EP3509636A1 - Réduction des quantités ou de l'activité de cellules t régulatrices systémiques pour le traitement de maladie et de lésion du snc - Google Patents

Réduction des quantités ou de l'activité de cellules t régulatrices systémiques pour le traitement de maladie et de lésion du snc

Info

Publication number
EP3509636A1
EP3509636A1 EP17848290.7A EP17848290A EP3509636A1 EP 3509636 A1 EP3509636 A1 EP 3509636A1 EP 17848290 A EP17848290 A EP 17848290A EP 3509636 A1 EP3509636 A1 EP 3509636A1
Authority
EP
European Patent Office
Prior art keywords
antibody
pharmaceutical composition
disease
use according
treatment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP17848290.7A
Other languages
German (de)
English (en)
Other versions
EP3509636A4 (fr
Inventor
Michal Eisenbach-Schwartz
Kuti BARUCH
Neta ROSENZWEIG
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Yeda Research and Development Co Ltd
Original Assignee
Yeda Research and Development Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US15/261,945 external-priority patent/US10519237B2/en
Priority claimed from PCT/IB2016/001433 external-priority patent/WO2017042633A2/fr
Application filed by Yeda Research and Development Co Ltd filed Critical Yeda Research and Development Co Ltd
Priority to EP23184844.1A priority Critical patent/EP4269439A3/fr
Publication of EP3509636A1 publication Critical patent/EP3509636A1/fr
Publication of EP3509636A4 publication Critical patent/EP3509636A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention relates in general to methods and compositions for treating disease, disorder, condition or injury of the Central Nervous System (CNS) by transiently reducing the level of systemic immunosuppression in the circulation.
  • CNS Central Nervous System
  • CNS central nervous system
  • AD Alzheimer's disease
  • amyloid-beta
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an active agent that causes reduction of the level of systemic immunosuppression in an individual for use in treating a disease, disorder, condition or injury of the CNS that does not include the autoimmune neuroinflammatory disease, relapsing-remitting multiple sclerosis (RRMS), wherein said pharmaceutical composition is for administration by a dosage regimen comprising at least two courses of therapy, each course of therapy comprising in sequence a treatment session followed by an interval session of non- treatment.
  • RRMS relapsing-remitting multiple sclerosis
  • the present invention provides a method for treating a disease, disorder, condition or injury of the Central Nervous System (CNS) that does not include the autoimmune neuroinflammatory disease relapsing-remitting multiple sclerosis (RRMS), said method comprising administering to an individual in need thereof a pharmaceutical composition comprising an active agent that causes reduction of the level of systemic immunosuppression according to the present invention, wherein said pharmaceutical composition is administered by a dosage regime comprising at least two courses of therapy, each course of therapy comprising in sequence a treatment session followed by an interval session of a non-treatment period.
  • CNS Central Nervous System
  • RRMS autoimmune neuroinflammatory disease relapsing-remitting multiple sclerosis
  • FIGs. 1 A-B depict the choroid plexus (CP) activity along disease progression in the 5XFAD transgenic mouse model of AD (AD-Tg).
  • error bars represent mean ⁇ s.e.m.; * , P ⁇ 0.05; ** , P ⁇ 0.01 ; *** , P ⁇ 0.001 .
  • error bars represent mean ⁇ s.e.m. ; * , P ⁇ 0.05; ** , P ⁇ 0.01 ; *** , P ⁇ 0.001 .
  • FIG. 4 shows gating strategy and representative flow cytometry plots of splenocytes from AD- Tg/Foxp3-DTR + " mice, 1 day after the last injection of DTx. DTx was injected i.p. for 4 constitutive days, achieving -99% depletion of Foxp3 + cells.
  • FIGs. 5A-G show the effects of transient depletion of Tregs in AD-Tg mice.
  • AD-Tg/Foxp3- DTR + which express the DTR transgene
  • AD-Tg/Foxp3- DTR " non-DTR-expressing AD-Tg littermate
  • Figs. 6A-E show the effect of transient depletion of Tregs on ⁇ plaques learning/memory performance.
  • A Representative microscopic images and
  • B quantitative analysis of the brains of 5- month old DTx-treated AD-Tg/Foxp3-DTR + and AD-Tg/Foxp3-DTR " control mice, 3 weeks after the last DTx injection, immunostained for ⁇ plaques and Hoechst nuclear staining (scale bar, 250 ⁇ ).
  • DG hippocampal dentate gyrus
  • FIGs. 8A-I show the therapeutic effect of administration of weekly Glatiramer acetate (GA) in AD- Tg mice.
  • A Schematic representation of weekly-GA treatment regimen. Mice (5-month old) were s.c. injected with GA ( ⁇ ⁇ ), twice during the first week (on day 1 and 4), and once every week thereafter, for an overall period of 4 weeks. The mice were examined for cognitive performance, 1 week (MWM), 1 month (RAWM) and 2 months (RAWM, using different experimental spatial settings) after the last injection, and for hippocampal inflammation.
  • MLM 1 week
  • RAWM 1 month
  • RAWM 2 months
  • pro-inflammatory cytokines such as TNF-a, IL-1 ⁇ and IL-12p40
  • C elevation of the anti-inflammatory cytokines IL-10 and TGF- ⁇
  • D the neurotropic factors
  • AD-Tg mice (5 months old) were treated with either weekly-GA or with vehicle (PBS), and compared to age-matched WT littermates in the MWM task at the age of 6m.
  • FIGs. 9A-H show further therapeutic effects of administration of weekly-GA in AD-Tg mice.
  • A-B shows 5XFAD AD-Tg mice that were treated with either weekly-GA, or vehicle (PBS), and were examined at the end of the 1 s ' week of the administration regimen (after a total of two GA injections).
  • Flow cytometry analysis for CD4 + Foxp3 + splenocyte frequencies (A), and CP IFN-y-expressing immune cells (B; intracellularly stained and pre-gated on CD45), in treated 6-month old AD-Tg mice, compared to age- matched WT controls (n 4-6 per group; one-way ANOVA followed by Newman-Keuls post hoc analysis).
  • FIG. 9D-E show representative images of brain sections from 6-month old AD- Tg/CX 3 CR1 GFP + BM chimeras following weekly-GA.
  • CX 3 CR1 GFP cells were localized at the CP of the third ventricle (3V; i), the adjacent ventricular spaces ( /), and the CP of the lateral ventricles (LV; Hi) in AD-Tg mice treated with weekly-GA (D; scale bar, 25 ⁇ ).
  • CX 3 CR1 GFP cells are co-localized with the myeloid marker IBA-1 in brains of GA-treated AD-Tg/CX 3 CR1 GFP + mice in the vicinity of ⁇ plaques, and co-expressing the myeloid marker, IBA-1 (scale bar, 25 ⁇ ).
  • Figs. 9G-H show representative flow cytometry plots of cells isolated from the hippocampus of 4-month old WT, untreated AD-Tg, and AD-Tg mice, on the 2 nd week of the weekly-GA regimen.
  • FIGs. 10A-H depict the therapeutic effect of administration of a p300 inhibitor (C646) in AD-Tg mice.
  • a p300 inhibitor C646
  • aged mice (18 months) were treated with either p300i or vehicle (DMSO) for a period of 1 week, and examined a day after cessation of treatment.
  • Representative flow cytometry plots showing elevation in the frequencies of CD4 + T cells expressing IFN- ⁇ in the spleen (A), and IFN- ⁇ - expressing immune cell numbers in the CP (B), following p300i treatment.
  • Figs. 10A-H depict the therapeutic effect of administration of a p300 inhibitor (C646) in AD-Tg mice.
  • DMSO vehicle
  • 10C-E show representative microscopic images (C), and quantitative analysis, of ⁇ plaque burden in the brains of 10- month old AD-Tg mice, which received either p300i or vehicle (DMSO) for a period of 1 week, and were subsequently examined after 3 additional weeks.
  • error bars represent mean ⁇ s.e.m.; * , P ⁇ 0.05; ** , P ⁇ 0.01 ; *** , P ⁇ 0.001 .
  • FIGs. 11 A-D show that PD-1 blockade augments percentage of IFN-y-producing CD4+ T-cells in the spleen, as well as IFN- ⁇ expression at the choroid plexus in AD-Tg mice.
  • 10-month old AD-Tg mice were i.p. injected on day 1 and day 4 with 250ug of either anti-PD-1 or control IgG, and examined at days 7-10 for the effect on the systemic immune response and CP activity.
  • C mRNA expression levels of ifn-g, measured by RT-qPCR in the CP of AD-Tg mice treated with anti-PD-1 when compared to IgG treated and untreated AD-Tg controls
  • FIGs. 12A-B show that PD-1 blockade mitigates cognitive decline in AD-Tg mice.
  • 10-month old AD-Tg mice were i.p. injected on day 1 and day 4 with 250 ug of either anti-PD-1 or control IgG, and examined 1 or 2 months later for the effect on pathology with (A) showing performance of AD-Tg mice in the RAWM after 1 treatment session with anti-PD-1 or IgG control and (B) showing effect of single anti- PD-1 treatment session, or 2 sessions with a 1 month interval on performance.
  • Single arrows indicate time points of treatment, and double arrows indicate time points of cognitive testing.
  • Fig. 14 shows the effect of different dosing and frequency of administration of anti-PD-1 antibody on cognitive decline in AD-Tg mice and illustrates the dosage scheme and the effect of anti-PD-1 antibody treatment on spatial learning and memory performance using the radial arm water maze (RAWM) task at 7 months of age.
  • Black arrows indicate time points of treatment, and illustrations indicate time points of cognitive testing.
  • FIGs. 15A-H show the effect of repeated administration of anti-PD-1 antibody on cognitive decline in AD-Tg mice.
  • 5XFAD mice were treated with either PD-1 -specific antibody or IgG control, starting at 3 months of age; treatment was continued once a month until the age of 5 months (total of three injections).
  • Experimental design is presented in (A). Black arrows indicate time points of treatment, and illustrations indicate time points of cognitive testing.
  • Fig. 16 shows the effect of a single administration of anti-TIM-3 antibody on cognitive decline in AD-Tg mice and illustrates the dosage scheme and the effect of anti-TIM-3 antibody treatment on spatial learning and memory performance using the radial arm water maze (RAWM) task at 7 months of age.
  • Black arrows indicate time points of treatment, and illustrations indicate time points of cognitive testing.
  • FIGs. 17A-J show that PD-L1 blockade mitigates cognitive decline in AD-Tg mice.
  • 5XFAD mice (7-months old) were treated with either PD-1 -specific antibody, PD-L1 -specific antibody or isotype matched control antibody (IgG).
  • Experimental design is presented in (A). Black arrow indicates time point of treatment, and illustrations indicate time points of cognitive scoring using the RAWM.
  • Figs. 18A-B show that PD-L1 expression increases at the CP with aging with (A) showing expression of PDL1 in the CP of young (left bar) and aged (right bar) mice, measured by RT-qPCR; and (B) showing immunohistochemical staining of epithelial expression of PD-L1 at the CP of young (left micrograph) and aged (right micrograph) mice. LV; lateral ventricle.
  • FIGs. 21 A-E show that PD-1 /PD-L1 axis blockade in DM-hTau mice enhances monocyte recruitment to the brain.
  • (A) Flow cytometry gating strategy for splenocytes, and (B) quantitative analysis of FoxP3 + regulatory T cells, and (C) CD44 + CD62L " l0W effector memory T (T EF ) cells, and (C) CD44 + CD62L " low effector memory T (T EF ) cells, versus CD44 + CD62L hi9h central memory T (T C M) cells.
  • (D) Flow cytometry gating strategy for brain CD45 low CD1 1 b + and CD45 h ' 9h CD1 1 b + myeloid cells.
  • FIGs. 22A-G shows that PD-1 /PD-L1 axis blockade in DM-hTau mice enhances monocyte recruitment to the brain.
  • Male mice expressing the human-tau gene with two mutations K257T/P301 S; double mutant, DM-hTAU) (average cohorts aged 8 months) were treated with anti-PD-1 -specific antibody, anti-PD-L1 -specific antibody, or isotype matched control antibody (IgG) (one i.p. injection of 0.5mg/mouse); experimental design is presented in (A).
  • Black arrow indicates time point of treatment, and illustrations indicate time points of cognitive testing.
  • FIGs. 23A-G show that blocking PD-1 /PD-L1 pathway reduces hyperphosphorylation in DM-tau mice.
  • Immunostaining of Neurofibrillary tangles (NFTs) in brains of 8 month old DM-hTAU mice 1 month after treatment with anti-PD-1 , anti-PD-L1 , or isotype matched control antibody or with isotype matched control antibody (IgG) A-F.
  • A, D Representative immunofluorescence images
  • B, C, E, F quantitative analysis of AT-100 and AT-180.
  • the effect of PD-L1 blockade on spatial memory was determined using the T maze task.
  • Fig. 24A-B shows PD-1 blockade enhances hippocampal neurogenesis in 5XFAD mice with (A) showing parasagittal brain sections immunostained for neuronal marker-NeuN (in green), DCX (in red), and hoechst nuclear staining (in blue); and (B) showing a graph quantitating the staining in anti-PD-1 treated animals, IgG immune controls and aged-matched wild-type controls.
  • Fig. 25A-B shows PD-1 blockade enhances hippocampal synaptic plasticity in 5XFAD mice with (A) showing parasagittal brain sections immunostained for VgluTI (red); and (B) showing a graph quantitating the staining in anti-PD-1 treated animals, IgG immune controls and aged-matched wild-type controls.
  • Fig. 26A-B shows PD-1 blockade reduces neuronal loss in the subiculum of 5XFAD mice with (A) showing parasagittal brain sections immunostained for neuronal marker-NeuN (in green); and (B) showing a graph quantitating the staining in anti-PD-1 treated animals, IgG immune controls and aged- matched wild-type controls.
  • Immune checkpoint mechanisms which include cell-intrinsic downregulation of activated T cell responsiveness and effector function by inhibitory receptors, maintain systemic immune homeostasis and autoimmune tolerance (Joller et al, 2012; Pardoll, 2012).
  • immune checkpoint blockade activates a cascade of immunological events that starts in the periphery and culminates in numerous activities inside the brain.
  • an immune response increases the availability of IFN- ⁇ at the secondary lymphoid organs (lymph nodes, spleen, etc.) and circulating monocytes in the periphery.
  • This immune response leads to the immunological activation of the brain's choroid plexus (CP), an epithelial layer at the brain ventricles, which forms the blood-cerebrospinal fluid-barrier (B-CSF-B), and serves as a selective gateway for leukocytes entering the CNS.
  • CP brain's choroid plexus
  • B-CSF-B blood-cerebrospinal fluid-barrier
  • the effect of the blockade of inhibitory immune checkpoints on CP gateway activity for leukocyte is mediated by the IFN-y-induced expression of leukocyte trafficking molecules (adhesion molecules and chemokines) by the CP epithelium, which enables leukocyte trafficking.
  • leukocyte trafficking molecules asdhesion molecules and chemokines
  • This increased expression leads to the recruitment of monocyte-derived macrophages and immunoregulatory cells to diseased sites within the brain.
  • monocyte-derived macrophages and immunoregulatory cells to diseased sites within the brain.
  • this recruitment results in a comprehensive effect on brain function, including reduced of plaque burden, restored of immunological balance, resolved local inflammation, reduced gliosis, reduced synaptic loss, increased neurogenesis, increased neuronal protection and enhanced neuronal survival, collectively leading to neuroprotection and/or reduction in cognitive decline.
  • Immune checkpoints are molecules in the immune system that either turn up a signal (co- stimulatory molecules) or turn down a signal.
  • Four stimulatory checkpoint molecules are members of the tumor necrosis factor (TNF) receptor superfamily - CD27, CD40, OX40, GITR and CD137.
  • TNF tumor necrosis factor
  • Another two stimulatory checkpoint molecules belongs to the B7-CD28 superfamily - CD28 itself and ICOS.
  • Many inhibitor checkpoint molecules are known, including, without limitation, A2aR, B7-H3, B7-H4, BTLA, CTLA-4, IDO, KIR, LAG -3, PD-1 , TIM-3 and VISTA.
  • the present invention provides a method for treating a disease, disorder, condition or injury of the Central Nervous System (CNS).
  • the disclosed method for treating a disease, disorder, condition or injury of the Central Nervous System (CNS) does not include the autoimmune neuroinflammatory disease relapsing-remitting multiple sclerosis (RRMS).
  • the disclose method comprising administering to an individual in need thereof an active agent that causes reduction of the level of systemic immunosuppression, wherein said active agent is administered by a dosage regime comprising at least two courses of therapy, each course of therapy comprising in sequence a treatment session followed by an interval session of non-treatment.
  • the present invention is directed to an active agent that causes reduction of the level of systemic immunosuppression in an individual, or a pharmaceutical composition comprising the active agent, for use in treating a disease, disorder, condition or injury of the CNS that does not include the autoimmune neuroinflammatory disease, relapsing-remitting multiple sclerosis (RRMS), wherein said pharmaceutical composition is for administration by a dosage regimen comprising at least two courses of therapy, each course of therapy comprising in sequence a treatment session followed by an interval session of non-treatment.
  • RRMS relapsing-remitting multiple sclerosis
  • the dosage regimen is calibrated such that the level of systemic immunosuppression is transiently reduced.
  • treating refers to means of obtaining a desired physiological effect. The effect may be therapeutic in terms of partially or completely curing a disease and/or symptoms attributed to the disease. The term refers to inhibiting the disease, i.e. arresting or slowing its development; or ameliorating the disease, i.e. causing regression of the disease.
  • systemic presence of regulatory or effector T cells refers to the presence of the regulatory or effector T cells (as measured by their level or activity) in the circulating immune system, i.e. the blood, spleen and lymph nodes. It is a well-known fact in the field of immunology that the cell population profile in the spleen is reflected in the cell population profile in the blood (Zhao et al, 2007).
  • the present treatment is applicable to both patients that show elevation of systemic immune suppression, as well as to patients that do not show such an elevation.
  • the individual in need for the treatment according to the present invention has a certain level of peripheral immunosuppression, which is reflected by elevated frequencies or numbers of Tregs in the circulation, and/or their enhanced functional activity and/or a decrease in IFNy-producing leukocytes and/or decreased proliferation of leukocytes in response to stimulation.
  • the elevation of frequencies or numbers of Tregs can be in total numbers or as percentage of the total CD4 cells. For example, it has been found in accordance with the present invention that an animal model of Alzheimer's disease has higher frequencies of Foxp3 out of CD4 cells as compared with wild-type mice.
  • the method of the present invention that reduces the level or activity of systemic immunosuppression is effective in treating disease, disorder, condition or injury of the CNS that does not include the autoimmune neuroinflammatory disease RRMS.
  • said systemic immune suppression can also involve additional immune cell types except of Tregs, such as myeloid-derived suppressor cells (MDSCs) (Gabrilovich & Nagaraj, 2009).
  • MDSCs myeloid-derived suppressor cells
  • the level of systemic immunosuppression may be detected by various methods that are well known to those of ordinary skill in the art.
  • the level of Tregs may be measured by flow cytometry analysis of peripheral blood mononuclear cells or T lymphocytes, immunostained either for cellular surface markers or nuclear intracellular markers of Treg (Chen & Oppenheim, 201 1 ), CD45, TCR- ⁇ , or CD4 markers of lymphocytes, and measuring the amount of antibody specifically bound to the cells.
  • Tregs may be measured by various assays; For example the thymidine incorporation assay is being commonly used, in which suppression of anti-CD3 mAb stimulated proliferation of CD4 + CD25 ⁇ T cells (conventional T cells) is measured by [ 3 H]thymidine incorporation or by using CFSE (5-(and 6)-carboxyfluorescein diacetate succinimidyl ester, which is capable of entering the cells; cell division is measured as successive halving of the fluorescence intensity of CFSE).
