Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHOD FOR TREATING AN AUTOIMMUNE AND INFLAMMATORY DISEASE
Document Type and Number:
WIPO Patent Application WO/2022/253794
Kind Code:
A2
Abstract:
The present invention relates to methods for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease (AIID) in a subject.

Inventors:
VIGOLO MICHELE (CH)
MAGNIN MORGANE (FR)
LAMY SEBASTIEN (FR)
LEHAL RAJWINDER (CH)
Application Number:
PCT/EP2022/064684
Publication Date:
December 08, 2022
Filing Date:
May 31, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CELLESTIA BIOTECH AG (CH)
International Classes:
A61K31/44; A61K31/351; A61K31/4412; A61P17/06; A61P19/02; A61P21/04; A61P29/00; A61P37/00
Domestic Patent References:
WO2013093885A12013-06-27
WO2020208139A12020-10-15
WO2012129353A12012-09-27
WO2014047372A12014-03-27
WO2020131998A12020-06-25
WO2005047366A12005-05-26
Foreign References:
US20040116404A12004-06-17
US7342118B22008-03-11
US7795447B22010-09-14
US7951958B22011-05-31
US4935465A1990-06-19
US6342244B12002-01-29
US20060074024A12006-04-06
US20050171328A12005-08-04
Other References:
CANCER RES, vol. 67, no. 5, 2007, pages 1879 - 82
MASSARD C ET AL., ANN ONCOL., vol. 29, no. 9, 2018, pages 1911 - 1917
HUYNH C ET AL., PLOS ONE, vol. 6, no. 9, 2011
HURTADO C ET AL., SCI REP, vol. 9, 2019, pages 10811
ASTUDILLO L. ET AL., CANCER RES, vol. 76, no. 12, 15 June 2016 (2016-06-15), pages 3593 - 603
J. MARCH: "Advanced Organic Chemistry", 1992, JOHN WILEY AND SONS
Attorney, Agent or Firm:
SCHÖLLHORN, ANDREAS (CH)
Download PDF:
Claims:
Claims

1) A T cell modulator (TCM) for use in a method for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease (AIID) in a subject.

2) The T cell modulator (TCM) for use according to claim 1, wherein the T cell modulator (TCM) is a NOTCH signaling pathway inhibitor.

3) The T cell modulator (TCM) for use according to claim 2, wherein the NOTCH signaling pathway inhibitor is selected from the group consisting of a g-secretase inhibitor, a blocking antibody against NOTCH receptors, a blocking antibody against NOTCH ligands, an inhibitor of NOTCH transcription complex and an inhibitor of intracellular trafficking of NOTCH ligands/receptors.

4) The T cell modulator (TCM) for use according to claim 1, wherein the T cell modulator (TCM) is an inhibitor of NOTCH transcription complex.

5) The T cell modulator (TCM) for use according to claim 1, wherein the T cell modulator (TCM) is a compound of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof, wherein X is selected from CH2, CF2, CHF, CO, CHOH, CHO(CI-C3) alkyl, NH, N(CI-C3 alkyl), S, SO and O; wherein Y1, Y2, and Y3 are each independently selected from N and C; wherein Z is NR10R11, wherein R10 and R11 are each independently selected from H and C1-C6 alkyl; wherein R1 is selected from H, halogen, C1-C6 alkyl, C3-C12 cycloalkyl, C3-C12 heterocyclyl, C1- C6 alkoxy, C1-C6 heteroalkyl, C0-C3 alkylOC0-C3 alkyl aryl wherein the aryl is optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl; C0-C3 alkylOC0-C3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3- C12 cycloalkyl, C3-C12 heterocyclyl; and C1-C6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl; wherein R2 is selected from C1-C6 alkyl, C3-C12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl, C(O)R12, C1-C6 alkyl C(O)R12; wherein R3 is selected from H, halogen, C1-C6 alkyl, C1-C6 heteroalkyl, C3-C12 cycloalkyl, C3- C12 heterocyclyl and C1-C6 alkoxy; wherein R4, R5 and R6 are each independently selected from H, halogen, CN, C1-C6 alkoxy, C1- C6-S-alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, C3-C12 cycloalkyl and C3-C12 heterocyclyl; wherein R7 is absent when Y1 is N or is selected from H, halogen, C1-C6 alkyl, C1-C6 heteroalkyl, C3-C12 cycloalkyl, C3-C12 heterocyclyl and C1-C6 alkoxy when Y1 is C; wherein R8 is absent when Y3 is N or is selected from H, halogen, C1-C6 alkyl, C1-C6 heteroalkyl, C3-C12 cycloalkyl, C3-C12 heterocyclyl and C1-C6 alkoxy when Y3 is C; wherein R9 is absent when Y2 is N or is selected from H, halogen, C1-C6 alkyl, C3-C12 cycloalkyl, C3-C12 heterocyclyl, C1-C6 alkoxy, C1-C6 heteroalkyl, C0-C3 alkylOC0-C3 alkyl aryl wherein the aryl is optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl; C0-C3 alkylOC0-C3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl; and C1-C6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl; wherein R12is selected from H, NH2, NHCI-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl and C3-C12 heterocyclyl; and optionally one or more pharmaceutically acceptable diluents, excipients or carriers.

6) The T cell modulator (TCM) for use according to claim 1, wherein the T cell modulator (TCM) is a compound of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof, wherein X is selected from CH2, CF2, CHF, CO, CHOH, CHO(CI-C3) alkyl, NH, N(CI-C3 alkyl), S, SO and O; wherein Y1, Y2, and Y3 are each independently selected from N and C; wherein Z is NR10R11, wherein R10 and R11 are each independently selected from H and C1-C6 alkyl; wherein R1 is selected from H, halogen, C1-C6 alkyl, C3-C12 cycloalkyl, C3-C12 heterocyclyl, C1- C6 alkoxy, C1-C6 heteroalkyl, C0-C3 alkylOC0-C3 alkyl aryl wherein the aryl is optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl; C0-C3 alkylOC0-C3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3- C12 cycloalkyl, C3-C12 heterocyclyl; and C1-C6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl; wherein R2 is selected from C1-C6 alkyl, C3-C12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl, C(O)R12, C1-C6 alkyl C(O)R12; wherein R3 is selected from H, halogen, C1-C6 alkyl, C1-C6 heteroalkyl, C3-C12 cycloalkyl, C3- C12 heterocyclyl and C1-C6 alkoxy; wherein R4, R5 and R6 are each independently selected from H, halogen, CN, C1-C6 alkoxy, C1- C6-S-alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, C3-C12 cycloalkyl and C3-C12 heterocyclyl; wherein R7 is absent when Y1 is N or is selected from H, halogen, C1-C6 alkyl, C1-C6 heteroalkyl, C3-C12 cycloalkyl, C3-C12 heterocyclyl and C1-C6 alkoxy when Y1 is C; wherein R8 is absent when Y3 is N or is selected from H, halogen, C1-C6 alkyl, C1-C6 heteroalkyl, C3-C12 cycloalkyl, C3-C12 heterocyclyl and C1-C6 alkoxy when Y3 is C; wherein R9 is absent when Y2 is N or is selected from H, halogen, C1-C6 alkyl, C3-C12 cycloalkyl, C3-C12 heterocyclyl, C1-C6 alkoxy, C1-C6 heteroalkyl, C0-C3 alkylOC0-C3 alkyl aryl wherein the aryl is optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl; C0-C3 alkylOC0-C3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl; and C1-C6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl; wherein R12is selected from H, NH2, NHCI-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl and C3-C12 heterocyclyl; and optionally one or more pharmaceutically acceptable diluents, excipients or carriers; with the proviso that if R2 is C1-C6 alkyl or C3-C12 cycloalkyl, Y1 is N.

7) The T cell modulator (TCM) for use according to claim 1, wherein the T cell modulator (TCM) is selected from the group consisting of 6-((4'-fluoro-[l,r-biphenyl]-4-yl)oxy)-2- methylpyridin-3 -amine, N-methyl-6-(4-(thiazol-5-yl)phenoxy)pyridin-3-amine, and 6-(4-(tert- butylphenoxy)pyri din-3 -amine or pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof.

8) The T cell modulator (TCM) for use according to claim 1, wherein the T cell modulator (TCM) is 6-(4-(tert-butylphenoxy)pyri din-3 -amine or pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof.

9) The T cell modulator (TCM) for use according to any one of claims 1-8, wherein the autoimmune and inflammatory disease is driven by a cytokine selected from the group consisting of IL-6, IFN-g, IL-17, IL12a, IL-10, and TGF-b, or any combination thereof.

10) The T cell modulator (TCM) for use according to any one of claims 1-8, wherein the autoimmune and inflammatory disease is selected from the group consisting of acute, chronic and steroid refractory Graft-versus-Host-Disease (GvHD), Type 1 diabetes, inflammatory bowel disease, rheumatoid arthritis, sjogren syndrome, asthma, allergies, atopic dermatitis, psoriasis, inflammation in organ transplantation, organ transplant rejection, Crohn’s disease, Ulcerative colitis, Psoriatic arthritis, Systemic lupus erythematosus/Lupus nephritis, Multiple sclerosis, Neuromyelitis optica, Pemphigus vulgaris, Chronic spontaneous urticaria,

Myasthenia gravis, Uveitis, autoimmune hepatitis, primary biliary cirrhosis, Behcet disease, Kawasaki disease, vasculitis, Felty syndrome, Guillain-Barre Syndrome, Autoimmune polyglandular syndrome 1, polyglandular syndrome type 2, Autoimmune pancreatitis, Celiac disease, Addison's disease, and autoimmune thyroiditis.

11) The T cell modulator (TCM) for use according to any one of claims 1-8, wherein the autoimmune and inflammatory disease is driven by Thl cells and is selected from the group consisting of acute GvHD, chronic GvHD, Type 1 diabetes, Inflammatory bowel disease, Crohn’s disease, Ulceritis, Rheumatoid arthritis, Psoriasis, Psoriatic arthritis, Multiple sclerosis, Sjogren syndrome.

12) The T cell modulator (TCM) for use according to any one of claims 1-8, wherein the autoimmune and inflammatory disease is driven by Th2 cells and is selected from the group consisting of Atopic dermatitis and Asthma or wherein the autoimmune and inflammatory disease is driven by Thl 7 cells and is Psoriasis.

13) The T cell modulator (TCM) for use according to any one of claims 1-8, wherein the autoimmune and inflammatory disease is driven by cells of innate immune system selected from the group consisting of macrophages and dendritic cells. 14) The T cell modulator (TCM) for use according to any one of claims 1-8, wherein the autoimmune and inflammatory disease is Graft-versus-Host-Disease (GvHD). 15) The T cell modulator (TCM) for use according to any one of claims 1-8, wherein the autoimmune and inflammatory disease is acute, chronic and steroid refractory Graft-versus- Host-Disease (GvHD).

16) A pharmaceutical composition for use in a method for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease comprising the T cell modulator

(TCM) according to any one of claims 1-8 and optionally one or more pharmaceutically acceptable diluents, excipients or carriers.

17) A kit for use in a method for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease in a subject, comprising comprising a container and a package insert, wherein the container comprises at least one dose of a medicament comprising the T cell modulator (TCM) according to any one of claims 1-8 or the pharmaceutical composition of claim 16, and the package insert comprises optionally instructions for treating a subject for an autoimmune and inflammatory disease (AIID) using the medicament.

Description:
Method for treating an autoimmune and inflammatory disease

Field of the invention

The present invention relates to methods for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease (AIID) in a subject.

Background of the invention

Autoimmune and inflammatory diseases (AIIDs) encompass >150 diseases, each resulting from a host immune response against self or non-self antigens, and affect up to 50 million individuals in the U.S. alone. AIIDs can be broadly characterized into five sub-categories based on type of immune response (e.g., adaptive vs. innate) and origin of the antigen (e.g., self vs. non-self) the host immune system is reacting to. The five sub-categories are inflammatory diseases; autoimmune diseases; allergic diseases; autoinflammatory diseases; and Graft-versus-Host- Disease (GvHD)/transplant rejection. New treatments targeting AIIDs are being investigating e.g., NLRP3/inflammasome inhibitors and antigen-specific immune tolerizing approaches, in order to meet the high medical need.

Summary of the invention

It has now unexpectedly been found that T cell modulators are useful for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease (AIID). Surprisingly, it has been found that the effect of T cell modulators in prevention, delay or treatment of AIID can be mediated via direct action on conventional T cells or immune suppressive regulatory T cells (Treg cells). T cell modulators such as 6-(4-tert-butylphenoxy)pyridin-3-amine led to expansion of Treg cells in human and mouse PBMC culture stimulation and treatment experiments. In addition, T cell modulators resulted in a significant lower cumulative GvHD score, prevented GvHD andenhanced GvHD free survival in vivo compared to the control.

