Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SELECTIVE POLYPEPTIDE SYNTHESIS STALLING ASSAY AND COMPOUND
Document Type and Number:
WIPO Patent Application WO/2021/078995
Kind Code:
A1
Abstract:
The invention relates to compounds, in particular pyrimidine-2,4-diamines, analogues and salts thereof and pharmaceutical compositions comprising pyrimidine-2,4-diamines analogues and salts thereof for use in the treatment and prevention of a disease, in particular a growth hormone receptor-dependent condition. The invention also relates to methods of using these compounds and compositions to treat physiological disorders related to the amount or activity of growth hormone. In a particular embodiment, the invention relates to a compound according to formula 1 for use in the treatment or prevention of a disease in a subject.

Inventors:
STROUS GERARDUS JACOBUS ANTONIUS MARIA (NL)
MOL JAN ADRIANUS (NL)
VAN DER VELDEN LIEKE MARIE (NL)
KLUMPERMAN JUDITH (NL)
MAAS PETRUS EMMANUEL MARIE (NL)
PIET DENNIS PATRICK (NL)
DE KLERK-SPRENKELS NANDA ELISABETH (NL)
TIJHUIS JOHANN HEINRICH (NL)
VIÉTOR HENDRIK ENGELBERTUS (NL)
MAURICE MADELON MARIA (NL)
VAN DER KRIFT FELIX (NL)
Application Number:
PCT/EP2020/080029
Publication Date:
April 29, 2021
Filing Date:
October 26, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UMC UTRECHT HOLDING BV (NL)
UNIV UTRECHT HOLDING BV (NL)
BIMINI BIOTECH B V (NL)
International Classes:
A61K31/505; C12Q1/6804; A61P3/10; A61P35/02; C12Q1/6883
Domestic Patent References:
WO2009046448A12009-04-09
WO2013070620A12013-05-16
WO2009025919A22009-02-26
WO1998020878A11998-05-22
WO2006050843A12006-05-18
WO2004101568A12004-11-25
WO2011153310A12011-12-08
WO1993013079A11993-07-08
WO1998020878A11998-05-22
Foreign References:
US20150225353A12015-08-13
Other References:
OZTEKIN ALGUL ET AL: "2,4-Diamino-5-(2-arylpropargyl)pyrimidine derivatives as new nonclassical antifolates for human dihydrofolate reductase inhibition", JOURNAL OF MOLECULAR GRAPHICS AND MODELLING, ELSEVIER SCIENCE, NEW YORK, NY, US, vol. 29, no. 5, 5 November 2010 (2010-11-05), pages 608 - 613, XP028141150, ISSN: 1093-3263, [retrieved on 20101111], DOI: 10.1016/J.JMGM.2010.11.004
AHN H-S ET AL: "Structure-activity relationships of pyrroloquinazolines as thrombin receptor antagonists", BIORGANIC & MEDICINAL CHEMISTRY LETTERS, ELSEVIER, AMSTERDAM , NL, vol. 9, no. 14, 19 July 1999 (1999-07-19), pages 2073 - 2078, XP004171640, ISSN: 0960-894X, DOI: 10.1016/S0960-894X(99)00339-X
KUNZE MICHAEL M ET AL: "sST2 translation is regulated by FGF2viaan hnRNP A1-mediated IRES-dependent mechanism", BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS, ELSEVIER, AMSTERDAM, NL, vol. 1859, no. 7, 8 May 2016 (2016-05-08), pages 848 - 859, XP029594462, ISSN: 1874-9399, DOI: 10.1016/J.BBAGRM.2016.05.005
FIELD JUDITH ET AL: "The MS Risk Allele of CD40 Is Associated with Reduced Cell-Membrane Bound Expression in Antigen Presenting Cells: Implications for Gene Function", PLOS ONE, vol. 10, no. 6, 11 June 2015 (2015-06-11), pages e0127080, XP055782003, DOI: 10.1371/journal.pone.0127080
MOON YUSEOK: "Ribosomal Alteration-Derived Signals for Cytokine Induction in Mucosal and Systemic Inflammation: Noncanonical Pathways by Ribosomal Inactivation", MEDIATORS OF INFLAMMATION., vol. 2014, 1 January 2014 (2014-01-01), GB, pages 1 - 10, XP055782009, ISSN: 0962-9351, Retrieved from the Internet DOI: 10.1155/2014/708193
SCHWARTZ ET AL., NAT REV RHEUMATOL, vol. 12, no. 1, January 2016 (2016-01-01), pages 25 - 36
LONGLEYJOHNSTON: "Molecular mechanisms of drug resistance", PATHOL, vol. 205, no. 2, January 2005 (2005-01-01), pages 275 - 92
ARUMUGAM ET AL., EXPERIMENTAL & MOLECULAR MEDICINE, vol. 51, 2019, pages 2
YI ET AL., KOREAN J PHYSIOL PHARMACOL, vol. 16, February 2012 (2012-02-01), pages 11 - 16
MORALES ET AL., ONCOGENE, vol. 24, 2005, pages 6842 - 6847
PETERS, CANCER DRUG RESIST, vol. 1, 2018, pages 1 - 5
BLOUNT ET AL., PROC NATL ACAD SCI USA., vol. 94, no. 7, 1 April 1997 (1997-04-01), pages 3290 - 5
ALGUL ET AL., J MOL GRAPH MODEL, vol. 29, no. 5, February 2011 (2011-02-01), pages 608 - 613
VISWANATHAN ET AL., PLOS ONE, vol. 7, no. 2, pages e29434
PAULKSEN ET AL., BIOORG MED CHEM, vol. 17, no. 14, 15 July 2009 (2009-07-15), pages 4866 - 4872
BASU, R.Y. QIANJ.J. KOPCHICK: "MECHANISMS IN ENDOCRINOLOGY: Lessons from growth hormone receptor gene disrupted mice: Are there benefits of endocrine defects?", EUR J ENDOCRINOL, 2018
BUCHMAN, M.S. BELLJ.J. KOPCHICK: "Growth Hormone Discovery and Structure", PEDIATR ENDOCRINOL REV, vol. 16, 2018, pages 2 - 10
CHHABRA, Y.WATERS, M.J.BROOKS, A.J.: "Role of the growth hormone-IGF-1 axis in cancer", EXPERT. REV.ENDOCRINOL. METAB., 2011, pages 71 - 84, XP009167646, DOI: 10.1586/eem.10.73
CHRISTGEN, M.P. DERKSEN.: "Lobular breast cancer: molecular basis, mouse, and cellular models", BREAST CANCER RES, vol. 17, 2015, pages 16, XP021213340, DOI: 10.1186/s13058-015-0517-z
DEHKHODA, F.C.M.M. LEEJ. MEDINAA.J. BROOKS: "The Growth Hormone Receptor: Mechanism of Receptor Activation, Cell Signaling, and Physiological Aspects", FRONTIERS IN ENDOCRINOLOGY, vol. 9, 2018, pages 35
FURQAN, M.N. MUKHIB. LEED. LIU: "Dysregulation of JAK-STAT pathway in hematological malignancies and JAK inhibitors for clinical application", BIOMORKER RESEARCH, vol. 1, 2013, pages 5, XP021138327, DOI: 10.1186/2050-7771-1-5
GUEVARA-AGUIRRE, J.P. BALASUBRAMANIANM. GUEVARA-AGUIRREWEI, F. MADIAC.W. CHENGD. HWANGA. MARTIN-MONTALVOJ. SAAVEDS. INGLESR. DE CA: "Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans", SCIENCE TRANSLATIONAL MEDICINE, vol. 3, 2011, pages 70
HOLLESTELLE, A.J.K. PEETERSM. SMIDM. TIMMERMANSL.C. VERHOOGP.J. WESTENENDA.A. HEINEA. CHANA.M. SIEUWERTSE.A. WIEMER: "Loss of E-cadherin is not a necessity for epithelial to mesenchymal transition in human breast cancer", BREAST CANCER RES TREAT, vol. 138, 2013, pages 47 - 57
JESSIMAN, A.G.D.D. MATSONF.D. MOORE: "Hypophysectomy in the treatment of breast cancer", N ENGL J MED, vol. 261, 1959, pages 1199 - 1207
LARON, Z: "Lessons from 50 Years of Study of Laron Syndrome", ENDOCRINE PRACTICE: OFFICIAL JOURNAL OF THE AMERICAN COLLEGE OF ENDOCRINOLOGY AND THE AMERICAN ASSOCIATION OF CLINICAL ENDOCRINOLOGISTS, vol. 21, 2015, pages 1395 - 1402
LU, M.J.U. FLANAGANR.J. LANGLEYM.P. HAYJ.K. PERRY: "Targeting growth hormone function: strategies and therapeutic applications", SIGNAL TRANSDUCTION AND TARGETED THERAPY, vol. 4, 2019, pages 3
MALERGUE, F.A. VAN AGTHOVENC. SCIFOD. EGANG.J. STROUS: "utomation of a phospho-STAT5 staining procedure for flow cytometry for application in drug discovery", J BIOMOL SCREEN, vol. 20, 2015, pages 416 - 421
MINOIA, M.E. GENTILIND. MOLEM. ROSSIC. FILIERIF. TAGLIATIA. BARONIM.R. AMBROSIOE. DEGLI UBERTIM.C. ZATELLI: "Growth hormone receptor blockade inhibits growth hormone-induced chemoresistance by restoring cytotoxic-induced apoptosis in breast cancer cells independently of estrogen receptor expression", J CLIN ENDOCRINOL METAB, vol. 97, 2012, pages E907 - 916, XP055475289, DOI: 10.1210/jc.2011-3340
NASHIRO, K.J. GUEVARA-AGUIRREM.N. BRASKIEG.W. HAFZALLAR. VELASCOP. BALASUBRAMANIANM. WEIP.M. THOMPSONM. MATHER M.D. NELSONA. GUEVA: "Brain Structure and Function Associated with Younger Adults in Growth Hormone Receptor-Deficient Humans", J NEUROSCI, vol. 37, 2017, pages 1696 - 1707
NESPITAL, T.L.M. VAN DER VELDENA. MENSINGAE.D. VAN DER VAARTG.J. STROUS: "Fos-Zippered GH Receptor Cytosolic Tails Act as Jak2 Substrates and Signal Transducers", MOL ENDOCRINOL, vol. 30, 2016, pages 290 - 301
RANKE, M.B.J.M. WIT.: "Growth hormone - past, present and future", NAT REV ENDOCRINOL, vol. 14, 2018, pages 285 - 300, Retrieved from the Internet
SEDEK, M.L.M. VAN DER VELDENG.J. STROUS: "Multimeric growth hormone receptor complexes serve as signaling platforms", J. BIOL. CHEM., vol. 289, 2014, pages 65 - 73
THEODOROPOULOU, M.G.K. STALLA: "Somatostatin receptors: from signaling to clinical practice", FRONTIERS IN NEUROENDOCRINOLOGY, vol. 34, 2013, pages 228 - 252
TIMMERMANS-SPRANGE.P.M.A. GRACANINJ.A. MOL: "Molecular Signaling of Progesterone, Growth Hormone, Wnt, and HE in Mammary Glands of Dogs, Rodents, and Humans: New Treatment Target Identification", FRONTIERS IN VETERINARY SCIENCE, vol. 4, 2017, pages 53
VERMEULEN, J.F.A.S. VAN BRUSSELA. ADAMSW.P. MALIE. VAN DER WALLP.J. VAN DIESTP.W. DERKSEN: "Near-infrared fluorescence molecular imaging of ductal carcinoma in situ with CD44v6-specific antibodies in mice: a preclinical study", MOLECULAR IMAGING AND BIOLOGY: MIB: THE OFFICIAL PUBLICATION OF THE ACADEMY OF MOLECULAR IMAGING, vol. 15, 2013, pages 290 - 298
ZHOU, Y.B.C. XUH.G. MAHESHWARIL. HEM. REEDM. LOZYKOWSKIS. OKADAL. CATALDOK. COSCHIGAMOT.E. WAGNER: "A mammalian model for Laron syndrome produced by targeted disruption of the mouse growth hormone receptor/binding protein gene (the Laron mouse", PROC NATL ACAD SCI U SA, vol. 94, 1997, pages 13215 - 13220, XP002312293, DOI: 10.1073/pnas.94.24.13215
Attorney, Agent or Firm:
WENSVOORT, Gert (NL)
Download PDF:
Claims:
CLAIMS

1. A polypeptide synthesis stalling assay comprising a protein-expression system provided with a ribonucleic acid encoding a polypeptide comprising at least a C-terminal part of a cytosolic tail of a type I/ll cytokine receptor and provided with a test molecule.

2. A polypeptide synthesis stalling assay according to claim 1 wherein said test molecule is selected from the group of molecules having general formula 1, formula 1 or having general formula 2, formula 2.

3. A polypeptide synthesis stalling assay according to claim 1 or 2 that is cell-free. 4. A polypeptide synthesis stalling assay according to anyone of claims 1 to 3 wherein said ribonucleic acid comprises ribonucleic acid encoding a leucine zipper 5'-terminal and/3'- terminal to said ribonucleic acid encoding said C-terminal part.

5. A polypeptide synthesis stalling assay according to anyone of claims 1 to 4 wherein said ribonucleic acid also encodes a reporter tag.

6. A polypeptide synthesis stalling assay according to anyone of claims 1 to 5 wherein box-1- mediated JAK binding activity is at least functionally deleted from said cytosolic tail.

7. A polypeptide synthesis stalling assay according to anyone of claims 1 to 6 wherein said receptor is a type I cytokine receptor.

8. A polypeptide synthesis stalling assay according to anyone of claims 1 to 7 wherein said receptor is a growth hormone receptor.

9. A polypeptide synthesis stalling assay according to anyone of claims 1 to 8 wherein said ribonucleic acid encodes a polypeptide provided with a least the last 44 C-terminal amino acids of said cytosolic tail of said receptor.

10. A polypeptide synthesis stalling assay according to claim 8 wherein said ribonucleic acid encodes a polypeptide provided with a C-terminal part of a growth hormone receptor at least comprising an amino acid sequence starting after position 594.

11. A set of at least two ribonucleic acids comprising a. a first ribonucleic acid encoding a polypeptide comprising at least a C-terminal part of a cytosolic tail of a type I/ll cytokine receptor and b. a second ribonucleic acid encoding a polypeptide provided with an at least functional deletion or truncation of a C-terminal part of said cytosolic tail of said cytokine receptor.

12. A set of at least two ribonucleic acids according to claim 11 wherein said functional deletion or truncation comprises a least the last 44 C-terminal amino acids of said C- terminal part of said cytosolic tail of said receptor.

13. A set of at least two ribonucleic acids according to claim 11 or 12 wherein said first and/or said second ribonucleic acid is or are also provided with at least one ribonucleic acid encoding a leucine zipper 5'-terminal and/or 3'-terminal to said ribonucleic acid. 14. A set of at least two ribonucleic acids according anyone of claims 11 to 13 wherein said first and/or said second ribonucleic acid is or are also provided with at least one ribonucleic acid encoding a reporter tag.

15. A set of at least two ribonucleic acids according to anyone of claims 11 to 14 wherein box- 1-mediated JAK binding activity is at least functionally deleted from said cytosolic tail.

16. A set of at least two ribonucleic acids according to anyone of claims 11 to 15 wherein said receptor is a type I cytokine receptor.

17. A set of at least two ribonucleic acids according to anyone of claims 11 to 16 wherein said receptor is a growth hormone receptor (GRH).

18. A set of at least two ribonucleic acids according anyone of claims 11 to 17 wherein said second ribonucleic acid encodes said cytosolic tail that is C-terminally truncated with at least the last 44 C-terminal amino acids of said cytosolic tail of said receptor.

19. A set of at least two ribonucleic acids according to claim 17 wherein said second ribonucleic acid is encoding a polypeptide comprising at least a C-terminally truncated part of said C-terminal part of said cytosolic tail, wherein said tail is C-terminally truncated at around position 594.

20. A set of at least two ribonucleic acids according anyone of claims 11 to 19 for use in a method for testing a molecule for the capacity to selectively inhibit synthesis of a receptor of the type I cytokine receptor family.

21. A set of at least two ribonucleic acids according anyone of claims 11 to 20 wherein said molecule is selected from the group of compounds with formula 1 or formula 2.

22. A set of at least two ribonucleic acids according anyone of claims 11 to 21 for use as target and control in a method for identifying a molecule capable of selectively inhibiting synthesis of a receptor of the type I cytokine receptor family,

23. A method for testing a molecule for the capacity to selectively inhibitsynthesis of a receptor of the type I cytokine receptor family, said method comprising a. providing a cytosol-like solution comprising a ribosome complex or polyribosome with a first ribonucleic acid encoding a polypeptide comprising at least a C- terminal part of a cytosolic tail of a type I cytokine receptor, b. providing said solution with a test molecule, c. determining the effect of the test molecule on translation of said first ribonucleic acid in synthesizing said polypeptide by determining the presence of said polypeptide in said solution. 24. A method according to claim 23 and determining the presence of said polypeptide translated in the absence of said test molecule.

25. A method according to claim 23 or 24 and a. providing a cytosol-like solution comprising a ribosome complex or polyribosome with a second ribonucleic acid encoding a polypeptide provided with an at least functional deletion or truncation of a C-terminal part of said cytosolic tail of a type I cytokine receptor b. providing said solution with a test molecule, c. determining the effect of the test molecule on translation of said second ribonucleic acid in synthesizing said polypeptide by determining the presence of said polypeptide in said solution.

26. A method according to anyone of claims 23 to 25 wherein said test molecule is selected from the group of molecules having general formula 1, formula 1 or having general formula 2, formula 2.

27. A method according to anyone of claims 23 to 26 wherein the solution is cell-free.

28. A method according to anyone of claims 23 to 26 wherein the solution is in a living cell in vitro.

29. A method according to anyone of claims 23 to 26 wherein the solution is in a living cell.

30. A method according to anyone of claims 23 to 29 wherein said ribonucleic acid comprises ribonucleic acid encoding a leucine zipper 5'-terminal and/3'-terminal to said ribonucleic acid encoding said C-terminal part.

31. A method according to anyone of claims 23 to 30 wherein said ribonucleic acid also encodes a reporter tag.

32. A method according to anyone of claims 23 to 31 wherein box-l-mediated JAK binding activity is at least functionally deleted from said cytosolic tail.

33. A method according to anyone of claims 23 to 32 wherein said receptor is a group 1 type I cytokine receptor.

34. A method according to anyone of claims 23 to 33 wherein said receptor is a growth hormone receptor.

35. A method according to anyone of claims 23 to 34 wherein said ribonucleic acid encodes a polypeptide provided with a least the last 44 C-terminal amino acids of said cytosolic tail of said receptor.

36. A method according to anyone of claims 23 to 35 wherein said ribonucleic acid encodes a polypeptide provided with a C-terminal part of a growth hormone receptor at least comprising an amino acid sequence starting after position 594. 37. A method according to anyone of claims 23-36 wherein said ribonucleic acid is selected from a set of at least two ribonucleic acids according to anyone of claims 11-22.

38. A method according to anyone of claims 23 to 37 wherein said ribonucleic acid at least encodes for a polypeptide encoding at least a part of a cytosolic tail having >70% homology, preferably >80% homology, preferably >90% homology, more preferably 95% homology with an polypeptide amino acid sequence 387-688 of preferably human GHR shown in figure 8.

39. A molecule capable of selectively stalling protein synthesis of a receptor of the type I cytokine receptor family.

40. A molecule capable of selectively stalling protein synthesis of a receptor of the group 1 type I cytokine receptor family.

41. A molecule capable of selectively stalling protein synthesis of a receptor of the growth hormone receptor.

42. A molecule according to claim 39 to 41 identifiable with an assay according to anyone of claims 1 to 10 or identifiable with a method according to anyone of claims 23 to 37.

43. A molecule according to anyone of claims 39 to 42, or functional equivalent thereof, selected from the group of molecules herein identified as AK-105/40836349, AK- 105/40837635, AA-504/32626008, AK-105/40836340, AK-105/40833746, AK- 105/40693663, AK-105/40833503, AK-105/40837674, AK-105/40836874, AK- 105/40833946, AK-105/40837563, AE-413/30061043, AK-105/40837629, AK- 105/40836387.

44. A molecule according to anyone of claims 39 to 42, or functional equivalent thereof, selected from the group of molecules herein identified as AK-105/40836349, AK- 105/40837635, AA-504/32626008, AK-105/40836340, AK-105/40833746, AK- 105/40693663, AK-105/40833503, AK-105/40837674, AK-105/40836874.

45. A molecule according to anyone of claims 39 to 42, or functional equivalent thereof, selected from the group of molecules herein identified as AK-105/40836349, AK- 105/40837635, AA-504/32626008, AK-105/40836340, AK-105/40833746, AK- 105/40693663. 46. A molecule according to anyone of claims 39 to 42, or functional equivalent thereof, selected from the group of molecules herein identified as AK-105/40836349, AK- 105/40837635, AA-504/32626008, AK-105/40836340.

47. A molecule according to anyone of claims 39 to 42, comprising AK-105/40836349 or functional equivalent thereof.

48. A molecule according to anyone of claims 39 to 42, comprising AK-105/40837635 or functional equivalent thereof.

49. A molecule according to anyone of claims 39 to 42, comprising AA-504/32626008 or functional equivalent thereof.

50. A molecule according to anyone of claims 39 to 42, comprising AK-105/40836340 or functional equivalent thereof.

51. A molecule according to anyone of claims 39 to 50, or functional equivalent thereof, for use in treatment of a subject having or suspected to having a type I cytokine receptor mediated disease.

52. A molecule according to anyone of claims 39 to 50, or functional equivalent thereof, for use in treatment of a subject having or suspected to having a group 1 type I cytokine receptor mediated disease.

53. A molecule according to anyone of claims 39 to 50, or functional equivalent thereof, for use in treatment of a subject having or suspected to having a growth hormone receptor dependent disease.

54. A molecule according to claim 53 wherein said disease comprises acromegaly.

55. A molecule according to claim 53 wherein said disease comprises increased levels of blood-glucose such as diabetes.

56. A molecule according to claim 53 wherein said disease comprises cancer.

57. A molecule according to claim 53 wherein said disease comprises secondary primary cancer.

58. A molecule according to claim 53 wherein said disease comprises metastatic cancer.

59. A molecule according to claim 53 wherein said disease comprises treatment-resistant cancer. 60. A molecule according to claim 53 wherein said disease comprises growth hormone assisted cancer.

61. A molecule according to anyone of claims 56 to 60 wherein said disease comprises chemotherapy-resistant cancer.

62. A pharmaceutical formulation comprising a molecule according to anyone of claims 39 to 61.

63. A pharmaceutical composition according to claim 62 and a pharmaceutically acceptable excipient.

64. A method of treatment of a subject having or suspected of having a type I cytokine receptor mediated disease comprising administering to said subject a molecule according to anyone of claims 39 to 61 or a pharmaceutical formulation according to claim 62 or 63.

