Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ADMINISTRATION OF CD22, CD33 AND/OR TIM-3 ANTIBODIES USING OMMAYA RESERVOIRS FOR THE TREATMENT OF CNS CANCER OR NEURODEGENERATIVE DISEASES
Document Type and Number:
WIPO Patent Application WO/2021/083467
Kind Code:
A1
Abstract:
The present invention relates to a method for diagnosing, treating and/or monitoring response of a CNS cancer and treating, and further preventing and/or alleviating the symptoms of a neurodegenerative disease or condition in an individual, wherein said method comprises a step of administration of an antibody or antigen binding fragment thereof, and wherein said antibody or antigen binding fragment thereof is delivered into the central nervous system (CNS) using a device allowing intra-cerebroventricular administration, such as an Ommaya reservoir.

Inventors:
SEQUEIRA SONIA (US)
MAHIUDDIN AHMED (US)
J MØLLER SAN-PEDRO CLAUS (DK)
Application Number:
PCT/DK2020/050291
Publication Date:
May 06, 2021
Filing Date:
October 23, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
Y MABS THERAPEUTICS INC (US)
International Classes:
A61K39/00; C07K14/725; C07K16/28
Domestic Patent References:
WO2019126725A12019-06-27
WO2019191704A12019-10-03
WO2013163519A12013-10-31
WO2014160871A22014-10-02
WO2018200562A12018-11-01
WO2018204873A12018-11-08
WO2020172621A12020-08-27
WO2020025532A12020-02-06
WO2018200562A12018-11-01
WO2018204873A12018-11-08
Other References:
K KRAMER ET AL: "Intraventricular radioimmunotherapy targeting B7H3 for CNS malignancies", 2019 ANNUAL MEETING OF THE AMERICAN SOCIETY OF CLINICAL ONCOLOGY, ASCO 2019, vol. 37, 1 May 2019 (2019-05-01), XP055770938, DOI: 10.1200/JCO.2019.37.15-suppl.e13592
KIM KRAMER ET AL: "Safety profile of long-term intraventricular access devices in pediatric patients receiving radioimmunotherapy for central nervous system malignancies", PEDIATRIC BLOOD AND CANCER, vol. 61, no. 9, 29 April 2014 (2014-04-29), US, pages 1590 - 1592, XP055770936, ISSN: 1545-5009, DOI: 10.1002/pbc.25080
SMITH D B ET AL: "Quantitative distribution of 131I-labelled monoclonal antibodies administered by the intra-ventricular route", EUROPEAN JOURNAL OF CANCER AND CLINICAL ONCOLOGY, PERGAMON PRESS LTD, vol. 26, no. 2, 1 February 1990 (1990-02-01), pages 129 - 136, XP026214014, ISSN: 0277-5379, [retrieved on 19900201], DOI: 10.1016/0277-5379(90)90294-4
PLUVINAGE JOHN V ET AL: "CD22 blockade restores homeostatic microglial phagocytosis in ageing brains", NATURE, MACMILLAN JOURNALS LTD, LONDON, vol. 568, no. 7751, 3 April 2019 (2019-04-03), pages 187 - 192, XP036755246, ISSN: 0028-0836, [retrieved on 20190403], DOI: 10.1038/S41586-019-1088-4
MIRIAM Y. KIM ET AL: "Genetic Inactivation of CD33 in Hematopoietic Stem Cells to Enable CAR T Cell Immunotherapy for Acute Myeloid Leukemia", CELL, vol. 173, no. 6, 1 May 2018 (2018-05-01), Amsterdam NL, pages 1439 - 1453.e19, XP055568383, ISSN: 0092-8674, DOI: 10.1016/j.cell.2018.05.013
PLUVINAGE ET AL.: "CD22 blockade restores homeostatic microglial phagocytosis in ageing brains", NATURE, April 2019 (2019-04-01)
GOTTENBERG ET AL.: "Efficacy of Epratuzumab, an Anti-CD22 Monoclonal IgG Antibody, in Systemic Lupus Erythematosus Patients with Associated Sjogren's Syndrome", ARTHRITIS & RHEUMATOLOGY, vol. 70, no. 5, May 2018 (2018-05-01)
DOUGLAS ET AL.: "Association of CD33 Polymorphism rs3865444 with Alzheimer's Disease Pathology and CD33 Expression in Human Cerebral Cortex", NEUROBIOL AGING., February 2015 (2015-02-01)
GRICUIC ET AL.: "TREM2 Acts Downstream of CD33 in Modulating Microglia Pathology in Alzhimer's Disease", NEURON, 2019
TANIMOTO ET AL.: "Restricted expression of an early myeloid and monocytic cell surface antigen defined by monoclonal antibody M195", LEUKEMIA, May 1989 (1989-05-01)
PEREZ-OLIVIA ET AL.: "Epitope mapping, expression and post-translational modifications of two isoforms of CD33", GLYCOBIOLOGY, 2011
KOH ET AL.: "The HIF-l/glial TIM-3 axis controls inflammation associated brain damage under hypoxia", NATURE COMMUNICATIONS, 2015
HAUBNER ET AL.: "Coexpression profile of leukemic stem cell markers for combinatorial targeted therapy in AML", LEUKEMIA, 2019
KIM ET AL.: "Combination Therapy with Anti-PD-1, Anti-TIM-3, and Focal Radiation Results in Regression of Murine Gliomas", CLIN CANCER RES, 2017
LABRIJN AFMEESTERS JIDE GOEIJ BEVAN DEN BREMER ETNEIJSSEN JVAN KAMPEN MD ET AL.: "Efficient generation of stable bispecific IgGl by controlled Fab-arm exchange", PROC NATL ACAD SCI U S A, vol. 110, no. 13, 2013
Attorney, Agent or Firm:
POULSEN, Niels Jakob (DK)
Download PDF:
Claims:
Claims

1. A method for treating, preventing and/or alleviating the symptoms of a disorder affecting the central nervous system (CNS), such as a neurodegenerative condition and/or disease, an inflammatory disease or cancer, in particular a metastatic cancer, in a subject, wherein said method comprises a step of administration to said subject of an antibody or an antigen binding fragment thereof, and wherein said antibody or antigen binding fragment thereof is delivered into CNS using a device allowing or adapted to provide intra-cerebroventricular administration.

2. The method of claim 1, wherein said device comprises a catheter.

3. The method according to any of the preceding claims, wherein said device comprises a reservoir.

4. The method according to any of the preceding claims, wherein said device comprises an Ommaya reservoir.

5. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment thereof is conjugated or bound to a radioactive isotope.

6. A method for treating, preventing and/or alleviating the symptoms of a condition in a subject, wherein said condition is characterized by an antigen or antigen expression selected among TIM-3, CD22 and CD33, and wherein said method comprises a step of administering a therapeutically effective amount of an antibody or an antigen binding fragment thereof and wherein said antibody or antigen binding fragment thereof is delivered into the central nervous system (CNS) using a device allowing or adapted to provide intra-cerebroventricular administration.

7. The method according to any of the preceding claims, wherein said condition is characterized by overexpression of CD22 and/or CD33. 8. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment comprises an antibody component that binds to an antigen, wherein said antigen is selected among CD22 and/or CD33.

9. The method according to any of the preceding claims, wherein a neurodegenerative disease is treated, and said antibody or antigen binding fragment is IgG1 with inactive Fc or lgG4.

10. The method according to any of the preceding claims, wherein a neurodegenerative disease is treated, and said antibody or antigen binding fragment is IgG1 heterodimer with inactive Fc.

11. The method according to claim 10, wherein said IgG1 heterodimer with inactive Fc comprises one or more of the following mutations: N297A, K322A, K409R, and K405L. 12. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment is selected among single anti-CD22 IgGs, single anti-CD33, a heterodimer anti-CD33xanti-CD22, and a heterodimer anti-TIM-3xanti-CD33.

13. The method according to any of the preceding claims, wherein a neurodegenerative disease is treated, and said antibody or antigen binding fragment does not comprise active Fc.

14. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment is a CD22 x CD33 IgG heterodimer.

15. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment is tri-specific CD22 x CD33 x TIM-3.

16. The method according to any of the preceding claims, wherein a neurodegenerative disease is treated, and wherein said antibody or antigen binding fragment blocks signaling and is internalized, preferably wherein said antibody or antigen binding fragment comprises an antibody component that binds to an antigen, wherein said antigen is CD22.

17. The method according to any of the preceding claims, wherein a CNS cancer is treated with radioimmunotherapy.

18. The method according to any of the preceding claims, wherein a metastatic tumor in the CNS is treated with a bispecific antibody, wherein said bispecific antibody binds to the antigens CD33 and CD3. 19. The method according to any of the preceding claims, wherein a metastatic tumor in the CNS is treated with a bispecific antibody, wherein said bispecific antibody binds to the antigens CD22 and CD3.

20. The method according to any of the preceding claims, wherein a metastatic tumor in the CNS is treated with at least two bispecific antibodies, wherein said bispecific antibody each bind to at least two of the antigens CD22, CD33 and CD3.

21. The method according to any of the preceding claims, wherein a metastatic tumor in the CNS is treated with at least two bispecific antibodies, wherein said bispecific antibody each bind to at least two of the antigens TIM-3, CD33 and CD3.