Taking these unexpected findings into account, the inventors herewith provide the present invention in its following aspects. In a first aspect, the present invention provides a T cell modulator (TCM) for use in a method for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease (AIID) in a subject.

In a second aspect, the present invention provides a pharmaceutical composition comprising a T cell modulator (TCM) and a pharmaceutically acceptable diluent, excipient, or carrier for use in a method for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease (AIID) in a subject.

In a third aspect, the present invention provides kit of parts comprising a container and a package insert, wherein the container comprises at least one dose of a medicament comprising a T cell modulator (TCM), and the package insert comprises optionally instructions for treating a subject for an autoimmune and inflammatory disease using the medicament.

In a fourth aspect, the present invention provides a T cell modulator (TCM) for use in a method for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease (AIID) in a subject in combination with standard of care or immunosuppressive drugs.

Brief description of the figures

Figure 1: In vitro 6-(4-tert-butylphenoxy)pyridin-3 -amine treatment causes an expansion of Treg cells in activated mouse splenocytes. Mouse splenocytes were harvested and single cell suspension stimulated in vitro with anti-CD3 and anti-CD28 antibodies for 96 hours. Treatment with 6-(4-tert-butylphenoxy)pyri din-3 -amine, during the same interval of time, caused an expansion of immunosuppressive regulatory T cells (Tregs). Each bar represents the mean ± SD (N=3 replicates).

Figure 2: In vivo 6-(4-tert-butylphenoxy)pyridin-3-amine dosing leads to an expansion of Treg cells in mouse spleen. C57BL6 wild type mice were treated with 6-(4-tert- butylphenoxy)pyridin-3 -amine at 60 mg/kg dose (QD). Treg cells (Eϋ4 + Oϋ25 w§:1i ROCR3 + ) number (a) and frequency (b) was significantly increased upon mice dosing with 6-(4-tert- butylphenoxy)pyridin-3 -amine. (*) p<0.05. Each bar represents the mean ± SEM (N=3 replicates). Figure 3: In vivo 6-(4-tert-butylphenoxy)pyridin-3-amine dosing leads to an upregulation of anti-inflammatory cytokine IL-10 mRNA in mouse spleen. Quantitative RT-PCR using mRNA extracted from splenocytes in vehicle and 6-(4-tert-butylphenoxy)pyri din-3 -amine treated mice showed an upregulation of immune suppressive cytokine IL-10 transcript. Each bar represents the mean ± SD (N=3 replicates). (*) p<0.05

Figure 4: In vivo 6-(4-tert-butylphenoxy)pyridin-3-amine dosing leads to a downregulation of pro-inflammatory cytokine IL-12a mRNA in mouse spleen. Quantitative RT-PCR using mRNA extracted from splenocytes in vehicle and 6-(4-tert-butylphenoxy)pyri din-3 -amine treated mice showed a downregulation of pro-inflammatory cytokine IL-12a transcript. Each bar represents the mean ± SD (N=3 replicates). (**) p <0.01

Figure 5: 6-(4-tert-butylphenoxy)pyridin-3-amine prevents development of GvHD. Following allogeneic bone marrow + splenocyte transplantation, mice were treated with vehicle, 6-(4-tert- butylphenoxy)pyri din-3 -amine and cyclosporin A. Compared to 40% vehicle treatment and 30% cyclosporin A treatment, 80% of 6-(4-tert-butylphenoxy)pyridin-3 -amine animals remained GvHD free.

Figure 6: 6-(4-tert-butylphenoxy)pyridin-3-amine downregulates cytokines involved in GvHD. Following allogeneic bone marrow + splenocyte transplantation, mice were treated with vehicle and 6-(4-tert-butylphenoxy)pyridin-3 -amine. Compared to vehicle treatment, 6-(4-tert- butylphenoxy)pyridin-3 -amine treatment led to a downregulation of IFNG, IL-17 and IL-6.

Detailed description of the invention

As outlined above, the present invention provides methods for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease (AIID) in a subject.

Thus, in a first aspect the present invention provides a T cell modulator (TCM) for use in a method for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease (AIID) in a subject.

For the purposes of interpreting this specification, the following definitions will apply and whenever appropriate, terms used in the singular will also include the plural and vice versa. It is to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. The terms "comprising", "having", and "including" are to be construed as open-ended terms (i.e., meaning "including, but not limited to,") unless otherwise noted.

Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein unless incompatible therewith. All of the features disclosed in this specification (including any accompanying claims, abstract and drawings), and/or all of the steps of any method or process so disclosed, may be combined in any combination, except combinations where at least some of such features and/or steps are mutually exclusive. The invention is not restricted to the details of any foregoing embodiments. The invention extends to any novel one, or any novel combination, of the features disclosed in this specification (including any accompanying claims, abstract and drawings), or to any novel one, or any novel combination, of the steps of any method or process so disclosed.

The term “autoimmune and inflammatory disease” also abbreviated herein as (AIID) refers to diseases caused by innate immune cells or adaptive immune cells. Autoimmune and inflammatory diseases as referred herein include alloimmune diseases in which an immune response is triggered against non-self antigens by members of the same species like Graft vs Host disease (GvHD) and allogeneic organ transplant rejection. Innate immune cells are cells of the immune system that are known to be activated by one or more agents (e.g., allergens, chemicals produced upon injury (e.g., opioids and alcohols), polymyxins, crosslinked IgE, crosslinked complement proteins, cytokines produced by T cells or other immune cells (e.g., interferon-g), DAMPs, or PAMPs) that activate downstream signaling pathway(s) in the innate immune cell and result in the activation of one or more functions of the innate immune cell. Adaptive immune cells relate to cells of the adaptive immune system. The adaptive immune system protects higher organisms against infections and other pathological events that may be attributable to foreign substances, using adaptive immune receptors, the antigen-specific recognition proteins that are expressed by hematopoietic cells of the lymphoid lineage and that are capable of distinguishing self from non-self molecules in the host. These lymphocytes may be found in the circulation and tissues of a host, and their recirculation between blood and the lymphatics has been described, including their extravasation via lymph node high endothelial venules, as well as at sites of infection, inflammation, tissue injury and other clinical insults.

The term “autoimmune and inflammatory disease driven by” or “diseases driven by” refers to AIIDs which are caused due to an aberrant activation of cells of immune system (innate or adaptive) against self-antigen or foreign antigen leading to damage to organs. The term “autoimmune and inflammatory disease driven by” or “diseases driven by” have the same meaning as “autoimmune and inflammatory disease caused by” or “diseases caused by” and the respective terms are used synonomously herein.

The term “inflammation” of the “inflammatory disease” refers to one of the biological reactions to harmful stimulants in living tissues, for example, pathogenic microorganisms, damaged cells, stimulants and the like. Inflammation can be caused by a variety of causes, such as infection by microorganisms or injuries, surgery, burns, frostbite, electrical stimulation, and chemicals. The inflammatory disease refers to a disease that involves inflammation as a major lesion. The Inflammatory disease may be selected from the group consisting of sepsis, gastritis, enteritis, nephritis, hepatitis, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis, hypersensitivity colorectal syndrome, inflammatory pain, migraine, headaches, back pain, fibromyalgia, fascial disease, viral infection, bacterial infection, fungal infection, bums, wounds due to surgery, including dental surgery, or accident, prostaglandin excessive syndrome, atherosclerosis, gout, Hodgkin's disease, pancreatitis, conjunctivitis, iritis, sclerotitis, uveitis, and eczema.

The term “autoimmune disease” refers to a disease in which an immune-function abnormality occurs, resulting in immune cells in the body attacking organs or tissues of the body. The autoimmune disease can be categorized as a disease associated with organ-specific autoantibodies and an organ non-specific (systemic) disease. The autoimmune disease may be selected from the group consisting of hemophagocytic lymphohistiocytosis, systemic lupus erythematosus, Kikuchi disease, vasculitis, adult onset Still's disease, rheumatoid arthritis, inflammatory myositis, Behcet disease, IgG4-related disease, Sjogren syndrome, giant cell arteritis, temporal arteritis, Type 1 diabetes, atopic dermatitis, Crohn's disease, systemic sclerosis, psoriasis, multiple sclerosis, and Graves hyperthyroidism. The term “Graft-versus-Host-Disease (GvHD)” refers to acute, chronic and steroid refractory Graft-versus-Host-Disease (GvHD) and includes graft-versus-host disease (GvHD) in patients undergoing allogenic hematopoietic stem cell (allow-bone marrow) transplantation (allo- HSCT).

The term "T cell modulator" also abbreviated herein as (TCM) refers to a molecule that modulates (e.g., increases or decreases) the numbers, proliferation, viability and/or activity of immune cells.

The term "NOTCH signaling pathway inhibitor" as used herein refers to a compound that is inhibiting the NOTCH signalling pathway. NOTCH signaling pathway inhibitors as used herein include a compound of formula (I) as shown below, a g-secretase inhibitor, a blocking antibody against NOTCH receptors, a blocking antibody against NOTCH ligands, an agent that blocks trafficking of NOTCH ligands/receptors and an inhibitor of NOTCH transcription complex.

The NOTCH signalling pathway represents a critical component in the molecular circuits that control cell fate during development, cell survival and cell proliferation (Shih IeM, Wang TL in Cancer Res 2007;67(5): 1879-82). Aberrant activation of this pathway contributes to tumorigenesis. The NOTCH family members are being revealed as oncogenes in an ever- increasing number of cancers. The role of NOTCH in human cancer has been highlighted recently by the presence of activating mutations and amplification of NOTCH genes in human cancer and by the demonstration that genes/proteins in the NOTCH signalling pathway could be potential therapeutic targets. It has become clear that one of the major therapeutic targets in the NOTCH pathway are the NOTCH receptors, in which g-secretase inhibitors prevent the generation of the oncogenic (intracellular) domain of NOTCH molecules and suppress the NOTCH activity. Though significant progress has been made in dissecting the complex workings of this signalling pathway, there are very limited options available for developing novel NOTCH inhibitors. However, the pioneering class of NOTCH inhibitors is already in clinical trials for few cancer types, such as g-secretase inhibitors AL101 and AL102 from Ayala Pharma (formerly BMS 906024 and BMS-986115 respectively), LY3039478 from Eli Lilly and, PF-03084014 (Nirogacestat) from Springworks Therapeutics, a synthetic small molecule, which inhibits the NOTCH signalling pathway, which may result in induction of growth arrest in tumor cells in which the NOTCH signalling pathway is overactivated.

The term “NOTCH receptors” as used herein refers to the NOTCH receptors NOTCH1, NOTCH2, NOTCH3 and NOTCH4. “A blocking antibody against NOTCH receptors” is a compound specifically binding to the extra-cellular part of the NOTCH receptors hence preventing either constitutive activation of the pathway or activation through ligand binding.

The term “NOTCH ligands” as used herein refers to the NOTCH ligands Delta like 1, Delta like 3, Delta like 4, Jagged, 1, Jagged 2. “A blocking antibody against NOTCH ligands” is a compound specifically binding to one of the ligands hence blocking binding to NOTCH receptors and preventing subsequent activation of the pathway.

The terms “an agent that blocks trafficking of NOTCH ligands/receptors” and “an inhibitor of intracellular trafficking of NOTCH ligands/receptors” which are used herein synonymously refer to a compound that prevents intracellular trafficking of NOTCH ligands or receptors (e.g inhibitors of SERCA2).

The term “inhibitor of NOTCH transcription complex” as used herein refers to a compound that prevents components of NOTCH transcription complex from assembling properly into a functional complex. Non-limiting examples of inhibitors of NOTCH transcription complex include compounds of formula I such as 6-(4-tert-butylphenoxy)pyri din-3 -amine, 6-((4'- fluoro-[l, r-biphenyl]-4-yl)oxy)-2-methylpyridin-3 -amine, and N-methyl-6-(4-(thiazol-5- yl)phenoxy)pyri din-3 -amine; and compounds such as 2-(2-fluorophenoxy)-4-( 1 -methyl- 1 H- pyrazol-5-yl)benzamide and 2-[2-Methoxy-4-[(4-oxo-2-thioxo-5- thiazolidinylidene)methyl]phenoxy]-acetic acid ethyl ester.

The term “gamma secretase inhibitor” (GSI) as used herein refers to a compound that blocks activity of the gamma secretase complex of proteins. Non-limiting examples of gamma secretase inhibitors include AL-101, AL-102, LY3039478, RO4929097, MK-0752, and PF- 03084014. The term “JAK/STAT signaling inhibitor” as used herein refers to inhibitors of JAK1-3 and TYK kinases or inhibitors of downstream mediators of JAK signaling (e.g inhibitors of STAT1-6 proteins). Non-limiting examples of JAK/STAT inhibitors include Ruxolitinib, Itacitinib, PF-06263276, AZD4205, BMS-911543, Ilginatinib, Tofacitinib, Oclacitinib, Fedratinib, Baricitinib, Solcitinib and Abrocitinib.

