Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOUNDS INDUCING PRODUCTION OF PROTEINS BY IMMUNE CELLS
Document Type and Number:
WIPO Patent Application WO/2023/203161
Kind Code:
A1
Abstract:
The present invention relates to a compound of formula (I) and its use for inducing the production of interleukin-10 (IL-10) by immune cells. The invention also relates to induced immune cells capable of producing interleukin 10. The invention also relates to the use of the induced immune cells in the prevention and/or treatment of immune-mediated diseases.

Inventors:
FILLATREAU SIMON (FR)
MANFROI BENOÎT (FR)
MAHUTEAU-BETZER FLORENCE (FR)
BEAUVINEAU CLAIRE (FR)
DANG VAN DUC (DE)
Application Number:
PCT/EP2023/060353
Publication Date:
October 26, 2023
Filing Date:
April 20, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV PARIS CITE (FR)
CENTRE NAT RECH SCIENT (FR)
INST NAT SANTE RECH MED (FR)
DEUTSCHES RHEUMA FORSCHUNGSZENTRUM BERLIN (DE)
INST CURIE (FR)
UNIV PARIS SACLAY (FR)
International Classes:
A61K31/4184; A61P29/00; A61P37/00; C07D235/18; C12N5/071
Domestic Patent References:
WO2012068406A22012-05-24
WO2014208354A12014-12-31
WO2012102937A22012-08-02
WO2018191146A12018-10-18
WO2021242790A12021-12-02
WO2011046954A12011-04-21
WO2012064569A12012-05-18
Foreign References:
JP2002318231A2002-10-31
US3705944A1972-12-12
Other References:
DATABASE Reaxys [online] Elsevier; 1 January 2022 (2022-01-01), ANONYMOUS: "Extract from Database: R1=R9=H, X3=C", XP093052600, Database accession no. -
TABARES-GUEVARA JORGE H. ET AL: "IL-10-Dependent Amelioration of Chronic Inflammatory Disease by Microdose Subcutaneous Delivery of a Prototypic Immunoregulatory Small Molecule", FRONTIERS IN IMMUNOLOGY, vol. 12, 8 July 2021 (2021-07-08), XP055962018, DOI: 10.3389/fimmu.2021.708955
RICHARDS M L ET AL: "Substituted 2-phenyl-benzimidazole derivatives: novel compounds that suppress key markers of allergy", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 41, no. 8, 1 August 2006 (2006-08-01), pages 950 - 969, XP024993949, ISSN: 0223-5234, [retrieved on 20060801], DOI: 10.1016/J.EJMECH.2006.03.014
DATABASE GEPRIS [online] DFG; 1 January 2018 (2018-01-01), FILLATREAU SIMON: "Characterization of IL-10-producing B cells and their suppressive activities during primary and secondary Salmonella typhimurium infection", XP055962035, retrieved from https://gepris.dfg.de/gepris/projekt/116594012 Database accession no. 116594012
THOMAS B SUNDBERG ET AL: "Small-molecule control of cytokine function: new opportunities for treating immune disorders", CURRENT OPINION IN CHEMICAL BIOLOGY, vol. 23, 1 December 2014 (2014-12-01), GB, pages 23 - 30, XP055336951, ISSN: 1367-5931, DOI: 10.1016/j.cbpa.2014.08.013
SARAIVA MARGARIDA ET AL: "The regulation of IL-10 production by immune cells", vol. 10, no. 3, 1 March 2010 (2010-03-01), London, pages 170 - 181, XP055962471, ISSN: 1474-1733, Retrieved from the Internet DOI: 10.1038/nri2711
SHUBIN KIRILL M ET AL: "Synthesis of benzo[4,5]imidazo[2,1-a]phthalazines", TETRAHEDRON LETTERS, vol. 45, no. 7, 9 February 2004 (2004-02-09), pages 1407 - 1408, XP029594162, ISSN: 0040-4039, DOI: 10.1016/J.TETLET.2003.12.070
YANG DAOSHAN ET AL: "Magnetically recoverable and reusable CuFe 2 O 4 nanoparticle-catalyzed synthesis of benzoxazoles, benzothiazoles and benzimidazoles using dioxygen as oxidant", RSC ADV., vol. 4, no. 34, 1 January 2014 (2014-01-01), pages 17832 - 17839, XP055960087, Retrieved from the Internet DOI: 10.1039/C4RA00559G
KUZNETSOV V A ET AL: "Efficient route to benzo[4,5]imidazo[2,1-a]phthalazines", TETRAHEDRON, ELSEVIER SIENCE PUBLISHERS, AMSTERDAM, NL, vol. 62, no. 42, 16 October 2006 (2006-10-16), pages 10018 - 10030, XP025002774, ISSN: 0040-4020, [retrieved on 20061016], DOI: 10.1016/J.TET.2006.07.089
ROWE F M ET AL: "A Reaction of Certain Diazosulphonates derived from beta-Naphthol-1-sulphonic Acid. Part X VII. Conversion of Nitro-3-aryl- and Nitro-3-aryl-4-methyl-phthalaz-1-ones into Corresponding Phthalaz-4-ones by Migration of the Nitroaryl Group, and Related Reactions.", JOURNAL OF THE CHEMICAL SOCIETY, 1 January 1937 (1937-01-01), pages 90 - 109, XP093052432, Retrieved from the Internet
G. KULKARNI RAVINDRA ET AL: "Synthesis, p38 Kinase Inhibitory and Anti-inflammatory Activity of New Substituted Benzimidazole Derivatives", MEDICINAL CHEMISTRY, vol. 9, no. 1, 1 February 2013 (2013-02-01), NL, pages 91 - 99, XP055824635, ISSN: 1573-4064, DOI: 10.2174/157340613804488440
HAKAN GÖKER ET AL: "Synthesis and In Vitro Activity of Polyhalogenated 2-phenylbenzimidazoles as a New Class of anti-MRSA and Anti-VRE Agents", CHEMICAL BIOLOGY & DRUG DESIGN, BLACKWELL MUNKSGAARD, HOBOKEN, USA, vol. 87, no. 1, 31 August 2015 (2015-08-31), pages 57 - 68, XP072380181, ISSN: 1747-0277, DOI: 10.1111/CBDD.12623
ARSHAD TANZILA ET AL: "Syntheses, in vitro evaluation and molecular docking studies of 5-bromo-2-aryl benzimidazoles as[alpha]-glucosidase inhibitors", MEDICINAL CHEMISTRY RESEARCH, BIRKHAEUSER, BOSTON, US, vol. 25, no. 9, 22 July 2016 (2016-07-22), pages 2058 - 2069, XP036063403, ISSN: 1054-2523, [retrieved on 20160722], DOI: 10.1007/S00044-016-1614-Y
"Remington's Pharmaceutical Sciences", 1980
JORDAO ET AL., SCIENCE, vol. 363, 2019, pages 6425
Attorney, Agent or Firm:
CABINET BEAU DE LOMENIE (FR)
Download PDF:
Claims:
CLAIMS 1. A compound of formula (I): wherein: - X1, X2 and X3 are each independently a nitrogen atom or a carbon atom; - R1 and R9 are each a hydrogen atom, when the double dotted line between R1 and R9 and the simple dotted line between R9 and Z1 are nothing; - or R1 is a nitrogen atom and R9 is a carbon atom, when the double dotted line is a double bond and the simple dotted line between R9 and Z1 is a simple bond, and Z1 is a (C1-C6)alkyl group or a phenyl group optionally substituted with a halogen atom or a (C1- C6)alkyl group; - R2 and R3 are each independently selected from: a hydrogen atom, a halogen atom, and a group selected from:

wherein: - Z3 is a hydrogen atom, a –COOH group, or a -NO2 group; - X4 is a carbon atom or a nitrogen atom; - Z2 is a hydroxyl group or a hydrogen atom; - q is selected from 0, 1, 2, 3, 4 and 5, and; - each Z4 is selected from a halogen atom, a (C1-C6)alkyl group, a (C1-C6)alkoxy group, an hydroxyl group, a –NH2 group, a –NHCOCH3 group, a -NO2 group, an alkylether group, a –SCH3 group, a -SO2N(CH3)2 group, a -SO2CH(CH3)2 group, a –SO2NHCH(CH3)2 group, a –NHCOOCH3 group, a -SOCH2CH3 group, and a –CH2NHCOCH3 group; - R4, R5, R6 and R10 are each independently a hydrogen atom or a halogen atom; - R8 is absent, when X2 is a nitrogen atom or R8 is a hydrogen atom, a halogen atom or a hydroxyl group, when X2 is a carbon atom; - R7 is absent, when X3 is a nitrogen atom, or R7 is selected from: a hydrogen atom, a halogen atom, a –OH group, a –OCH3 group, and a group selected from: wherein: - p is selected from 0, 1, 2, 3, 4 and 5 ; - i is selected from 0, 1, 2, 3 and 4; - Z5 is selected from a halogen atom, a (C1-C6)alkyl group, a (C1-C6)alkoxy group, a hydroxyl group, a –NH2 group, a –NHCOCH3 group, a - NO2 group and a –N(CH3)2 group; - Z6 is a hydrogen atom, a –COOH group or a -NO2 group; - Z8 is a hydrogen atom or a –OH group; - Z9 is a hydrogen atom or a halogen atom; - X5 is a nitrogen atom or a carbon atom; provided that:

(i) R3 and R7 cannot simultaneously represent a group and a group

(ii) R3 and R7 cannot simultaneously represent a group and a group (iii) the compound of formula (I) is not one of the following compounds:

N-(2-(3-fluorophenyl)-1 H-benzo[d]imidazol-6-yl)benzamide;

N-[2-(2-Chlorophenyl)-1 H-benzimidazol-6-yl]-2-(4-morpholinyl)benzamide;

6-bromo-2-(2-fluorophenyl)-1 H-benzimidazole;

N-[5-(4-Methylphenyl)benzimidazo[2,1 -a]phthalazin-10-yl]benzamide; 2-Chloro-N-[5-(4-methylphenyl)benzimidazo[2,1 -a]phthalazin-10-yl]benzamide;

2-lodo-N-[5-(4-methylphenyl)benzimidazo[2,1 -a]phthalazin-10-yl]benzamide;

2-Methoxy-N-[5-(4-methylphenyl)benzimidazo[2,1 -a]phthalazin-10-yl]benzamide. 2. The compound of claim 1 , wherein:

- X1 ; X2, X3 are a carbon atom;

- Ri is a hydrogen atom;

- R2 and R3 are each independently selected from a hydrogen atom, a halogen atom, and a group selected from: wherein:

- q is selected from 0, 1 , 2, 3 , 4 and 5;

- each Z4 is selected from a hydrogen atom, a halogen atom, a (Ci-C6)alkoxy group, an alkylether group, a -SCH3 group, and a -SO2N(CH3)2 group ;

- R4, R5, Re, R?, Rs, Rg and Rw are each independently a hydrogen atom or a halogen atom.

3. The compound of claim 1 , which is selected from: . The compound of claim 1 , which is selected from:

5. The compound of claim 1 , which is selected from: 6. The compound of claim 1 , which is:

7. The compound of claim 1 , wherein:

- X1 ; X2, X3are a carbon atom; - Ri is a hydrogen atom; - R2, and R3 are each independently selected from a hydrogen atom, a halogen atom, wherein :

-q is selected from 0, 1 , 2, 3, 4 and 5;

- Z4 is a - NH2 group or a -NHCOCH3 group;

- X4 is a carbon atom and Z2 is a hydroxyl group;

- R7 is selected from: a hydrogen atom, a halogen atom, wherein:

- p is selected from 0, 1 , 2, 3, 4 and 5;

- i is 0;

- each Z5 is independently a -NH2 group or a -NHCOCH3 group;

- X5 is a carbon atom and Z8 is a hydroxyl group;

- R4, R5, R6, R8, R9 and Rw are each independently a hydrogen atom or a halogen atom.

8. The compound of claim 1 , wherein:

- Xj and X3 are a carbon atom;

- X2 is a nitrogen atom;

- Ri is a hydrogen atom;

- R4, R5, R6, R7, R9 and Rw are a hydrogen atom;

- R2 and R3 are each independently selected from a hydrogen atom, wherein q is selected from 0, 1 , 2, 3, 4 and 5, and Z4 is (Ci-C6) alkoxy group.

9. The compound of claim 1 or claim 8, which is selected from:

10. The compound of claim 1 , wherein:

- Xb X2, X3 are a carbon atom;

- Ri is a hydrogen atom;

- R2, R3, R4, R5, R6, R?, Rs, Rg and Rw are each independently a hydrogen atom or a halogen atom.

11. The compound of claim 1 , wherein:

- X1 ; X2, X3 are a carbon atom;

- Ri is a nitrogen atom and R9 is a carbon atom;

- the double dotted line is a double bond;

- the simple dotted line between R9 and ZA is a simple bond;

- ZA is a (Ci-C6)alkyl group, or a phenyl group optionally substituted with a halogen atom;

- R2, R are a hydrogen atom;

- R3 is: wherein:

- q is selected from 0, 1 , 2 , 3 , 4 and 5; and

- each Z4 is independently a halogen atom or a (Ci-C6)alkoxy group. 12. An in vitro method for inducing the production of a protein by immune cells, the method comprising contacting immune cells with a compound of formula (I), whereby induced immune cells are obtained:. wherein: - X1 ; X2 and X3 are each independently a nitrogen atom or a carbon atom;

- Ri and R9 are each a hydrogen atom, when the double dotted line between Ri and R9 and the simple dotted line between R9 and ZA are nothing;

- or R, is a nitrogen atom and R9 is a carbon atom, when the double dotted line is a double bond and the simple dotted line between R9 and ZA is a simple bond, and ZA is a (Ci-C6)alkyl group or a phenyl group optionally substituted with a halogen atom or a (Cr C6)alkyl group;

- R2 and R3 are each independently selected from: a hydrogen atom, a halogen atom, and a group selected from:

wherein:

- Z3 is a hydrogen atom, a -COOH group, or a -NO2 group; - X4 is a carbon atom or a nitrogen atom;

- Z2 is a hydroxyl group or a hydrogen atom;

- q is selected from 0, 1 , 2, 3, 4 and 5, and;

- each Z4 is selected from a halogen atom, a (Ci-C6)alkyl group, a (Ci-C6)alkoxy group, an hydroxyl group, a -NH2 group, a -NHCOCH3 group, a -NO2 group, an alkylether group, a -SCH3 group, a -SO2N(CH3)2 group, a -SO2CH(CH3)2 group, a -SO2NHCH(CH3)2 group, a -NHCOOCH3 group, a - SOCH2CH3 group, and a -CH2NHCOCH3 group;

- R4, R5, R6and Rw are each independently a hydrogen atom or a halogen atom;

- R8 is absent, when X2 is a nitrogen atom or R8 is a hydrogen atom, a halogen atom or a hydroxyl group, when X2 is a carbon atom; - R7 is absent, when X3 is a nitrogen atom, or R7 is selected from: a hydrogen atom, a halogen atom, a -OH group, a -OCH3 group, and a group selected from: wherein: - p is selected from 0, 1 , 2, 3, 4 and 5 ;

- i is selected from 0, 1 , 2, 3 and 4;

- Z5 is selected from a halogen atom, a (Ci-C6)alkyl group, a (Ci-C6)alkoxy group, a hydroxyl group, a -NH2 group, a -NHCOCH3 group, a - NO2 group and a -N(CH3)2 group;

- Z6 is a hydrogen atom, a -COOH group or a -NO2 group; - Z8 is a hydrogen atom or a -OH group;

- Z9 is a hydrogen atom or a halogen atom;

- X5 is a nitrogen atom or a carbon atom.

13. The method of claim 12, wherein in formula (I): - Xi, X2, X3 are a carbon atom;

- Ri is a hydrogen atom;

- R2 and R3 are each independently selected from a hydrogen atom, a halogen atom, and a group selected from:

wherein: - q is selected from 0, 1, 2, 3 , 4 and 5; - each Z4 is selected from a hydrogen atom, a halogen atom, a (C1-C6)alkoxy group, an alkylether group, a –SCH3 group, and a -SO2N(CH3)2 group ; - R4, R5, R6, R7, R8, R9 and R10 are each independently a hydrogen atom or a halogen atom. 14. The method of claim 12, wherein the compound of formula (I) is selected from:

15. The method of claim 12, wherein the compound of formula (I) is selected from:

16. The method of claim 12, wherein the compound of formula (I) is selected from: preferably the compound of formula (I) is

17. The method of claim 12, wherein in formula (I):

- X1 ; X2, X3 are a carbon atom; - Ri is a hydrogen atom;

- R2 and R3 are each independently selected from: a hydrogen atom, a halogen atom, wherein:

- q is selected from 0, 1 , 2, 3, 4 and 5; - each Z4 is independently a -NH2 group or a -NHCOCH3 group;

- X4 is a carbon atom and Z2 is a hydroxyl group;

- R7 is selected from: a hydrogen atom, a halogen atom,

wherein:

- p is selected from 0, 1 , 2, 3, 4 and 5;

- i is 0; - each Z5 is independently a -NH2 group or a -NHCOCH3 group;

- X5 is a carbon atom and Z8 is a hydroxyl group;

- R4, Rs, Re, Rs, R9 and Rw are each independently a hydrogen atom or a halogen atom.

18. The method of claim 12, wherein the compound of formula (I) is selected from:

19. The method of claim 12, wherein in formula (I):

- Xi and X3 are a carbon atom,

- X2 is a nitrogen atom;

- R1 is a hydrogen atom; - R4, R5, R6, R7, R9 and Rw are a hydrogen atom;

- R2 and R3 are each independently selected from a hydrogen atom, wherein q is selected from 0, 1 , 2, 3 , 4 and 5 and Z4 is a (Ci-C6)alkoxy group.

20. The method of claim 12, wherein the compound of formula (I) is selected from:

21 . The method of claim 12, wherein in formula (I):

- Xi, X2, X3 are a carbon atom;

- R1 is a hydrogen atom;

- R2, R3, R4, R5, R6, R7, RS, R9 and Rw are each independently a hydrogen atom or a halogen atom.

22. The method of claim 1 , wherein in formula (I):

- Xi, X2, X3 are a carbon atom;

- R1 is a nitrogen atom and R9 is a carbon atom;

- the double dotted line is a double bond;

- the simple dotted line between R9 and ZA is a simple bond;

- ZA is a (Ci-C6)alkyl group, or a phenyl group optionally substituted with a halogen atom;

- R2, R4, R5, R6, R7, R8, and Rw are a hydrogen atom;

- q is selected from 0, 1 , 2 , 3 , 4 and 5; and

- each Z4 is independently a halogen atom or a (Ci-C6)alkoxy group.

23. The method of claim 12, wherein the compound of formula (I) is selected from: preferably the compound of formula (I) is

24. The method of any one of claims 12 to 23, wherein the immune cells are T lymphocytes, B lymphocytes, dendritic cells, natural killer cells, innate lymphoid cells, mesenchymal cells or myeloid cells, in particular the immune cells are B lymphocytes. 25. The method of any one of claims 12 to 24, wherein the protein is interleukin-10.

26. Induced immune cells obtained by the method of claim 25.

27. A pharmaceutical composition comprising a dose of induced immune cells according to claim 26, and at least one pharmaceutically acceptable carrier.

