (CSS); Respiratory distress syndrome (RDS); gastroenteritis and respiratory infections; pneumonia. In a particular embodiment, the SARS-CoV-2 is COVID-19.

As used herein, the term “subject” refers to any mammals or birds, such as a rodent, a feline, a canine, a bat and a primate. In a particular embodiment, the subject is human. Particularly, in the present invention, the subject has or is susceptible to have SARS-CoV-2 infection. In a particular embodiment, the subject has or is susceptible to have and SARS-CoV- 2 related disorders and/or complications.

As used herein, the term “NLRP3” refers to Nucleotide-binding oligomerization domain-like receptor including a pyrin domain 3. Nucleotide-binding oligomerization domain like receptors ("NLRs") include a family of intracellular receptors that detects pathogen- associated molecular patterns ("PAMPs") and endogenous signal danger molecules. NLRPs represent a subfamily of NLRs that include a Pyrin domain and are constituted by proteins such as NLRP l , NLRP3, NLRP4, NLRP6, NLRP7, and NLRP l 2. NLRPs are involved in the formation of multiprotein complexes termed inflammasomes. The NLRP3 inflammasome forms a molecular platform inside macrophages and microglial cells, catalyzing the activation of the protease Caspase-1. Caspase-1 is responsible for converting the potent pro-inflammatory cytokine interleukin- 1 beta (IL-Ib) from an inactive to an active secreted form.

As used herein the term "IL-1 beta" has its general meaning in the art and refers to Interleukin- 1 beta. IL-1 beta is a member of the Interleukin 1 cytokine family. This cytokine is produced as a proprotein, which is proteolytically processed to its active form by Caspase 1 (CASP 1/ICE). This cytokine is an important mediator of the inflammatory response, and is involved in a variety of cellular activities, including cell proliferation, differentiation, and apoptosis.

As used herein, the term “an inhibitor of NLRP3” refers to a natural or synthetic compound that has a biological effect to inhibit the activity or the expression of NLRP3. More particularly, such inhibitor inhibits the recruitment of the associated protein Nek7 and/or the adapter protein the apoptosis-associated speck-like (ASC) pro-caspase-1 leading to caspase-1 production and subsequent IL-Ib maturation and release.

In a particular embodiment, the inhibitor of NLRP3 is a peptide, peptidomimetic, small organic molecule, antibody, aptamers, siRNA or antisense oligonucleotide. The term “peptidomimetic” refers to a small protein-like chain designed to mimic a peptide. In a particular embodiment, the inhibitor of NLRP3 is an aptamer. Aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition. Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.

In a particular embodiment, the inhibitor of NLRP3 is a small organic molecule. The term “small organic molecule” refers to a molecule of a size comparable to those organic molecules generally used in pharmaceuticals. The term excludes biological macro molecules (e.g., proteins, nucleic acids, etc.). Preferred small organic molecules range in size up to about 5000 Da, more preferably up to 2000 Da, and most preferably up to about 1000 Da.

In a particular embodiment, the inhibitor of NLRP3 is MCC950. MCC950 blocks the release of IL-lbeta induced by NLRP3 activators, such as ATP, MSU and Nigericin, by preventing oligomerization of the inflammasome adaptor protein ASC (apoptosis-associated speck-like protein containing CARD). MCC950 is well known in the art and has the cas number 210826-40-7 and chemical formula: C20H24N2O5S. In a particular embodiment, the inhibitor of NLRP3 is Colchicine. Colchicine is well known in the art and has the cas number 64-86-8 and chemical formula: C22H25N06.

In a particular embodiment, the inhibitor of NLRP3 is described in the following patent applications: WO2017/129897; W02013/007763; WO2016/12322; W02017/031161;

WO20 17/017469; WO2017/184746; WO2019/025467; WO2019/034693.

In a particular embodiment, the inhibitor of NLRP3 is selected from the group consisting of but not limited to: a sufonylurea drug such as glyburide, including functionally equivalent derivatives thereof, for example, glyburide precursors or derivatives that lack the cyclohexylurea moiety, or functionally equivalent precursors or derivatives that contain the sulfonyl and benamido groups. Examples include 5-chloro-2-methoxy-N-[2-(4- sulfamoylphenyl)-ethyl]- benzamide and l-[(4-methylbenzene)-sulfonyl]-lH-l,3-benzodiazol- 2-amine. Functionally equivalent precursors or derivatives of glyburide include precursors or derivatives that retain the activity of glyburide, at least in part, to inhibit or reduce the activity of NLRP3 inflammasome, e.g. retain at least about 25% of the activity of glyburide, preferably about 50% of glyburide activity, for example, at least about 70%, 80%, or 90% if glyburide activity.