22. The method according to any of the preceding claims, wherein a metastatic tumor in the CNS is treated, and wherein said antibody or antigen binding fragment comprises an antibody component that binds to an antigen, wherein said antigen is selected among CD22 and/or CD33, and wherein said antibody or antigen-binding fragment is bound or conjugated to a payload selected among a drug, a toxin and an isotope.

23. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment comprises at least one sequence selected among a heavy chain variable region CDR1 according to SEQ ID No. 38, 44 and 50, a heavy chain variable region CDR2 according to SEQ ID No. 39, 45 and 51, a heavy chain variable region CDR3 according to SEQ ID No. 40, 46 and 52, a light chain variable region CDR1 according to SEQ ID No. 35, 41 and 47, a light chain variable region CDR2 according to SEQ ID No. 36, 42 and 48 and a light chain variable region CDR3 according to SEQ ID No. 37, 43 and 49.

24. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment comprises a heavy chain sequence according to SEQ ID No. 12, 14 or 16 and/or a light chain sequence according to SEQ ID No. 11, 13 or 15.

25. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment comprises a heavy chain sequence that is at least about 80 %, about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 12, 14 or 16 and/or a light chain sequence that is at least about 80 %, about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. il, 13 or 15.

26. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment comprises at least one sequence selected among a heavy chain variable region CDR1 according to SEQ ID No. 20, 26 and 32, a heavy chain variable region CDR2 according to SEQ ID No. 21, 27 and 33, a heavy chain variable region CDR3 according to SEQ ID No. 22, 28 and 34, a light chain variable region CDR1 according to SEQ ID No. 17, 23 and 29, a light chain variable region CDR2 according to SEQ ID No. 18,24 and 30 and a light chain variable region CDR3 according to SEQ ID No. 19, 25 and 31.

27. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment comprises a heavy chain sequence according to SEQ ID No. 2, 4, 6, 8 or 10 and/or a light chain sequence according to SEQ ID No. 1, 3, 5, 7 or 9.

28. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment comprises a heavy chain sequence that is at least about 80 %, about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 2, 4, 6, 8 or 10 and/or a light chain sequence that is at least about 80 %, about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 1, 3, 5, 7 or 9.

29. The method according to any of the preceding claims, wherein said antigen binding fragment is a single chain variable fragment (scFv).

30. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment is a murine antibody or an antigen binding fragment thereof.

31. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment is a chimeric antibody or an antigen binding fragment thereof.

32. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment is a humanized antibody or an antigen binding fragment thereof.

33. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment is radiolabeled with a radioactive isotope.

34. The method according to claim 33, wherein said radioactive isotope is selected among a PET label and a SPECT label.

35. The method according to claim 34, wherein said PET label is selected among 124l, 225Ac and 89Zr.

36. The method according to claim 34, wherein said SPECT label is selected among 131l,

177Lu, 99mTc, 64Cu and 89Zr.

37. The method according to any of the preceding claims, wherein said antibody or antigen binding fragment thereof is conjugated to a chelator compound.

38. The method according to claim 37, wherein said chelator compound is bound to a radioactive isotope.

39. The method according to claim 38, wherein said radioactive isotope is selected among 124l, 131l and 177Lu or 99mTc, 64Cu and 89Zr.

40. The method according to any of the claims 37- 39, wherein said chelator compound is selected among DOTA, DTPA, NOTA and DFO.

41. The method according to claim 40, wherein said DOTA is a variant of DOTA, such as

Benzyl-DOTA.

42. The method according to claim 40, wherein said DTPA is a variant of DTPA, such as CHX-A"-DTPA.

43. The method according to any of the preceding claims, wherein said neurodegenerative disease is selected among dementia, a motor neuro disease, a lysosomal storage disease, a Parkinson's disease-related disorder, a prion disease and an ataxia.

44. The method according to any of the preceding claims, wherein said neurodegenerative disease is selected among Alzheimer's disease, vascular dementia, frontotemporal dementia, Lewy body dementia, amyotrophic lateral sclerosis, spinal muscular atrophy, Niemann-Pick type C56, Parkinson's disease, Creutzfeldt-Jakob disease, Huntington's Chorea and spinocerebellar ataxia.

45. The method according to any of the preceding claims, wherein said neurodegenerative disease is Alzheimer's disease. 46. The method according to any of the preceding claims, wherein said administration of said antibody or antigen binding fragment thereof is repeated with an interval of 1- 12 hours, 12-24 hours, 1-2 days, 2-3 days, 3-4 days, 4-5 days, 5-6 days, 6-7 days, 7-8 days, 8-9 days, 9-10 days, 10-11 days, 11-12 days, 12-13 days, 13-14 days, 14-15 days, 15-16 days, 16-17 days, 17-18 days, 18-19 days, 19-20 days, 20-21 days, 21-22 days, 22-23 days, 23-24 days, 24-25 days, 25-26 days, 26-27 days, 27-28 days, 28-29 days,

29-30 days, 30-31 days, 31-32 days, 32-33 days, 33-34 days, 34-35 days, 35-36 days, 36-37 days, 37-38 days, 38-39 days or 39-40 days.

47. The method according to any of the preceding claims, wherein said subject is a human subject.

48. The method according to any of the preceding claims, wherein said subject is an animal, preferably a mouse, a monkey or a rat.

49. The method according to any of the preceding claims, wherein said subject is older than 15 years, 17.5 years, 20 years, 22.5 years, 25 years, 27.5 years, 30 years, 32.5 years, 35 years, 37.5 years, 40 years, 42.5 years, 45 years, 47.5 years, 50 years, 52.5 years, 55 years, 57.5 years, 60 years, 62.5 years, 65 years, 67.5 years, 70 years, 72.5 years, 75 years, 77.5 years, 80 years, 82.5 years, 85 years, 87.5 years or older than 90 years.

50. The method according to any of the preceding claims for improving cognitive function and/or motoric function.

51. The method according to any of the preceding claims for increasing the phagocytosis in the CNS.

52. The method of claim 51, wherein said phagocytosis is performed by microglia cells.

53. The method according to any of the preceding claims for inhibiting the activity of a protein, wherein said protein is selected among CD22, CD33 and/or a sialic acid, preferably a-2,6-linked sialic acid. 54. The method according to claim 53, wherein said inhibition is preformed directly and/or indirectly.

55. The method according to any of the preceding claims, for reducing the amount of protein aggregates and/or cellular debris, such as myelin debris, amyloid-b, Oligomeric Ab and extracellular a-synuclein fibrils in the CNS.

56. An antibody or antigen binding fragment thereof, for use in a treatment according to any of the preceding claims. 57. An antibody or an antigen binding fragment thereof, wherein said antibody or antigen binding fragment thereof comprises at least one of the sequences selected among a heavy chain variable region CDR1 according to SEQ ID No. 38, 44 and 50, a heavy chain variable region CDR2 according to SEQ ID No. 39, 45 and 51, a heavy chain variable region CDR3 according to SEQ ID No. 40, 46 and 52, a light chain variable region CDR1 according to SEQ ID No. 35, 41 and 47, a light chain variable region CDR2 according to SEQ ID No. 36, 42 and 48 and a light chain variable region CDR3 according to SEQ ID No. 37,43 and 49.

58. The antibody or antigen binding fragment according to claim 57, wherein said antibody or antigen binding fragment thereof comprises a heavy chain sequence according to SEQ ID No. 12, 14 or 16 and a light chain sequence according to SEQ ID No. 11, 13 or 15.

59. The antibody or antigen binding fragment according to claim 57 or 58, wherein said antibody or antigen binding fragment thereof comprises a heavy chain sequence that is at least about 80 %,10 about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 12, 14 or 16 and/or a light chain sequence that is at least about 80 %, about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 11, 13 or 15.

60. An antibody or antigen binding fragment thereof, wherein said antibody or antigen binding fragment comprises at least one of the sequences selected among a heavy chain variable region CDR1 according to SEQ ID No. 20, 26 and 32, a heavy chain variable region CDR2 according to SEQ ID No. 21, 27 and 33, a heavy chain variable region CDR3 according to SEQ ID No. 22, 28 and 34, a light chain variable region CDR1 according to SEQ ID No. 17,23 and 29, a light chain variable region CDR2 according to SEQ ID No. 18, 24 and 30 and a light chain variable region CDR3 according to SEQ ID No. 19, 25 and 31.

61. The antibody or antigen binding fragment according to claim 60, wherein said antibody or antigen binding fragment thereof comprises a heavy chain sequence according to SEQ ID No. 2, 4, 6, 8 or 10 and a light chain sequence according to SEQ ID No. 1, 3, 5, 7 or 9.

62. The antibody or antigen binding fragment according to claim 60 or 61, wherein said antibody or antigen binding fragment thereof comprises a heavy chain sequence that is at least about 80 %,10 about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 2, 4, 6,

8 or 10 and/or a light chain sequence that is at least about 80 %, about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 1, 3, 5, 7 or 9.

63. The antibody or antigen binding fragment thereof according to claim 60 - 62, wherein said antibody or antigen binding fragment thereof comprises an antibody component that binds an antigen, wherein said antigen is selected among CD22 and/or CD33.