  • CFSE 5-(and 6)-carboxyfluorescein diacetate succinimidyl ester
  • the number of IFNy-producing leukocytes or their activity or their proliferation capacity can easily be assessed by a skilled artisan using methods known in the art;
  • the level of IFNy-producing leukocytes may be measured by flow cytometry analysis of peripheral blood mononuclear cells, following short ex-vivo stimulation and golgi-stop, and immunostaining by IFNy intracellular staining (using e.g., BD Biosciences Cytofix/cytopermTM fixation/permeabilization kit), by collecting the condition media of these cells and quantifying the level of secreted cytokines using ELISA, or by comparing the ratio of different cytokines in the condition media, for example IL2/IL10, IL2/IL4, INFy/TGFp, etc.
  • the levels of MDSCs in the human peripheral blood easily can be assessed by a skilled artisan, for example by using flow cytometry analysis of frequency of DR LIN CD1 1 b+, DR LIN CD1 5+, DR7LIN7CD33+ and DR(7low)/CD14+ cells, as described (Kotsakis et al, 2012).
  • the peripheral/systemic immunosuppression may be considered elevated when the total number of Tregs in the circulation is higher than 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% or more than in a healthy control population, the percentage of Treg cells out of the total CD4+ cells is elevated by 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% or more than in a healthy control population, or the functional activity of Tregs is elevated by 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% or more than in a healthy control population.
  • the peripheral/systemic immunosuppression may be considered elevated when the level of IFNy-producing leukocytes or their activity is reduced relative to that of a healthy control population by 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100%; or the proliferation of leukocytes in response to stimulation is reduced relative to that of a healthy control population by 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100%.
  • An agent may be considered an agent that causes reduction of the level of systemic immunosuppression when, upon administration of the agent to an individual, the total number of Tregs in the circulation of this individual is reduced by 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100% as compared with the level before administration of the agent, the percentage of Treg cells out of the total CD4+ cells drops by 1 0, 20, 30, 40, 50, 60, 70, 80, 90 or 1 00% relative to that of a healthy control population or the functional activity of Tregs is reduced by 1 0, 20, 30, 40, 50, 60, 70, 80, 90 or 100% as compared with the level before administration of the agent.
  • an agent may be considered an agent that causes reduction of the level of systemic immunosuppression when, upon administration of the agent to an individual, the total number of IFNy-producing leukocytes or their activity is increased by 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% or more; or the proliferation of leukocytes in response to stimulation is increased relative to that of a healthy control population by 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100% or more.
  • the active agent causes reduction of the level of systemic immunosuppression by release of a restraint imposed on the immune system by one or more immune checkpoints, for example by blockade of the one or more immune checkpoints.
  • the reduction of the level of systemic immunosuppression is associated with an increase in systemic presence or activity of IFNy-producing leukocytes.
  • the active agent causes reduction of the level of systemic immunosuppression and thereby an increase in the systemic presence or activity of effector T cells.
  • the reduction of the level of systemic immunosuppression is associated with an increase in systemic presence or activity of an IFNy cytokine.
  • the reduction of the level of systemic immunosuppression is associated with a decrease in systemic presence or activity of regulatory T-cells.
  • the reduction of the level of systemic immunosuppression is associated with a decrease in systemic presence or activity of an IL-10 cytokine.
  • the reduction of the level of systemic immunosuppression is associated with a decrease in systemic presence or activity of myeloid-derived suppressor cells (MDSCs).
  • the active agent causes reduction of the level of systemic immunosuppression and thereby an increase in the systemic presence or activity of effector T cells.
  • the checkpoints that may be manipulated to release the systemic immunosuppression are referred to herein as a pair of an immune checkpoint receptor and its native ligand or either one of the two partners.
  • PD-1 which has two known ligands is referred to herein as “ PD-L1 " and " PD- L2", while B7H3, the ligand of which has not yet been identified, is referred to simply by "B7H3".
  • the checkpoints that may be manipulated to release the systemic immunosuppression in accordance with the present invention include, without limitation, PD-1 -PD-L1 , PD-1 -PD-L2, CD28-CD80, CD28-CD86, CTLA- 4-CD80, CTLA-4-CD86, ICOS-B7RP1 , B7H3, B7H4, B7H7, B7-CD28-like molecule, BTLA-HVEM, KIR- MHC class I or II, LAG3-MHC class I or II, CD137-CD137L, OX40-OX40L, CD27-CD70, CD40L-CD40, TIM3-GAL9, V-domain Ig suppressor of T cell activation (VISTA), STimulator of INterferon Genes (STING), T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT), glucocorticoid-induced tumor necrosis factor receptor related protein (GITR
  • the active agent that may be used according to the present invention may be an antibody.
  • An antibody as disclosed herein can be a polyclonal antibody, a monoclonal antibody, a dimer, a multimer, a multispecific antibody, a human antibody, a humanized antibody, a recombinant antibody, a chimeric antibody, bi-functional antibody, a cell-associated antibody like an Ig receptor, a linear antibody, a diabody, a minibody or a nanobody, so long as the fragment exhibits the desired biological activity, and single chain derivatives of the same.
  • An antibody can be a full-length immunoglobulin molecule comprising the VH and VL domains, as well as a light chain constant domain (CL) and heavy chain constant domains, CH1 , CH2 and CH3, or an immunologically active fragment of a full-length immunoglobulin molecule, such as, e.g., a single domain antibody (sdAb), a single-chain variable fragment (scFv), a Fab fragment, a F(ab')2 fragment, a Fc fragment, a Fd fragment, a Fv fragment.
  • sdAb single domain antibody
  • scFv single-chain variable fragment
  • An antibody can be derived from any vertebrate species (e.g., human, goat, horse, donkey, murine, rat, rabbit, or chicken), and can be of any type (e.g., IgG, IgE, IgM, IgD, and IgA), class (e.g., IgA, IgD, IgE, IgG, and IgM) or subclass (lgG1 , lgG2, lgG3, lgG4, lgA1 and lgA2).
  • an antibody disclosed herein may be an antagonist antibody, meaning an antibody that inhibits a biological activity or an antibody disclosed herein may be an agonist antibody, meaning an antibody that stimulates a biological activity.
  • an antibody disclosed herein may be a neutralizing antibody, meaning an antibody that can block or neutralize a biological activity.
  • a neutralizing antibody meaning an antibody that can block or neutralize a biological activity.
  • An antibody disclosed herein may be, without limitation, an anti-PD-1 , an anti-PD-L1 , an anti-PD- L2, an anti-CTLA-4, an anti-CD80,an anti-CD86, an anti-B7RP1 , an anti-B7-H3, an anti-B7-H4, an anti- B7-H7, an anti-BTLA, an anti-HVEM, an anti-CD-27, an anti-CD40, an anti-CD40L, an anti-CD70, an anti- CD80, an anti-CD86, an anti-CD137, an anti-CD137L, an anti-OX40, an anti-OX40L, an anti-TIM-3, an anti-Galectin9, an anti-KIR, an anti-LAG-3, an anti-ICOS, an anti-VISTA, an anti-STING, an anti-TIGIT, anti-GITR or any combination thereof.
  • An antibody disclosed herein may be administered to a human at a dosage of for example about 0.1 mg/kg - 20 mg/kg, 0.1 mg/kg - 15 mg/kg, 0.1 mg/kg - 10 mg/kg, 0.1 mg/kg - 5 mg/kg, 0.2 mg/kg - 20 mg/kg, 0.2 mg/kg - 15 mg/kg, 0.2 mg/kg - 10 mg/kg, 0.2 mg/kg - 6 mg/kg, 0.2 mg/kg - 5 mg/kg, 0.3 mg/kg - 20 mg/kg, 0.3 mg/kg - 15 mg/kg, 0.3 mg/kg - 10 mg/kg, 0.3 mg/kg - 5 mg/kg, , 1 mg/kg - 20 mg/kg, 1 mg/kg - 15 mg/kg, 1 mg/kg - 10 mg/kg, 1 mg/kg - 5 mg/kg, 1 .5 mg/kg - 20 mg/kg, 1 .5 mg/kg - 15 mg/kg, 1 .5
  • Programmed cell death protein 1 also known as PD-1 and CD279 (cluster of differentiation 279), is a cell surface receptor that belongs to the immunoglobulin superfamily and is expressed on T cells and pro-B cells.
  • PD-1 binds two ligands, PD-L1 and PD-L2. Functioning as an immune checkpoint, PD-1 plays an important role in down regulating the immune system by preventing the activation of T-cells, which in turn reduces autoimmunity and promotes self-tolerance.
  • the inhibitory effect of PD-1 is accomplished through a dual mechanism of promoting apoptosis (programmed cell death) in antigen specific T-cells in lymph nodes while simultaneously reducing apoptosis in regulatory T cells (suppressor T cells).
  • compounds which inhibit PD-1 function such as PD-1 inhibitors, PD-L1 inhibitors and/or PD-L2 inhibitors, serve to activate the immune system.
  • One class of PD-1 inhibitors includes antagonist or neutralizing anti-PD-1 , anti-PD-L1 and anti-PD-L2 antibodies. Many antagonist or neutralizing anti-PD-1 , anti-PD-L1 and anti-PD-L2 antibodies are known in the art.
  • the anti- PD-1 antibody used in accordance with the present invention may be selected from those disclosed in Ohaegbulam et al. (Ohaegbulam et al, 201 5), the entire contents of which being hereby incorporated herein by reference.
  • human or humanized anti-PD-1 antibodies include, without limitation, CD279 (human anti-PD1 monoclonal antibody, Bio X Cell), MEDI0680 (AMP-514; humanized lgG4 anti- PD-1 monoclonal antibody; AstraZeneca), Nivolumab (BMS-936558; human lgG4 anti-PD1 monoclonal antibody; Bristol-Myers Squibb), Pembrolizumab (Lambrolizumab, MK-3475; humanized lgG4 anti-PD1 monoclonal antibody; Merck), Pidilizumab (CT-01 1 ; humanized lgG1 anti-PD1 monoclonal antibody; Medivation) and TSR-042 (humanized lgG4 anti-PD-1 monoclonal antibody; Tesaro).
  • CD279 human anti-PD1 monoclonal antibody, Bio X Cell
  • MEDI0680 AMP-514; humanized lgG4 anti- PD-1 monoclo
  • human or humanized anti-PD-L1 antibodies include, without limitation, Avelumab (MSB0010718C; human lgG1 anti-PD-L1 monoclonal antibody; Merck-Serono), Atezolizumab (MPDL3280A, RG7446; human IgG anti-PD-L1 monoclonal antibody; Hoffmann-La Roche), BMS-936559 (MDX-1 105; human lgG4 anti-PD- L1 monoclonal antibody; Bristol-Myers Squibb), Durvalumab (MEDI4736; humanized lgG1 anti-PD-L1 monoclonal antibody; AstraZeneca), KN035 (anti-PD-L1 monoclonal antibody; 3D Medicines) and LY3300054 (anti-PD-L1 monoclonal antibody; Eli Lilly).
  • Avelumab MSB0010718C
  • human lgG1 anti-PD-L1 monoclonal antibody Merck-Serono
  • an anti-PD-1 antibody, an anti-PD-L1 antibody and/or an anti-PD-L2 antibody may be administered to a human at a dosage of for example about 0.1 mg/kg - 20 mg/kg, 0.1 mg/kg - 15 mg/kg, 0.1 mg/kg - 10 mg/kg, 0.1 mg/kg - 5 mg/kg, 0.2 mg/kg - 20 mg/kg, 0.2 mg/kg - 15 mg/kg, 0.2 mg/kg - 10 mg/kg, 0.2 mg/kg - 6 mg/kg, 0.2 mg/kg - 5 mg/kg, 0.3 mg/kg - 20 mg/kg, 0.3 mg/kg - 15 mg/kg, 0.3 mg/kg - 10 mg/kg, 0.3 mg/kg - 5 mg/kg, , 1 mg/kg
  • Pidilizumab may be administered to a human at a dosage of 0.2-6 mg/kg or between 1 .5-6 mg/kg; Pembrolizumab may be administered to a human at a dosage of 1 -10 mg/kg; Nivolumab may be administered to a human at a dosage of 0.3-20 mg/kg, 0.3-10 mg/kg, 1 -10 mg/kg or at 1 or 3 mg/kg; BMS-936559 may be administered to a human at a dosage of 0.3-10 mg/kg; Atezolizumab may be administered to a human at a dosage of 1 -20 mg/kg; Durvalumab may be administered to a human at a dosage of 0.1 -15 mg/kg; and Avelumab may be administered to a human at a dosage of 1 -20 mg/kg.
  • T-cell immunoglobulin and mucin domain-3 (TIM-3) is a Th1 -specific cell surface protein that acts as an immune checkpoint that inhibits lymphocyte activity by down regulating macrophage activation and playing an important role in CD8+ T cell exhaustion that takes place in chronic immune conditions.
  • TIM-3 acts as a negative regulator of Th1 /Tc1 function by triggering cell death upon interaction with its ligand, galectin-9 (Gal9).
  • Gal9 galectin-9
  • compounds which inhibit TIM-3 function such as TIM-3 inhibitors and/or Gal9 inhibitors, serve to activate the immune system.
  • One class of TIM-3 inhibitors includes antagonist or neutralizing antibodies against TIM-3 and/or Gal-9.
  • human or humanized anti-TIM-3 antibodies include, without limitation, AF2365 (human IgG anti-TIM-3 monoclonal antibody; R&D Systems), CD366 (human lgG1 anti-TIM-3 monoclonal antibody; BioLegend), F38-2E2 (human lgG1 anti-TIM-3 monoclonal antibody; R&D Systems), L3D (human lgG1 anti-TIM-3 monoclonal antibody; CN 102492038 B), MAB2365 (human lgG2a anti-TIM-3 monoclonal antibody; R&D Systems), MAB23651 (human lgG1 anti- TIM-3 monoclonal antibody; R&D Systems) and TSR-022 (humanized lgG4 anti-TIM-3 monoclonal antibody; Tesaro).
  • AF2365 human IgG anti-TIM-3 monoclonal antibody; R&D Systems
  • CD366 human lgG1 anti-TIM-3 monoclonal antibody; BioLegend
  • an anti-TIM-3 antibody and/or an anti-Gal9 antibody may be administered to a human at a dosage of for example about 0.1 mg/kg - 20 mg/kg, 0.1 mg/kg - 15 mg/kg, 0.1 mg/kg - 10 mg/kg, 0.1 mg/kg - 5 mg/kg, 0.2 mg/kg - 20 mg/kg, 0.2 mg/kg - 15 mg/kg, 0.2 mg/kg - 10 mg/kg, 0.2 mg/kg - 6 mg/kg, 0.2 mg/kg - 5 mg/kg, 0.3 mg/kg - 20 mg/kg, 0.3 mg/kg - 15 mg/kg, 0.3 mg/kg - 10 mg/kg, 0.3 mg/kg - 5 mg/kg, , 1 mg/kg - 20 mg/kg, 1 mg/kg - 15 mg/kg, 1 mg/kg - 10 mg/kg, 1 mg/kg - 5 mg/kg, 1 .5 mg/kg - 20 mg/kg, 1 .
  • CTLA-4 Cytotoxic T-lymphocyte-associated protein 4
  • CD152 cluster of differentiation 152
  • CTLA-4 is a protein receptor functioning as an immune checkpoint that downregulates immune responses.
  • CTLA-4 is constitutively expressed in Tregs but only upregulated in conventional T cells after activation.
  • CTLA-4 acts as an "off" switch when bound to CD80 or CD86 on the surface of antigen- presenting cells.
  • compounds which inhibit CTLA-4 function such as CTLA-4 inhibitors, CD80 inhibitors and/or CD86 inhibitors, serve to activate the immune system.
  • CTLA-4 inhibitors includes antagonist or neutralizing antibodies against CTLA-4, CD80 and/or CD86.
  • anti-CTLA-4, anti-CD80 and anti-CD86 antibodies are known in the art.
  • human or humanized anti-CTLA-4 antibodies include, without limitation, Ipilimumab (human lgG1 anti-CTLA-4 monoclonal antibody; Bristol-Myers Squibb) and Tremelimumab (human lgG2 anti-CTLA-4 monoclonal antibody; Pfizer).
  • an anti-CTLA-4 antibody, an anti-CB80 antibody and/or an anti-CD86 antibody may be administered to a human at a dosage of for example about 0.1 mg/kg - 20 mg/kg, 0.1 mg/kg - 15 mg/kg, 0.1 mg/kg - 10 mg/kg, 0.1 mg/kg - 5 mg/kg, 0.2 mg/kg - 20 mg/kg, 0.2 mg/kg - 15 mg/kg, 0.2 mg/kg - 10 mg/kg, 0.2 mg/kg - 6 mg/kg, 0.2 mg/kg - 5 mg/kg, 0.3 mg/kg - 20 mg/kg, 0.3 mg/kg - 15 mg/kg, 0.3 mg/kg - 10 mg/kg, 0.3 mg/kg - 5 mg/kg, , 1 mg/kg - 20 mg/kg, 1 mg/kg - 15 mg/kg, 1 mg/kg - 10 mg/kg, 1 mg/kg - 5 mg/kg, 1 .5 mg/kg -
  • KIRs Killer-cell immunoglobulin-like receptors
  • MHC major histocompatibility
  • KIRs are inhibitors of lymphocyte activity.
  • compounds which inhibit KIR function such as KIR inhibitors, serve to activate the immune system.
  • One class of KIR inhibitors includes antagonist or neutralizing antibodies against KIR. Many antagonist or neutralizing anti-KIR antibodies are known in the art.
  • an anti-KIR antibody may be administered to a human at a dosage of for example about 0.1 mg/kg - 20 mg/kg, 0.1 mg/kg - 15 mg/kg, 0.1 mg/kg - 10 mg/kg, 0.1 mg/kg - 5 mg/kg, 0.2 mg/kg - 20 mg/kg, 0.2 mg/kg - 15 mg/kg, 0.2 mg/kg - 10 mg/kg, 0.2 mg/kg - 6 mg/kg, 0.2 mg/kg - 5 mg/kg, 0.3 mg/kg - 20 mg/kg, 0.3 mg/kg - 15 mg/kg, 0.3 mg/kg - 10 mg/kg, 0.3 mg/kg - 5 mg/kg, , 1 mg/kg - 20 mg/kg, 1 mg/kg - 15 mg/kg, 1 mg/kg - 15 mg/kg, 1 mg/kg - 20 mg/kg, 1 mg/kg - 15 mg/kg, , 1 mg/kg - 20 mg/kg, 1 mg/kg - 15
  • Lymphocyte-activation gene 3 (LAG-3), also known as cluster of differentiation 223 (CD223), is a cell surface molecule with diverse biologic effects on T cell function.
  • LAG-3 is an immune checkpoint that inhibits lymphocyte activity by suppressing an immune response by action to Tregs as well as direct effects on CD8+ T cells.
  • compounds which inhibit LAG-3 function such as LAG-3 inhibitors, serve to activate the immune system.
  • One class of LAG-3 inhibitors includes antagonist or neutralizing antibodies against LAG-3. Many antagonist or neutralizing anti-LAG-3 antibodies are known in the art. Examples of human or humanized anti-LAG-3 antibodies include, without limitation, BMS-986016 (human anti-LAG-3 monoclonal antibody; Bristol-Myers Squibb).
  • an anti-LAG-3 antibody may be administered to a human at a dosage of for example about 0.1 mg/kg - 20 mg/kg, 0.1 mg/kg - 15 mg/kg, 0.1 mg/kg - 10 mg/kg, 0.1 mg/kg - 5 mg/kg, 0.2 mg/kg - 20 mg/kg, 0.2 mg/kg - 15 mg/kg, 0.2 mg/kg - 10 mg/kg, 0.2 mg/kg - 6 mg/kg, 0.2 mg/kg - 5 mg/kg, 0.3 mg/kg - 20 mg/kg, 0.3 mg/kg - 15 mg/kg, 0.3 mg/kg - 10 mg/kg, 0.3 mg/kg - 5 mg/kg, , 1 mg/kg - 20 mg/kg, 1 mg/kg - 15 mg/kg, 1 mg/kg - 10 mg/kg, 1 mg/kg - 5 mg/kg, 1 .5 mg/kg - 20 mg/kg, 1 .5 mg/kg - 15 mg/kg/kg,
  • OX40 also known as cluster of differentiation 134 (CD134)
  • CD134 cluster of differentiation 134
  • OX40 promotes the expansion of effector and memory T cells, however it is also noted for its ability to suppress the differentiation and activity of T-regulatory cells, and also for its regulation of cytokine production. Being transiently expressed after T-cell receptor engagement, OX40 is only upregulated on the most recently antigen-activated T cells within inflammatory lesions.
  • Its ligand is OX40L, also known as cluster of differentiation 252 (CD252).
  • OX40 activators and/or OX40L activators serve to activate the immune system.
  • OX40 activators includes agonist antibodies against OX40 and OX40L. Many agonist antibodies against OX40 and/or OX40L are known in the art. Examples of human or humanized anti-OX40 antibodies include, without limitation, GSK3174998; humanized lgG1 anti-OX40 monoclonal antibody; GlaxoSmithKline), MEDI0562 (humanized anti-OX40 monoclonal antibody; Medlmmune) and MEDI6383 (human OX40 fusion protein; Medlmmune). Other anti-OX40 antibodies include, without limitation, MEDI6469 (9B12; murine anti-OX40 monoclonal antibody; Medlmmune).
  • an anti-OX40 antibody and/or an anti-OX40L antibody may be administered to a human at a dosage of for example about 0.1 mg/kg - 20 mg/kg, 0.1 mg/kg - 15 mg/kg, 0.1 mg/kg - 10 mg/kg, 0.1 mg/kg - 5 mg/kg, 0.2 mg/kg - 20 mg/kg, 0.2 mg/kg - 15 mg/kg, 0.2 mg/kg - 10 mg/kg, 0.2 mg/kg - 6 mg/kg, 0.2 mg/kg - 5 mg/kg, 0.3 mg/kg - 20 mg/kg, 0.3 mg/kg - 15 mg/kg, 0.3 mg/kg - 10 mg/kg, 0.3 mg/kg - 5 mg/kg, , 1 mg/kg - 20 mg/kg, 1 mg/kg - 15 mg/kg, 1 mg/kg - 10 mg/kg, 1 mg/kg - 5 mg/kg, 1 .5 mg/kg - 20 mg/kg, 1
  • Anti-GITR antibodies target glucocorticoid-induced tumor necrosis factor receptor related protein (GITR), which is regularly expressed on the surface of regulatory T-cells (Tregs) and is expressed on the surface of effector T-cells after their activation.
  • GITR glucocorticoid-induced tumor necrosis factor receptor related protein
  • Tregs regulatory T-cells
  • Anti-GITR antibodies block the interaction of GITR, found on multiple types of T cells, with its ligand, thereby inducing both the activation of tumor-antigen-specific T effector cells, as well as abrogating the suppression induced by inappropriately activated T regulatory cells.
  • compounds which activate or stimulate GITR function such as GITR activators, serve to activate the immune system.
  • One class of GITR activators includes agonist antibodies against GITR.