The term inhibitor of pro-inflammatory cytokines” as used herein refers to inhibitors of IL-6, IL-6R, IFNG, TNFa, IL-17, IL-4 and IL-13. Non-limiting examples include, Siltuximab, Sarilumab, Tocilizumab, Infliximab, Adalimumab, Etanercept, Secukinumab, Ixekizumab, Brodalumab, Omalizumab, Dupilumab, Pascolizumab Anrukinzumab, Lebrikizunab and Tralokinumab.

The term “enhancer of anti-inflammatory cytokines” as used herein refers to IL-10, TGF , low dose IL-2 and IL-2 mutein.

The terms "individual," "subject" or "patient" are used herein interchangeably. In certain embodiments, the subject is a mammal. Mammals include, but are not limited to primates (including human and non-human primates). In a preferred embodiment, the subject is a human.

The term "pharmaceutically acceptable diluents, excipients or carriers" as used herein refers to diluents, excipients or carriers that are suitable for use with humans and/or animals without undue adverse side effects (such as toxicity, irritation, and allergic response) commensurate with a reasonable benefit/risk ratio. "Diluents" are agents which are added to the bulk volume of the active agent making up the solid composition. As a result, the size of the solid composition increases, which makes it easier to handle. Diluents are convenient when the dose of drug per solid composition is low and the solid composition would otherwise be too small. "Excipients" can be binders, lubricants, glidants, coating additives or combinations thereof. Thus, excipients are intended to serve multiple purposes. "Carriers" can be solvents, suspending agents or vehicles, for delivering the instant compounds to a subject.

The term "pharmaceutically acceptable salt" of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. Such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxy- benzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4- chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]-oct-2-enel-carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxy naphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e. g. an alkaline metal ion, an alkaline earth metal ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.

The term "about" as used herein refers to +/- 10% of a given measurement.

Thus, in a first aspect the present invention provides a T cell modulator (TCM) for use in a method for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease (AIID) in a subject.

T cell modulator (TCM)

In one embodiment the T cell modulator (TCM) is selected from the group consisting of a NOTCH signaling pathway inhibitor, JA /STAT signaling inhibitor, inhibitors of pro- inflammatory cytokines and enhancer of anti-inflammatory cytokines.

In one embodiment the T cell modulator (TCM) is a NOTCH signaling pathway inhibitor.

In one embodiment the T cell modulator (TCM) is a NOTCH signaling pathway inhibitor selected from the group consisting of a g-secretase inhibitor, a blocking antibody against NOTCH receptors, a blocking antibody against NOTCH ligands, an inhibitor of NOTCH transcription complex and an inhibitor of intracellular trafficking of NOTCH ligands/receptors. In one embodiment the T cell modulator (TCM) is selected from the group consisting of a y- secretase inhibitor and an inhibitor of NOTCH transcription complex and is preferably an inhibitor of NOTCH transcription complex.

In one embodiment the T cell modulator (TCM) is selected from the group consisting of a y- secretase inhibitor, IMR-1, IMR-1 A and a compound of formula (I) as shown below. Preferably the g-secretase inhibitor is selected from the group consisting of AL-101, AL-102, LY3039478, RO4929097, MK-0752, and PF-03084014, more preferably selected from the group consisting of AL-101, AL-102, PF-03084014 and LY3039478.

In one embodiment the T cell modulator (TCM) is selected from the group consisting of a y- secretase inhibitor and a compound of formula (I) as shown below. Preferably the g-secretase inhibitor is selected from the group consisting of AL-101, AL-102, LY3039478, RO4929097, MK-0752, and PF-03084014, more preferably selected from the group consisting of AL-101, AL-102, PF-03084014 and LY3039478.

In a further embodiment the T cell modulator (TCM) is a g-secretase inhibitor; or a compound selected from the group consisting of 2-(2-fluorophenoxy)-4-( 1 -methyl- 1 //-pyrazol-5- yl)benzamide, 2-[2-Methoxy-4-[(4-oxo-2-thioxo-5-thiazolidinylidene)methyl] phenoxy]-acetic acid ethyl ester, IMR-1, IMR-1 A and a compound of formula (I) as shown below.

In a further embodiment the T cell modulator (TCM) is a g-secretase inhibitor; or a compound selected from the group consisting of 2-(2-fluorophenoxy)-4-( 1 -methyl- 1 //-pyrazol-5- yl)benzamide, 2-[2-Methoxy-4-[(4-oxo-2-thioxo-5-thiazolidinylidene)methyl] phenoxy]-acetic acid ethyl ester and a compound of formula (I) as shown below.

In a further embodiment the T cell modulator (TCM) is a g-secretase inhibitor selected from the group consisting of AL-101, AL-102, LY3039478, RO4929097, MK-0752, and PF- 03084014; or a compound selected from the group consisting of 2-(2-fluorophenoxy)-4-(l- ethyl- 1 //-pyrazol-5-yl)benzamide, 2-[2-Methoxy-4-[(4-oxo-2-thioxo-5- thiazolidinylidene)methyl]phenoxy]-acetic acid ethyl ester, IMR-1, IMR-IA and a compound of formula (I) as shown below.

In a further embodiment the T cell modulator (TCM) is a g-secretase inhibitor selected from the group consisting of AL-101, AL-102, LY3039478, RO4929097, MK-0752, and PF- 03084014; or a compound selected from the group consisting of 2-(2-fluorophenoxy)-4-(l- ethyl- 1 //-pyrazol-5-yl)benzamide, 2-[2-Methoxy-4-[(4-oxo-2-thioxo-5- thiazolidinylidene)methyl]phenoxy]-acetic acid ethyl ester and a compound of formula (I) as shown below.

In a further embodiment the T cell modulator (TCM) is a compound selected from the group consisting of of 2-(2-fluorophenoxy)-4-( 1 -methyl- 1 //-pyrazol-5-yl)benzamide, 2-[2-Methoxy- 4-[(4-oxo-2-thioxo-5-thiazolidinylidene)methyl]phenoxy]-acet ic acid ethyl ester, IMR-1, IMR- 1 A and a compound of formula (I) as shown below.

In a further embodiment the T cell modulator (TCM) is a compound selected from the group consisting of 2-(2-fluorophenoxy)-4-( 1 -methyl- 1 //-pyrazol-5-yl)benzamide, 2-[2-Methoxy-4- [(4-oxo-2-thioxo-5-thiazolidinylidene)methyl]phenoxy]-acetic acid ethyl ester and a compound of formula (I) as shown below.

In a further embodiment the T cell modulator (TCM) is a g-secretase inhibitor selected from the group consisting of AL-101, AL-102, LY3039478, RO4929097, MK-0752, and PF- 03084014; IMR-1, IMR-1 A or a compound of formula (I) as shown below.

In a further embodiment the T cell modulator (TCM) is a g-secretase inhibitor selected from the group consisting of AL-101, AL-102, LY3039478, RO4929097, MK-0752, and PF- 03084014; or a compound of formula (I) as shown below.

In a preferred embodiment the T cell modulator (TCM) is a compound of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof, wherein X is selected from CH 2 , CF 2 , CHF, CO, CHOH, CHO(C 1 -C 3 ) alkyl, NH, N(C 1 -C 3 alkyl), S, SO and O; wherein Y1 , Y2 , and Y3 are each independently selected from N and C; wherein Z is NR10R11 , wherein R10 and R11 are each independently selected from H and C 1 -C 6 alkyl; wherein R1 is selected from H, halogen, C1-C6 alkyl, C3-C12 cycloalkyl, C3-C12 heterocyclyl, C1- C 6 alkoxy, C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C3-C12 heterocyclyl; C0-C3 alkylOC0-C3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C -C cycloalkyl, C -C heteroc 12 3 12 3 12 yclyl, C(O)R , C 1 -C 6 alkyl C(O)R12; wherein R3 is selected from H, halogen, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 12 cycloalkyl, C 3 - C 12 heterocyclyl and C 1 -C 6 alkoxy; wherein R4 , R5 and R6 are each independently selected from H, halogen, CN, C1-C6 alkoxy, C1- C 6 -S-alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 12 cycloalkyl and C 3 -C 12 heterocyclyl; wherein R7 is absent when Y1 is N or is selected from H, halogen, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C -C cycloa 1 3 12 lkyl, C 3 -C 12 heterocyclyl and C 1 -C 6 alkoxy when Y is C; wherein R8 is absent when Y3 is N or is selected from H, halogen, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C -C 3 3 12 cycloalkyl, C 3 -C 12 heterocyclyl and C 1 -C 6 alkoxy when Y is C; wherein R9 is absent when Y2 is N or is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 -C 6 alkoxy, C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH2, OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R12 is selected from H, NH 2 , NHC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 12 cycloalkyl and C 3 -C 12 heterocyclyl; and optionally one or more pharmaceutically acceptable diluents, excipients or carriers. In a more preferred embodiment the T cell modulator (TCM) is a compound of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof, wherein X is selected from CH 2 , CF 2 , CHF, CO, CHOH, CHO(C 1 -C 3 ) alkyl, NH, N(C 1 -C 3 alkyl), S, SO and O; wherein Y1 , Y2 , and Y3 are each independently selected from N and C; wherein Z is NR10R11 , wherein R10 and R11 are each independently selected from H and C 1 -C 6 alkyl; wherein R1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy, C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R2 is selected from C1-C6 alkyl, C3-C12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C -C cycloalkyl, C -C heteroc 12 3 12 3 12 yclyl, C(O)R , C 1 -C 6 alkyl C(O)R12; wherein R3 is selected from H, halogen, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 12 cycloalkyl, C 3 - C 12 heterocyclyl and C 1 -C 6 alkoxy; wherein R4 , R5 and R6 are each independently selected from H, halogen, CN, C 1 -C 6 alkoxy, C 1 - C 6 -S-alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 12 cycloalkyl and C 3 -C 12 heterocyclyl; wherein R7 is absent when Y1 is N or is selected from H, halogen, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C -C cycloalkyl, C -C heterocyclyl a 1 3 12 3 12 nd C 1 -C 6 alkoxy when Y is C; wherein R8 is absent when Y3 is N or is selected from H, halogen, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C -C cycloalkyl, C -C heterocyclyl 3 3 12 3 12 and C1-C6 alkoxy when Y is C; wherein R9 is absent when Y2 is N or is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 -C 6 alkoxy, C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R12 is selected from H, NH 2 , NHC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 12 cycloalkyl and C 3 -C 12 heterocyclyl; and optionally one or more pharmaceutically acceptable diluents, excipients or carriers; with the proviso that if R2 is C 1 -C 6 alkyl or C -C c 1 3 12 ycloalkyl, Y is N. The compounds of formula (I) and their synthesis is described in WO2013093885A1 and WO2020208139A1, respectively. The term "alkyl" as used herein refers to a saturated straight or branched chain group of carbon atoms derived from an alkane by the removal of one hydrogen atom. C 1 -C 3 alkyl comprises for example methyl, ethyl, n-propyl, i-propyl and comprises preferably non-branched C 1 -C 3 alkyl. C 1 -C 4 alkyl comprises for example methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, tert-butyl and comprises preferably non-branched C 1 -C 4 alkyl. C 1 -C 6 alkyl comprises for example methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, tert-butyl, n-pentyl, and n-hexyl and comprises preferably non-branched C 1 -C 6 alkyl. C 1 -C 10 alkyl comprises for example methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, tert-butyl, n-pentyl, n-hexyl, n-heptyl, n- octyl, n- nonyl or n-decyl and comprises preferably non-branched C1-C10 alkyl. The term “C0 alkyl “as used herein refers to a covalent bond. Thus e.g. the term “C 0 alkylOC 0 alkyl aryl” refers to Oaryl. The term “C 0 -C 3 alkylOC 0 -C 3 alkyl aryl” as used herein refers to Oaryl as defined herein when both C 0 -C 3 alkyl groups are C 0 alkyl. The term refers to OC 0 -C 3 alkyl aryl when the first C 0 -C 3 alkyl group is C 0 alkyl. The term refers to C 0 -C 3 alkylOaryl when the second C 0 -C 3 alkyl group is C 0 alkyl. Preferably C 0 -C 3 alkylOC 0 -C 3 alkyl aryl is C 0 -C 3 alkylOaryl, more preferably Oaryl or C 1 -C 3 alkylOaryl. The term “C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl” as used herein refers to Oheteroaryl as defined herein when both C 0 -C 3 alkyl groups are C 0 alkyl. The term refers to OC 0 -C 3 alkyl heteroaryl when the first C 0 -C 3 alkyl group is C 0 alkyl. The term refers to C 0 -C 3 alkylOheteroaryl when the second C 0 -C 3 alkyl group is C 0 alkyl. Preferably C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl is C 0 -C 3 alkylOheteroaryl, more preferably Oheteroaryl or C 1 -C 3 alkylOheteroaryl, most preferably Oheteroaryl. The aryl and the heteroaryl of C 0 -C 3 alkylOC 0 - C3 alkyl aryl and C0-C3 alkylOC0-C3 alkyl heteroaryl are optionally substituted by NH2, OC1-C6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably are optionally substituted by NH 2 . The term "heteroalkyl" as used herein refers to an alkyl radical as defined herein wherein one, two, three or four hydrogen atoms have been replaced with a substituent independently selected from the group consisting of OR a , C(O)0R a , NR b R c , C(O)NR b R c , S(O) n R d (where n is an integer from 0 to 2) and halogen, with the understanding that the point of attachment of the heteroalkyl radical is through a carbon atom, wherein R a is H, C1-C3 alkylcarbonyl, C1-C3 alkyl, or C3-7 cycloalkyl; R b and R c are each independently H, C1-C3 alkylcarbonyl, C1-C3 alkyl, C3-7 cycloalkyl or NR b R c is guanidinyl; and when n is 0, R d is H, C1-C3 alkyl or C3-7 cycloalkyl, and when n is 1 or 2, R d is C1-C3 alkyl or C3-7 cycloalkyl. Preferably, the term "heteroalkyl" or “heteroalkanediyl” as used herein refers to an alkyl radical or an alkanediyl radical as defined herein wherein one, two, three or four hydrogen atoms have been replaced with a substituent independently selected from the group consisting of OH, NH 2 , guanidinyl and halogen, more preferably wherein one or two hydrogen atoms have been replaced with a substituent independently selected from the group consisting of OH, NH 2 and halogen. Representative examples include, but are not limited to, 2-hydroxy ethyl, 2-hydroxypropyl, 3 -hydroxy propyl, 2- hydroxy-l-hydroxymethylethyl, 2-hydroxy- 1 -methyl ethyl, 2,3-dihydroxypropyl, 1- hydroxymethylethyl, 3-hydroxybutyl, 2,3-dihydroxybutyl, l-hydroxy-2-methylpropyl, 3- hydroxy-1 -(2-hydroxy ethyl)-propyl, 2-hydroxy- 1 -methyl propyl, 1,1,1-trifluoroethyl, 1,1,1- trifluoromethy 1, 2, 2, 3 , 3 -tetrafluoropropyl .