In some embodiments, the inhibitor of NLRP3 is an antibody. As used herein, the term “antibody” is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity. The term includes antibody fragments that comprise an antigen binding domain such as Fab', Fab, F(ab')2, single domain antibodies (DABs), TandAbs dimer, Fv, scFv (single chain Fv), dsFv, ds-scFv, Fd, linear antibodies, minibodies, diabodies, bispecific antibody fragments, bibody, tribody (scFv-Fab fusions, bispecific or trispecific, respectively); sc-diabody; kappa(lamda) bodies (scFv-CL fusions); BiTE (Bispecific T-cell Engager, scFv-scFv tandems to attract T cells); DVD-Ig (dual variable domain antibody, bispecific format); SIP (small immunoprotein, a kind of minibody); SMIP ("small modular immunopharmaceutical" scFv-Fc dimer; DART (ds-stabilized diabody "Dual Affinity ReTargeting"); small antibody mimetics comprising one or more CDRs and the like. The techniques for preparing and using various antibody-based constructs and fragments are well known in the art (see Rabat et ak, 1991, specifically incorporated herein by reference). Diabodies, in particular, are further described in EP 404, 097 and WO 93/1 1 161; whereas linear antibodies are further described in Zapata et al. (1995). Antibodies can be fragmented using conventional techniques. For example, F(ab')2 fragments can be generated by treating the antibody with pepsin. The resulting F(ab')2 fragment can be treated to reduce disulfide bridges to produce Fab' fragments. Papain digestion can lead to the formation of Fab fragments. Fab, Fab' and F(ab')2, scFv, Fv, dsFv, Fd, dAbs, TandAbs, ds-scFv, dimers, minibodies, diabodies, bispecific antibody fragments and other fragments can also be synthesized by recombinant techniques or can be chemically synthesized. Techniques for producing antibody fragments are well known and described in the art. For example, each of Beckman et al, 2006; Holliger & Hudson, 2005; Le Gall et ah, 2004; Reff & Heard, 2001 ; Reiter et al., 1996; and Young et al., 1995 further describe and enable the production of effective antibody fragments. In some embodiments, the antibody is a “chimeric” antibody as described in U.S. Pat. No. 4,816,567. In some embodiments, the antibody is a humanized antibody, such as described U.S. Pat. Nos. 6,982,321 and 7,087,409. In some embodiments, the antibody is a human antibody. A “human antibody” such as described in US 6,075,181 and 6,150,584. In some embodiments, the antibody is a single domain antibody such as described in EP 0368 684, WO 06/030220 and WO 06/003388.

In a particular embodiment, the inhibitor is a monoclonal antibody. Monoclonal antibodies can be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture. Techniques for production and isolation include but are not limited to the hybridoma technique, the human B-cell hybridoma technique and the EBV-hybridoma technique.

In a particular, the inhibitor is an intrabody having specificity for NLRP3. As used herein, the term "intrabody" generally refer to an intracellular antibody or antibody fragment. Antibodies, in particular single chain variable antibody fragments (scFv), can be modified for intracellular localization. Such modification may entail for example, the fusion to a stable intracellular protein, such as, e.g., maltose binding protein, or the addition of intracellular trafficking/localization peptide sequences, such as, e.g., the endoplasmic reticulum retention. In some embodiments, the intrabody is a single domain antibody. In some embodiments, the antibody according to the invention is a single domain antibody. The term “single domain antibody” (sdAb) or "VHH" refers to the single heavy chain variable domain of antibodies of the type that can be found in Camelid mammals which are naturally devoid of light chains. Such VHH are also called “nanobody®”. According to the invention, sdAb can particularly be llama sdAb.

In some embodiments, the NLRP3 inhibitor is an inhibitor of NLRP3 expression. An "inhibitor of expression" refers to a natural or synthetic compound that has a biological effect to inhibit the expression of a gene. In a particular embodiment of the invention, the inhibitor of gene expression is a siRNA, an antisense oligonucleotide or a ribozyme.

For example, anti-sense oligonucleotides, including anti-sense RNA molecules and anti- sense DNA molecules, would act to directly block the translation of NLRP3 mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of NLRP3, and thus activity, in a cell.