28. The pharmaceutical composition of claim 27, wherein the pharmaceutical composition further comprises at least one other active ingredient.

29. The pharmaceutical composition of claim 27 or claim 28 for use in the prevention or treatment of an immune-mediated disease.

30. The pharmaceutical composition for use according to claim 39, wherein the immune-mediated disease is selected from: a neurodegenerative disease, an inflammatory disease, an allergic disease, an adverse event related to transplantation, an autoimmune disease, a metabolic disease with an inflammatory component, inflammation facilitating cancer onset or progression, and aging.

31. A compound of formula (I) as defined in any one of claims 12 to 23 for use in a method of treatment of an immune-mediated disease, the method comprising (i) contacting immune cells with said compound of formula (I), whereby induced immune cells are obtained, and (ii) administering the induced immune cells to a patient in need thereof.

Description:
COMPOUNDS INDUCING PRODUCTION OF PROTEINS BY IMMUNE CELLS

FIELD OF THE INVENTION

The present invention relates to compounds inducing production of proteins, in particular interleukin-10 (IL-10) by immune cells and its use in the prevention and/or treatment of immune-mediated diseases, in particular neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer or progression, and aging.

BACKGROUND OF THE INVENTION

Immune-mediated diseases are chronic inflammatory diseases perpetuated by antibodies and cellular immunity. The immune response damages healthy organs either inadvertently as a result of attacking foreign ingredients that have entered the body, or by attacking self- tissues, a process called autoimmunity. These diseases include many forms of arthritis (e.g., rheumatoid arthritis and psoriatic arthritis), inflammatory bowel diseases (e.g., ulcerative colitis and Crohn's disease), endocrinopathies (e.g., type 1 diabetes and Graves disease), neurodegenerative diseases (e.g., multiple sclerosis, autistic spectrum disorder, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), Parkinson's disease, Huntington's Disease, Guillain-Barre syndrome, myasthenia gravis, and chronic idiopathic demyelinating disease (CID)), and vascular diseases (e.g., autoimmune hearing loss, systemic vasculitis, and atherosclerosis). These diseases are common and have a major socioeconomic impact.

Currently, the primary focus of therapy is to suppress immunity and inhibit inflammation, by using immunosuppressive drugs, blockers of cytokine or cytokine receptor signaling, or by the transient depletion of leukocyte subsets. However, these approaches do not take into account the need to restore dominant immune regulation, which is often defective in these pathologies, to restore a healthy and stable immune system. A large fraction of patients treated by these drugs do not respond, or become resistant after some time, or relapse upon treatment cessation.

Thus, there is a need for new effective forms of therapy, in particular treatments that may provide a controlled, sustained therapy, offered alone or in combination with other therapies, whilst improving patient quality of life and reducing side effects related to treatment.

To prevent and/or treat immune-mediated diseases, the inventors have developed a new compound inducing production of proteins, in particular interleukin-10 (IL-10), by immune cells, enabling the generation of IL-10-producing immune cells, which can be used in adoptive cell therapy. SUMMARY OF THE INVENTION The present invention provides a compound inducing production of proteins, in particular interleukin-10 (IL-10) by immune cells of formula (I): wherein: - X 1 , X 2 and X 3 are each independently a nitrogen atom or a carbon atom; - R 1 and R 9 are each a hydrogen atom, when the double dotted line between R 1 and R 9 and the simple dotted line between R 9 and Z 1 are nothing; - or R 1 is a nitrogen atom and R 9 is a carbon atom, when the double dotted line is a double bond and the simple dotted line between R 9 and Z 1 is a simple bond, and Z 1 is a (C 1 -C 6 )alkyl group or a phenyl group optionally substituted with a halogen atom or a (C 1 - C 6 )alkyl group; - R 2 and R 3 are identical or different, are each independently selected from: a hydrogen atom, a halogen atom, and a group selected from:

, wherein: - Z 3 is a hydrogen atom, a –COOH group, or a -NO 2 group; - X 4 is a carbon atom or a nitrogen atom; - Z 2 is a hydroxyl group or a hydrogen atom; - q is selected from 0, 1, 2, 3, 4 and 5, and; - each Z 4 is selected from a halogen atom, a (C 1 -C 6 )alkyl group, a (C 1 -C 6 )alkoxy group, an hydroxyl group, a –NH 2 group, a –NHCOCH 3 group, a -NO 2 group, an alkylether group, a –SCH 3 group, a -SO 2 N(CH 3 ) 2 group, a -SO 2 CH(CH 3 ) 2 group, a –SO 2 NHCH(CH 3 ) 2 group, a –NHCOOCH 3 group, a - SOCH 2 CH 3 group, and a –CH 2 NHCOCH 3 group; - R 4, R 5, R 6 and R 10 are each independently a hydrogen atom or a halogen atom; - R 8 is absent, when X 2 is a nitrogen atom or R 8 is a hydrogen atom, a halogen atom or a hydroxyl group, when X 2 is a carbon atom; - R 7 is absent, when X 3 is a nitrogen atom or R 7 is selected from: a hydrogen atom, a halogen atom, a –OH group, a –OCH 3 group, and a group selected from: wherein: - p is selected from 0, 1, 2, 3, 4 and 5 ; - i is selected from 0, 1, 2, 3 and 4; - Z 5 is selected from a halogen atom, a (C 1 -C 6 )alkyl group, a (C 1 -C 6 )alkoxy group, a hydroxyl group, a –NH 2 group, a –NHCOCH 3 group, a - NO 2 group and a –N(CH 3 ) 2 group; - Z 6 is a hydrogen atom, a –COOH group or a -NO 2 group; - Z 8 is a hydrogen atom or a –OH group; - Z 9 is a hydrogen atom or a halogen group; - X 5 is a nitrogen atom or a carbon atom. The present invention also provides a method for inducing immune cells characterized in that said immune cells are contacting with at least one compound of formula (I) inducing production of proteins, in particular interleukin-10. The present invention also provides an immune cell capable of producing proteins, in particular interleukin-10. The present invention also provides a pharmaceutical composition comprising a dose of induced immune cell, and at least one pharmaceutically acceptable carrier and its use in the prevention and/or treatment of immune-mediated diseases, in particular neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammatory reactions predisposing to cancer or aggravating cancer progression, and aging.

DESCRIPTION OF FIGURES

Figure 1 : Common B cell stimuli induce a low level of IL-10 expression in resting mouse B cells.

Figure 2: Identification of novel IL-10-inducers in a high throughput flow cytometry-based phenotypic screen.

Figure 3: C3 and N6 upregulate IL-10 expression in TLR-, CD40-, and BCR-stimulated B cells

Figure 4: N6 increases IL-10-production by CD4 + T cells.

Figure 5: N6 induces a quick upregulation of IL-10 mRNA expression in stimulated mouse B cells

Figure 6: N6 and C3 increase IL-10 transcription in unstimulated B cells.

Figure 7A: Generation of B220low/CD138high plasma cells in cultures of spleen CD19+, follicular (Fo) and marginal zone (MZ) B cells after induction with Cpg/DMSO, CpG/C3 or CpG/N6.

Figure 7B: IL-10 expression in spleen CD19+ B cells, Fo B cells, MZ cells and blood CD19+ B cells after induction with CpG/DMSO, CpG/C3 or CpG/N6.

Figure 7C: IL-10 concentration in cell culture supernatant from spleen CD19+, Fo B cells and MZ cells after induction with CpG/DMSO, CpG/C3 or CpG/N6.

Figure 8: N6 increases IL-10 expression in mouse B cells without augmenting their IL-6 production.

Figure 9: Establishment of an expansion protocol to generate an almost pure population of IL-10-expressing B cells with the IL-10 inducing compound.

Figure 10: Establishment of a feeder cell-free expansion protocol to generate an almost pure population of IL-10-expressing B cells.

Figure 11 : The established protocol generates an almost pure population of IL-10- expressing B cells from various starting B cell subsets.

Figure 12A: B cells obtained using the 2 steps culture protocol and displaying an almost pure expression of IL-10, have a stable IL-10 expression upon subsequent culture without IL-10-inducing compound in unstimulated condition (RPMI). Results obtained by flow cytometry. Figure 12B: B cells obtained using the 2 steps culture protocol and displaying an almost pure expression of IL-10, have a stable IL-10 expression upon subsequent culture without IL-10-inducing compound in CpG condition. Results obtained by flow cytometry.

Figure 12C: B cells obtained using the 2 steps culture protocol and displaying an almost pure expression of IL-10, have a stable IL-10 expression upon subsequent culture without IL-10-inducing compound in LPS condition. Results obtained by flow cytometry.

Figure 12D: B cells obtained using the 2 steps culture protocol and displaying an almost pure expression of IL-10, have a stable IL-10 expression upon subsequent culture without IL-10-inducing compound in R848 condition. Results obtained by flow cytometry.

Figure 12E: B cells obtained using the 2 steps culture protocol and displaying an almost pure expression of IL-10, have a stable IL-10 expression upon subsequent culture without IL-10-inducing compound in anti-CD40 condition. Results obtained by flow cytometry.

Figure 12F: B cells obtained using the 2 steps culture protocol and displaying an almost pure expression of IL-10, have a stable IL-10 expression upon subsequent culture without IL-10-inducing compound in anti-BCR condition. Results obtained by flow cytometry.

Figure 12G: B cells obtained using the 2 steps culture protocol and displaying an almost pure expression of IL-10, have a stable IL-10 expression upon subsequent culture without IL-10-inducing compound in anti-BCR+anti-CD40 condition. Results obtained by flow cytometry.

Figure 13: N6 induces a quick and specific remodeling of DNA accessibility (including at 1110 gene) in murine B cells.

Figure 14: B cells expanded with L47/CpG/N6 protect mice from EAE development in a prophylactic setting.

Figure 15: B cells expanded with L47/CpG/N6 protect mice from EAE development in a therapeutic setting.

Figure 16: The therapeutic effect of B cells expanded with L47/CpG/N6 in EAE requires their IL-10 expression and is independent of their MHC-II expression.

Figure 17: B cells expanded with L47/CpG/N6 can therapeutically restore recovery from EAE in mice depleted of CD4 + FOXP3 + Tregs that otherwise develop a severe chronic disease

Figure 18: B cells expanded with L47/CpG/N6 accumulate in the CNS during EAE where they show an IL-10-producing plasmocyte phenotype

Figure 19: B cells expanded with L47/CpG/N6 rapidly enter the inflamed CNS of treated mice during EAE where they locally persist

Figure 20: B cells expanded with L47/CpG/N6 dampen the activation of CNS-associated macrophages and microglial cells. Figure 21 : B cells expanded with L47/CpG/N6 express IL-10 and display a plasmocyte transcriptome in the CNS of mice treated by adoptive B cell therapy at the peak of EAE, whereas microglia and CNS-associated macrophages/monocytes are the major IL-10 receptor-expressing cell types locally.

Figure 22: B cells expanded with L47/CpG/N6 dampen disease severity in an acute DSS- induced colitis model.

Figure 23A: Effect of compounds 1 , 4, 7, 22, 30, 31 , 32, 40, 43, 45, 52, 54, 56, 57, 58, 60, 61 , 67, 107, 109, 110, 111 , 116, N4, N6, 117, 120, 124, 136, 145, 153 and 157 on IL-10 induction by human B cells.

Figure 23B: Effect of compounds 366, 371 and 375 on IL-10 induction by human B cells. Figure 24: Effect of induction of human B cells with compounds 1 , 4, 7, 22, 30, 31 , 32, 40, 43,45, 52, 54, 56, 57, 58, 60, 61 , 67, 107, 109, 110, 111 , 116, N4, N6, 117 and 120 on IL- 10 production by human B cells.

Figure 25: Effect of induction of human B cells with compounds 124, 136, 145, 153 and 157 on IL-10 production by human B cells.

Figures 26-27: Differentially expressed genes in lymphocytes in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 28-29: Differentially expressed genes in granulocytes in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 30A-30G: Differentially expressed genes in disease-associated microglia in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 31A-C: Differentially expressed genes in disease-associated microglia in brain from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figure 32: Differentially expressed genes in disease-associated microglia 2 in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figure 33: Differentially expressed genes in disease-associated microglia 2 in brain from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 34A-F: Differentially expressed genes in disease-associated microglia 3 in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 35A-D: Differentially expressed genes in disease-associated microglia 3 in brain from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figure 36: Differentially expressed genes in disease-associated microglia 4 in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figure 37: Differentially expressed genes in disease-associated microglia 4 in brain from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6. Figure 38: Differentially expressed genes in disease-associated perivascular macrophages in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 39A-B: Differentially expressed genes in disease-associated perivascular macrophages in brain from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 40A-D: Differentially expressed genes in homeostatic microglia in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figure 41 : Differentially expressed genes in homeostatic microglia in brain from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 42A-D: Differentially expressed genes in homeostatic perivascular macrophages in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 43A-E: Differentially expressed genes in homeostatic perivascular macrophages in brain from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figure 44: Differentially expressed genes in perivascular and parenchymal monocyte- derived cells 1 in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 45A-B: Differentially expressed genes in perivascular and parenchymal monocyte-derived cells 1 in brain from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figure 46: Differentially expressed genes in perivascular and parenchymal monocyte- derived cells 2 in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 47A-D: Differentially expressed genes in perivascular and parenchymal monocyte-derived cells 2 in brain from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 48A-B: Differentially expressed genes in perivascular and parenchymal monocyte-derived cells 3 in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figure 49: Differentially expressed genes in perivascular and parenchymal monocyte- derived cells 3 in brain from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figure 50: Differentially expressed genes in perivascular and parenchymal monocyte- derived cells 4 in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6. Figure 51 : Differentially expressed genes in perivascular and parenchymal monocyte- derived cells 4 in brain from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figure 52: Differentially expressed genes in perivascular and parenchymal monocyte- derived cells 5 in spinal cord from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

Figures 53A-B: Differentially expressed genes in perivascular and parenchymal monocyte-derived cells 5 in brain from EAE mice treated with IL-10-producing B cells generated with L47/CpG/N6.

DETAILLED DESCRIPTION

The present invention provides a compound inducing production of proteins, in particular interleukin 10 (IL-10) by immune cells of formula (I):

- Xi, X 2 and X 3 are each independently a nitrogen atom or a carbon atom;

- Ri and R 9 are each a hydrogen atom, when the double dotted line between Ri and R 9 and the simple dotted line between R 9 and Zi are nothing;

- or RT is a nitrogen atom and R 9 is a carbon atom, when the double dotted line is a double bond and the simple dotted line between R 9 and 7_ y is a simple bond, and 7_ y is a (Ci-C 6 )alkyl group or a phenyl group optionally substituted with a halogen atom or a (C r C 6 )alkyl group;

- R 2 and R 3 are identical or different, are each independently selected from: a hydrogen atom, a halogen atom, and a group selected from:

wherein:

- Z 3 is a hydrogen atom, a -COOH group, or a -NO 2 group; - X 4 is a carbon atom or a nitrogen atom;

- Z 2 is a hydroxyl group or a hydrogen atom;

- q is selected from 0, 1 , 2, 3, 4 and 5, and;

- each Z 4 is selected from a halogen atom, a (Ci-C 6 )alkyl group, a (Ci-C 6 )alkoxy group, an hydroxyl group, a -NH 2 group, a -NHCOCH 3 group, a -NO 2 group, an alkylether group, a -SCH 3 group, a -SO 2 N(CH 3 ) 2 group, a -SO 2 CH(CH 3 ) 2 group, a -SO 2 NHCH(CH 3 ) 2 group, a -NHCOOCH 3 group, a - SOCH 2 CH 3 group, and a -CH 2 NHCOCH 3 group;

- R 4 , R 5 , R 6 and R w are each independently a hydrogen atom or a halogen atom;

- R 8 is absent, when X 2 is a nitrogen atom or R 8 is a hydrogen atom, a halogen atom or a hydroxyl group, when X 2 is a carbon atom; - R 7 is absent, when X 3 is a nitrogen atom or R 7 is selected from: a hydrogen atom, a halogen atom, a -OH group, a -OCH 3 group, and a group selected from: wherein:

- p is selected from 0, 1 , 2, 3, 4 and 5 ;

- i is selected from 0, 1 , 2, 3 and 4;

- Z 5 is selected from a halogen atom, a (Ci-C 6 )alkyl group, a (Ci-C 6 )alkoxy group, a hydroxyl group, a -NH 2 group, a -NHCOCH 3 group, a - NO 2 group and a -N(CH 3 ) 2 group;

- Z 6 is a hydrogen atom, a -COOH group or a -NO 2 group;

- Z 8 is a hydrogen atom or a -OH group;

- Z 9 is a hydrogen atom or a halogen group;

- X 5 is a nitrogen atom or a carbon atom.

The inventors have shown that the compound of formula (I) is capable of inducing the production of proteins, in particular interleukin 10 (IL-10) by immune cells. Advantageously, the compound of formula (I) enables the production of pure and stable IL-10-producing immune cells. In particular, the inventors have shown that the compound of formula (I) increases interleukin production, in particular IL-10 production, by immune cells, in particular B cells, activated via TLR4, TLR7/8, TLR9, CD40, and IgM, indicating that the IL-10 production is not restricted to any particular activation condition. Moreover, the inventors have shown that compound of formula (I) induces a distinct molecular program in immune cell, in particular B cell, with an up-regulation of transcriptional regulators known to promote IL-10 expression in these cells. The IL-10-expressing B cells induced by the compound of formula (I) up-regulated genes known to be associated with plasmocyte differentiation including Jchain, Xbp1 , Mzb1 , Prdml , and Irf4.

The inventors have shown that immune cells induced with a compound of formula (I) continue to express proteins, in particular IL-10 homogeneously, when subsequently washed and re-cultured in various conditions with the compound of formula (I), while the immune cells were refractory to IL-6 production.

As used herein, the term “compound inducing production of proteins” refer to a compound enables to increase proteins production by immune cell. Advantageously, a compound inducing production of proteins enables to produce immune cell cultures, wherein more than 5%, advantageously more than 10%, advantageously more than 15%, advantageously more than 20%, advantageously more than 25%, advantageously more than 30%, advantageously more than 35%, advantageously more than 40%, advantageously more than 45%, advantageously more than 50%, advantageously more than 55%, advantageously more than 60%, advantageously more than 65%, advantageously more than 70%, advantageously more than 75%, advantageously more than 80%, advantageously more than 85%, advantageously more than 90%, advantageously more than 91%, advantageously more than 92%, advantageously more than 93%, advantageously more than 94%, advantageously more than 95%, advantageously more than 96%, advantageously more than 97%, advantageously more than 98%, advantageously more than 99% of the immune cells of the culture are capable of producing proteins. Advantageously, the compound of the invention induces production of proteins, in particular of one of the following proteins: interleukin-10, PDL1 , PDL2, CD200, KLRA4 and LAG-3, advantageously the production of interleukin-10.

As used herein, the term “interleukin-10”, also known as human cytokine synthesis inhibitory factor (CSIF), is an anti-inflammatory cytokine. As used herein, the terms “interleukin-10” or “IL10” or “IL-10” are interchangeable.

As used herein, the term "Ct-Cz" means a carbon chain optionally containing from t to z carbon atoms in which t and z can take values from 1 to 10; for example, Ci-C 6 is a carbon chain optionally containing 1 to 6 carbon atoms.