The term " C 3 -i 2 cycloalkyl " and “C3-7 cycloalkyl” as used herein refers to a monocyclic, bicyclic, tricyclic or tetracyclic hydrocarbon group, usually to a monovalent saturated monocyclic or bicyclic hydrocarbon group, preferably a monovalent saturated monocyclic goup of 3-12 or 3-7 carbons, respectively derived from a cycloalkane by the removal of a single hydrogen atom. “C3.7 cycloalkyl” includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. The term "C 3 - 12 cycloalkyl " and “C3-7 cycloalkyl” as used herein also includes cycloalkyl groups that comprise a C1.3 alkyl radical. Examples of such "C3-7 cycloalkyl" groups comprise cyclopropylmethyl, 2-cyclopropylethyl, cyclobutylmethyl, 2-cyclobutylethyl, cyclopentylmethyl, 2-cyclopentylethyl. Cycloalkyl groups of this invention can be optionally substituted.

The term "aryloxy" or “Oaryl” which are used interchangeably herein refers to a radical -OR where R is an aryl as defined herein, e. g. phenoxy. The term "C 1 -C 6 alkoxy" or “OC 1 -C 6 alkyl” which are used interchangeably herein refers to a radical -OR where R is a C 1 -C 6 alkyl as defined herein. Examples are methoxy, ethoxy, propoxy, butoxy. The term "aryl" as used herein refers to a mono- or bicyclic carbocyclic ring system having one or two aromatic rings, and is preferably a monocyclic carbocyclic ring system. The aryl group can also be fused to a cyclohexane, cyclohexene, cyclopentane, or cyclopentene ring or to a cyclohexane, cyclohexene, cyclopentane, or cyclopentene ring comprising a carbonyl group. The aryl groups of this invention can be optionally substituted as further described below. A preferred aryl group and optionally substituted aryl group, respectively of this invention is a phenyl group or substituted phenyl group. Substituents can be e.g. NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C -C heteroalkyl, halogen, CN, C -C c 12 1 6 3 12 ycloalkyl, C 3 -C 12 heterocyclyl, C(O)R , C 1 -C 6 alkyl C(O)R12. The term "heteroaryl" as used herein refers to substituted and unsubstituted aromatic 5-, or 6- membered monocyclic groups and 9- or 10-membered bicyclic groups, preferably a substituted and unsubstituted aromatic 5-, or 6- membered monocyclic group, which have at least one heteroatom (O, S or N) in at least one of the ring(s). Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom. The fused rings completing the bicyclic group may contain only carbon atoms and may be saturated, partially saturated, or unsaturated. Heteroaryl groups must include at least one fully aromatic ring but the other fused ring or rings may be aromatic or non-aromatic. The heteroaryl group may be attached at any available nitrogen or carbon atom of any ring. Heteroaryl groups of this invention can be optionally substituted as further described below. Usually, a heteroaryl group and optionally substituted heteroaryl group, respectively of this invention is selected from the group consisting of substituted and/or unsubstituted aromatic 5-, or 6- membered monocyclic groups, which have at least one heteroatom (O, S or N), preferably one or two heteroatoms selected from S and N in the ring, more preferably one S and one N in the ring, or one or two N in the ring. A preferred heteroaryl group is an optionally substituted heteroaryl group, selected from the group consisting of an optionally substituted pyridinyl group, an optionally substituted pyrimidinyl group, an optionally substituted di- or triazine group, an optionally substituted thiazole group, an optionally substituted oxazole group, and an optionally substituted imidazole group. An even more preferred heteroaryl group is an optionally substituted pyridinyl group, an optionally substituted pyrimidinyl group, an optionally substituted imidazole group or an optionally substituted thiazole group. Most preferably an optionally substituted pyridinyl group, an optionally substituted imidazole group or an optionally substituted thiazole group, is used as heteroaryl group in the present invention. Optional substituents can be e.g. NH 2 , OC 1 -C 6 alkyl, C -C alkyl, C -C heteroalkyl, halogen, CN, C -C cycloalkyl, C 12 1 6 1 6 3 12 3 -C 12 heterocyclyl, C(O)R , C1-C6 alkyl C(O)R12 , or NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3- C 12 cycloalkyl, C 3 -C 12 heterocyclyl. The term "heterocyclyl" as used herein means a saturated, monocyclic ring with 3 to 12, preferably with 3 to 7, more preferably 5 to 6 ring atoms which contains up to 3, preferably 1 or 2 heteroatoms selected independently from nitrogen, oxygen or sulfur, and wherein the remaining ring atoms being carbon atoms. Examples of such saturated heterocycles include [1,3]dioxanyl, [1,3]dioxolanyl, pyrrolidinyl, morpholinyl, piperazinyl, piperidinyl, oxazolidinyl, thiazolidinyl, azepanyl and the like. Preferably such heterocyclyl groups are unsubstituted. The terms "halo" or "halogen" as used herein refers to F, Cl, Br, or I and is preferably F, Cl, or Br, more preferably F. The term ”optionally substituted” or “substituted” means that the referenced group is substituted with one or more additional group(s), preferably with one additional group, individually and independently selected from the listed groups. AL101 (formerly BMS 906024), which has the chemical name (2R,3S)-N1-((S)-1-methyl-2- oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepin-3-yl)-2,3- bis(3,3,3- trifluoropropyl)succinamide is described e.g. in WO2012129353A1 or NCT03691207 and is represented by the structural formula indicated below:

AL102 (BMS 986115), which has the chemical name (2S,3R)-N'-[(3S)-5-(3-fluorophenyl)-9- methyl-2-oxo-1,3-dihydro-1,4-benzodiazepin-3-yl]-2,3-bis(3,3 ,3-trifluoropropyl)butanediamide is described e.g. in WO2014047372A1and is represented by the structural formula indicated below: LY3039478 which has the chemical name 4,4,4-trifluoro-N-[(2S)-1-[[(7S)-5-(2-hydroxyethyl)- 6-oxo-7H-pyrido[2,3-d][3]benzazepin-7-yl]amino]-1-oxopropan- 2-yl]butanamide is described e.g. in Massard C et al.. Ann Oncol.2018;29(9):1911-1917 or in WO2020131998A1 and is represented by the structural formula indicated below: RO4929097 which has the chemical name 2,2-dimethyl-N-[(7S)-6-oxo-5,7- dihydrobenzo[d][1]benzazepin-7-yl]-N'-(2,2,3,3,3-pentafluoro propyl)propanediamide is described e.g. in Huynh C et al. PLoS One .2011;6(9) or in WO2020131998A1and is represented by the structural formula indicated below: MK-0752 which has the chemical name 3-[4-(4-chlorophenyl)sulfonyl-4-(2,5- difluorophenyl)cyclohexyl]propanoic acidis described e.g. in US2004116404A1and is represented by the structural formula indicated below: PF-03084014 which has the chemical name (2S)-2-[[(2S)-6,8-difluoro-1,2,3,4- tetrahydronaphthalen-2-yl]amino]-N-[1-[1-(2,2-dimethylpropyl amino)-2-methylpropan-2- yl]imidazol-4-yl]pentanamideis described e.g. in US7342118, US7795447 and US7951958 and is represented by the structural formula indicated below: 2-(2-fluorophenoxy)-4-(1-methyl-1H-pyrazol-5-yl)benzamide also referred to as RBPJ INhibitor1 (RIN1) is described e.g. in Hurtado C et al., 2019 Sci Rep 9, 10811 and is represented by the structural formula indicated below: 2-[2-Methoxy-4-[(4-oxo-2-thioxo-5-thiazolidinylidene)methyl] phenoxy]-acetic acid ethyl ester ) is obtainable from e.g. Sigma Aldrich and is represented by the structural formula indicated below: IMR-1 is described e.g. in Astudillo L. et al., 2016, Cancer Res Jun 15;76(12):3593-603 and is represented by the structural formula indicated below: IMR-1A is described e.g. in Astudillo L. et al., 2016, Cancer Res Jun 15;76(12):3593-603 and is represented by the structural formula indicated below:

“6-(4-Tert-Butylphenoxy)Pyridin-3-Amine” is represented by the structural formula indicated below: 6-(4-Tert-Butylphenoxy)Pyridin-3-Amine is a synthetic small molecule (Molecular Mass: 242.32 g/mol) and is described e.g. in WO2013093885A1. The present invention also encompasses chemical modifications of the compounds of the present invention to prolong their circulating lifetimes. Non-limiting examples of methods for transiently, or reversibly, pegylating drugs, including polypeptide-based drugs, are provided in U.S. Pat. Nos.4,935,465 (issued in Jun.19, 1990) and 6,342,244 (issued Jan.29, 2002); and in U.S. published applications number US2006/0074024. One skilled in the art would typically find more details about PEG-based reagents in, for example, published applications WO2005047366, US2005171328, and those listed on the NEKTAR PEG Reagent Catalog® 2005-2006 (Nektar Therapeutics, San Carlos, Calif.). The invention also relates to salts, hydrates or solvates of the compounds of the present invention. Preferably, these salts, hydrates and/or solvates are pharmaceutically acceptable. The invention also relates to stereoisomers of the compounds of formula (I). “Stereoisomer” or “stereoisomers” refer to compounds that differ in the chirality of one or more stereocentres. Stereoisomers include enantiomers and diastereomers. A compound of formula (I) may exist in stereoisomeric form if they possess one or more asymmetric centres or a double bond with asymmetric substitution and, therefore, can be produced as individual stereoisomers or as mixtures. Unless otherwise indicated, the description is intended to include individual stereoisomers as well as mixtures. The methods for the determination of stereochemistry and the separation of stereoisomers are well-known in the art (see discussion in Chapter 4 of Advanced Organic Chemistry, 4th ed., J. March, John Wiley and Sons, New York, 1992). A skilled person will know that, if the compounds of the present invention contain charged group, a suitable counterion will be derived from an organic or inorganic acid. Such counterions include halide (such as chloride, bromide, fluoride, iodide), sulfate, phosphate, acetate, succinate, citrate, lactate, maleate, fumarate, palmitate, cholate, glutamate, glutarate, tartrate, stearate, salicylate, methanesulfonate, benzenesulfonate, sorbate, picrate, benzoate, cinnamate, and the like. If the polar moiety is a negatively charged group, a suitable counterion will be selected from sodium, ammonium, barium, calcium, copper, iron, lithium, potassium and zinc, and the like. When R1 is C0-C3 alkylOC0-C3 alkyl aryl or C0-C3 alkylOC0-C3 alkyl heteroaryl, the optional substitutions of the aryl and the heteroaryl group are preferably in para position. When R2 is aryl or heteroaryl, the optional substitutions are preferably in ortho or meta position, provided that the substitutents are not halogen, OC1-C6 alkyl or methyl and are in para position when the substituents are halogen, OC 1 -C 6 alkyl or methyl. When R9 is C 0 -C 3 alkylOC 0 -C 3 alkyl aryl or C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl the optional substitutions of the aryl and the heteroaryl group are preferably in para position. In one embodiment X is selected from CH 2 , CF 2 , CHF, NH, N(C 1 -C 3 alkyl), S, SO and O. In a further embodiment X is selected from CO, CHOH, CHO(C 1 -C 3 ) alkyl, S, SO and O. In a preferred embodiment X is selected from CH 2 , NH, and O. In a more preferred embodiment X is selected from CH 2 , CO, CHOH, CHO(C 1 -C 3 ) alkyl, NH, and O. In an even more preferred embodiment X is selected from CH 2 , CO, CHOH, CHO(C 1 -C 3 ) alkyl, and O. In a particular preferred embodiment X is selected from CH 2 , CO, CHOH, CHOCH 3 , and O. In a more particular preferred embodiment X is selected from CH 2 and O. In an even more particular preferred embodiment X is O. In one embodiment R1 is selected from H, halogen, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, preferably is optionally substituted by NH 2 ; and C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl, preferably is optionally substituted by NH . In a fu 1 2 rther embodiment R is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 -C 6 alkoxy and C 1 -C 6 heteroalkyl. In a preferred embodiment R1 is selected from H, halogen and C 1 -C 4 alkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, preferably is optionally substituted by NH 2 ; and C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 . In a further preferred embodiment R1 is selected from H, halogen, C 1 -C 6 alkyl and C -C 6 heteroalkyl. In a more pre 1 1 ferred embodiment R is selected from H, C 1 -C 6 alkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , C 1 -C 6 alkyl, C1-C6 heteroalkyl, halogen, CN, preferably is optionally substituted NH2; C0-C3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, preferably is optionally substituted by NH 2. In an even more preferred embodiment R1 is selected from H, C1-C4 alkyl and C0-C3 alkylOC0-C3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, preferably is optionally substituted by NH 2 ; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH . In a further more p 1 2 referred embodiment R is selected from H, halogen and C 1 -C 4 alkyl. In an even more preferred embodiment R1 is selected from H, methyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, preferably is optionally substituted by NH 2 ; and C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 . In a further even more preferred embodiment R1 is selected from C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 ; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 ; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH2, OC1-C6 alkyl, C1-C6 alkyl, C1-C6 heteroalkyl, halogen, CN, C3-C12 cycloalkyl, C3-C12 heterocyclyl, preferably is optionally substituted by NH 2 . In a further even more preferred embodiment R1 is selected from C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 ; and C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 1 2. In a further even more preferred embodiment R is selected from H and methyl. In one embodiment R2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl is substituted by a substituent selected from NH2, OC1-C6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C(O)R12 and C 1 -C 6 alkyl C(O)R12. In a further embodiment R2 is selected from C 1 -C 6 alkyl, C 3 - C12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH , OC -C alkyl, C -C alkyl, C -C heteroalkyl, hal 12 2 1 6 1 6 1 6 ogen, CN, C(O)R , C 1 -C 6 alkyl C(O)R12. In a preferred embodiment R2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , C 1 -C 6 alkyl, C -C heteroalkyl, halogen 12 1 6 , CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C(O)R and C 1 -C 6 alkyl C(O)R12. In a more preferred embodiment R2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH , 12 12 2 C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C(O)R , C 1 -C 6 alkyl C(O)R . In a further more preferred embodiment R2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl. In a particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by C 1 -C 6 alkyl, halogen, C 1 -C 6 heteroalkyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 . In a further particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by C 1 -C 6 alkyl, halogen. In a further particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , halogen, CN. In a further particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 . Among the particular preferred embodiments, the embodiment wherein R 2 is selected from aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by C 1 -C 6 alkyl, halogen, C 1 -C 6 heteroalkyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 is preferred. In an even more particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, C 1 -C 6 alkyl C(O)R 12 .

In an even more particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, phenyl, pyridyl, imidazole and thiazole, wherein the phenyl, pyridyl, imidazole and thiazole each are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 . In a further even more particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, phenyl, thiazole, pyridyl and imidazole wherein the phenyl, thiazole, pyridyl and imidazole each are optionally substituted by NH 2 , C I -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 . In a further even more particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, phenyl, pyridyl, imidazole and thiazole wherein the phenyl, pyridyl, imidazole and thiazole each are optionally substituted by NH 2 , C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 . In a further even more particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, phenyl, pyridyl, imidazole and thiazole wherein the phenyl, pyridyl, imidazole and thiazole each are optionally substituted by C 1 -C 6 alkyl, halogen, C 1 -C 6 heteroalkyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 . In a further even more particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, phenyl, pyridyl, imidazole and thiazole wherein the phenyl, pyridyl, imidazole and thiazole each are optionally substituted by C 1 -C 6 alkyl, halogen. In a further even more particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, phenyl, pyridyl, imidazole and thiazole wherein the phenyl, pyridyl, imidazole and thiazole each are optionally substituted by NH 2 , halogen, CN. In a further even more particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, phenyl, pyridyl, imidazole and thiazole wherein the phenyl, pyridyl, imidazole and thiazole each are optionally substituted by C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 . Among the even more particular preferred embodiments the embodiment wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, phenyl, pyridyl, imidazole and thiazole wherein the phenyl, pyridyl, imidazole and thiazole each are optionally substituted by C 1 -C 6 alkyl, halogen, C 1 -C 6 heteroalkyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 is preferred. In a further even more particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, phenyl, pyridyl, imidazole and thiazole wherein the phenyl, pyridyl, imidazole and thiazole each are optionally substituted by C 1 -C 6 alkyl, halogen. In a further even more particular preferred embodiment R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, phenyl, pyridyl, imidazole and thiazole wherein the phenyl, pyridyl, imidazole and thiazole each optionally substituted by C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 . In a further even more particular preferred R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, phenyl, pyridyl, imidazole and thiazole, wherein the phenyl, pyridyl, imidazole and thiazole each are optionally substituted by OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, C 1 -C 6 alkyl C(O)R 12 . In a most preferred embodiment R 2 is C 1 -C 6 alkyl, more preferably tert-butyl.

In a preferred embodiment when R 2 is heteroaryl the heteroaryl is an optionally substituted aromatic 5-, or 6- membered monocyclic group.

In one embodiment R 3 is selected from H, halogen, C 1 -C 6 alkyl, and C 3 -C 12 cycloalkyl. In a preferred embodiment R 3 is selected from H, halogen, C 1 -C 4 alkyl, and C 3 -C 7 cycloalkyl. In a more preferred embodiment R 3 is selected from H, halogen and C 1 -C 4 alkyl. In an even more preferred embodiment R 3 is H.

In one embodiment R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl. In a preferred embodiment R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 4 alkyl and C 1 -C 4 heteroalkyl. In an even more preferred embodiment R 4 , R 5 and R 6 are each independently selected from H, halogen, and C 1 -C 4 alkyl.

In a particular preferred embodiment R 4 , R 5 and R 6 are each independently selected from H, halogen, and methyl. In a more particular preferred embodiment R 4 is selected from H and halogen and/or R 5 and/or R 6 are selected from H and C 1 -C 6 alkyl, in particular from H and C 1 - C 4 alkyl, more particular from H and methyl.

In one embodiment R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C. In a preferred embodiment R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 4 alkyl and C 3 -C 7 cycloalkyl when Y 1 is C. In a more preferred embodiment R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl, preferably selected from H, halogen, and methyl when Y 1 is C. In an even more preferred embodiment R 7 is absent when Y 1 is N or is selected from H, halogen, and methyl when Y 1 is C. In a particular preferred embodiment R 7 is absent when Y 1 is N or is selected from H and halogen when Y 1 is C. In a more particular preferred embodiment R 7 is absent.

In one embodiment R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C. In a preferred embodiment R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 4 alkyl and C 3 -C 7 cycloalkyl when Y 3 is C. In a more preferred embodiment R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl preferably selected from H, halogen, and methyl when Y 3 is C. In an even more preferred embodiment R 8 is absent when Y 3 is N or is selected from H, halogen, and methyl when Y 3 is C. In a particular preferred embodiment R 8 is absent when Y 3 is N or is selected from H and halogen when Y 3 is C. In a particular preferred embodiment R 8 is H.

In one embodiment R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C. In a preferred embodiment R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 4 alkyl and C 3 -C 7 cycloalkyl when Y 2 is C. In a more preferred embodiment R 9 is absent when Y 2 is N or is selected from H and C 1 -C 4 alkyl preferably selected from H, halogen, and methyl when Y 2 is C. In an even more preferred embodiment R 9 is absent when Y 2 is N or is selected from H, halogen, and methyl when Y 2 is C. In a particular preferred embodiment R 9 is absent when Y 2 is N or is selected from H and halogen, preferably H, when Y 2 is C.

In one embodiment R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C, preferably selected from H, halogen, and methyl when Y 1 is C, R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl, preferably selected from H, halogen, and methyl when Y 3 is C, and R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl, preferably selected from H, halogen, and methyl when Y 2 is C.

In a preferred embodiment R 7 is absent when Y 1 is N or is selected from H and halogen when

Y 1 is C, R 8 is absent when Y 3 is N or is H when Y 3 is C, and R 9 is absent when Y 2 is N or is selected from H and methyl when Y 2 is C.

In one embodiment at least one of R 10 and R 11 is C 1 -C 6 alkyl, preferably C 1 -C 4 alkyl, more preferably methyl. In a preferred embodiment R 10 and R 11 are independently selected from H and methyl. In a more preferred embodiment R 10 is H and R 11 is selected from H and C 1 -C 4 alkyl. In an even more preferred embodiment R 10 is H and R 11 is H or methyl. In a particular preferred embodiment R 10 is H and R 11 is C 1 -C 6 alkyl. In a more particular preferred embodiment R 10 is H and R 11 is C 1 -C 4 alkyl. In an even more particular preferred embodiment R 10 is H and R 11 is methyl.

In one embodiment R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl. In a preferred embodiment R 12 is selected from NH 2 , C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl. In a more preferred embodiment R 12 is NH 2 .

In one embodiment Y 1 is N. In a preferred embodiment Y 1 and Y 2 are each independently selected from N and C and Y 3 is C. In a more preferred embodiment Y 1 is selected from N and C and Y 2 and Y 3 are selected from N and C with the proviso that one of Y 2 and Y 3 is C. In a particular preferred embodiment Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C. In a more particular preferred embodiment Y 1 is N and R 7 is absent.

In one embodiment at least one of the substituents selected from R 1 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , and R 9 is not H. In a further embodiment at least one of the substituents selected from R 1 , R 3 , R 8 , and R 9 is not H. Preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O, preferably selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy; C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC 1 -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N. Further preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O, preferably selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy; C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl C 3 -C 12 heterocyclyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC 1 -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; with the proviso that when R 1 is selected from C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C 1 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 1 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, R 2 is selected from aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , halogen, CN, preferably wherein the aryl and the heteroaryl are not substituted; preferably with the further proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

More preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O, preferably selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy and C 1 -C 6 heteroalkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further more preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O, preferably selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy and C 1 -C 6 heteroalkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C; and wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC 1 -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl, with the proviso that when R 2 is selected from aryl and heteroaryl wherein the aryl and the heteroaryl are substituted by C 1 -C 6 alkyl, halogen, C 1 -C 6 heteroalkyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 , R 1 is selected from H, halogen, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl, preferably is H or methyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C3-C12 cycloalkyl, Y 1 is N.