65. A method of treatment of a subject having or suspected to having a group 1 type I cytokine receptor mediated disease comprising administering to said subject a molecule according to anyone of claims 39 to 61 or a pharmaceutical formulation according to claim 62 or 63.

66. A method of treatment of a subject having or suspected to having a growth hormone receptor mediated disease comprising administering to said subject a molecule according to anyone of claims 39 to 61 or a pharmaceutical formulation according to claim 62 or 63.

67. A method of treatment according to claim 66 wherein said disease comprises acromegaly.

68. A method of treatment according to claim 66 wherein said disease comprises increased levels of blood-glucose such as diabetes.

69. A method of treatment according to claim 66 wherein said disease comprises cancer.

70. A method of treatment according to claim 66 wherein said disease comprises secondary primary cancer.

71. A method of treatment according to claim 66 wherein said disease comprises treatment- resistant cancer.

72. A method of treatment according to claim 66 wherein said disease comprises growth hormone assisted cancer.

73. A method of treatment according to anyone of claims 69 to 72 wherein said disease comprises chemotherapy-resistant cancer. 74. A compound according to formula 1, or functional equivalent thereof for use in the treatment or prevention of a disease in a subject,

Formula 1

With

A and E represent carbon atoms with binding between A and E with a single bond or a double bond in E-configuration or a triple bond.

U, V, X, Y and Z represent carbon atoms or one of them represents an unsubstituted N to form a pyridine,

R1 is (lC-3C)-alkyl or 3,3,3-trifluoropropyl,

R2 is methyl and R3 is hydrogen or methyl, or R2 and R3 form together with the carbon to which they are attached a cyclopropane, or R2 and R3 form together with the carbon to which they are attached a cyclopentane,

R4, R7, R8, R9and R10 are hydrogen, halogen, trifluoromethyl, methoxy, trifluoromethoxy, morpholine or methylsulfonyl,

R5 and R6 are hydrogen, ethyl or R5 or R6 are pyridin-2-ylmethyl, or a pharmaceutically acceptable salt thereof,

75. A compound according to claim 74 wherein R5 is pyridin-2-ylmethyl.

76. A compound according to claim 74 or 75 wherein R1 is methyl,

R2 and R3 form together with the carbon to which they are attached a cyclopropane and

R4 is chloro or trifluoromethyl. R1 is methyl,

R2 and R3 form together with the carbon to which they are attached a cyclopropane and

R4 is chloro or trifluoromethyl.

77. A compound according to anyone of claims 74 to 76 wherein said disease is a growth hormone dependent disease

78. A compound according to formula 2, or functional equivalent thereof for use in the treatment or prevention of a disease in a subject,

Formula 2

With

R1 is hydrogen or (lC-3C)alkyl or (lC-4C)alkyloxy; and wherein

R2 is phenyl optionally substituted with one or more groups selected from (lC-3C)alkyl, (1C- 3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R2 is N-methyl-benzyl-amine optionally substituted with one or more groups selected from (1C- 3C)alkyl, (lC-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or

R2 is benzyl-amine optionally substituted with one or more groups selected from (lC-3C)alkyl, (1C-

3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or

R2 is anilinomethyl optionally substituted with one or more groups selected from (lC-3C)alkyl,

(lC-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or

R2 is benzenesulfinyl optionally substituted with one or more groups selected from (lC-3C)alkyl,

(lC-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or

R2 is benzenesulfide optionally substituted with one or more groups selected from (lC-3C)alkyl,

(lC-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or or a pharmaceutically acceptable salt thereof.

79. A compound according to claim 78 wherein R1 and R2 together form 2,2-dimethyl-3H- furo[2,3-f]

80. A compound according to claim 78 wherein R1 and R2 together form 7- (cyclopropylmethyl)pyrrolo[3,2-f]

81. A compound according to anyone of claims 78 to 80 wherein said disease is a growth hormone dependent disease.

82. A method of treatment of a subject having or suspected to having a growth hormone receptor mediated disease comprising administering to said subject a compound according to anyone of claims 74 to 81

83. A method of treatment according to claim 82 wherein said disease comprises acromegaly.

84. A method of treatment according to claim 82 wherein said disease comprises increased levels of blood-glucose such as diabetes.

85. A method of treatment according to claim 82 wherein said disease comprises cancer.

86. A method of treatment according to claim 82 wherein said disease comprises secondary primary cancer.

87. A method of treatment according to claim 82 wherein said disease comprises treatment- resistant cancer.

88. A method of treatment according to claim 82 wherein said disease comprises growth hormone assisted cancer.

89. A method of treatment according to anyone of claims 85 to 88 wherein said disease comprises chemotherapy-resistant cancer.

Description:
Selective polypeptide synthesis stalling assay and compound FIELD The invention relates to the field of medicinal chemistry and candidate-drug-testing and development. BACKGROUND Cytokines are grouped into superfamilies based on shared structural elements of the receptors they bind (Schwartz et al., Nat Rev Rheumatol.2016 Jan;12(1):25-36.). The major cytokine families are: the type I/II cytokines, the TNF family, the IL-1 family, the IL-17 cytokines, the stem cell factor/receptor tyrosine kinase (STF/RTK) cytokines, the transforming growth factor (TGF)-β family cytokines, and chemokines. Cytokines are major drivers of autoimmunity, and biologic agents targeting cytokines have revolutionized the treatment of disease. Despite the effectiveness of these drugs, they do not induce complete remission in all patients, prompting the development of alternative strategies — including targeting of intracellular signal transduction pathways downstream of cytokines. Many cytokines that bind type I and type II cytokine receptors are critical regulators of immune-mediated diseases and employ the Janus kinase (JAK) and signal transducer and activator of transcription (STAT) pathway to exert their effect. When a type I/II cytokine binds to its cognate receptor, the receptor becomes activated. JAKs autophosphorylate and transphosphorylate, causing STATs to be recruited to the activated receptor where they, in turn, are phosphorylated. The STATs then dimerize and translocate to the nucleus where they initiate transcription. Pharmacological inhibition of JAKs blocks the actions of type I/II cytokines, and within the past 3 years therapeutic JAK inhibitors, or Jakinibs, have become available. Jakinibs have proven effective for the treatment of rheumatoid arthritis and other inflammatory diseases. Adverse effects of these agents are largely related to their mode of action and include infections and hyperlipidaemia. Jakinibs are currently being investigated for several new indications, and second-generation selective Jakinibs are being developed and tested. Type I and type II cytokines (sometimes also terms as class 1 and 2 cytokines are used) are molecules produced by a cell that generally act within said cell (autocrine) or on another cell (paracrine or endocrine). Paracrine signaling acts on nearby cells, endocrine signaling uses the circulatory system to transport ligands, and autocrine signaling acts on the signaling cell itself. Type I and type II cytokine receptors are grouped together as they associate as homo- or heterodimers including common polypeptide chains and signal via JAK/STAT pathways. These receptors present a high level of complexity in cytokine–receptor interactions, that results from the fact that one particular cytokine can bind to different receptor complexes, and that one particular receptor complex can bind several cytokines. Typical other cytokine receptors such as IG-type, TNFR-type and chemokine type do not show such dimerization depended JAK/STAT signaling. The term cytokine applies not only to molecules with immunologic functions but to hormones as well. Therewith, growth hormone, prolactin, and leptin are classified as type I cytokines. These and other type I cytokines, (including interleukin 2–9, 11–13, and 15; erythropoietin; thrombopoietin; and granulocyte-colony–stimulating factor) are characterized by a four α-helical bundle structure and signal via type I cytokine receptors. Type II cytokines include the interferons and IL-10 and seem not to involve hormones. Prolactin, leptin, and growth hormone belong to a subclass of type I cytokines having a long cytosolic tail. Other type I cytokines, including IL-2 and stem cell factor, have a short cytosolic tail, generally less than 60 amino acids long. Such a long cytosolic tail (long intracellular domain) enables activation of intracellular signaling. The short forms have similar extracellular structure but for the intracellular domain (short-cytosolic tail). The role of the short form is still unclear. In activation and signaling, these receptors show a dimerization of the cytosolic tails, some show homomerization, others show heteromerization, where typically short-cytosolic tail receptor molecules heteromerize with a long-cytosolic tail variant to allow signaling. In disease, type I cytokine signaling typically act via a restricted number of Jak-Stat pathways that positively and negatively regulate cell types involved in the initiation, propagation, and resolution of an array of cellular responses. Three major cell types involved in inflammatory responses are for example T cells, neutrophils, and macrophages. Similarly, type I cytokines play critical roles in the regulation of a wide range of functions leading to cellular proliferation, differentiation, and survival, as well as in specialized cellular functions involved in cancer development and progression. The predominant intracellular signaling pathway triggered by type I cytokines is the JAK-signal transducer and activator of transcription (STAT) pathway. Type I cytokine receptors are complexes of single pass transmembrane domain containing proteins. Many cytokine-specific receptor chains contain a cytoplasmic tail region with little known function, but each functional receptor complex always consists of at least two (often precisely two) individual receptor chains with long intracellular regions (cytosolic tails of several hundred amino acids in length) that are the scaffolds upon which signaling is initiated. These unstructured cytoplasmic domains on long cytosolic tails exist to provide sequence-specific docking sites for JAKs and STATs. The JAK-binding regions are known historically as the Box 1 and Box 2 motifs and are membrane proximal whilst the STAT binding motifs are located towards the C-terminus, distal to the membrane. Knockout mice and clinical human studies have provided evidence that JAK-STAT proteins regulate the immune system, as well as maintain immune tolerance and tumour surveillance. Group 1 of type I cytokine receptor chains contains the erythropoietin receptor (EPOR), thrombopoietin receptor (TPOR), prolactin receptor (PRLR), and growth hormone receptor (GHR) chains that each form homodimers in the presence of their respective ligands. Prolactin (PRL), placental lactogen (PL) and growth hormone (GH) receptors are well-studied members of group 1 of the type I cytokine receptor superfamily, and are activated by ligand binding that induces a conformational change that cause the transmembrane domains to transition from a parallel interaction to a left-handed crossover interaction. There is currently no evidence for a specific PL- receptor, and PL is thought to act through either the GHR, the PRLR, or both. PRL, PL and GH receptors are typically non-kinase receptors of the type I cytokine receptor family whose activation of signaling pathways requires participation of receptor-associated kinases, such as Janus kinases and Src kinases. Signal transduction by these receptors mainly involves the JAK/Stat pathway. In addition to the GHR, GH can bind and activate the PRLR, and the GHR can form heterodimers with PRLR. These structural changes cause a separation of the long intracellular domains to the Box1 and Box2 motifs and associated JAK2 molecules. On the basis of this mechanism of activation, antagonists have been developed that block the receptors in an inactive conformation. Furthermore, GHR cross-talks and/or forms complexes with several other type I cytokine (growth factor and hormone) receptors, such as EphA4, EGFR, and IGF1R, which enhances the stimulation of downstream signaling pathways. Growth hormone (GH), placental lactogen (PL), and prolactin (PRL) regulate an extensive variety of important physiological functions. Human growth hormone (GH) is a peptide hormone that is secreted from the anterior pituitary. The history of growth hormone (GH) has been described by Buchman, Bell and Kopchick (Buchman et al., 2018). The state-of-the-art of the GH-field has been reviewed in Basu et al., 2018; Dehkhoda et al., 2018, and Ranke and Wit, 2018. Growth hormone has a central function of regulating postnatal growth and metabolism and exhibits pleiotropic effects on various human tissues. Chronic hypersecretion of GH into the circulation, usually from a GH-secreting pituitary adenoma, is classically associated with acromegaly, a debilitating disease characterized by excessive skeletal growth, soft tissue enlargement, insulin resistance, and cardiovascular and gastrointestinal morbidities. The pea- sized pituitary gland, located at the sella turcica, is responsible for secreting a multitude of hormones, including GH or somatotropin, the action of which is controlled by a complex feedback mechanism. The most common cause of acromegaly is the presence of a benign tumour or adenoma originating from pituitary somatotroph cells and secreting excess GH. This excessive secretion of GH leads to a persistent elevation of insulin-like growth factor-1 (IGF-1), which is produced by the liver, kidney, pituitary gland, muscle, and gastrointestinal tract; with liver being the primary source. IGF-1 facilitates the growth-promoting effects of GH. Increased GH levels have also been implicated in cancer and diabetes. There is only one GHR-based therapy using the GH- antagonist, namely Pegvisomant. It is used in the treatment of acromegaly and in cancer cell lines. A major disadvantage of Pegvisomant is that it only acts endocrine, while the cancer driver activity of GH occurs in autocrine mode. Pegvisomant, a GH analogue, is the only clinically used antagonist of the GH receptor (GHR). However, other antagonists are in clinical trials or preclinical development. Once released into the circulation, GH binds and activates the cell-surface GHR, as well as the related prolactin receptor in target tissues such as liver, muscle, bone, and adipose tissue. It is the key regulator of insulin-like growth factor 1 (IGF1), which is secreted from target tissues, particularly the liver. Growth hormone (GH) and insulin-like growth factor-1 (IGF-1, together identified as the GH/IGF-1 axis) play a central role in development, differentiation, growth, and metabolism in mammals. Increased serum GH and IGF1 produce feedback loops that lead to inhibition of GHRH, release of somatostatin, and consequently inhibition of GH secretion from the pituitary. Whereas the endocrine system is the main secretory pathway, GH is also expressed in many extra pituitary tissues in which it has autocrine and paracrine effects. For example, in breast development, growth hormone (GH) has important roles in ductal elongation and the differentiation of ductal epithelia into terminal end buds, whereas prolactin is necessary for normal lobular epithelial cell proliferation and secretory function. Attention has focused on prolactin as a potential breast tumour promoter because it was early shown to act in this way with rat mammary carcinomas, and overexpression of prolactin in mammary tissue induces tumours in mice. The situation in human breast cancer is not so clear, because both prolactin and GH are able to activate the prolactin receptor in humans, and the homologous GH and prolactin receptors activate remarkably similar signaling pathways. These class 1 cytokine receptors activate not only the Janus kinase 2 and signal transducers and activators of transcription JAK2/STAT5 and JAK2/STAT3 pathways, but also Src family kinases, leading to phospholipase C γ, extracellular signal-regulated kinase (ERK), and phosphatidylinositol 3-kinase (PI 3-kinase) pathway activation. These pathways are capable of upregulating tumorous characteristics such as cell proliferation, survival, and motility, and ERK activation has been implicated in metastasis. Overall, GH receptor (GHR) acts as a modulator of cellular metabolism, whose loss is not lethal, but results in sub-optimal health with short stature, decreased bone mineral density, decreased muscle strength, thin skin and hair, increased adiposity, and hepatic steatosis. Interestingly, people without GHR signaling live normal lives, but are highly resistant to cancer and diabetes type 2. In a well-studied Ecuadorian cohort of 100 persons, no cancer deaths were observed (Guevara-Aguirre et al., 2011) in the study period. They also performed significantly better in memory tasks and had lower cognitive impairment compared to their unaffected relatives (Nashiro et al., 2017). In adults, hypersecretion of GH causes acromegaly, and strategies that aim to block the release of GH (e.g. with GH-antagonist Pegvisomant) or that aim to inhibit GHR activation are the primary forms of medical therapy for this disease. Typically, inhibitors of GHR activity include an antagonist-GHBP fusion protein; a GH ligand or a GHR antagonist fused to GH-binding protein (GHBP), the extracellular domain of the GHR, which is proteolytically cleaved from the receptor and exists in the circulation. Also, anti-GHR antibodies have been reported, which inhibit the activation of GHR and block downstream signaling. Another approach is atesidorsen (ATL1103), an antisense oligonucleotide (ASO), that binds and induces the degradation of GHR mRNA. Small molecule compounds may also have applications; however, there are currently limited reports in this area. Overproduction of GH has also been linked to cancer and diabetes. GH abuse has been widespread among athletes for more than 20 years, with consequences such as oedema, carpal tunnel syndrome, arthralgias, myalgias and glucose intolerance with susceptibility for development of cancer and diabetes. However, studies to investigate the therapeutic potential of GHR antagonism in these diseases have been limited, most likely due to difficulty in accessing therapeutic tools to study the pharmacology of the receptor in vivo (Lu et al., 2019). IGF-1 is a single-chain polypeptide growth factor that is related to insulin and IGF-2. IGF-1 stimulates cell growth, proliferation, and differentiation, and is essential for normal organismal growth and development. IGF-1 binds to the insulin-like growth factor 1 receptor (IGF-1R), which is a tyrosine kinase receptor. IGF-1 has a higher binding affinity than IGF-2 for IGF-1R. IGF-1R initiates a cascade of downstream signal transduction pathways known to be involved in cell growth, proliferation, and cancer, including Ras/Raf/ERK and PI3K/Akt/mTOR. The majority of IGF- 1 found in the circulation is produced by the liver, functioning as an endocrine hormone. IGF-1 is also produced in other organs where autocrine or paracrine mechanisms have a role. Ample evidence indicates that IGF-1 and IGF-1R are important for growth and survival of cancer cells. The expression of the IGF-1 gene is primarily regulated by growth hormone (GH), and to a smaller extent by various other hormones. Cancer survival rates are increasing, and as a result, more cancer survivors are exposed to the risk of developing a second primary cancer (SPC). One of the underlying mechanisms for this risk are increased levels in insulin-like growth factor-1 (IGF-1). Current epidemiological evidence and identifies IGF-1 to play a distinct role in the development of SPCs. IGF-1 is known to promote cancer development by inhibiting apoptosis and stimulating cell proliferation. Epidemiological studies have reported a positive association between circulating IGF-1 levels and various primary cancers, such as breast, colorectal, and prostate cancer. Treatment resistance presents a significant hurdle for successful cancer treatment, especially in the metastatic setting where it accounts for 90% of therapy failure [Longley and Johnston., Molecular mechanisms of drug resistance, Pathol.2005 Jan;205(2):275-92.]. Numerous mechanisms can lead to the development of chemoresistance, among which transporter- mediated drug efflux is one of the most thoroughly validated. ATP-binding cassette (ABC) transporters utilize ATP to efflux various compounds across cellular membranes, including a wide range of anti-cancer drugs with different structures and properties. A number of ABC transporters are strongly implicated in chemoresistance of numerous solid tumours, including breast cancer. In particular, multidrug-resistant protein-1 (ABCC1/MRP1), breast cancer resistance protein (ABCG2/BCRP) and multidrug-resistant protein-8 (ABCC11/MRP8) were expressed significantly more, and more frequently in so-called triple-negative breast cancer cells (TNBC) compared to other breast cancer subtypes. The growth hormone receptor dependent GH/IGF-1 axis is also inducing chemoresistance in human melanoma by driving MITF-regulated and ABC-transporter- mediated drug clearance pathways. Moreover, as reviewed by Basu and Kopchick (The effects of growth hormone on therapy resistance in cancer. Cancer Drug Resist.2019 Summer’2:827-84), numerous reports demonstrate growth hormone assisted or growth hormone receptor dependent cancer development, in particular in development of therapeutic resistance in human cancers and provide evidence of the anti-apoptotic effect of growth hormone receptor dependent GH/IGF-1 axis activity in enhancing resistance of cancer against common cancer treatment, such as enhancing resistance to chemotherapy or radiation therapy. Basu and Kopchick (see their Table 1) provide an extensive list of reports demonstrating growth hormone in development of therapeutic resistance in human cancers, e.g. as diagnosed in patients with therapy resistance due to or mediated by for example deregulated apoptosis, epithelial-to-mesenchymal transition (EMT), drug efflux via ABC- transporters, stemness (cancer stem cell) and radiation resistance. Studies have reported that chemotherapy, induced by apoptosis via JNK expression and phosphorylation, was blocked by GH - an effect reversed by GHR-antagonist, Pegvisomant-, in triple-negative breast cancer (TNBC) cells. Their studies also found that in TNBC cells, GH did induce drug-resistance independent of IGF1, by directly inducing c-fos and suppressing apoptosis. In a recent study, GH was found to confer chemoresistance from doxorubicin, paclitaxel, and cisplatin in human endometrial adenocarcinoma. In human endometrial cancer, GH was found to suppress caspase 3/7 activation and appeared to function differentially either through the ERK1/2 or PKC pathways depending upon the drug or the cell line; again Pegvisomant was found to reverse the effects in cells provided with an external source of GH. Bogazzi had proposed another mechanism of the anti- apoptotic effects of GH where it blocks the expression of pro-apoptotic PPARγ and Bax in colon cancer cells. This survival advantage of tumours bestowed upon by GH to evade the DNA damaging effects of therapy and avoid apoptosis, were also reported in pancreatic cancer, and breast cancer. Arumugam et al., (Experimental & Molecular Medicine (2019) 51:2 ) found growth hormone receptor (GHR) to play a vital role in breast cancer chemoresistance and metastasis and determined that GHR is potential therapeutic target for oestrogen - receptor negative (ER−ve) breast cancer, which are highly chemoresistant and metastatic. Furthermore, activation or overexpression of GHR increased chemoresistance and metastasis. Among others, malignant breast cancer cell lines MDAMB231 and MDAMB468 were used by Arumugam et al., to demonstrate mechanisms of GHR-dependent or -assisted chemoresistance, and in particular growth hormone receptor dependent or assisted GH-IGF-1 axis induced conditions of chemoresistance. Arumugam et al. conclude that inhibition of GHR signaling sensitized breast cancer cells to chemotherapy but do not identify useful GHR-signaling inhibitors. Similarly, melanoma remains one of the most therapy-resistant forms of human cancer despite recent introductions of highly efficacious targeted therapies. The intrinsic therapy resistance of human melanoma is largely due to abundant expression of a repertoire of xenobiotic efflux pumps of the ATP-binding cassette (ABC) transporter family. Basu et al., (HORM CANC DOI 10.1007/s12672-017-0292-7) report that GH action is also a key mediator of chemotherapeutic (cisplatin, doxorubicin, oridonin, paclitaxel or vemurafenib mediated)) resistance, and in particular growth hormone receptor dependent or assisted GH-IGF-1 axis induced resistance in human melanoma cells. GHR-knockdown was found to lead to significantly higher drug retention and sensitizes human melanoma cells to low doses of chemotherapy. Among others, human melanoma cell lines SKMEL28, SKMEL5 and MDAMB435 were used by Basu et al., to demonstrate mechanisms of GHR-dependent or -assisted chemoresistance. Basu et al. conclude that combination of GHR inhibition and chemotherapy can not only markedly improve the efficacy of available anti-melanoma drugs but may also assist the development of novel chemotherapeutic compounds, but do not identify useful GHR-inhibitors. Yi et al., (Korean J Physiol Pharmacol Vol 16: 11-16, February 2012) show 5-fluoracil and methotrexate resistance (with increased IL-6-receptor expression) in non-small cell lung cancer (NSCLC) cell lines, among which NCI 460, and Morales et al., (Oncogene (2005) 24, 6842–6847) show methotrexate resistance in colon cancer cell lines HCT116 and HT-29 and conclude results indicate that multiple mechanisms operate in MTX resistance in lung and colon cancer cells, and that both the capacity to develop resistance and the mechanism(s) involved are associated with the genetic features of the on tumour. Neither Yi nor Morales provide methods to overcome said resistance. There appears to be a consensus over the anti-apoptotic effects of GH, which is harnessed by proliferative tumour cells, while the details of molecular events converging to the net effect of escaping cell death are overlapping and continue to emerge. To put these findings in cells to practical use in treatment` of cancer patients, however, is -as yet- not possible, Pegvisomant not capable of antagonizing autocrine signaling of the cancers in the individuals involved. There is a clear need in the field of treatment of cancer-resistance to provide compounds that can target autocrine GHR-mediated signaling. Currently, the cancer field is equipped with multiple methods of detection as well as multiple modalities of therapy using surgery, radiation, chemotherapy, targeted-therapy, immunotherapy and combinations of these - leading to a 26% drop in cancer death rates in United States since 1991. However, there is still a “standing order” to decipher and overcome the hurdle of therapy refractoriness in cancer, a multi-factorial process with diverse underlying mechanisms (Peters; Cancer Drug Resist 2018;1:1–5.). In conclusion, a substantial body of evidence supports a role for the growth hormone receptor dependent GH-IGF-1 axis in cancer incidence, recurrence, metastasis, resistance to treatment, and fatal progression. This includes epidemiological evidence relating elevated plasma IGF-1 to cancer incidence as well as a lack of certain cancers in GH/IGF-1 deficiency. While GH receptor expression is elevated in many cancers, autocrine GH is present in several types, and overexpression of autocrine GH can induce further cell transformation. While the mechanism of autocrine action is not clear, it does involve both STAT5 and STAT3 activation, and probably nuclear translocation of the GH receptor. Development of a more potent inhibitor of growth hormone receptor dependent or assisted GH-IGF-1 axis induced cellular activity than presently is available is in particular considered warranted for cancer therapy. INVENTION The invention relates collectively to type I and type II cytokine receptors that herein are grouped together as they associate as homo- or heterodimers including common chains and signal via JAK/STAT pathways. The invention puts growth hormone dependent or assisted conditions and in particular growth hormone receptor dependent or assisted GH-IGF-1 axis induced conditions at rest. The invention provides a molecule capable of selectively stalling polypeptide synthesis of a growth hormone receptor, and a functional equivalent molecule capable of selectively stalling polypeptide synthesis of a growth hormone receptor. Stalling herein relates to hampering, inhibiting, or preventing polypeptide synthesis, preferably at the ribosomal level of translation. Such a molecule is particularly provided for use in treatment of a subject having or suspected to having a growth hormone receptor dependent disease, in particular growth hormone receptor dependent GH-IGF-1 axis induced or assisted disease. Such a molecule as provided herein is particularly useful wherein said hormone receptor dependent GH-IGF-1 axis induced or assisted disease comprises acromegaly and/or said hormone receptor dependent GH-IGF-1 axis induced or assisted disease comprises increased levels of blood-glucose such as diabetes. Such a molecule as provided herein is also particularly useful wherein said hormone receptor dependent GH-IGF-1 axis induced or assisted disease comprises cancer, such as wherein said hormone receptor dependent GH-IGF-1 axis induced or assisted disease comprises secondary primary cancer and/or wherein said hormone receptor dependent GH-IGF-1 axis induced or assisted disease comprises metastatic cancer, or wherein said hormone receptor dependent GH-IGF-1 axis induced or assisted disease comprises treatment-resistant cancer, or said hormone receptor dependent GH- IGF-1 axis induced or assisted disease comprises growth hormone assisted cancer and/or wherein hormone receptor dependent GH-IGF-1 axis induced or assisted said disease comprises chemotherapy-resistant cancer. The invention also provides a method of treatment of a subject having or suspected to having a growth hormone receptor mediated disease comprising administering to said subject a molecule according to the invention. Such a method of treatment is particularly provided for use in treatment of a subject having or suspected to having a growth hormone receptor dependent disease, in particular growth hormone receptor dependent GH-IGF-1 axis induced or assisted disease. Such a method of treatment as provided herein is particularly useful wherein said hormone receptor dependent GH- IGF-1 axis induced or assisted disease comprises acromegaly and/or said hormone receptor dependent GH-IGF-1 axis induced or assisted disease comprises increased levels of blood-glucose such as diabetes. Such a method of treatment as provided herein is also particularly useful wherein said hormone receptor dependent GH-IGF-1 axis induced or assisted disease comprises cancer, such as wherein said hormone receptor dependent GH-IGF-1 axis induced or assisted disease comprises secondary primary cancer and/or wherein said hormone receptor dependent GH-IGF-1 axis induced or assisted disease comprises metastatic cancer, or wherein said hormone receptor dependent GH-IGF-1 axis induced or assisted disease comprises treatment-resistant cancer, or said hormone receptor dependent GH-IGF-1 axis induced or assisted disease comprises growth hormone assisted cancer and/or wherein hormone receptor dependent GH-IGF-1 axis induced or assisted said disease comprises chemotherapy-resistant cancer. In a first preferred embodiment, aiming to obtain detect, test or identify a functional equivalent molecule according to the invention, the invention provides a selective polypeptide synthesis stalling assay and ribonucleic acid for use in candidate drug testing and provides a selective polypeptide synthesis stalling compound, in particular a small molecule identifiable in a selective polypeptide synthesis stalling assay as provided herein. The invention also provides a molecule useful in a method of treating type I or type II cytokine receptor mediated disease and a method of treatment a cytokine receptor mediated disease comprising treating a mammal, preferably a human with a molecule identifiable as selective polypeptide synthesis stalling molecule, preferably identifiable with an assay as provided herein. In a preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system, preferably for use in candidate drug testing, said assay provided with a ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I/II cytokine receptor, preferably a cytosolic tail derived from a receptor of the group of type I/II receptors having a long cytosolic tail, and provided with a drug-candidate test molecule. Polypeptide herein comprises protein as well as peptide. In a preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system according to the invention wherein said test molecule is selected from the group of molecules having general formula 1, formula 1