As used herein, the term “alkyl group” refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds). In particular, a “(Ci-C 6 )alkyl group” refers an alkyl group having 1 to 6 carbon atoms. By way of examples, mention may be made of methyl, ethyl, n-propyl, isopropyl, butyl, n-butyl, isobutyl, tert-butyl, pentyl, neopentyl, n-hexyl and the like. “Ci alkyl” refers to methyl, “C 2 alkyl” refers to ethyl, “C 3 alkyl” refers to propyl and “C 4 alkyl” refers to butyl.

As used herein, the term “(Ci-C 6 )alkoxy group” refers to a straight or branched hydrocarbon chain group comprising the specified number of carbon atoms, advantageously between 1 and 6 carbon atoms, and an oxygen atom (-O-C). By way of examples, mention may in particular be made of a methoxy, ethoxy, propoxy, isopropoxy, butoxy, tert-butoxy, pentoxy, and hexoxy. “Ci alkoxy” refers to methoxy, “C 2 alkoxy” refers to ethoxy, “C 3 alkoxy” refers to propoxy and“C 4 alkoxy” refers to butoxy. Further, as used herein, “OMe” refers to methoxy and “OEt” refers to ethoxy.

As used herein, the term “alkylether group” refers to any alkyl group as defined above, wherein at least one carbon-carbon bond is replaced with a carbon-oxygen bond. The carbon-oxygen bond may be on the terminal end (as in an alkoxy group) or the carbon oxygen bond may be internal (i.e., C — O — C).

As used herein, the term “hydroxyl” refers to a monovalent group of formula -OH.

As used herein, the term “halogen atom” refers to an atom selected from fluorine, chlorine, bromine, or iodine. The halogen atom may advantageously be a chlorine atom, a fluorine atom or a bromine atom.

In the context of the present disclosure, the various embodiments described herein can be combined.

According to the present invention, the compound of formula (I) according to the invention can be used in the form of a pharmaceutically acceptable salt and can comprise salts, hydrates and solvates prepared according to conventional methods, well known to those skilled in the art. By way of examples, mention may in particular be made of salts derived from carboxylic acids, such as carboxylates (COO ), but also sulfonates (-SO3 ), phosphonates (PO 3 2 ), quaternary ammoniums (NR 4 + ) or sulfonamines (SO 2 NH 3 + ). Advantageously, suitable salts include pharmaceutically or physiologically acceptable acid addition salts. Advantageously, suitable salts include acid addition salts formed with various pharmaceutically or physiologically acceptable free acids. Examples of acids may include, but are not limited to, hydrochloric acid, bromic acid, sulfuric acid, phosphoric acid, citric acid, acetic acid, lactic acid, tartaric acid, fumaric acid, formic acid, propionic acid, oxalic acid, trifluoroacetic acid, methanesulfonic acid, benzenesulfonic acid, maleic acid, benzoic acid, gluconic acid, glycolic acid, succinic acid, 4-morpholineethanesulfonic acid, camphorsulfonic acid, nitrobenzenesulfonic acid, hydroxy-O-sulfonic acid, 4- toluenesulfonic acid, ruktu knife, EMBO acid, glutamic acid, aspartic acid, etc.

Additionally, pharmaceutically acceptable metal salts can be prepared using bases. For example, alkali metal or alkaline earth metal salts can be obtained by dissolving the compound in an excess of an alkali metal hydroxide or alkaline earth metal hydroxide solution, filtering the compound salts undissolved and evaporating and drying the filtrate. Here, sodium, potassium, lithium, cesium, magnesium or calcium salts are pharmaceutically suitable metal salts, but not limited to these. In addition, silver salts corresponding to metal salts can be obtained by reacting alkali metals or alkaline earth metals with appropriate silver salts (eg nitrates). Mention may also be made of ammoniums (NH 4 + ) or amines in ammonium form such as diethylamine (EDTA), pyrrolidine, piperidine and pyridine.

In some embodiments, R 3 and R 7 cannot simultaneously (and respectively) represent a

In some embodiments, R 3 and R 7 cannot simultaneously (and respectively) represent a

In some embodiments, the compound of formula (I) is not one of the following compounds:

N-(2-(3-fluorophenyl)-1 H-benzo[d]imidazol-6-yl)benzamide;

N-[2-(2-Chlorophenyl)-1 H-benzimidazol-6-yl]-2-(4-morpholinyl)benzamide;

6-bromo-2-(2-fluorophenyl)-1 H-benzimidazole;

N-[5-(4-Methylphenyl)benzimidazo[2,1 -a]phthalazin-10-yl]benzamide;

2-Chloro-N-[5-(4-methylphenyl)benzimidazo[2,1 -a]phthalazin-10-yl]benzamide;

2-lodo-N-[5-(4-methylphenyl)benzimidazo[2,1 -a]phthalazin-10-yl]benzamide;

2-Methoxy-N-[5-(4-methylphenyl)benzimidazo[2,1 -a]phthalazin-10-yl]benzamide. In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is constituted by a subgroup wherein:

- X 1 ; X 2 , X 3 are a carbon atom; - Ri is a hydrogen atom;

- R 2 and R 3 are each independently selected from: a hydrogen atom, a halogen atom, and a group selected from: wherein:

- q is selected from 0, 1 , 2, 3 , 4 and 5 ;

- each Z 4 is selected from a hydrogen atom, a halogen atom, a (Ci-C 6 )alkoxy group, an alkylether group, a -SCH 3 group, and a -SO 2 N(CH 3 ) 2 group;

- R 4 , R 5 , Re, R?, Rs, Rg and R w are each independently a hydrogen atom or a halogen atom.

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is selected from:

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is selected from:

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is constituted by a subgroup wherein:

- X b X 2 , X 3 are a carbon atom;

- Ri is a hydrogen atom;

- R 2 and R 3 are each independently selected from: a hydrogen atom, a halogen atom, wherein:

- q is selected from 0, 1 , 2, 3, 4 and 5;

- each Z 4 is independently a -NH 2 group or a -NHCOCH 3 group;

- X 4 is a carbon atom and Z 2 is a hydroxyl group;

- R 7 is selected from: a hydrogen atom, a halogen atom, wherein:

- p are each independently selected from 0, 1 , 2, 3, 4 and 5;

- i is 0;

- each Z 5 is independently a -NH 2 group or a -NHCOCH 3 group;

- X 5 is a carbon atom and Z 8 is a hydroxyl group;

- R 4 , R 5 , R 6 , R 8 , R 9 and R w are each independently a hydrogen atom or a halogen atom.

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is selected from:

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is:

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is constituted by a subgroup wherein:

- Xi and X 3 are a carbon atom,

- X 2 is a nitrogen atom;

- Ri is a hydrogen atom;

- R 4 , R 5 , R 6 , R 7 , R 9 and R w are a hydrogen atom;

- R 2 and R 3 are each independently selected from a hydrogen atom, wherein q is selected from 0, 1 , 2, 3 , 4 and 5 and Z 4 is a (Ci-C 6 )alkoxy group. In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is selected from:

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is constituted by a subgroup wherein:

- Xi, X 2 , X 3 are a carbon atom;

- Ri is a hydrogen atom;

- R 2 , R3, R4, Rs, Re, R7, Rs, R9 and R w are each independently a hydrogen atom or a halogen atom.

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is selected from:

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is selected from:

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is constituted by a subgroup wherein:

- X 1 ; X 2 , X 3 are a carbon atom;

- R1 is a nitrogen atom and R 9 is a carbon atom;

- the double dotted line is a double bond;

- the simple dotted line between R 9 and Z1 is a simple bond;

- Z1 is a (Ci-C 6 )alkyl group, or a phenyl group optionally substituted with a halogen atom;

- R 2 , R 4 , R 5 , Re, R7, Rs, and R w are a hydrogen atom; wherein:

- q is selected from 0, 1 , 2, 3, 4 and 5; and

- each Z 4 is independently a halogen atom or a (Ci-C 6 )alkoxy group.

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is selected from:

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is:

In some embodiments, the compound of formula (I) according to the invention, inducing the production of proteins, in particular interleukin-10 (IL-10), is selected from:

The chemical structures and some spectroscopic data of preferred compounds of inducing production of interleukin-10 (IL-10) of formula (I) are illustrated in the following Table 1 .

Table 1

Synthesis

The following general scheme may be implemented for obtaining a variety of compounds of formula (I), wherein R 1 ; R 3 , R 4 , R 10 are a hydrogen atom and X 1 ; X 2 , X 3 , R 5 , R 6 , R 7 , R 8 and R 9 are defined as above and R 2 is a -NHCOR a group or a -CONHR a , wherein R a is a phenyl group substituted with (Z 4 ) q , starting from a compound of formula (1), as set out in scheme 1 herein after:

The synthesis is based on the reaction of 1 ,2-phenylenediamine derivative of formula (1) with substituted aldehydes of formula (2) to form a benzimidazole derivative of formula (II) via according to route (A), following the incorporation of benzamide group through the Pd- catalytic coupling between halogenated compound of formula (II) and a benzamide derivative of formula (3) according to route (B). The reaction between a carboxylic acid derivative of formula (II) and an amine derivative of formula (4) via a peptide coupling follows to incorporate a reversed amide group (in comparison to the molecule of formula (la)) according to route (C).

Scheme 1

Route (A)

According to route (A), the compound of formula (1 ), wherein R 2 is a halogen atom, such as a bromine, or a carboxylic acid group may be placed in an aprotic polar solvent, such as dimethylformamide. An aldehyde compound (2), in which X! or X 2 or X 3 is a nitrogen atom and R 5 =R 6 =R 7 =R 8 =R 9 are a hydrogen atom or, in which X b X 2 and X 3 are a carbon atom and R 5 or R 6 or R 7 or R 8 or R 9 is a halogen atom, such as a fluorine atom, may then be added for example in a molar ratio ranging from 0.8 to 1 with respect to the compound of formula (1) in presence of an inorganic acid, such as Na 2 S 2 O 5 for example in a molar ratio ranging from 1 to 2 with respect to the compound of formula (1). The reaction mixture can be heated at a temperature ranging from 70°C to 130°C, for example at 1 10°C and stirred for a time for example ranging from 1 to 18 hours, for example during 4 hours. Or, the reaction can be irradiated under microwave at a temperature ranging from 70°C to 130°C, for example at 110°C and stirred for a time for example ranging from 10 min to 1 hour, for example during 30 min. The reaction mixture can be cooled and poured on ice to precipitate the compound of formula (II). Then the precipitate can be washed by water and dried under vacuum overnight. Or, the reaction mixture can be concentrated under reduce pressure and the residue can be diluted with an organic solvent such as ethyl acetate. The organic phase can be washed with water, decanted and dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue can be dried under vacuum overnight or purified by column chromatography on silica gel to give product (II) where X b X 2 , X 3 , R 5 , R 6 , R 7 , R 8 ,R 9 and (Z 4 ) q are as defined above and R 3 , R 4 and R w are a hydrogen atom and R 2 is a halogen atom. The aldehyde compounds (2) are available or can be prepared according to methods known to the person skilled in the art. Route (B)

According to route (B), the compound of formula (II), wherein X 1 ; X 2 , X 3 , R 5 , R 6 , R 7 , R 8 , R 9 and (Z 4 ) q are as defined above, R 3 , R 4 and R w are a hydrogen atom and R 2 is a halogen atom, such as a bromine, may be placed in a non-polar solvent, such as 1 ,4-dioxane. The benzamide compound of formula (3) in which (Z 4 ) q is as defined above, may then be added for example in a molar ratio ranging from 1 to 1 .5 with respect to the compound of formula (II) in presence of an inorganic base, such as K 3 PO 4 for example in a molar ratio ranging from 2 to 4 with respect to the compound of formula (II), in the presence of diphosphine, such as Me 4 tBuXphos (di-tert-butyl[3,4,5,6-tetramethyl-2',4',6'-tris(propan-2- yl)-[1 ,1 '-biphenyl]-2-yl]phosphane) in amount ranging from 5 mol% to 20 mol% relative to the total amount of compound of formula (II), and in the presence of an organometallic catalyst, such as Pd 2 dba 3 in an amount ranging from 2 mol% to 5 mol% relative to the total amount of compound of formula (II) . The reaction mixture can then be heated at a temperature ranging from 80°C to 140°C, for example at 130°C and stirred for a time for example ranging from 15 to 25 hours, for example during 18 hours in sealed tube under inert gas and for example argon. The reaction mixture can be concentrated under reduce pressure and the residue can be diluted with an organic solvent such as ethyl acetate. The organic phase can be washed with water, decanted and dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue can be dried under vacuum overnight or purified by column chromatography on silica gel to give product (I), wherein R 1 ; R 3 , R 4 , R 10 are a hydrogen atom and X 1 ; X 2 , X 3 , R 5 , R 6 , R 7 , R 8 and R 9 are defined as above and R 2 is a -NHCOR a group, wherein R a is a phenyl group substituted with (Z 4 ) q . The starting compound of formula (3) is available or can be prepared according to methods known to the person skilled in the art.

Route (C)

According to the route (C ), the compound of formula (II), wherein X b X 2 , X 3 , R 5 , R 6 , R 7 , R 8 , R 9 and (Z 4 ) q are as defined above, R 3 , R 4 and R w are a hydrogen atom and R 2 is a carboxylic acid group, may be placed in an aprotic polar solvent, such as dimethylformamide. The aniline compound of formula (4) in which (Z 4 ) q is as defined above, may then be added for example in a molar ratio ranging from 1 to 1.5 with respect to the compound of formula (II) in presence of an organic base, such as DIPEA for example in a molar ratio ranging from 3 to 5 with respect to the compound of formula (II) and a coupling reagent, such as HATU (Hexafluorophosphate Azabenzotriazole Tetramethyl Uronium) for example in a molar ratio ranging from 1.2 to 2 with respect to the compound of formula (II). The reaction mixture can then be stirred at a room temperature for a time for example ranging from 3 to 25 hours, for example during 18 hours. Water can be poured on the reaction mixture to precipitate the compound of formula (lb). Then the precipitate can be washed by water and dried under vacuum overnight. Or, the reaction mixture can be diluted with an organic solvent such as ethyl acetate. The organic phase can be washed with water, decanted and dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue can be dried under vacuum overnight or purified by column chromatography on silica gel to give product (I), wherein R b R 3 , R 4 , R w are a hydrogen atom and X b X 2 , X 3 , R 5 , R 6 , R 7 , R 8 and R 9 are defined as above and R 2 is a -NHCOR a group, wherein R a is a phenyl group substituted with (Z 4 ) q . The starting compound of formula (4) is available or can be prepared according to methods known to the person skilled in the art.

The examples that follow illustrate the present invention without limiting the scope thereof. The following examples illustrate in detail the preparation of some compounds according to the invention. The structures of the products obtained have been confirmed by NMR analyses and mass spectroscopy.

The following abbreviations are used:

“EtOAc” means ethyl acetate, “Et 2 O” means diethyl ether

“DMF” means dimethylformamide,

“MgSO 4 ” means magnesium sulfate,

“Na 2 S 2 O 5 ” means sodium metabisulfite,

“DIPEA” means N,N-diisopropylethylamine, “HATLI” means 1-[Bis(dimethylamino)methylene]-1 H-1 ,2,3-triazolo[4,5-b]pyridinium 3- oxide hexafluorophosphate.

Synthesis of compound (110)

According to route (A), 4-bromo-1 ,2-benzenediamine (1g, 5,35 mmol, 1 ,2 eq) was placed in anhydrous DMF (14 mL). 3-fluorobenzaldehyde (461 pL, 4,35 mmol, 1 eq) was then added in the presence of Na 2 S 2 O 5 (901 mg, 4,74 mmol, 1 ,1 eq). The reaction mixture was heated at 110°C and stirred during 2h30 hours. The reaction mixture was concentrated under reduced pressure and the residue was diluted with EtOAc. The organic phase was washed with water, decanted and dried over MgSO 4 , filtered and concentrated under reduced pressure. The solid residue was triturated with Et 2 O and dried under vacuum overnight to give 6-Bromo-2-(3-fluorophenyl)-1 H-benzimidazole (110) (753 mg, 60%). Synthesis of compound (297)

According to route (A), 3,4-diaminobenzoic acid (200 mg, 1 ,31 mmol, 1 eq) was placed in anhydrous DMF (2 mL) in a MW tube. Benzaldehyde (133 pL, 1 ,31 mmol, 1 eq) was then added in the presence of Na 2 S 2 O 5 (250 mg, 1 ,31 mmol, 1 eq). The reaction mixture was irradiated during 25 min at 100°C. The reaction mixture was concentrated under reduced pressure and the residue was diluted with EtOAc. The organic phase was washed with water, decanted and dried over MgSO 4 , filtered and concentrated under reduced pressure. The solid residue was purified by preparative HPLC on C18 silica column to give 2-phenyl-1 H-benzo[d]imidazole-6-carboxylic acid (297) (115 mg, 37%).

Synthesis of compound (117)

According to route (B), 6-Bromo-2-(3-fluorophenyl)-1 H-benzimidazole (110) (100 mg, 0.343 mmol, 1 eq) was placed in anhydrous 1 ,4-dioxane (4 mL) in a sealed tube. Benzamide (46 mg, 0.378 mmol, 1.1 eq) was added in the presence of K 3 PO 4 (218 mg, 1.03 mmol, 3 eq), Me4tBuXphos (25 mg, 0.051 mmol, 0.15 eq), and Pd 2 dba 3 (9.4 mg, 0.01 mmol, 0.03 eq). The reaction mixture was heated at 130°C and stirred during 18 hours under argon. The reaction mixture was concentrated under reduced pressure and the residue was diluted with EtOAc. The organic phase was washed with water and brine, decanted and dried over MgSO 4 , filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography on silica gel to give N-[2-(3- fluorophenyl)-1 H-1 ,3-benzodiazol-5-yl]benzamide (117) (60 mg, 53%). Synthesis of compound (366)

According to the route (C), 2-phenyl-1 H-benzo[d]imidazole-6-carboxylic acid (297) (40 mg, 0.17 mmol, 1 eq) and o-anisidine (21 pL, 0.18 mmol, 1 ,1 eq) were placed in anhydrous DMF (2 mL). Then, HATLI (96 mg, 0.25 mmol, 1.5eq) and DIPEA (145 pL, 0.84 mmol, 5 eq) were added to the reaction mixture. The reaction mixture was stirred at a room temperature for 4 hours. The reaction mixture was diluted with EtOAc. The organic phase was washed with water, decanted and dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography on silica gel to give N-(2-methoxyphenyl)-2-phenyl-1 H- benzo[d]imidazole-6-carboxamide (366) (57 mg, 98%).

Further synthetic processes are gathered in the following Table 2, giving the reactions conditions under which they were carried out.

Table 2

Method for inducing immune cells

In some embodiments, the immune cells of the invention are selected among T lymphocytes, B lymphocytes, dendritic cells, natural killer cells, innate lymphoid cells, mesenchymal cells and myeloid cells. In a particular embodiment, the immune cells are T lymphocytes. In a particular embodiment, the immune cells are B lymphocytes. In a particular embodiment, the immune cells are myeloid cells. Another aspect of the invention relates to a method for inducing production of proteins, in particular interleukin-10, by immune cells characterized in that said immune cells are contacting with at least one compound inducing production of interleukin-10 of formula (I) as described above.