Further more preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C 1 -Ck) alkyl, NH, and O, preferably selected from CH 2 , CO, CHOH, CHO(C 1 -Ck) alkyl, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy and C 1 -C 6 heteroalkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C; and wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC 1 -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N. Further more preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those , wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O, preferably selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy; C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 -C 6 alkoxy; C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC I -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , halogen, CN; wherein R 3 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; and wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N. Even more preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O, preferably selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 4 alkyl and C 1 -C 4 heteroalkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 4 alkyl and C 3 -C 7 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 4 alkyl and

C 1 -C 4 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 4 alkyl and C 3 -C 7 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 4 alkyl and C 3 -C 7 cycloalkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 4 alkyl and C 3 -C 7 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC 1 -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O, preferably selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl, preferably independently selected from H and methyl, more preferably R 10 is H and R 11 is H or methyl; wherein R 1 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen and C1-C4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C | -C,) alkyl, NH, and O, preferably selected from CH 2 , CO, CHOH, CH0(C | -C,) alkyl, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl, preferably independently selected from H and methyl, preferably R 10 is H and R 11 is H or methyl; wherein R 1 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by C 1 -C 6 heteroalkyl; wherein R 3 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O, preferably selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl, preferably independently selected from H and methyl, preferably R 10 is H and R 11 is H or methyl; wherein R 1 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by C 1 -C 6 alkyl, halogen; wherein R 3 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N. Further particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O, preferably selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl, preferably independently selected from H and methyl, preferably R 10 is H and R 11 is H or methyl; wherein R 1 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O, preferably selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl, preferably independently selected from H and methyl, preferably R 10 is H and R 11 is H or methyl; wherein R 1 is selected from C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 ; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 ; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 , wherein R 1 is preferably selected from C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 ; and C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC I -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 ; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , halogen, C 1 -C 6 alkyl, CN, preferably optionally substituted by halogen, C 1 -C 6 alkyl; wherein R 3 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; and wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C.

Preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , NH and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy; C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , NH and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy; C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl C 3 -C 12 heterocyclyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; with the proviso that when R 1 is selected from C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C 12 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, R 2 is selected from aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , halogen, CN, preferably wherein the aryl and the heteroaryl are not substituted; preferably with the further proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

More preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , NH and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy and C 1 -C 6 heteroalkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further more preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , NH and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy and C 1 -C 6 heteroalkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C; and wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl, with the proviso that when R 2 is selected from aryl and heteroaryl wherein the aryl and the heteroaryl are substituted by C 1 -C 6 alkyl, halogen, C 1 -C 6 heteroalkyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 , R 1 is selected from H, halogen, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl, preferably is H or methyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further more preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , NH and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy and C 1 -C 6 heteroalkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C; and wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further more preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those , wherein X is selected from CH 2 , NH and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy; C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 -C 6 alkoxy; C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC I -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , halogen, CN; wherein R 3 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; and wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Even more preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , NH and O, and is preferably O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 4 alkyl and C 1 -C 4 heteroalkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 4 alkyl and C 3 -C 7 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 4 alkyl and C 1 -C 4 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 4 alkyl and C 3 -C 7 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 4 alkyl and C 3 -C 7 cycloalkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C1-C4 alkyl and C3-C7 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , NH and O, and is preferably O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl, preferably independently selected from H and methyl, more preferably R 10 is H and R 11 is H or methyl; wherein R 1 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , NH and O, and is preferably O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl, preferably independently selected from H and methyl, preferably R 10 is H and R 11 is H or methyl; wherein R 1 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by C 1 -C 6 heteroalkyl; wherein R 3 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , NH and O, and is preferably O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl, preferably independently selected from H and methyl, preferably R 10 is H and R 11 is H or methyl; wherein R 1 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by C 1 -C 6 alkyl, halogen; wherein R 3 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , NH and O, and is preferably O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl, preferably independently selected from H and methyl, preferably R 10 is H and R 11 is H or methyl; wherein R 1 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N. Further particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , NH and O, and is preferably O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C, preferably Y 1 and Y 2 are each independently selected from N and C and Y 3 is C, more preferably Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl, preferably independently selected from H and methyl, preferably R 10 is H and R 11 is H or methyl; wherein R 1 is selected from C 0 -C 3 alkylOC (l -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 ; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 ; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 , wherein R 1 is preferably selected from C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 ; and C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC I -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, preferably is optionally substituted by NH 2 ; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , halogen, C 1 -C 6 alkyl, CN, preferably optionally substituted by halogen, C 1 -C 6 alkyl; wherein R 3 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; and wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy and C 1 -C 6 heteroalkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

More particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy, C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 3 is C; wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further more particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O; wherein Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, halogen, C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C 1 - C 6 alkoxy, C 1 -C 6 heteroalkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 - C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 1 -C 6 alkyl substituted by aryl or heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl, C(O)R 12 , C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, CN, C 1 -C 6 alkyl and C 1 -C 6 heteroalkyl; wherein R 8 is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl; wherein R 9 is absent when Y 2 is N or is selected from H, halogen, C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl when Y 2 is C; and wherein R 12 is selected from NH 2 , NHC I -C 6 alkyl, C 1 -C 6 alkyl, and C 1 -C 6 heteroalkyl; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further more particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, C 1 -C 6 alkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 ; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 ; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is H; wherein R 4 is selected from H and halogen; wherein R 5 and R 6 are each independently selected from H and C 1 -C 6 alkyl; wherein R 7 is absent when Y 1 is N or is H when Y 1 is C; wherein R 8 is absent when Y 3 is N or is H when Y 3 is C; wherein R 9 is absent when Y 2 is N or is H when Y 2 is C; and wherein R 12 is NH 2 ; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further more particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O; wherein Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H, C 1 -C 6 alkyl, C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 ; C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 ; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is H; wherein R 4 is selected from H and halogen; wherein R 5 and R 6 are each independently selected from H and C 1 -C 6 alkyl; wherein R 8 is H; wherein R 9 is absent when Y 2 is N or is H when Y 2 is C; and wherein R 12 is NH 2 ; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N. Further more particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H and C 1 -C 6 alkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C3-C12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is H; wherein R 4 is selected from H and halogen; wherein R 5 and R 6 are each independently selected from H and C 1 -C 6 alkyl; wherein R 7 is absent when Y 1 is N or is H when Y 1 is C; wherein R 8 is absent when Y 3 is N or is H when Y 3 is C; wherein R 9 is absent when Y 2 is N or is H when Y 2 is C; and wherein R 12 is NH 2 ; preferably with the proviso that if R 2 is C 1 -C 6 alkyl or C 3 -C 12 cycloalkyl, Y 1 is N.

Further more particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O; wherein Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H and C 1 -C 6 alkyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, C 1 -C 6 alkyl C(O)R 12 ; wherein R 3 is H; wherein R 4 is selected from H and halogen; wherein R 5 and R 6 are each independently selected from H and C 1 -C 6 alkyl; wherein R 8 is H; wherein R 9 is absent when Y 2 is N or is H when Y 2 is C; and wherein R 12 is NH 2 .

Further more particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O; wherein Y 1 , Y 2 , and Y 3 are each independently selected from N and C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from C 0 -C 3 alkylOC 0 -C 3 alkyl aryl wherein the aryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; and C 0 -C 3 alkylOC 0 -C 3 alkyl heteroaryl wherein the heteroaryl is optionally substituted by NH 2 , OC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, halogen, CN, C 3 -C 12 cycloalkyl, C 3 -C 12 heterocyclyl; wherein R 2 is selected from C 1 -C 6 alkyl, C 3 -C 12 cycloalkyl, aryl and heteroaryl wherein the aryl and the heteroaryl are optionally substituted by NH 2 , halogen, C 1 -C 6 alkyl, CN; wherein R 3 is selected from H, halogen and C 1 -C 4 alkyl; wherein R 4 , R 5 and R 6 are each independently selected from H, halogen, and C 1 -C 4 alkyl; wherein R 7 is absent when Y 1 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 1 is C; wherein R 8 is absent when Y 3 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 3 is C; and wherein R 9 is absent when Y 2 is N or is selected from H, halogen and C 1 -C 4 alkyl when Y 2 is C.

Further even more particular preferred compounds of formula (I) pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof are those, wherein X is selected from CH 2 , CO, CHOH, CHO(C I -C 3 ) alkyl, NH, and O, and is preferably

O; wherein Y 1 is N and R 7 is absent, Y 2 is selected from N and C and Y 3 is C; wherein Z is NR 10 R 11 , wherein R 10 and R 11 are each independently selected from H and C 1 -C 6 alkyl; wherein R 1 is selected from H and C 1 -C 6 alkyl; wherein R 2 is selected from C 1 -C 6 alkyl and C 3 -C 12 cycloalkyl, and is preferably C 1 -C 6 alkyl; wherein R is H; wherein R 4 is selected from H and halogen; wherein R 5 and R 6 are each independently selected from H and C 1 -C 6 alkyl; wherein R is H; wherein R 9 is absent when Y 2 is N or is H when Y 2 is C; and wherein R 12 is N¾.

Even more particular preferred compounds of the compound of formula (I) are selected from the group consisting of

Even more particular preferred compounds of the compound of formula (I) are selected from the group consisting of

Even more particular preferred compounds of the compound of formula (I) are selected from the group consisting of

Even more particular preferred compounds of the compound of formula (I) are selected from the group consisting of Most particular preferred compounds of the compound of formula (I) are selected from the group consisting of The most particular preferred T cell modulator (TCM) is selected from the group consisting of 6-((4'-fluoro-[ 1 , 1 '-biphenyl] -4-yl)oxy)-2-methylpyridin-3 -amine

6 -(4-(tert-butylphenoxy)pyri din-3 -amine or pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof.

The most preferred T cell modulator (TCM) is selected from the group consisting of 6-((4'- fluoro-[ 1 , 1 ’-biphenyl] -4-yl)oxy)-2-methylpyridin-3 -amine ? and 6 -(4-(tert-butylphenoxy)pyri din-3 -amine or pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof, in particular 6-(4-(tert-butylphenoxy)pyri din-3 -amine or pharmaceutically-acceptable salts, hydrates, solvates, or stereoisomers thereof.

Pharmaceutical compositions

As outlined above, the invention also relates to a pharmaceutical composition comprising a T cell modulator (TCM) and optionally a pharmaceutically acceptable diluent, excipient, or carrier for use in a method for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease (AIID) in a subject.

The term “pharmaceutical composition” refers to a fixed-dose combination (FDC) that includes the T cell modulator in a single dosage form, having a predetermined combination of respective dosages.

The pharmaceutical composition further may be used as add-on therapy. As used herein, "addon" or "add-on therapy" means an assemblage of reagents for use in therapy, the subject receiving the therapy begins a first treatment regimen of one or more reagents prior to beginning a second treatment regimen of one or more different reagents in addition to the first treatment regimen, so that not all of the reagents used in the therapy are started at the same time.

The amount of the T cell modulator to be administered will vary depending upon factors such as the particular compound, disease condition and its severity, according to the particular circumstances surrounding the case, including, e.g., the specific T cell modulator being administered, the route of administration, the condition being treated, the target area being treated, and the subject or host being treated.

In one embodiment, the invention provides a pharmaceutical composition comprising a T cell modulator, wherein said T cell modulator is present in a therapeutically effective amount.

The expression “effective amount” or “therapeutically effective amount” as used herein refers to an amount capable of invoking one or more of the following effects in a subject receiving the composition of the present invention: (i) expands regulatory T cells (Treg cells) (ii) activates Treg cells (iii) inhibits proliferation of conventional T cells (iv) inhibits activation of conventional T cells (vi) inhibits activity or proliferation of cells of innate immune cells (e.g dendritic cells, macrophages etc), (vii) inhibits production or secretion of pro-inflammatory cytokines and chemokines (viii) upregulates production or enhance secretion of antiinflammatory cytokines and chemokines.

Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.

In one embodiment, the invention provides a pharmaceutical composition comprising a T cell modulator, wherein the amount of said T cell modulator in the composition is from about 1 to about 1000 mg.

Formulations and modes of administration

A pharmaceutical composition according to the invention is, preferably, suitable for enteral administration, such as oral administration to a subject and comprises a therapeutically effective amount of the active ingredient and one or more suitable pharmaceutically acceptable diluent, excipient, or carrier.

If not indicated otherwise, a pharmaceutical composition according to the invention is prepared in a manner known per se, e.g. by means of conventional mixing, granulating, coating, dissolving or lyophilizing processes. In preparing a composition for an oral dosage form, any of the usual pharmaceutical media may be employed, for example water, glycols, oils, alcohols, carriers, such as starches, sugars, or microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid pharmaceutical carriers are obviously employed.

In one embodiment, the pharmaceutical composition according to the invention is a composition for enteral administration. Preferred are compositions for oral administration. As indicated above, said pharmaceutical composition is preferably a pharmaceutical composition, i.e. fixed-dose composition.

A pharmaceutical composition for enteral administration is, for example, a unit dosage form, such as a tablet, a capsule or a suppository.

In one embodiment, the invention provides a pharmaceutical composition comprising a T cell modulator and at least one pharmaceutically acceptable diluent, excipient, or carrier, wherein the composition is a tablet or a capsule, preferably a tablet.

In a preferred embodiment, the invention provides a pharmaceutical composition comprising a T cell modulator and at least one pharmaceutically acceptable diluent, excipient, or carrier, wherein the composition is a sustained release tablet.