or having general formula 2 formula 2. In a further preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system according to the invention wherein said test molecule is selected from the group of molecules having general formula 1. The inventors herein identity various useful drug candidate molecules that selectively inhibit protein expression, said selectivity herein characterized by the finding that said expression of said cytosolic tail is inhibited or hampered by said test molecule when a C-terminal part of said cytosolic tail carries a near C- terminal polypeptide drug target whereas said expression of said cytosolic tail is not inhibited or hampered by said test molecule when said polypeptide drug target sequence is at least functionally deleted or C-terminally truncated. Preferably the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system according to the invention wherein A and E of said test molecule according to formula 1 represent carbon atoms with binding between A and E with a single bond or a double bond in E-configuration or a triple bond, furthermore wherein U, V, X, Y and Z represent carbon atoms or one of them represents an unsubstituted N to form a pyridine, R1 is (1C-3C)-alkyl or 3,3,3-trifluoropropyl or 2-methoxyethoxy, R2 is methyl and R3 is hydrogen or methyl, or R2 and R3 form together with the carbon to which they are attached a cyclopropane, or R2 and R3 form together with the carbon to which they are attached a cyclopentane, or R2 and R3 form together with the carbon to which they are attached a cyclobutane, R4, R7, R8, R9 and R10 are hydrogen, halogen, trifluoromethyl, methoxy, trifluoromethoxy, morpholine or methylsulfonyl, hydroxy, piperazine R5 and R6 are hydrogen, methyl, ethyl or R5 or R6 are pyridin-2-ylmethyl, or a pharmaceutically acceptable salt thereof. It is in particular preferred that A and E of said test molecule according to formula 1 represent carbon atoms with binding between A and E with a single bond or a double bond. In another preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system according to the invention wherein said test molecule is selected from the group of molecules having general formula 2. Preferably the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system according to the invention wherein R1 of said test molecule according to formula 2 is hydrogen or (1C-3C)alkyl or (1C-4C)alkyloxy; and wherein R2 is phenyl optionally substituted with one or more groups selected from (1C-3C)alkyl, (1C- 3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R2 is N-methyl-benzyl-amine optionally substituted with one or more groups selected from (1C- 3C)alkyl, (1C-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R2 is benzyl-amine optionally substituted with one or more groups selected from (1C-3C)alkyl, (1C-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R2 is anilinomethyl optionally substituted with one or more groups selected from (1C-3C)alkyl, (1C-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R2 is benzenesulfinyl optionally substituted with one or more groups selected from (1C-3C)alkyl, (1C-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R2 is benzenesulfide optionally substituted with one or more groups selected from (1C-3C)alkyl, (1C-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R1 and R2 together form 2,2-dimethyl-3H-furo[2,3-f] or R1 and R2 together form 7-(cyclopropylmethyl)pyrrolo[3,2-f]; In a further preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a cell-free protein-expression system for use in candidate drug testing, said cell-free system provided with a ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I cytokine receptor and provided with a drug-candidate test molecule. Cell-free protein expression is the in vitro synthesis of a protein using translation- compatible extracts of whole cells. In principle, whole cell extracts contain all the macromolecules and components needed for transcription, translation, and even post-translational modification. These components generally include RNA polymerase, regulatory protein factors, transcription factors, ribosomes and tRNA. When supplemented with cofactors, nucleotides and the specific ribonucleic acid or gene template, these extracts can synthesize proteins of interest in a few hours. Several cell-free-expression systems of both prokaryotic and eukaryotic origin are known in the art, the invention preferably provides a polypeptide synthesis stalling assay comprising a cell- free protein-expression system for use in candidate drug testing that is of eukaryotic origin. Several eukaryotic cell-free-expression systems are available, for example a commercially available Hela-cell free expression system may be used. In a further preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a cell-free protein-expression system for use in candidate drug testing, said, preferably cell-free, system provided with a ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I cytokine receptor and provided with a drug- candidate test molecule, wherein said ribonucleic acid also comprises ribonucleic acid encoding a leucine zipper 5’-terminal and 3’-terminal to said ribonucleic acid encoding said C-terminal part. A pcDNA3 vector DNA expressing Fos-zippered GHR cytosolic tails as described in Nespital et al, 2016 was used herein to show that various compounds act on protein translation at the ribosomal level in the cell-free system. Using increasing concentrations reveals that compound BM001 is effectively inhibiting synthesis at concentrations > 0.1 µM. Translation of other proteins nor of C- terminal cytosolic tail wherein the polypeptide drug target was at least functionally deleted or truncated was affected. In a further preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system for use in candidate drug testing, said, preferably cell- free, system provided with a ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I cytokine receptor and provided with a drug- candidate test molecule, wherein said ribonucleic acid also encodes a reporter tag, preferably wherein said reporter tag is selected from the group of polyhistidine (His-tag), Glutathione-S- Transferase (GST-tag), epitope tags such as hemaglutinine-(HA)-tag, 11 amino acid peptide tag called HiBiT fluorescent with LgBiT protein, luciferase, c-myc-tag, strep-tag, VSV-G-tag, V5-tag. Various tag-systems are commercially available. In a further preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system for use in candidate drug testing, said, preferably cell- free, system provided with a ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I cytokine receptor and provided with a drug- candidate test molecule, wherein box-1-mediated JAK binding activity is at least functionally deleted from said cytosolic tail. Such a functional deletion may for example be achieved by replacing a codon or codons encoding a proline with a codon or codons encoding an alanine in said ribonucleic acid segment encoding box-1. In a further preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system for use in candidate drug testing, said, preferably cell- free, system provided with a ribonucleic acid encoding at least a part of a cytosolic tail having >70% homology, preferably >80% homology, preferably >90% homology, more preferably 95% homology with a polypeptide amino acid sequence comprising at least a C-terminal part of a cytosolic tail of a type I/II cytokine receptor and provided with a drug-candidate test molecule. In preferred embodiment, said cytosolic tail is of interleukin-6 receptor subunit beta and said at least C-terminal part comprises amino acid sequence 5, more preferably sequence 6, more preferably sequence 7, more preferably sequence 8, more preferably sequence 3, most preferably sequence 4, or functional equivalents thereof, as listed in example 10. Useful control interleukin-6 receptor subunit beta polypeptide sequences are sequences 9, 10, 11 or 12, or functional equivalents thereof, as listed in example 10. In another preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system for use in candidate drug testing, said, preferably cell- free, system provided with a ribonucleic acid encoding at least a part of a cytosolic tail having >70% homology, preferably >80% homology, preferably >90% homology, more preferably 95% homology with a polypeptide amino acid sequence comprising at least a C-terminal part of a cytosolic tail of a type I/II cytokine receptor and provided with a drug-candidate test molecule. In preferred embodiment, said cytosolic tail is of granulocyte colony-stimulating factor receptor and said at least C-terminal part comprises amino acid sequence 29, more preferably sequence 30, more preferably sequence 31, more preferably sequence 27, most preferably sequence 28, or functional equivalents thereof, as listed in example 10. Useful control granulocyte colony-stimulating factor receptor polypeptide sequences are sequences 32, 33 or 34, or functional equivalents thereof, as listed in example 10. In another preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system for use in candidate drug testing, said, preferably cell- free, system provided with a ribonucleic acid encoding at least a part of a cytosolic tail having >70% homology, preferably >80% homology, preferably >90% homology, more preferably 95% homology with a polypeptide amino acid sequence comprising at least a C-terminal part of a cytosolic tail of a type I/II cytokine receptor and provided with a drug-candidate test molecule. In preferred embodiment, said cytosolic tail is of leukemia inhibitory factor receptor and said at least C-terminal part comprises amino acid sequence 39, more preferably sequence 40, more preferably sequence 41, more preferably sequence 37, most preferably sequence 38, or functional equivalents thereof, as listed in example 10. Useful control leukemia inhibitory factor receptor polypeptide sequences are sequences 42, 43 or 44, or functional equivalents thereof, as listed in example 10. In a further preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system for use in candidate drug testing, said, preferably cell- free, system provided with a ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I cytokine receptor and provided with a drug- candidate test molecule wherein said receptor is a group 1 type I cytokine receptor and wherein said cytosolic tail is of leptin receptor and said at least C-terminal part comprises amino acid sequence 17, more preferably sequence 18, more preferably sequence 19, more preferably sequence 20, more preferably sequence 15, most preferably sequence 16, or functional equivalents thereof, as listed in example 10. Useful control leptin receptor polypeptide sequences are sequences 65, 66, 67 or 68, or functional equivalents thereof, as listed in example 10. In a further preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system for use in candidate drug testing, said, preferably cell- free, system provided with a ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I cytokine receptor and provided with a drug- candidate test molecule wherein said receptor is a group 1 type I cytokine receptor and wherein said cytosolic tail is of erythropoietin receptor and said at least C-terminal part comprises amino acid sequence 73, more preferably sequence 74, more preferably sequence 75, more preferably sequence 71, most preferably sequence 72, or functional equivalents thereof, as listed in example 10. Useful control erythropoietin receptor polypeptide sequences are sequences 76, 77, or 78, or functional equivalents thereof, as listed in example 10. In a further preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system for use in candidate drug testing, said, preferably cell- free, system provided with a ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I cytokine receptor and provided with a drug- candidate test molecule wherein said receptor is a group 1 type I cytokine receptor and wherein said cytosolic tail is of prolactin receptor and said at least C-terminal part comprises amino acid sequence 61, more preferably sequence 62, more preferably sequence 63, more preferably sequence 64, more preferably sequence 59, most preferably sequence 60, or functional equivalents thereof, as listed in example 10. Useful control prolactin receptor polypeptide sequences are sequences 65, 66, 67 or 68, or functional equivalents thereof, as listed in example 10. In a further preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system for use in candidate drug testing, said, preferably cell- free, system provided with a ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I cytokine receptor and provided with a drug- candidate test molecule wherein said receptor is a group 1 type I cytokine receptor and wherein said cytosolic tail is of prolactin receptor and said at least C-terminal part comprises amino acid sequence 61, more preferably sequence 62, more preferably sequence 63, more preferably sequence 64, more preferably sequence 59, most preferably sequence 60, or functional equivalents thereof, as listed in example 10. Useful control prolactin receptor polypeptide sequences are sequences 65, 66, 67 or 68, or functional equivalents thereof, as listed in example 10. In a most preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system for use in candidate drug testing, said, preferably cell- free, system provided with a ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I cytokine receptor and provided with a drug- candidate test molecule wherein said receptor is a group 1 type I cytokine receptor and wherein said cytosolic tail is of growth hormone receptor and said at least C-terminal part comprises amino acid sequence 49, more preferably sequence 50, more preferably sequence 87, more preferably sequence 88, more preferably sequence 89, more preferably sequence 90, more preferably sequence 91, more preferably sequence 92, more preferably sequence 93, more preferably sequence 94, more preferably sequence 95, more preferably sequence 51, more preferably sequence 52, more preferably sequence 47, most preferably sequence 48, or functional equivalents thereof, as listed in example 10. Useful control growth hormone receptor polypeptide sequences are sequences 53, 54, 55 or 56, or functional equivalents thereof, as listed in example 10. In a further preferred embodiment, the invention provides a polypeptide synthesis stalling assay comprising a protein-expression system for use in candidate drug testing, said, preferably cell- free, system provided with a ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I cytokine receptor and provided with a drug- candidate test molecule wherein said ribonucleic acid (herein also called the first ribonucleic acid or the polypeptide drug target ribonucleic acid) encodes a polypeptide provided with a least the last 44 C-terminal amino acids of said cytosolic tail, preferably with at least the last 117 amino acids, more preferably with at least the last 167 amino acids, more preferably with at least the last 251 amino acids of said C-terminal cytosolic tail of said receptor. In a preferred embodiment, the invention provides a polypeptide synthesis stalling assay according to the invention wherein said ribonucleic acid encodes a polypeptide provided with a C-terminal tail part of a growth hormone receptor, said polypeptide at least comprising an amino acid sequence starting after position 594, preferably after position 521, preferably after position 471, more preferably after position 387, as indicated in figure 8. It is preferred that said tail is encoded up to the C-terminal end of the cytosolic tail of GHR. The invention also provides a set of at least two ribonucleic acids, preferably for use as target and control ribonucleic acid in a polypeptide synthesis stalling assay comprising a protein-expression system according to the invention, comprising a first ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I cytokine receptor and a second ribonucleic acid encoding a polypeptide provided with an at least functional deletion or truncation of a C-terminal part of said cytosolic tail of a type I cytokine receptor. In a preferred embodiment, said functional deletion or truncation in said second (control) nucleic acid comprises a least the last 44 C-terminal amino acids of said C-terminal part of said cytosolic tail of said receptor, more preferably the last 117 amino acids, more preferably the last 167 amino acids, more preferably the last 251 amino acids of said C-terminal cytosolic tail of said receptor. Herewith, the invention provides recombinant cytosolic tails with as well as without the polypeptide drug target, for use as target as well as control in a polypeptide synthesis stalling assay according to the invention. Therewith, the invention also provides a type I cytokine receptor polypeptide drug target, with method and means to identify a molecule capable of selectively inhibiting, hampering, or stalling synthesis of said type I cytokine receptor. Such compounds that typically act on the target but not on the control have the desired selective stalling (inhibitory) activity are useful for treating and/or preventing type I cytokine receptor-dependent disease. The invention also provides a molecule (herein also identified as compound) identifiable with said polypeptide synthesis stalling assay according to the invention. In a preferred embodiment, the invention provides a type I cytokine receptor polypeptide drug target, with method and means to identify a compound for treating and/or preventing type I cytokine receptor-dependent disease and compounds identifiable with said method and means, wherein said molecule or compound is selected from general formula 1. Test results according to the invention are provided wherein certain molecules, selected from the group of molecules having general formula 1 shown herewith, were found to inhibit synthesis of a polypeptide comprising at least a C-terminal part or fragment of a cytosolic tail of a type I cytokine receptor. Synthesis of a C-terminally truncated or deleted, but otherwise homologous control polypeptide comprising said C-terminal part of said cytosolic tail provided with said C-terminal truncation or C-terminal deletion, and thus lacking an ultimate C-terminal polypeptide drug target part, synthesis of said cytosolic tail (or fragment) of type I cytokine receptor was not inhibited by said molecule. The invention also provides a molecule (herein also identified as compound) identifiable with said method and/or means according to the invention. In a preferred embodiment, the invention provides a type I cytokine receptor polypeptide drug target, with method and means to identify a compound for treating and/or preventing type I cytokine receptor-dependent disease and compounds identifiable with said method and means, wherein said molecule or compoundis selected from general formula 2.