In some embodiments, immune cells were cultivated for two consecutive days in the presence of anti-IgM plus CpG in presence of at least one compound of formula (I) at 0,25 pM final concentration inducing the production of interleukin-10.

As used herein, the expression “immune cells induced by at least one compound of formula (I) or “induced immune cells” or “immune cells capable of producing interleukin 10” refers to immune cells having being in contact with at least one compound of formula (I) and after their incubation with such compound of formula (I) enabled to produce interleukin-10 (IL-10).

In some embodiments, the present invention also provides immune cells capable of producing interleukin 10 obtained by the method as defined above. Advantageously, the immune cells obtained by the method as defined above exhibit an up-regulation of at least one gene selected from : 1110, Jchain, Ighm, Hsp90b1 , Xbp1 , Mzb1 , Tmed6, Manf, A430035B10Rik, Igkc, Pdia6, Hspa5, Tent5c, Calr, Ly6d, Selenos, FtH , Pdia4, H13, Creld2, Derl3, Sdf2l1 , Ssr4, Ddost, Pdia3, Ppib, Sec61b, Herpudl , Spcs2, Ostc, Ssr2, Hdlbp, Phgdh, Eaf2, Dnajc3, Lmanl , Sec11c, Pafah1 b3, Edeml , Ppp3cc, Rpn1 , Dnase1 l3, lft20, Topi , Ly6a, Sub1 , Aldoa, P4hb, E130308A19Rik, Irf4, EII2, Sec61g, TrnedlO, Prdml , Glccil , Selenok, Krtcap2, Rgcc, Mpc2, Pkm, Dnajb11 , Sqstml , Spcsl , Ssr3, Dusp4, Cope, Rilpl2, Edem2, Prdx4, Klra4, Rpn2, Tmem258, Nucbl , Itm2b, Senp6, Plaat3, Lmo4, Tmem123, Kdelr2, Rrbpl , Tmed2, A530032D15Rik, Nars, Plpp5, Rgs10, Ctsd, Cd81 , Trabd, Trp53inp1 , Pim1 , Rexo2, Spcs3, Utp3, Ccnd2, Tmed9, BnipSI, Derll , AW1 12010, Os9, Cd36 and Ggh, compared to the same immune cells non-induced by at least one compound of formula (I) as defined above.

In some embodiments, the immune cells obtained by the method as defined above exhibit an up-regulation of at least one gene selected from: Jchain, Xbp1 , Mzb1 , Irf4 and Prdml , compared to the same immune cells non-induced by at least one compound of formula (I) as defined above.

In some embodiments, the immune cells obtained by the method as defined above exhibit a down-regulation of at least one gene selected from : Cd37, Btg1 , lfi30, Nfkbia, Ms4a6c, Ms4a1 , Zeb2, Vim, Ctsh, Ptpn6, Napsa, Lmo2, Ly86, Cd83, Bankl , Stapl , Dek, Siglecg, Gimap3, Mndal, Gm15987, Serpinbla, Klf2, Cd22, Fcgr2b, Fam111 a, Cd74, Gpx1 , Pold4, Irf8, CD45R B220, Tespal , Cd53, Pfn1 , lfi203, Aldh2, Akap13, Bell 1 a, Slfn2, Lcp1 , H2- DMb2, Ptpnl , Ebf 1 , Sh3bgrl3, Gimap4, Rac2, Pax5, H2-DMa, H2-Eb1 , Corol a, 1121 r, Jak1 , Cripl , Cfp, Cdkn2d, H2-Aa, CD69, Pou2f2, Gimap5, Camk2d, Gm31243, Sh3bp5, Ms4a4c, Fam49b, Fus, Hclsl , Stk38, Tubal b, Lat2, Lpxn, Clic1 , Samd9l, H2-Oa, Abliml , Ptpn18, Bcl2a1 b, Limd2, Il4i1 , Bach2, Grb2, Gm26740, Fchsd2, Tpm3, Bini , Lyn, Blvrb, Msn, Zfp36l1 , Ifitm2, Anp32a, Tnfaip8l2, Bazl a, Gm8369, Ncf4, Ciita, Swap70, Ncf1 , Gm20559, Apobec3, Rhoh and Socs3, compared to the same immune cells non-induced by at least one compound of formula (I) as defined above.

In some embodiments, the immune cells obtained by the method as defined above exhibit an up-regulation of at least one transcriptional regulators selected from : Xbp1 , Hspa5, Sub1 , P4hb, Irf4, Prdml , Pkm, Nucbl , Atf4, Canx, Srp9, Nfe2l2, Nme2, Atf5, Cebpb, Pck2, Hbp1 , Pou2af1 , Mxd4, Tpi1 , Snd1 , Arid3a, Rufy3, Aff1 , Creb3l2, Zbtb38, Tcf3, Rere, Akr1 a1 , Mbnl2, Camtai , Foxa3, Bptf, Klf3, Plek, Jun, Rab2a, Sp140, Ddit3, Preb, Zfp945, Gimp, Kdm5b, Foxn3, Zfp706, Mxi1 , Smarca4, Tcf12, GpbpW and Ets2, compared to the same immune cells non-induced by at least one compound of formula (I) as defined above.

In some embodiments, the immune cells obtained by the method as defined above exhibit an down-regulation of at least one transcriptional regulators selected from : Zeb2, Lmo2, Klf2, Irf8, Bell 1 a, Ebf1 , Pax5, Pou2f2, Hclsl , Bach2, Zfp36l1 , Hmgb2, Purb, Sp100, Mef2c, Rel, Psma6, Ran, Plscrl , Spib, Smap2, Foxpl , Relb, Cenps, Zfp608, Yeats4, Bcl3, Tsc22d4, H2afz, Hmgbl , Nfkbl , Batf, Fubpl , Rbckl , Smarcbl , Rfc2, Spi1 , Runxl , Nap1 l1 , Cebpd, Dnmtl , Sfpq, Hhex, Lrrfipl , Statl , Zfp318, H2afy, Cnbp, Mybll , Lyl1 , compared to the same immune cells non-induced by at least one compound of formula (I) as defined above.

Another object of the invention relates to the immune cells induced by at least one compound of formula (I) as defined above for use as a medicament. In particular, the present invention relates to the immune cells induced by at least one compound of formula (I) as defined above for use in the prevention and/or the treatment of neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset and progression, and aging.

Another object of the invention relates to the use of immune cells induced by at least one compound of formula (I) as defined above in the preparation of a medicament intended for the prevention and/or the treatment of neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset and progression, or aging. According to some embodiments, the immune cells induced by at least one compound of formula (I) as defined above are particularly useful for preventing and/or treating neurodegenerative diseases. The term "neurodegenerative disease", as used herein, refers to a condition or disorder in which neuronal cells are lost due to cell death bringing about a deterioration of cognitive functions or result in damage, dysfunction, or complications that may be characterized by neurological, neurodegenerative, physiological, psychological, or behavioral aberrations. Neurodegenerative diseases include, without limitation, multiple sclerosis, autistic spectrum disorder, depression, age- related macular degeneration, Creutzfeldt-Jakob disease, Alzheimer's Disease, radiotherapy induced dementia, axon injury, acute cortical spreading depression, alpha- synucleinopathies, brain ischemia, Huntington's disease, Guillain-Barre syndrome, permanent focal cerebral ischemia, peripheral nerve regeneration, post-status epilepticus model, spinal cord injury, sporadic amyotrophic lateral sclerosis, transmissible spongiform encephalopathy, myasthenia gravis, obsessive-compulsive disorder, optic neuritis, retinal degeneration, dry eye syndrome DES, Sjogren's syndrome, chronic idiopathic demyelinating disease (CID), anxiety, compulsive disorders (such as compulsive obsessive disorders), meningitis, neuroses, psychoses, insomnia and sleeping disorder, sleep apnoea, and drug abuse.

According to some embodiments, the immune cells induced by at least one compound of formula (I) as defined above are particularly useful for preventing and/or the treating inflammatory diseases. Inflammatory diseases can include inflammatory diseases of the digestive apparatus, especially including the intestine (and particularly the colon in the case of irritable bowel syndrome, ulcero-haemorrhagic rectocolitis or Crohn's disease); pancreatitis, hepatitis (acute and chronic), inflammatory bladder pathologies and gastritis; inflammatory diseases of the locomotor apparatus, including rheumatoid arthritis, osteoarthritis, osteoporosis, traumatic arthritis, post-infection arthritis, muscular degeneration and dermatomyositis; inflammatory diseases of the urogenital apparatus and especially glomerulonephritis; inflammatory diseases of the cardiac apparatus and especially pericarditis and myocarditis and diseases including those for which inflammation is an underlying factor (such as atherosclerosis, transplant atherosclerosis, peripheral vascular diseases, inflammatory vascular diseases, intermittent claudication or limping, restenosis, strokes, transient ischaemic attacks, myocardial ischaemia and myocardial infarction), hypertension, hyperlipidaemia, coronary diseases, unstable angina (or angina pectoris), thrombosis, platelet aggregation induced by thrombin and/or the consequences of thrombosis and/or of the formation of atheroma plaques; inflammatory diseases of the respiratory and ORL apparatus, especially including asthma, acute respiratory distress syndrome, hayfever, allergic rhinitis and chronic obstructive pulmonary disease; inflammatory diseases of the skin, and especially urticaria, scleroderma, contact dermatitis, atopic dermatitis, psoriasis, ichthyosis, acne and other forms of folliculitis, rosacea and alopecia; the treatment of pain, irrespective of its origin, such as post- operative pain, neuromuscular pain, headaches, cancer-related pain, dental pain or osteoarticular pain; modulating pigmentation, for the treatment of: diseases with pigmentation disorders and especially benign dermatoses such as vitiligo, albinism, melasma, lentigo, ephelides, melanocytic naevus and all post-inflammatory pigmentations; and also pigmented tumours such as melanomas and their local (permeation nodules), regional or systemic metastases; photo-protection for the purpose of preventing: the harmful effects of sunlight, such as actinic erythema, cutaneous ageing, skin cancer (spinocellular, basocellular and melanoma) and especially diseases that accelerate its occurrence (xeroderma pigmentosum, basocellular naevus syndrome and familial melanoma); photodermatoses caused by exogenous photosensitizers and especially those caused by contact photosensitizers (for example furocoumarins, halogenated salicylanilides and derivatives, and local sulfamides and derivatives) or those caused by systemic photosensitizers (for example psoralenes, tetracyclines, sulfamides, phenothiazines, nalidixic acid and tricyclic antidepressants); bouts or outbreaks of dermatosis with photosensitivity and especiallylight-aggravated dermatoses (for example lupus erythematosus, recurrent herpes, congenital poikilodermal or telangiectatic conditions with photosensitivity (Bloom's syndrome, Cockayne's syndrome or Rothmund- Thomson syndrome), actinic lichen planus, actinic granuloma, superficial disseminated actinic porokeratosis, acne rosacea, juvenile acne, bullous dermatosis, Darier's disease, lymphoma cutis, psoriasis, atopic dermatitis, contact eczema, Chronic Actinic Dermatosis (CAD), follicular mucinosis, erythema multiforme, fixed drug eruption, cutaneous lymphocytoma, reticular erythematous mucinosis, and melasma) ;dermatoses with photosensitivity by deficiency of the protective system with anomalies of melanin formation or distribution (for example oculocutaneous albinism, phenylketonuria, hypopituitarism, vitiligo and piebaldism) and with deficiency of the DNA repair systems (for example xeroderma pigmentosum and Cockayne's syndrome), dermatoses with photosensitivity via metabolic anomalies, for instance cutaneous porphyria (for example tardive cutaneous porphyria, mixed porphyria, erythropoietic protoporphyria, congenital erythropoietic porphyria (Gunther's disease), and erythropoietic coproporphyria), pellagra or pellagroid erythema (for example pellagra, pellagroid erythemas and tryptophan metabolism disorders); bouts or outbreaks of idiopathic photodermatoses and especially PMLE (polymorphic light eruption), benign summer light eruption, actinic prurigo, persistent photosensitizations (actinic reticuloid, remanent photosensitizations and photosensitive eczema), solar urticaria, hydroa vacciniforme, juvenile spring eruption and solar pruritus;modifying the colour of the skin or head hair and bodily hair, and especially by tanning the skin by increasing melanin synthesis or bleaching it by interfering with melanin synthesis, but also by preventing the bleaching or greying of head hair or bodily hair (for example canities and piebaldism); and also modifying the colour of head hair and bodily hair in cosmetic indications;modifying the sebaceous functions, and especially the treatment of: hyperseborrhoea complaints and especially acne, seborrhoeic dermatitis, greasy skin and greasy hair, hyperseborrhoea in Parkinson's disease and epilepsy and hyperandrogenism; complaints with reduction of sebaceous secretion and especially xerosis and all forms of dry skin; benign or malignant proliferation of sebocytes and the sebaceous glands; inflammatory complaints of the pilosebaceous follicles and especially acne, boils, anthrax and folliculitis; male or female sexual dysfunctions; male sexual dysfunctions including, but not limited to, impotence, loss of libido and erectile dysfunction; female sexual dysfunctions including, but not limited to, sexual stimulation disorders or desire-related disorders, sexual receptivity, orgasm, and disturbances of the major points of sexual function; pain, premature labour, dysmenorrhoea, excessive menstruation, and endometriosis; disorders related to weight but not limited to obesity and anorexia (such as modification or impairment of appetite, metabolism of the spleen, or the vocable irreproachable taking of fat or carbohydrates); diabetes mellitus (by tolerance to glucose doses and/or reduction of insulin resistance); cancer and in particular lung cancer, prostate cancer, bowel cancer, breast cancer, ovarian cancer, bone cancer or angiogenesis disorders including the formation or growth of solid tumours.

According to some embodiments, the immune cells induced by at least one compound of formula (I) as defined above are particularly useful for preventing and/or the treating immune-mediated diseases. Immune-mediated diseases include, for example, but not limited to, asthma, allergies, arthritis (e.g., rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis), juvenile arthritis, inflammatory bowel diseases (e.g., ulcerative colitis and Crohn's disease), endocrinopathies (e.g., type 1 diabetes and Graves' disease), vascular diseases (e.g., autoimmune hearing loss, systemic vasculitis, and atherosclerosis), and skin diseases (e.g., acne vulgaris dermatomyositis, pemphigus, systemic lupus erythematosus (SLE), discoid lupus erthematosus, scleroderma, psoriasis, plaque psoriasis, vasculitis, vitiligo and alopecias). Hashimoto's thyroiditis, pernicious anemia, Cushing's disease, Addison's disease, chronic active hepatitis, polycystic ovary syndrome (PCOS), celiac disease, pemphigus, transplant rejection (allograft transplant rejection), graft-versus-host disease (GVDH). According to some embodiments, the immune cells induced by at least one compound of formula (I) as defined above are particularly useful for preventing and/or the treating allergic diseases. As used herein, the term "allergy" or "allergies" or "allergic reaction" or "allergic response" refers to a disorder (or improper reaction) of the immune system. Allergic reactions occur to normally harmless environmental substances known as allergens; these reactions are acquired, predictable, and rapid. Strictly, allergy is one of four forms of hypersensitivity and is called type I (or immediate) hypersensitivity. It is characterized by excessive activation of certain white blood cells called mast cells and basophils by a type of antibody known as IgE, resulting in an extreme inflammatory response. Common allergic reactions include eczema, hives, hay fever, asthma, food allergies, and reactions to the venom of stinging insects such as wasps and bees. Mild allergies like hay fever are highly prevalent in the human population and cause symptoms such as allergic conjunctivitis, itchiness, and runny nose. Allergies can play a major role in conditions such as asthma. In some people, severe allergies to environmental or dietary allergens or to medication may result in life-threatening anaphylactic reactions and potentially death. The treatment of allergies via immune suppression is therefore one embodiment of the present invention.

According to some embodiments, the immune cells induced by at least one compound of formula (I) as defined above are particularly useful for preventing and/or the treating adverse events related to transplantation. Adverse events related to transplantation relate to any medical condition surrounding rejection of transplanted material, or an immune response caused by transplantation of tissue to a recipient subject. Advantageously, adverse events related to transplantation can be directed to the kidney, liver, heart, lung, pancreas, bone marrow, cornea, intestine or skin (skin allograft, homograft or heterograft, etc.).