  • human or humanized anti-TIGR antibodies include, without limitation, GWN323 (humanized anti-GITR monoclonal antibody; Novartis) and TRX518 (humanized anti-GITR monoclonal antibody; GITR, Inc.).
  • an anti-GITR antibody may be administered to a human at a dosage of for example about 0.1 mg/kg - 20 mg/kg, 0.1 mg/kg - 15 mg/kg, 0.1 mg/kg - 10 mg/kg, 0.1 mg/kg - 5 mg/kg, 0.2 mg/kg - 20 mg/kg, 0.2 mg/kg - 15 mg/kg, 0.2 mg/kg - 10 mg/kg, 0.2 mg/kg - 6 mg/kg, 0.2 mg/kg - 5 mg/kg, 0.3 mg/kg - 20 mg/kg, 0.3 mg/kg - 15 mg/kg, 0.3 mg/kg - 10 mg/kg, 0.3 mg/kg - 5 mg/kg, , 1 mg/kg - 20 mg/kg, 1 mg/kg - 15 mg/kg, 1 mg/kg - 10 mg/kg, 1 mg/kg - 5 mg/kg, 1 .5 mg/kg - 20 mg/kg, 1 .5 mg/kg - 15 mg/kg,
  • CD27 is a member of the tumor necrosis factor receptor superfamily. CD27 activity is governed by the transient availability of its ligand, CD70, on lymphocytes and dendritic cells. Activation of CD27 plays a key role in regulating B-cell activation and immunoglobulin synthesis, supports antigen-specific expansion of na ' ive T cells, is required for generation and long-term maintenance of T cell immunity and is a memory marker of B cells. CD27 transduces signals that lead to the activation of NF- ⁇ and MAPK8/JNK. As such, compounds which activate or stimulate CD27 function, such as CD27 activators and/or CD70 activators, serve to activate the immune system.
  • CD27 activators includes agonist antibodies against CD27 and/or CD70. Many agonist antibodies against CD27 and/or CD70 are known in the art. Examples of human or humanized anti-CD27 antibodies include, without limitation, Varlilumab (CDX-1 127; human anti-CD27 monoclonal antibody; Celldex Therapeutics).
  • an anti-CD27 antibody and/or an anti-CD70 antibody may be administered to a human at a dosage of for example about 0.1 mg/kg - 20 mg/kg, 0.1 mg/kg - 15 mg/kg, 0.1 mg/kg - 10 mg/kg, 0.1 mg/kg - 5 mg/kg, 0.2 mg/kg - 20 mg/kg, 0.2 mg/kg - 15 mg/kg, 0.2 mg/kg - 10 mg/kg, 0.2 mg/kg - 6 mg/kg, 0.2 mg/kg - 5 mg/kg, 0.3 mg/kg - 20 mg/kg, 0.3 mg/kg - 15 mg/kg, 0.3 mg/kg - 10 mg/kg, 0.3 mg/kg - 5 mg/kg, , 1 mg/kg - 20 mg/kg, 1 mg/kg - 15 mg/kg, 1 mg/kg - 10 mg/kg, 1 mg/kg - 5 mg/kg, 1 .5 mg/kg - 20 mg/kg, 1 .5
  • Inducible T-cell COStimulator also known as cluster of differentiation 278 (CD278), is a CD28-superfamily costimulatory cell-surface receptor that is expressed on activated T cells. It is an activator of T cell function.
  • compounds which activate or stimulate ICOS function such as ICOS activators or B7RP1 activators, serve to activate the immune system.
  • One class of ICOS activators includes agonist antibodies against ICOS and/or B7RP1 . Many agonist anti-ICOS and anti-B7RP1 antibodies are known in the art.
  • an anti-ICOS antibody and/or an anti-B7RP1 antibody may be administered to a human at a dosage of for example about 0.1 mg/kg - 20 mg/kg, 0.1 mg/kg - 15 mg/kg, 0.1 mg/kg - 10 mg/kg, 0.1 mg/kg - 5 mg/kg, 0.2 mg/kg - 20 mg/kg, 0.2 mg/kg - 15 mg/kg, 0.2 mg/kg - 10 mg/kg, 0.2 mg/kg - 6 mg/kg, 0.2 mg/kg - 5 mg/kg, 0.3 mg/kg - 20 mg/kg, 0.3 mg/kg - 15 mg/kg, 0.3 mg/kg - 10 mg/kg, 0.3 mg/kg - 5 mg/kg, , 1 mg/kg - 20 mg/kg, 1 mg/kg - 15 mg/kg, 1 mg/kg - 10 mg/kg, 1 mg/kg - 5 mg/kg, 1 .5 mg/kg - 20 mg/kg, 1
  • combinations of antibodies may be used such as but not limited to: CT- 01 1 in combination with Rituximab (trade names Rituxan, MabThera and Zytux) a chimeric monoclonal antibody against the protein CD20, for example, each at 3mg/kg; Nivolumab (for example 1 mg/kg) in combination with Ipilimumab; for example at 3 mg/kg); or Nivolumab (e.g. 1 -10 mg/kg) in combination with a an HLA-A * 0201 -restricted multipeptide vaccine (Weber et al, 2013).
  • Rituximab trade names Rituxan, MabThera and Zytux
  • Nivolumab for example 1 mg/kg
  • Ipilimumab for example at 3 mg/kg
  • Nivolumab e.g. 1 -10 mg/kg
  • the active agent that may be used according to the present invention may be an antibody mimetic.
  • Antibody mimetics can specifically bind antigens like antibodies, but that are not structurally related to antibodies. They are usually artificial peptides or proteins with a molar mass of about 3 kDa to 20 kDa.
  • an antibody mimetic disclosed herein may be an affibody molecule; an affilin; an affimer; an affitin; an alphabody; an anticalin; an avimer; a DARPin; a fynomer; a Kunitz domain peptide; or a monobody.
  • Non-limiting examples of antibody mimetics are presented in Table 1 .
  • the active agent that may be used according to the present invention may be an aptamer.
  • Aptamers are oligonucleotide or peptide molecules that bind to a specific target molecule.
  • an aptamer disclosed herein may be a DNA aptamer, a RNA aptamer, a XNA aptamer or a peptide aptamer.
  • an antibody disclosed herein may be in combination with an adjuvant.
  • An adjuvant is any substance or mixture of substances that increases or diversifies the immune response.
  • An adjuvant may serve to reduce the number of immunizations or the amount of antigen required for protective immunization.
  • Non-limiting adjuvants include, e.g., liposomes, oily phases, including, without limitation, the Freund type of adjuvants, such as, e.g., Freund's complete adjuvant (FCA); Freund's incomplete adjuvant (FIA); sapogenin glycosides, such as, e.g., saponins; carbopol; N-acetylmuramyl-L- alanyl-D-isoglutamine (commonly known as muramyl dipeptide or "MDP"); and lipopolysaccharide (LPS).
  • FCA Freund's complete adjuvant
  • FIA Freund's incomplete adjuvant
  • sapogenin glycosides such as, e.g., saponins; carbopol; N-acetylmuramyl-L- alanyl-D-isoglutamine (commonly known as muramyl dipeptide or "MDP"); and lipopolysaccharide (LPS
  • inorganic salts include aluminum hydroxide, zinc sulfate, colloidal iron hydroxide, calcium phosphate or calcium chloride.
  • an antibody disclosed herein may be combined with, e.g., an anti-CTLA-4 antibody in combination with an anti-OX40 antibody and a TLR9 ligand such as CpG (Marabelle et al, 2013).
  • the active agent that may be used according to the present invention may be a small molecule.
  • a small molecule disclosed herein may be: (a) a p300 inhibitor (Liu et al, 2013), such as gemcitabine (low dose) or C646 or analogs thereof, i.e. a compound of the formula I:
  • R 2 is selected from H, -C0 2 R 6 , -CONR 6 R 7 , -S0 3 H, or -S0 2 NR 6 R 7 ;
  • R 2 is selected from H, -C0 2 R 6 , or halogen, preferably CI;
  • R 3 is selected from halogen, preferably F, -N0 2 , -CN, -C0 2 R 6 , preferably C0 2 CH 3 or C0 2 CH 2 CH 3 , or -CH 2 OH;
  • R 4 and R 5 each independently is H or -C r C 6 alkyi, preferably methyl ;
  • R 6 is H or -Ci-C 6 alkyi, preferably H, methyl or ethyl;
  • R 7 is H or -Ci-C 6 alkyi, preferably H or methyl ;
  • the agent is a p300 inhibitor, which formulas are listed in Table 2, i.e. C646 (4-(4-((5-(4,5-dimethyl-2-nitrophenyl)furan-2-yl)methylene)-3-methyl-5-oxo-4,5-dihydro-1 H-pyrazol- 1 -yl)benzoic acid), C146 (4-hydroxy-3-(((2-(3-iodophenyl)benzo[d]oxazol-5-yl)imino)methyl)benzoic acid) or C375 (2-chloro-4-(5-((2,4-dioxo-3-(2-oxo-2-(p-tolylamino)ethyl)thiazolidin-5-ylidene)methyl)furan-2- yl)benzoic acid).
  • the p300 inhibitor is C646.
  • the adenosine receptor antagonist may be CGS15943 (9-Chloro-2-(2- furanyl)-[1 ,2,4]triazolo[1 ,5-c]quinazolin-5-amine); the adenosine A1 receptor antagonist may be PSB 36 (1 -Butyl-8-(hexahydro-2,5-methanopentalen-3a(1 H)-yl)-3,7-dihydro-3-(3-ydroxypropyl)-1 H-purine-2,6- dione); the adenosine A2a receptor antagonist may be SCH58261 (5-Amino-7-(2-phenylethyl)-2-(2-furyl)- pyrazolo(4,3-e)-1 ,2,4-triazolo(1 ,5-c)pyrimidine), SYN1 15 (4-Hydroxy-N-[4-methoxy-7-(4-morpholinyl
  • the small molecule inhibitor of the indoleamine-2,3-dioxygenase pathway may be Indoximod (NSC-721782/NLG-9189 (1 -Methyl-D-tryptophan), NewLink Genetics), INCB024360 ((4E)-4-[(3-chloro-4-fluoroanilino)-nitrosomethylidene]-1 ,2,5-oxadiazol-3-amine, Incyte) or NLG-919 (1 - Cyclohexyl-2-(5H-imidazo[5,1 -a]isoindol-5-yl)ethanol), NewLink Genetics).
  • the HIF-1 regulator may be M30, (5-[N-methyl-N-propargylaminomethyl]-8-hydroxyquinoline) described in Zheng et al. (Zheng et al, 201 5).
  • an active agent that may be used according to the present invention may be any combination of an antibody disclosed herein and a small molecule disclosed herein. In aspects of this embodiment, an active agent may be any combination of antibody disclosed herein and small molecule disclosed herein. [075] In certain embodiments, an active agent that may be used according to the present invention may be a protein selected from the group consisting of: (a) Neem leaf glycoprotein (NLGP; (Roy et al, 2013)); and/or (b) sCTLA-4 (soluble isoform of CTLA-4) (Ward et al, 2013).
  • an active agent that may be used according to the present invention may be a silencing molecule.
  • a silencing molecule is selected from the group consisting of miR-126 antisense (Qin et al, 2013) and anti-galectin-1 (Gal-1 ; (Dalotto-Moreno et al, 2013)).
  • an active agent that may be used according to the present invention may be an OK-432 (lyophilized preparation of Streptococcus pyogenes) (Hirayama et al, 2013).
  • an active agent that may be used according to the present invention may be a combination of IL-12 and anti-CTLA-4.
  • the agent can be derived from a broad spectrum of antibiotics which targets gram-positive and gram-negative bacteria, and thereby facilitating immunomodulation of Tregs, e.g. vancomycin which targets gram-positive bacteria and has been shown to reduce Treg levels/activity (Brestoff & Artis, 2013; Smith et al, 2013).
  • the active agent is administered by a dosage regime comprising at least two courses of therapy, each course of therapy comprising in sequence a treatment session followed by an interval session of non-treatment.
  • treatment session is used interchangeably herein with the terms “treatment period” or “period of treatment” and refers to a session during which one or more active agents disclosed herein are administered to the individual being treated.
  • a treatment session can be a single dosing event, or can be a multiple dosing regimen that occurs over a period of time.
  • a treatment session results in a therapeutically effective amount of an active agent disclosed herein to be consistently maintained throughout the treatment session.
  • non-treatment session is used interchangeably herein with the terms “non-treatment period”, “period of no treatment”, “interval session” or “interval session of non-treatment” and refers to a period of time during which no active agent disclosed herein is administered to the individual being treated.
  • the cessation of active agent administration during the non-treatment session results in the reduction of an active agent disclosed herein to sub-therapeutic levels in the individual being treated.
  • a “non-treatment session” is not the same event as a period of time that intervenes between a dosing event making up a multiple dosing regimen that occurs over a period of time during a treatment session. If administration of one or more active agents disclosed herein during a treatment session is a repeated administration, the non-treatment session is longer than the intervening period between these repeated administrations during the treatment session.
  • the dosage regime may be determined in a number of ways.
  • the level of immunosuppression may be calibrated to a desired level for each patient who is being treated (personalized medicine), by monitoring the level or activity of IFN-y-producing leukocytes or proliferation rate of leukocytes in response to stimulation individually, and adjusting the treatment session, the frequency of administration and the interval session empirically and personally as determined from the results of the monitoring.
  • the treatment session may comprise administering the active agent or pharmaceutical composition to the individual and the treatment session is maintained at least until the systemic presence or level of IFN-y-producing leukocytes, or the rate of proliferation of leukocytes in response to stimulation rises above a reference, the administering is paused during the interval session, and the interval session is maintained as long as the level is above the reference, wherein the reference is selected from (a) the level of systemic presence or activity of IFN-y-producing leukocytes, or the rate of proliferation of leukocytes in response to stimulation, measured in the most recent blood sample obtained from said individual before said administering; or (b) the level of systemic presence or activity of IFN-y-producing leukocytes, or the rate of proliferation of leukocytes in response to stimulation, characteristic of a population of individuals afflicted with a disease, disorder, condition or injury of the CNS.
  • the reference is selected from (a) the level of systemic presence or activity of IFN-y-producing leukocytes, or the rate of proliferation of
  • the length of the treatment and non-treatment or interval sessions may be determined by physicians in clinical trials directed to a certain patient population and then applied consistently to this patient population, without the need for monitoring the level of immunosuppression on a personal basis.
  • the treatment session comprises administering the active agent to the individual and the treatment session is maintained at least until the systemic presence of the active agent reaches therapeutic levels, the administering is paused during the interval session, and the interval session is maintained as long as the level is above about 95%, 90%, 80%, 70%, 60% or 50% of said therapeutic level.
  • therapeutic level refers to generally accepted systemic levels of drugs used to block immune checkpoints in known therapies, such as cancer therapy (see above).
  • the treatment session comprises administering the active agent to the individual and the treatment session is maintained at least until the systemic presence or activity of the active agent reaches a therapeutic level, at which point the administration is then stopped, and a non- treatment period is maintained as long as the systemic presence or activity of the active agent is maintained above a threshold therapeutic level.
  • a threshold therapeutic level is a level that is, e.g. at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, of the therapeutic level.
  • therapeutic level refers to generally accepted systemic levels of drugs used to block immune checkpoints in known therapies, such as cancer therapy (see above).
  • the active agent is an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti- B7RP1 antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-B7-H7 antibody, an anti-BTLA antibody, an anti-HVEM antibody, an anti-CD-27 antibody, an anti-CD40 antibody, an anti-CD40L antibody, an anti-CD70 antibody, an anti-CD80 antibody, an anti-CD86 antibody, an anti-CD137 antibody, an anti-CD137L antibody, an anti-OX40 antibody, an anti-OX40L antibody, an anti-TIM-3 antibody, an anti-Galectin9 antibody, an anti-KIR antibody, an anti-LAG-3 antibody, an anti-ICOS antibody, an anti-VISTA antibody, an anti-STING, an anti-TIGIT, anti-GITR or any combination thereof.
  • the treatment session comprises administering the active agent to the individual and the treatment session is maintained at least until the systemic presence or activity of the active agent reaches a therapeutic level, at which point the administration is then stopped, and a non- treatment period is maintained as long as a beneficial effect on cognition is maintained above the level before treatment commencement.
  • a beneficial effect on cognition is maintained is one that shows an improvement of, e.g. at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, above the cognition level before treatment commencement.
  • the active agent is an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-B7RP1 antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-B7-H7 antibody, an anti-BTLA antibody, an anti-HVEM antibody, an anti-CD-27 antibody, an anti-CD40 antibody, an anti-CD40L antibody, an anti-CD70 antibody, an anti-CD80 antibody, an anti-CD86 antibody, an anti-CD137 antibody, an anti-CD137L antibody, an anti-OX40 antibody, an anti-OX40L antibody, an anti-TIM-3 antibody, an anti-Galectin9 antibody, an anti-KIR antibody, an anti-LAG-3 antibody, an anti-ICOS antibody, an anti-VISTA antibody, an anti-STING, an anti-TIGIT, anti-GITR or any combination thereof.
  • the treatment session comprises administering the active agent to the individual and the treatment session is maintained at least until the systemic presence or activity of the active agent reaches a therapeutic level, at which point the administration is then stopped, and a non- treatment period is maintained as long as a beneficial effect on vision is maintained above the level before treatment commencement.
  • a beneficial effect on vision is maintained is one that shows an improvement of, e.g. at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, above the vision level before treatment commencement.
  • the active agent is an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-B7RP1 antibody, an anti- B7-H3 antibody, an anti-B7-H4 antibody, an anti-B7-H7 antibody, an anti-BTLA antibody, an anti-HVEM antibody, an anti-CD-27 antibody, an anti-CD40 antibody, an anti-CD40L antibody, an anti-CD70 antibody, an anti-CD80 antibody, an anti-CD86 antibody, an anti-CD137 antibody, an anti-CD137L antibody, an anti-OX40 antibody, an anti-OX40L antibody, an anti-TIM-3 antibody, an anti-Galectin9 antibody, an anti-KIR antibody, an anti-LAG-3 antibody, an anti-ICOS antibody, an anti-VISTA antibody, an anti-STING, an anti-TIGIT, anti-GITR or any combination thereof.
  • the treatment session may be a single administration or it may comprise multiple administrations given during a prescribed period of time.
  • a treatment session may be multiple administrations given in the course of between, e.g., 1 day to four weeks, 2 days to four weeks, 3 days to four weeks, 4 days to four weeks, 5 days to four weeks, 6 days to four weeks, one week and four weeks, 10 days and four weeks, two weeks and four weeks, 17 days and four weeks or three weeks and four weeks.
  • the treatment session may comprise two administrations both given within one week, such as, e.g., the second administration given 1 , 2, 3, 4, 5 or 6 days after the first administration.
  • the treatment session may comprise three administrations all given within one week such as, e.g., given 1 , 2 or 3 days after the preceding administration.
  • the treatment session may comprise three administrations all given within two week such as, e.g., given 1 , 2, 3, 4 or 5 days after the preceding administration.
  • the treatment session may comprise four administrations all given within two week such as, e.g., given 1 , 2, 3 or 4 days after the preceding administration.
  • the treatment session may comprise four administrations all given within three week such as, e.g., given 1 , 2, 3, 4, 5 or 6 days after the preceding administration.
  • the treatment session may comprise five administrations all given within three week such as, e.g., given 1 , 2, 3, 4 or 5 days after the preceding administration.
  • the interval session of non-treatment may be between one week and six months, for example between 2 weeks to 4 weeks, 3 weeks to 4 weeks, 2 weeks to 6 weeks, 3 weeks to 6 weeks, 4 weeks to 6 weeks, 5 weeks to 6 weeks, 2 weeks to 2 months, 3 weeks to 2 months, 4 weeks to 2 months, 5 weeks to 2 months, 6 weeks to 2 months, 7 weeks to 2 months, 2 months to 3 months, 2 months to 4 months, 3 months to 4 months, 3 months to 5 months, 3 months to 5 months, 4 months to 5 months, 1 week to 6 months, 2 weeks to 6 months, 3 weeks to 6 months, 4 weeks to 6 months, 6 weeks to 6 months, 2 months to 6 months, 3 months to 6 months, 4 months to 6 months or 5 months to 6 months.
  • the interval session of non-treatment may be 1 to 2 months in length, 1 to 3 months in length or 2 to 3 months in length.
  • the administration of the active agent or pharmaceutical composition may be a single administration or repeated administration, for example the active agent or pharmaceutical composition may be administered only once and then immediately followed by an interval, or it may be administered daily, or once every two, three, four, five or six days, or once weekly, once every two weeks, once every three weeks or once every four weeks.