Test results according to the invention are provided wherein certain molecules, selected from the group of molecules having general formula 2 shown herewith, were found to inhibit synthesis of a polypeptide comprising at least a C-terminal part or fragment of a cytosolic tail of a type I cytokine receptor. Synthesis of a C-terminally truncated or deleted, but otherwise homologous control polypeptide comprising said C-terminal part of said cytosolic tail provided with said C-terminal truncation or C- terminal deletion, and thus lacking an ultimate C-terminal polypeptide drug target part, synthesis of said cytosolic tail (or fragment) of type I cytokine receptor was not inhibited by said molecule. Generally, during eukaryotic transcription and translation, the enzyme RNA polymerase uses ribonucleic acid DNA as a template to produce a ribonucleic acid pre-mRNA transcript. The pre- mRNA is processed to form a mature ribonucleic acid mRNA molecule that subsequently can be translated by ribosomes to synthesize the protein or peptide molecule (polypeptide) encoded by the original template ribonucleic acid DNA. It is provided herein that said C-terminally truncated or deleted part of said cytosolic polypeptide tail comprises at least an essential, but hitherto unrecognized, part of a polypeptide drug target located at or at around, or at least partly including said C-terminal tail fragment. When said polypeptide drug target part is contacted with said molecule, (cell free) translation or synthesis of said polypeptide sufficiently stalls to hamper or inhibit synthesis, and a resulting polypeptide is not detectable. When said cytosolic tail lacks said essential part of said polypeptide drug target, no stalling of synthesis during translation by said molecule is observed, and the resulting peptide is detectable. Thus, stalling is selectively depending on the presence or absence of said C-terminally located polypeptide drug target sequence of said cytosolic tail, and such desired selective polypeptide synthesis stalling of a drug- candidate is preferably identified by testing said drug-candidate molecule in the polypeptide synthesis stalling assay according to the invention on the presence as well absence of said C- terminally located polypeptide drug target sequence of said cytosolic tail. For screening purposes, testing in the presence of said drug target is preferred, for further identification, testing in the presence of a control polypeptide lacking said target is recommended. The invention also provides a set of at least two ribonucleic acids (target and control) according to the invention wherein said first (target) and/or said second (control) ribonucleic acid is or are also provided with at least one ribonucleic acid encoding a leucine zipper 5’-terminal and/or 3’- terminal to said ribonucleic acid. It is preferred that both said first and said second ribonucleic acid are also provided with at least one ribonucleic acid encoding a leucine zipper 5’-terminal to said ribonucleic acid. It is moreover preferred that both said first and said second ribonucleic acid are also provided with at least one ribonucleic acid encoding a leucine zipper 3’-terminal to said ribonucleic acid. In a further preferred embodiment, the invention also provides a set of at least two ribonucleic acids according to the invention wherein said first and/or said second ribonucleic acid is or are also provided with at least one ribonucleic acid encoding a reporter tag, preferably wherein said reporter tag is selected from the group of His-tags or epitope tags such as HA-tags or luciferase- tags. In a further preferred embodiment, the invention also provides a set of at least two ribonucleic acids according to the invention wherein box-1-mediated JAK binding activity of said first and said second ribonucleic acid is at least functionally deleted from said cytosolic tail. It is preferred that said receptor is a group 1 type I cytokine receptor, it is particularly preferred that said receptor is a growth hormone receptor (GRH). It is moreover preferred that said second ribonucleic acid is C-terminally truncated or at least functionally deleted with at around 44 C-terminal amino acids, preferably with at around amino acids, more preferably with at around 167 amino acids, more preferably with at around 251 amino acids. It is particularly preferred that said second ribonucleic acid is encoding a control GHR polypeptide comprising at least a C-terminally truncated part of said C-terminal part of said cytosolic tail, wherein said tail is C-terminally truncated at around position 594, preferably at around position 521, preferably at around position 471, more preferably at around position 387, as indicated in figure 8. The invention also provides a set of at least two ribonucleic acids according to the invention comprising a first and a second ribonucleic acid, respectively, for use in synthesizing two polypeptides as target polypeptide and control polypeptide, respectively, for use in a method for testing a molecule for the capacity to selectively inhibit synthesis of a receptor of the type I cytokine receptor family. Preferably said set of at least two ribonucleic acids according to the invention is used in a polypeptide synthesis assay wherein a drug-candidate test molecule is tested, preferably wherein said molecule is selected from the group of compounds with general formula 1 or formula 2. The inventors set out to investigate if a small molecule can be identified and defined that is capable to control the GH/IGF-1 axis. The invention provides a ribonucleic acid encoding a polypeptide comprising at least a C-terminal part of a cytosolic tail of an cytokine type 1 receptor and provides use and methods of use of said ribonucleic acid in identifying a molecule capable of inhibiting synthesis of said polypeptide. The inventors screened close to 39000 small molecule compounds (the 39K tested compounds group) and detected various small molecules that inhibited signaling via a cytokine type 1 receptor and its downstream JAK/Stat pathway. Various of said compounds tested were found to belong to the group of compounds having a structure according to formula 1 or formula 2 as shown herein. Surprisingly, however, when testing a ribonucleic acid encoding a polypeptide comprising at least a C-terminal part of a cytosolic tail of an cytokine type 1 receptor in identifying a molecule capable of inhibiting synthesis of said polypeptide, the inventors detected specific and selective inhibitory activity of several of said 39K tested compounds, and selected analogues thereof, that appeared independent of DHFR-inhibitory activity or of JAK/STAT- or other protein kinase mediated signaling that may be attributed to some of the molecules selected from the group of compounds having a structure according to formula 1 or formula 2 as shown herein. In general, DHFR-inhibitors broadly affect all cell divisions, lacking specific anti-cancer activity as a whole. Also, Jak inhibitors, or other protein kinase inhibitors, are less useful as cancer drugs since these broadly inhibit all (cytokine and other) receptors dependent on protein kinase activity and negatively affect for example immunity and blood cell formation. The invention, in one embodiment, thus provides a means and methods to detect a compound with hitherto unknown activities independent of DHFR- or JAK/STAT or protein kinase inhibitory activity. Such a compound as provided therewith according to the invention may be advantageously be used as a novel therapeutics in novel methods of treatment at large, especially since such compound as provided herein act on type I cytokine receptor activity both from outside (endocrine/paracrine) as from inside (autocrine) at the same time. In particular, such a compound with a polypeptide drug target residing in the growth hormone receptor as provided therewith according to the invention may be advantageously be used as a therapeutics in methods of treatment in cancer therapy at large, especially since an compound as provided herein acts on GHR synthesis both from outside (endocrine/paracrine) as from inside (autocrine) at the same time. Moreover, a polypeptide drug GHR targeting compound as provided according to the invention may particularly be useful for therapy in patients with malignancies, hematological or otherwise, that show resistance against DHFR-inhibitor and/or JAK2 inhibitor therapy, such as resistance to methotrexate. In particular, a compound as provided according to the invention is typically especially useful in treatment of second primary cancer (SPC) or tumors that may arise. In a particular preferred embodiment, the invention provides a compound capable of selectively inhibiting GHR synthesis for use in SPC treatment of a cancer patient, or subject deemed in need of such treatment, preferably wherein said SPC is, or is considered to be, caused by increased activity of the growth hormone receptor dependent GH-IGF-1 axis. Also, the invention provides a method of treatment of a cancer patient resistant to cancer treatment, such as resistant to chemotherapy or radiotherapy, or a subject deemed in need of such treatment, in particular wherein said resistance includes resistance against DHFR-inhibition or JAK/STAT inhibition, such as often is observed with commonly used DHFR-inhibitors of JAK-inhibitors, such as for example resistance to methotrexate or other DHFR-inhibitors of JAK-inhibitors having a structure according to formula 1 or formula 2 as shown herein. Various compounds in the tested 39K group have earlier been demonstrated inhibitors of dihydrofolate reductase (DHFR). The synthesis of folates in both eukaryotic and prokaryotic cells is strictly dependent on the activities of two enzymes: DHFR and dihydrofolate synthase (DHFS), whose inhibition leads to cell death. From a medicinal chemistry perspective, the ubiquitous enzyme DHFR is of particular interest since it is essential for folate metabolism and purine and thymidylate synthesis. Poor DHFR activity causes tetrahydrofolate deficiency and cell death (Blount et al., Proc Natl Acad Sci U S A.1997 Apr 1;94(7):3290-5.). Dihydrofolate reductase inhibitors are an important class of drugs, as evidenced by their use as antibacterial, antimalarial, antifungal, and anticancer agents. These compounds broadly inhibit DHFR thus reducing the level of tetrahydrofolates required for the synthesis of pyrimidine and purines used in ribonucleic acid transcription. Consequently, overall RNA and DNA transcriptional activities broadly stop and treated cells may not have sufficient and appropriate ribonucleic acid to continue with translation. As a result, such cells typically suffer from lack of overall polypeptide production and availability, show faltered cell division, and in the end may die. For example, trimethoprim, aminopterin and methotrexate represent early antibacterial or chemotherapy agents that have been attributed as competitive inhibitors of DHFR. Methotrexate is a cancer chemotherapeutic agent, whilst trimethoprim is an antibiotic. The reason for this disparity, despite both being dihydrofolate reductase inhibitors, is that methotrexate has affinity for mammalian dihydrofolate reductase, while trimethoprim has affinity for the bacterial enzyme. DHFR inhibitors are among the most used classes of anticancer agents to stop RNA and DNA transcription, and finding novel agents with a promising pharmacological profile still remains one of the major challenges for medicinal chemists, as testified by the literature trend of the last 20 years wherein more and more variants of DHFR inhibitors surface. In particular such interest is sparked due to increasingly found resistance of several cancers against DHFR-inhibition, as drug resistance is often a limiting factor in successful chemotherapy. Methotrexate resistance is a specific example of said resistance. Various compounds in the tested 39K group have been also been demonstrated to act as inhibitors of the JAK/STAT pathway. For example, ruxolitinib, and also methotrexate (indeed independent from methotrexate’s DHFR-inhibitory activity) has been attributed as inhibitor of JAK/STAT pathways. Ruxolitinib is an orally bioavailable Janus-associated kinase (JAK) inhibitor with potential antineoplastic and immunomodulating activities. Ruxolitinib binds to and inhibits protein tyrosine kinases JAK 1 and 2. Methotrexate activity independently of dihydrofolate reductase (DHFR) is comparable to the activity of ruxolitinib; methotrexate suppresses human JAK/STAT as well. The JAK/STAT pathway plays an important role in cytokine type I receptor-mediated signal transduction via specific binding of JAK to the BOX 1 element in the cytosolic tail of said receptors and subsequent activation of downstream signal transducers and activators of transcription (STAT), phosphatidylinositol 3-kinase (PI3K), and mitogen-activated protein kinase (MAPK) pathways. However, recent reports have identified many cancer patients exhibiting resistance to JAK2 inhibitor therapy. Furthermore, JAK2 inhibitor therapy is not cancer specific, considering that JAK2 is a major signaling actor for many cytokine receptors involved Again, methotrexate resistance is a specific example of said resistance. WO2013070620 is concerned with compounds that inhibit dihydrofolate reductase (DHFR) and methods of using the compounds for treating pathogenic infections and neoplasia’s. DHFR as part of its molecular mechanism reduces dihydrofolate to tetrahydrofolate. Thus, the inhibition of DHFR deprives the cell of tetrahydrofolate, without which the cell cannot produce 5,10- methylenetetrahydrofolate. Although compounds provided in WO2013070620 may or may not be useful as anti-cancer, or anti-neoplastic, agents, there is no indication of GH or IGF-1 in WO2013070620, nor mention of compounds capable of targeting a cytosolic tail of a type I cytokine receptor and/or capable of targeting the IGH/IGF-1 axis, nor mention a compound capable of selectively inhibiting polypeptide synthesis of a cytosolic tail of a type I cytokine receptor or selectively inhibit paracrine signaling of a type I cytokine. WO2009025919 is concerned with compounds that inhibit dihydrofolate reductase (DHFR) and methods of using the compounds for treating pathogenic infections and neoplasia’s. The compounds provided herein are antifungal and antibacterial agents in vitro using cultures of organisms (B. subtilis, B. cereus, C. albicans, C. glabrata and C neoformans). Compounds herein may or may not be potent against the mammalian DHFR enzyme and may or may not be useful as anti- cancer therapeutics. Although most of the antipathogenic inhibitors described in WO2009025919 are nontoxic for mammalian cells, at least one of the newly developed inhibitors displays broad mammalian cell toxicity and is, therefore, considered useful as an anticancer agent. In WO2009025919 there is no indication of GH nor IGF-1, nor mention of compounds capable of targeting a cytosolic tail of a type I cytokine receptor and/or capable of targeting the IGH/IGF-1 axis, nor mention a compound capable of selectively inhibiting polypeptide synthesis of a cytosolic tail of a type I cytokine receptor or selectively inhibit paracrine signaling of a type I cytokine. Algul et al, J Mol Graph Model.2011 February; 29(5): 608–613, is concerned with hydrofolate reductase (DHFR) that has been a well-recognized target for the development of therapeutics for human cancers for several decades. It furthermore shows that classical inhibitors of DHFR use an active transport mechanism to gain access to the cell; disabling this mechanism creates a pathway for resistance. In response, recent research focuses on nonclassical lipid soluble DHFR inhibitors that are designed to passively diffuse through the membrane. Algul et al., display a series of propargyl-linked antifolates investigated as potential nonclassical human DHFR inhibitors. Algul et al. provide no indication of GH nor IGF-1, nor mention of compounds capable of targeting a cytosolic tail of a type I cytokine receptor and/or capable of targeting the IGH/IGF-1 axis, nor mention a compound capable of selectively inhibiting polypeptide synthesis of a cytosolic tail of a type I cytokine receptor or selectively inhibit paracrine signaling of a type I cytokine. Viswanathan et al, PLoS ONE 7(2): e29434. doi: 10.1371/journal.pone.0029434 is concerned with DHFR as a critical enzyme in the recycling of folate cofactors that are essential for the synthesis of deoxythymidine monophosphate and several amino acids. Since inhibition of DHFR depletes the pool of available thymidine. Viswanathan et al., consider DHFR to be an excellent drug target and broadly reactive for rapidly proliferating bacteria, protozoa, and cancer cells. Viswanathan et al., provide no indication of GH nor IGF-1, nor mention of compounds capable of targeting a cytosolic tail of a type I cytokine receptor and/or capable of targeting the IGH/IGF-1 axis, nor mention a compound capable of selectively inhibiting polypeptide synthesis of a cytosolic tail of a type I cytokine receptor or selectively inhibit paracrine signaling of a type I cytokine. Paulksen et al., Bioorg Med Chem.2009 July 15; 17(14): 4866–4872 is concerned with the aim to develop new antifungal agents effective against two species of Candida with a series of dihydrofolate reductase (DHFR) inhibitors. Paulksen et al., explore the structure-activity relationships of inhibitors toward Candida albicans DHFR by evaluating enzyme inhibition, antifungal activity, and toxicity to mammalian cells. Analysis of docked complexes of the enzyme and inhibitors yields the structural basis of relative potency. According to Paulksen et al., the meta-biphenyl series of this class exhibits the greatest enzyme inhibition, selectivity, and antifungal activity. Paulksen et al., provide no indication of GH nor IGF-1, nor mention of compounds capable of targeting a cytosolic tail of a type I cytokine receptor and/or capable of targeting the IGH/IGF-1 axis, nor mention of a compound capable of selectively inhibiting polypeptide synthesis of a cytosolic tail of a type I cytokine receptor WO1998020878 is concerned with 5-substituted-2,4-diaminopyrimidine compounds (hereinafter "2,4-diaminopyrimidines") and compositions containing the same which are useful for controlling agricultural pests such as insects and acarids. Still more particularly, WO1998020878 relates to certain 2,4- diaminopyrimidine compounds and compositions, and their use as acaricides, and as insecticides, particularly of the order Lepidoptera such as the tobacco budworm, and Coleoptera, such as the Mexican bean beetle, and does not related to cancer treatment. WO1998020878 provides no indication of GH nor IGF-1, nor mention of compounds capable of targeting a cytosolic tail of a type I cytokine receptor and/or capable of targeting the IGH/IGF-1 axis, nor mention a compound capable of selectively inhibiting polypeptide synthesis of a cytosolic tail of a type I cytokine receptor or selectively inhibit paracrine signaling of a type I cytokine. WO2006050843 is concerned with quinazoline compounds for use in medicaments as protein tyrosine phosphatase PTP-1 B inhibitors. WO2006050843 provides no indication of GH nor IGF-1, nor mention of compounds capable of targeting a cytosolic tail of a type I cytokine receptor and/or capable of targeting the IGH/IGF-1 axis, nor mention a compound capable of selectively inhibiting polypeptide synthesis of a cytosolic tail of a type I cytokine receptor or selectively inhibit paracrine signaling of a type I cytokine. WO2004101568 is concerned with quinazoline compounds as protein tyrosine phosphatase inhibitors. WO2004101568 provides no indication of GH nor IGF-1, nor mention of compounds capable of targeting a cytosolic tail of a type I cytokine receptor and/or capable of targeting the IGH/IGF-1 axis, nor mention a compound capable of selectively inhibiting polypeptide synthesis of a cytosolic tail of a type I cytokine receptor or selectively inhibit paracrine signaling of a type I cytokine. WO2011153310 is concerned with quinazoline compounds as DHFR-inhibitors. WO2011153310 provides no indication of GH nor IGF-1, nor mention of compounds capable of targeting a cytosolic tail of a type I cytokine receptor and/or capable of targeting the IGH/IGF-1 axis, nor mention a compound capable of selectively inhibiting polypeptide synthesis of a cytosolic tail of a type I cytokine receptor or selectively inhibit paracrine signaling of a type I cytokine. WO1993013079 is concerned with quinazoline compounds as DHFR-inhibitors. WO1993013079 provides no indication of GH nor IGF-1, nor mention of compounds capable of targeting a cytosolic tail of a type I cytokine receptor and/or capable of targeting the IGH/IGF-1 axis, nor mention a compound capable of selectively inhibiting polypeptide synthesis of a cytosolic tail of a type I cytokine receptor or selectively inhibit paracrine signaling of a type I cytokine. The invention also provides a ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a long cytosolic tail of a cytokine type 1 receptor, and provides use and methods of use of said ribonucleic acid in identifying a molecule capable of non-DHFR and/or non-JAK/STAT mediated inhibition of synthesis of said polypeptide drug target. In a preferred embodiment, Box-1 mediated JAK-binding is at least functionally deleted from said polypeptide drug target. When testing a ribonucleic acid encoding a polypeptide comprising at least a C-terminal part of a cytosolic tail of an cytokine type 1 receptor in identifying a molecule capable of inhibiting synthesis of said polypeptide, the inventors surprisingly detected specific and selective inhibitory activity of several of tested 39K compounds. Even more surprisingly, said inhibitory activity of such compounds appeared independent of DHFR-inhibitory activity or of JAK/STAT-mediated signaling activity, but is provided herein to be specifically and selectively directed against said C- terminal part of a cytosolic tail of an cytokine type 1 receptor, instead. For one, such a compound, with novel activities independent of DHFR- or JAK/STAT inhibitory activity, as provided according to the invention may be advantageously be used as a therapeutic in methods of treatment in cancer therapy, preferably in mammals, at large. For two, it may particularly be useful for therapy in patients with malignancies, hematological or otherwise, that show resistance against DHFR-inhibitor and/or JAK2 inhibitor therapy, such as resistance to methotrexate. For three, it might be used as a general medicine to postpone chronic diseases, associated with the process of aging. In another embodiment, it can be used in domestic animals in all three cases mentioned above. The invention also provides a set of at least two ribonucleic acids according the invention for use as target and control in a method for identifying a molecule capable of polypeptide cytosolic tail sequence specific and selectively inhibiting synthesis of a receptor of the type I cytokine receptor family. In a preferred embodiment, the invention provides a method for testing a molecule for the capacity to selectively inhibit synthesis of a receptor of the type I cytokine receptor family, said method comprising providing a cytosol-like solution comprising a ribosome complex or polyribosome with a first ribonucleic acid encoding a polypeptide drug target comprising at least a C-terminal part of a cytosolic tail of a type I cytokine receptor, providing said solution with a test molecule, determining the effect of the test molecule on translation of said first ribonucleic acid in synthesizing said polypeptide by determining the presence of said polypeptide in said solution. In a preferred embodiment said determining is also comprising and preferably executed in comparison with determining the presence of said polypeptide translated in the absence of said test molecule. In a further preferred embodiment, said determining also comprising and preferably executed in comparison with providing a cytosol-like solution comprising a ribosome complex or polyribosome with a second ribonucleic acid encoding a polypeptide provided with an at least functional deletion or truncation of a C-terminal part of said cytosolic tail of a type I cytokine receptor, providing said solution with a test molecule, and determining the effect of the test molecule on translation of said second ribonucleic acid in synthesizing said polypeptide by determining the presence of said polypeptide in said solution. The invention provides in a preferred embodiment a method according to the invention wherein said test molecule is selected from the group of molecules having general formula 1,