In a further preferred embodiment, the pharmaceutical composition according to the invention is for oral administration, wherein the composition is adapted to provide sustained release of the active pharmaceutical ingredients (API). Thus, the composition may increase T max or reduce C max , or both increase T max and reduce C max , as compared to an immediate release composition.

"C max " means the the peak concentration of the drug in the plasma. "T max " means the time from administration to reach C max ·

Additional or alternative, e.g. alternative materials which may be included in the composition to provide sustained release are hydrophobic polymers, for example ethyl cellulose or a methacrylic acid polymer, or a combination thereof. Such polymers, whether used singly or in combination, may be comprised in a coating or may be included in admixture with the API (i.e. may be used as a matrix-former), or may be present both in a coating and in admixture with the API.

The unit content of active ingredients in an individual dose need not in itself constitute a therapeutically effective amount, since such an amount can be reached by the administration of a plurality of dosage units. A composition according to the invention may contain, e.g., from about 10% to about 100% of the therapeutically effective amount of the active ingredients.

Dosing regimen

An exemplary treatment regime entails administration once daily, twice daily, three times daily, every second day, twice per week, once per week. The composition of the invention is usually administered on multiple occasions. Intervals between single dosages can be, for example, less than a day, daily, every second day, twice per week, or weekly. The composition of the invention may be given as a continous uninterrupted treatment. The composition of the invention may also be given in a regime in which the subject receives cycles of treatment interrupted by a drug holiday or period of non-treatment. Thus, the composition of the invention may be administered according to the selected intervals above for a continuous period of one week or a part thereof, for two weeks, for three weeks, for four weeks, for five weeks or for six weeks and then stopped for a period of one week, or a part thereof, for two weeks, for three weeks, for four weeks, for five weeks, or for six weeks. The composition of the treatment interval and the non-treatment interval is called a cycle. The cycle may be repeated one or more times. Two or more different cycles may be used in combination for repeating the treatment one or more times. Intervals can also be irregular as indicated by measuring blood levels of said T cell modulator in the patient. In a preferred embodiment, the pharmaceutical composition according to the invention is administered once daily. In an exemplary treatment the T cell modulator can be administered from 1 - 1000 mg per day.

Using the T cell modulator or a pharmaceutical composition thereof to treat AIID

According to an aspect the present invention provides a T cell modulator or a pharmaceutical composition thereof as described herein, for use in a method for the prevention, delay of progression or treatment of AIID in a subject.

Also provided is the use of a T cell modulator or a pharmaceutical composition thereof as described herein for the manufacture of a medicament for the prevention, delay of progression or treatment of AIID in a subject. Also provided is the use of a T cell modulator or a pharmaceutical composition thereof as described herein for the prevention, delay of progression or treatment of AIID in a subject.

Also provided is a method for the prevention, delay of progression or treatment of AIID in a subject, comprising administering to said subject a therapeutically effective amount of a T cell modulator or a pharmaceutical composition therof as described herein.

The terms “treatment’ ’/’’treating” as used herein includes: (1) delaying the appearance of clinical symptoms of the state, disorder or condition developing in an animal, particularly a mammal and especially a human, that may be afflicted with or predisposed to the state, disorder or condition but does not yet experience or display clinical or subclinical symptoms of the state, disorder or condition; (2) inhibiting the state, disorder or condition (e.g. arresting, reducing or delaying the development of the disease, or a relapse thereof in case of maintenance treatment, of at least one clinical or subclinical symptom thereof); and/or (3) relieving the condition (i.e. causing regression of the state, disorder or condition or at least one of its clinical or subclinical symptoms). The benefit to a patient to be treated is either statistically significant or at least perceptible to the patient or to the physician. However, it will be appreciated that when a medicament is administered to a patient to treat a disease, the outcome may not always be effective treatment.

As used herein, "delay of progression" means increasing the time from symptoms to worsening of symptoms of AIID and includes reversing or inhibition of disease progression. "Inhibition" of disease progression or disease complication in a subject means preventing or reducing the disease progression and/or disease complication in the subject.

Preventive treatments comprise prophylactic treatments. In preventive applications, the pharmaceutical composition of the invention is administered to a subject suspected of having, or at risk for developing AIID. In therapeutic applications, the pharmaceutical composition is administered to a subject such as a patient already suffering from AIID, in an amount sufficient to cure or at least partially arrest the symptoms of the disease. Amounts effective for this use will depend on the severity and course of the disease, previous therapy, the subject's health status and response to the drugs, and the judgment of the treating physician. In the case wherein the subject's condition does not improve, the pharmaceutical composition of the invention may be administered chronically, which is, for an extended period of time, including throughout the duration of the subject's life in order to ameliorate or otherwise control or limit the symptoms of the subject's disease or condition.

In the case wherein the subject's status does improve, the pharmaceutical composition may be administered continuously; alternatively, the dose of drugs being administered may be temporarily reduced or temporarily suspended for a certain length of time (i.e., a “drug holiday”).

Once improvement of the patient's condition has occurred, a maintenance dose of the pharmaceutical composition of the invention is administered if necessary. Subsequently, the dosage or the frequency of administration, or both, is optionally reduced, as a function of the symptoms, to a level at which the improved disease is retained.

In one embodiment the autoimmune and inflammatory disease is driven by cells selected from the group consisting of Thl cells, Th2 cells, Thl7 cells and cells of innate immune system, preferably the autoimmune and inflammatory disease is driven by cells selected from the group consisting of Thl cells, Th2 cells and Thl7 cells.

In one embodiment the autoimmune and inflammatory disease is driven by Thl cells and is selected from the group consisting of acute GvHD, chronic GvHD, Type 1 diabetes, Inflammatory bowel disease, Crohn’s disease, Ulceritis, Rheumatoid arthritis, Psoriasis, Psoriatic arthritis, Multiple sclerosis and Sjogren syndrome; Th2 cells and is selected from the group consisting of Atopic dermatitis and Asthma; Thl 7 cells and is Psoriasis; cells of innate immune system; and/or by loss or absence Treg cells or imbalance Treg/Tconv cell ratio.

In one embodiment the autoimmune and inflammatory disease is driven by Thl cells and is selected from the group consisting of acute GvHD, chronic GvHD, Type 1 diabetes, Inflammatory bowel disease, Crohn’s disease, Ulceritis, Rheumatoid arthritis, Psoriasis, Psoriatic arthritis, Multiple sclerosis, Sjogren syndrome.

In one embodiment the autoimmune and inflammatory disease is driven by Th2 cells and is selected from the group consisting of Atopic dermatitis and Asthma. In one embodiment the autoimmune and inflammatory disease is driven by Thl7 cells and is Psoriasis.

In one embodiment the autoimmune and inflammatory disease is driven by loss or absence of Treg cells or due to imbalance of Treg/Tconv cell ratio and is selected from the group consisting of acute GvHD, chronic GvHD, steroid refractory GvHD, Type 1 diabetes, Inflammatory bowel disease, Crohn’s disease, Ulceritis, Rheumatoid arthritis, Psoriasis, Psoriatic arthritis, Multiple sclerosis, Sjogren syndrome, Atopic dermatitis, Asthma, ALS.

In one embodiment the autoimmune and inflammatory disease is driven by cells of innate immune system selected from the group consisting of macrophages and dendritic cells.

In one embodiment the autoimmune and inflammatory disease is driven by a cytokine selected from the group consisting of IL-6, IFN-g, IL-17, IL12a, IL-10, and TGF-b, or any combination thereof. In one embodiment the autoimmune and inflammatory disease is driven by IL-6. In one embodiment the autoimmune and inflammatory disease is driven by IFN-g. In one embodiment the autoimmune and inflammatory disease is driven by IL-17. In one embodiment the autoimmune and inflammatory disease is driven by IL12a. In one embodiment the autoimmune and inflammatory disease is driven by TGF-b.

In one embodiment the autoimmune and inflammatory disease is selected from the group consisting of inflammatory diseases; autoimmune diseases; allergic diseases; autoinflammatory diseases; and Graft-versus-Host-Disease (GvHD)/transplant rejection. The term “transplant rejection” as referred herein includes organ transplants e.g. kidney transplants, liver transplants and heart transplants. In a preferred embodiment the autoimmune and inflammatory disease is selected from the group consisting of autoimmune diseases; allergic diseases; autoinflammatory diseases; and Graft-versus-Host-Disease (GvHD). In a more preferred embodiment the autoimmune and inflammatory disease is selected from the group consisting of autoimmune diseases; allergic diseases; and Graft-versus-Host-Disease (GvHD). In an even more preferred embodiment the autoimmune and inflammatory disease is selected from the group consisting of autoimmune diseases; and Graft-versus-Host-Disease (GvHD). In a preferred embodiment the autoimmune and inflammatory disease is selected from the group consisting of acute, chronic and steroid refractory Graft-versus-Host-Disease (GvHD), Type 1 diabetes, inflammatory bowel disease, rheumatoid arthritis, Sjogren syndrome, asthma, allergies, atopic dermatitis, psoriasis, inflammation in organ transplantation, organ transplant rejection, Crohn’s disease, Ulcerative colitis, Psoriatic arthritis, Systemic lupus erythematosus/Lupus nephritis, Multiple sclerosis, Neuromyelitis optica, Pemphigus vulgaris, Chronic spontaneous urticaria, Myasthenia gravis, Uveitis, autoimmune hepatitis, primary biliary cirrhosis, Behcet disease, Kawasaki disease, vasculitis, Felty syndrome, Guillain-Barre Syndrome, Autoimmune polyglandular syndrome 1, polyglandular syndrome type 2, Autoimmune pancreatitis, Celiac disease, Addison's disease, and autoimmune thyroiditis

In one embodiment the autoimmune and inflammatory disease is selected from the group consisting of acute, chronic and steroid refractory Graft-versus-Host-Disease (GvHD), Type 1 diabetes, sjogren syndrome, asthma, allergies, Ulcerative colitis, Psoriatic arthritis, Systemic lupus erythematosus/Lupus nephritis, Neuromyelitis optica, Pemphigus vulgaris, Chronic spontaneous urticaria, Myasthenia gravis, Uveitis, autoimmune hepatitis, primary biliary cirrhosis, Behcet disease, Kawasaki disease, vasculitis, Felty syndrome, Guillain-Barre Syndrome, Autoimmune polyglandular syndrome 1, polyglandular syndrome type 2, Autoimmune pancreatitis, Celiac disease, Addison's disease, and autoimmune thyroiditis.

In a more preferred embodiment the autoimmune and inflammatory disease is selected from the group consisting of acute, chronic and steroid refractory Graft-versus-Host-Disease (GvHD).

In a more preferred embodiment the autoimmune and inflammatory disease is acute GvHD.

In a more preferred embodiment the autoimmune and inflammatory disease is chronic GvHD.

In a more preferred embodiment the autoimmune and inflammatory disease is steroid refractory GvHD.

In a more preferred embodiment the autoimmune and inflammatory disease is selected from the group consisting of acute, chronic and steroid refractory Graft-versus-Host-Disease (GvHD), Ulcerative colitis, Psoriatic arthritis, Systemic lupus erythematosus/Lupus nephritis, Neuromyelitis optica, Pemphigus vulgaris, Chronic spontaneous urticaria, Myasthenia gravis, Uveitis, autoimmune hepatitis, primary biliary cirrhosis, Behcet disease, Kawasaki disease, vasculitis, Felty syndrome, Guillain-Barre Syndrome, Autoimmune polyglandular syndrome 1, polyglandular syndrome type 2, Autoimmune pancreatitis, Celiac disease, Addison's disease, and autoimmune thyroiditis.

In a more preferred embodiment the autoimmune and inflammatory disease is selected from the group consisting of Ulcerative colitis, Psoriatic arthritis, Systemic lupus erythematosus/Lupus nephritis, Neuromyelitis optica, Pemphigus vulgaris, Chronic spontaneous urticaria,

Myasthenia gravis, Uveitis, autoimmune hepatitis, primary biliary cirrhosis, Behcet disease, Kawasaki disease, vasculitis, Felty syndrome, Guillain-Barre Syndrome, Autoimmune polyglandular syndrome 1, polyglandular syndrome type 2, Autoimmune pancreatitis, Celiac disease, Addison's disease, and autoimmune thyroiditis.

In a more preferred embodiment the autoimmune and inflammatory disease is Type 1 diabetes.

In an even more preferred embodiment the autoimmune and inflammatory disease is acute, chronic and steroid refractory Graft-versus-Host-Disease (GvHD), in particular acute and chronic GvHD.

In a particular preferred embodiment the autoimmune and inflammatory disease is Graft- versus-Host-Disease (GvHD) developed following allo-hematopoetic stem cell transplant (allo- HSCT).