64. The antibody or antigen binding fragment thereof according to claim 56 - 63, wherein said antibody or antigen biding fragment thereof binds to CD22 and/or CD33 with an affinity having a KD value of less than 250, less than 240, less than 230, less than 220, less than 210, less than 200, less than 190, less than 180, less than 170, less than 160, less than 150, less than 140, less than 130, less than 120, less than 110, less than 100, less than 90, less than 80, less than 70, less than 60, less than 50, less than 40, less than 30, less than 20, less than 10, less than 9, less than 8, less than 7, less than 6, less than 5, less than 4, less than 3, less than 2, less than 1, or preferably less than

0.5.

65. The antibody or antigen biding fragment according to claim 57 - 64, for use in the method according to any of the preceding claims.

66. The antigen binding fragment according to claim 57 - 65, wherein said antigen binding fragment is a single chain variable fragment (scFv). 67. The antibody or antigen binding fragment according to claim 57 - 66, wherein said antibody or antigen binding fragment thereof is a murine antibody or antigen binding fragment thereof.

68. The antibody or antigen binding fragment according to claim 57 - 67, wherein said antibody or antigen binding fragment thereof is a chimeric antibody or antigen binding fragment thereof.

69. The antibody or antigen binding fragment thereof according to claim 57 - 68, wherein said antibody or antigen binding fragment is a humanized antibody or antigen binding fragment thereof.

70. The antibody or antigen binding fragment according to claim 57 - 69, wherein said antibody or antigen binding fragment thereof is radiolabeled with a radioactive isotope.

71. The antibody or antigen binding fragment according to claim 70, wherein said radioactive isotope is selected among a PET label and a SPECT label.

72. The antibody or antigen binding fragment according to claim 71, wherein said PET label is selected among 124l, 225Ac and 89Zr.

73. The antibody or antigen binding fragment according to claim 71, wherein said SPECT label is selected among 131l, 177Lu, 99mTc, 64Cu and 89Zr.

74. The antibody or antigen binding fragment according to claims 57 - 73, wherein said antibody or antigen binding fragment thereof is conjugated to a chelator compound. 75. The antibody or antigen binding fragment according to claim 74, wherein said chelator compound is bound to a radioactive isotope.

76. The antibody or antigen binding fragment according to claim 75, wherein said radioactive isotope is selected among 124l, 131l and 177Lu or 99mTc, 64Cu and 89Zr.

77. The antibody or antigen binding fragment according to claim 74 - 76, wherein said chelator compound is selected among DOTA, DTPA, NOTA and DFO.

78. The antibody or antigen binding fragment according to claim 77, wherein said DOTA is a variant of DOTA, such as Benzyl-DOTA.

79. The antibody or antigen binding fragment according to claim 77, wherein said DTPA is a variant of DTPA, such as CHX-A"-DTPA. 80. A bispecific binding antibody comprising a first antigen binding site, for binding to a first antigen, wherein said first antigen is selected among CD22 and/or CD33, and wherein said bispecific binding antibody further comprises a second antigen binding site, for binding to a second antigen.

81. The bispecific binding antibody of claim 80, wherein said first antigen is CD22.

82. The bispecific binding antibody of claim 80, wherein said first antigen is CD33.

83. The bispecific binding antibody of claim 80, wherein said bispecific binding antibody comprises a first antibody or antigen binding fragment thereof according to any of the claims 56 - 79, and a second antibody or antigen binding fragment for binding to a second antigen, and wherein said antibody is adapted for use in a method of treatment according to any of the preceding claims.

84. The bispecific binding antibody of claim 83, wherein said second antibody or antigen binding fragment thereof binds to DOTA.

85. A self-assembly disassembly (SADA) polypeptide, wherein said polypeptide is linked to an antibody or antigen binding fragment thereof according to claim 56 - 84, and wherein said polypeptide is for use in a method of treatment according to any of the preceding claims.

86. The self-assembly disassembly (SADA) polypeptide according to claim 85, wherein said self-assembly disassembly (SADA) polypeptide has an amino acid sequence that shows at least 75% identity with that of a human homo-multimerizing polypeptide and being characterized by one or more multimerization dissociation constants (KD).

87. A polypeptide conjugate, wherein said conjugate comprising the self-assembly disassembly (SADA) polypeptide of claim 85 or 86, and wherein said conjugate further comprises the bispecific antibody of claim 80 - 84, wherein said first antigen is CD22 and wherein said second antigen is DOTA.

88. A polypeptide conjugate, wherein said conjugate comprises the self-assembly disassembly (SADA) polypeptide of claim 85 or 86, and wherein said conjugate further comprises the bispecific antibody of claim 80 - 84, wherein said first antigen is CD33 and wherein said second antigen is DOTA.

89. A polypeptide conjugate comprising a self-assembly disassembly (SADA) polypeptide, and at least a first binding domain that binds to a first target and is covalently linked to the SADA polypeptide, wherein said polypeptide conjugate is for use in a method of treatment according to any of the preceding claims.

90. The polypeptide conjugate according to claim 89, wherein said self-assembly disassembly (SADA) polypeptide has an amino acid sequence that shows at least 75% identity with that of a human homo-multimerizing polypeptide and being characterized by one or more multimerization dissociation constants (KD); and wherein said conjugate is being constructed and arranged so that it adopts a first multimerization state and one or more higher-order multimerization states, wherein: the first multimerization state is less than about -70 kDa in size, at least one of the higher-order multimerization states is a homo-tetramer or higher-order homo- multimer greater than 150 kDa in size, wherein the higher-order homo-multimerized conjugate is stable in aqueous solution when the conjugate is present at a concentration above the SADA polypeptide KD, and the conjugate transitions from the higher-order multimerization state(s) to the first multimerization state under physiological conditions when the concentration of the conjugate is below the SADA polypeptide KD.

91. A chimeric antigen receptor (CAR) comprising an antibody or antigen binding fragment thereof according to any of the preceding claims, wherein said chimeric antigen receptor is used in the method according to any of the preceding claims.

92. A CAR-T cell expressing a CAR according to claim 91.

93. A population of CAR-T cells according to claim 92.

94. A composition comprising the population of CAR-T cells of claim 93.

95. A composition comprising the antibody or antigen binding fragment thereof according to any of the preceding claims.

96. The composition according to claim 95, wherein said composition is for use in the method according to any of the preceding claims.

97. Use of the composition according to any of the claims 94 - 96 in the manufacturing of a medicament for the treatment of a neurodegenerative disease and/or a method according to any of the preceding claims.

98. Use of the antibody or antigen binding fragment thereof of claim 56 - 79 for the manufacturing of a medicament for the treatment of a neurodegenerative disease or for a method of treatment according to any of the preceding claims.

99. An in vitro use of an antibody or antigen binding fragment thereof, for inhibiting the activity of a protein, wherein said protein is selected among CD22, CD33 and/or α- 2,6-linked sialic acid.

100. A kit of parts, wherein said kit comprises an antibody or antigen binding fragment thereof according to any of the preceding claims, and wherein said kit further comprises a device allowing intra-cerebroventricular administration of said antibody or antigen binding fragment thereof, and wherein said kit is adapted to be used in a method according to any of the preceding claims.

Description:
ADMINISTRATION OF CD22, CD33 AND/OR TIM-3 ANTIBODIES USING OMMAYA RESERVOIRS FOR THE TREATMENT OF CNS CANCER OR NEURODEGENERATIVE DISEASES

The present invention relates to administration of antibodies, such as CD22, CD33 and/or TIM-3 antibodies, into the central nervous system (CNS) using an Ommaya reservoir. In particular, the present invention relates to a method for diagnosing, treating and/or monitoring a cancer and/or treating, preventing and/or alleviating the symptoms of a neurodegenerative disease or condition in an individual, wherein said method comprises intra-cerebroventricular delivery via a device consisting of an indwelling catheter connected to an Ommaya reservoir, of a radiolabeled or non-radiolabeled antibody, bispecific antibody or antigen binding fragment thereof, to kill tumor cells expressing specific proteins or to block cell signaling activated by specific proteins involved in neuronal or microglial degeneration. The invention further relates to an IgG antibody or an antigen binding fragment thereof, and/or a bispecific antibody or antigen binding fragment thereof, for use as a single agent or in combination, for use in a method of the invention. In addition, the invention relates to a kit of parts adapted to carry out the inventive method. A particular utility comes from treating a neurodegenerative disease in an individual, wherein the treatment comprises a step of administration to said individual of an antibody or antigen binding fragment thereof according to the invention, and wherein said antibody or antigen biding fragment thereof is delivered into the CNS using a device comprising a catheter and an Ommaya reservoir.

Technical Background

Current research suggests that using multi-target therapies may overcome downregulation of antigens, tumor-escape mechanisms, tumor or disease heterogeneity and patient-to- patient heterogeneity as well as increase efficacy.