For example, antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence encoding NLRP3 can be synthesized, e.g., by conventional phosphodiester techniques. Methods for using antisense techniques for specifically inhibiting gene expression of genes whose sequence is known are well known in the art (e.g. see U.S. Pat. Nos. 6,566, 135; 6,566,131; 6,365,354; 6,410,323; 6,107,091; 6,046,321; and 5,981,732). Small inhibitory RNAs (siRNAs) can also function as inhibitors of expression for use in the present invention. NLRP3 gene expression can be reduced by contacting a subject or cell with a small double stranded R A (dsPvNA), or a vector or construct causing the production of a small double stranded R A, such that NLRP3 gene expression is specifically inhibited (i.e. RNA interference or RNAi). Antisense oligonucleotides, siRNAs, shRNAs and ribozymes of the invention may be delivered in vivo alone or in association with a vector. In its broadest sense, a "vector" is any vehicle capable of facilitating the transfer of the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid to the cells and typically cells expressing NLRP3. Typically, the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector. In general, the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid sequences. Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rous sarcoma virus; adenovirus, adeno-associated virus; SV40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and RNA virus such as a retrovirus. One can readily employ other vectors not named but known to the art. In some embodiments, the inhibitor consists in a vector that comprises the CRISPR/cas 9 protein and the appropriate RNA guide for disrupting the expression level of the gene encoding for NLRP3. In some embodiments, the endonuclease is CRISPR-Cpfl which is the more recently characterized CRISPR from Provotella and Francisella 1 (Cpfl) in Zetsche et al. (“Cpfl is a Single RNA-guided Endonuclease of a Class 2 CRISPR-Cas System (2015); Cell; 163, 1-13).

In a particular embodiment, the inhibitor of NLRP3 is not Tranilast, melatonin, ascorbic acid or nitric oxide.

As used herein the terms "administering" or "administration" refer to the act of injecting or otherwise physically delivering a substance as it exists outside the body (e.g., NLRP3 inhibitor) into the subject, such as by oral, mucosal, intradermal, intravenous, subcutaneous, intramuscular delivery and/or any other method of physical delivery described herein or known in the art. When a disease, or a symptom thereof, is being treated, administration of the substance typically occurs after the onset of the disease or symptoms thereof. When a disease or symptoms thereof, are being prevented, administration of the substance typically occurs before the onset of the disease or symptoms thereof. More particularly, the PAK-1 and/or PAK- 2 inhibitors are formulated for an oral administration is performed to the subject. In a further embodiment, intravenous administration is performed to the subject. In a particular embodiment, the NLRP3 inhibitor is according to the invention is formulated for a nasal administration.

By a "therapeutically effective amount" is meant a sufficient amount of a NLRP3 inhibitor for use in a method for the treatment of SARS-CoV-2 infection at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide employed; and like factors well known in the medical arts. For example, it is well known within the skill of the art to start doses of the compound at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. However, the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day. Typically, the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic 20 adjustment of the dosage to the subject to be treated. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, typically from 1 mg to about 100 mg of the active ingredient. An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.

Combined preparation:

The NLRP3 inhibitor as described above is also combined with a classical treatment.

As used herein, the term “classical treatment” refers to antiviral treatment, antibiotics, anti-parasitic treatment, immunosuppressive corticosteroids or non-steroidal therapies; immunotherapy: recombinant human IL-1B receptor antagonist; neutralizing monoclonal anti- IL-Ib antibody; PAK-l/PAK-2 inhibitors; or caspase-1 inhibitor.

Accordingly, in a second aspect, the invention relates to i) a NLRP3 inhibitor and ii) a classical treatment used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof.

In a particular embodiment, the combined preparation according to the invention, wherein the SARS-CoV-2 is COVID-19.

In a particular embodiment, the invention relates to i) a NLRP3 inhibitor and ii) antiviral treatment used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof.

As used herein, the term “antiviral treatment” to an inhibitor which inhibits the viral entry, viral internalization, viral replication and/or viral release. Such inhibitor prevents or reduces the occurrence of viral resistance, and the like. Typically, the antiviral treatment includes but not limited to interferons (e.g., interferon-alpha, pegylated interferon-alpha), ribavirin, anti-HCV, (monoclonal or polyclonal) antibodies, RNA polymerase inhibitors, protease inhibitors, IRES inhibitors, helicase inhibitors, antisense compounds, ribozymes, neuraminidase inhibitor, nucleoside analogues of guanine, nucleoside analogue of thymidine, nucleoside reverse transcriptase inhibitors (NRTI), nucleotide reverse transcriptase inhibitor (NtRTI), protease inhibitors and any combination thereof.