According to some embodiments, the immune cells induced by at least one compound of formula (I) as defined above are particularly useful for preventing and/or the treating autoimmune diseases. Examples of autoimmune disease include, but are not limited to arthritis (rheumatoid arthritis, juvenile-onset rheumatoid arthritis, osteoarthritis, psoriatic arthritis, and ankylosing spondylitis), psoriasis, dermatitis including atopic dermatitis, chronic idiopathic urticaria, including chronic autoimmune urticaria, polymyositis/ dermatomyositis, toxic epidermal necrolysis, scleroderma (including systemic scleroderma), sclerosis such as progressive systemic sclerosis, inflammatory bowel disease (IBD) (for example, Crohn's disease, ulcerative colitis, autoimmune inflammatory bowel disease), pyoderma gangrenosum, erythema nodosum, primary sclerosing cholangitis, episcleritis), respiratory distress syndrome, including adult respiratory distress syndrome (ARDS), meningitis, IgE-mediated diseases such as anaphylaxis and allergic and atopic rhinitis, encephalitis such as Rasmussen's encephalitis, uveitis or autoimmune uveitis, colitis such as microscopic colitis and collagenous colitis, glomerulonephritis (GN) such as membranous GN (membranous nephropathy), idiopathic membranous GN, membranous proliferative GN (MPGN), including Type I and Type II, and rapidly progressive GN, allergic conditions, allergic reaction, eczema, asthma, conditions involving infiltration of T cells and chronic inflammatory responses, atherosclerosis, autoimmune myocarditis, leukocyte adhesion deficiency, systemic lupus erythematosus (SLE) such as cutaneous SLE, subacute cutaneous lupus erythematosus, lupus (including nephritis, cerebritis, pediatric, non-renal, discoid, alopecia), juvenile onset (Type I) diabetes mellitus, including pediatric insulin-dependent diabetes mellitus (IDDM), adult onset diabetes mellitus (Type II diabetes), multiple sclerosis (MS) such as spino-optical MS, immune responses associated with acute and delayed hypersensitivity mediated by cytokines and T-lymphocytes, tuberculosis, sarcoidosis, granulomatosis including lymphomatoid granulomatosis, Wegener's granulomatosis, agranulocytosis, vasculitis (including large vessel vasculitis (including polymyalgia rheumatica and giant cell (Takayasu's) arteritis), medium vessel vasculitis (including Kawasaki's disease and polyarteritis nodosa), CNS vasculitis, systemic necrotizing vasculitis, and ANCA- associated vasculitis, such as Churg-Strauss vasculitis or syndrome (CSS)), temporal arteritis, aplastic anemia, Coombs positive anemia, Diamond Blackfan anemia, hemolytic anemia or immune hemolytic anemia including autoimmune hemolytic anemia (AIHA), pernicious anemia, pure red cell aplasia (PRCA), Factor VIII deficiency, hemophilia A, autoimmune neutropenia, pancytopenia, leukopenia, diseases involving leukocyte diapedesis, CNS inflammatory disorders, multiple organ injury syndrome, antigen- antibody complex mediated diseases, anti-glomerular basement membrane disease, antiphospholipid antibody syndrome, allergic neuritis, Bechet's or Behcet's disease, Castleman's syndrome, Goodpasture's syndrome, Reynaud's syndrome, Sjogren's syndrome, Stevens-Johnson syndrome, pemphigoid such as pemphigoid bullous, pemphigus (including vulgaris, foliaceus, and pemphigus mucus-membrane pemphigoid), autoimmune polyendocrinopathies, Reiter's disease, immune complex nephritis, chronic neuropathy such as IgM polyneuropathies or IgMmediated neuropathy, thrombocytopenia (as developed by myocardial infarction patients, for example), including thrombotic thrombocytopenic purpura (TTP) and autoimmune or immune-mediated thrombocytopenia such as idiopathic thrombocytopenic purpura (ITP) including chronic or acute ITP, autoimmune disease of the testis and ovary including autoimune orchitis and oophoritis, primary hypothyroidism, hypoparathyroidism, autoimmune endocrine diseases including thyroiditis such as autoimmune thyroiditis, chronic thyroiditis (Hashimoto's thyroiditis), or subacute thyroiditis, autoimmune thyroid disease, idiopathic hypothyroidism, Addison's disease, Grave's disease, polyglandular syndromes such as autoimmune polyglandular syndromes (or polyglandular endocrinopathy syndromes), paraneoplastic syndromes, including neurologic paraneoplastic syndromes such as Lambert-Eaton myasthenic syndrome or Eaton-Lambert syndrome, stiff-man or stiff-person syndrome, encephalomyelitis such as allergic encephalomyelitis, myasthenia gravis, cerebellar degeneration, limbic and/or brainstem encephalitis, neuromyotonia, opsoclonus or opsoclonus myoclonus syndrome (QMS), and sensory neuropathy, Sheehan's syndrome, autoimmune hepatitis, chronic hepatitis, lupoid hepatitis, chronic active hepatitis or autoimmune chronic active hepatitis, lymphoid interstitial pneumonitis, bronchiolitis obliterans (non-transplant) vs NSIP, GuillainBarre syndrome, Berger's disease (IgA nephropathy), primary biliary cirrhosis, celiac sprue (gluten enteropathy), refractory sprue, dermatitis herpetiformis, cryoglobulinemia, amyotrophic lateral sclerosis (ALS; Lou Gehrig's disease), coronary artery disease, autoimmune inner ear disease (AIED) or autoimmune hearing loss, opsoclonus myoclonus syndrome (OMS), polychondritis such as refractory polychondritis, pulmonary alveolar proteinosis, amyloidosis, giant cell hepatitis, scleritis, a non-cancerous lymphocytosis, a primary lymphocytosis, which includes monoclonal B cell lymphocytosis (e.g., benign monoclonal gammopathy and monoclonal gammopathy of undetermined significance, MGUS), peripheral neuropathy, paraneoplastic syndrome, channelopathies such as epilepsy, migraine, arrhythmia, muscular disorders, deafness, blindness, periodic paralysis, and channelopathies of the CNS, autism, inflammatory myopathy, focal segmental glomerulosclerosis (FSGS), endocrine ophthalmopathy, uveoretinitis, autoimmune hepatological disorder, fibromyalgia, multiple endocrine failure, Schmidt's syndrome, adrenalitis, gastric atrophy, presenile dementia, demyelinating diseases, Dressier's syndrome, alopecia areata, CREST syndrome (calcinosis, Raynaud's phenomenon, esophageal dysmotility, sclerodactyly, and telangiectasia), male and female autoimmune infertility, ankylosing spondolytis, mixed connective tissue disease, Chagas' disease, rheumatic fever, recurrent abortion, farmer's lung, erythema multiforme, post-cardiotomy syndrome, Cushing's syndrome, bird-fancier's lung, Alport's syndrome, alveolitis such as allergic alveolitis and fibrosing alveolitis, interstitial lung disease, transfusion reaction, leprosy, malaria, leishmaniasis, kypanosomiasis, schistosomiasis, ascariasis, aspergillosis, Sampler's syndrome, Caplan's syndrome, dengue, endocarditis, endomyocardial fibrosis, endophthalmitis, erythema elevatum et diutinum, erythroblastosis fetalis, eosinophilic faciitis, Shulman's syndrome, Felty's syndrome, flariasis, cyclitis such as chronic cyclitis, heterochronic cyclitis, or Fuch's cyclitis, Henoch-Schonlein purpura, human immunodeficiency virus (HIV) infection, echovirus infection, cardiomyopathy, Alzheimer's disease, parvovirus infection, rubella virus infection, post-vaccination syndromes, congenital rubella infection, Epstein-Barr virus infection, mumps, Evan's syndrome, autoimmune gonadal failure, Sydenham's chorea, post-streptococcal nephritis, thromboangitis ubiterans, thyrotoxicosis, tabes dorsalis, and giant cell polymyalgia. The treatment of autoimmune disorders via immune suppression is therefore one embodiment of the present invention.

According to some embodiments, the immune cells induced by at least one compound of formula (I) as defined above are particularly useful for preventing and/or the treating metabolic diseases with an inflammatory component, such as arteriosclerosis with resulting coronary heart diseases, adiposity or diabetes.

According to some embodiments, the immune cells induced by at least one compound of formula (I) as defined above are particularly useful for preventing and/or the treating aging.

Advantageously, when administered to the patient, the immune cells induced by at least one compound of formula (I) rapidly enter the inflamed central nervous system (CNS) upon administration, where they suppressed the deleterious activation of microglia and CNS-associated macrophages. Thus, the immune cells induced by at least one compound of formula (I) suppress the deleterious activation of myeloid cells in tissues targeted by deleterious immune responses and inflammation.

Pharmaceutical composition

Another object of the invention concerns a pharmaceutical composition comprising at least a dose of induced immune cell as defined above.

In some embodiments, the dose of immune cells is provided as a composition or formulation, such as a pharmaceutical composition or formulation. Such compositions can be used in accord with the provided methods and/or with the provided articles of manufacture or compositions, such as in the treatment of diseases, conditions, and disorders.

The term “pharmaceutical formulation” refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.

A “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject. A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.

In some embodiments, the subject is a human being, man or woman, regardless of age.

In some embodiments, the patient is a non-human animal, for example a non-human mammal, such as a dog, a cat, a mouse, a rat, a hamster, a rabbit, a chinchilla, a horse, a cow, a pig, a sheep, a goat or a primate.

In some embodiments, the pharmaceutical composition comprises a dose of immune cell induced by at least one compound of formula (I) as defined above, and at least one pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition comprises a dose of immune cells induced by a compound of formula (I) and at least one pharmaceutically acceptable carrier, wherein the compound of formula (I) is one of the following compounds:

Carrier

In some embodiments, the immune cells are formulated with a pharmaceutically acceptable carrier. In some aspects, the choice of carrier is determined in part by the particular cell or agent and/or by the method of administration. Accordingly, there are a variety of suitable formulations. For example, the pharmaceutical composition can contain preservatives. Suitable preservatives may include, for example, methylparaben, propylparaben, sodium benzoate, and benzalkonium chloride. In some aspects, a mixture of two or more preservatives is used. The preservative or mixtures thereof are typically present in an amount of about 0.0001% to about 2% by weight of the total composition. Carriers are described, e.g., by 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).

Buffering agents in some aspects are included in the compositions. Suitable buffering agents include, for example, citric acid, sodium citrate, phosphoric acid, potassium phosphate, and various other acids and salts. In some aspects, a mixture of two or more buffering agents is used. The buffering agent or mixtures thereof are typically present in an amount of about 0.001% to about 4% by weight of the total composition.

The formulations can include aqueous solutions. The formulation or composition may also contain at least one other active ingredient useful for the particular indication, disease, or condition being treated with the induced immune cells, where the respective activities do not adversely affect one another. Such at least one other active ingredient is suitably present in combination in amounts that are effective for the purpose intended. In some embodiments, the pharmaceutical composition according to the invention also comprises at least a second active ingredient. In some embodiments, the at least one other active ingredient can interact synergically with the immune cell induced by at least one compound of formula (I) according to the invention. Examples of other active ingredients include, but are not limited to, an estrogen receptor modulator; an androgen receptor modulator; a glucocorticoid, a retinoid receptor modulator; a cytotoxic agent; an anti- proliferative agent comprises: adriamycin, dexamethasone, vincristine, cyclophosphamide, fluorouracil, topotecan, taxol, interferons, or platinum derivatives; an anti-inflammatory agent comprises: corticosteroids, TNF blockers, IL-1 RA, azathioprine, cyclophosphamide, JAK inhibitors (e.g. tofacitinib and baricitinib), SYK inhibitors, BTK inhibitors or sulfasalazine; a prenyl-protein transferase inhibitor; an HMG-CoA reductase inhibitor; an HIV protease inhibitor; a reverse transcriptase inhibitor; an angiogenesis inhibitor comprises: sorafenib, sunitinib, pazopanib, or everolimus; an immunomodulatory or immunosuppressive agents comprises: cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, interferons, corticosteroids, cyclophophamide, azathioprine, or sulfasalazine; a PPAR-y agonist comprising thiazolidinediones; a PPAR-b agonist; an inhibitor of inherent multidrug resistance; an agent for the treatment of anemia, comprising: erythropoiesis-stimulating agents, vitamins, or iron supplements; an anti- emetic agent including 5-HT3 receptor antagonists, dopamine antagonists, NK1 receptor antagonist, H1 histamine receptor antagonists, cannabinoids, benzodiazepines, anticholinergic agents, or steroids; an agent for the treatment of neutropenia; an immunologic-enhancing agents; a proteasome inhibitors; an HDAC inhibitors; an inhibitor of the chemotrypsin-like activity in the proteasome; a E3 ligase inhibitors; a modulator of the immune system including interferon-alpha, Bacillus Calmette-Guerin (BCG), or ionizing radition (LIVB) that can induce the release of cytokines, interleukins, TNF, or induce release of death receptor ligands including TRAIL; a modulator of death receptors TRAIL, or TRAIL agonists including humanized antibodies HGS-ETR1 , or HGS-ETR2; neurotrophic factors selected from the group of: cetylcholinesterase inhibitors, MAO inhibitors, interferons, anti-convulsants, ion channel blockers, or riluzole; anti-Parkinsonian agents comprising anticholinergic agents, or dopaminergic agents, including dopaminergic precursors, monoamine oxidase B inhibitors, COMT inhibitors, dopamine receptor agonists; agents for treating cardiovascular disease comprises beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers, or statins; agents for treating liver disease comprises corticosteroids, cholestyramine, or interferons; anti-viral agents, including nucleoside reverse transcriptase inhibitors, non-nucleoside reverse transcriptase inhibitors, protease inhibitors, integrase inhibitors, fusion inhibitors, chemokine receptor antagonists, polymerase inhibitors, viral proteins synthesis inhibitors, viral protein modification inhibitors, neuraminidase inhibitors, fusion or entry inhibitors; agents for treating blood disorders comprising: corticosteroids, anti-leukemic agents, or growth factors; agents for treating immunodeficiency disorders comprising gamma globulin, adalimumab, etarnecept, tocilizumab or infliximab; a HMG-CoA reductase inhibitors including torvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin, simvastatin, or pitavastatin, or in combination, or sequentially with radiation, or at least one chemotherapeutic agents or immune modulatory agents e.g., asparaginase, busulfan, carboplatin, cisplatin, daunorubicin, doxorubicin, fluorouracil, gemcitabine, hydroxyurea, methotrexate, paclitaxel, rituximab, vinblastine, vincristine, etc.

In some embodiments, when the pharmaceutical composition comprises another active ingredient, the pharmaceutical composition is adapted for a simultaneous administration or a sequential administration of the immune cell induced by at least one compound of formula (I) as defined above and of the other active ingredient.

Within the meaning of the present invention, by “simultaneous administration” is meant an administration of the immune cell induced by at least one compound of formula (I) as defined above and of the other active ingredient at the same time, at the same moment, to a patient in therapeutically effective quantities in order to permit the synergic effect of the pharmaceutical composition. This does not necessarily mean that the immune cell induced by at least one compound of formula (I) as defined above and the other active ingredient must be administered in the form of a mixture; they can in fact be administered simultaneously but separately, in the form of distinct compositions.

By “present in two distinct compositions” is meant that the immune cell induced by at least one compound of formula (I) as defined above and the other active ingredient are physically separate. They are then implemented, administered separately in therapeutically effective quantities, to permit the synergic effect of the pharmaceutical composition, without prior mixing, in several (at least two) dosage forms (for example two distinct compositions).

In some embodiments, the immune cells induced by at least one compound of formula (I) as defined above and the other active ingredient can be present in one and the same composition in therapeutically effective quantities in order to permit the synergic effect of the pharmaceutical composition. Within the meaning of the present invention, by “present in one and the same composition” is meant the physical combination of the immune cells induced by at least one compound of formula (I) as defined above and the other active ingredient. The immune cells induced by at least one compound of formula (I) as defined above and the other active ingredient must then be administered simultaneously, as they are administered together in the form of a mixture, in one and the same dosage form and in therapeutically effective quantities, to permit the synergic effect of the pharmaceutical composition.

Within the meaning of the present invention, by the expression “sequential administration” is meant that the immune cells induced by at least one compound of formula (I) as defined above and the other active ingredient are administered in therapeutically effective quantities in order to permit the synergic effect of the pharmaceutical composition, not simultaneously but separately over time, one after another. The terms “precede” or “preceding” and “follow” or “following” then apply. The term “precede” or “preceding” is used when a component of the pharmaceutical composition according to the invention is administered a few minutes or a few hours, or even a few days before the administration of the other component(s) of the pharmaceutical composition. Conversely, the term “follow” or “following” is used when a component of the pharmaceutical composition according to the invention is administered a few minutes or a few hours, or even a few days after the administration of the other component(s) of the pharmaceutical composition. In a particularly advantageous embodiment of the invention, the immune cells induced by at least one compound of formula (I) as defined above are administered several days before the other active ingredient.

In some embodiments, when the pharmaceutical composition comprises another active ingredient, the pharmaceutical composition is adapted for a sequential administration of the immune cells induced by at least one compound of formula (I) as defined above and of the other active ingredient.

The pharmaceutical composition in some embodiments comprises the induced immune cells in amounts effective to treat the disease or condition, such as a therapeutically effective or prophylactically effective amount. Within the meaning of the present invention, the terms “therapeutically effective” or “effective quantity for treatment”, or “pharmaceutically effective” denote the quantity of immune cell induced by at least one compound of formula (I) as defined above necessary to inhibit or reverse a disease, advantageously for treating a disease selected among neurodegenerative disease, inflammatory disease, immune-mediated disease, allergic disease, adverse events related to transplantation, autoimmune disease, metabolic diseases with an inflammatory component, inflammation facilitating cancer or progression, and aging. The determination of a therapeutically effective quantity depends specifically on factors such as the toxicity and efficacy of the medicament. These factors will differ as a function of other factors such as the activity, the relative bioavailability, the body weight of the patient, the severity of the adverse effects and the preferred administration route. The toxicity can be determined by using methods that are well known in the art. The same applies for the efficacy. A pharmaceutically effective quantity is consequently a quantity that is considered by the clinician as toxicologically tolerable but effective. Therapeutic efficacy in some embodiments is monitored by periodic assessment of treated subjects. For repeated administrations over several days or longer, depending on the condition, the treatment is repeated until a desired suppression of disease symptoms occurs. However, other dosage regimens may be useful and can be determined. The desired dosage can be delivered by a single bolus administration of the composition, by multiple bolus administrations of the composition, or by continuous infusion administration of the composition

The induced immune cells of the invention may be administered using standard administration techniques, formulations, and/or devices. Provided are formulations and devices, such as syringes and vials, for storage and administration of the compositions. With respect to cells, administration can be autologous or heterologous. For example, immunoresponsive cells or progenitors can be obtained from one subject, and administered to the same subject or a different, compatible subject. Peripheral blood derived immunoresponsive cells or their progeny (e.g., in vivo, ex vivo or in vitro derived) can be administered via localized injection, including catheter administration, systemic injection, localized injection, intravenous injection, or parenteral administration. When administering a therapeutic composition (e.g., a pharmaceutical composition comprising the induced immune cells of the invention), it will generally be formulated in a unit dosage injectable form (solution, suspension, emulsion).

Formulations include those for oral, intravenous, intraperitoneal, subcutaneous, pulmonary, transdermal, intradermal, intratumoral, intralesional, intramuscular, intranasal, buccal, sublingual, topical, local, otic, inhalational, retinal, intraocular, periocular, subretinal, intravitreal, suppository, trans-septal, subscleral, intrachoroidal, intracameral, subconjectval, subconjuntival, sub-Tenon’s, retrobulbar, peribulbar, or posterior juxtascleral administration. In some embodiments, the agent or cell populations are administered parenterally. The term “parenteral,” as used herein, includes intravenous, intramuscular, subcutaneous, rectal, vaginal, and intraperitoneal administration. In some embodiments, the agent or cell populations are administered to a subject using peripheral systemic delivery by intravenous, intraperitoneal, or subcutaneous injection.

Compositions in some embodiments are provided as sterile liquid preparations, e.g., isotonic aqueous solutions, suspensions, emulsions, dispersions, or viscous compositions, which may in some aspects be buffered to a selected pH. Liquid preparations are normally easier to prepare than gels, other viscous compositions, and solid compositions. Additionally, liquid compositions are somewhat more convenient to administer, especially by injection. Viscous compositions, on the other hand, can be formulated within the appropriate viscosity range to provide longer contact periods with specific tissues. Liquid or viscous compositions can comprise carriers, which can be a solvent or dispersing medium containing, for example, water, saline, phosphate buffered saline, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol) and suitable mixtures thereof.

Sterile injectable solutions can be prepared by incorporating the cells in a solvent, such as in admixture with a suitable carrier, diluent, or excipient such as sterile water, physiological saline, glucose, dextrose, or the like.

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.

Dosing

In some embodiments, a dose of induced immune cells of the invention is administered to subjects in accord with the provided methods, and/or with the provided articles of manufacture or compositions. In some embodiments, the size or timing of the doses is determined as a function of the particular disease or condition in the subject. In some cases, the size or timing of the doses for a particular disease in view of the provided description may be empirically determined.

In some embodiments, the dose of induced immune cells of the invention comprises between at or about 2x10 5 of the cells/kg and at or about 9x10 14 of the cells/kg, advantageously between at or about 2x10 5 of the cells/kg and at or about 9x10 13 of the cells/kg, advantageously between at or about 2x10 5 of the cells/kg and at or about 9x10 12 of the cells/kg, advantageously, between at or about 2x10 5 of the cells/kg and at or about 9x10 11 of the cells/kg, advantageously between at or about 2x10 5 of the cells/kg and at or about 9x10 1 ° of the cells/kg, advantageously between at or about 2x10 5 of the cells/kg and at or about 9x10 9 of the cells/kg, advantageously between at or about 2x10 5 of the cells/kg and at or about 9x10 8 of the cells/kg.