According to Pluvinage et al. (Pluvinage et al., CD22 blockade restores homeostatic microglial phagocytosis in ageing brains, Nature, April 2019) the function of microglia deteriorates in the ageing brain and in brains affected by neurodegenerative diseases. Microglia maintain homeostasis in the central nervous system through phagocytic clearance of protein aggregates and cellular debris, thus protecting the neurons of the central nervous system (CNS). Specifically, CD22, a sialic acid-binding immunoglobin-like-lectin (siglec) expressed on mature B cells in healthy individuals, is significantly upregulated in aged microglia; approximately three times more CD22 surface molecule than younger microglia, and comparable to the number of molecules on immature B-cells. CD22 has also been shown to be highly upregulated in mouse models of amyotrophic lateral sclerosis, a motor neuron disease, and Niemann-Pick type C56, a lysosomal storage disease.

Knockout of CD22 in the microglia-derived cell line BV2 cells promotes phagocytosis. Reversely, knockout of N-acylneuraminate cytidylyltransferase (CMAS), a key enzyme in sialic acid synthesis or PTPN6 (which codes for SHP-1) or removal of sialic acid by treatment with sialidase or 3FAX-Neu5Ac, an inhibitor of sialic acid synthesis, robustly promotes phagocytosis. Pluvinage et al. describes implantation of osmotic pumps in aged mice to continuously infuse a CD22 blocking antibody or an IgG control antibody directly into the cerebrospinal fluid for one month. CD22 mediates the anti-phagocytic effect of a2,6-linked sialic acid, and inhibition of CD22, either via antibody blockade or genetic ablation, promotes the clearance of myelin debris, Ab oligomers and a-synuclein fibrils in vivo. Long-term CD22 blockade partially reverses the transcriptional signature of age- and disease-related microglia, increases markers of neuronal activation, and, ultimately, improves cognitive function in aged mice. Future investigations of CD22 on microglia may inform, therapeutic strategies to treat or prevent neurodegenerative diseases such as Alzheimer's and Parkinson's disease via restoration of homeostasis in the CNS.

CD22 is overexpressed on B-ALL and B-cell lymphomas. Targeting systemic tumors with anti- CD22 antibodies and cell therapies has been extensively evaluated in oncology patients.

Current FDA approved CD22 immunotherapies include Moxetumomab pasudotox (Astrazeneca), an antibody-toxin conjugate approved for the treatment of hairy cell leukemia, Inotuximab ozogamicin (Pfizer), approved for CD19 resistant or relapsed B-ALL and ADC-Polatuzumab vedotin (Roche), approved for use together with rituximab and bendamustine for the treatment of B cell lymphoma. The CD22 humanized monoclonal antibody Epratuzumab by Immunomedics is currently under clinical development for non- Hodgkin's lymphoma as well as autoimmune diseases (Gottenberg et al. Efficacy of Epratuzumab, an Anti-CD22 Monoclonal IgG Antibody, in Systemic Lupus Erythematosus Patients with Associated Sjogren's Syndrome, Arthritis & Rheumatology Vol. 70, No. 5, May 2018).

CD33 is another Siglec expressed on cells of the myeloid immune lineage, including monocytes, macrophages, microglia, neutrophils, eosinophils, and dendritic cells. CD33 has been shown to regulate microglial phagocytosis and suppress amyloid-b (Ab) clearance in Alzheimer's disease. The CD33 can exist in two isoforms (a long form known as CD33M and truncated form known as CD33m). According to Douglas et al. (Douglas et al., Association of CD33 Polymorphism rs3865444 with Alzheimer's Disease Pathology and CD33 Expression in Human Cerebral Cortex, Neurobiol Aging., February 2015) it has been shown that the single nucleotide polymorphism rs3865444 results in these two isoforms, where the minor CD33m form has been associated with a decreased risk for developing Alzheimer's disease. Gricuic et al (Gricuic et al., TREM2 Acts Downstream of CD33 in Modulating Microglia Pathology in Alzhimer's Disease, Neuron, 2019) have shown that knockout of CD33 attenuated amyloid beta (Ab) pathology and improved cognition in 5xFAD mice. RNA-seq profiling of microglia revealed that genes related to phagocytosis and signaling (IL-6, IL-8, acute phase response) are upregulated in 5xFAD; CD33 -/- mice.

Targeting systemic tumors with anti-CD33 antibodies and cell therapies has been evaluated in oncology patients. The antibody drug conjugate gemtuzumab ozogamicin (based on murine clone M195) has been approved to treat AML patients. Other anti-CD33 clones have also been developed including M195 according to Tanimoto et al. (Tanimoto et al. Restricted expression of an early myeloid and monocytic cell surface antigen defined by monoclonal antibody M195, Leukemia, May 1989) and HIM3-4 according to Perez-Olivia et al., Epitope mapping, expression and post-translational modifications of two isoforms of CD33 (CD33M and CD33m on lymphoid and myeloid human cells, Glycobiology, 2011).

Antibodies targeting CD33 including the therapeutic treatment of leukemia, such as acute myeloid leukemia (AML), has been described in International Patent Application Publication No. W02018/200562.

The transmembrane T-cell immunoglobulin and mucin domain protein TIM-3 is a type I protein expressed by sub-types of lymphoid cells, such as lymphocytes Th1, Th17, Tc1, NK, as well as in myeloid cells. TIM-3 may act as a negative regulator of T lymphocyte activation, cytokine production, cell activation, and the capture of apoptotic bodies. TIM-3 has been associated to inflammation and subsequent brain damage after ischaemia. TIM-3 is highly expressed in hypoxic brain regions of a mouse cerebral hypoxia-ischaemia (H/l) model and distinctively upregulated in activated microglia and astrocytes, brain resident immune cells, in a hypoxia-inducible factor (HIF)-1-dependent manner. Notably, blockade of TIM-3 markedly reduces infarct size, neuronal cell death, edema formation and neutrophil infiltration in H/l mice (Koh et al., The HIF-1/glial TIM-3 axis controls inflammation associated brain damage under hypoxia, Nature Communications, 2015).

Haubner et al (Haubner et al., Coexpression profile of leukemic stem cell markers for combinatorial targeted therapy in AML, Leukemia, 2019) have suggested that combinatorial targeting of AML cells can enhance therapeutic efficacy without increasing toxicity. By generating a protein expression profile based on flow cytometry of primary AML (n = 356) , normal bone marrow samples (n = 34), and a an integrated normal tissue proteomic data set they analyzed antigen expression levels of CD33, CD123, CLL1, TIM3, CD244 and CD7 on AML bulk and leukemic stem cells at initial diagnosis (n = 302) and relapse (n = 54). CD33, CD123, CLL1, TIM3 and CD244 were ubiquitously expressed on AML bulk cells at initial diagnosis and relapse, irrespective of genetic characteristics. When they analyzed the coexpression of these targets in all dual combinations (n = 15), they found that CD33/TIM3 and CLL1/TIM3 were highly expressed in AML compared with normal hematopoiesis and non-hematopoietic tissues. Kim et al 2017 (Kim et al., Combination Therapy with Anti-PD-1, Anti-TIM-3, and Focal Radiation Results in Regression of Murine Gliomas, Clin Cancer Res. 2017), have also shown that combinatorial treatment with Anti-PD-1, Anti-TIM-3, and focal radiation results in regression of TIM-3 expressing gliomas in mice.

Summary of the invention

Existing treatment paradigms suffer inter alia from the drawback that access to the brain is limited by the blood brain barrier.

Neurodegenerative diseases and cancer share common cellular features such as inflammation and aberrant overexpression of immunomodulating proteins. CD33, CD22 and TIM-3 are transmembrane proteins that regulate self/non-self-protective mechanisms and promote favorable tumor growth conditions. CD33, CD22 and TIM-3 based immunotherapies have been explored for the systemic treatment of autoimmune diseases, B cell acute lymphoid leukemia (B-ALL) and B cell lymphomas however, to the best of our knowledge, such therapies have not been explored in the context of CNS disease in humans, whether neurodegenerative diseases or cancer. This is partially due to the limitations of safely accessing the CNS. Yet both pediatric and adult B-ALL as well as B-cell lymphomas relapse in the CNS and the current standard of care is prophylactic chemotherapy delivered intrathecally. We have shown that antibody delivery via intracerebroventricular delivery is feasible, safe and effective for the treatment of relapsed neuroblastoma to the CNS. There is a need for a method of treating CD22 and/or CD33 and/or TIM-3 CNS cancers such as metastatic B-ALL and/or lymphoma, and in particular parenchymal masses such as gliomas.

There is a need for a method of treating neurodegenerative diseases by targeting protein involved in the regulation of neuron-immune cell surveillance. CD22 and/or CD33 and/or TIM-3 are molecules that have been identified to play a role in this mechanism. Many pharmaceuticals have a limited access to the brain, because of the blood brain barrier. There is a need for a method of treating neurodegenerative diseases, that circumvent the blood brain barrier.

Targeting CD22, TIM-3 and CD33 using an antibody product that carries a drug/toxin/radioisotope payload administered via an Ommaya catheter for intra- cerebroventricular administration is a novel strategy to treat CD22 and CD33 positive tumors that have metastasized to the CNS.

Furthermore, targeting CD22, TIM-3 and CD33 using blocking antibodies administered via an Ommaya catheter for intra-cerebroventricular administration is a novel strategy to alleviate Azheimer's and other neurodegenerative diseases.