Typically, the antiviral treatment is selected from the group consisting of but not limited to Abacavir, Acyclovir (Aciclovir), Adefovir, Amantadine, Ampligen, Amprenavir (Agenerase), Arbidol, Atazanavir, Atripla, Balavir, Baloxavir marboxil (Xofluza), Biktarvy, Boceprevir (Victrelis), Cidofovir, Cobicistat (Tybost), Combivir, Daclatasvir (Daklinza), Darunavir, Delavirdine, Descovy, Didanosine, Docosanol Dolutegravir, Doravirine (Pifeltro), Ecoliever, Edoxudine, Efavirenz, Elvitegravir, Emtricitabine, Enfuvirtide, Entecavir, Etravirine (Intelence), Famciclovir, Fomivirsen, Fosamprenavir, Foscamet, Fosfonet, Fusion inhibitor, Ganciclovir (Cytovene), Ibacitabine, Ibalizumab (Trogarzo), Idoxuridine, Imiquimod, Imunovir, Indinavir, Inosine, Integrase inhibitor, Interferon type I, Interferon type II, Interferon type III, Interferon, Lamivudine, Letermovir (Prevymis), Lopinavir, Loviride, Maraviroc, Methisazone, Moroxydine, Nelfmavir, Nevirapine, Nexavir, Nitazoxanide, Norvir, Nucleoside analogues, Oseltamivir (Tamiflu), Peginterferon alfa-2a, Peginterferon alfa-2b, Penciclovir, Peramivir (Rapivab), Pleconaril, Podophyllotoxin, Protease inhibitor, Pyramidine, Raltegravir, Remdesivir, Reverse transcriptase inhibitor Ribavirin, Rilpivirine (Edurant), Rimantadine, Ritonavir, Saquinavir, Simeprevir (Olysio), Sofosbuvir, Stavudine, Synergistic enhancer, Telaprevir, Telbivudine (Tyzeka), Tenofovir alafenamide, Tenofovir disoproxil, Tenofovir, Tipranavir, Trifluridine, Trizivir, Tromantadine, Truvada, Valaciclovir (Valtrex), Valganciclovir, Vicriviroc, Vidarabine, Viramidine, Zalcitabine, Zanamivir (Relenza) and/or Zidovudine.

In a particular embodiment, the invention relates to i) a NLRP3 inhibitor and ii) an antibiotic used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof.

As used herein, the term “antibiotic” refers to an antimicrobial substance active against bacteria and is the most important type of antibacterial agent for fighting bacterial infections. Each antibiotic belongs to an antibiotic class which is a grouping of different drugs that have similar chemical and pharmacologic properties. Their chemical structures may look comparable, and drugs within the same class may kill the same or related bacteria. Typically, the antibiotic class includes but not limited to Penicillins, Tetracyclines, Cephalosporins, Quinolones, Lincomycins, Macrolides, Sulfonamides, Glycopeptides, Aminoglycosides, and Carbapenems.

Typically, the antibiotic is selected from the group consisting of but not limited to amoxicillin, amoxicillin and clavulanate, ampicillin, dicloxacillin, oxacillin, penicillin V potassium, demeclocycline, doxycycline, eravacycline, minocycline, omadacycline, tetracycline, cefaclor, cefdinir, cefotaxime, ceftazidime, ceftriaxone, cefuroxime, ciprofloxacin, levofloxacin, moxifloxacin, clindamycin, lincomycin, azithromycin, clarithromycin, erythromycin, sulfamethoxazole and trimethoprim, sulfasalazine, dalbavancin, oritavancin, telavancin, vancomycin, gentamicin, tobramycin, amikacin, imipenem and cilastatin, meropenem, doripenem and ertapenem.

In a particular embodiment, the invention relates to i) a NLRP3 inhibitor and ii) an anti- parasitic treatment used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof. As used herein, the term “anti-parasitic treatment” refers to a treatment of parasitic diseases, such as those caused by helminths, amoeba, ectoparasites, parasitic fungi, and protozoa. Antiparasitics target the parasitic agents of the infections by destroying them or inhibiting their growth.

In a particular embodiment, the anti-parasitic drug is selected from the group consisting of but not limited to: chloroquine, amodiaquine, mefloquine, halofantrine, artemether, artesunate, arteminol, sulfadoxine, pyrimethamine, proguanil, atovaquone, quinine, abamectine, albendazole, diethylcarbamazine, mebendazole, niclosamide, ivermectin, suramine, thiabendazole, levamisole, praziquantel, triclabendazole, flubendazole, metronidazole, tinidazole, secnidazole, tenonitrozole, pyrimethamine, amphotericin B, pentamidine, miltefosine, nifurtimox, benznidazole, amphotericin B, ketoconazole, econazole, griseofulvin, miconazole orfluconazole.

In a particular embodiment, the anti-parasitic drug is an anti-malaria drug. Typically, the anti-parasitic drug is chloroquine.

In a particular embodiment, the NLRP3 inhibitor and ii) chloroquine used as a combined preparation for treating SARS-CoV-2 infection in a subject.

Accordingly, in a particular embodiment, the invention relates to a NLRP3 inhibitor and ii) an anti-parasitic treatment used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof, wherein the NLRP3 inhibitor is MCC950 and the anti- parasitic drug is chloroquine.