In some embodiments, the dose of induced immune cells of the invention comprises at least or at least about or at or about 2x10 5 of the cells (e.g. IL10-expressing cells) per kilogram body weight of the subject (cells/kg), such as at least or at least about or at or about 3x10 5 cells/kg, at least or at least about or at or about 4x10 5 cells/kg, at least or at least about or at or about 5x10 5 cells/kg, at least or at least about or at or about 6x10 5 cells/kg, at least or at least about or at or about 7x10 5 cells/kg, at least or at least about or at or about 8x10 5 cells/kg, at least or at least about or at or about 9x10 5 cells/kg, at least or at least about or at or about 1 x10 6 cells/kg, or at least or at least about or at or about 2x10 6 cells/kg, at least or at least about or at or about 3x10 6 cells/kg, at least or at least about or at or about 4x10 6 cells/kg, at least or at least about or at or about 5x10 6 cells/kg, at least or at least about or at or about 6x10 6 cells/kg, at least or at least about or at or about 7x10 6 cells/kg, at least or at least about or at or about 8x10 6 cells/kg, at least or at least about or at or about 9x10 6 cells/kg, at least or at least about or at or about 1 x10 7 cells/kg, or at least or at least about or at or about 2x10 7 cells/kg, at least or at least about or at or about 3x10 7 cells/kg, at least or at least about or at or about 4x10 7 cells/kg, at least or at least about or at or about 5x10 7 cells/kg, at least or at least about or at or about 6x10 7 cells/kg, at least or at least about or at or about 7x10 7 cells/kg, at least or at least about or at or about 8x10 7 cells/kg, at least or at least about or at or about 9x10 7 cells/kg, at least or at least about or at or about 1 x10 8 cells/kg, or at least or at least about or at or about 2x10 8 cells/kg, at least or at least about or at or about 3x10 8 cells/kg, at least or at least about or at or about 4x10 8 cells/kg, at least or at least about or at or about 5x10 8 cells/kg, at least or at least about or at or about 6x10 8 cells/kg, at least or at least about or at or about 7x10 8 cells/kg, at least or at least about or at or about 8x10 8 cells/kg, at least or at least about or at or about 9x10 8 cells/kg, at least or at least about or at or about 1 x10 9 cells/kg, or at least or at least about or at or about 2x10 9 cells/kg, at least or at least about or at or about 3x10 9 cells/kg, at least or at least about or at or about 4x10 9 cells/kg, at least or at least about or at or about 5x10 9 cells/kg, at least or at least about or at or about 6x10 9 cells/kg, at least or at least about or at or about 7x10 9 cells/kg, at least or at least about or at or about 8x10 9 cells/kg, at least or at least about or at or about 9x10 9 cells/kg, at least or at least about or at or about 1 x10 1 ° cells/kg, or at least or at least about or at or about 2x10 1 ° cells/kg, at least or at least about or at or about 3x10 1 ° cells/kg, at least or at least about or at or about 4x1 O 10 cells/kg, at least or at least about or at or about 5x10 1 ° cells/kg, at least or at least about or at or about 6x10 1 ° cells/kg, at least or at least about or at or about

7x10 1 ° cells/kg, at least or at least about or at or about 8x10 1 ° cells/kg, at least or at least about or at or about 9x10 1 ° cells/kg, at least or at least about or at or about

1 x10 11 cells/kg, or at least or at least about or at or about 2x10 11 cells/kg, Dosages may vary depending on attributes particular to the disease or disorder and/or patient and/or other treatments. In some embodiments, the dose of cells is a flat dose of cells or fixed dose of cells such that the dose of cells is not tied to or based on the body surface area or weight of a subject.

In some embodiments, the dose of induced immune cells of the invention, is administered to the subject as a single dose or is administered only one time within a period of two weeks, one month, three months, six months, 1 year or more. In the context of adoptive cell therapy, administration of a given “dose” encompasses administration of the given amount or number of cells as a single composition and/or single uninterrupted administration, e.g., as a single injection or continuous infusion, and also encompasses administration of the given amount or number of cells as a split dose or as a plurality of compositions, provided in multiple individual compositions or infusions, over a specified period of time, such as over no more than 3 days. Thus, in some contexts, the dose is a single or continuous administration of the specified number of cells, given or initiated at a single point in time. In some contexts, however, the dose is administered in multiple injections or infusions over a period of no more than three days, such as once a day for three days or for two days or by multiple infusions over a single day period.

Thus, in some aspects, the cells of the dose are administered in a single pharmaceutical composition. In some embodiments, the induced immune cells of the dose are administered in a plurality of compositions, collectively containing the cells of the dose.

In some embodiments, the term “split dose” refers to a dose that is split so that it is administered over more than one day. This type of dosing is encompassed by the present methods and is considered to be a single dose.

Thus, the dose of induced immune cells may be administered as a split dose, e.g., a split dose administered over time. For example, in some embodiments, the dose may be administered to the subject over 2 days or over 3 days. Exemplary methods for split dosing include administering 25% of the dose on the first day and administering the remaining 75% of the dose on the second day. In other embodiments, 33% of the dose may be administered on the first day and the remaining 67% administered on the second day. In some aspects, 10% of the dose is administered on the first day, 30% of the dose is administered on the second day, and 60% of the dose is administered on the third day. In some embodiments, the split dose is not spread over more than 3 days.

In some embodiments, the administration of the composition or dose, e.g., administration of the plurality of cell compositions, involves administration of the cell compositions separately. In some aspects, the separate administrations are carried out simultaneously, or sequentially, in any order. In some embodiments, the dose comprises a first composition and a second composition, and the first composition and second composition are administered within 48 hours of each other, such as no more than 36 hours of each other or not more than 24 hours of each other. In some embodiments, the first composition and second composition are administered 0 to 12 hours apart, 0 to 6 hours apart or 0 to 2 hours apart. In some embodiments, the initiation of administration of the first composition and the initiation of administration of the second composition are carried out no more than 2 hours, no more than 1 hour, or no more than 30 minutes apart, no more than 15 minutes, no more than 10 minutes or no more than 5 minutes apart. In some embodiments, the initiation and/or completion of administration of the first composition and the completion and/or initiation of administration of the second composition are carried out no more than 2 hours, no more than 1 hour, or no more than 30 minutes apart, no more than 15 minutes, no more than 10 minutes or no more than 5 minutes apart.

In some embodiments, the first composition and the second composition is mixed prior to the administration into the subject. In some embodiments, the first composition and the second composition is mixed shortly (e.g., within 6 hours, 5 hours, 4 hours, 3 hours, 2 hours, 1.5 hours, 1 hour, or 0.5 hour) before the administration, In some embodiments, the first composition and the second composition is mixed immediately before the administration.

In some embodiments, the subject receives multiple doses, e.g., two or more doses or multiple consecutive doses, of the induced immune cells. In some embodiments, two doses are administered to a subject. In some embodiments, the subject receives the consecutive dose, e.g., second dose, is administered approximately 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 days after the first dose. In some embodiments, multiple consecutive doses are administered following the first dose, such that an additional dose or doses are administered following administration of the consecutive dose. In some aspects, the number of cells administered to the subject in the additional dose is the same as or similar to the first dose and/or consecutive dose. In some embodiments, the additional dose or doses are larger than prior doses.

Prevention treatment

Another aspect of the invention relates to a pharmaceutical composition comprising immune cells induced by at least one compound of formula (I) for its use in the prevention of immune-mediated diseases. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer or progression, and aging.

As used herein, the term “prevention” or “prophylaxis” or “preventative treatment” or “prophylactic treatment” comprises a treatment leading to the prevention of a disease as well as a treatment reducing and/or delaying the incidence of a disease or the risk of occurrence of the disease.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient, for its use in the prevention of immune- mediated diseases. Advantageously, the immune-mediated disease is selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer or progression, and aging.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and and at least one pharmaceutically acceptable carrier as defined above, for its use in the prevention of immune-mediated diseases. Advantageously, the immune-mediated disease is selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer or progression, and aging.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and and at least one pharmaceutically carrier as defined above, for its use in the prevention of neurodegenerative diseases. The term "neurodegenerative disease", as used herein, refers to a condition or disorder in which neuronal cells are lost due to cell death bringing about a deterioration of cognitive functions or result in damage, dysfunction, or complications that may be characterized by neurological, neurodegenerative, physiological, psychological, or behavioral aberrations. Neurodegenerative diseases include, without limitation, multiple sclerosis, autistic spectrum disorder, depression, age-related macular degeneration, Creutzfeldt-Jakob disease, Alzheimer's Disease, radiotherapy induced dementia, axon injury, acute cortical spreading depression, alpha-synucleinopathies, brain ischemia, Huntington's disease, Guillain-Barre syndrome, permanent focal cerebral ischemia, peripheral nerve regeneration, post-status epilepticus model, spinal cord injury, sporadic amyotrophic lateral sclerosis, transmissible spongiform encephalopathy, myasthenia gravis, obsessive- compulsive disorder, optic neuritis, retinal degeneration, dry eye syndrome DES, Sjogren's syndrome, chronic idiopathic demyelinating disease (CID), anxiety, compulsive disorders (such as compulsive obsessive disorders), meningitis, neuroses, psychoses, insomnia and sleeping disorder, sleep apnoea, and drug abuse.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and and at least one pharmaceutically acceptable carrier as defined above, for its use in the prevention of inflammatory diseases. Advantageously, the inflammatory diseases are selected among: Inflammatory diseases can include inflammatory diseases of the digestive apparatus, especially including the intestine (and particularly the colon in the case of irritable bowel syndrome, ulcero- haemorrhagic rectocolitis or Crohn's disease); pancreatitis, hepatitis (acute and chronic), inflammatory bladder pathologies and gastritis; inflammatory diseases of the locomotor apparatus, including rheumatoid arthritis, osteoarthritis, osteoporosis, traumatic arthritis, post-infection arthritis, muscular degeneration and dermatomyositis; inflammatory diseases of the urogenital apparatus and especially glomerulonephritis; inflammatory diseases of the cardiac apparatus and especially pericarditis and myocarditis and diseases including those for which inflammation is an underlying factor (such as atherosclerosis, transplant atherosclerosis, peripheral vascular diseases, inflammatory vascular diseases, intermittent claudication or limping, restenosis, strokes, transient ischaemic attacks, myocardial ischaemia and myocardial infarction), hypertension, hyperlipidaemia, coronary diseases, unstable angina (or angina pectoris), thrombosis, platelet aggregation induced by thrombin and/or the consequences of thrombosis and/or of the formation of atheroma plaques; inflammatory diseases of the respiratory and ORL apparatus, especially including asthma, acute respiratory distress syndrome, hayfever, allergic rhinitis and chronic obstructive pulmonary disease; inflammatory diseases of the skin, and especially urticaria, scleroderma, contact dermatitis, atopic dermatitis, psoriasis, ichthyosis, acne and other forms of folliculitis, rosacea and alopecia; the treatment of pain, irrespective of its origin, such as post-operative pain, neuromuscular pain, headaches, cancer-related pain, dental pain or osteoarticular pain; modulating pigmentation, for the treatment of:diseases with pigmentation disorders and especially benign dermatoses such as vitiligo, albinism, melasma, lentigo, ephelides, melanocytic naevus and all post- inflammatory pigmentations; and also pigmented tumours such as melanomas and their local (permeation nodules), regional or systemic metastases; photo-protection for the purpose of preventing :the harmful effects of sunlight, such as actinic erythema, cutaneous ageing, skin cancer (spinocellular, basocellular and melanoma) and especially diseases that accelerate its occurrence (xeroderma pigmentosum, basocellular naevus syndrome and familial melanoma); photodermatoses caused by exogenous photosensitizers and especially those caused by contact photosensitizers (for example furocoumarins, halogenated salicylanilides and derivatives, and local sulfamides and derivatives) or those caused by systemic photosensitizers (for example psoralenes, tetracyclines, sulfamides, phenothiazines, nalidixic acid and tricyclic antidepressants); bouts or outbreaks of dermatosis with photosensitivity and especiallylight-aggravated dermatoses (for example lupus erythematosus, recurrent herpes, congenital poikilodermal or telangiectatic conditions with photosensitivity (Bloom's syndrome, Cockayne's syndrome or Rothmund- Thomson syndrome), actinic lichen planus, actinic granuloma, superficial disseminated actinic porokeratosis, acne rosacea, juvenile acne, bullous dermatosis, Darier's disease, lymphoma cutis, psoriasis, atopic dermatitis, contact eczema, Chronic Actinic Dermatosis (CAD), follicular mucinosis, erythema multiforme, fixed drug eruption, cutaneous lymphocytoma, reticular erythematous mucinosis, and melasma) ;dermatoses with photosensitivity by deficiency of the protective system with anomalies of melanin formation or distribution (for example oculocutaneous albinism, phenylketonuria, hypopituitarism, vitiligo and piebaldism) and with deficiency of the DNA repair systems (for example xeroderma pigmentosum and Cockayne's syndrome), dermatoses with photosensitivity via metabolic anomalies, for instance cutaneous porphyria (for example tardive cutaneous porphyria, mixed porphyria, erythropoietic protoporphyria, congenital erythropoietic porphyria (Gunther's disease), and erythropoietic coproporphyria), pellagra or pellagroid erythema (for example pellagra, pellagroid erythemas and tryptophan metabolism disorders); bouts or outbreaks of idiopathic photodermatoses and especially PMLE (polymorphic light eruption), benign summer light eruption, actinic prurigo, persistent photosensitizations (actinic reticuloid, remanent photosensitizations and photosensitive eczema), solar urticaria, hydroa vacciniforme, juvenile spring eruption and solar pruritus; modifying the colour of the skin or head hair and bodily hair, and especially by tanning the skin by increasing melanin synthesis or bleaching it by interfering with melanin synthesis, but also by preventing the bleaching or greying of head hair or bodily hair (for example canities and piebaldism); and also modifying the colour of head hair and bodily hair in cosmetic indications; modifying the sebaceous functions, and especially the treatment of: hyperseborrhoea complaints and especially acne, seborrhoeic dermatitis, greasy skin and greasy hair, hyperseborrhoea in Parkinson's disease and epilepsy and hyperandrogenism; complaints with reduction of sebaceous secretion and especially xerosis and all forms of dry skin; benign or malignant proliferation of sebocytes and the sebaceous glands; inflammatory complaints of the pilosebaceous follicles and especially acne, boils, anthrax and folliculitis; male or female sexual dysfunctions; male sexual dysfunctions including, but not limited to, impotence, loss of libido and erectile dysfunction; female sexual dysfunctions including, but not limited to, sexual stimulation disorders or desire-related disorders, sexual receptivity, orgasm, and disturbances of the major points of sexual function; pain, premature labour, dysmenorrhoea, excessive menstruation, and endometriosis; disorders related to weight but not limited to obesity and anorexia (such as modification or impairment of appetite, metabolism of the spleen, or the vocable irreproachable taking of fat or carbohydrates); diabetes mellitus (by tolerance to glucose doses and/or reduction of insulin resistance); cancer and in particular lung cancer, prostate cancer, bowel cancer, breast cancer, ovarian cancer, bone cancer or angiogenesis disorders including the formation or growth of solid tumours.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above, for its use in the prevention of immune-mediated disease. Immune-mediated diseases include, for example, but not limited to, asthma, allergies, arthritis (e.g., rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis), juvenile arthritis, inflammatory bowel diseases (e.g., ulcerative colitis and Crohn's disease), endocrinopathies (e.g., type 1 diabetes and Graves' disease), neurodegenerative diseases (e.g., multiple sclerosis (MS)), depression, obsessive-compulsive disorder, optic neuritis, retinal degeneration, dry eye syndrome DES, Sjogren's syndrome, vascular diseases (e.g., autoimmune hearing loss, systemic vasculitis, and atherosclerosis), skin diseases (e.g., acne vulgaris dermatomyositis, pemphigus, systemic lupus erythematosus (SLE), discoid lupus erthematosus, scleroderma, psoriasis, plaque psoriasis, vasculitis, vitiligo and alopecias), Hashimoto's thyroiditis, pernicious anemia, Cushing's disease, Addison's disease, chronic active hepatitis, polycystic ovary syndrome (PCOS), celiac disease, pemphigus, transplant rejection (allograft transplant rejection), graft-versus-host disease (GVDH).