According to an aspect, the invention concerns a method for treating, preventing and/or alleviating the symptoms of a disorder affecting the central nervous system (CNS), such as a neurodegenerative condition and/or disease, and inflammatory disease or cancer, in particular a metastatic cancer, in a subject, wherein said method comprises a step of administration to said subject of an antibody or an antigen binding fragment thereof, and wherein said antibody or antigen binding fragment thereof is delivered into CNS using a device allowing or adapted to provide intra-cerebroventricular administration. According to another aspect, the invention concerns a method for treating, preventing and/or alleviating the symptoms of a condition in a subject, wherein said condition is characterized by an antigen selected among TIM-3, CD22 and CD33, and wherein said method comprises a step of administering a therapeutically effective amount of an antibody or an antigen binding fragment thereof and wherein said antibody or antigen binding fragment thereof is delivered into the central nervous system (CNS) using a device allowing or adapted to provide intra-cerebroventricular administration.

According to another aspect, the invention concerns an antibody or antigen binding fragment thereof, for use in a treatment of the invention.

According to another aspect, the invention concerns an antibody or an antigen binding fragment thereof, wherein said antibody or antigen binding fragment thereof comprises at least one of the sequences selected among a heavy chain variable region CDR1 according to SEQ ID No. 38, 44 and 50, a heavy chain variable region CDR2 according to SEQ ID No. 39, 45 and 51, a heavy chain variable region CDR3 according to SEQ ID No. 40, 46 and 52, a light chain variable region CDR1 according to SEQ ID No. 35, 41 and 47, a light chain variable region CDR2 according to SEQ ID No. 36, 42 and 48 and a light chain variable region CDR3 according to SEQ ID No. 37,43 and 49.

According to another aspect, the invention concerns an antibody or antigen binding fragment thereof, wherein said antibody or antigen binding fragment comprises at least one of the sequences selected among a heavy chain variable region CDR1 according to SEQ ID No. 20, 26 and 32, a heavy chain variable region CDR2 according to SEQ ID No. 21, 27 and 33, a heavy chain variable region CDR3 according to SEQ ID No. 22, 28 and 34, a light chain variable region CDR1 according to SEQ ID No. 17,23 and 29, a light chain variable region CDR2 according to SEQ ID No. 18, 24 and 30 and a light chain variable region CDR3 according to SEQ ID No. 19, 25 and 31.

According to another aspect, the invention concerns a bispecific binding antibody comprising a first antigen binding site, for binding to a first antigen, wherein said first antigen is selected among CD22 and/or CD33, and wherein said bispecific binding antibody further comprises a second antigen binding site, for binding to a second antigen. According to another aspect, the invention concerns a self-assembly disassembly (SADA) polypeptide, wherein said polypeptide is linked to an antibody or antigen binding fragment, and wherein said polypeptide is for use in a method of the invention.

According to another aspect, the invention concerns a polypeptide conjugate, wherein said conjugate comprising the self-assembly disassembly (SADA) polypeptide, and wherein said conjugate further comprises the bispecific antibody, wherein said first antigen is CD22 and wherein said second antigen is DOTA.

According to another aspect, the invention concerns a polypeptide conjugate, wherein said conjugate comprises the self-assembly disassembly (SADA) polypeptide, and wherein said conjugate further comprises the bispecific antibody, wherein said first antigen is CD33 and wherein said second antigen is DOTA.

According to another aspect, the invention concerns a polypeptide conjugate comprising a self-assembly disassembly (SADA) polypeptide, and at least a first binding domain that binds to a first target and is covalently linked to the SADA polypeptide, wherein said polypeptide conjugate is for use in a method of treatment according to the invention.

According to another aspect, the invention concerns a chimeric antigen receptor (CAR) comprising an antibody or antigen binding fragment thereof, wherein said chimeric antigen receptor is used in the method according to the invention.

According to another aspect, the invention concerns a CAR-T cell expressing a CAR. According to another aspect, the invention concerns a population of CAR-T cells.

According to another aspect, the invention concerns a composition comprising the population of CAR-T cells.

According to another aspect, the invention concerns a composition comprising the antibody or antigen binding fragment thereof of the invention. According to another aspect, the invention concerns use of the composition according to the invention in the manufacturing of a medicament for the treatment of a neurodegenerative disease and/or a method according to the invention.

According to another aspect, the invention concerns use of the antibody or antigen binding fragment thereof of according to the invention for the manufacturing of a medicament for the treatment of a neurodegenerative disease or for a method of treatment according to the invention.

According to another aspect, the invention concerns an in vitro use of an antibody or antigen binding fragment thereof, for inhibiting the activity of a protein, wherein said protein is selected among CD22, CD33 and/or a-2,6-linked sialic acid.

According to another aspect, the invention concerns a kit of parts, wherein said kit comprises an antibody or antigen binding fragment thereof according to the invention, and wherein said kit further comprises a device allowing intra-cerebroventricular administration of said antibody or antigen binding fragment thereof, and wherein said kit is adapted to be used in a method according to the invention.

Detailed Disclosure

In order to provide a clear and consistent understanding of the specification and claims, including the scope to be given such terms, the following definitions are provided.

Affinity: As is known in the art, "affinity" is a measure of the tightness with a particular ligand (e.g., an antibody) binds to its partner (e.g., an epitope). Affinities can be measured in difference ways.

Antibody: The term "antibody" is art-recognized terminology and is intended to include molecules or active fragments of molecules that bind to known antigens. Examples of active fragments of molecules that bind to known antigens include Fab and F(ab')2fragments.

These active fragments can be derived from an antibody of the present invention by a number of techniques. For example, purified monoclonal antibodies can be cleaved with an enzyme, such as pepsin, and subjected to HPLC gel filtration. The appropriate fraction containing Fab fragments can then be collected and concentrated by membrane filtration and the like. The term "antibody" also includes bispecific and chimeric antibodies and other available formats.

Antibody fragment: An antibody fragment is a portion of an antibody such as F(ab')2, F(ab)2,

Fab', Fab, Fv, sFv and the like. Regardless of structure, an antibody fragment binds with the same antigen that is recognized by the intact antibody. For example, an 3F8 monoclonal antibody fragment binds with an epitope recognized by 3F8. The term "antibody fragment" also includes any synthetic or genetically engineered protein that acts like an antibody by binding to a specific antigen to form a complex. For example, antibody fragments include isolated fragments consisting of the variable regions, such as the "Fv" fragments consisting of the variable regions of the heavy and light chains, recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker ("scFv proteins"), and minimal recognition units consisting of the amino acid residues that mimic the hypervariable region.

Bispecific antibody: A bispecific antibody is an antibody that can bind simultaneously to two targets which are of different structure. Bispecific antibodies (bsAb) and bispecific antibody fragments (bsFab) have at least one arm that specifically binds to an antigen, for example, GD2 and at least one other arm that specifically binds to another antigen, for example a targetable conjugate that bears a therapeutic or diagnostic agent. A variety of bispecific fusion proteins can be produced using molecular engineering. In one form, the bispecific fusion protein is divalent, consisting of, for example, a scFv with a single binding site for one antigen and a Fab fragment with a single binding site for a second antigen. In another form, the bispecific fusion protein is tetravalent, consisting of, for example, an IgG with two binding sites for one antigen and two identical scFv for a second antigen.

Chimeric antibody: A chimeric antibody is a recombinant protein that contains the variable domains including the complementarity-determining regions (CDRs) of an antibody derived from one species, for example a rodent antibody, while the constant domains of the antibody molecule is derived from those of a human antibody. The constant domains of the chimeric antibody may also be derived from that of other species, such as a cat or dog.

Effective amount: As used herein, the term "effective amount" refers to an amount of a given compound, conjugate or composition that is necessary or sufficient to realize a desired biologic effect. An effective amount of a given compound, conjugate or composition in accordance with the methods of the present invention would be the amount that achieves this selected result, and such an amount can be determined as a matter of routine by a person skilled in the art, without the need for undue experimentation.

Heterodimer: As used herein, the term "heterodimer" refers to a dimer formed by two different proteins. Humanized antibody: A humanized antibody is a recombinant protein in which the CDRs from an antibody from one species; e.g., a rodent antibody, is transferred from the heavy and light variable chains of the rodent antibody into human heavy and light variable domains. The constant domain of the antibody molecule is derived from those of a human antibody.

A human antibody may be an antibody obtained from transgenic mice that have been "engineered" to produce specific human antibodies in response to antigenic challenge. In this technique, elements of the human heavy and light chain locus are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci. The transgenic mice can synthesize human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas.

Prevent: As used herein, the terms "prevent", "preventing" and "prevention" refer to the prevention of the recurrence or onset of one or more symptoms of a disorder in a subject as result of the administration of a prophylactic or therapeutic agent.