In a particular embodiment, the invention relates to a NLRP3 inhibitor and ii) an anti- parasitic treatment used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof, wherein the NLRP3 inhibitor is Colchicine and the anti-parasitic drug is chloroquine.

In a particular embodiment, the invention relates to i) a NLRP3 inhibitor and ii) immunosuppressive corticosteroids used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof.

As used herein, the term “corticosteroid” is well known in the art and refers to class of steroid hormones that are produced in the adrenal cortex as well as the synthetic analogues of these hormones. Two types of classes of corticosteroid exist in the art: glucocorticoids and mineralocorticoids. The corticosteroid for use in the invention is selected from the group consisting of: Flugestone (flurogestone); Fluorometholone; Medrysone; Prebediolone acetate; chlormadinone acetate, cyproterone acetate, medrogestone, medroxyprogesterone acetate, megestrol acetate, and segesterone acetate; Chloroprednisone; Cloprednol; Difluprednate; Fludrocortisone; Fluocinolone; Fluperolone; Fluprednisolone; Loteprednol; Methylprednisolone; Prednicarbate; Prednisolone; Prednisone; Tixocortol; Triamcinolone; Alclometasone; Beclometasone; Betamethasone; Clobetasol; Clobetasone; Clocortolone; Desoximetasone; Dexamethasone; Diflorasone; Difluocortolone; Fluclorolone; Flumetasone; Fluocortin; Fluocortolone; Fluprednidene; Fluticasone; Fluticasone furoate; Halometasone; Meprednisone; Mometasone; Mometasone furoate; Paramethasone; Prednylidene; Rimexolone; Ulobetasol (halobetasol); Amcinonide; Budesonide; Ciclesonide; Deflazacort; Desonide; Formocortal (fluoroformylone); Fluclorolone acetonide (flucloronide); Fludroxycortide (flurandrenolone, flurandrenolide); Flunisolide; Fluocinolone acetonide; Fluocinonide; Halcinonide; Triamcinolone acetonide; Cortivazol; RU-28362.

In a particular embodiment, the invention relates to i) a NLRP3 inhibitor and ii) non steroidal drug used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof.

As used herein, the term “nonsteroidal drug” refers to a class of drugs which decrease inflammation. The nonsteroidal drug for use in the invention is selected from the group consisting of: Aspirin (acetylsalicylic acid); Diflunisal (Dolobid); Salicylic acid and other salicylates Salsalate (Disalcid); Ibuprofen; Dexibuprofen ; Naproxen ; Fenoprofen ; Ketoprofen ; Dexketoprofen ; Flurbiprofen ; Oxaprozin; Loxoprofen; Indomethacin; Tolmetin; Sulindac; Etodolac; Ketorolac; Diclofenac; Aceclofenac; Nabumetone; Piroxicam; Meloxicam; Tenoxicam; Droxicam; Lornoxicam; Phenylbutazone; Mefenamic acid; Meclofenamic acid; Flufenamic acid; Tolfenamic acid; Celecoxib; Clonixin.

In a particular embodiment, the invention relates to i) a NLRP3 inhibitor and ii) an immunotherapy treatment used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof.

As used herein, the term “immunotherapy” has its general meaning in the art and refers to the treatment that consists in administering an immunogenic agent i.e. an agent capable of inducing, enhancing, suppressing or otherwise modifying an immune response.

In a further embodiment, the invention relates to i) a NLRP3 inhibitor and ii) a neutralizing monoclonal anti-IL-Ib antibody used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof.

As used herein, the term “a neutralizing monoclonal anti-IL-Ib antibody” refers to an antibody that blocks or reduces at least one activity of a polypeptide comprising the epitope to which the antibody specifically binds. The neutralizing antibody reduces IL-Ib biological activity in in cellulo and/or in vivo tests. In the context of the invention, the neutralizing monoclonal anti-IL-Ib antibody is canakinumab (trade name Ilaris, developed by Novartis).

In a particular embodiment, the invention relates i)a NLRP3 inhibitor and ii) a recombinant human IL-1B receptor antagonist used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof.

As used herein, the term “a recombinant human IL-1B receptor antagonist” refers to an inhibitor which inhibits the activity of IL-la and IL-Ib by competitively blocking their binding to type I and type II receptors. IL-1RA is produced by corneal epithelial cells, monocytes, neutrophils, macrophages, and fibroblasts. In the context of the invention, the recombinant human IL-1B receptor antagonist is Anakinra (marketed as Kineret® by Swedish Orphan Biovitru).

In a particular embodiment, the invention relates i) a NLRP3 inhibitor and ii) a PAK-1 and/or PAK-2 inhibitor used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof.