  • These frequencies are applicable to any active agent, may be based on commonly used practices in the art, and may finally be determined by physicians in clinical trials.
  • the frequency of the repeated administration in the treatment session could be adapted according to the nature of the active agent, wherein for example, a small molecule may be administered daily and an antibody may be administered once every 3 days.
  • an agent when administered during a treatment session at a relatively low frequency, for example once per week during a treatment session of one month, or once per month during a treatment session of six months, this treatment session is followed by a non-treatment interval session, the length of which is longer than the period between the repeated administrations during the treatment session (i.e. longer than one week or one month, respectively, in this example).
  • the pause of one week or one month between the administrations during the treatment session in this example is not considered an interval session.
  • the lengths of the treatment session and the non-treatment or interval session may be adjusted to the frequency of the administration such that, for example, a frequency of administering the active agent once every 3 days may result in a treatment session of 6 or 9 days and an interval session that is commenced accordingly.
  • the dosage regimen is determined by the length of the non-treatment interval, so that a single administration is followed by a non-treatment interval of 7, 8, 9, 10, 14, 18, 21 , 24 or 28 days or longer before the next single-administration treatment session.
  • the dosage regimen consists of single administrations interspersed with non- treatment intervals of non-treatment of 2, 3 or 4 weeks.
  • the dosage regimen may consist of single administrations interspersed with non-treatment intervals of non-treatment of 2 to 4 weeks, 2 to 3 weeks or 3 to 4 weeks.
  • the dosage regimen is determined by the length of the non-treatment interval, so that multiple administrations given within one week is followed by a non-treatment interval of 7, 10, 14, 1 8, 21 , 24 or 28 days or longer before the next multiple- administration treatment session.
  • the dosage regimen may consist of multiple administrations given within one week interspersed with non-treatment intervals of non-treatment of 2 or 3 or 4 weeks.
  • the dosage regimen may consist of multiple administrations given within one week interspersed with intervals of non-treatment of 2 to 4 weeks, 2 to 3 weeks or 3 to 4 weeks.
  • the dosage regimen may comprise multiple administrations given within two weeks followed by a non-treatment interval of 2 weeks, 3 weeks or 1 , 2, 3 or 4 months or longer before the next multiple-administration treatment session.
  • the dosage regimen may consist of multiple administrations given within two weeks interspersed with intervals of non-treatment of 1 , 2, 3 or 4 months.
  • the dosage regimen may consists of multiple administrations given within two week interspersed with intervals of non-treatment of 1 to 2 months, 1 to 3 months, 1 to 4 months, 2 to 3 months, 2 to 4 months or 3 to 4 months.
  • the dosage regimen may comprise multiple administrations given within three week followed by 1 , 2, 3, 4, 5 or 6 months or longer of non-treatment before the next multiple- administration treatment session.
  • the dosage regimen may consist of multiple administrations given within three weeks interspersed with intervals of non-treatment of 1 , 2, 3, 4, 5 or 6 months.
  • the dosage regimen may consists of multiple administrations given within three weeks interspersed with intervals of non-treatment of 1 to 2 months, 1 to 3 months, 1 to 4 months, 1 to 5 months, 1 to 6 months, 2 to 3 months, 2 to 4 months, 2 to 5 months, 2 to 6 months, 3 to 4 months, 3 to 5 months, 3 to 6 months, 4 to 5 months, 4 to 6 months or 5 to 6 months.
  • a flexible dosage regimen is envisioned that starts with a certain regimen and is replaced with another.
  • treatment sessions each one including 2 single administrations 3 days apart, with an interval of for example 1 week between the treatment sessions, could be replaced when considered appropriate by a dosage regimen including treatment sessions of single administrations separated by for example 2, 3 or 4 weeks intervals.
  • treatment sessions, each one including 2 single administrations 7 days apart, with an interval of for example 2 weeks between the treatment sessions could be replaced when considered appropriate by a dosage regimen including treatment sessions of single administrations separated by for example 2, 3, 4, 5 or 6 weeks intervals.
  • treatment sessions, each one including 3 single administrations 3 days apart, with an interval of for example 2 weeks between the treatment sessions could be replaced when considered appropriate by a dosage regimen including treatment sessions of single administrations separated by for example 2, 3, 4, 5 or 6 weeks intervals.
  • the dosage regimen i.e. the length of the treatment session and the interval session, is calibrated such that the reduction in the level of immunosuppression, for example as measured by a reduction in the level of systemic presence or activity of regulatory T cells or the increase in the level of systemic presence or activity of IFN- ⁇ producing leukocytes in the individual, is transient.
  • the method, active agent or pharmaceutical composition according to the present invention may be for treating a disease, disorder or condition of the CNS that is a neurodegenerative disease, disorder or condition.
  • the neurodegenerative disease, disorder or condition is Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease Huntington's disease, primary progressive multiple sclerosis; secondary progressive multiple sclerosis, corticobasal degeneration, Rett syndrome, a tauopathy, a retinal degeneration disorder; anterior ischemic optic neuropathy; glaucoma; uveitis; depression; trauma-associated stress or post-traumatic stress disorder, frontotemporal dementia, Lewy body dementias, mild cognitive impairments, posterior cortical atrophy, primary progressive aphasia or progressive supranuclear palsy.
  • the condition of the CNS is aged-related dementia.
  • the condition of the CNS is Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease Huntington's disease, primary progressive multiple sclerosis; secondary progressive
  • Tauopathies are a clinically, morphologically and biochemically heterogeneous class of neurodegenerative diseases characterized by a pathological aggregation of tau protein in neurofibrillary or gliofibrillary tangles in the human brain.
  • Tau is a microtubule-associated protein (MAP) that binds to microtubules and promotes their polymerization. It plays an important role in maintaining axonal transport and neuronal integrity but has a physiological role in dendrites, and it is expressed at low levels in glial cells.
  • a tauopathy tangles are formed by hyperphosphorylation of tau causing it to aggregate in an insoluble form.
  • Non-limiting examples of tauopathies include Alzheimer's disease, argyrophilic grain disease, chronic traumatic encephalopathy, corticobasal degeneration, dementia pugilistica, frontotemporal dementia, frontotemporal lobar degeneration, Hallervorden-Spatz disease, Huntington's disease, ganglioglioma, gangliocytoma, globular glial tauopathy, lead encephalopathy, lipofuscinosis, Lytico-Bodig disease (Parkinson-dementia complex of Guam), meningioangiomatosis, Parkinsonism disease linked to chromosome 17, Pick's disease, primary age-related tauopathy (PART), formerly known as neurofibrillary tangle-only dementia (NFT-dementia), postencephalitic parkinsonism, progressive supranuclear palsy, subacute sclerosing panencephalitis and tuberous sclerosis.
  • PART primary age-related tauopathy
  • Retinal degeneration disorders are ones that result in the deterioration of the retina due to the death of photoreceptor cells.
  • retinal degeneration There are several causes for retinal degeneration, including artery or vein occlusion, diabetic retinopathy, retrolental fibroplasia/retinopathy of prematurity, or disease (usually hereditary).
  • Symptoms include, without limitation, impaired vision, night blindness, retinal detachment, light sensitivity, glare sensitivity, tunnel vision, loss of depth perception, loss of contrast, night blindness, loss of central vision, loss of peripheral vision and total loss of vision.
  • Retinal degeneration disorders include, without limitation, Age-Related Macular Degeneration (wet and dry), Retinitis Pigmentosa, Choroideremia, Cone-Rod Retinal Dystrophy, Gyrate Atrophy, Juvenile Retinoschisis, Vitelliform Macular Dystrophy (Best's Disease), Abetalipoproteinemia (Bassen-Kornzweig Disease), Bardet-Biedl Syndrome, Blue Cone Monochromatism Disease, Dominant Drusen, Goldman-Favre Vitreoretinal Dystrophy (Enhanced S-cone Syndrome), Kearns-Sayre Syndrome, Laurence-Moon Syndrome, Leber's Congenital Amaurosis, Leber's Refsum disease, Oguchi Disease, Peripapillary (pericentral) Choroidal Dystrophy, Pigment Pattern Dystrophy, Sorsby Macular Dystrophy, Stargardt's Disease, Stickler's Syndrome, Usher Syndrome and Wagner'
  • VISTA V-domain
  • the method, active agent and pharmaceutical composition according to the present invention may further be for treating an injury of the CNS selected from spinal cord injury, closed head injury, blunt trauma, penetrating trauma, hemorrhagic stroke, ischemic stroke, cerebral ischemia, optic nerve injury, myocardial infarction, organophosphate poisoning and injury caused by tumor excision.
  • an injury of the CNS selected from spinal cord injury, closed head injury, blunt trauma, penetrating trauma, hemorrhagic stroke, ischemic stroke, cerebral ischemia, optic nerve injury, myocardial infarction, organophosphate poisoning and injury caused by tumor excision.
  • VISTA V-domain
  • the present invention improves the cognitive function in mice that emulates Alzheimer's disease.
  • the method, active agent and pharmaceutical composition may be for use in improving CNS motor and/or cognitive function, for example for use in alleviating age-associated loss of cognitive function, which may occur in individuals free of a diagnosed disease, as well as in people suffering from neurodegenerative disease.
  • the method, active agent and pharmaceutical composition may be for use in alleviating loss of cognitive function resulting from acute stress or traumatic episode.
  • the cognitive function mentioned herein above may comprise learning, memory or both.
  • mice that emulates Alzheimer's disease
  • 5XFAD AD-Tg mice begin to display cerebral plaque pathology at the ages of 2.5 months and cognitive deficits at the ages of 5 months (Oakley et al, 2006).
  • Example 2 the inventors describe the therapeutic effect in 5XFAD mice at 6 months of age, in Example 5 they characterize the therapeutic effect in 5XFAD mice at 1 1 and 12 months of age - an extremely progressive stage of amyloid beta plaque deposition and cognitive deficits in this model.
  • Stage 1 - Mild/Early lasts 2-4 years
  • Stage 2 - Moderate/Middle lasts 2-1 0 years
  • Stage 3 - Severe/Late lasts 1 -3+ years.
  • CNS function refers, inter alia, to receiving and processing sensory information, thinking, learning, memorizing, perceiving, producing and understanding language, controlling motor function and auditory and visual responses, maintaining balance and equilibrium, movement coordination, the conduction of sensory information and controlling such autonomic functions as breathing, heart rate, and digestion.
  • Cognition is formed in multiple areas of the brain such as hippocampus, cortex and other brain structures. However, it is assumed that long term memories are stored at least in part in the cortex and it is known that sensory information is acquired, consolidated and retrieved by a specific cortical structure, the gustatory cortex, which resides within the insular cortex.
  • cognitive function may be measured by any know method, for example and without limitation, by the clinical global impression of change scale (CIBIC-plus scale); the Mini Mental State Exam (MMSE); the Neuropsychiatric Inventory (NPI) ; the Clinical Dementia Rating Scale (CDR); the Cambridge Neuropsychological Test Automated Battery (CANTAB) or the Sandoz Clinical Assessment- Geriatric (SCAG).
  • Cognitive function may also be measured indirectly using imaging techniques such as Positron Emission Tomography (PET), functional magnetic resonance imaging (fMRI), Single Photon Emission Computed Tomography (SPECT), or any other imaging technique that allows one to measure brain function.
  • An improvement of one or more of the processes affecting the cognition in a patient will signify an improvement of the cognitive function in said patient, thus in certain embodiments improving cognition comprises improving learning, plasticity, and/or long term memory.
  • improving cognition comprises improving learning, plasticity, and/or long term memory.
  • the term "learning” relates to acquiring or gaining new, or modifying and reinforcing, existing knowledge, behaviors, skills, values, or preferences.
  • plasticity relates to synaptic plasticity, brain plasticity or neuroplasticity associated with the ability of the brain to change with learning, and to change the already acquired memory.
  • One measurable parameter reflecting plasticity is memory extinction.
  • Memory relates to the process in which information is encoded, stored, and retrieved. Memory has three distinguishable categories: sensory memory, short-term memory, and long-term memory.
  • Long term memory is the ability to keep information for a long or unlimited period of time.
  • Long term memory comprises two major divisions: explicit memory (declarative memory) and implicit memory (non-declarative memory).
  • Long term memory is achieved by memory consolidation which is a category of processes that stabilize a memory trace after its initial acquisition. Consolidation is distinguished into two specific processes, synaptic consolidation, which occurs within the first few hours after learning, and system consolidation, where hippocampus-dependent memories become independent of the hippocampus over a period of weeks to years.
  • the present invention provides methods for reducing ⁇ -plaque burden in a patient diagnosed with Alzheimer's disease, comprising administering to said patient an active agent or pharmaceutical composition as defined herein above that causes reduction of the level of systemic immunosuppression by release of a restraint imposed on the immune system by one or more immune checkpoints.
  • the present invention provides a method for reducing hippocampal gliosis in a patient diagnosed with Alzheimer's disease, comprising administering to said patient an active agent or pharmaceutical composition as defined herein above that causes reduction of the level of systemic immunosuppression by release of a restraint imposed on the immune system by one or more immune checkpoints.
  • compositions for use in accordance with the present invention may be formulated in conventional manner using one or more physiologically acceptable carriers or excipients.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof.
  • Methods of administration include, but are not limited to, parenteral, e.g., intravenous, intraperitoneal, intramuscular, subcutaneous, mucosal (e.g., oral, intranasal, buccal, vaginal, rectal, intraocular), intrathecal, topical and intradermal routes. Administration can be systemic or local.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the active agent is administered.
  • the carriers in the pharmaceutical composition may comprise a binder, such as microcrystalline cellulose, polyvinylpyrrolidone (polyvidone or povidone), gum tragacanth, gelatin, starch, lactose or lactose monohydrate; a disintegrating agent, such as alginic acid, maize starch and the like; a lubricant or surfactant, such as magnesium stearate, or sodium lauryl sulphate; and a glidant, such as colloidal silicon dioxide.
  • a binder such as microcrystalline cellulose, polyvinylpyrrolidone (polyvidone or povidone), gum tragacanth, gelatin, starch, lactose or lactose monohydrate
  • a disintegrating agent such as alginic acid, maize starch and the like
  • a lubricant or surfactant such as
  • the pharmaceutical preparation may be in liquid form, for example, solutions, syrups or suspensions, or may be presented as a drug product for reconstitution with water or other suitable vehicle before use.
  • Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid).
  • suspending agents e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats
  • emulsifying agents e.g., lecithin or acacia
  • non-aqueous vehicles e.g., almond oil, oily esters, or fractionated vegetable oils
  • preservatives e.g
  • compositions may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinized maize starch, polyvinyl pyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulphate).
  • binding agents e.g., pregelatinized maize starch, polyvinyl pyrrolidone or hydroxypropyl methylcellulose
  • fillers e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate
  • lubricants e.g., magnesium stearate, talc or silica
  • disintegrants e.g., potato starch
  • Preparations for oral administration may be suitably formulated to give controlled release of the active compound.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • compositions may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen free water, before use.
  • compositions may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • compositions for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or
  • a method of treating a treating a disease, disorder, condition or injury of the Central Nervous System to an individual in need thereof comprising administering to the individual a composition comprising an active agent that causes reduction of the level of systemic immunosuppression by release of a restraint imposed on the immune system by one or more immune checkpoints, wherein the composition is administered by a dosage regime comprising at least one course of therapy, each course of therapy comprising in sequence a treatment session where the composition is administered to the individual followed by a non-treatment session where the composition is not administered to the individual, wherein the non-treatment period is longer than the treatment session; wherein, if administration of the composition during the treatment session is a repeated administration, the non- treatment period is longer than the period between repeated administrations during the treatment session; wherein administration of the composition transiently reduces levels of systemic immunosuppression and increases choroid plexus gateway activity in facilitating selective recruitment of immune cells into the central nervous system, thereby treating the individual.
  • An active agent, or a pharmaceutical composition comprising the active agent for use in treating a treating a disease, disorder, condition or injury of the Central Nervous System, wherein the active agent causes reduction of the level of systemic immunosuppression by release of a restraint imposed on the immune system by one or more immune checkpoints, wherein the active agent or pharmaceutica composition is administered by a dosage regime comprising at least one course of therapy, each course of therapy comprising in sequence a treatment session where the composition is administered to the individual followed by a non-treatment session where the composition is not administered to the individual, wherein the non-treatment period is longer than the treatment session; wherein, if administration of the composition during the treatment session is a repeated
  • the non-treatment period is longer than the period between repeated administrations during the treatment session; wherein administration of the composition transiently reduces levels of systemic immunosuppression and increases choroid plexus gateway activity in facilitating selective recruitment of immune cells into the central nervous system, thereby treating the individual.