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and and at least one pharmaceutically acceptable carrier as defined above, for its use in the prevention of allergic diseases. As used herein, the term "allergy" or "allergies" or "allergic reaction" or "allergic response" refers to a disorder (or improper reaction) of the immune system. Allergic reactions occur to normally harmless environmental substances known as allergens; these reactions are acquired, predictable, and rapid. Strictly, allergy is one of four forms of hypersensitivity and is called type I (or immediate) hypersensitivity. It is characterized by excessive activation of certain white blood cells called mast cells and basophils by a type of antibody known as IgE, resulting in an extreme inflammatory response. Common allergic reactions include eczema, hives, hay fever, asthma, food allergies, and reactions to the venom of stinging insects such as wasps and bees. Mild allergies like hay fever are highly prevalent in the human population and cause symptoms such as allergic conjunctivitis, itchiness, and runny nose. Allergies can play a major role in conditions such as asthma. In some people, severe allergies to environmental or dietary allergens or to medication may result in life-threatening anaphylactic reactions and potentially death. The treatment of allergies via immune suppression is therefore one embodiment of the present invention.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and and at least one pharmaceutically acceptable carrier as defined above, for its use in the prevention of adverse events related to transplantation.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and and at least one pharmaceutically acceptable carrier as defined above, for its use in the prevention of autoimmune diseases. Examples of autoimmune disease include, but are not limited to, arthritis (rheumatoid arthritis, juvenile-onset rheumatoid arthritis, osteoarthritis, psoriatic arthritis, and ankylosing spondylitis), psoriasis, dermatitis including atopic dermatitis, chronic idiopathic urticaria, including chronic autoimmune urticaria, polymyositis/ dermatomyositis, toxic epidermal necrolysis, scleroderma (including systemic scleroderma), sclerosis such as progressive systemic sclerosis, inflammatory bowel disease (IBD) (for example, Crohn's disease, ulcerative colitis, autoimmune inflammatory bowel disease), pyoderma gangrenosum, erythema nodosum, primary sclerosing cholangitis, episcleritis), respiratory distress syndrome, including adult respiratory distress syndrome (ARDS), meningitis, IgE- mediated diseases such as anaphylaxis and allergic and atopic rhinitis, encephalitis such as Rasmussen's encephalitis, uveitis or autoimmune uveitis, colitis such as microscopic colitis and collagenous colitis, glomerulonephritis (GN) such as membranous GN (membranous nephropathy), idiopathic membranous GN, membranous proliferative GN (MPGN), including Type I and Type II, and rapidly progressive GN, allergic conditions, allergic reaction, eczema, asthma, conditions involving infiltration of T cells and chronic inflammatory responses, atherosclerosis, autoimmune myocarditis, leukocyte adhesion deficiency, systemic lupus erythematosus (SLE) such as cutaneous SLE, subacute cutaneous lupus erythematosus, lupus (including nephritis, cerebritis, pediatric, non-renal, discoid, alopecia), juvenile onset (Type I) diabetes mellitus, including pediatric insulin- dependent diabetes mellitus (IDDM), adult onset diabetes mellitus (Type II diabetes), multiple sclerosis (MS) such as spino-optical MS, immune responses associated with acute and delayed hypersensitivity mediated by cytokines and T-lymphocytes, tuberculosis, sarcoidosis, granulomatosis including lymphomatoid granulomatosis, Wegener's granulomatosis, agranulocytosis, vasculitis (including large vessel vasculitis (including polymyalgia rheumatica and giant cell (Takayasu's) arteritis), medium vessel vasculitis (including Kawasaki's disease and polyarteritis nodosa), CNS vasculitis, systemic necrotizing vasculitis, and ANCA-associated vasculitis, such as Churg-Strauss vasculitis or syndrome (CSS)), temporal arteritis, aplastic anemia, Coombs positive anemia, Diamond Blackfan anemia, hemolytic anemia or immune hemolytic anemia including autoimmune hemolytic anemia (AIHA), pernicious anemia, pure red cell aplasia (PRCA), Factor VIII deficiency, hemophilia A, autoimmune neutropenia, pancytopenia, leukopenia, diseases involving leukocyte diapedesis, CNS inflammatory disorders, multiple organ injury syndrome, antigen-antibody complex mediated diseases, anti- glomerular basement membrane disease, antiphospholipid antibody syndrome, allergic neuritis, Bechet's or Behcet's disease, Castleman's syndrome, Goodpasture's syndrome, Reynaud's syndrome, Sjogren's syndrome, Stevens-Johnson syndrome, pemphigoid such as pemphigoid bullous, pemphigus (including vulgaris, foliaceus, and pemphigus mucus- membrane pemphigoid), autoimmune polyendocrinopathies, Reiter's disease, immune complex nephritis, chronic neuropathy such as IgM polyneuropathies or IgMmediated neuropathy, thrombocytopenia (as developed by myocardial infarction patients, for example), including thrombotic thrombocytopenic purpura (TTP) and autoimmune or immune-mediated thrombocytopenia such as idiopathic thrombocytopenic purpura (ITP) including chronic or acute ITP, autoimmune disease of the testis and ovary including autoimune orchitis and oophoritis, primary hypothyroidism, hypoparathyroidism, autoimmune endocrine diseases including thyroiditis such as autoimmune thyroiditis, chronic thyroiditis (Hashimoto's thyroiditis), or subacute thyroiditis, autoimmune thyroid disease, idiopathic hypothyroidism, Addison's disease, Grave's disease, polyglandular syndromes such as autoimmune polyglandular syndromes (or polyglandular endocrinopathy syndromes), paraneoplastic syndromes, including neurologic paraneoplastic syndromes such as Lambert-Eaton myasthenic syndrome or Eaton- Lambert syndrome, stiff-man or stiff-person syndrome, encephalomyelitis such as allergic encephalomyelitis, myasthenia gravis, cerebellar degeneration, limbic and/or brainstem encephalitis, neuromyotonia, opsoclonus or opsoclonus myoclonus syndrome (QMS), and sensory neuropathy, Sheehan's syndrome, autoimmune hepatitis, chronic hepatitis, lupoid hepatitis, chronic active hepatitis or autoimmune chronic active hepatitis, lymphoid interstitial pneumonitis, bronchiolitis obliterans (non-transplant) vs NSIP, GuillainBarre syndrome, Berger's disease (IgA nephropathy), primary biliary cirrhosis, celiac sprue (gluten enteropathy), refractory sprue, dermatitis herpetiformis, cryoglobulinemia, amyotrophic lateral sclerosis (ALS; Lou Gehrig's disease), coronary artery disease, autoimmune inner ear disease (AIED) or autoimmune hearing loss, opsoclonus myoclonus syndrome (QMS), polychondritis such as refractory polychondritis, pulmonary alveolar proteinosis, amyloidosis, giant cell hepatitis, scleritis, a non-cancerous lymphocytosis, a primary lymphocytosis, which includes monoclonal B cell lymphocytosis (e.g., benign monoclonal gammopathy and monoclonal gammopathy of undetermined significance, MGUS), peripheral neuropathy, paraneoplastic syndrome, channelopathies such as epilepsy, migraine, arrhythmia, muscular disorders, deafness, blindness, periodic paralysis, and channelopathies of the CNS, autism, inflammatory myopathy, focal segmental glomerulosclerosis (FSGS), endocrine ophthalmopathy, uveoretinitis, autoimmune hepatological disorder, fibromyalgia, multiple endocrine failure, Schmidt's syndrome, adrenalitis, gastric atrophy, presenile dementia, demyelinating diseases, Dressier's syndrome, alopecia areata, CREST syndrome (calcinosis, Raynaud's phenomenon, esophageal dysmotility, sclerodactyly, and telangiectasia), male and female autoimmune infertility, ankylosing spondolytis, mixed connective tissue disease, Chagas' disease, rheumatic fever, recurrent abortion, farmer's lung, erythema multiforme, post- cardiotomy syndrome, Cushing's syndrome, bird-fancier's lung, Alport's syndrome, alveolitis such as allergic alveolitis and fibrosing alveolitis, interstitial lung disease, transfusion reaction, leprosy, malaria, leishmaniasis, kypanosomiasis, schistosomiasis, ascariasis, aspergillosis, Sampler's syndrome, Caplan's syndrome, dengue, endocarditis, endomyocardial fibrosis, endophthalmitis, erythema elevatum et diutinum, erythroblastosis fetalis, eosinophilic faciitis, Shulman's syndrome, Felty's syndrome, flariasis, cyclitis such as chronic cyclitis, heterochronic cyclitis, or Fuch's cyclitis, Henoch-Schonlein purpura, human immunodeficiency virus (HIV) infection, echovirus infection, cardiomyopathy, Alzheimer's disease, parvovirus infection, rubella virus infection, post-vaccination syndromes, congenital rubella infection, Epstein-Barr virus infection, mumps, Evan's syndrome, autoimmune gonadal failure, Sydenham's chorea, post-streptococcal nephritis, thromboangitis ubiterans, thyrotoxicosis, tabes dorsalis, and giant cell polymyalgia. The treatment of autoimmune disorders via immune suppression is therefore one embodiment of the present invention.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and and at least one pharmaceutically acceptable carrier as defined above, for its use in the prevention of metabolic diseases with an inflammatory component such as arteriosclerosis with resulting coronary heart diseases, adiposity or diabetes.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and and at least one pharmaceutically acceptable carrier as defined above, for its use in the prevention of aging.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and and at least one pharmaceutically acceptable carrier as defined above, for its use in the prevention of inflammation facilitating cancer onset or progression

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and and at least one pharmaceutically acceptable carrier as defined above and another active ingredient, for its use in the prevention of immune-mediated diseases. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression, and aging.

Another aspect of the invention relates to a method for the preventive treatment of immune-mediated diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and and at least one pharmaceutically acceptable carrier as defined above. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression and aging.

According to the present invention, the immune cells induced by at least one compound of formula (I) are particularly effective for preventing such diseases in adoptive cell therapy, since the immune cells induced by at least one compound of formula (I) act directly in the target organ, where they rapidly home and locally intercept the activation of microglia and CNS-associated macrophages. The immune cells induced by at least one compound of formula (I) directly intercept the inflammatory reaction in the target organ. The immune cells induced by at least one compound of formula (I) directly produce IL-10 at inflamed sites.

In some embodiments, the present invention relates to a method for the preventive treatment of immune-mediated diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression and aging. In some embodiments, the present invention relates to a method for the preventive treatment of neurodegenerative diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In a some embodiments, the present invention relates to a method for the preventive treatment of inflammatory diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to a method for the preventive treatment of immune-mediated diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to a method for the preventive treatment of allergic diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to a method for the preventive treatment of adverse events related to transplantation, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In a some embodiments, the present invention relates to a method for the preventive treatment of metabolic diseases with an inflammatory component, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to a method for the preventive treatment of aging, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to a method for the preventive treatment of inflammation facilitating cancer onset or progression, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the prevention of immune-mediated diseases. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression, and aging.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the prevention of neurodegenerative diseases.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the prevention of inflammatory diseases. In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the prevention of immune-mediated disease.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the prevention of allergic diseases.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the prevention of adverse events related to transplantation.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the prevention of autoimmune diseases.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the prevention of metabolic diseases with an inflammatory component.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the prevention of aging.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the prevention of inflammation facilitating cancer onset or progression.

Curative treatment

Another aspect of the invention relates to a pharmaceutical composition comprising immune cells induced by at least one compound of formula (I) for its use in the treatment of immune-mediated diseases. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression, and aging.

As used herein, the term “treatment” or “curative treatment” is defined as a treatment leading to recovery or treatment that relieves, ameliorates and/or eliminates, reduces and/or stabilizes the symptoms of a disease or the suffering that it elicits.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient, for its use in the treatment of immune- mediated diseases. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression, and aging.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and and at least one pharmaceutically acceptable carrier as defined above, for its use in the treatment of immune-mediated diseases. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression, and aging.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above, for its use in the treatment of neurodegenerative diseases.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above, for its use in the treatment of inflammatory diseases.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above, for its use in the treatment of immune-mediated disease.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above, for its use in the treatment of allergic diseases. In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above, for its use in the treatment of adverse events related to transplantation.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above, for its use in the treatment of autoimmune diseases.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above, for its use in the treatment of metabolic diseases with an inflammatory component.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above, for its use in the treatment of aging.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above, for its use in the treatment of inflammation facilitating cancer onset or progression.

In some embodiments, the present invention relates to a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient, at least one pharmaceutically acceptable carrier as defined above, and another active ingredient, for its use in the treatment of immune-mediated diseases. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression, and aging.

Another aspect of the invention relates to a method for the curative treatment of immune- mediated diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression and aging.

In some embodiments, the present invention relates to a method for the curative treatment of immune-mediated diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, and aging.

In some embodiments, the present invention relates to a method for the curative treatment of neurodegenerative diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to a method for the curative treatment of inflammatory diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to a method for the curative treatment of immune-mediated diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to a method for the curative treatment of allergic diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to a method for the curative treatment of adverse events related to transplantation, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to a method for the curative treatment of metabolic diseases with an inflammatory component, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to a method for the curative treatment of aging, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

In some embodiments, the present invention relates to a method for the curative treatment of inflammation facilitating cancer onset or progression, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above.

Another aspect of the invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the treatment of immune-mediated diseases. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression and aging.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the treatment of neurodegenerative diseases.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the treatment of inflammatory diseases.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the treatment of immune-mediated disease.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the treatment of allergic diseases.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the treatment of adverse events related to transplantation.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the treatment of autoimmune diseases.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the treatment of metabolic diseases with an inflammatory component.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the treatment of aging.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the treatment of inflammation facilitating cancer onset or progression. Another subject of the invention relates to a method for the curative treatment of immune- mediated diseases, comprising administration to said patients of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient and at least one pharmaceutically acceptable carrier as defined above. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression, and aging.

Another subject of the invention relates to a method for the curative treatment of immune- mediated diseases, comprising administration to a patient of immune cells induced by at least one compound of formula (I) or of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient, at least one pharmaceutically acceptable carrier as defined above, and another active ingredient as defined above. Advantageously, the immune-mediated diseases are selected from : neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression, and aging.

In some embodiments, the present invention relates to the use of a pharmaceutical composition comprising a therapeutically effective quantity of immune cells induced by at least one compound of formula (I) as active ingredient in the manufacture of a medicament intended for the treatment of immune-mediated diseases. Advantageously, the immune-mediated diseases are selected from: neurodegenerative diseases, inflammatory diseases, allergic diseases, adverse events related to transplantation, autoimmune diseases, metabolic diseases with an inflammatory component, inflammation facilitating cancer onset or progression, and aging.

Another aspect of the invention relates to a compound selected from:

EXEMPLES

Example 1 : Identification of compound of the invention with a comparable IL-10 inducing capacity in human B cells.

Human B cells isolated magnetically through negative selection (B Cell Isolation Kit II, human, Miltenyi ref: 130-091 -151) were cultivated for 2 consecutive days in the presence of anti-IgM plus CpG in the presence of the indicated molecule (see table 1) at 0.25pM final concentration, or DMSO as control. IL-10 was measured in cell culture supernatants, and the IL-10 induction fold was calculated for each molecule compared to the DMSO control.

Table 1 : Tested Molecules

Example 2: Common B cell stimuli induce a low level of IL-10 expression in resting mouse B cells

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43+ cells and subjected to different cell culture conditions for 3 consecutive days. B cells from C57BL/6 mice were used to determine autofluorescence in eGFP channel and IL- 10. eGFP-IL-6.tdTomato mice were used to monitor IL-10 expression by flow cytometry. The percentage of viable/singlets/CD19+/IL-10+ cells is shown. Data are presented as mean ± SD. n>3 independent experiments. The results are presented in Figure 1.

Conclusion: B cell activation using classical stimuli (agonists of BCR, CD40 ,TLR, and inflammatory cytokines such as IL-1 p, IL-6, IL-21 , BAFF) induces IL-10 only a in small proportion of treated cells (less than 30%).

Example 3: Identification of novel IL-10-inducers in a high throughput flow cytometry-based phenotypic screen

Resting mouse B cells were isolated from spleen of IL-10.eGFP mice by magnetic depletion of CD43+ cells, and stimulated for 72 hours in 384-well plates in the presence of chemical compounds at 10 pM final. The chemical library originated from the Leibniz- Forschungsinstitut fur Molekulare Pharmakologie (FMP) (Berlin, Germany). The frequency of IL-10-expressing B cells was then measured in each well by flow cytometry.

Data show the amount of IL-10 expressing B cells in unstimulated B cells (negative control), test condition with LPS and DMSO but no compound, and the result obtained for the compound C3 identified as a novel IL-10 inducer. The results are presented in Figure 2.

Conclusion: compound C3 markedly increases IL-10 expression in stimulated B cells.

Example 4: C3 and N6 upregulate IL-10 expression in TLR-, CD40-, and BCR- stimulated B cells

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and stimulated through TLR4, TLR7/8, TLR9, CD40, and IgM as indicated for 3 consecutive days. Cell culture medium was supplemented with DMSO-vehicle control, C3 or N6 at 0.25 pM. B cells from C57BL/6 mice were used to determine autofluorescence in eGFP channel and IL-10.eGFP-IL-6.tdTomato mice were used to monitor IL-10 expression by flow cytometry at 72 hours. The percentage of viable/singlets/CD197IL-10 + cells is shown. Data are presented as mean ± SD. n>3 independent experiments. The results are presented in Figure 3.

Conclusion: compounds C3 and N6 increase IL-10 expression in B cells across a broad range of stimulation protocols. Their effect is therefore not limited to a specific modality of B cell activation.

Example 5: N6 increases IL-10-production by CD4 + T cells

CD4 murine T cells were isolated from spleen by magnetic positive selection of CD4 + cells and stimulated with anti-CD3/anti-CD28 and IL-27 for 3 consecutive days. Cell culture medium was supplemented with DMSO-vehicle or N6 (0.25 pM). IL-10 secretion was quantified by ELISA in cell-free supernatants. The results are presented in Figure 4.

Conclusion: N6 increases IL-10 production by mouse CD4 + T cells Example 6: N6 induces a quick upregulation of 1110 mRNA expression in unstimulated and stimulated mouse B cells

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells. B cells were incubated with DMSO-vehicle control or N6 (0.25 pM), as such, or in combination with CpG. mRNA was extracted at different time-points and H10/ actin ratio was determined. The results are presented in Figure 5.

Conclusion: N6 induces IL-10 transcription in unstimulated (non-CpG stimulated) and CpG-stimulated cells within a few hours.

Example 7: N6 and C3 increase 1110 transcription in unstimulated B cells

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells. B cells were incubated with DMSO vehicle control, or C3 or N6 at 0.25 pM for the indicated time, and 1110 mRNA induction was quantified by quantitative RT-PCR. The results are presented in Figure 6.

Conclusion: N6 induces IL-10 transcription in unstimulated B cells within 3 hours of exposition.

Example 8: C3 and N6 upregulate IL-10 expression in various mouse B cell subsets

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and total CD19 + B cells, follicular B cells (CD197CD21 l0W 7CD23 hi9 7CD1 d l0W ) and marginal zone B cells (CD197CD21 hi9 7CD23 l0W/ 7CD1d hi ) were purified by FACS. Total CD19 + blood B cells were directly purified by FACS after red blood cell lysis. Purified cells were then subjected to CpG-based culture supplemented with DMSO, C3 or N6 for 3 consecutive days (C3 or N6 at 0.25pM). B cells from C57BL/6 mice were used to determine autofluorescence in eGFP channel and IL-10.eGFP-IL-6.tdTomato mice were used to monitor IL-10 expression by flow cytometry. IL-10 secretion was quantified in cell-free supernatants of C57BL/6 WT B cells. Generation of B220 lo 7CD138 hl9h plasma cells was also quantified by flow cytometry. Data are presented as mean ± SD. n>3 independent experiments. The results are presented in Figures 7A, 7B and 7C.

Conclusion: N6 and C3 increase the differentiation of B cells into plasma cells (Fig. 7A), increase the proportion of activated B cells expressing IL-10 (Fig. 7B), and increase the amount of IL-10 produced by stimulated B cells (Fig. 7C).

Example 9: N6 increases IL-10 expression in mouse B cells without augmenting their IL-6 production

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and subjected to TLR9-based cell culture conditions for 3 consecutive days. Cell culture medium was supplemented with DMSO-vehicle control or N6 (0.25 pM). B cells from C57BL/6 mice were used to determine autofluorescence in eGFP channel and IL- 10.eGFP-IL-6.tdTomato mice were used to monitor IL-6 and IL-10 expression by flow cytometry. The percentages of viable/singlets/CD197IL-10 + , viable/singlets/CD197IL-6 + , and viable/singlets/CD197IL-6 + IL-10 + cells are shown. IL-10 and IL-6 secretion was quantified in cell-free supernatants of C57BL/6 WT B cells. Data are presented as mean ± SD. n>3 independent experiments. The results are presented in Figure 8.

Conclusion: N6 increases IL-10 expression in B cells without modulating their IL-6 provision. It is thus not simply a booster of B cell activation, but instead a modulator of activated B cell polarization towards IL-10 expression.

Example 10: Establishment of an expansion protocol to generate an almost pure population of IL-10-expressing B cells

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on CD40L-expressing feeder cells (L47) and medium was supplemented with IL-4/BAFF, CpG/DMSO or CpG/N6 for 4 additional days. Cell fold expansion was determined during the second step of the culture, from day 3 to day 7. Cells were phenotyped by flow cytometry for B220, CD138 and PD-L1 expression. Light and dark histograms represent, FMO and PD-L1 staining, respectively. B cells from C57BL/6 mice were used to determine autofluorescence in eGFP channel and IL-10.eGFP-IL-6.tdTomato mice were used to monitor IL-10 and IL-6 expression by flow cytometry. The percentage of viable/singlets/CD197IL-10 + and/or IL-6 + cells is shown. IL-10 and IL-6 secretion was quantified in cell-free supernatants of C57BL/6 WT B cells. Data are presented as mean ± SD. n>3 independent experiments. The results are presented in Figure 9.