As used herein, the term "PAK-1" has its general meaning in the art and refers to P21- Activated Kinase 1, also known as Serine/threonine-protein kinase PAK-1, or P21 protein (Cdc42/Rac)-activated kinase 1. PAK-1 is a member of p21 -activated kinases family (PAKs) involved in the ERK activation, MAPK pathway activation and that are critical effectors that link the Rho GTPases to cytoskeleton reorganization and nuclear signaling and have been implicated in a wide range of biological activities.

As used herein, the term "PAK-1 inhibitor" refers to any compound that is able to inhibit the activity or expression of PAK-1. In particular the PAK-1 inhibitor inhibits the kinase activity of PAK-1. Typically, the PAK-1 inhibitor blocks PAK-1 interaction with proteins involved in ERK pathway and MAPK pathway such as RAF-1 (CRAF), inhibits its phosphorylation, or blocks MAPK cascade. The term "PAK-1 antagonist" refers to a compound that selectively blocks or inactivates PAK-1. As used herein, the term "selectively blocks or inactivates" refers to a compound that preferentially binds to and blocks or inactivates PAK-1 with a greater affinity and potency, respectively, than its interaction with the other sub-types or isoforms of the PAKs family.

Example of PAK-1 inhibitors include the compounds described in W02004007504, W02006072831, W02007023382, W02007072153, W02009086204, W02010071846, WO20 11044264, WO2011044535, WO2011156640, WO2011156646, WO2011156775, WO201 1156780, WO2011156786, and WO 2013026914. Additional examples of PAK-1 inhibitors include, but are not limited to, staurosporine, 3 -hydroxy staurosporine, K252a, CEP-1347, OSU-03012, DW12, FL172 (disclosed in Yi et al., Biochemical Pharmacology, 2010, 80:683-689, the disclosure of which with respect to PAK-1 inhibitor compounds is hereby incorporated herein by reference), IP A3 (commercially available from Tocris), PF-3758309, PAK10 (available from Calbiochem), EKB569, TKI258, FRAX- 597 (available from Tocris) and SU-14813. In some embodiments, the PAK-1 inhibitor is a macrocyclic lactone. As used herein, the term "macrocyclic lactones" has its general meaning in the art and refers to macrocyclic lactones and macrocyclic lactones derivatives described in Lespine A. Lipid-like properties and pharmacology of the anthelmintic macrocyclic lactones. Expert Opin Drug Metab Toxicol. 2013 Dec; 9(12): 1581-95. Macrocyclic lactones, like ivermectin, are capable of inhibiting PAK-1 activity (e.g. HASMIMOTO ET AL: "Ivermectin inactivates the kinase PAK-1 and blocks the PAK-1 dependent growth of human ovarian cancer and NF2 tumor cell lines", DRUG DISCO V. THERAPEUTICS, vol. 3, no. 6, 2009, - 2009, pages 243-246). Examples of macrocyclic lactones include those described in WO 2012078605, WO 2012150543, WO2011075592, W0199316189, and WO2012028556.

In some embodiments, examples of macrocyclic lactones include but are not limited to Ivermectin (Stromectol), Doramectin, Selamectin, Moxidectin, Milbemycin, Abamectin, Nemadectin and Eprinomectin. In a particular embodiment, the inhibitor of PAK-1 is AZ13711265. AZ13711265 is well known in the art, its CAS number is 2016806-55-4 and has the following chemical formula and structure in the art C28H35FN603S:

In a particular embodiment, the invention relates i) a NLRP3 inhibitor and ii) a caspase- 1 inhibitor used as a combined preparation for treating SARS-CoV-2 infection in a subject in need thereof.