In one embodiment Treg cells are expanded in a subject who receives a TCM compared to a control or compared to a subject who receives a vehicle. A “vehicle” as referred herein is a substance without preventive and/or therapeutic action. Thus, if a T cell modulator or a pharmaceutical composition thereof is administered to a subject according to the method of the present invention, the Treg cells of this subject or the Treg cells in an organ of the subject are expanded compared to a control or compared to cells of the subject who receives a vehicle. In a further aspect the present invention provides a kit for use in a method for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease in a subject, comprising a container and a package insert, wherein the container comprises at least one dose of a medicament comprising a T cell modulator (TCM) or a pharmaceutical composition thereof, and the package insert comprises optionally instructions for treating a subject for an autoimmune and inflammatory disease (AIID) using the medicament.

In a further aspect the present invention provides a T cell modulator (TCM) for use in a method for the prevention, delay of progression or treatment of an autoimmune and inflammatory disease (AIID) in a subject in combination with standard of care or immunosuppressive drugs. “Standard of care” as refered herein describes a treatment process approved in clinics for a certain disease and includes treatment with Ruxolitinib, Tocilizumab, Infliximab, Secukinumab, Dupilumab. Immunosuppressive drugs as refered herein includes different types of compounds which have the effect of dampening the response of the immune system. Corticosteroids are an example of immunosuppressive drugs.

Examples

The present examples are intended to illustrate the present invention without restricting it.

Material and Methods:

Human PBMCs and mouse splenocyte in vitro stimulation and treatment: Human peripheral blood mononuclear cells (PBMCs) and mouse splenocytes from donors were stimulated in vitro with anti-CD3 and anti-CD28 antibodies. The stimulated cells were simultaneously treated with compounds for 96 hours. The T cells were profiled for different cell surface and intracellular markers and proliferation by flow cytometry.

In vivo treatment of mice and pharmacodynamic response: Mice experiments were performed in compliance with authorization to perform animal experiments (VD3323). Briefly, 6-8 weeks old wild type C57B16 mice were subcutaneously or intraperitoneally dosed with vehicle or compounds for different durations. Mice were sacrificed, spleens were harvested 24 hours post-last dosing. Splenocytes were analyzed for cell surface and intracellular markers expression using flow cytometry. RNA expression analyses were performed using quantitative real time PCR.

Humanized GvHD mouse model: 6-8 weeks old NSG mice were intraperitoneally injected with 10 million human peripheral blood mononuclear cells (PBMCs) as below:

Ex vivo treatment of PBMCs: 10 million human PBMCs from healthy donor #1 were ex-vivo treated with DMSO and 10 mM of 6-(4-tert-butylphenoxy)pyridin-3 -amine for 6 hours. Ex- vivo treated PBMCs were intraperitoneally injected into mice (10 million per mouse). 10 NSG mice were injected with DMSO pre-treated PBMCs and 10 NSG mice were injected with 6-(4- tert-butylphenoxy)pyridin-3 -amine pre-treated PBMCS. Following PBMC injections mice were monitored for up to 42 days for symptoms of GvHD development as measured by standardized GvHD clinical score system. No in-vivo drug treatment was administered.

Animals were scored according to GvHD clinical scoring system as shown in table 1 below:

Table 1: GvHD clinical score

A cumulative GvHD clinical score was calculated by addition of scores for individual parameters (e.g cumulative score = BWL + posture + activity† fur texture + skin integrity + paleness).

Ex-vivo pre-treatment of PBMCs and in-vivo treatment of mice: Human PBMCs were ex- vivo treated with either DMSO or 6-(4-tert-butylphenoxy)pyri din-3 -amine and then intraperitoneally injected into NSG mice. Mice were in vivo treated with different compounds as below: i. DMSO ex vivo hPBMCs treatment (7 hrs), plus vehicle in vivo treatment ii. DMSO ex vivo hPBMCs treatment (7 hrs), plus 6-(4-tert-butylphenoxy)pyridin-3- amine in vivo treatment for 3 days iii. 6-(4-tert-butylphenoxy)pyri din-3 -amine ex vivo hPBMCs treatment (7 hrs), plus 6-(4- tert-butylphenoxy)pyri din-3 -amine in vivo treatment for 3 days iv. DMSO ex vivo PBMCs treatment (7 hrs) plus Ruxolitinib in vivo treatment for 3 days v. 6-(4-tert-butylphenoxy)pyridin-3-amine ex vivo hPBMCs treatment (7 hrs), plus Ruxolitinib in vivo treatment for 3 days

Following PBMC injections, mice were treated with vehicle or 6-(4-tert-butylphenoxy)pyridin- 3-amine or Ruxolitinib starting at day 0. Mice were monitored from 60-85 days for symptoms of GvHD and scored according to a standard clinical scoring system for GvHD (Naserian et al., 2018) (see table 1) and a cumulative clinical score was obtained by addition of clinical score for individual parameters. A maximum score of 6 is measured when reaching endpoint.

Cell proliferation assay: The effect of compounds on T cell proliferation was quantified using Cell trace violet (CTV). PBMCs/splenocytes were incubated with CTV for 20 minutes at 37°C. Cells were washed twice with RPMI complete medium. CTV labelled cells were stimulated with anti-CD3 and anti-CD28 antibodies and treated with control or compounds and cultured at 37°C for 96 hours. The cell division index was measured by flow cytometry.

Flow cytometry analysis: Flow cytometry analysis was performed to quantify cell surface and intracellular markers expression and measure cell division index.

Viability staining: Cells were transferred into a 96 well conical bottom plate and washed with serum free PBSlx. Cells were incubated with Zombie NIR (Biolegend #423105) + FC block (CD16/31) for 20 minutes at 4°C. Cells were washed with staining medium (SM) (lx HBSS, 2.5mM EDTA, 2% NBS

Cell surface marker staining: Cells were stained with antibodies against cell surface markers (CD3, CD4, CD8, CD25, Biolegend or ThermoFisher) for 20 minutes at 4°C. Cells were washed with SM before continuing with nuclear staining.

FOXP3 nuclear staining: Cells were fixed and permeabilized with Foxp3 Transcription Factor Staining Buffer Set (ThermoFisher #00-5523) for 20 minutes at room temperature. Following washing with lx permeabilization buffer, cells were incubated with blocking solution for 15 minutes at room temperature. The appropriate dilution of FOXP3 antibody (Biolegend or ThermoFisher) was the added to the blocking solution. Cells were then resuspended and incubated for 30 minutes at room temperature. Following washing with lx permeabilization buffer, cells were resuspended in staining media for FACS acquisition by Cytoflex S (Beckman Coulter). Results are finally analysed with the software FlowJo v.10.7.1.

ELISA assay: Supernatant from activated human PBMC cultures described above was separated from cells at the end of culture time. Key cytokines (IL-6, IFN-g, IL-17) were quantified using sandwich ELISA reagents set (Diaclone) following manufacturer’s instruction manual.

Cytokine profiling in human plasma samples: Plasma samples from patients treated with 6- (4-tert-butylphenoxy)pyri din-3 -amine were collected and stored at -80 °C until analysed. Cytokine and chemokine profiling was performed using MesoScale Discovery (MSD) platform and three assay panels: 1) Pro-inflammatory panel human, 2) Human IL-6 assay S-Plex, 3) Human IL-10 assay S-Plex.

Allogeneic Bone marrow transplant: Recipient Balb/c mice were irradiated twice 3.75Gy (T=0 and T=4h) in an X-ray source irradiator before transplantation at T=8h. Treatment with antibiotics (Nopil) and analgesic (Dafalgan) was supplemented to drinking water 4 days before injection. Irradiated recipient Balb/c mice were transplanted with 5 million T cell depleted bone marrow+ 1 million splenocytes. Allogeneic recipient animals were treated with vehicle or 6-(4- tert-butylphenoxy)pyridin-3 -amine subcutaneously at 20 mg/kg for two days immediately after allo-BM transplant, given a 10 days treatment holiday to recover body weight loss due to whole body irradiation and then treatment continued at 20 mg/kg . A last group was treated intraperitoneally with Cyclosporin A at 5 mg/kg for 2 days (QD), and then treatment continued at 5 mg/kg (QDX5) after break for 10 days. Animals were monitored for symptoms of GvHD development (severe body weight loss, alopecia and diarrhea.

Cytokine analysis in allogeneic Bone marrow transplant: The recipient animals were bled on day 36 and blood plasma was extracted to determine inflammatory cytokine concentration (IFNY, IL-6, IL-17a) by LEGENDplexTM multiplexed assay.

Results:

Expansion of Treg cells

As can be seen from Table 2 below and Figures 1 and 2, respectively, compounds lead to an expansion of Treg cells in activated mouse splenocytes and in vivo treated mice.

Table 2: Compounds leads to an expansion of Treg cells in activated human PBMC cultures.

Inhibition of proliferation of activated CD4 T cells

As can be seen from Table 3 below compounds inhibit proliferation of activated CD4 T cells.

Table 3: Compounds inhibit proliferation of activated CD4 T cells: Percentage inhibition of CD4 T cells.

Inhibition of proliferation of activated CD8 T cells As can be seen from Table 4 below compounds inhibit proliferation of activated CD8 T cells.

Table 4: Compounds inhibit proliferation of activated CD8 T cells. Percentage inhibition of CD8 T cells.

Inhibition of pro-inflammatory cytokine expression As can be seen from Table 5 below compounds inhibit pro-inflammatory cytokine expression.

Table 5: Compounds inhibit pro-inflammatory cytokine expression. Percentage downregulation of pro-inflammatory cytokines in activated human PBMC cultures.

Inhibition of pro-inflammatory cytokine expression As can be seen from Table 6 below 6-(4-tert-butylphenoxy)pyridin-3-amine inhibits pro- inflammatory cytokines in human patients.

Table 6: 6-(4-tert-butylphenoxy)pyridin-3-amine inhibits pro-inflammatory cytokines in human patients. Percentage downregulation of pro-inflammatory cytokines in blood plasma of human patients treated with 6-(4-tert-butylphenoxy)pyri din-3 -amine. Upregulation of anti- inflammatory cytokine

As can be seen from Table 7 below and Figures 3 and 4, respectively, 6-(4-tert- butylphenoxy)pyri din-3 -amine upregulates anti- inflammatory cytokine in human patients.

Table 7: 6-(4-tert-butylphenoxy)pyridin-3-amine upregulates anti- inflammatory cytokine in human patients. Percentage upregulation of IL-10 in blood plasma of human patient treated with 6-(4-tert-butylphenoxy)pyri din-3 -amine.

Prophylactic treatment with 6-(4-tert-butylphenoxy)pyridin-3-amine treats humanized mouse model of GvHD: As can be seen from Table 8 below treatment with 6-(4-tert-butylphenoxy)pyri din-3 -amine decreased cumulative GvHD score significantly in animals injected with human PBMCs pretreated (ex- vivo) with DMSO and 6-(4-tert-butylphenoxy)pyri din-3 -amine.

Table 8: GvHD clinical score at day 21 in animals injected with human PBMCs pre-treated (ex- vivo) with DMSO and 6-(4-tert-butylphenoxy)pyri din-3 -amine. As can be seen from Table 9 below treatment with 6-(4-tert-butylphenoxy)pyridin-3-amine decreased cumulative GvHD score significantly in NSG mice injected with ex- vivo treated human PBMCs followed by in vivo treatment with 6-(4-tert-butylphenoxy)pyri din-3 -amine and Ruxolitinib.

Table 9: GvHD clinical score in NSG mice injected with ex- vivo treated human PBMCs followed by in vivo treatment with 6-(4-tert-butylphenoxy)pyri din-3 -amine and Ruxolitinib.

Alleviation of GvHD in an allo-bone marrow transplant model: Graft vs host disease (GvHD) is an example of alloimmune disease that develops following allo-hematopoetic stem cell transplant (allo-HSCT) in humans. The activity of 6-(4-tert- butylphenoxy)pyri din-3 -amine and a calcineurin inhibitor Cyclosprin A was investigated in a mouse model of GvHD following allo-bone marrow transplant. In this model, T cell depleted bone marrow cells (TCD-BM) + splenocytes from C57B16 mice were transplanted into irradiated Balb/c mice. As shown in figure 5, while 60 % of vehicle treated animals died due to GvHD, only 20 % of 6-(4-tert-butylphenoxy)pyridin-3-amine treated developed GvHD. Cyclosporin A, which is used as standard of care for the prevention of GvHD did not perform better than vehicle treated animals. This data shows that in an allogeneic-hematopoetic stem cell transplanted (or allo-bone marrow transplant) setting, 6-(4-tert-butylphenoxy )pyri din-3 - amine prevents development of GvHD. In addition, treatment of allo-BM + splenocyte transplanted mice with 6-(4-tert-butylphenoxy)pyri din-3 -amine led to a downregulation of cytokines implicated in GvHD pathophysiology (INFG, IL-17 and IL-6, figure 6) and an expansion of regulatory T cells.