  • the active agent is an antibody, an antibody mimetic, an aptamer, a small molecule, a Neem leaf glycoprotein, a sCTLA-4, a silencing molecule, an OK-432, a combination of IL-12 and anti-CTLA-4, or any combination thereof.
  • the antibody is an anti-PD-1 , an anti-PD-L1 , an anti-PD-L2, an anti-CTLA-4, an anti-CD80,an anti-CD86, an anti-B7RP1 , an anti-B7-H3, an anti-B7-H4, an anti-B7-H7, an anti-BTLA, an anti- HVEM, an anti-CD-27, an anti-CD40, an anti-CD40L, an anti-CD70, an anti-CD80, an anti-CD86, an anti-CD137, an anti-CD137L, an anti-OX40, an anti-OX40L, an anti-TIM-3, an anti-Galectin9, an anti- KIR, an anti-LAG-3, an anti-ICOS, an anti-VISTA, an anti-STING, an anti-TIGIT, anti-
  • the antibody mimetic is an affibody molecule, an affilin, an affimer, an affitin, an alphabody, an anticalin, an avimer, a DARPin, a fynomer, a Kunitz domain peptide, or a monobody.
  • the aptamer is a DNA aptamer, a RNA aptamer, a XNA aptamer or a peptide aptamer.
  • the small molecule is a p300 inhibitor, a Sunitinib, a Polyoxometalate-1 , an ⁇ , ⁇ -methyleneadenosine 5'-diphosphate, an arsenic trioxide, a GX15-070, a retinoic acid antagonist, a CCR4 antagonist, an adenosine receptor antagonist, an adenosine A1 receptor antagonist; an adenosine A2a receptor, an adenosine A2b receptor antagonist, an A3 receptor antagonist, an antagonist of indoleamine-2,3-dioxygenase or an HIF-1 regulator.
  • the small molecule is a p300 inhibitor, a Sunitinib, a Polyoxometalate-1 , an ⁇ , ⁇ -methyleneadenosine 5'-diphosphate, an arsenic trioxide, a GX15-070, a retinoic acid antagonist, a CCR4 antagonist, an a
  • the one or more immune checkpoints includes a PD-1 -PD-L1 , a PD-1 -PD-L2, a CD28-CD80, a CD28-CD86, a CTLA-4-CD80, a CTLA-4-CD86, an ICOS-B7RP1 , a B7H3, a B7H4, a B7H7, a B7-CD28-like molecule, a BTLA- HVEM, a KIR-MHC class I or II, a LAG3-MHC class I or II, a CD137-CD137L, an OX40-OX40L, a CD27-CD70, a CD40L-CD40, a TIM3-GAL9, a V-domain Ig suppressor of T cell activation (VISTA), a STimulator of INterferon Genes (STING),
  • VISTA V-domain Ig suppressor of T cell activation
  • STING STimulator of INterfer
  • any one of embodiments 1 -35, the active agent or pharmaceutical composition according to any one of embodiments 2-35, or the use according to any one of embodiments 3-35 wherein the administration of the composition during the treatment session is maintained at least until a systemic presence or activity of IFNy-producing leukocytes and/or an IFNy cytokine rises above a reference, at which point the administration is stopped, and the non-treatment period is maintained as long as the systemic presence or activity of IFNy-producing leukocytes and/or an IFNy cytokine is above the reference, wherein the reference includes a) a level of a systemic presence or activity of IFNy-producing leukocytes and/or an IFNy cytokine measured in the most recent blood sample obtained from the individual before the administering; or b) a level of a systemic presence or activity of IFNy-producing leukocytes and/or an IFNy cytokine characteristic of a population of individuals afflicted with the disease, disorder, condition or
  • a cerebral level of soluble amyloid beta peptide is reduced in the individual
  • a cerebral amyloid beta ( ⁇ ) plaque burden is reduced or cleared in the individual
  • a hippocampal gliosis is reduced in the individual
  • a cerebral level of a pro-inflammatory cytokine is reduced in the individual
  • a brain inflammation is decreased in the individual and/or a cognitive function is improved in the individual.
  • the method according to embodiment 37, the active agent or pharmaceutical composition according to embodiments 37, or the use according to embodiment 37, wherein the improved cognitive function is learning, memory, creation of imagery, plasticity, thinking, awareness, reasoning, spatial ability, speech and language skills, language acquisition, capacity for judgment attention or any combination thereof.
  • T cells include regulatory T cells.
  • the disease, disorder, condition or injury of the Central Nervous System is Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease Huntington's disease, primary progressive multiple sclerosis, secondary progressive multiple sclerosis, corticobasal degeneration, Rett syndrome, anterior ischemic optic neuropathy, glaucoma, uveitis, depression, trauma-associated stress or post-traumatic stress disorder, frontotemporal dementia, Lewy body dementias, mild cognitive impairments, posterior cortical atrophy, primary progressive aphasia or progressive supranuclear palsy
  • tauopathy is Alzheimer's disease, argyrophilic grain disease, chronic traumatic encephalopathy, corticobasal degeneration, dementia pugilistica, frontotemporal dementia, frontotemporal lobar degeneration, Hallervorden-Spatz disease, Huntington's disease, ganglioglioma, gangliocytoma, globular glial tauopathy, lead encephalopathy, lipofuscinosis, Lytico-Bodig disease (Parkinson-dementia complex of Guam), meningioangiomatosis, Parkinsonism disease linked to chromosome 17, Pick's disease, primary age- related tauopathy (PART), formerly known as neurofibrillary tangle-only dementia (NFT-dementia), postencephalitic parkinsonism, progressive supranuclear palsy, subacute sclerosing panencepha
  • the retinal degeneration disorder is wet Age-Related Macular Degeneration, dry Age-Related Macular Degeneration, Retinitis Pigmentosa, Choroideremia, Cone-Rod Retinal Dystrophy, Gyrate Atrophy, Juvenile Retinoschisis, Vitelliform Macular Dystrophy (Best's Disease), Abetalipoproteinemia (Bassen-Kornzweig Disease), Bardet-Biedl Syndrome, Blue Cone Monochromatism Disease, Dominant Drusen, Goldman-Favre Vitreoretinal Dystrophy (Enhanced S-cone Syndrome), Kearns-Sayre Syndrome, Laurence-Moon Syndrome, Leber's Congenital Amaurosis, Leber's Refsum disease, Oguchi Disease, Peripapillary (pericentral) Choroidal Dystrophy, Pigment Pattern Dy
  • mice Animals. 5XFAD transgenic mice (Tg6799) that co-overexpress familial AD mutant forms of human APP (the Swedish mutation, K670N/M671 L; the Florida mutation, 1716V; and the London mutation, V717I) and PS1 (M146L/L286V) transgenes under transcriptional control of the neuron-specific mouse Thy-1 promoter (Oakley et al, 2006), and AD double transgenic B6.Cg-Tg (APPswe, PSEN1 dE9) 85Dbo/J mice (Borchelt et al, 1997) were purchased from The Jackson Laboratory. Genotyping was performed by PCR analysis of tail DNA, as previously described (Oakley et al, 2006).
  • Heterozygous mutant cx 3 cr1 GFPI+ mice (Jung et al, 2000) (B6.129P-cx3cr7 im ' L ' j, in which one of the CX 3 CR1 chemokine receptor alleles was replaced with a gene encoding GFP) were used as donors for BM chimeras.
  • Foxp3.LuciDTR mice (Suffner et al, 2010) were bred with 5XFAD mice to enable conditional depletion of Foxp3 + Tregs. Animals were bred and maintained by the Animal Breeding Center of the Weizmann Institute of Science. All experiments detailed herein complied with the regulations formulated by the Institutional Animal Care and Use Committee (IACUC) of the Weizmann Institute of Science.
  • RNA purification, cDNA synthesis, and quantitative real-time PCR analysis Total RNA of the hippocampal dentate gyrus (DG) was extracted with TRI Reagent (Molecular Research Center) and purified from the lysates using an RNeasy Kit (Qiagen). Total RNA of the choroid plexus was extracted using an RNA MicroPrep Kit (Zymo Research). mRNA (1 ⁇ g) was converted into cDNA using a High Capacity cDNA Reverse Transcription Kit (Applied Biosystems). The expression of specific mRNAs was assayed using fluorescence-based quantitative real-time PCR (RT-qPCR).
  • RT-qPCR fluorescence-based quantitative real-time PCR
  • RT-qPCR reactions were performed using Fast-SYBR PCR Master Mix (Applied Biosystems). Quantification reactions were performed in triplicate for each sample using the standard curve method. Peptidylprolyl isomerase A (pp/ ' a) was chosen as a reference (housekeeping) gene. The amplification cycles were 95 °C for 5 s, 60°C for 20 s, and 72°C for 15 s. At the end of the assay, a melting curve was constructed to evaluate the specificity of the reaction.
  • the cDNA was pre-amplified in 14 PCR cycles with non-random PCR primers, thereby increasing the sensitivity of the subsequent real-time PCR analysis, according to the manufacturer's protocol (PreAmp Master Mix Kit; Applied Biosystems).
  • mRNA expression was determined using TaqMan RT-qPCR, according to the manufacturer's instructions (Applied Biosystems). All RT-qPCR reactions were performed and analyzed using StepOne software V2.2.2 (Applied Biosystems). The following TaqMan Assays-on-DemandTM probes were used: Mm02342430_g1 (ppia) and Mm01 168134_m1 (ifn- ⁇ ).
  • mice were transcardially perfused with PBS prior to tissue excision and fixation.
  • CP tissues were isolated under a dissecting microscope (Stemi DV4; Zeiss) from the lateral, third, and fourth ventricles of the brain.
  • tissue were fixated with 2.5% paraformaldehyde (PFA) for 1 hour at 4°C, and subsequently transferred to PBS containing 0.05% sodium azide.
  • PFA paraformaldehyde
  • the dissected tissues were washed with PBS and blocked (20% horse serum, 0.3% Triton X-100, and PBS) for 1 h at room temperature.
  • mice anti- ⁇ (1 :300, Covance, #SIG-39320); rabbit anti-GFP (1 :1 00, MBL, #598); rat anti-CD68 (1 :300, eBioscience, #14-0681 ); rat anti-ICAM-1 (1 :200, Abeam, #AB2213); goat anti-GFP (1 :100, Abeam, #ab6658); rabbit anti-IBA-1 (1 :300, Wako, #019-19741 ); goat anti-IL-1 0 (1 :20, R&D systems, #AF519); rat anti-Foxp3 (1 :20, eBioscience, #13-5773- 80); rabbit anti-CD3 (1 :500, Dako, #IS503);; mouse anti-ZO-1 , mouse anti-E-Cahedrin, and rabbit anti- Claudin-1 (all 1 :100, Invitrogen, #33-9100, #33-4000, #51 -9000); rabbit
  • Intracellular labeling of cytokines was done with BD Cytofix/CytopermTM Plus fixation/permeabilization kit (cat. no. 555028).
  • Cytofix/CytopermTM Plus fixation/permeabilization kit cat. no. 555028.
  • eBioscience FoxP3 staining buffer set cat. no. 00-5523-00 was used.
  • fluorochrome-labeled monoclonal antibodies were purchased from BD Pharmingen, BioLegend, R&D Systems, or eBiosciences, and used according to the manufacturers' protocols: PE or Alexa Fluor 450-conjugated anti-CD4; PE-conjugated anti-CD25; PerCP-Cy5.5-conjugated anti-CD45; FITC-conjugated anti-TCRP; APC-conjugated anti-IFN- ⁇ ; APC-conjugated anti-FoxP3; Brilliant-violet- conjugated anti-CD45.
  • Cells were analyzed on an LSRII cytometer (BD Biosciences) using FlowJo software. In each experiment, relevant negative control groups, positive controls, and single stained samples for each tissue were used to identify the populations of interest and to exclude other populations.
  • BM chimeras were prepared as previously described (Shechter et al, 2009; Shechter et al, 2013). In brief, gender-matched recipient mice were subjected to lethal whole- body irradiation (950 rad) while shielding the head (Shechter et al, 2009). The mice were then injected intravenously with 5x10 6 BM cells from CX 3 CR1 GFP + donors. Mice were left for 8-10 weeks after BM transplantation to enable reconstitution of the hematopoietic lineage, prior to their use in experiments.
  • the percentage of chimerism was determined by FACS analysis of blood samples according to percentages of GFP expressing cells out of circulating monocytes (CD1 1 b + ). In this head-shielded model, an average of 60% chimerism was achieved, and CNS-infiltrating GFP + myeloid cells were verified to be CD45 h ' 9h /CD1 1 b h ' 9h , representing monocyte-derived macrophages and not microglia (Shechter et al, 2013).
  • the inter-trial interval for each mouse was 10 min. On day 5, the platform was removed, and mice were given a single trial lasting 60s without available escape. On days 6 and 7, the platform was placed in the quadrant opposite the original training quadrant, and the mouse was retrained for three sessions each day. Data were recorded using the EthoVision V7.1 automated tracking system (Noldus Information Technology). Statistical analysis was performed using analysis of variance (ANOVA) and the Bonferroni post-hoc test. All MWM testing was performed between 10 a.m. and 5 p.m. during the lights-off phase.
  • RAWM radial-arm water maze
  • the radial-arm water maze (RAWM) was used to test spatial learning and memory, as was previously described in detail (Alamed et al, 2006). Briefly, six stainless steel inserts were placed in the tank, forming six swim arms radiating from an open central area. The escape platform was located at the end of one arm (the goal arm), 1 .5 cm below the water surface, in a pool 1 .1 m in diameter. The water temperature was kept between 21 -22°C. Water was made opaque with milk powder. Within the testing room, only distal visual shape and object cues were available to the mice to aid in location of the submerged platform. The goal arm location remained constant for a given mouse.
  • mice were trained for 15 trials (spaced over 3 h), with trials alternating between a visible and hidden platform, and the last 4 trails with hidden platform only.
  • mice were trained for 15 trials with the hidden platform. Entry into an incorrect arm, or failure to select an arm within 15 sec, was scored as an error. Spatial learning and memory were measured by counting the number of arm entry errors or the escape latency of the mice on each trial. Training data were analyzed as the mean errors or escape latency, for training blocks of three consecutive trials.
  • ATRA treatment All-trans retinoic acid (ATRA) administration to mice was performed similarly to previously described (Walsh et al, 2014). ATRA (Sigma) was dissolved in DMSO and injected i.p. (8 mg kg "1 d "1 ) every other day over the course of 1 week. Vehicle-treated mice were similarly injected with DMSO.
  • Soluble ⁇ (sAP) protein isolation and quantification Tissue homogenization and sAp protein extraction was performed as previously described (Schmidt et al, 2005). Briefly, cerebral brain parenchyma was dissected and snap-frozen and kept at -80 S C until homogenization.
  • Proteins were sequentially extracted from samples to obtain separate fractions containing proteins of differing solubility.
  • Samples were homogenized in 10 volumes of ice-cold tissue homogenization buffer, containing 250mM of sucrose, 20mM of Tris base, 1 mM of ethylenediaminetetraacetic acid (EDTA), and 1 mM of ethylene glycol tetraacetic acid (pH 7.4), using a ground glass pestle in a Dounce homogenizer. After six strokes, the homogenate was mixed 1 :1 with 0.4% diethylamine (DEA) in a 100-mM NaCI solution before an additional six strokes, and then centrifuged at 135,000g at 4 S C for 45 min.
  • DEA diethylamine
  • the supernatant (DEA-soluble fraction containing extracellular and cytosolic proteins) was collected and neutralized with 10% of 0.5Mof Tris-HCI (pH 6.8).
  • ⁇ 1-40 and Ap ⁇ were individually measured by enzyme-linked immunosorbent assay (ELISA) from the soluble fraction using commercially available kits (Biolegend; #SIG-38954 and #SIG- 38956, respectively) according to the manufacturer instructions.
  • [159] ⁇ plaque quantitation. From each brain, 6 ⁇ coronal slices were collected, and eight sections per mouse, from four different pre-determined depths throughout the region of interest (dentate gyrus or cerebral cortex) were immunostained. Histogram-based segmentation of positively stained pixels was performed using the Image-Pro Plus software (Media Cybernetics, Bethesda, MD, USA). The segmentation algorithm was manually applied to each image, in the dentate gyrus area or in the cortical layer V, and the percentage of the area occupied by total Ap immunostaining was determined. Plaque numbers were quantified from the same 6 ⁇ coronal brain slices, and are presented as average number of plaques per brain region. Prior to quantification, slices were coded to mask the identity of the experimental groups, and plaque burden was quantified by an observer blinded to the identity of the groups.
  • Example 1 Choroid plexus (CP) gateway activity along disease progression in the mouse model of AD.
  • AD-Tg 5XFAD transgenic mouse model of AD
  • WT age-matched wild-type mice
  • Example 2 The functional relationships between Treg-mediated systemic immune suppression, CP gateway activity, and AD pathology.
  • Tregs Regulatory T cells
  • Regulatory T cells play a pivotal role in suppressing systemic effector immune responses (Sakaguchi et al, 2008).
  • Treg-mediated systemic immune suppression affects IFN- ⁇ availability at the CP, and therefore focused on the involvement of Tregs in AD pathology.
  • Example 3 Weekly administration of Copolymer- 1 reduces Treg-mediated systemic immune suppression, improves CP gateway activity, and mitigates AD pathology.
  • Example 4 Interference with Treg activity using a small molecule histone acetyltransferase inhibitor.
  • mice treated with p300i compared to vehicle (DMSO) treated controls, showed elevated levels of systemic I FN- ⁇ - expressing cells in the spleen (Fig. 10A), as well as in the CP (Fig. 10B).
  • AD-Tg mice with either p300i or vehicle over the course of 1 week, and examined the animals 3 weeks later for cerebral ⁇ plaque burden. Immunohistochemical analysis revealed a significant reduction in cerebral ⁇ plaque load in the p300i treated AD-Tg mice (Fig. 10C-E). We also tested whether the effect on plaque pathology following one course of treatment would last beyond the 3 weeks, and if so, whether additional courses of treatment would contribute to a long-lasting effect. We therefore compared AD-Tg mice that received a single course of p300i treatment and were examined 2 month later, to an age-matched group that received two courses of treatments during this period, with a 1 -month interval in between (schematically depicted in Fig. 10F).
  • Example 5 Therapeutic potential of PD-1 immune checkpoint blockade in Alzheimer's disease.
  • AD-Tg mice 5XFAD AD transgenic mice
  • AD-Tg mice 5XFAD AD transgenic mice
  • I.p. mice at the age of 1 0 months, a time point at which cerebral pathology is advanced, were administrated with two intraperitoneal (i.p.) injections of either blocking antibodies directed at PD-1 (anti-PD-1 ) or IgG control antibodies, on days 1 and 4, and then examined on day 7.
  • Flow cytometry analysis revealed that blockade of the PD-1 pathway resulted in elevated frequencies of I FN-y-producing CD4 + T splenocytes (Fig. 11 A, B).
  • Table 3. GO annotation, related to Figure 11.
  • AD-Tg mice One month following treatment (two i.p. injections with 3-day interval), anti-PD1 treated AD-Tg mice exhibited a significant improvement in cognitive function relative to IgG-treated or untreated age- matched controls, reaching cognitive levels similar to that of age-matched WT mice (Fig. 12A).
  • Control groups included WT mice, untreated AD-Tg mice, and AD-Tg mice that received two sessions of IgG treatment.
  • AD-Tg mice that received two sessions of anti-PD-1 displayed cognitive performance similar to that of WT mice, at the end of the 2-month timeframe (Fig. 12B).
  • Aged matched wild-type (WT) mice were used as additional control group.
  • Black arrows indicate time points of treatment, and illustrations indicate time points of cognitive testing. Repeated-measures were analyzed using two-way ANOVA and Dunnett post-test. Error bars represent mean ⁇ s.e.m.
  • anti-PD-1 -treated (1 injection) versus IgG-treated controls One month following treatment (two i.p. injections with 3-day interval), anti-PD1 treated AD-Tg mice exhibited a significant improvement in cognitive function relative to IgG-treated or untreated age-matched controls, reaching cognitive levels similar to that of age-matched WT mice (Fig. 14).
  • mice Male 5XFAD AD transgenic mice were treated in a repeated treatment session, once a month, with either anti-PD-1 -specific antibody (lgG2a anti-mouse PD-1 ) or IgG control (Rat lgG2a).
  • the first injection was at the age of 3 months, the second at the age of 4 months, and the third at the age of 5 months. Dosage is indicated in the scheme of the experimental design (Fig. 15A).
  • WT Aged matched wild- type mice were used as additional control group.