formula 1 or having general formula 2, formula 2. In another preferred embodiment the solution is cell-free. Several cell-free-expression systems of both prokaryotic and eukaryotic origin are known in the art, the invention preferably provides a polypeptide synthesis stalling assay comprising a cell-free protein-expression system for use in candidate drug testing that is of eukaryotic origin. Several eukaryotic cell-free-expression systems are available, for example a commercially available Hela-cell free expression system may be used. In another preferred embodiment the invention provides a method according to the invention wherein the solution is in a living cell in vitro, such as a HEK293 or COS cell or organoid or other cell systems show herein. In another preferred embodiment the invention provides a method according to the invention wherein the solution is in a living cell in vivo, preferably in a mammal, more preferably in an experimental animal, such as a mouse. In another preferred embodiment the invention provides a method according to the invention wherein said ribonucleic acid comprises ribonucleic acid encoding a leucine zipper 5’-terminal and/3’-terminal to said ribonucleic acid encoding said C-terminal part. It is preferred that said ribonucleic acid also encodes a reporter tag. In another preferred embodiment the invention provides a method according to the invention wherein box-1-mediated JAK binding activity is at least functionally deleted from said cytosolic tail. It is wherein said receptor is a group 1 type I cytokine receptor, preferably a long-cytosolic tail type I cytokine receptor. It is preferred that that said receptor is is a growth hormone receptor. It is moreover preferred that said ribonucleic acid encodes a polypeptide provided with a least the last 44 C-terminal amino acids of said cytosolic tail of said receptor, in particular wherein said ribonucleic acid encodes a polypeptide provided with a C-terminal part of a growth hormone receptor at least comprising an amino acid sequence starting after position 594. It is moreover preferred that said (first and/or second) ribonucleic acid or use in a method according to the invention comprises a ribonucleic acid is selected from a set of at least two ribonucleic acids according to the invention as provided herein. The invention also provides a method according to the invention wherein said first and/or second ribonucleic acid at least encodes for a polypeptide encoding at least a part of a cytosolic tail having >70% homology, preferably >80% homology, preferably >90% homology, more preferably 95% homology with a polypeptide amino acid sequence 387-688 of preferably human GHR shown in figure 8. The invention also provides a molecule capable of selectively stalling protein synthesis, preferably for use in stalling synthesis of a receptor of the type I cytokine receptor family. In a preferred embodiment, the invention provides a molecule according to the invention identifiable with an assay according to the invention or identifiable with a method according to the invention. In a particular preferred embodiment, the invention provides a molecule according to the invention, or functional equivalent thereof, selected from the group of molecules herein identified as AK- 105/40836349, AK-105/40837635, AA-504/32626008, AK-105/40836340, AK-105/40833746, AK- 105/40693663, AK-105/40833503, AK-105/40837674, AK-105/40836874, AK-105/40833946, AK- 105/40837563, AE-413/30061043, AK-105/40837629, AK-105/40836387. In a particular preferred embodiment, the invention provides a molecule according to the invention, or functional equivalent thereof, selected from the group of molecules herein identified as AK-105/40836349, AK-105/40837635, AA-504/32626008, AK-105/40836340, AK- 105/40833746, AK-105/40693663, AK-105/40833503, AK-105/40837674, AK-105/40836874. In a particular preferred embodiment, the invention provides a molecule according to the invention, or functional equivalent thereof, selected from the group of molecules herein identified as AK- 105/40836349, AK-105/40837635, AA-504/32626008, AK-105/40836340, AK-105/40833746, AK- 105/40693663. In a particular preferred embodiment, the invention provides a molecule according to the invention,, or functional equivalent thereof, selected from the group of molecules herein identified as AK-105/40836349, AK-105/40837635, AA-504/32626008, AK- 105/40836340. In a most particular preferred embodiment, the invention provides a molecule according to the invention, comprising AK-105/40836349 or functional equivalent thereof. In a most preferred embodiment, the invention provides a molecule according to the invention comprising AK-105/40837635 or functional equivalent thereof. In a most preferred embodiment, the invention provides a molecule according to the invention comprising AA-504/32626008 or functional equivalent thereof. In a most preferred embodiment, the invention provides a molecule according to the invention comprising AK-105/40836340 or functional equivalent thereof. In another most preferred embodiment, a molecule or functional equivalent thereof according to the invention is provided for use in treatment of a subject having or suspected to having a type I cytokine receptor mediated disease, in particular wherein said disease comprises cancer, in particular wherein said disease comprises secondary primary cancer, more in particular wherein said disease comprises metastatic cancer, more in particular wherein said disease comprises treatment-resistant cancer, more in particular said disease comprises growth hormone assisted cancer, more in particular wherein said disease comprises chemotherapy-resistant cancer. The invention also provides a pharmaceutical formulation comprising a molecule according to the invention, preferably accompanied with a pharmaceutically acceptable excipient. In another embodiment, the invention provides a method of treatment of a subject having or suspected to having a type I cytokine receptor mediated disease comprising administering to said subject a molecule according the invention or a pharmaceutical formulation according to the invention, in particular wherein said disease comprises cancer, in particular wherein said disease comprises secondary primary cancer, more in particular wherein said disease comprises metastatic cancer, more in particular wherein said disease comprises treatment-resistant cancer, more in particular said disease comprises growth hormone assisted cancer, more in particular wherein said disease comprises chemotherapy-resistant cancer. In a further embodiment the invention provides a method for obtaining a molecule capable of inhibiting synthesis of a receptor of the GH/prolactin receptor family and of such a compound use in disease under control of the growth hormone receptor dependent GH/IGF-1 axis. Compounds capable of inhibiting or stalling the synthesis of a GHR have hitherto not been provided for use in disease under control of the GH/IGF-1 axis. The present invention also provides GHR-synthesis inhibiting or stalling compounds, in particular related to GHR-synthesis inhibiting or stalling pyrimidine-2,4-diamines and to GHR- synthesis inhibiting or stalling 2,4-quinazolinediamines, and to analogues and salts thereof and pharmaceutical compositions comprising GHR-synthesis stalling pyrimidine-2,4-diamines analogues and GHR-synthesis stalling 2,4-quinazolinediamines, and salts thereof for use in the treatment and prevention of a disease, in particular a growth hormone receptor-dependent condition. The invention also relates to methods of using these compounds and compositions to treat physiological disorders related to the activity of growth hormone , more particularly for the treatment of cancer, preventing or treating cancer metastasis, treatment or prevention of diabetes, treatment of chronic inflammation such as rheumatoid arthritis and Crohn's disease, treatment of acromegaly, treatment or prevention of neurodegenerative diseases, treatment or prevention of idiopathic pulmonary disease and other occurrences of fibrosis, and the treatment or prevention of autoimmune diseases, such as Lupus erythematosus and macular degeneration, improving long- and short-term memory and increasing health span. In particular, the invention relates to the treatment or prevention of GHR-dependent cancer such as melanoma, acute myeloid leukaemia, chronic lymphocytic leukaemia, colorectal cancer, renal cell carcinoma, breast cancer, lung cancer, ovarian cancer, fallopian tube carcinoma, primary peritoneal carcinoma, cervical cancer, gastric cancer, liver cancer, pancreatic cancer, thyroid cancer, glioma, non-Hodgkin's lymphoma, and Hodgkin's lymphoma. The present invention solves one or more problems of the prior art by providing in at least one embodiment, a compound or composition for treating diseases or conditions by causing inhibition of the synthesis of the growth hormone receptor (GHR). The compound according to this embodiment has structural formula 1 Formula 1 wherein A and E represent carbon atoms with binding between A and E with a single bond or a double bond in E-configuration or a triple bond. U, V, X, Y and Z represent carbon atoms or one of them represents an unsubstituted N to form a pyridine, R 1 is (1C-3C)-alkyl or 3,3,3-trifluoropropyl, R 2 is methyl and R 3 is hydrogen or methyl, or R 2 and R 3 form together with the carbon to which they are attached a cyclopropane, or R 2 and R 3 form together with the carbon to which they are attached a cyclopentane, R 4 , R 7 , R 8 , R 9 and R 10 are hydrogen, halogen, trifluoromethyl, methoxy, trifluoromethoxy, morpholine or methylsulfonyl, R 5 and R 6 are hydrogen, ethyl or R 5 or R 6 are pyridin-2-ylmethyl, or a pharmaceutically acceptable salt thereof. The present invention solves one or more problems of the prior art by providing in at least another embodiment, a compound or composition for treating diseases or conditions by causing inhibition of the synthesis of the growth hormone receptor (GHR). The compound according to this embodiment has structural formula 2 Formula 2 wherein R1 of said test molecule according to formula 2 is hydrogen or (1C-3C)alkyl or (1C- 4C)alkyloxy; and wherein R2 is phenyl optionally substituted with one or more groups selected from (1C-3C)alkyl, (1C- 3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R2 is N-methyl-benzyl-amine optionally substituted with one or more groups selected from (1C- 3C)alkyl, (1C-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R2 is benzyl-amine optionally substituted with one or more groups selected from (1C-3C)alkyl, (1C-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R2 is anilinomethyl optionally substituted with one or more groups selected from (1C-3C)alkyl, (1C-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R2 is benzenesulfinyl optionally substituted with one or more groups selected from (1C-3C)alkyl, (1C-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R2 is benzenesulfide optionally substituted with one or more groups selected from (1C-3C)alkyl, (1C-3C)alkyloxy, ethyl-3-oxypropanoate, cyano, halogen, sulfonyl, trifluoromethyl, or R1 and R2 together form 2,2-dimethyl-3H-furo[2,3-f] or R1 and R2 together form 7-(cyclopropylmethyl)pyrrolo[3,2-f]; or a pharmaceutically acceptable salt thereof. Legend to the figures Figure 1. Diagram of the primary screening method for candidate compounds. Depicted are GHR fos-zipped cytosolic tails, amino acid 288-638 with fos zippers (fos), box-1 sequence (B1), and Jak2 (FERM, SH2, pseudokinase and kinase domains), as described in Nespital et al., 2016; Sedek et al, 2014). If the dimerized tails and Jak2 bind via FERM and Box-1, Jak2 gets activated and phosphorylates itself, the dimerized tails and STAT5b. After phosphorylation of specific tyrosine residues on GHR, Jak2 and STAT5, the complex dissociates. As fos-pGHRct is protease-resistant it accumulates in the cells. Unphosphorylated fos-GHRct is rapidly degraded. Steady state levels of Fos-pGHRct are lower if its synthesis, degradation, or Jak2-interaction is changed by a candidate compound. The cell-based assay was performed in γ2A cells carrying inducible Jak2 (iJak2) or binding-deficient iJak2 Y119E (as control). Figure 2. Effect of test compounds on mice, xenografted with MDA-MB-231 cells. Athymic nude BALB/c mice were transplanted with luciferase-expressing MDA-MB-231 cells at week -5. Starting at week 0, compound BM001 was injected 3 times a week for 3 weeks; group size: 9, intraperitoneally (IP) at 1 or 5 mg/kg as indicated. Anesthetized mice were IP-injected with luciferin and the bioluminescence was imaged. The animals received the last drug injections 2 hours before they were euthanized, then blood was collected, and livers were weighed. Mice treated with compound BM001 showed a significantly lowered serum IGF-1 (27%) and liver weight (85%), whereas total body weights remained the same. Panel A: Quantification of bioluminescence imaging of mice treated with compound BM001, BM002 and BM003 using near-infrared fluorescence molecular imaging as described in Vermeulen et al., 2013. Panel B: Tumour growth of control and mice treated with compound BM001 using bioluminescence imaging (PhotonIMAGER, Biospace Lab, Paris, France). Panel C: Plasma IGF-1 levels of control and mice treated with compound BM001. Panel D: Liver weights of control and mice treated with compound BM001. Figure 3: Effect of compound BM001 on GHR/Jak/STAT functions in canine mammary organoids derived from tumour cells. Organoids were cultured in Matrigel in the presence of a variety of growth factors, Wnt activators and MPA. Organoids were treated for 48 h with 100 nM of compound BM001 followed by staining with DAPI (nuclei) or specific antibodies against GHR or cytokeratin 8. Figure 4A: Effect of compounds on GHR-levels. Hek293T cells transfected with GHR were treated overnight with 10 µM, as indicated in the figure. The amounts of GHR both at the plasma membrane (PM) and in the ER (ER) were decreased in the drug-treated cells. BM001 had the strongest effect. The cells were treated for GH for 10 min before lysis to be able to monitor the pY signals, after GHR isolation by immunoprecipitation (middle panels). The effect of the drugs on the pY signals reflected those of the GHR levels (only PM species were visible). In the presence of the Jak2 inhibitor, ruxolitinib, the protein levels (upper panel) were unchanged, while the pY label (middle panel) was absent. The lower panel (actin) shows that the amounts of cells were identical across the experiment. Figure 4B: BM001 concentration effects on the synthesis of fos-zippered GHR cytosolic tails in an HeLa cell cell-free system at 30°C for 120 min, and the blot was detected with a rabbit anti-GHR antiserum. Synthesis was inhibited irrespective of the presence of a functional box-1 sequence (EC500.1-1.0 µM). In the right panel the 4 proline residues in Box-1 were replaced by alanine residues. Empty vector (pcDNA, lane 11) was used as control. Figure 4C: In vitro translation as in Figure 4B, using various drugs as described in the experimental part at a concentration of 1 µM. BM001 (lanes 2, 12, 13) was the strongest inhibitor; BM007, GHR016, and GHR36 showed some inhibition, pcDNA3 vector served as control. Methotrexate (MTX) was tested in the in vitro translation as a negative control (lane 1). Figure 5: Schematic view of GHR signaling. Top panel, downstream signaling pathways with their main expressed genes and enzymatic activities; middle panel, resulting effect; lower panel, major diseased states if unbalanced. Figure 6: Drugs do not affect GHR with C-terminally shortened cytosolic tail BM001 targets the fos-GHRct. Hek293T cells transfected with full-length GHR (19-638) were treated for 4 h with DMSO (lane 1), 10 µM Ruxolitinib (lane 2) and 10 µM BM001 (lane 3). Chinese hamster cells (ts20), expressing GHR, truncated at amino acid 387 were treated as above. While in the presence of BM001 the full-length GHR receptor was virtually absent both in the ER and at the plasma membrane, the drug had no effect on the truncated GHR species. In both cases JAK- inhibitor Ruxolitinib did not affect GHR levels Figure 7: Drugs do not affect GHR with C-terminally shortened cytosolic tail C-terminally HA-tagged GHR constructs were made and expressed in MDA-MB-453 cells in the presence and absence of BM001. Quantification of expression results as shown in figure 9. Figure 8. Growth hormone receptor cytosol tails of relevant species. Growth hormone receptor cytosol tails of relevant species. Of relevance are the sequences downstream of amino acid 387, using the human sequence as internal reference. Fig.6 shows that the drugs target the synthesis of the GHR downstream of amino acid 387. This part of the sequence contains 3 conserved stretches of amino acids (underlined). To identify the targeted sequence 3 truncations were made downstream of amino acid 387, indicated in red, and shown in Fig.9 for the human receptor. Figure 9. C-terminally HA-tagged GHR constructs were made as indicated in the top panel and expressed in MDA-MB-453 cells in the presence and absence of BM001 as indicated in the figure. Both the precursor and the mature form of the GHR disappeared in the presence of BM001 if the full- length GHR construct was expressed. Synthesis of the truncated GHR species was not affected, indicating that the drug targets the last 50-60 amino acid sequence of the GHR. Top: C-terminal sequence (position 579-638) of several GHR species with an at least partial C- terminal stalling sit indicated, as tested on GHR of rabbit, mice, dog and human. Middle: GHR constructs used C-terminally HA-tagged. Bottom: This shows that if you change, replace, truncate, or delete the last 44 amino acids by for example an HA tag the translation is not affected anymore by BM001. This indicates that the stalling sequence is in the last 50-60 amino acids. This part of the GHR sequence is very much conserved among species. This correlates with results obtained with the different species that show an effect of BM001 on GHR synthesis, and signaling results as in dog organoids, human IM9 cells, rabbit cDNA derived synthesis, mice IGF1 levels and effect on mice liver size. Figure 10 a In vitro profiling of compound BM001 (identified in figure 10 as BIMI001) in chemoresistant cell lines relating to SMALL CELL LUNG CANCER (NCIH82), LARGE CELL LUNG CANCER (NCIH460), BLADDER CARCINOMA (UMUC3), COLON CARCINOMA, (HCT116), COLON ADENOCARCINOMA (HCT15), CUTANEOUS MELANOMA (SKMEL5) was performed using six 10-fold compound dilutions prepared in DMSO. Treatment duration was 72 hours. Growth inhibition was measured by using Sulforhodamin B, a protein staining assay. Sulforhodamin B assay has been established and recommended by the DTP NCI/NIH (USA). IC50 was measured for all cells as indicated in the figure. Figure 10 b In vitro profiling of compound BM001 (identified in figure 10 as BIMI001) in chemoresistant cell lines relating to AMELANOTIC MELANOMA (A375), ADULT HEPATOCELLULAR CARCINOMA (SKHEP1), EWING SARCOMA (RDES), EWING SARCOMA (MHHES1), TRIPLE NEGATIVE BREAST CANCER (MDAMB231), TRIPLE NEGATIVE BREAST CANCER (MDAMB435), MELANOMA (MDAMB435) was performed using six 10-fold compound dilutions prepared in DMSO. Treatment duration was 72 hours. Growth inhibition was measured by using Sulforhodamin B, a protein staining assay. IC50 was measured for all cells as indicated in the figure. Figure 10c In vitro profiling of compound BM001 (identified in figure 10 as BIMI001) in chemoresistant cell lines relating to MELANOMA (SKMEL28), and MELANOMA (SKMEL5) and to non-resistant control cell cultures (here results with peripheral blood mononuclear cells (PBMC) is shown) was performed using six 10-fold compound dilutions prepared in DMSO. Treatment duration was 72 hours. Growth inhibition was measured by using Sulforhodamin B, a protein staining assay. IC50 was measured for all cells as indicated in the figure. Peripheral blood mononuclear cells and other non-resistant cells were not significantly affected by BMI001. Figure 11 Domain structures of class 1 cytokine receptors showing disulphide bonded domain 1 (yellow) and domain 2 (red), with Box 1 and 2 sequences and the conserved WSxWS motif (figure from Waters: The growth hormone receptor, Growth Hormone & IGF Research. (2015). The growth hormone (GH) receptor was the first type (class) I cytokine receptor to be cloned, and is an exemplar for the around 30 receptors in this class, which include the receptors for erythropoietin, prolactin, leptin, thrombopoietin, LIF, CTNF, oncostatin-M, carditropin-1 and most of the interleukins, together with many of the haematopoietic colony stimulating factors. To acquire an active state, these receptors form homodimers, or heterodimers with accessory proteins such as gp 130, common beta and common gamma chains. Long-cytosolic tail and short-cytosolic tail type I cytokine receptors and their Box 1 motifs are shown, as well as common subunits shared by heteromeric receptors. As provided herein, targeting a polypeptide drug target of a long-cytosolic tail type receptor (preferably inhibiting its synthesis) suffices to target homodimer as well as heterodimer type I cytokine receptors. Figure 12 Overview of current best compounds as provided herein having selective protein synthesis stalling activity. AK-105/40836349 BM001 30 nM AK-105/40837635 BM002 50 nM AA-504/32626008 BM003 50 nM AK-105/40836340 BM004 80 nM AK-105/40833746 BM005 200 nM AK-105/40693663 BM006 200 nM AK-105/40833503 BM007 400 nM AK-105/40837674 BM008 500 nM AK-105/40836874 BM009 600 nM AK-105/40833946 BM010 800 nM AK-105/40837563 BM011 900 nM AE-413/30061043 BM012 900 nM AK-105/40837629 BM013 900 nM AK-105/40836387 BM014 900 nM