Radioactive isotope: Examples of radioactive isotopes that can be conjugated to antibodies for use diagnostically or therapeutically include, but are not limited to, 211 At, 14 C, 51 Cr, 57 Co, 58 Co, 67 Cu, 152 Eu, 67 Ga, 3 H, 111 ln, 59 Fe, 212 Pb, 177 Lu, 32 P, 223 Ra, 224 Ra, 186 Re, 188 Re, 75 Se, 35 S, 99m Tc, 227 Th, 89 Zr, 90 Y, 123 l, 124 l, 125 l, 131 l, 94m Tc, 64 Cu, 68 Ga, 66 Ga, 76 Br, 86 Y, 82 Rb, 110m ln, 13 N, 11 C, 18 F and alpha-emitting particles. Non-limiting examples of alpha-emitting particles include

209 Bi, 211 Bi, 212 Bi, 213 Bi, 210 Po, 211 Po, 212 Po, 214 Po, 215 Po, 216 Po, 218 Po, 211 At, 215 At, 217 At, 218 At, 218 Rn, 219 Rn, 220 Rn, 222 Rn, 226 Rn, 221 Fr, 223 Ra, 224 Ra, 226 Ra, 225 Ac, 227 Ac, 227 Th, 228 Th, 229 Th, 230 Th, 232 Th, 231 Pa, 233 U, 234 U, 235 U, 236 U, 238 U, 237 Np, 238 Pu, 239 Pu, 240 Pu, 244 Pu, 241 Am, 244 Cm, 245 Cm, 248 Cm, 249 Cf, and 252 Cf.

Subject: By "subject" or "individual" or "animal" or "patient" or "mammal," is meant any subject, particularly a mammalian subject, for whom diagnosis, prognosis, or therapy is desired. Mammalian subjects include humans and other primates, domestic animals, farm animals, and zoo, sports, or pet animals such as dogs, cats, guinea pigs, rabbits, rats, mice, horses, cattle, cows, and the like.

Treatment: As used herein, the terms "treatment, " "treat, " "treated" or "treating" refer to prophylaxis and/or therapy, particularly wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder, such as the progression of multiple sclerosis. Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.

According to an embodiment, the invention concerns a method for treating, preventing and/or alleviating the symptoms of a disorder affecting the central nervous system (CNS), such as a neurodegenerative condition and/or disease, and inflammatory disease or cancer, in particular a metastatic cancer, in a subject, wherein said method comprises a step of administration to said subject of an antibody or an antigen binding fragment thereof, and wherein said antibody or antigen binding fragment thereof is delivered into CNS using a device allowing or adapted to provide intra-cerebroventricular administration.

According to an embodiment, the invention concerns the method, wherein said device comprises a catheter.

According to an embodiment, the invention concerns the method, wherein said device comprises a reservoir.

According to an embodiment, the invention concerns the method, wherein said device comprises an Ommaya reservoir.

An Ommaya reservoir is a device that is adapted to be placed under the scalp. It may be connected to a catheter, that may be placed in one of the ventricles in the brain. The Ommaya reservoir can be used for aspiration (taps) of cerebrospinal fluid (CSF) or for delivery of pharmaceuticals into the cerebrospinal fluid. An Ommaya reservoir may provide direct access to the cerebrospinal fluid, and may allow medication to bypass the blood-brain barrier. Ommaya reservoirs have been used to administer chemotherapy. An Ommaya reservoir is sometimes referred to as an Ommaya shunt. According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment thereof is conjugated or bound to a radioactive isotope.

According to an embodiment, the invention concerns a method for detecting, diagnosing, selecting individuals that will benefit from treatment, treating, preventing and/or alleviating the symptoms of a condition in a subject, wherein said condition is characterized by the overexpression of an antigen selected among TIM-3, CD22 and CD33, and wherein said method comprises a step of administering a therapeutically effective amount of an antibody or an antigen binding fragment thereof and wherein said antibody or antigen binding fragment thereof is delivered into the central nervous system (CNS) using a device allowing or adapted to provide intra-cerebroventricular administration.

According to an embodiment, the invention concerns the method, wherein said condition is characterized by overexpression of CD22 and/or CD33 and/or TIM-3.

According to an embodiment, protein overexpression may be detected by an immunohistochemistry method. Any of the antibodies and/or fragments of the invention may be used in this context. According to an embodiment, the methods of the invention may be used for imaging.

Overexpression may refer to synthesis of excess gene product. Overexpression is often due to the amplification or deregulation of the gene which encodes the gene product. Overexpression of certain proteins or other substances may play a role in e.g. cancer development.

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment comprises an antibody component that binds to an antigen, wherein said antigen is selected among CD22 and/or CD33 and/or TIM-3. A bispecific antibody may target CD22, CD33 and TIM-3 individually or in combination.

According to an embodiment, the invention concerns the method, wherein a neurodegenerative disease is treated, and said antibody or antigen binding fragment is IgG1 with inactive Fc or lgG4.

According to an embodiment, the invention concerns the method, wherein a neurodegenerative disease is treated, and said antibody or antigen binding fragment is IgG1 heterodimer with inactive Fc. According to an embodiment, the invention concerns the method, wherein said IgG1 heterodimer with inactive Fc comprises one or more of the following mutations N297A, K322A, K409R, and K405L.

Inactive Fc may be defined as not having FcgammaR binding and no complement C1q binding or having negligible FcgammaR binding and/or negligible complement C1q binding. IgG1 with inactive Fc can include N297A (aglycosylation) and K322A (no complement) mutations.

Duobody mutations may provide a heterodimer, one Fc has K409R, and the other Fc has K405L (se also Labrijn AF, Meesters J I, de Goeij BE, van den Bremer ET, Neijssen J, van Kampen MD, et al. Efficient generation of stable bispecific IgG1 by controlled Fab-arm exchange. Proc Natl Acad Sci U S A (2013) 110(13):5145-50.10.1073/pnas.1220145110.

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment is selected among single anti-CD22 IgGs, single anti-CD33, single TIM-3, a heterodimer anti-CD33xanti-CD22 and a heterodimer anti-TIM-3xanti-CD33. According to an embodiment, the invention concerns the method, wherein a neurodegenerative disease is treated, and said antibody or antigen binding fragment does not comprise active Fc.

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment is a CD22 x CD33 IgG heterodimer. The antibody may be a CD22 x CD33 IgG heterodimer, preferably without active-Fc. The antibody may be N297A or K322A. Alternatively, a TIM-3 x CD33 may be used.

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment is tri-specific CD22 x CD33 x TIM-3.

According to an embodiment, the invention concerns the method, wherein a neurodegenerative disease is treated, and wherein said antibody or antigen binding fragment blocks signaling and is internalized, preferably wherein said antibody or antigen binding fragment comprises an antibody component that binds to an antigen, wherein said antigen is CD22. According to an embodiment, the invention concerns the method, wherein a CNS cancer is treated with radioimmunotherapy.

According to an embodiment, the invention concerns the method, wherein a metastatic tumor in the CNS is treated with a bispecific antibody, wherein said bispecific antibody binds to the antigens CD33 and CD3.

According to an embodiment, the invention concerns the method, wherein a metastatic tumor in the CNS is treated with a bispecific antibody, wherein said bispecific antibody binds to the antigens CD22 and CD3.

According to an embodiment, the invention concerns the method, wherein a metastatic tumor in the CNS is treated with at least two bispecific antibodies, wherein said bispecific antibody each bind to at least two of the antigens CD22, CD33 and CD3.

According to an embodiment, the invention concerns the method, wherein a metastatic tumor in the CNS is treated with at least two bispecific antibodies, wherein said bispecific antibody each bind to at least two of the antigens TIM-3, CD33 and CD3.

According to an embodiment, the invention concerns the method, wherein a metastatic tumor in the CNS is treated, and wherein said antibody or antigen binding fragment comprises an antibody component that binds to an antigen, wherein said antigen is selected among CD22 and/or CD33, and wherein said antibody or antigen-binding fragment is bound or conjugated to a payload selected among a drug, a toxin and an isotope.

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment comprises at least one sequence selected among a heavy chain variable region CDR1 according to SEQ ID No. 38, 44 and 50, a heavy chain variable region CDR2 according to SEQ ID No. 39, 45 and 51, a heavy chain variable region CDR3 according to SEQ ID No. 40, 46 and 52, a light chain variable region CDR1 according to SEQ ID No. 35, 41 and 47, a light chain variable region CDR2 according to SEQ ID No. 36, 42 and 48 and a light chain variable region CDR3 according to SEQ ID No. 37, 43 and 49.

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment comprises a heavy chain sequence according to SEQ ID No. 12, 14 or 16 and/or a light chain sequence according to SEQ ID No. 11, 13 or 15. According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment comprises a heavy chain sequence that is at least about 80 %, about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 12, 14 or 16 and/or a light chain sequence that is at least about 80 %, about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 11, 13 or 15.

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment comprises at least one sequence selected among a heavy chain variable region CDR1 according to SEQ ID No. 20, 26 and 32, a heavy chain variable region CDR2 according to SEQ ID No. 21, 27 and 33, a heavy chain variable region CDR3 according to SEQ ID No. 22, 28 and 34, a light chain variable region CDR1 according to SEQ ID No. 17, 23 and 29, a light chain variable region CDR2 according to SEQ ID No. 18,24 and 30 and a light chain variable region CDR3 according to SEQ ID No. 19, 25 and 31.

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment comprises a heavy chain sequence according to SEQ ID No. 2, 4, 6,

8 or 10 and/or a light chain sequence according to SEQ ID No. 1, 3, 5, 7 or 9.