Conclusion: the two steps culture protocol established here (step 1 : CpG+N6 for 3 days; step 2: CpG+N6+L47 cells expressing CD40L for 4 days) robustly expands activated B cells (expansion fold around 35), and generates a quasi-pure population of IL-10- expressing B cells on day 7 (%IL-10+ B cells around 95%) that do not produce the pro- inflammatory cytokine IL-6. These cells additionally express the suppressive molecule PD- L1.

Example 11 : Establishment of a feeder cell-free expansion protocol to generate an almost pure population of IL-10-expressing B cells

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) or stimulated with an antibody-agonist of CD40 (clone FGK4.5) and medium was supplemented with IL-4/BAFF, CpG/DMSO or CpG/N6 for 4 additional days. B cells from C57BL/6 mice were used to determine autofluorescence in eGFP channel and IL-10.eGFP-IL-6.tdTomato mice were used to monitor IL-10 and IL-6 expression by flow cytometry. Representative dot plots gated on viable/singlets/CD457CD19 + are shown. Data are presented as mean ± SD. n>4 independent experiments. The results are presented in Figure 10.

Conclusion: the two steps culture protocol (step 1 : CpG+N6 for 3 days; step 2: CpG+N6+anti-CD40 for 4 days) induces an almost pure population of IL-10-producing B cells.

Example 12: The established protocol generates an almost pure population of IL-10- expressing B cells from various starting B cell subsets

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and total CD19 + B cells, follicular B cells (CD197CD21 l0W 7CD23 hi9 7CD1 d l0W ) and marginal zone B cells (CD197CD21 hi9 7CD23 l0W/ 7CD1d hi ) were purified by FACS. Total CD19 + blood B cells were directly purified by FACS after red blood cell lysis. Cell were stimulated with CpG/N6 for 3 consecutive days and next transferred on top of CD40L-expressing feeder cells (L47). Medium was supplemented in this second step with IL-4/BAFF, CpG/DMSO or CpG/N6 for 4 additional days. B cells from C57BL/6 mice were used to determine autofluorescence in eGFP channel and IL-10.eGFP-IL-6.tdTomato mice were used to monitor IL-10 expression by flow cytometry. The percentage of viable/singlets/CD197IL- 10 + is shown. Data are presented as mean ± SD. n>3 independent experiments. The results are presented in Figure 11 .

Conclusion: N6 can induce quasi-pure populations of IL-10-expressing B cells starting from any B cell subset.

Example 13: The IL-10-expressing B cells induced by TLR9 agonist, CD40 agonist, and N6 have acquired a stable IL-10-expression phenotype

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) and medium was supplemented with IL-4/BAFF, CpG/DMSO or CpG/N6 for 4 additional days. The B cells were then extensively washed, and re-cultured as follows: left unstimulated (RPMI) or stimulated with various stimuli as indicated for 3 additional days and analyzed daily for IL-10 and IL-6 expression by flow cytometry. B cells from C57BL/6 mice were used to determine autofluorescence in eGFP channel and IL-10.eGFP-IL-6.tdTomato mice were used to monitor IL-10 and IL-6 expression by flow cytometry. Data are presented as mean ± SD. n>3 independent experiments. The data show the percentage of IL-10 and IL-6 expressing B cells at indicated days, depending on the culture condition of origin. The results are presented in Figures 12 A to 12G. Conclusion: IL-10-expressing B cells induced in the 7-day long, two-step protocol, display a stable IL-10 expression upon subsequent exposure to various stimuli in the absence of N6, suggesting a memorization of IL-10 expression.

Example 14: N6 induces a quick and specific remodeling of DNA accessibility (including at IL-10 gene) in murine B cells

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) and medium was supplemented with IL-4/BAFF, CpG/DMSO or CpG/N6 for 4 additional days. Cells were harvested at indicated time- points during this culture scheme, and nuclei were prepared by treating them with Tn5 transposase before subjecting the samples to ATAC-sequencing. Principal Component Analysis (PCA) of the samples is shown (left panel). Accessibility of the 1110 genomic region is also shown (right panel). The results are presented in Figure 13.

Conclusion: N6 induces the rapid remodeling of chromatin accessibility in treated cells, in particular leading to the higher accessibility of the 1110 locus.

Example 15: B cells expanded with L47/CpG/N6 protect mice from EAE development in a prophylactic setting

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) and medium was supplemented with IL-4/BAFF, CpG/DMSO or CpG/N6 for 4 additional days. The different activated B cells were adoptively transferred at day -1 before EAE induction by a single intravenous injection of 10 7 cells per mouse. EAE was induced by immunizing the mice with MOG ( 33. 55) peptide followed by PTX injection at DO and D2. Mice were monitored daily based on the following scores: 0=healthy; 1 =flaccid tail; 2=impaired righting reflex and/or abnormal gait; 3=partial hind leg paralysis; 4=total hind leg paralysis; 5=hind leg paralysis with partial front leg paralysis. Weight was monitor daily and weight loss (%) is presented. Cumulative and maximal scores are also shown. Data are presented as mean ± SD. n>10 mice/group. The results are presented in Figure 14.

Conclusion: IL-10-producing B cells generated with N6 protect recipient mice from EAE in a prophylactic setting.

Example 16: B cells expanded with L47/CpG/N6 protect mice from EAE development in a therapeutic setting

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) and medium was supplemented with CpG/N6 for 4 additional days. Cells were adoptively transferred at day -1 before EAE induction or at D10 or at D17 after EAE induction by a single intravenous injection of 10 7 cells per mouse. EAE was induced by immunizing the mice with MOG ( 33. 55) peptide followed by PTX injection at DO and D2. Mice were monitored daily based on the following scores: 0=healthy; 1 =flaccid tail; 2=impaired righting reflex and/or abnormal gait; 3=partial hind leg paralysis; 4=total hind leg paralysis; 5=hind leg paralysis with partial front leg paralysis. Weight was monitor daily and weight loss (%) is presented. Cumulative and maximal scores are also shown. Data are presented as mean ± SD. n>10 mice/group. The results are presented in Figure 15.

Conclusion: IL-10-producing B cells generated with N6 protect recipient mice from EAE in a therapeutic setting upon administration to recipients either just before the onset of clinical signs (day 10) or at the peak of the disease (day 17).

Example 17: The therapeutic effect of B cells expanded with L47/CpG/N6 in EAE requires their IL-10 expression and is independent of their MHC-II expression

Resting mouse B cells from WT B6, H1 (J 1 ' or H2-AB 1 mice were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) and medium was supplemented with CpG/N6 for 4 additional days. EAE was induced by immunizing the mice with MOG ( 33. 55) peptide followed by PTX injection at DO and D2. B cells were adoptively transferred at D17 by a single intravenous injection of 10 7 cells per mouse. Mice were monitored daily based on the following scores: 0=healthy; 1 =flaccid tail; 2=impaired righting reflex and/or abnormal gait; 3=partial hind leg paralysis; 4=total hind leg paralysis; 5=hind leg paralysis with partial front leg paralysis. Weight was monitor daily and weight loss (%) is presented. Cumulative and maximal scores are also shown. Data are presented as mean ± SD. n>10 mice/group. The results are presented in Figure 16.

Conclusion: IL-10-producing B cells generated with N6 protect recipient mice from EAE in a therapeutic setting via an IL-10-dependent mechanisms, and independently of any cognate interaction with CD4 + T cells.

Example 18: B cells expanded with L47/CpG/N6 can therapeutically restore recovery from EAE in mice depleted of CD4 + FOXP3 + Tregs that otherwise develop a severe chronic disease

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) and medium was supplemented with CpG/N6 for 4 additional days. B cells were adoptively transferred at D-1 before EAE induction or at D10 or at D17 after EAE induction by a single intravenous injection of 10 7 cells per mouse. CD25 + Tregs were antibody-depleted by a single intravenous injection of 200pg anti-CD25 (PC.61.5.3 clone) 5 days before EAE induction. EAE was induced by immunizing the mice with MOG(33-55) peptide followed by PTX injection at DO and D2. Mice were monitored daily based on the following scores: 0=healthy; 1 =flaccid tail; 2=impaired righting reflex and/or abnormal gait; 3=partial hind leg paralysis; 4=total hind leg paralysis; 5=hind leg paralysis with partial front leg paralysis. Weight was monitor daily and weight loss (%) is presented. Cumulative and maximal scores are also shown. Data are presented as mean ± SD. n>5 mice/group. The results are presented in Figure 17.

Conclusion: IL-10-producing B cells generated with N6 restrain EAE induction in Treg- depleted mice, and therapeutically restore recovery from disease upon administration at the peak of clinical signs.

Example 19: B cells expanded with L47/CpG/N6 accumulate in the CNS during EAE where they show an IL-10-producing plasmocyte phenotype

Resting mouse B cells from IL-10.eGFP-IL-6.tdTomato mice were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) and medium was supplemented with CpG/N6 for 4 additional days. 10 7 Ly5.2 + B cells from WT mice and IL-10.eGFP-IL-6.tdTomato mice were adoptively transferred in Ly5.1 recipient at D-1 before EAE induction. EAE was induced by immunizing the mice with MOG ( 33. 55) peptide followed by PTX injection at DO and D2. Indicated organs were harvested at D7, D14 and D28. Percentage of infiltrating CD45.2 + donor-derived B cells is shown. Data are presented as mean ± SD. n=4 mice/time-point. Representative dot plots and histograms are shown at D28. Square denotes CD45.2 infiltrating B cells. Dots plots are gated on viable/singlet cells. Dark and light histograms represent autofluorescence (obtained from WT Ly5.2 + B cells treated mice) and IL-10 expression (obtained from Ly5.2 + IL-10.eGFP- IL-6.tdTomato B cells treated mice) respectively. The results are presented in Figure 18.

Conclusion: IL-10-producing B cells generated with N6 give rise to and persist as IL10- producing-plasmocytes in the CNS of treated mice.

Example 20: B cells expanded with L47/CpG/N6 rapidly enter the inflamed CNS of treated mice during EAE where they locally persist

Resting mouse B cells from Ly5.1 mice were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) and medium was supplemented with CpG/N6 for 4 additional days. EAE was induced by immunizing the mice with MOG ( 33. 55) peptide followed by PTX injection at DO and D2. 10 7 Ly5.1 + B cells were adoptively transferred in CD45.2 recipient at D17 after EAE induction. Brain and spinal cord were harvested and content analyzed at D19 and D30. Percentage of infiltrating CD45.1 + B cells is shown. Data are presented as mean ± SD. n=4 mice/time- point. Representative dot plot is shown at D28. Square denotes CD45.1 infiltrating eBregs. Dots plot is gated on viable/singlet cells. The results are presented in Figure 19. Conclusion: IL-10-producing B cells generated with N6 rapidly home to the CNS of treated mice upon administration at the peak of EAE and locally persist.

Example 21 : B cells expanded with L47/CpG/N6 dampen the activation of CNS- associated macrophages and microglial cells

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) and medium was supplemented with CpG/N6 for 4 additional days. EAE was induced by immunizing mice with MOG ( 33. 55) peptide followed by PTX injection at DO and D2. They received 10 7 B cells adoptively transferred at D17 after EAE induction. Brain and spinal cord were harvested at D19 and D30. Activation of CNS- associated macrophages (CAMs, CD45 high /CD1 1 b hl9h /CX3CR1 int /Ly6C + ) and microglial cells (CD45 int /CD1 1 b int /CX3CR1 hl9h /Ly6C ) was analyzed by flow cytometry. Mean fluorescence intensity of P2RY12 expression in and percentage of CD86 + and CD68 + microglial cells in indicated organs is shown. Percentage of double CD867MHC-IF CAMs in indicated organs is shown. Data are presented as mean ± SD. n=4 mice/time-point. The results are presented in Figure 20.

Conclusion: IL-10-producing plasmocytes derived from B cells generated with N6 locally suppress the activation of microglia and monocytes/ macrophages in the CNS of treated mice.

Example 22: B cells expanded with L47/CpG/N6 express IL-10 and display a plasmocyte transcriptome in the CNS of mice treated by adoptive B cell therapy at the peak of EAE, whereas microglia and CNS-associated macrophages are the major IL-10 receptor-expressing cell types locally

Resting mouse B cells from CD45.1 + mice were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) and medium was supplemented with CpG/N6 for 4 additional days. EAE was induced by immunizing CD45.2 + mice with MOG ( 33. 55) peptide followed by PTX injection at day 0 and day 2. They received 10 7 CD45.1 + B cells (activated as indicated) adoptively transferred at day 17 after EAE induction. At day 19, brain and spinal cords of EAE mice were collected, CD45 + cells were isolated by FACS and processed by single cell RNAseq (10x Genomics). Adoptively transferred B cells were identified through CITE-Seq using an anti-CD45.1. Data show that these cells express Cd19, 1110, Irf4, Prdml, and Xbp1, highlighting their IL-10- expressing plasmocytes characteristics. Microglia were identified through the expression of Cx3cr1. Data show that together with CNS-associated macrophages, they are the major cell types expressing the two chains of the IL- 10 receptor, namely 111 Ora and 111 Orb. The results are presented in Figure 21 .

Conclusion: IL-10-producing B cells generated with N6 rapidly home to the CNS following administration where they display a transcriptome profile characteristic of plasmocytes.

Example 23: B cells expanded with L47/CpG/N6 target CNS-associated macrophages and microglial cells

Resting mouse B cells from Ly5.1 mice were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) and medium was supplemented with CpG/N6 for 4 additional days. EAE was induced by immunizing the mice with MOG ( 33. 55) peptide followed by PTX injection at DO and D2. 10 7 B cells were adoptively transferred into CD45.2 recipient at D17 after EAE induction. Brain and spinal cord were harvested at D19. CD45 + total cells were purified by FACS and subjected to library preparation for scRNA-sequencing analysis (10x Genomics). Subsets of cells were identified as described by Jordao et a/.(2019, Science 363: 6425), and analyzed for differentially expressed genes between non-treated (NT) and eBreg-treated (T) conditions. Genes expression in treated samples are presented in Figures 26 to 53B. Analyzed organs are indicated (SC: spinal cord, brain). daMG: disease-associated microglia; dapvMF: disease-associated perivascular macrophages; hMG: homeostatic microglia; hpvMF: homeostatic perivascular macrophages; pMC: perivascular and parenchymal monocyte-derived cells. Cells from hMG2 subset were not present in enough quantity to perform analysis.

Conclusion: IL-10-producing plasmocytes derived from B cells generated with N6 locally suppress the activation of microglia and monocytes/ macrophages in the CNS within 2 days of treatment.

Example 24: B cells expanded with L47/CpG/N6 dampen disease severity in an acute DSS-induced colitis model

Resting mouse B cells were isolated from spleen by magnetic depletion of CD43 + cells and stimulated with CpG/N6 for 3 consecutive days. Cells were next transferred on top of CD40L-expressing feeder cells (L47) and medium was supplemented with CpG/N6 for 4 additional days. Cells were injected intravenously one day before treating the mice with 4% DSS in water during 7 consecutive days. Colitis severity was scored by daily observation of the following parameters and scores. Weight loss: 0=no weight loss; 1 = 5%— 10% weight loss; 2= 1 %— 15% weight loss; 3=16%-20% weight loss; 4=>20% weight loss. Stool consistency: 0=normal and well formed; 2= very soft and unformed; 4= watery stool. Blood in stool: 0=normal color; 2=reddish color; 4=bloody. The disease activity index (DAI) was calculated as the combined scores of weight loss, bleeding, stool consistency, with a maximum score of 12. After 7 days mice were killed and colon harvested to measure its length. Data are presented as mean ± SD. n>4 mice/group. The results are presented in Figure 22.

Conclusion: IL-10-producing B cells generated with N6 limit the severity of inflammatory bowel disease in treated mice.

Example 25: Effect of compounds 1 , 4, 7, 22, 30, 31 , 32, 40, 43, 45, 52, 54, 56, 57, 58, 60, 61 , 67, 107, 109, 110, 111 , 116, N4, N6, 117, 120, 124, 136, 145, 153, 157, 366, 371 and 375 on IL-10 induction by human B cells

Human B cells isolated magnetically through negative selection (B Cell Isolation Kit II, human, Miltenyi ref: 130-091 -151 ) were cultivated for 2 consecutive days in the presence of anti-IgM plus CpG in the presence of DMSO (as control) or in the presence of the tested molecule at 0.25 pM final concentration. IL-10 was measured in cell culture supernatants. NS: non stimulated B cell culture condition. The results are presented in Figures 23A and 23B.

Conclusion: We have identified novel molecules analog to N6 that increase IL-10 induction by human B cells.

Example 26: Effect of the induction of human B cells with compounds 1 , 4, 7, 22, 30, 31 , 32, 40, 43,45, 52, 54, 56, 57, 58, 60, 61 , 67, 107, 109, 110, 111 , 116, N4, N6, 117, 120, 124, 136, 145, 153, and 157 on IL-10 production by human B cells

Human B cells isolated magnetically through negative selection (B Cell Isolation Kit II, human, Miltenyi ref: 130-091 -151 ) were cultivated for 2 consecutive days in the presence of anti-IgM plus CpG in the presence of DMSO (as control) or in the presence of the tested molecule at 0.25 pM final concentration. IL-10 was measured in cell culture supernatants. NS: non stimulated B cell culture condition. The results are presented in Figures 24 and 25.

Conclusion: We have identified novel molecules analog to N6 that increase IL-10 production by human B cells.

Example 27: List of genes up-and down-regulated in CpG-stimulated mouse B cells cultivated in the presence of N6 compared to the control condition (no N6)

Mouse B cells were activated for 48h in the presence of CpG with or without N6, and their molecular profile was analyzed using single cell RNAseq (10X Genomics). The table below shows a selection of the top 101 up- and down-regulated genes in N6-treated cells compared to controls. Fold change indicates expression level in N6-treated cells compared to control.

Example 28: List of genes up-and down-regulated in IL-10-expressing B cells compared to IL-10-negative B cells within the culture of B cells stimulated for 48h with CpG in the presence of N6 B cells were activated for 48h in the presence of CpG with N6, and their molecular profile was analyzed using single cell RNAseq (10X Genomics). The transcriptomes of IL-10- positive and IL- 10-negative B cells in these cultures were then analyzed. The table below shows a selection of the top 101 up- and down-regulated genes in IL- 10-positive versus IL-10-negative B cells. Fold change indicates expression level in IL-10-positive cells compared to I L-10-negative B cells.

Example 29: Lists of transcriptional regulators up-regulated and down-regulated in N6-treated CpG-stimulated B cells compared to control cells

B cells were activated for 48h in the presence of CpG with or without N6, and their molecular profile was analyzed using single cell RNAseq (10X Genomics). The table below shows top up- and down-regulated transcriptional regulators in N6-treated cells compared to controls. Fold change indicates expression level in N6-treated cells compared to control.

Example 30: Lists of transcriptional regulators up-regulated and down-regulated in IL-10-positive B cells compared to IL-10-negative B cells from N6-treated cultures at 48h post-activation with CpG B cells were activated for 48h in the presence of CpG with N6, and their molecular profile was analyzed using single cell RNAseq (10X Genomics). The transcriptomes of IL-10- positive and IL- 10-negative B cells in these cultures were then analyzed. Shown is the selection of the top up- and down-regulated transcriptional regulators in IL- 10-positive versus IL-10-negative B cells. Fold change indicates expression level in IL- 10-positive cells compared to IL-10-negative B cells.