Detailed description of the invention Cytokines represent a diverse group of small soluble proteins that when secreted by one cell can act on the same cell, in an autocrine fashion, or on another cell, in a paracrine or endocrine fashion. Through binding to specific receptors, they initiate signals that are critical to a diverse spectrum of functions, including induction of immune responses, cell proliferation, differentiation, and apoptosis. On the basis of common structural features, the cytokine receptors are grouped into five major families: Type I cytokine receptors with type II cytokine receptors, TNF receptors, IL-1 receptors, tyrosine kinase receptors, and chemokine receptors. All type I cytokine receptors have four conserved cysteine residues, fibronectin type II modules, a Trp-Ser-X-Trp-Ser motif in the extracellular domain, and a proline-rich Box 1/Box 2 region in the cytoplasmic domain. With the exception of stem cell factor, type I cytokine receptors do not contain catalytic domains such as kinases. The prolactin receptor (PRLR) is a member of the type 1 cytokine receptor family and its nearest relative is the growth hormone receptor (GHR). Several hematopoietic cytokine receptors, such as those for erythropoietin, most interleukins, and granulocyte-macrophage colony-stimulating factor, are also very similar to PRL and GH receptors. Type I cytokine receptors also include the interleukin-2 (IL-2) receptor (IL-2R), interleukin-3 receptor (IL-3R), interleukin-6 receptor (IL-6R), and interleukin-12 receptor (IL-12R) subfamilies, Cytokines signaling within subfamilies exhibit strikingly different activities, underscoring a central issue: the means by which specificity is achieved through similar receptor types. Type I cytokine receptors generally require homodimerization for activation. First, the ligand may bind a monomeric receptor. Then, the ligand interacts with a second receptor to induce receptor dimerization and activation. Activated receptors then stimulate members of the Janus family of tyrosine kinases (Jak kinases) to phosphorylate tyrosine residues in itself and the cytoplasmic region of the receptors. Signal transducers and activators of transcription (STATs) then dock on the phosphorylated cytoplasmic receptor domains or Jak kinases via an SH2 domain and are tyrosine phosphorylated. The phosphorylated STATs then dissociate from the receptors or Jak kinases, form homo- or heterodimers, and translocate to the nucleus. In the nucleus, the STAT dimers bind and alter the activity of regulatory regions of target DNA. Several hematopoietic cytokine receptors, such as those for erythropoietin, most interleukins, and granulocyte-macrophage colony-stimulating factor (GM-CSF), are also very similar to PRL and GH receptors. Reference is made to preferred compositions, embodiments, and methods of the present invention, which constitute the best modes of practicing the invention presently known to the inventors. The disclosed embodiments are merely exemplary of the invention that may be embodied in various and alternative forms. Therefore, specific details disclosed herein are not to be interpreted as limiting, but merely as a representative basis for any aspect of the invention and/or as a representative basis for teaching one skilled in the art to variously employ the present invention. As used herein, the singular form "a," "an," and "the" comprise plural referents unless the context clearly indicates otherwise. Reference to a component in the singular is intended to comprise a plurality of components. Where publications are referenced, the disclosures of these publications in their entireties are hereby incorporated by reference into this application to more fully describe the state of the art to which this invention pertains. The ribosome is a complex molecule made of ribosomal RNA molecules and proteins that form a factory for protein synthesis in cells. In 1955, George E. Palade discovered ribosomes and described them as small particles in the cytoplasm that preferentially associated with the endoplasmic reticulum membrane. Along with other scientists, Palade discovered that ribosomes performed protein synthesis in cells, and he was awarded the Nobel Prize in 1974 for his work. Each ribosome has a large component and a small component that together form a single unit composed of several ribosomal RNA molecules and dozens of proteins. The ribosome is responsible for translating encoded messages from messenger RNA molecules to synthesize proteins from amino acids. The ribosome translates each codon, or set of three nucleotides, of the mRNA template and matches it with the appropriate amino acid in a process called translation. The amino acid is provided by a transfer RNA (tRNA) molecule. Each newly translated amino acid is then added to the growing protein chain until the ribosome completes the process of protein synthesis. An extended number of studies in animals indicate the involvement of endocrine GH and IGF-1 in tumour growth promotion and demonstrate that effective therapeutic options for cancer treatment need to drastically lower serum IGF-1 (Chhabra, 2011). Attempts to successfully develop effective IGF-1 inhibitor failed, as IGF-1 (and somatostatin) act in an auto-inhibitory loupe with GH in the pituitary: interrupted IGF-1 signaling causes upregulation of the Jak/STAT pathway via high concentrations of GH in the blood. Early attempts to treat breast cancer include hypophysectomy (Jessiman et al., 1959) and application of somatostatin receptor ligands, such as octreotide, lanreotide, pasireotide, somatoprim, etc., that inhibit GH secretion (Theodoropoulou and Stalla, 2013). Treatment with the GH antagonist, Pegvisomant, is currently the only way to reduce GH activity (Basu et al., 2018). The actions of GH are a result of activation of its receptor, and loss of function of this receptor may have consequences. Because GH is a modulator, such a loss is not considered lethal. Herein we describe the identification of compounds, in particular pyrimidine-2,4- diamines according to formula 1, as the first small molecular inhibitor of the integral growth hormone receptor dependent GH/IGF-1 axis. Formula 1. wherein A and E represent carbon atoms with binding between A and E with a single bond or a double bond in E-configuration or a triple bond. U, V, X, Y and Z represent carbon atoms or one of them represents an unsubstituted N to form a pyridine, R 1 is (1C-3C)-alkyl or 3,3,3-trifluoropropyl, R 2 is methyl and R 3 is hydrogen or methyl, or R 2 and R 3 form together with the carbon to which they are attached a cyclopropane, or R 2 and R 3 form together with the carbon to which they are attached a cyclopentane, R 4 , R 7 , R 8 , R 9 and R 10 are hydrogen, halogen, trifluoromethyl, methoxy, trifluoromethoxy, morpholine or methylsulfonyl, R 5 and R 6 are hydrogen, ethyl or R 5 or R 6 are pyridin-2-ylmethyl, or a pharmaceutically acceptable salt thereof. These molecules act on the synthesis of the growth hormone receptor (GHR) and deplete cells from GH signaling activity. Consequently, mice treated with the molecule have low serum levels of IGF-1. In addition, the molecule inhibits the growth of human triple-negative breast cancer cells both in tissue culture and in xenografted mice. We screened a 38,720-compound library and in two consecutive rounds of analogue selection we detected a small molecule that inhibited signaling via the GH receptor (GHR) and its downstream effectors Jak2 and STAT5. This molecule is identified herein as compound BM001. We then synthesized and tested several analogues of compounds according to formula 1 or 2 and came up with more active compounds (table 1 and 2). An active compound in this respect is herein defined as a compound according to formula 1 or 2 that shows either: 1. A lower plasma level of IGF-1 in a model of xenografted mice according to example 5, 2. Anti-tumour activity in a model of xenografted mice according to example 5, 3. Anti-proliferation activity with an EC50 value of 2000 nM or less (wherein less means a value smaller than 2000 nM) in an assay according to example 4, 4. A value of less than 1 in the viability assay according to example 7 on either MDA- MB-231 cells or MDA-MB-453 cells. In this way a set of 28 compounds according to formula 1 were synthesized and tested (example 8). All 28 compounds showed the desired activity. These compounds prevent the translation of GHR (Figure 4) and the mice treated with these compounds showed the same phenotype as GHR knock-out mice (GHR-KO) and people with inherited Laron disease who lack a functional GHR (Laron, 2015). As a consequence of that, their length growth is hampered. Yet, they live normal lives, are fertile, and have a healthy normal life span. However, they do not suffer from cancer or diabetes type 2. Numerous studies on GHR KO mice have confirmed these phenotypes. These mice are resistant to cancer incidence and progression. In addition, these mice have reduced serum IGF-1, increased insulin sensitivity, are resistant to High-Fat-Diet-induced diabetes, and to diabetic kidney disease, have a 21%-40% longer lifespan, a slower age-related neuromusculoskeletal deterioration, improved mitochondrial biogenesis, are resistant to oxidative stress, have less senescent cells, less fibrosis, less macrophage inflammation, lower hepatic inflammation and steatosis, increased brain size compared to body, increased brain NMDA-receptors, better long-and short-term memory, and better cognitive performance. In humans, it is generally appreciated that the growth hormone receptor dependent GH/IGF-1 axis is a driver for all these diseases and conditions. Consequently, a small molecule that is able to down-regulate the GH/IGF-1 axis is useful in the treatment or prevention of each of these conditions, as a principal drug or in combination with other treatments. The compounds and compositions disclosed herein may therefore be used to inhibit cancer and/or prevent/inhibit cancer metastasis, to prevent or cure diabetes, to act as geroprotectors and senolytics and to increase health span. Harnessing the activity of GH has been a long-time goal. Initially, the actions were aimed at fighting the activity in GH-overproducing individuals called acromegaly. Recently, it became clear that the growth hormone receptor dependent GH/IGF-1 axis constitutes a driver behind many conditions that shortens health-span in humans. Solid evidence shows that in many tissues’ cancer growth depends on the activity of the growth hormone receptor dependent GH/IGF-1 axis. Now it is generally accepted that both GH and IGF-1 are drivers of the major human cancers. In addition, growth hormone receptor dependent GH/IGF-1 axis appears to have a positive role in onset and progress of other chronic diseases as indicated above, Several efforts have aimed or aim at the development of an IGF-1 inhibitor. The problem is that IGF-1 acts in an auto-inhibitory loop to control GH activity. If IGF-1 activity is artificially decreased, the levels of GH remain high, causing hyperstimulation of the Jak2/Stat and Src pathways, which in itself contribute to cancer and diabetes. There are many efforts to inhibit the GH/IGF-1 axis downstream of their receptors, e.g. aimed at: Jak1, Jak2, STAT, AKT, mTOR, glucose control, or protein synthesis. They all have the disadvantage that they act on general 'house-hold' enzyme systems and lack specificity. An exception is metformin. Metformin has been used as a safe and effective treatment for diabetes type 2 for over half a century, yet the precise mechanism of action of this drug remains elusive. The anti-hyperglycaemic properties of metformin are chiefly mediated by suppressing hepatic gluconeogenesis and it is generally accepted that this is achieved via inhibition of complex I in the mitochondrial respiratory chain. This impedes gluconeogenic flux, interferes with glucagon signaling and promotes activation of the major metabolic regulator AMPK. There are no other small molecules known to regulate the GH/IGF-1 axis The drug development protocol described herein resulted in a number of pyrimidine- 2,4-diamines and quinazolines that were found to be effective in inhibiting the translation of GHR. The assays as described in the examples were designed to find drugs that inhibit GHR signaling and was based on the observation that inhibiting the growth hormone receptor dependent GH/IGF-1 pathway will inhibit cancer growth. The results of the mice experiments of Fig.2A and 2B present the first supporting evidence for this purpose. The drugs were also shown to decrease liver weight and IGF-1 levels, as in GHR knockout mice and Laron patients. GHR knock- out mice have low circulating IGF-1 and high GH levels due to the lack of GHR activity and less negative feedback of IGF-I on the pituitary gland (Zhou et al., 1997). Figures 2C and 2D show that the same phenotype was found in mice treated with the compounds and compositions according to the present invention. The animals of Fig.2A received the last drug injections 2 h before they were euthanized, then blood was collected, and livers were weighed. Only animals treated with the compounds according to the invention showed significant effects. The drugs indeed lowered serum IGF-1 (27%) and liver weight (85%), while total body weights remained the same. The results of Fig.2 add another important piece of information: they prove that the compounds according to the invention work systemic on the growth hormone receptor dependent GH/IGF-1 axis in mice, in addition to the human GHR system (as in the MDA-MB-231 cells), and the rabbit GHR (as in the primary screen and the transfected Hek293T cells). Next, we determined that also growth of dog breast cancer cells, grown as organoids, could be inhibited with the compounds according to the invention, in the best case (compound BM001) with an EC50 of about 20 nM, see also figure 3. Hence, the invention relates to the medical use of a group of compounds according to formula 1 or a composition comprising such a compound in the treatment or prevention of a disease in a subject Formula 1 wherein A and E represent carbon atoms with binding between A and E with a single bond or a double bond in E-configuration or a triple bond. U, V, X, Y and Z represent carbon atoms or one of them represents an unsubstituted N to form a pyridine, R 1 is (1C-3C)-alkyl or 3,3,3-trifluoropropyl, R 2 is methyl and R 3 is hydrogen or methyl, or R 2 and R 3 form together with the carbon to which they are attached a cyclopropane, or R 2 and R 3 form together with the carbon to which they are attached a cyclopentane, R 4 , R 7 , R 8 , R 9 and R 10 are hydrogen, halogen, trifluoromethyl, methoxy, trifluoromethoxy, morpholine or methylsulfonyl, R 5 and R 6 are hydrogen, ethyl or R 5 or R 6 are pyridin-2-ylmethyl, or a pharmaceutically acceptable salt thereof. One member of this group (compound BM001) is known as a pesticide and described in WO 98/20878. The compounds described herein are administered to the subject in a therapeutically effective amount so that growth hormone receptor dependent conditions may be treated or prevented. The invention also provides a pharmaceutical formulation comprising a compound according to the invention a and a pharmaceutical formulation comprising a compound according to the invention with a pharmaceutical excipient such as a salt or a carrier. Examples of such conditions or diseases that may be treated or prevented by such a formulation are cancer, cancer metastasis, diabetes, chronic inflammation, rheumatoid arthritis, Crohn's disease, acromegaly, neurodegenerative diseases, idiopathic pulmonary disease, fibrosis, autoimmune diseases, Lupus erythematosus and macular degeneration. The compounds may also be used for improving long- and short-term memory and increasing health span. The compounds as described herein may form pharmaceutically acceptable salts with both organic and inorganic acids or bases. For example, the acid addition salts of the basic compounds are prepared either by dissolving the free base in aqueous or aqueous alcohol solution or other suitable solvents containing the appropriate acid and isolating the salt by evaporating the solution. Examples of pharmaceutically acceptable salts are hydrochlorides, hydrobromides, hydrosulfates, etc. as well as sodium, potassium, and magnesium, etc. salts. The invention includes the individual diastereomers or enantiomers, and the mixtures thereof. The individual diastereomers or enantiomers may be prepared or isolated by methods already well- known in the art. Pharmaceutical compositions include the compounds as described herein or a salt thereof and a pharmaceutical carrier. Typically, the pharmaceutical compositions are divided into dosage units. Examples of dosage unit forms include, but are not limited to, pills, powders, tablets, capsules, aqueous and non-aqueous oral solutions and suspensions, and parenteral solutions. Examples of suitable pharmaceutical carriers include, but are not limited to, water, sugars (e.g., lactose and sucrose), starches (e.g., corn starch and potato starch), cellulose derivatives (e.g., sodium carboxymethyl cellulose, and methyl cellulose), gelatin, talc, stearic acid, magnesium stearate, vegetable oils (e.g., peanut oil, cottonseed oil, sesame oil, olive oil, etc.), propylene glycol, glycerin, sorbitol, polyethylene glycol, water, agar, alginic acid, saline, and other pharmaceutically acceptable materials. The percentage of the active ingredients in the pharmaceutical compositions can be varied within wide limit. For example, a concentration of at least 10% in a solid composition and at least 2% in a primary liquid composition is used. Routes of administration of the compound or its salts are oral or parenteral. For example, a useful intravenous dose is between 1 and 50 mg and a useful oral dosage is between 5 and 800 mg. Compounds as described herein may advantageously be used as a medicament for treating a disease, in particular when the disease is a growth hormone receptor-dependent disease. The term “growth hormone receptor dependent disease” is used herein to indicate a disease that is associated with an increased amount or activity of growth hormone receptor or caused by an increased amount or activity of growth hormone receptor. Moreover, the term “growth hormone receptor dependent disease” may also indicate a disease that is associated with an altered activity of the growth hormone receptor or caused by a locally increased amount or activity of growth hormone receptor. More in particular, the term “growth hormone receptor dependent disease” is used herein to indicate a disease selected from the group consisting of cancer, cancer metastasis, diabetes, chronic inflammation, rheumatoid arthritis, Crohn's disease, acromegaly, neurodegenerative diseases, idiopathic pulmonary disease, fibrosis, autoimmune diseases, Lupus erythematosus and macular degeneration. The compounds may also be used for improving long- and short-term memory and increasing health span. The compounds as disclosed and described herein may be used for treatment and/or prevention of diseases as disclosed herein wherein treatment or prevention is selected from the group consisting of the treatment or prevention of cancer, preventing cancer metastasis, treatment or prevention of diabetes, treatment of chronic inflammation such as rheumatoid arthritis and Crohn's disease, treatment of acromegaly, treatment or prevention of neurodegenerative diseases, treatment and prevention of idiopathic pulmonary disease and other occurrences of fibrosis, treatment and prevention of autoimmune diseases, such as Lupus erythematosus and macular degeneration, improving long- and short-term memory and increasing health span. Preferred use is the use of a compound as disclosed herein for the treatment or prevention of cancer. Preferably, it is used to prevent metastasis of cancer cells. Particularly preferred is the use of a compound as described herein in the treatment of cancer wherein the cancer is selected from the group consisting of melanoma, acute myeloid leukaemia, chronic lymphocytic leukaemia, colorectal cancer, renal cell carcinoma, breast cancer, lung cancer, ovarian cancer, fallopian tube carcinoma, primary peritoneal carcinoma, cervical cancer, gastric cancer, liver cancer, pancreatic cancer, thyroid cancer, glioma, non-Hodgkin's lymphoma, and Hodgkin's lymphoma. In a preferred embodiment, the invention relates to a compound as disclosed above, wherein R5 is pyridin-2-ylmethyl and its medical use as disclosed herein. In a further preferred embodiment, the invention relates to a compound as disclosed above wherein R5, R6, R7, R8 and R9 are hydrogen and its medical use as disclosed herein. In an even further preferred embodiment, the invention relates to a compound as described above wherein R1 is methyl, R2 and R3 form together with the carbon to which they are attached a cyclopropane and R4 is chloro- or trifluoromethyl. In an even further preferred embodiment, the invention relates to a compound selected from the group consisting of 6-methyl-5-[2-{1-[4- (trifluoromethyl)phenyl]cyclopropyl}ethenyl]pyrimidine-2,4-d iamine, 5-{[1-(4- chlorophenyl)cyclopropyl]ethynyl}-6-methylpyrimidine-2,4-dia mine, 5-{[1-(4- chlorophenyl)cyclopropyl]ethynyl}-6-methylpyrimidine-2,4-dia mine, 5-[3-(4-fluorophenyl)butyl]- 6-methylpyrimidine-2,4-diamine, 5-{3-methyl-3-[4-(trifluoromethyl)phenyl]but-1-en-1-yl}-6-(3 ,3,3- trifluoropropyl)pyrimidine-2,4-diamine, 6-methyl-5-[3-methyl-3-[4-(trifluoromethyl)phenyl]but-1- en-1-yl]- N4-[(pyridin-2-yl)methyl]pyrimidine-2,4-diamine, 6-methyl-5-{3-methyl-3-[4- (trifluoromethyl)phenyl]but-1-en-1-yl}-N2-[(pyridin-2-yl)met hyl]pyrimidine-2,4-diamine and 6- methyl-5-{3-[4-(trifluoromethyl)phenyl]but-1-en-1-yl}pyrimid ine-2,4-diamine and the medical uses thereof as described herein.. The invention also relates to compositions comprising a compound as described herein and their medical uses. The term “subject” as used herein is intended to refer to a human or an animal, a mammal, a primate, a higher primate, a sheep, a dog, a rodent, a mouse, a rat, a guinea pig, a goat, a pig, a cat, a rabbit, or a cow.

Examples Example 1: In vitro screening method In our previous studies (Nespital et al., 2016; Sedek et al., 2014) we showed that soluble fos-zipped GHR cytosolic tails (fos-GHRct) can serve as signaling complexes when co- expressed with Jak2 (Figure 1). In this system, Fos-GHRct expressed without Jak2 is immediately degraded by the ubiquitin system. Phosphorylated fos-GHRct (fos-pGHRct) detaches from Jak2 and unzips. Only unzipped fos-pGHR cytosolic tail are protected from proteasomal degradation and accumulate in the cell. This system therefore offers a very sensitive cell-based assay for high- throughput screening with 4 functional parameters, namely: fos-pGHRct using both anti-GHR (a) and anti-pY (b), as well as anti-pJak2 (c) and anti-pSTAT5b (d) (Figure 1). The assay is herein used as a candidate-drug-test to screen for 6 different activities that will lead to a decreased or abolished signal: if a candidate compound (1) inhibits Jak2 kinase activity, (2) inhibits the fos-GHRct synthesis, (3) inhibits the fos-GHRct:Jak2 interaction, (4) induces fos-pGHRct degradation, (5) induces Jak2 degradation, and (6) induces fos-pGHRct dephosphorylation. Each of these 6 possibilities will result in lower signals a-d mentioned above (Nespital et al., 2016; Sedek et al., 2014). In the primary screen we used γ2A cells with inducible Jak2 (iJak2) or iJak2-119E as binding-deficient Jak2 control (Figure 1). Example 2: Flow cytometer-based assay 109 primary hits from the assay described in example 1 were selected and the best 17 were tested in a flow cytometer-based assay of whole human blood, supplemented with human IM-9 lymphoblasts that have detectable levels of endogenous GH receptor (Dr. van Agthoven, Beckman-Coulter, Marseille, paid service) (Malergue et al., 2015). This allowed simultaneous testing of GH- and GM-CSF activity. The GH/GM-CSF cytometric test added two important criteria to GH signaling: The test prioritized compounds that target the GH-driven signaling at the expense of another Jak2-dependent cytokine signaling (GM- CSF); equally important: it selected compounds that exert a rapid effect on the growth hormone receptor dependent GH/IGF-axis as the response time for STAT5 phosphorylation was limited to 20-30 min. Given the short resident time of the GHR both in the synthesis (endoplasmic reticulum) and the signaling compartment (the plasma membrane (PM) of less than 30 min, candidate hits must have an immediate and strong effect on the signaling capacity via the PM. This screening yielded 10 compounds out of the original 109 primary hits. Example 3 A: Screening with Hek293T cells expressing GHR Analogues of these 10 compounds were synthesized and 122 analogues were tested in Hek293T cells expressing GHR. GH-induced GH-GHR-Jak2 signaling was probed by GHR pY staining in concentration series (0-20 µM, 1 h drug-treated, 10 min GH). Based on data combined with again the GH/GM-CSF cytometric test, we selected 2 hits (Compounds BM001 and BM002) that both inhibited the phosphorylation of fos-GHRct (fos-pGHRct), of the active site of Jak2 (p1007/1008) and of STAT5 (pSTAT5) and were inactive towards GM-CSF signaling. Compound BM001 showed the same growth hormone receptor dependent potential (EC50 ~ 0,3 µM) as Ruxolitinib (EC50, 0.1-0.5 μM) (Furqan et al., 2013). Ruxolitinib is a Jak1/Jak2 inhibitor that is being used for the treatment of intermediate or high-risk myelofibrosis, a type of myeloproliferative disorder that affects the bone marrow, and for polycythemia vera when there has been an inadequate response to or intolerance of hydroxyurea. A set of 28 compounds according to formula 1 was then synthesized and tested for activity. Example 3B: Effect of compound BM001 on the in vitro translation of fos-zippered GHR cytosolic tails in an HeLa cell cell-free system. pcDNA3 vector DNA expressing Fos-zippered GHR cytosolic tails as described in Nespital et al, 2016 was used to show that the compounds act on protein translation at the ribosomal level. Using increasing concentrations reveals that compound BM001 is effective at concentrations > 0.1 µM. Translation of other proteins was not affected by the drugs (not shown). We used a cell-free translation system based on HeLa cell extract (Promega) Empty vector (pcDNA) was used as control. The blots were detected with an anti-GHR antibody raised in rabbits against the cytosolic tails. Example 4: Proliferation test We used the triple-negative human breast cancer line, MDA-MB-231 and quantified DNA in vital cells. Dose-response curves were made after 24, 48 & 72 h. The 28 compounds inhibited the cell line, with EC50 values varying from 30-2000 nM (Table 1). The compound with the lowest EC50 (Compound BM001) was tested in vivo. Example 5: In vivo test system in mice Mice were xenografted with MDA-MB-231 cells as described. This cell line was chosen as it is GH-responsive, Pegvisomant can block doxorubicin-induced apoptosis, it lacks E- cadherin due to hypermethylation, proliferates fast, has a basal-like expression profile and originates from infiltrating ductal breast cancer (Christgen and Derksen, 2015; Hollestelle et al., 2013; Minoia et al., 2012). Patients with this type of breast cancer, not driven by epidermal growth factor receptor 2 (HER-2), estrogen receptors (ER), or progesterone receptors (PR) have a poor prognosis. We used a protocol of 3 injections per week (1 and 5 mg/kg) for 3 weeks (9 mice/group). The treatment started after 5 weeks at a tumor volume of about 60 mm 3 , and the animals were killed after 3 weeks at a tumor volume of 130 mm 3 in the control group. The drug had an immediate effect on the tumors: growth halted, and the tumor sizes were reduced (Figure 2A and 2B). We also showed that the mice treated with compound BM001 had a decreased level of plasma IGF-1 (figure 2C) and a reduced liver weight (figure 2D). Neither control nor xenografted animals showed any discomfort during the 3-week treatments. Example 6: In vitro test system in dogs Canine mammary organoids were derived from tumor cells and cultured in Matrigel in the presence of a variety of growth factors, Wnt activators and MPA (Timmermans-Sprang et al., 2017). Organoids were treated for 48 h with 100 nM of compound BM001, followed by staining with DAPI (nuclei) or specific antibodies against GHR or cytokeratin 8 (CK8). The results are shown in figure 3. Example 7: Viability assay on MDA-MB-231 and MDA-MB-453 cells Compounds were assessed for effects on viability of the breast cancer cell lines MDA- MB-231 and MDA-MB-453. The cells were incubated with the compounds for 46 hours at a concentration of 5 uM. DMSO was used as vehicle control. Cell viability was assessed after approximately 46 hours by using a luminescent cell viability assay. This assay generates a luminescent signal, proportional to the amount of ATP that is present as a readout of metabolically active cells. Relative viability was calculated by expressing the luciferase signal as a value normalized to untreated cells, meaning that the measured luciferase signal in untreated cells equals 1. Example 8: Synthesis of small molecules. General scheme for the synthesis of vinylpyrimidine-2-4-diamines:

Scheme 1: a) 1-bromo-2-chloroethane; b) DIBAL-H; c) dimethyl (1-diazo-2-oxopropyl)phosphonate; d) tetrakisPPh3 palladium (0) : R = 4-Cl, 4-OMe, 4-OCF3, 2-CF3, 3-CF3, 4-morpholino. Synthesis of compound BM001. The synthesis of compound BM001 was performed as described in WO 98/20878.