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment comprises a heavy chain sequence that is at least about 80 %, about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 2, 4, 6, 8 or 10 and/or a light chain sequence that is at least about 80 %, about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 1, 3, 5, 7 or 9. According to an embodiment, the invention concerns the method, wherein said antigen binding fragment is a single chain variable fragment (scFv).

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment is a murine antibody or an antigen binding fragment thereof. According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment is a chimeric antibody or an antigen binding fragment thereof.

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment is a humanized antibody or an antigen binding fragment thereof.

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment is radiolabeled with a radioactive isotope.

A radioactive isotope may be selected among 124 l, 225 Ac and 89 Zr. 131 l, 177 Lu, 99 mTc and 64 Cu.

According to an embodiment, the invention concerns the method, wherein said radioactive isotope is selected among a PET label and a SPECT label.

According to an embodiment, the invention concerns the method, wherein said PET label is selected among 124 l, 225 Ac and 89 Zr.

According to an embodiment, the invention concerns the method, wherein said SPECT label is selected among 131 l, 177 Lu, 99 mTc, 64 Cu and 89 Zr.

According to an embodiment, the invention concerns the method, wherein said antibody or antigen binding fragment thereof is conjugated to a chelator compound. According to an embodiment, the invention concerns the method, wherein said chelator compound is bound to a radioactive isotope.

According to an embodiment, the invention concerns the method, wherein said radioactive isotope is selected among 124 l, 131 l and 177 Lu or 99 mTc, 64 Cu and 89 Zr. According to another embodiment it may be selected among 124 l, 131 l and 177 Lu or 99 mTc, 64 Cu chelated to NOTA; and 89 Zr chelated to DFO.

According to an embodiment, the invention concerns the method, wherein said chelator compound is selected among DOTA, DTPA, NOTA and DFO. According to an embodiment, the invention concerns the method, wherein said DOTA is a variant of DOTA, such as Benzyl-DOTA.

According to an embodiment, the invention concerns the method, wherein said DTPA is a variant of DTPA, such as CHX-A"-DTPA. According to an embodiment, the invention concerns the method, wherein said disorder affecting the central nervous system (CNS) is a neurodegenerative condition and/or disease.

According to an embodiment, the invention concerns the method, wherein said disorder affecting the central nervous system (CNS) is an inflammatory disease.

According to an embodiment, the invention concerns the method, wherein said disorder affecting the central nervous system (CNS) is a cancer, in particular a metastatic cancer.

According to an embodiment, the invention concerns the method, wherein said neurodegenerative disease is selected among dementia, a motor neuro disease, a lysosomal storage disease, a Parkinson's disease-related disorder, a prion disease and an ataxia.

According to an embodiment, the invention concerns the method, wherein said neurodegenerative disease is selected among Alzheimer's disease, vascular dementia, frontotemporal dementia, Lewy body dementia, amyotrophic lateral sclerosis, spinal muscular atrophy, Niemann-Pick type C 56 , Parkinson's disease, Creutzfeldt-Jakob disease, Huntington's Chorea and spinocerebellar ataxia.

According to an embodiment, the invention concerns the method, wherein said neurodegenerative disease is Alzheimer's disease.

According to an embodiment, the invention concerns the method, wherein said administration of said antibody or antigen binding fragment thereof is repeated with an interval of 1- 12 hours, 12-24 hours, 1-2 days, 2-3 days, 3-4 days, 4-5 days, 5-6 days, 6-7 days, 7-8 days, 8-9 days, 9-10 days, 10-11 days, 11-12 days, 12-13 days, 13-14 days, 14-15 days, 15-16 days, 16-17 days, 17-18 days, 18-19 days, 19-20 days, 20-21 days, 21-22 days, 22-23 days, 23-24 days, 24-25 days, 25-26 days, 26-27 days, 27-28 days, 28-29 days, 29-30 days, 30-31 days, 31-32 days, 32-33 days, 33-34 days, 34-35 days, 35-36 days, 36-37 days, 37-38 days, 38-39 days or 39-40 days. According to an embodiment, the invention concerns the method, wherein said subject is a human subject.

According to an embodiment, the invention concerns the method, wherein said subject is an animal, preferably a mouse, a monkey or a rat. According to an embodiment, the invention concerns the method, wherein said subject is older than 15 years, 17.5 years, 20 years, 22.5 years, 25 years, 27.5 years, 30 years, 32.5 years, 35 years, 37.5 years, 40 years, 42.5 years, 45 years, 47.5 years, 50 years, 52.5 years, 55 years, 57.5 years, 60 years, 62.5 years, 65 years, 67.5 years, 70 years, 72.5 years, 75 years, 77.5 years, 80 years, 82.5 years, 85 years, 87.5 years or older than 90 years. According to an embodiment, the invention concerns the method for improving cognitive function and/or motoric function.

According to an embodiment, the invention concerns the method for increasing the phagocytosis in the CNS. Phagocytosis may be measured in vitro.

According to an embodiment, the invention concerns the method, wherein said phagocytosis is performed by microglia cells.

According to an embodiment, the invention concerns the method for inhibiting the activity of a protein, wherein said protein is selected among CD22, CD33 and/or a sialic acid, preferably a-2,6-linked sialic acid.

According to an embodiment, the invention concerns the method, wherein said inhibition is preformed directly and/or indirectly.

Sialic acid inhibits phagocytosis via CD22. CD22 is canonically expressed on B cells, where it negatively regulates B cell receptor signaling by binding sialic acid and recruiting SHP-1 or SHIP-1 via immunoreceptor tyrosine-based inhibitory mortifs (ITIMs). Removal of sialic acid by treatment with sialidase or 3F A x-Neu5Ac, an inhibitor of sialic acid synthesis, robustly promotes phagocytosis.

According to an embodiment, the invention concerns the method for reducing the amount of protein aggregates and/or cellular debris, such as myelin debris, amyloid-b, Oligomeric Ab and extracellular a-synuclein fibrils in the CNS. According to an embodiment, the invention concerns an antibody or antigen binding fragment thereof, for use in a treatment according to the invention.

According to an embodiment, the invention concerns an antibody or an antigen binding fragment thereof, wherein said antibody or antigen binding fragment thereof comprises at least one of the sequences selected among a heavy chain variable region CDR1 according to SEQ ID No. 38, 44 and 50, a heavy chain variable region CDR2 according to SEQ ID No. 39, 45 and 51, a heavy chain variable region CDR3 according to SEQ ID No. 40, 46 and 52, a light chain variable region CDR1 according to SEQ ID No. 35, 41 and 47, a light chain variable region CDR2 according to SEQ ID No. 36, 42 and 48 and a light chain variable region CDR3 according to SEQ ID No. 37, 43 and 49.

According to an embodiment, the invention concerns an antibody or antigen binding fragment, wherein said antibody or antigen binding fragment thereof comprises a heavy chain sequence according to SEQ ID No. 12, 14 or 16 and a light chain sequence according to SEQ ID No. 11, 13 or 15.

According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said antibody or antigen binding fragment thereof comprises a heavy chain sequence that is at least about 80 %,10 about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 12, 14 or 16 and/or a light chain sequence that is at least about 80 %, about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 11, 13 or 15.

According to an embodiment, the invention concerns an antibody or antigen binding fragment thereof, wherein said antibody or antigen binding fragment comprises at least one of the sequences selected among a heavy chain variable region CDR1 according to SEQ ID No. 20, 26 and 32, a heavy chain variable region CDR2 according to SEQ ID No. 21, 27 and 33, a heavy chain variable region CDR3 according to SEQ ID No. 22, 28 and 34, a light chain variable region CDR1 according to SEQ ID No. 17,23 and 29, a light chain variable region CDR2 according to SEQ ID No. 18, 24 and 30 and a light chain variable region CDR3 according to SEQ ID No. 19, 25 and 31.

According to an embodiment the invention concerns the antibody or antigen binding fragment, wherein said antibody or antigen binding fragment thereof comprises a heavy chain sequence according to SEQ ID No. 2, 4, 6, 8 or 10 and a light chain sequence according to SEQ ID No. 1, 3, 5, 7 or 9.

According to an embodiment the invention concerns the antibody or antigen binding fragment, wherein said antibody or antigen binding fragment thereof comprises a heavy chain sequence that is at least about 80 %,10 about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID No. 2, 4, 6, 8 or 10 and/or a light chain sequence that is at least about 80 %, about 81 %, about 82 %, about 83 %, about 84 %, about 85 %, about 86 %, about 87 %, about 88 %, about 89 %, about 90 %, about 91 % about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 % or about 99% sequence identity to the sequence set forth in SEQ ID Nol, 3, 5, 7 or 9.

According to an embodiment, the invention concerns the antibody or antigen binding fragment thereof, wherein said antibody or antigen binding fragment thereof comprises an antibody component that binds an antigen, wherein said antigen is selected among CD22 and/or CD33.