  • Treatment effect on spatial learning and memory performance was evaluated using the radial arm water maze (RAWM) task, at two different time points - the age of 5 months (Fig. 15B), and the age of 6 months (Fig. 15C).
  • RAWM radial arm water maze
  • Black arrows indicate time points of treatment, and illustrations indicate time points of cognitive testing.
  • Repeated- measures were analyzed using two-way ANOVA and Dunnett post-test. Error bars represent mean ⁇ s.e.m.; * P ⁇ 0.05, ** P ⁇ 0.01 , *** P ⁇ 0.001 , anti-PD-1 -treated versus IgG-treated controls.
  • the control IgG-treated mice have not fully lost spatial learning/memory skills, and thus exhibited some learning on the last trial of the second day (Fig.
  • FIG. 15B whereas at the age of 6 months, disease progression was observed, with a further decrease in functional performance (Fig. 15C,D).
  • Figure 15D illustrates the decline in the IgG-treated mice between the ages of 5 and 6 months, while the anti-PD-1 antibody treated group retained learning capacity.
  • mice treated with anti-PD-1 antibody showed a reduction in activated Caspase-3 immunoreactivity in Neu-N + neurons, as compared to the IgG-treated group (Fig. 15G, H), further substantiating the effect of the anti-PD-1 antibody treatment on neuronal survival.
  • Example 6 Therapeutic potential of TIM-3 immune checkpoint blockade in Alzheimer's disease.
  • Example 7 Therapeutic potential of PD-L1 immune checkpoint blockade in Alzheimer's disease, and comparison to anti-PD-1 treatment.
  • PD-1 is an inhibitory receptor expressed by numerous immune cells, among which are effector CD4 T cells, while its ligand, PD-L1 , is expressed by dendritic cells, epithelial cells and regulatory T cells.
  • PD-L1 its ligand
  • dendritic cells a cell that express CD4 T cells
  • dendritic cells a cell that express CD4 T cells
  • PD-L1 a ligand that is expressed by dendritic cells, epithelial cells and regulatory T cells.
  • Fig. 17A 6-month old 5XFAD mice were first treated with a single dose of anti PD-L1 antibody (0.1 mg, 0.5mg, or 1 .5mg per mouse), injected intraperitoneal ⁇ . Mice were assessed 1 month later, at the age of 7 months, using RAWM.
  • PD-L1 is expressed by activated immune cells such as T cells, B cells, macrophages, dendritic cells and microglia, as well as by non-immune cells such as endothelial and epithelial cells.
  • activated immune cells such as T cells, B cells, macrophages, dendritic cells and microglia
  • non-immune cells such as endothelial and epithelial cells.
  • the expression of PD-L1 by the CP epithelium might contribute to the down regulation of the trafficking of leukocytes to the CNS, by dampening the activity of IFN- ⁇ producing T-cells which express PD-1 upon communication with PD-L1 -expressing epithelial cells within the CP.
  • Immunohistochemical analysis shows that in aged mice the CP epithelium expressed significantly higher levels of PD-L1 , compared to young mice (Fig. 18).
  • Example 8 Therapeutic potential of immune checkpoint blockade in Alzheimer's disease.
  • AD-Tg mice are treated at ages between 6 to10-month old with one of the following anti-checkpoint antibodies: anti-ICOS, anti-B7RP1 , anti-VISTA, anti-CD40, anti-CD40L, anti-CD80, anti-CD86, anti-B7-H3, anti-B7-H4, B7-H7, anti-BTLA, anti-HVEM, anti-CD137, anti-CD137L, anti-OX40L, anti-CD-27, anti-CD70, anti-STING, anti- TIGIT antibody or anti-GITR antibody.
  • anti-ICOS anti-B7RP1
  • anti-VISTA anti-CD40
  • anti-CD40L anti-CD80
  • anti-CD86 anti-B7-H3, anti-B7-H4, B7-H7, anti-BTLA, anti-HVEM
  • anti-CD137, anti-CD137L, anti-OX40L anti-CD-27, anti-CD70, anti-STING, anti- TIGIT
  • mice are treated with anti-PD-1 antibody as positive control, IgG control as negative control or combinations of anti-PD1 and one of the other anti-checkpoint antibodies mentioned above.
  • Treatment effect on spatial learning and memory performance, using the radial arm water maze (RAWM) task, ⁇ plaque burden by immunohistochemistry for ⁇ and hippocampal astrogliosis by immunohistochemistry for glial fibrillary acid protein (GFAP) will be measured one month following treatment.
  • RAWM radial arm water maze
  • ⁇ plaque burden by immunohistochemistry for ⁇ and hippocampal astrogliosis by immunohistochemistry for glial fibrillary acid protein (GFAP) will be measured one month following treatment.
  • GFAP glial fibrillary acid protein
  • Example 9 Therapeutic potential of PD-1 in combination with CTLA-4 immune checkpoint blockade in Alzheimer's disease.
  • AD Alzheimer's' disease
  • Tg 5XFAD Alzheimer's' disease (AD) transgenic mice are injected i.p. with either 25( ⁇ g of anti-PD1 (RMP1 -14; #BE0146; Bioxcell Lifesciences Pvt. LTD.) and 25( ⁇ g anti-CTLA-4 (InVivoMAb anti-mCD152; #BE0131 ; Bioxcell Lifesciences Pvt. LTD.) or control IgG (lgG2a, #BE0089 or Polyclonal Syrian Hamster IgG, #BE0087; Bioxcell Lifesciences Pvt. LTD.) antibodies, on day 1 and day 4 of the experiment, and are examined 3 weeks after for their cognitive performance by radial arm water maze (RAWM) spatial learning and memory task, as described above.
  • RAWM radial arm water maze
  • mice receive an additional treatment session with an interval session of 3 weeks.
  • Control groups are either treated with IgG or untreated, and all groups of mice are tested for their cognitive performance 3 weeks later.
  • mice treated with the combination of antibodies display significant cognitive improvement in comparison to IgG-treated and untreated AD-Tg mice as well as a significant reduction of cerebral plaque load.
  • Example 10 Therapeutic potential of immune checkpoint blockade approach in PTSD pathology.
  • mice are habituated for 10 days to a reverse day/night cycle, inflicted with two episodes of electrical shocks (the trauma and the trigger), referred to as a "PTSD induction", and evaluated at different time points subsequent to trauma. Following the traumatic event mice are injected with said compound which blocks immune checkpoints. The mice are treated according to one of the following regimens:
  • mice are treated with one of the following anti-checkpoint antibodies: anti-ICOS, anti-B7RP1 , anti-VISTA, anti-CD40, anti-CD40L, anti-CD80, anti-CD86, anti-B7-H3, anti-B7-H4, B7-H7, anti-BTLA, anti-HVEM, anti-CD137, anti-CD137L, anti-OX40L, anti-CD-27, anti-CD70, anti-STING, anti-GITR or anti- TIGIT antibody alone or in combination with an anti-CTLA-4 antibody.
  • Some mice are treated with anti- PD-1 antibody as positive control, IgG control as negative control or combinations of anti-PD1 and one of the other anti-checkpoint antibodies mentioned above.
  • mice receive an additional treatment session with an appropriate interval session.
  • mice that receive the treatment do not display anxiety behavior associated with PTSD in this experimental model, as assessed by time spent exploring and risk assessing in dark/light maze or the other behavioral tasks described in (Lebow et al, 2012).
  • mice are treated at the progressive stages of disease according to one of the following regimens:
  • PD-Tg mice are treated with one of the following anti-checkpoint antibodies: anti-ICOS, anti- B7RP1 , anti-VISTA, anti-CD40, anti-CD40L, anti-CD80, anti-CD86, anti-B7-H3, anti-B7-H4, B7-H7, anti- BTLA, anti-HVEM, anti-CD137, anti-CD137L, anti-OX40L, anti-CD-27, anti-CD70, anti-STING, anti-GITR or anti-TIGIT antibody alone or in combination with an anti-CTLA-4 antibody.
  • Some mice are treated with anti-PD-1 antibody as positive control, IgG control as negative control or combinations of anti-PD1 and one of the other anti-checkpoint antibodies mentioned above.
  • mice treated with one treatment session show significant improved motor performance, compared to IgG-treated or vehicle treated control group, or untreated group.
  • PD-Tg mice which receive two courses of therapy, and examined after an appropriate interval session are expected to show a long-lasting therapeutic effect. To maintain this therapeutic effect mice are subjected to an active session of treatment with an appropriate interval session of non-treatment between each treatment session.
  • Example 12 Therapeutic potential of PD-1 in combination with CTLA-4 immune checkpoint blockade in Huntington's disease pathology.
  • the model used in these experiments may be the Huntington's disease (HD) R6/2 transgenic mice (Tg) test system.
  • R6/2 transgenic mice over express the mutated human huntingtin gene that includes the insertion of multiple CAG repeats mice at the progressive stages of disease. These mice show progressive behavioral-motor deficits starting as early as 5-6 weeks of age, and leading to premature death at 10-13 weeks.
  • the symptoms include low body weight, clasping, tremor and convulsions.
  • mice are treated according to one of the following regimens when they are 45 days old:
  • mice are treated with one of the following anti-checkpoint antibodies: anti-ICOS, anti-B7RP1 , anti-VISTA, anti-CD40, anti-CD40L, anti-CD80, anti-CD86, anti-B7-H3, anti-B7-H4, B7-H7, anti-BTLA, anti-HVEM, anti-CD137, anti-CD137L, anti-OX40L, anti-CD-27, anti-CD70, anti-STING, anti-GITR or anti- TIGIT antibody alone or in combination with an anti-CTLA-4 antibody.
  • Some mice are treated with anti- PD-1 antibody as positive control, IgG control as negative control or combinations of anti-PD1 and one of the other anti-checkpoint antibodies mentioned above.
  • HD-Tg mice treated with one treatment session show significant improved motor performance, compared to IgG-treated or vehicle treated control group, or untreated group.
  • HD-Tg mice which receive which receive two courses of therapy, and examined after an appropriate interval session are expected to show a long-lasting therapeutic effect.
  • mice are subjected to an active session of treatment with an appropriate interval session of non-treatment between each treatment session.
  • Example 13 Therapeutic potential of immune checkpoint blockade approach in amyotrophic lateral sclerosis pathology.
  • the model used in this experiment may be the transgenic mice overexpressing the defective human mutant SOD1 allele containing the Gly93 ⁇ Ala (G93A) gene (B6SJL-TgN (SOD1 -G93A)1 Gur (herein "ALS mice”).
  • This model develop motor neuron disease and thus constitute an accepted animal model for testing ALS.
  • mice are treated according to one of the following regimens when they are 75 days old:
  • mice are treated with one of the following anti-checkpoint antibodies: anti-ICOS, anti-B7RP1 , anti-VISTA, anti-CD40, anti-CD40L, anti-CD80, anti-CD86, anti-B7-H3, anti-B7-H4, B7-H7, anti-BTLA, anti-HVEM, anti-CD137, anti-CD137L, anti-OX40L, anti-CD-27, anti-CD70, anti-STING, anti-GITR or anti- TIGIT antibody alone or in combination with an anti-CTLA-4 antibody.
  • Some mice are treated with anti- PD-1 antibody as positive control, IgG control as negative control or combinations of anti-PD1 and one of the other anti-checkpoint antibodies mentioned above.
  • mice are allowed to grasp and hold onto a vertical wire (2 mm in diameter) with a small loop at the lower end.
  • a vertical wire allows mice to use both fore- and hindlimbs to grab onto the wire.
  • the wire is maintained in a vertically oriented circular motion (the circle radius was 1 0 cm) at 24 rpm. The time that the mouse is able to hang onto the wire is recorded with a timer.
  • mice treated with one treatment session show significant improved motor performance, compared to IgG-treated or vehicle treated control group, or untreated group.
  • ALS mice which receive which receive two courses of therapy, and examined after an appropriate interval session are expected to show a long-lasting therapeutic effect.
  • mice are subjected to an active session of treatment with an appropriate interval session of non-treatment between each treatment session.
  • Example 14 Dose effect experiments to determine minimal and maximal dose range and experiments to determine treatment regimen and its long lasting therapeutic effect.
  • mice treated with the anti-PD-1 antibodies display significant reduction in cerebral amyloid beta plaque load in comparison to untreated AD-Tg mice or to control IgG-treated mice.
  • 5XFAD AD-Tg mice are injected with the drug at a dosage that will be determined according to the previous study results. Mice will be injected and their cognitive performance is monitored using the radial arm water maze learning and memory task during and after the study period. Histological examination of the brain for amyloid plaque burden is also performed.
  • mice are injected repeatedly with single injections (or double injections 3 days apart as described in Example 5) with 2, 3 or 4 weeks intervals of non-treatment (Table 4). The mice are monitored as described above at one, two or three months after the initial treatment.
  • Example 15 Systemic administration of anti-PD-1 monoclonal antibody in RCS rats attenuates retinal degeneration.
  • the aim of this experiment was to determine whether systemic administration of an anti-PD1 antibody attenuates the degeneration of the outer nuclear layer in an animal model of a retinal degeneration disease.
  • RCS rats an accepted animal model of dry AMD and Retinitis Pigmentosa and other retinal degenerative diseases and conditions, carry a deletion mutation in the gene encoding the MerTK protein which leads to retinal degeneration and complete loss of sight by the age of three months.
  • a significant and rapid deterioration in the thickness of the retinal outer nuclear layer (ONL) is observed starting at the age of 4 weeks. Preservation of ONL thickness in this model is thus considered in the scientific literature to be directly correlated to preservation of sight.
  • Example 16 Local administration of anti-PD-1 monoclonal antibody directly into the vitreous in RCS rats attenuates retinal degeneration.
  • the aim of this experiment was to determine whether local administration of an anti-PD1 antibody directly into the vitreous of the eye attenuates the degeneration of the outer nuclear layer in an animal model of a retinal degeneration disease.
  • Example 17 Local PD-1 blockade through intravitreal injection of PD-1 or PD-L1 monoclonal antibodies attenuates retinal degeneration.
  • Anti-PD-L1 monoclonal antibodies or anti-PD-1 monoclonal antibodies will be injected directly into the vitreous of RCS rats at 4 weeks of age. Throughout the 8 following weeks, assessment of visual function of the animals in response to visual stimuli will be taken. Importantly, only a single eye of each animal will receive the treatment while the contra-lateral eye will be left untreated and will serve as an additional control.
  • Example 18 Targeting PD-1/PD-L1 pathway in a mouse model of Tau pathology enhances recruitment of monocyte-derived macrophages to the brain parenchyma.
  • Example 19 Blockade of the PD-1/PD-L1 axis in a mouse model of Tau pathology mitigates cognitive deficits and cerebral pathology.
  • mice Age matched wild type mice were assessed as an additional control for normal learning/memory performance in the tests used in this study.
  • T maze and Y maze tests which measure short-term spatial memory, are commonly used.
  • the two groups that received either anti-PD-1 or anti-PD-L1 antibodies demonstrated increased preference for the novel arm in the T-maze assay as compared to the IgG-treated control group, 1 month after a single injection of the antibodies (Fig. 22B, C).
  • both treatment groups exhibited improved cognitive performance in the Y-maze test, compared to the IgG-treated control group (Fig. 22D).
  • mice treated with 0.5 or 1 .5 mg /mouse showed performance approaching that of WT mice; the anti-PD-L1 dose of 1 .5 mg/mouse was slightly more effective, but the difference between the doses was not significnat (Fig. 23G).
  • Example 20 PD-1 blockade enhances hippocampal neurogenesis in 5XFAD mice.
  • Doublecortin (DCX) is a microtubule-associated protein expressed by neuronal precursor cells and immature neurons. In the adult neuronal tissue DCX is used as a marker for neurogenesis since it is expressed almost solely by developing neurons.
  • Fig. 24A Brain sections were immunostained for neuronal marker-NeuN (in green), DCX (in red), and hoechst nuclear staining (in blue). DCX+ cells were quantified in a double-blinded manner from 6m thick brain slices (Fig. 24B). Repeated measures were analyzed using one-way ANOVA and Dunnett post-test. Error bars represent mean ⁇ s.e.m. ; * P ⁇ 0.05, ** P ⁇ 0.01 , *** P ⁇ 0.001 .
  • Example 21 PD-1 blockade enhances hippocampal synaptic plasticity in 5XFAD mice.
  • VGLUT1 Vesicular glutamate transporters 1 (VGLUT1 ), expressed by glutamatergic neurons, mediate glutamate uptake into synaptic vesicles and was shown to contribute to hippocampal synaptic plasticity and hippocampus-dependent spatial learning.
  • Aged matched wild-type (WT; n 9) mice were used as additional control group. Parasagittal brain sections from representative animals were prepared and the Subiculum region was marked (Fig.
  • Example 22 PD-1 blockade reduces neuronal loss in the Subiculum of 5XFAD mice.
  • the 5XFAD transgenic mouse model is one of the few amyloid animal models that exhibits significant neuron loss, similar to AD progression in human patients. Neuronal loss in 5XFAD mice was characterized in the Subiculum and cortical Layer 5.
  • Aged matched wild-type (WT; n 9) mice were used as additional control group. Parasagittal brain sections from representative animals were prepared and the Subiculum region was marked (Fig. 26A).
  • the meaning of the open-ended transitional phrase “comprising” is being defined as encompassing all the specifically recited elements, limitations, steps and/or features as well as any optional, additional unspecified ones.
  • the meaning of the closed-ended transitional phrase “consisting of” is being defined as only including those elements, limitations, steps and/or features specifically recited in the claim whereas the meaning of the closed-ended transitional phrase “consisting essentially of” is being defined as only including those elements, limitations, steps and/or features specifically recited in the claim and those elements, limitations, steps and/or features that do not materially affect the basic and novel characteristic(s) of the claimed subject matter.
  • the open-ended transitional phrase “comprising” includes within its meaning, as a limiting case, claimed subject matter specified by the closed-ended transitional phrases “consisting of” or “consisting essentially of.”
  • claimed subject matter specified by the closed-ended transitional phrases “consisting of” or “consisting essentially of.”
  • embodiments described herein or so claimed with the phrase “comprising” are expressly or inherently unambiguously described, enabled and supported herein for the phrases “consisting essentially of” and “consisting of.”
  • Butovsky O Koronyo-Hamaoui M, Kunis G, Ophir E, Landa G, Cohen H, Schwartz M (2006) G!atiramer acetate fights against Alzheimer's disease by inducing dendr!tic-iike microglia expressing insulin-like growth factor 1 .
  • Butovsky O Kunis G, Koronyo-Hamaoui M, Schwartz M (2007) Selective ablation of bone marrow-derived dendritic ceils increases amyloid plaques in a mouse Alzheimer's disease model.
  • Blocking retinoic acid receptor-alpha enhances the efficacy of a dendritic cell vaccine against tumours by suppressing the induction of regulatory T cells. Cancer immunology, immunotherapy: Cli 62: 1273-1282
  • Tim-3/galectin-9 pathway involves in the homeostasis of hepatic Tregs in a mouse mode! of concanavalin A-induced hepatitis.
  • Neem leaf glycoprotein overcomes indoleamine 2,3 dioxygenase mediated tolerance in dendritic cells by attenuating hyperactive regulatory T ceils in cervical cancer stage 1MB patients.
  • Torres K C Araujo Pereira P, Lima G S, Bozzi ! C, Rezende V B, Blcaiho M A, Moraes E N, Miranda D M, Romano-Sliva M A (2013) Increased frequency of T cells expressing IL-10 in Alzheimer disease but not in late-onset depression patients.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Ophthalmology & Optometry (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Saccharide Compounds (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