Scheme 1: Total synthesis of compound BM001. a) 1-bromo-2-chloroethane; b) DIBAL-H; c) dimethyl (1-diazo-2-oxopropyl)phosphonate; d) ICl; e) tetrakisPPh3 palladium (0). In a further preferred embodiment, the invention provides a method to produce a 1-[4-(trifluoromethyl)phenyl]cyclopropane carbonitrile (2): A mixture of benzyl triethylammonium chloride (137mg, 0.58mmol), 1-bromo-2-chloroethane (3.65mL, 43.75mmol), 1 (5.40g, 29.17mmol) and NaOH (7.00g, 175mmol) in 7ml of water was stirred at 50°C under a nitrogen atmosphere. Additional water was added to facilitate stirring if needed. After 16h TLC (10% EtOAc in PE (40-60)) indicated complete consumption of the starting material. The reaction mixture was diluted with EtOAc and washed twice with water. The combined aqueous layers were extracted with EtOAc and the combined organic layers were washed with brine, dried over MgSO 4 and evaporated to give 2 (6.21g, 29.17mmol, 100%) as a red liquid which crystalizes on standing. ppm 7.62 (d, 2H), 7.40 (d, 2H), 1.69-1.97 (m, 2H), 1.33-1.53 (m, 2H). Herewith the invention provides a 1-[4-(trifluoromethyl)phenyl]cyclopropane carbonitrile. In a further preferred embodiment, the invention provides a method to produce a 1-[4- (trifluoromethyl)phenyl]cyclopropane carbaldehyde (3): To a solution of 2 (4.99g, 23.6mmol) in 20mL of DCM, cooled on an ice bath under a nitrogen atmosphere, DIBAL-H (1M in DCM) (5.04g, 35.5mL, 35.5mmol) was added dropwise. After the reaction mixture was stirred for 16h, TLC (10% EtOAc in PE (40-60)) indicated complete consumption of the starting material.40mL 1N HCl solution was added dropwise to the reaction mixture at 0°C. Additional DCM was added to facilitate stirring. The layers were separated, and the organic fraction was washed twice with water. The combined aqueous layers were extracted with DCM and the combined organic layers were washed with brine, dried over MgSO 4 and evaporated. The residue was purified by flash column chromatography (silica, 50%DCM/PE (40/60)) to give 3 (3.72g, 17.37mmol, 74%) as a yellow liquid which crystallizes on standing. 1 H NMR (400 MHz, CDCl 3 ) δ ppm 9.16 (s, 1H), 7.53- 7.68 (m, 2H), 7.30-7.50 (m, 2H), 1.54-1.69 (m, 2H), 1.33-1.52 (m, 2H). Herewith the invention provides a 1-[4-(trifluoromethyl)phenyl]cyclopropane carbaldehyde In a further preferred embodiment, the invention provides a method to produce a 1-(1- ethynylcyclopropyl)-4-(trifluoromethyl)benzene (4): To a solution of 3 (6.03g, 28.2mmol) in 5mL MeOH, cooled on an ice bath under a nitrogen atmosphere, a solution of dimethyl (1-diazo-2- oxopropyl)phosphonate (5.5mL, 36.6mmol) in 5mL MeOH was added followed by the addition of K 2 CO 3 (8.17g, 58.8mmol). After stirring 16h TLC (5% EtOAc in PE (40-60)) indicated complete consumption of the starting material. The reaction mixture was filtered over celite and the cake was washed with Et 2 O. The filtrate was washed twice with water. The combined aqueous layers were extracted with Et 2 O and the combined organic layers were washed with brine, dried over MgSO 4 and evaporated. The residue was purified by flash column chromatography (silica, PE (40- 60)) to give 4 (5.34g, 25.42mmol, 90%) as a colourless oil. 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.55 (m, 2H), 7.43 (m, 2H), 2.16 (s, 1H), 1.40-1.64 (m, 2H), 1.16-1.40 (m, 2H). Herewith the invention provides a 1-(1-ethynylcyclopropyl)-4-(trifluoromethyl)benzene In a further preferred embodiment, the invention provides a method to produce a [2-[1-[4- (trifluoromethyl)phenyl]cyclopropyl]vinyl]boronic acid (5): To a solution of 4 (1.55g, 7.38mmol) in 10mL THF a solution of catecholborane (0.97g, 0.87mL, 8.12 mmol) in 5mL THF was added and the resulting mixture was stirred for 6h at 70°C. TLC (30% EtOAc in PE (40-60)) indicated complete consumption of the starting material after which 50mL of water was added to the reaction mixture followed by stirring for 16h at room temperature. The reaction mixture was diluted with EtOAc and washed twice with water. The combined aqueous layers were extracted with EtOAc and the combined organic layers were washed with brine, dried over MgSO 4 , evaporated to give a mixture of catechol and 5 (2.32g) as a brown solid. 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.50-7.62 (m, 2H), 7.31-7.49 (m, 2H), 6.58 (d, 1H), 4.93 (d, 1H), 1.12-1.33 (m, 4H). Herewith the invention provides a [2-[1-[4-(trifluoromethyl)phenyl]cyclopropyl]vinyl]boronic acid In a further preferred embodiment, the invention provides a method to produce a 5-iodo-6- methyl-pyrimidine-2,4-diamine (6): To a solution of AB-323/25048181 (1.50g, 12.06mmol) in 15mL AcOH a solution of ICl (2.54g, 15.64mmol) in 15mL AcOH was added. The reaction mixture was stirred at room temperature for 3h after which TLC (EtOAc) indicated complete consumption of the starting material. The reaction was basified with a 4N NaOH solution and extracted twice with EtOAc. The combined organic layers are washed with brine, dried over MgSO 4 and evaporated to give 6 (2.70g, 10.80mmol, 89%) as a brown solid. 1 H NMR (400 MHz, CDCl 3 ) δ ppm 5.14 (br. s., 2H), 4.70 (br. s., 2H), 2.44 (s, 3H). Herewith the invention provides a 5-iodo-6-methyl-pyrimidine-2,4-diamine In a further preferred embodiment, the invention provides a method to produce a 6-methyl-5-[2- [1-[4-(trifluoromethyl)phenyl]cyclopropyl]vinyl]pyrimidine-2 ,4-diamine (7, compound BM001): 6 (264mg, 1.05 mmol) was added to a solution of 5 (456mg) in 15mL of THF and the reaction mixture is purged with nitrogen.5mL of a nitrogen purged stock solution of 2N Na 2 CO 3 solution was added to the reaction mixture. The air was evacuated under high vacuum and filled with a nitrogen atmosphere. This procedure was repeated twice after which tetrakisPPh3 palladium (0) (277mg, 0.24mmol) was added. The reaction was stirred at 70°C for 16h under a nitrogen atmosphere. TLC (20% i-PrOH/EtOAc) indicated complete consumption of starting material. The reaction mixture was diluted with EtOAc and washed twice with water. The combined aqueous layers were extracted with EtOAc and the combined organic layers were washed with brine, dried over MgSO 4 and evaporated. The residue was purified by column chromatography (silica, 20% i- PrOH/EtOAc) to give a mixed fraction of 7 (compound BM001) and 6, as a yellow solid. The mixture is dissolved in 5mL isopropanol and injected into 150mL demineralized water. The formed solids are filtered over a glass filter and dried to give pure 7 (compound BM001, 184mg, 0.55mmol, 46%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.61 (m, 2H), 7.47 (m, 2H), 5.71 (s, 1H), 5.66 (s, 1H), 4.64 (br. s., 2H), 4.57 (br. s., 2H), 2.13 (s, 3H), 1.17-1.32 (m, 2H), 1.05-1.17 (m, 2H). Herewith the invention provides a 6-methyl-5-[2-[1-[4- (trifluoromethyl)phenyl]cyclopropyl]vinyl]pyrimidine-2,4-dia mine Synthesis of 5-[2-[1-(4-chlorophenyl)cyclopropyl]vinyl]-6-methylpyrimidin e-2,4-diamine (GHR_0011) as a general example In a further preferred embodiment, the invention provides a method to produce a 1-(4- chlorophenyl)cyclopropanecarbonitrile: A mixture of benzyl triethylammoniumchloride (133mg, 0.58mmol), 1-bromo-2-chloroethane (6.29g, 43.83mmol), 2-(4-chlorophenyl)acetonitrile (4.43g, 29.22mmol) and NaOH (7.00g, 175mmol) in 7ml of water was stirred at 50°C under a nitrogen atmosphere. Additional water was added to facilitate stirring if needed. After 16h TLC (15% EtOAc in PE (40-60)) indicated complete consumption of the starting material. The reaction mixture was diluted with EtOAc and washed twice with water. The combined aqueous layers were extracted with EtOAc and the combined organic layers were washed with brine, dried over MgSO 4 and evaporated to give 1-(4- chlorophenyl)cyclopropanecarbonitrile (4.85g, 29.17mmol, 100%) as a red liquid which crystalizes on standing. 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.32 (d, 2H), 7.22 (d, 2H), 1.74 (m, 2H), 1.38 (m, 2H). Herewith the invention provides a 1-(4-chlorophenyl)cyclopropanecarbonitrile In a further preferred embodiment, the invention provides a method to produce a 1-[4- (trifluoromethoxy)phenyl]cyclopropanecarbonitrile: As above. From 2-(4-trifluoromethoxyphenyl)acetonitrile (4.36g, 21.68mmol) to yield 1-[4- (trifluoromethoxy)phenyl]cyclopropanecarbonitrile (4.08g, 17.96mmol, 83%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.33 (d, 2H), 7.22 (d, 2H), 1.71 (m, 2H), 1.40 (m, 2H). Herewith the invention provides a 1-[4-(trifluoromethoxy)phenyl]cyclopropanecarbonitrile In a further preferred embodiment, the invention provides a method to produce a 1-[2- (trifluoromethyl)phenyl]cyclopropanecarbonitrile: As above: From 2-(2-trifluoromethylphenyl)acetonitrile (5.00g, 27.01mmol) to yield 1-[2- (trifluoromethyl)phenyl]cyclopropanecarbonitrile (5.58g, 26.42mmol, 98%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.72-7.48 (m, 4H), 1.77 (m, 2H), 1.44 (m, 2H). Herewith the invention provides a 1-[2-(trifluoromethyl)phenyl]cyclopropanecarbonitrile In a further preferred embodiment, the invention provides a method to produce a 1-[3- (trifluoromethyl)phenyl]cyclopropanecarbonitrile: As above: From 2-(3-trifluoromethylphenyl)acetonitrile (5.00g, 27.01mmol) to yield 1-[3- (trifluoromethyl)phenyl]cyclopropanecarbonitrile (5.05g, 23.91mmol, 89%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.57-7.49 (m, 4H), 1.80 (m, 2H), 1.46 (m, 2H). Herewith the invention provides a 1-[3-(trifluoromethyl)phenyl]cyclopropanecarbonitrile In a further preferred embodiment, the invention provides a method to produce a 1-(4- morpholinophenyl)cyclopropanecarbonitrile: To a mixture of 18ml of concentrated H2SO4 and 18ml of 65% HNO3 solution, cooled on an ice bath and under a nitrogen atmosphere, was added neat 1-phenylcyclopropanecarbonitrile (10.00g, 69.84mmol). The reaction mixture was stirred for 15 minutes on ice, warmed to room temperature and stirred for an additional 30 minutes after which TLC (25% EtOAc in PE (40-60)) indicated complete consumption of the starting material. The reaction mixture was poured in water, the solids were filtered, washed with water, dried and recrystallized from EtOH to give 1- (4-nitrophenyl)cyclopropanecarbonitrile (9.47g, 50.32mmol, 72%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 8.21 (d, 2H), 7.43 (d, 2H), 1.91 (m, 2H), 1.54 (m, 2H). To a mixture of the above nitro compound (2.95g, 15.96mmol) in 50ml EtOH and 50ml water containing NH4Cl (8.52g, 159.6mmol) at 50°C under a nitrogen atmosphere was added portionwise iron powder (4.45g, 79.65mmol). The resulting reaction mixture was stirred for 45 minutes after which TLC (25% EtOAc in PE (40-60)) indicated complete consumption of the starting material. The reaction mixture was cooled, filtered over celite and the filter cake was washed with EtOH. The filtrate was evaporated and the residue was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc and the combined organic layers were washed with brine, dried over MgSO 4 and evaporated to give 1-(4-aminophenyl)cyclopropanecarbonitrile (2.27g, 14.35mmol, 90%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.09 (d, 2H), 6.64 (d, 2H), 3.70 (bs, 2H), 1.60 (m, 2H), 1.28 (m, 2H). A mixture of the above amine (2.25g, 14.22mmol), 1-chloro-2-(2-chloroethoxy)ethane (2.44g, 17.07mmol), sodium iodide (5.33g, 35.56mmol) and K2CO3 (3.93g, 28.45mmol) in 25ml NMP was stirred for 3h at 120°C under a nitrogen atmosphere after which TLC (25% EtOAc in PE (40-60)) indicated complete consumption of the starting material. The residue was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc and the combined organic layers were washed with brine, dried over MgSO 4 and evaporated. The residue was purified by column chromatography (silica, 5% EtOAc in DCM) to give 1-(4-morpholinophenyl)- cyclopropanecarbonitrile (2.60g, 11.39mmol, 80%) as a crystalline solid. NMR (400 MHz, CDCl 3 ) δ ppm 7.21 (d, 2H), 6.87 (d, 2H), 3.85 (d, 4H), 3.15 (d, 4H), 1.64 (m, 2H), 1.30 (m, 2H). Herewith the invention provides a 1-(4-morpholinophenyl)cyclopropanecarbonitrile In a further preferred embodiment, the invention provides a method to produce a 1-(4- chlorophenyl]cyclopropanecarbaldehyde: To a solution of 1-(4-chlorophenyl)cyclopropanecarbonitrile (2.48g, 13.96mmol) in 215mL of DCM, cooled on an ice bath under a nitrogen atmosphere, DIBAL-H (1M in DCM) (18mL, 18mmol) was added dropwise. After the reaction mixture was stirred for 16h, TLC (10% EtOAc in PE (40-60)) indicated complete consumption of the starting material.1N HCl solution (40ml) was added dropwise to the reaction mixture at 0°C. Additional DCM was added to facilitate stirring. The layers were separated, and the organic fraction was washed twice with water. The combined aqueous layers were extracted with DCM and the combined organic layers were washed with brine, dried over MgSO 4 and evaporated. The residue was purified by flash column chromatography (silica, 15% EtOAc in PE (40/60)) to give 1-(4- chlorophenyl]cyclopropanecarbaldehyde (2.10g, 11.63mmol, 83%) as a yellow oil which crystallizes on standing. 1 H NMR (400 MHz, CDCl 3 ) δ ppm 9.16 (s, 1H), 7.33 (d, 2H), 7.23 (d, 2H), 1.58 (m, 2H), 1.38 (m, 2H). Herewith the invention provides a 1-(4-chlorophenyl]cyclopropanecarbaldehyde In a further preferred embodiment, the invention provides a method to produce a 1-(4- methoxyphenyl)cyclopropanecarbaldehyde: As above. From 1-(4-methoxyphenyl)cyclopropanecarbonitrile (3.51g, 20.20mmol) to give 1-(4- methoxyphenyl)cyclopropanecarbaldehyde (2.69g, 15.27mmol, 76%) after purification by column chromatography (silica, 5% EtOAc in DCM). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 9.23 (s, 1H), 7.23 (d, 2H), 6.90 (d, 2H), 3.81 (s, 3H), 1.54 (m, 2H), 1.36 (m, 2H). Herewith the invention provides 1-(4-methoxyphenyl)cyclopropanecarbaldehyde In a further preferred embodiment, the invention provides a method to produce a 1-(4- trifluoromethoxyphenyl)cyclopropanecarbaldehyde: As above. From 1-(4-trifluoromethoxyphenyl)cyclopropanecarbonitrile (2.50g, 11.00mmol) to give 1-(4-methoxyphenyl)cyclopropanecarbaldehyde (2.05g, 8.91mmol, 81%) after purification by column chromatography (silica, 15% EtOAc in PE(40-60)). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 9.23 (s, 1H), 7.23 (d, 2H), 6.90 (d, 2H), 3.81 (s, 3H), 1.54 (m, 2H), 1.36 (m, 2H). Herewith the invention provides a 1-(4-trifluoromethoxyphenyl)cyclopropanecarbaldehyde In a further preferred embodiment, the invention provides a method to produce a 1-(2- trifluorophenyl)cyclopropanecarbaldehyde: As above. From 1-(2-trifluorophenyl)cyclopropanecarbonitrile (2.50g, 11.84mmol) to give 1-(2- trifluorophenyl)cyclopropanecarbaldehyde (1.83g, 8.54mmol, 72%) after purification by column chromatography (silica, 15% EtOAc in PE(40-60)). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 9.13 (s, 1H), 7.73-7.23 (m, 4H), 1.70 (m, 2H), 1.49 (m, 2H). Herewith the invention provides a 1-(2-trifluorophenyl)cyclopropanecarbaldehyde In a further preferred embodiment, the invention provides a method to produce a 1-(3- trifluorophenyl)cyclopropanecarbaldehyde: As above. From 1-(3-trifluorophenyl)cyclopropanecarbonitrile (2.50g, 11.84mmol) to give 1-(3- trifluorophenyl)cyclopropanecarbaldehyde (2.27g, 10.60mmol, 90%) after purification by column chromatography (silica, 5% EtOAc in DCM). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 9.15 (s, 1H), 7.58- 7.47 (m, 4H), 1.63 (m, 2H), 1.45 (m, 2H). Herewith the invention provides a 1-(3-trifluorophenyl)cyclopropanecarbaldehyde In a further preferred embodiment, the invention provides a method to produce a 1-(4- morpholinophenyl)cyclopropanecarbaldehyde: As above. From 1-(4-morpholinophenyl)cyclopropanecarbonitrile (2.10g, 9.20mmol) to give 1-(4- morpholinophenyl)cyclopropanecarbaldehyde (1.69g, 7.31mmol, 79%) after purification by column chromatography (silica, 10% EtOAc in DCM). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 9.25 (s, 1H), 7.21 (d, 2H), 6.90 (d, 2H), 3.86 (d, 4H), 3.16 (d, 4H), 1.53 (m, 2H), 1.35 (m, 2H). Herewith the invention provides 1-(4-morpholinophenyl)cyclopropanecarbaldehyde In a further preferred embodiment, the invention provides a method to produce a 1-chloro-4-(1- ethynylcyclopropyl)benzene: To a solution of 1-(4-chlorophenyl]cyclopropanecarbaldehyde (2.03g, 11.24mmol) in 8mL MeOH, cooled on an ice bath under a nitrogen atmosphere, a solution of dimethyl (1-diazo-2- oxopropyl)phosphonate (2.59g, 13.49mmol) in 5mL MeOH was added followed by the addition of K2CO3 (3.11g, 22.48mmol). After stirring 16h TLC (15% EtOAc in PE (40-60)) indicated complete consumption of the starting material. The reaction mixture was filtered over a celite cake and washed with MeOH. The filtrate was evaporated and diluted with EtOAc and washed with water, brine, dried over MgSO 4 and evaporated. The residue was purified by flash column chromatography (silica, PE (40-60)) to give 1-chloro-4-(1-ethynylcyclopropyl)benzene (576mg, 3.26mmol, 29%) as a colourless oil. 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.26 (s, 4H), 2.11 (s, 1H), 1.46 (m, 2H), 1.22 (m, 2H). Herewith the invention provides a 1-chloro-4-(1-ethynylcyclopropyl)benzene In a further preferred embodiment, the invention provides a method to produce a 1-(1- ethynylcyclopropyl)-4-methoxybenzene: As above. From 1-(4-methoxyphenyl)cyclopropanecarbaldehyde (2.69g, 15.30mmol) to give 1-(1- ethynylcyclopropyl)-4-methoxybenzene (2.51g, 14.57mmol, 95%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.27 (d, 2H), 6.84 (d, 2H), 3.79 (s, 3H), 2.06 (s, 1H), 1.39 (m, 2H), 1.17 (m, 2H). Herewith the invention provides 1-(1-ethynylcyclopropyl)-4-methoxybenzene In a further preferred embodiment, the invention provides a method to produce a 1-(1- ethynylcyclopropyl)-4-(trifluoromethoxy)benzene: As above. From 1-(4-trifluoromethoxyphenyl)cyclopropanecarbaldehyde (1.56g, 6.78mmol) to give 1-(1-ethynylcyclopropyl)-4-(trifluoromethoxy)benzene (1.48g, 6.54mmol, 97%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.35 (d, 2H), 7.14 (d, 2H), 2.12 (s, 1H), 1.48 (m, 2H), 1.23 (m, 2H). Herewith the invention provides a 1-(1-ethynylcyclopropyl)-4-(trifluoromethoxy)benzene In a further preferred embodiment, the invention provides a method to produce a 1-(1- ethynylcyclopropyl)-2-(trifluoromethyl)benzene: As above. From 1-(2-trifluorophenyl)cyclopropanecarbaldehyde (1.83g, 8.54mmol) to give 1-(1- ethynylcyclopropyl)-2-(trifluoromethyl)benzene (764mg, 3.55mmol, 42%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.66-7.38 (m, 4H), 1.93 (s, 1H), 1.45 (m, 2H), 1.25 (m, 2H). Herewith the invention provides a 1-(1-ethynylcyclopropyl)-2-(trifluoromethyl)benzene In a further preferred embodiment, the invention provides a method to produce a 1-(1- ethynylcyclopropyl)-3-(trifluoromethyl)benzene: As above. From 1-(3-trifluorophenyl)cyclopropanecarbaldehyde (2.27g, 10.60mmol) to give 1-(1- ethynylcyclopropyl)-3-(trifluoromethyl)benzene (1.77g, 8.42mmol, 79%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.58-7.40 (m, 4H), 1.99 (s, 1H), 1.46 (m, 2H), 1.25 (m, 2H). Herewith the invention provides 1-(1-ethynylcyclopropyl)-3-(trifluoromethyl)benzene In a further preferred embodiment, the invention provides a method to produce a 4-[4-(1- ethynylcyclopropyl)phenyl]morpholine: As above. From 1-(4-morpholinophenyl)cyclopropanecarbaldehyde (1.69g, 7.40mmol) to give 4- [4-(1-ethynylcyclopropyl)phenyl]morpholine (1.66g, 7.30mmol, 99%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.26 (d, 2H), 6.85 (d, 2H), 3.85 (d, 4H), 3.12 (d, 4H), 2.06 (s, 1H), 1.38 (m, 2H), 1.17 (m, 2H). Herewith the invention provides a 4-[4-(1-ethynylcyclopropyl)phenyl]morpholine In a further preferred embodiment, the invention provides a method to produce a [2-[1-(4- chlorophenyl)cyclopropyl]vinyl]boronic acid: To a solution of 1-chloro-4-(1-ethynylcyclopropyl)benzene (576mg, 3.26mmol) in 6mL THF a solution of catecholborane (430mg, 3.59mmol) in 2mL THF was added and the resulting mixture was stirred overnight at 70°C under a nitrogen atmosphere. After cooling, water was added and the reaction mixture was stirred for 3h at room temperature. The resulting solids were filtered and dried to give [2-[1-(4-chlorophenyl)cyclopropyl]vinyl]boronic acid (585mg, 2.63mmol, 81%) which was used directly in the next step. Herewith the invention provides a [2-[1-(4-chlorophenyl)cyclopropyl]vinyl]boronic acid In a further preferred embodiment, the invention provides a method to produce a [2-[1-(4- methoxyphenyl)cyclopropyl]vinyl]boronic acid: As above: From 1-(1-ethynylcyclopropyl)-4-methoxybenzene (1.51g, 8.74mmol). No precipitation was observed after the hydrolysis is water. The slurry was extracted into EtOAc, washed with brine, dried over MgSO 4 and evaporated to give a mixture of the boronic acid and catechol which was used directly in the next step. Herewith the invention provides a [2-[1-(4-methoxyphenyl)cyclopropyl]vinyl]boronic acid In a further preferred embodiment, the invention provides a method to produce a [2-[1-(4- trifluoromethoxyphenyl)cyclopropyl]vinyl]boronic acid: As above: From 1-(1-ethynylcyclopropyl)-4-trifluoromethoxybenzene (556mg, 2.46mmol). No precipitation was observed after the hydrolysis is water. The slurry was extracted into EtOAc, washed with brine, dried over MgSO 4 and evaporated to give a mixture of the boronic acid and catechol which was used directly in the next step. Herewith the invention provides a [2-[1-(4-trifluoromethoxyphenyl)cyclopropyl]vinyl]boronic acid In a further preferred embodiment, the invention provides a method to produce a [2-[1-(2- trifluoromethylphenyl)cyclopropyl]vinyl]boronic acid: As above: From 1-(1-ethynylcyclopropyl)-2-trifluoromethylbenzene (530mg, 2.52mmol). No precipitation was observed after the hydrolysis is water. The slurry was extracted into EtOAc, washed with brine, dried over MgSO 4 and evaporated to give a mixture of the boronic acid and catechol which was used directly in the next step. Herewith the invention provides a [2-[1-(2-trifluoromethylphenyl)cyclopropyl]vinyl]boronic acid In a further preferred embodiment, the invention provides a method to produce a [2-[1-(3- trifluoromethylphenyl)cyclopropyl]vinyl]boronic acid: As above: From 1-(1-ethynylcyclopropyl)-3-trifluoromethylbenzene (586mg, 2.88mmol). No precipitation was observed after the hydrolysis is water. The slurry was extracted into EtOAc, washed with brine, dried over MgSO 4 and evaporated to give a mixture of the boronic acid and catechol which was used directly in the next step. Herewith the invention provides a [2-[1-(3-trifluoromethylphenyl)cyclopropyl]vinyl]boronic acid In a further preferred embodiment, the invention provides a method to produce a [2-[1-(4- morpholinophenyl)cyclopropyl]vinyl]boronic acid: As above. From 4-[4-(1-ethynylcyclopropyl)phenyl]morpholine (571mg, 2.51mmol) to give [2-[1- (4-morpholinophenyl)cyclopropyl]vinyl]boronic acid (554mg, 2.03mmol, 81%) which was used directly in the next step. Herewith the invention provides a [2-[1-(4-morpholinophenyl)cyclopropyl]vinyl]boronic acid In a further preferred embodiment, the invention provides a method to produce a 5-[2-[1-(4- chlorophenyl)cyclopropyl]vinyl]-6-methylpyrimidine-2,4-diami ne (GHR_0011): [2-[1-(4-chlorophenyl)cyclopropyl]vinyl]boronic acid (585mg, 2.63mmol) was added to a solution of 5-iodo-6-methyl-pyrimidine-2,4-diamine (500mg, 2.00mmol) in 25mL of THF and the resulting reaction mixture is purged with nitrogen. A solution of 10ml 2N Na2CO3 solution was added, the air was evacuated under high vacuum and filled with a nitrogen atmosphere. This procedure was repeated twice, (PPh 3 ) 4 Pd(0) (575mg, 0.50mmol) was added and the reaction mixture was stirred at 70°C for 16h under a nitrogen atmosphere. The reaction mixture was diluted with EtOAc and washed twice with water. The combined aqueous layers were extracted with EtOAc and the combined organic layers were washed with brine, dried over MgSO 4 and evaporated. The residue was purified by column chromatography (silica, 20% i-PrOH/EtOAc), the fractions containing the product were pooled, evaporated, dissolved in a minimum amount of warm i-PrOH and injected into water. The solids were filtered, washed with water and dried to give 5-[2-[1-(4- chlorophenyl)cyclopropyl]vinyl]-6-methylpyrimidine-2,4-diami ne (340mg, 1.13mmol, 57%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.34 (s, 4H), 5.88-5.73 (m, 6H), 2.01 (s, 3H), 1.16 (m, 2H), 1.04 (m, 2H). Herewith the invention provides a 5-[2-[1-(4-chlorophenyl)cyclopropyl]vinyl]-6- methylpyrimidine-2,4-diamine (GHR_0011) In a further preferred embodiment, the invention provides a method to produce a 5-[2-[1-(4- methoxyphenyl)cyclopropyl]vinyl]-6-methylpyrimidine-2,4-diam ine (GHR_0003): As above. From the 4-methoxyboronic acid-catechol mixture (890mg), 5-iodo-6- methylpyrimidine-2,4-diamine (810mg, 3.27mmol) and (PPh3)4Pd(0) (473mg, 0.41mmol) to give 5- [2-[1-(4-methoxyphenyl)-cyclopropyl]vinyl]-6-methylpyrimidin e-2,4-diamine (200mg, 0.68mmol). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.29 (s, 4H), 6.88 (d, 1H), 5.62 (m, 1H), 4.68 (m, 4H), 3.81 (s, 3H), 2.13 (s, 3H), 1.20 (m, 2H), 1.14 (m, 2H). Herewith the invention provides a 5-[2-[1-(4-methoxyphenyl)cyclopropyl]vinyl]-6- methylpyrimidine-2,4-diamine (GHR_0003) In a further preferred embodiment, the invention provides a method to produce a 6-methyl-5-[2- [1-[4-(trifluoromethoxy)phenyl]cyclopropyl]vinyl]pyrimidine- 2,4-diamine (GHR_0016): As above. From the 4-trifluoromethoxyboronic acid-catechol mixture (650mg), 5-iodo-6- methylpyrimidine-2,4-diamine (418mg, 1.67mmol) and (PPh 3 ) 4 Pd(0) (552mg, 0.48mmol) to give 6- methyl-5-[2-[1-[4-(trifluoromethoxy)phenyl]cyclopropyl]vinyl ]pyrimidine-2,4-diamine (77mg, 0.22mmol). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.39-7.18 (m, 5H), 5.55 (dd, 1H), 4.66 (m, 4H), 2.13 (s, 3H), 1.18 (m, 2H), 1.08 (m, 2H). Herewith the invention provides a 6-methyl-5-[2-[1-[4- (trifluoromethoxy)phenyl]cyclopropyl]vinyl]pyrimidine-2,4-di amine (GHR_0016) In a further preferred embodiment, the invention provides a method to produce a 6-methyl-5-[2- [1-[2-(trifluoromethyl)phenyl]cyclopropyl]vinyl]pyrimidine-2 ,4-diamine (GHR_0043): As above. From the 2-trifluoromethylboronic acid-catechol mixture (708mg), 5-iodo-6- methylpyrimidine-2,4-diamine (380mg, 1.52mmol) and (PPh3)4Pd(0) (639mg, 0.55mmol) to give 6- methyl-5-[2-[1-[2-(trifluoromethyl)phenyl]cyclopropyl]vinyl] pyrimidine-2,4-diamine (206mg, 0.62mmol). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.69-7.40 (s, 4H), 5.54 (d, 1H), 5.32 (d, 1H), 4.71 (m, 4H), 2.16 (s, 3H), 1.31 (m, 2H), 1.16 (m, 2H). Herewith the invention provides a 6-methyl-5-[2-[1-[2- (trifluoromethyl)phenyl]cyclopropyl]vinyl]pyrimidine-2,4-dia mine (GHR_0043) In a further preferred embodiment, the invention provides a method to produce a 6-methyl-5-[2- [1-[3-(trifluoromethyl)phenyl]cyclopropyl]vinyl]pyrimidine-2 ,4-diamine (GHR_0013): As above. From the 3-trifluoromethylboronic acid-catechol mixture (736mg), 5-iodo-6- methylpyrimidine-2,4-diamine (431mg, 1.72mmol) and (PPh3)4Pd(0) (650mg, 0.58mmol) to give 6- methyl-5-[2-[1-[3-(trifluoromethyl)phenyl]cyclopropyl]vinyl] pyrimidine-2,4-diamine (54mg, 0.17mmol). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.62-7.36 (s, 4H), 5.71 (d, 1H), 5.62 (d, 1H), 4.61 (m, 4H), 2.12 (s, 3H), 1.21 (m, 2H), 1.11 (m, 2H). Herewith the invention provides a 6-methyl-5-[2-[1-[3- (trifluoromethyl)phenyl]cyclopropyl]vinyl]pyrimidine-2,4-dia mine (GHR_0013) In a further preferred embodiment, the invention provides a method to produce a 6-methyl-5-[2- [1-(4-morpholinophenyl)cyclopropyl]vinyl]pyrimidine-2,4-diam ine (GHR_0017): As above. From [2-[1-(4-morpholinophenyl)cyclopropyl]vinyl]boronic acid (554mg, 2.03mmol), 5- iodo-6-methylpyrimidine-2,4-diamine (380mg, 1.52mmol) and (PPh 3 ) 4 Pd(0) (468mg, 0.41mmol) to give 6-methyl-5-[2-[1-(4-morpholinophenyl)cyclopropyl]vinyl]pyrim idine-2,4-diamine (167mg, 0.48mmol, 32%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.18 (d, 2H), 6.87 (d, 2H), 5.70 (m, 6H), 3.71 (bs, 4H), 3.05 (bs, 4H), 1.98 (s, 3H), 1.06 (m, 2H), 0.97 (m, 2H). Herewith the invention provides a 6-methyl-5-[2-[1-(4- morpholinophenyl)cyclopropyl]vinyl]pyrimidine-2,4-diamine (GHR_0017) General scheme for the synthesis of ethynylpyrimidine-2-4-diamines: Scheme 2: a) bis(triphenylphosphine)palladium(II) chloride, Cu(I)I, piperidine : R = 4-CF3, 4-OCF3, 3- CF3, 4-morpholin o. Synthesis of 6-methyl-5-[2-[1-[4-(trifluoromethyl)phenyl]cyclopropyl]ethy nyl]pyrimidine-2,4- diamine as a general example In a further preferred embodiment, the invention provides a method to produce a 6-methyl-5-[2- [1-[4-(trifluoromethyl)phenyl]cyclopropyl]ethynyl]pyrimidine -2,4-diamine (GHR_0012): A mixture of 1-(1-ethynylcyclopropyl)-4-(trifluoromethyl)benzene (504mg, 2.40mmol), 5-iodo-6- methylpyrimidine-2,4-diamine (383mg, 1.53mmol), Cu(I)I (18mg, 0.10mmol) and piperidine (2.1ml, 26.86mmol) in 20ml DMF was degassed under high vacuum and purged with nitrogen. This procedure was repeated twice. Under a nitrogen atmosphere, bis(triphenylphosphine)palladium(II) chloride (93mg, 0.13mmol) was added and the resulting mixture was stirred over night at room temperature. The mixture was diluted with water, extracted twice with EtOAc and the combined organic layers were washed 3 times with 5% LiCl solution, dried over MgSO 4 , evaporated and purified by column chromatography (silica, 5% MeOH in DCM) to give 6-methyl-5-[2-[1-[4- (trifluoromethyl)phenyl]cyclopropyl]ethynyl]pyrimidine-2,4-d iamine (440mg, 1.32mmol, 87%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.57 (d, 2H), 7.47 (d, 2H), 5.10 (bs, 2H), 4.81 (bs, 2H), 2.38 (s, 3H), 1.61 (m, 2H), 1.42 (m, 2H). Herewith the invention provides 6-methyl-5-[2-[1-[4- (trifluoromethyl)phenyl]cyclopropyl]ethynyl]pyrimidine-2,4-d iamine (GHR_0012 In a further preferred embodiment, the invention provides a method to produce a 6-methyl-5-[2- [1-[4-(trifluoromethoxy)phenyl]cyclopropyl]ethynyl]pyrimidin e-2,4-diamine (GHR_0039): As above. From 1-(1-ethynylcyclopropyl)-4-(trifluoromethoxy)benzene (466mg, 2.06mmol), 5- iodo-6-methylpyrimidine-2,4-diamine (360mg, 1.44mmol), Cu(I)I (12mg, 0.06mmol), piperidine (2.75ml, 37.55mmol) and bis(triphenylphosphine)palladium(II) chloride (72mg, 0.10mmol) to give 6-methyl-5-[2-[1-[4-(trifluoromethoxy)phenyl]cyclopropyl]eth ynyl]pyrimidine-2,4-diamine (211mg, 0.61mmol, 42%). 1 H NMR (400 MHz, CDCl 3 ) δ ppm 7.38 (d, 2H), 7.16 (d, 2H), 5.16 (bs, 2H), 4.92 (bs, 2H), 2.37 (s, 3H), 1.54 (m, 2H), 1.36 (m, 2H). Herewith the invention provides a 6-methyl-5-[2-[1-[4- (trifluoromethoxy)phenyl]cyclopropyl]ethynyl]pyrimidine-2,4- diamine (GHR_0039) In a further preferred embodiment, the invention provides a method to produce a 6-methyl-5-[2- [1-[3-(trifluoromethyl)phenyl]cyclopropyl]ethynyl]pyrimidine -2,4-diamine (GHR_0042): As above. From 1-(1-ethynylcyclopropyl)-3-trifluoromethylbenzene (506mg, 2.41mmol), 5-iodo-6- methylpyrimidine-2,4-diamine (421mg, 1.69mmol), Cu(I)I (14mg, 0.07mmol), piperidine (2.5ml, 34.14mmol) and bis(triphenylphosphine)palladium(II) chloride (85mg, 0.12mmol) to give 6- methyl-5-[2-[1-[3-(trifluoromethyl)phenyl]cyclopropyl]ethyny l]pyrimidine-2,4-diamine (377mg, 1.13mmol, 67%). 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 7.76 (s, 1H), 7.65 (m, 3H), 6.25 (bs, 2H), 6.15 (s, 2H), 2.21 (s, 3H), 1.65 (m, 2H), 1.44 (m, 2H). Herewith the invention provides a 6-methyl-5-[2-[1-[3- (trifluoromethyl)phenyl]cyclopropyl]ethynyl]pyrimidine-2,4-d iamine (GHR_0042) In a further preferred embodiment, the invention provides a method to produce a 6-methyl-5-[2- [1-(4-morpholinophenyl)cyclopropyl]ethynyl]pyrimidine-2,4-di amine (GHR_0041): As above. From 4-[4-(1-ethynylcyclopropyl)phenyl]morpholine (423mg, 1.86mmol), 5-iodo-6- methylpyrimidine-2,4-diamine (326mg, 1.30mmol), Cu(I)I (11mg, 0.06mmol), piperidine (2.5ml, 34.14mmol) and bis(triphenylphosphine)palladium(II) chloride (65mg, 0.09mmol) to give 6- methyl-5-[2-[1-(4-morpholinophenyl)cyclopropyl]ethynyl]pyrim idine-2,4-diamine: (350mg, 1.00mmol, 77%). 1 H NMR (400 MHz, CDCl 3 ) 7.28 (d, 2H), 6.86 (d, 2H), 5.10 (bs, 2H), 4.79 (bs, 2H), 3.85 (t, 4H), 3.13 (t, 4H), 2.36 (s, 3H), 1.30 (m, 2H), 1.21 (m, 2H). Herewith the invention provides a a 6-methyl-5-[2-[1-(4- morpholinophenyl)cyclopropyl]ethynyl]pyrimidine-2,4-diamine (GHR_0041) In a further preferred embodiment, the invention provides a method to produce a 3-(2,4- diamino-5-methyl-quinazolin-6-yl)benzonitrile (GHR_0038). 2-amino-6-methylbenzonitrile (5.39g, 40.81mmol) was dissolved in 60ml of DMF at room temperature under a nitrogen atmosphere. To the reaction mixture was added dropwise a solution of N-bromosuccinimide (7.26g, 40.81mmol) in 60ml of DMF and stirring was continued over night after which TLC (silica, 25% EtOAc in PE 40/60) indicated complete consumption of the starting material. The reaction mixture was poured into 750ml of water and the resulting solids were filtered off, washed with water, and dried to give 6-amino-3-bromo-2-methylbenzonitrile (8.12g, 38.67mmol, 95%) as a solid. 1 H-NMR (CDCl 3 ): d 7.42 (d, 1H), 6.48 (d, 1H), 4.41 (bs, 2H), 2.52 (s, 3H). A solution of 6-amino-3-bromo-2-methylbenzonitrile (3.94g, 18.76mmol), (3-cyanophenyl)boronic acid (3.92g, 26.64mmol), 75ml of toluene and 20ml 2M Na 2 CO 3 -solution in a 250ml Schlenk-vessel was evacuated in vacuo and filled with nitrogen. This procedure was repeated three times. Under a stream of nitrogen tetrakis(triphenylphosphine)-palladium(0) (1.63g, 1.41mmol) was added and the reaction was heated to 100°C over night after which TLC (silica, 25% EtOAc in PE 40/60) indicated almost complete consumption of the starting material. The reaction mixture was cooled, diluted with EtOAc and washed twice with water. The combined aqueous layers were extracted twice with EtOAc and the combined organic layers were washed with brine, dried over MgSO 4 and evaporated. The remaining residue was triturated in 90% EtOH to give 6-amino-3-(3- cyanophenyl)-2-methylbenzonitrile (3.94g, 16.90mmol, 90%) as a solid. 1 H-NMR (DMSO-d 6 ): d 7.75 (m, 2H), 7.61 (m, 2H), 7.19 (d, 1H), 6.70 (d, 1H), 6.12 (bs, 2H), 2.25 (s, 3H). A solution of 6-amino-3-(3-cyanophenyl)-2-methylbenzonitrile (3.94g, 16.90mmol) and carbamimidic chloride hydrochloride (2.93g, 25.49mmol) in 75ml of diglyme was stirred over night at 165°C under a nitrogen atmosphere after which TLC (silica, 25% EtOAc in PE 40/60) indicated almost complete consumption of the starting material. The reaction mixture was cooled, ether was added, and the resulting solids were filtered off, suspended in 10ml of EtOH and added to 150ml of 1M NaOH-solution. After stirring for 4h, the solids were filtered off, washed with water and dried to give 3-(2,4-diamino-5-methylquinazolin-6-yl)benzonitrile (GHR_0038) (3.39g, 12.32mmol, 73%) as a solid.. 1 H-NMR (DMSO-d 6 ): d 8.20 (bs, 2H), 7.90 (m, 1H), 7.80 (s, 1H), 7.67 (m, 2H), 7.58 (d, 1H), 7.34 (m, 3H), 2.25 (s, 3H). Herewith the invention provides a 3-(2,4-diamino-5-methyl-quinazolin-6-yl)benzonitrile (GHR_0038) Example 9 Effects of a compound capable of selectively inhibiting stalling protein synthesis as provided herein on growth-inhibition of chemoresistant and non-resistant cancer-cell lines. Experiment performed with Oncolead GmbH (Germany) Experimental set up. As discussed above, Arumugam et al., (Experimental & Molecular Medicine (2019) 51:2 ) used malignant breast cancer cell lines MDAMB231 and MDAMB468 (available at ATCC) to demonstrate mechanisms of GHR-dependent or -assisted chemoresistance. Similarly, Basu et al., (HORM CANC DOI 10.1007/s12672-017-0292-7) report that human melanoma cell lines SKMEL28, SKMEL5 and MDAMB435 (available at ATCC) show mechanisms of GHR-dependent or -assisted chemoresistance. Yi et al., (Korean J Physiol Pharmacol Vol 16: 11-16, February 2012) show resistance (with increased IL-6-receptor expression) in non-small cell lung cancer (NSCLC) cell line NCI 460, and Morales et al., (Oncogene (2005) 24, 6842–6847) show resistance in colon cancer cell lines HCT116 and HT-29. In vitro profiling of compound BM001 in 92 cell lines (among which the cell lines mentioned above) was performed using six 10-fold compound dilutions prepared in DMSO. Treatment duration was 72 hours. Growth inhibition was measured by using Sulforhodamin B, a protein staining assay. Sulforhodamine B assay has been established and recommended by the DTP NCI/NIH (USA). IC50 was measured for all cells. Results (see also figure 10 a, b, and c) show that many of the collection of resistant cancer cells (with proven resistance to varied anti-cancer substances such as methotrexate, 5-urofluoacil, cisplatin, doxorubicin, oridonin, paclitaxel or vemurafenib), show clear sensitivity to the growth inhibitory anti-cancer activity of the molecule capable of selectively inhibiting stalling protein synthesis as provided herein, whereas non-cancer cells (such as PBMC and breast or muscle cell lines) were not significantly affected by said compound.