In another embodiment, the inhibitor is a Caspase-1 inhibitor. The Caspase-1 inhibitor may be a direct inhibitor of Caspase-1 enzymatic activity, or may be an indirect inhibitor that inhibits initiation of inflammasome assembly or inflammasome signal propagation. Caspase-1 inhibitors for use in the present invention may be antioxidants, including reactive oxygen species (ROS) inhibitors. Examples of such Caspase-1 inhibitors include, but are not limited to, flavonoids including flavones such as apigenin, luteolin, and diosmin; flavonols such as myricetin, fisetin and quercetin; flavanols and polymers thereof such as catechin, gallocatechin, epicatechin, epigallocatechin, epigallocatechin-3- gallate and theaflavin; isoflavone phytoestrogens; and stilbenoids such as resveratrol. Also included are phenolic acids and their esters such as gallic acid and salicyclic acid; terpenoids or isoprenoids such as andrographolide and parthenolide; vitamins such as vitamins A, C and E; vitamin cofactors such as co-enzyme Q10, manganese and iodide, other organic antioxidants such as citric acid, oxalic acid, phytic acid and alpha-lipoic acid, and Rhus verniciflua stokes extract. The Caspase-1 inhibitor may be a combination of these compounds, for example, a combination of a-lipoic acid, co-enzyme Q10 and vitamin E, or a combination of a Caspase 1 inhibitor(s) with another inflammasome inhibitor such as glyburide or a functionally equivalent precursor or derivative thereof. The Caspase-1 inhibitor may be a small molecule inhibitor, as one of skill in the art will appreciate. Non-limiting examples include cyanopropanate-containing molecules such as (S)-3-((S)-l- ((S)-2-(4-amino-3-chlorobenzamido)-3,3-dimethylbutanoyl)pyrr olidine-2-carboxamido)-3- cyano- propanoic acid, as well as other small molecule caspase-1 inhibitors such as (S)-1-((S)- 2-{[l-(4-amino- 3 -chloro-phenyl)-methanoyl] -amino } -3 ,3 -dimethyl-butanoyl)-pyrrolidine- 2-carboxylic acid ((2R,3 S)- 2-ethoxy-5-oxo-tetrahydro-furan-3-yl)-amide. Such inhibitors may be chemically synthesized.

As used herein, the terms “combined treatment”, “combined therapy” or “therapy combination” refer to a treatment that uses more than one medication. The combined therapy may be dual therapy or bi-therapy.

In a particular embodiment, i) a NLRP3 inhibitor and ii) a classical treatment as a combined preparation according to the invention for simultaneous, separate or sequential use in the treatment of SARS-CoV-2 infection in a subject in need thereof.

In a particular embodiment, the invention relates to i) a NLRP3 inhibitor and ii) antiviral treatment used as a combined preparation according to the invention for simultaneous, separate or sequential use in the treatment of SARS-CoV-2 infection in a subject in need thereof.

In a particular embodiment, the invention relates to i) a NLRP3 inhibitor and ii) an antibiotic used as a combined preparation according to the invention for simultaneous, separate or sequential use in the treatment of SARS-CoV-2 infection in a subject in need thereof.

In a particular embodiment, the invention relates to i) a NLRP3 inhibitor and ii) an anti- parasitic treatment used as a combined preparation according to the invention for simultaneous, separate or sequential use in the treatment of SARS-CoV-2 infection in a subject in need thereof. In a particular embodiment, i) a NLRP3 inhibitor and ii) a neutralizing monoclonal anti- IL-Ib antibody as a combined preparation according to the invention for simultaneous, separate or sequential use in the treatment of SARS-CoV-2 infection in a subject in need thereof.

In a particular embodiment, i) a NLRP3 inhibitor and ii) recombinant human IL-1B receptor antagonist as a combined preparation according to the invention for simultaneous, separate or sequential use in the treatment of SARS-CoV-2 infection in a subject in need thereof.

In a particular embodiment, i) a NLRP3 inhibitor and ii) a PAK-1 and/or PAK-2 inhibitor as a combined preparation according to the invention for simultaneous, separate or sequential use in the treatment of SARS-CoV-2 infection in a subject in need thereof.

In a particular embodiment, i) a NLRP3 inhibitor and ii) a caspase-1 inhibitor as a combined preparation according to the invention for simultaneous, separate or sequential use in the treatment of SARS-CoV-2 infection in a subject in need thereof.

As used herein, the term “administration simultaneously” refers to administration of 2 active ingredients by the same route and at the same time or at substantially the same time. The term “administration separately” refers to an administration of 2 active ingredients at the same time or at substantially the same time by different routes. The term “administration sequentially” refers to an administration of 2 active ingredients at different times, the administration route being identical or different.

Pharmaceutical composition:

The NLRP3 inhibitor for use according to the invention alone and/or combined with NLRP3 inhibitor and classical treatment as described above may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form pharmaceutical compositions.

Accordingly, in a further aspect, the invention relates to a pharmaceutical composition comprising a NLRP3 inhibitor for treating SARS-CoV-2 infection in a subject in need thereof.

In a particular embodiment, the pharmaceutical composition according the invention, wherein the NLRP3 inhibitor is MCC950.

In a particular embodiment, the pharmaceutical composition according the invention, wherein the NLRP3 inhibitor is colchicine.

In a particular embodiment, the pharmaceutical composition according the invention comprising i) a NLRP3 inhibitor and ii) a classical treatment. In a particular embodiment, the pharmaceutical composition according the invention comprising i) a NLRP3 inhibitor and ii) an antiviral treatment.

In a particular embodiment, the pharmaceutical composition according the invention comprising i) a NLRP3 inhibitor and ii) an antibiotic.