La présente invention concerne une composition pharmaceutique comprenant un agent actif qui provoque la réduction du niveau de l'immunosuppression systémique chez un individu, destinée à être utilisée dans le traitement d'une maladie, d'un trouble, d'une affection ou d'une lésion du système nerveux central SNC. Cette composition pharmaceutique est administrée par un régime posologique comprenant au moins un cycle de traitement, chaque cycle de traitement comprenant successivement une session de traitement suivie d'une session intermédiaire sans traitement.
EP17848290.7A 2016-09-10 2017-09-08 Réduction des quantités ou de l'activité de cellules t régulatrices systémiques pour le traitement de maladie et de lésion du snc Withdrawn EP3509636A4 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP23184844.1A EP4269439A3 (fr) 2016-09-10 2017-09-08 Réduction des quantités ou de l'activité de cellules t régulatrices systémiques pour le traitement de maladie et de lésion du snc

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US15/261,945 US10519237B2 (en) 2014-03-12 2016-09-10 Reducing systemic regulatory T cell levels or activity for treatment of disease and injury of the CNS
PCT/IB2016/001433 WO2017042633A2 (fr) 2015-09-10 2016-09-10 Réduction des quantités ou de l'activité de cellules t régulatrices systémiques pour le traitement de maladie et de lésion du snc
PCT/IL2017/051011 WO2018047178A1 (fr) 2016-09-10 2017-09-08 Réduction des quantités ou de l'activité de cellules t régulatrices systémiques pour le traitement de maladie et de lésion du snc