Example 10 LISTING OF POLYPEPTIDE SEQUENCE DETAILS OF 7 CYTOKINE RECEPTORS

REFERENCES Basu, R., Y. Qian, and J.J. Kopchick.2018. MECHANISMS IN ENDOCRINOLOGY: Lessons from growth hormone receptor gene disrupted mice: Are there benefits of endocrine defects? Eur J Endocrinol. Buchman, M., S. Bell, and J.J. Kopchick.2018. Growth Hormone Discovery and Structure. Pediatr Endocrinol Rev.16:2-10. Chhabra, Y., Waters, M.J., Brooks, A.J.2011. Role of the growth hormone–IGF-1 axis in cancer. Expert. Rev.Endocrinol. Metab.:71-84. Christgen, M., and P. Derksen.2015. Lobular breast cancer: molecular basis, mouse, and cellular models. Breast Cancer Res.17:16. Dehkhoda, F., C.M.M. Lee, J. Medina, and A.J. Brooks.2018. The Growth Hormone Receptor: Mechanism of Receptor Activation, Cell Signaling, and Physiological Aspects. Frontiers in endocrinology.9:35. Furqan, M., N. Mukhi, B. Lee, and D. Liu.2013. Dysregulation of JAK-STAT pathway in hematological malignancies and JAK inhibitors for clinical application. Biomarker research.1:5. Guevara-Aguirre, J., P. Balasubramanian, M. Guevara-Aguirre, Wei, F. Madia, C.W. Cheng, D. Hwang, A. Martin-Montalvo, J. Saaved , S. Ingles, R. de Cabo, P. Cohen, and V.D. Longo.2011. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Science translational medicine.3:70 . Hollestelle, A., J.K. Peeters, M. Smid, M. Timmermans, L.C. Verhoog, P.J. Westenend, A.A. Heine, A. Chan, A.M. Sieuwerts, E.A. Wiemer, J.G. Klijn, P.J. van der Spek, J.A. Foekens, M. Schutte, M.A. den Bakker, and J.W. Martens.2013. Loss of E-cadherin is not a necessity for epithelial to mesenchymal transition in human breast cancer. Breast Cancer Res Treat.138:47-57. Jessiman, A.G., D.D. Matson, and F.D. Moore.1959. Hypophysectomy in the treatment of breast cancer. N Engl J Med.261:1199-1207. Laron, Z.2015. Lessons from 50 Years of Study of Laron Syndrome. Endocrine practice: official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists.21:1395-1402. Lu, M., J.U. Flanagan, R.J. Langley, M.P. Hay, and J.K. Perry.2019. Targeting growth hormone function: strategies and therapeutic applications. Signal transduction and targeted therapy.4:3 Malergue, F., A. van Agthoven, C. Scifo, D. Egan, and G.J. Strous.2015. utomation of a phospho- STAT5 staining procedure for flow cytometry for application in drug discovery. J Biomol Screen. 20:416-421. Minoia, M., E. Gentilin, D. Mole, M. Rossi, C. Filieri, F. Tagliati, A. Baroni, M.R. Ambrosio, E. degli Uberti, and M.C. Zatelli.2012. Growth hormone receptor blockade inhibits growth hormone- induced chemoresistance by restoring cytotoxic-induced apoptosis in breast cancer cells independently of estrogen receptor expression. J Clin Endocrinol Metab.97:E907-916. Nashiro, K., J. Guevara-Aguirre, M.N. Braskie, G.W. Hafzalla, R. Velasco, P. Balasubramanian, M. Wei, P.M. Thompson, M. Mather M.D. Nelson, A. Guevara, E. Teran, and V.D. Longo.2017. Brain Structure and Function Associated with Younger Adults in Growth Hormone Receptor-Deficient Humans. J Neurosci. 37:1696-1707. Nespital, T., L.M. van der Velden, A. Mensinga, E.D. van der Vaart, and G.J. Strous.2016. Fos- Zippered GH Receptor Cytosolic Tails Act as Jak2 Substrates and Signal Transducers. Mol Endocrinol.30:290-301. Ranke, M.B., and J.M. Wit.2018. Growth hormone - past, present and future. Nat Rev Endocrinol. 14:285-300 http://www.ncbi.nlm.nih.gov/pubmed/29546874. Sedek, M., L.M. van der Velden, and G.J. Strous.2014. Multimeric growth hormone receptor complexes serve as signaling platforms. J. Biol. Chem.289:65-73. Theodoropoulou, M., and G.K. Stalla.2013. Somatostatin receptors: from signaling to clinical practice. Frontiers in neuroendocrinology.34:228-252. Timmermans-Sprang, E.P.M., A. Gracanin, and J.A. Mol.2017. Molecular Signaling of Progesterone, Growth Hormone, Wnt, and HE in Mammary Glands of Dogs, Rodents, and Humans: New Treatment Target Identification. Frontiers in veterinary science.4:53. Vermeulen, J.F., A.S. van Brussel, A. Adams, W.P. Mali, E. van der Wall, P.J. van Diest, and P.W. Derksen.2013. Near-infrared fluorescence molecular imaging of ductal carcinoma in situ with CD44v6-specific antibodies in mice: a preclinical study. Molecular imaging and biology: MIB : the official publication of the Academy of Molecular Imaging.15:290-298. Zhou, Y., B.C. Xu, H.G. Maheshwari, L. He, M. Reed, M. Lozykowski, S. Okada, L. Cataldo, K. Coschigamo, T.E. Wagner, G. Baumann, and J.J. Kopchick.1997. A mammalian model for Laron syndrome produced by targeted disruption of the mouse growth hormone receptor/binding protein gene (the Laron mouse). Proc Natl Acad Sci U S A.94:13215-13220.