In a particular embodiment, the pharmaceutical composition according the invention comprising i) a NLRP3 inhibitor and ii) an anti-parasitic drug.

In a particular embodiment, the pharmaceutical composition according the invention comprising i) a NLRP3 inhibitor and ii) immunosuppressive corticosteroids.

In a particular embodiment, the pharmaceutical composition according the invention comprising i) a NLRP3 inhibitor and ii) non-steroidal drug.

In a particular embodiment, the pharmaceutical composition according the invention comprising i) a NLRP3 inhibitor and ii) a neutralizing monoclonal anti-IL-Ib antibody.

In a particular embodiment, the pharmaceutical composition according the invention comprising i) a NLRP3 inhibitor and ii) a recombinant human IL-1B receptor antagonist.

In a particular embodiment, the pharmaceutical composition according the invention comprising i) a NLRP3 inhibitor and ii) PAK-1 and/or PAK-2 inhibitor.

In a particular embodiment, the pharmaceutical composition according the invention comprising i) a NLRP3 inhibitor and ii) a caspase-1 inhibitor.

As used herein, the terms "pharmaceutically" or "pharmaceutically acceptable" refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate. A pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. The pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, transdermal, local or rectal administration, the active principle, alone or in combination with another active principle, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings. Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms. Typically, the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected. These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. The polypeptide (or nucleic acid encoding thereof) can be formulated into a composition in a neutral or salt form. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. The carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin. Sterile injectable solutions are prepared by incorporating the active polypeptides in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed. For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intrap eritoneal administration. In this connection, sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage could be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.

In a particular embodiment, pharmaceutical composition according to the invention is suitable for a nasal formulation.

Method for screening:

In a further aspect, the invention relates to a method of screening a drug suitable for the treating SARS-CoV-2 infection comprising i) providing a test compound and ii) determining the ability of said test compound to inhibit the expression or activity of NLRP3.

Any biological assay well known in the art could be suitable for determining the ability of the test compound to inhibit the activity or expression of NLRP3. In some embodiments, the assay first comprises determining the ability of the test compound to bind to NLRP3. In some embodiments, a population of cells then contacted and activated so as to determine the ability of the test compound to inhibit the activity or expression of NLRP3. In particular, the effect triggered by the test compound is determined relative to that of a population of immune cells incubated in parallel in the absence of the test compound or in the presence of a control agent either of which is analogous to a negative control condition. The term "control substance", "control agent", or "control compound" as used herein refers a molecule that is inert or has no activity relating to an ability to inhibit a biological activity or expression. It is to be understood that test compounds capable of inhibiting the activity or expression of NLRP3, as determined using in vitro methods described herein, are likely to exhibit similar modulatory capacity in applications in vivo. Typically, the test compound is selected from the group consisting of peptides, petptidomimetics, small organic molecules, antibodies (e.g. intraantibodies), aptamers or nucleic acids. For example the test compound according to the invention may be selected from a library of compounds previously synthesised, or a library of compounds for which the structure is determined in a database, or from a library of compounds that have been synthesised de novo.

The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.

FIGURES:

Figure 1. (A-C) Whole peripheral blood cells of COVID-19 patients were treated with MCC950 (IOmM), Colchicine (IOmM), AZ13711265 (ImM) or IPA-3 (ImM) for 3h. The indicated subsets were analyzed for the percent of FAM-FLICA positive cells.

EXAMPLE:

Material & Methods

The whole blood of control or COVID-19 patients is incubated with or without NLRP3 inhibitors at 37°C for 3h. Leukocytes are isolated and the caspase-1 activity measured using FAM-FLICA probe using flow cytometry and IL-lbeta cytokines secretion are measured in the serum. Nigericin, a bona fide NLRP3 activator, is used as a positive control.

Results

Whole peripheral blood cells of COVID-19 patients were treated with MCC950 (IOmM), Colchicine (IOmM), AZ13711265 (ImM) or IPA-3 (ImM) for 3h. Samples were stained for active caspase-1 (detected using the FAM-FLICA probe) and for CD45, CD14, CD 16, and CD66b markers. Cells were immunophenotyped by flow cytometry. Leukocytes were defined as CD45+ and were analyzed for granulocyte and monocytes surface markers. Granulocytes were defined as CD66b+ and the different subsets were gated as indicated using CD66b and CD 16 markers (Figures 1A to 1C). Inhibition of the FAM-FLICA signal by NLRP3 inhibitors indicates the potential of using these drugs to block the NLRP3 triggered IL-lbeta secretion.

Thus, inventors demonstrate that targeting NLRP3 allows to inhibit the IL-lbeta secretion and thus to prevent storm cytokine.

REFERENCES:

Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.