According to an embodiment, the invention concern the antibody or antigen binding fragment thereof, wherein said antibody or antigen biding fragment thereof binds to CD22 and/or CD33 with an affinity having a KD value of less than 250, less than 240, less than 230, less than 220, less than 210, less than 200, less than 190, less than 180, less than 170, less than 160, less than 150, less than 140, less than 130, less than 120, less than 110, less than 100, less than 90, less than 80, less than 70, less than 60, less than 50, less than 40, less than 30, less than 20, less than 10, less than 9, less than 8, less than 7, less than 6, less than 5, less than 4, less than 3, less than 2, less than 1, or preferably less than 0.5. Binding affinity is measured as the equilibrium dissociation constant KD. KD is a ration of Koff/Kon, between the antibody and its antigen. The smaller the KD, the greater the binding affinity of the ligand for its target.

According to an embodiment, the invention concerns the antibody or antigen biding fragment, for use in the method according to the invention.

According to an embodiment, the invention concerns the antigen binding fragment, wherein said antigen binding fragment is a single chain variable fragment (scFv).

According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said antibody or antigen binding fragment thereof is a murine antibody or antigen binding fragment thereof.

According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said antibody or antigen binding fragment thereof is a chimeric antibody or antigen binding fragment thereof.

According to an embodiment, the invention concerns the antibody or antigen binding fragment thereof, wherein said antibody or antigen binding fragment is a humanized antibody or antigen binding fragment thereof.

According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said antibody or antigen binding fragment thereof is radiolabeled with a radioactive isotope.

According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said radioactive isotope is selected among a PET label and a SPECT label.

According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said PET label is selected among 124 l, 225 Ac and 89 Zr.

According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said SPECT label is selected among 131 l, 177 Lu, 99 mTc, 64 Cu and 89 Zr.

According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said antibody or antigen binding fragment thereof is conjugated to a chelator compound. According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said chelator compound is bound to a radioactive isotope.

According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said radioactive isotope is selected among 124 l, 131 l and 177 Lu or 99 mTc, 64 Cu and 89 Zr. Alternatives include 124 l, 131 l and 177 Lu or 99 mTc, 64 Cu chelated to NOTA; and 89 Zr chelated to DFO.

According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said chelator compound is selected among DOTA, DTPA, NOTA and DFO.

According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said DOTA is a variant of DOTA, such as Benzyl-DOTA.

According to an embodiment, the invention concerns the antibody or antigen binding fragment, wherein said DTPA is a variant of DTPA, such as CFIX-A"-DTPA.

According to an embodiment, the invention concerns a bispecific binding antibody comprising a first antigen binding site, for binding to a first antigen, wherein said first antigen is selected among CD22 and/or CD33, and wherein said bispecific binding antibody further comprises a second antigen binding site, for binding to a second antigen.

According to an embodiment, the invention concerns the bispecific binding antibody, wherein said first antigen is CD22.

According to an embodiment, the invention concerns the bispecific binding antibody, wherein said first antigen is CD33.

According to an embodiment, the invention concerns the bispecific binding antibody, wherein said bispecific binding antibody comprises a first antibody or antigen binding fragment thereof, and a second antibody or antigen binding fragment for binding to a second antigen, and wherein said antibody is adapted for use in a method of treatment according to the invention.

According to an embodiment, the invention concerns the bispecific binding antibody, wherein said second antibody or antigen binding fragment thereof binds to DOTA.

According to an embodiment, the invention concerns a self-assembly disassembly (SADA) polypeptide, wherein said polypeptide is linked to an antibody or antigen binding fragment thereof, and wherein said polypeptide is for use in a method of treatment according to the invention.

According to an embodiment, the invention concerns the self-assembly disassembly (SADA) polypeptide, wherein said self-assembly disassembly (SADA) polypeptide has an amino acid sequence that shows at least 75% identity with that of a human homo-multimerizing polypeptide and being characterized by one or more multimerization dissociation constants (KD).

According to an embodiment, the invention concerns a polypeptide conjugate, wherein said conjugate comprising the self-assembly disassembly (SADA) polypeptide, and wherein said conjugate further comprises the bispecific antibody, wherein said first antigen is CD22 and wherein said second antigen is DOTA.

According to an embodiment, the invention concerns a polypeptide conjugate, wherein said conjugate comprises the self-assembly disassembly (SADA) polypeptide, and wherein said conjugate further comprises the bispecific antibody, wherein said first antigen is CD33 and wherein said second antigen is DOTA.

According to an embodiment, the invention concerns a polypeptide conjugate comprising a self-assembly disassembly (SADA) polypeptide, and at least a first binding domain that binds to a first target and is covalently linked to the SADA polypeptide, wherein said polypeptide conjugate is for use in a method of treatment according to the invention.

According to an embodiment, the invention concerns the polypeptide conjugate, wherein said self-assembly disassembly (SADA) polypeptide has an amino acid sequence that shows at least 75% identity with that of a human homo-multimerizing polypeptide and being characterized by one or more multimerization dissociation constants (KD); and wherein said conjugate is being constructed and arranged so that it adopts a first multimerization state and one or more higher-order multimerization states, wherein: the first multimerization state is less than about -70 kDa in size, at least one of the higher- order multimerization states is a homo-tetramer or higher-order homo-multimer greater than 150 kDa in size, wherein the higher-order homo-multimerized conjugate is stable in aqueous solution when the conjugate is present at a concentration above the SADA polypeptide KD, and the conjugate transitions from the higher-order multimerization state(s) to the first multimerization state under physiological conditions when the concentration of the conjugate is below the SADA polypeptide KD.

In certain embodiments, the antibody of antigen binding fragment thereof is linked to a self- assembly disassembly (SADA) polypeptide disclosed in International Patent Application Publication No. WO2018204873, all of which is incorporated by reference in its entirety.

According to an embodiment, the invention concerns a chimeric antigen receptor (CAR) comprising an antibody or antigen binding fragment thereof, wherein said chimeric antigen receptor is used in the method according to the invention. According to an embodiment, the invention concerns a CAR-T cell expressing a CAR.

According to an embodiment, the invention concerns a population of CAR-T cells.

According to an embodiment, the invention concerns a composition comprising the population of CAR-T cells.

According to an embodiment, the invention concerns a composition comprising the antibody or antigen binding fragment thereof according to the invention.

According to an embodiment, the invention concerns the composition, wherein said composition is for use in the method according to the invention.

According to an embodiment, the invention concerns use of the composition of the invention in the manufacturing of a medicament for the treatment of a neurodegenerative disease and/or a method according to the invention.

According to an embodiment, the invention concerns use of the antibody or antigen binding fragment thereof for the manufacturing of a medicament for the treatment of a neurodegenerative disease or for a method of treatment according to the invention.

According to an embodiment, the invention concerns an in vitro use of an antibody or antigen binding fragment thereof, for inhibiting the activity of a protein, wherein said protein is selected among CD22, CD33 and/or a-2,6-linked sialic acid.

According to an embodiment, the invention concerns a kit of parts, wherein said kit comprises an antibody or antigen binding fragment thereof, and wherein said kit further comprises a device allowing intra-cerebroventricular administration of said antibody or antigen binding fragment thereof, and wherein said kit is adapted to be used in a method according to the invention.

According to an embodiment, the invention concerns a CD22 based immunotherapy for the treatment of a pediatric cancer expressing CD22. According to an embodiment, the invention concerns the antibody, wherein said antibody is a bispecific antibody combined with an exogenous T-cell (armed T-cell).

According to an embodiment, the invention concerns the bispecific antibody, wherein said antibody activates resident T-cells in the brain.

According to an embodiment, the invention concerns the method, wherein said method comprises administration of a CD22 x CD33 bispecific IgG antibody for dual target blockade of signaling, wherein said method is for use in a method according to the invention.

According to an embodiment, the invention concerns the antibody, wherein said antibody binds to an antigen, wherein said antigen is CD20.

According to an embodiment, the invention concerns the method, wherein said method comprises treatment of a neurodegenerative disease with an antibody that blocks signaling and is internalized.

According to an embodiment, the invention concerns the method, wherein said method comprises treatment of a CNS cancer, wherein said antibody is labeled with a radioactive isotope. According to an embodiment, the invention concerns the method, wherein said method comprises treatment of a CNS cancer, wherein said antibody is binding to an antigen, wherein said antigen is CD22 or CD33.

According to an embodiment, the invention concerns the method, wherein said method comprises treatment of a CNS cancer, wherein said antibody is an IgG or comprises a self- assembly disassembly (SADA) polypeptide.

All cited references are incorporated by reference. The accompanying Examples are provided to explain rather than limit the present invention. It will be clear to the person skilled in the art that aspects, embodiments, claims and any items of the present invention may be combined.

Unless otherwise mentioned, all percentages are in weight/weight. Unless otherwise mentioned, all measurements are conducted under standard conditions (ambient temperature and pressure). Unless otherwise mentioned, test conditions are according to European Pharmacopoeia 8.0.

Examples

Safety and efficacy of the treatment may be shown, e.g. initially by an animal model and by determination of in vitro binding. Safety may be shown by cross-reactivity to normal CNS tissue, and further biodistribution may be investigated.

Table 1. VL and VH sequences of antibody products that can be administered to target CD22/CD33 via Ommaya catheter for intra-cerebroventricular administration Table 2. IMGT CDR sequences of antibody products that can be administered to target CD22/CD33 via Ommaya catheter for intra-cerebroventricular administration