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP23184844.1A Division EP4269439A3 (fr) 2016-09-10 2017-09-08 Réduction des quantités ou de l'activité de cellules t régulatrices systémiques pour le traitement de maladie et de lésion du snc

Publications (2)

Publication Number Publication Date
EP3509636A1 true EP3509636A1 (fr) 2019-07-17
EP3509636A4 EP3509636A4 (fr) 2020-12-16

Family

ID=61561788

Family Applications (2)

Application Number Title Priority Date Filing Date
EP23184844.1A Pending EP4269439A3 (fr) 2016-09-10 2017-09-08 Réduction des quantités ou de l'activité de cellules t régulatrices systémiques pour le traitement de maladie et de lésion du snc
EP17848290.7A Withdrawn EP3509636A4 (fr) 2016-09-10 2017-09-08 Réduction des quantités ou de l'activité de cellules t régulatrices systémiques pour le traitement de maladie et de lésion du snc

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP23184844.1A Pending EP4269439A3 (fr) 2016-09-10 2017-09-08 Réduction des quantités ou de l'activité de cellules t régulatrices systémiques pour le traitement de maladie et de lésion du snc

Country Status (9)

Country Link
EP (2) EP4269439A3 (fr)
JP (2) JP7074662B2 (fr)
KR (2) KR20240064742A (fr)
CN (2) CN108348607B (fr)
AU (1) AU2017322656B2 (fr)
CA (1) CA2997578A1 (fr)
IL (4) IL300982B1 (fr)
MX (1) MX2018003028A (fr)
WO (1) WO2018047178A1 (fr)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
US9567399B1 (en) 2016-06-20 2017-02-14 Kymab Limited Antibodies and immunocytokines
JP7198752B2 (ja) 2016-08-09 2023-01-04 カイマブ・リミテッド 抗icos抗体
EP3534947A1 (fr) 2016-11-03 2019-09-11 Kymab Limited Anticorps, combinaisons comprenant des anticorps, biomarqueurs, utilisations et procédés
GB201709808D0 (en) 2017-06-20 2017-08-02 Kymab Ltd Antibodies
US11629189B2 (en) 2017-12-19 2023-04-18 Kymab Limited Bispecific antibody for ICOS and PD-L1
EP3801571A4 (fr) * 2018-05-25 2022-08-10 Memorial Sloan Kettering Cancer Center Diagnostic et traitement d'une neurotoxicité induite par une immunothérapie
CN112638944A (zh) 2018-08-23 2021-04-09 西进公司 抗tigit抗体
CN111956799A (zh) * 2020-09-18 2020-11-20 生物抗素公司 抗pd-1抗体和/或pd-l1抗体在制备治疗帕金森病药物中的应用
CN112569351B (zh) * 2020-12-16 2021-10-15 南通大学 Cd44抗体在制备治疗帕金森疾病的药物中的应用

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050106586A1 (en) * 2003-06-13 2005-05-19 Mount Sinai Hospital Detection of neurodegenerative diseases
CN1838950A (zh) * 2003-06-23 2006-09-27 神经化学(国际)有限公司 治疗蛋白质聚集疾病的方法
DK1678194T3 (da) * 2003-10-10 2013-10-07 Alchemia Oncology Pty Ltd Modulering af syntese og degradering af hyaluronan i behandlingen af sygdom
US8993731B2 (en) * 2010-03-11 2015-03-31 Ucb Biopharma Sprl PD-1 antibody
CN102492038B (zh) 2011-12-09 2014-05-28 中国人民解放军军事医学科学院基础医学研究所 抗人Tim-3的中和性单克隆抗体L3D及其用途
EP3013355B1 (fr) * 2013-06-25 2019-08-07 ICM - Institut du Cerveau et da la Moelle Epinière Il-2 pour utilisation dans le traitement de la maladie d'alzheimer et de troubles associes
BR112016020919A2 (pt) * 2014-03-12 2018-01-23 Yeda Res & Dev redução dos níveis ou da atividade sistêmica de células t regulatórias para o tratamento de doença e lesão do snc
WO2017042633A2 (fr) * 2015-09-10 2017-03-16 Yeda Research And Development Co. Ltd Réduction des quantités ou de l'activité de cellules t régulatrices systémiques pour le traitement de maladie et de lésion du snc
US9394365B1 (en) * 2014-03-12 2016-07-19 Yeda Research And Development Co., Ltd Reducing systemic regulatory T cell levels or activity for treatment of alzheimer's disease
TWI664190B (zh) * 2014-06-27 2019-07-01 美商C2N醫療診斷有限責任公司 人類化抗-tau抗體
CN115960235A (zh) * 2015-08-28 2023-04-14 艾利妥 抗siglec-7抗体及其使用方法

Also Published As

Publication number Publication date
IL291784A (en) 2022-06-01
AU2017322656B2 (en) 2024-02-29
MX2018003028A (es) 2018-05-28
JP7451596B2 (ja) 2024-03-18
CN116492457A (zh) 2023-07-28
EP4269439A3 (fr) 2024-01-17
IL257885A (en) 2018-06-28
EP4269439A2 (fr) 2023-11-01
KR20190050931A (ko) 2019-05-14
JP2022106983A (ja) 2022-07-20
WO2018047178A1 (fr) 2018-03-15
CN108348607B (zh) 2023-06-23
IL257885B (en) 2022-05-01
EP3509636A4 (fr) 2020-12-16
IL291784B1 (en) 2023-05-01
KR20240064742A (ko) 2024-05-13
CA2997578A1 (fr) 2018-03-10
CN108348607A (zh) 2018-07-31
IL311841A (en) 2024-05-01
IL300982B1 (en) 2024-04-01
JP2019530635A (ja) 2019-10-24
IL291784B2 (en) 2023-09-01
IL300982A (en) 2023-04-01
JP7074662B2 (ja) 2022-05-24
AU2017322656A1 (en) 2018-04-19

Similar Documents

Publication Publication Date Title
US11884728B2 (en) Reducing systemic regulatory T cell levels or activity for treatment of amyotrophic lateral sclerosis
AU2017322656B2 (en) Reducing systemic regulatory T cell levels or activity for treatment of disease and injury of the CNS
US10519237B2 (en) Reducing systemic regulatory T cell levels or activity for treatment of disease and injury of the CNS
US11492405B2 (en) Reducing systemic regulatory t cell levels or activity for treatment of disease and injury of the CNS
WO2017042633A2 (fr) Réduction des quantités ou de l'activité de cellules t régulatrices systémiques pour le traitement de maladie et de lésion du snc
RU2815892C1 (ru) Снижение системных уровней или активности регуляторных т-клеток для лечения заболевания и поражения цнс
AU2024203073A1 (en) Reducing systemic regulatory T cell levels or activity for treatment of disease and injury of the CNS

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20180410

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 27/00 20060101ALI20200714BHEP

Ipc: A61P 25/28 20060101ALI20200714BHEP

Ipc: A61K 39/395 20060101AFI20200714BHEP

Ipc: C07K 16/28 20060101ALI20200714BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20201117

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 25/28 20060101ALI20201111BHEP

Ipc: A61P 27/00 20060101ALI20201111BHEP

Ipc: A61K 39/395 20060101AFI20201111BHEP

Ipc: C07K 16/28 20060101ALI20201111BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20230103

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20230712