WO2023111560A1 - Nouvelles méthode et composition comprenant de l'il-2 et une fraction de ciblage spécifique d'un tissu ou d'un organe - Google Patents

Nouvelles méthode et composition comprenant de l'il-2 et une fraction de ciblage spécifique d'un tissu ou d'un organe Download PDF

Info

Publication number
WO2023111560A1
WO2023111560A1 PCT/GB2022/053228 GB2022053228W WO2023111560A1 WO 2023111560 A1 WO2023111560 A1 WO 2023111560A1 GB 2022053228 W GB2022053228 W GB 2022053228W WO 2023111560 A1 WO2023111560 A1 WO 2023111560A1
Authority
WO
WIPO (PCT)
Prior art keywords
tissue
organ
cells
pharmaceutical composition
ageing
Prior art date
Application number
PCT/GB2022/053228
Other languages
English (en)
Inventor
James Dooley
Matthew Holt
Pierre LEMAITRE
Adrian LISTON
Emanuela PASCIUTO
Samar TAREEN
Lidia YSHII
Original Assignee
Babraham Institute
Vib Vzw
Katholieke Universiteit Leuven
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
Application filed by Babraham Institute, Vib Vzw, Katholieke Universiteit Leuven filed Critical Babraham Institute
Publication of WO2023111560A1 publication Critical patent/WO2023111560A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/761Adenovirus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/55IL-2
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination

Definitions

  • the invention relates to a pharmaceutical composition for use in a method of treating, preventing and/or reducing ageing, said composition comprising IL-2 and a targeting moiety specific for a tissue or organ of a subject, such as the brain and wherein the ageing is neurological ageing. Also provided are methods for treating, preventing and/or reducing ageing (e.g. neurological ageing) or cognitive decline, said methods comprising administration of a pharmaceutical composition as defined herein.
  • Aging is an irreversible process associated with physical deterioration and a progressive decline in memory, orientation, attention, motivation, and cognition (Hof & Morrison (2004), doi: https://doi.org/10.1016/i.tins.2004.07.013). Tissues composed primarily of post-mitotic cells, such as the brain, appear to be especially sensitive to the effects of ageing. Age-related cognitive decline has been attributed to several molecular processes, including chronic inflammation, impaired autophagy, macromolecular damage, and mitochondrial dysfunction and senescence (Hou et al. (2019), doi: https://doi.org/10.1038/s41582-019-0244-7).
  • Microglia from aged animals are hyper-reactive to inflammatory stimuli, with higher basal levels of cytokine expression and exaggerated responses to activation (Deczkowska et al. (2017), doi: https://doi.org/ 10.1838/s41467-017-00769-0.
  • Aged microglia exhibit shortening of processes, slower process movement, and enlarged soma volumes (Hefendehl et al. (2014), doi: https://doi.org/10.1111/acel.12149).
  • microglia Functional decline of microglia is hypothesized to be progressive over the lifespan, due to accumulation of oxidative DNA damage and non-degradable protein and lipid aggregates.
  • This inflamed and dysfunctional microglial profile of old age plays a role in the development of age-associated neurodegenerative diseases (Spittau (2017), doi: https://doi.org/10.3389/fnagi.2017.00194).
  • the ageing phenotype of microglia is driven in part by the breakdown of myelin (Safaiyan et al. (2016), doi: https://doi.org/10.1038/nn.4325) a phenomenon paralleled by the ageing of oligodendrocytes.
  • Oligodendrocytes are derived from specific neural progenitor cells, oligodendrocyte progenitor cells (OPCs), with the primary role of myelin production. OPC heterogeneity increases with age (Spitzer et al. (2019), doi: https://doi.org/10,1016/
  • CNS leukocytes may also play a role in age- related inflammation and neurodegeneration (Baruch, et al. (2013), doi: https: ://doi.org/10.1073/pnas.1211270110, Prokop et al. (2015), doi: https://doi_.org/10.1084/ jem.20150479, and Ritzel et al. (2016), doi: https ://doi.org/ 10.4049/iimmunol,1502021).
  • the accumulation of clonally expanded T cells is observed in the aged brain, accompanied by production of IFNy (Dulken et al.
  • AD Alzheimer's disease
  • PD Parkinson's disease
  • ALS amyotrophic lateral sclerosis
  • PPMS primary progressive multiple sclerosis
  • T cells have been observed in the substantia nigra of patients, and pharmacological manipulation of T cell responses alters the outcome of mouse models (Baird et al. (2019), doi: htps://doi.org/10.1016/ j.parkreldis 2018.10.029).
  • T cell infiltration is observed at sites of motor neuron loss in ALS patients, with modified disease progression in T cell-deficient mouse models of ALS (Zhao et al. (2013), doi: htps://doi.org/10.1007/s11481-013-9489-x).
  • the association of HLA alleles to AD and PD susceptibility supports a causative role of T cells in the process (International Parkinson Disease Genomics Consortium (2011), doi: https : //doi.org/10,1016/ s0140 ⁇ 6736(10)62345-8), Lambert et al. (2013), doi: httgs://doi.org/101038/ng.2802, and Hamza et al. (2010), doi: https: //doi .org/10.1038/ng.642) .
  • Tregs Regulatory T cells
  • Tregs reside in both lymphoid and in non-lymphoid organs, where they exert diverse functions regulating tissue homeostasis and contributing to tissue repair (Liston & Gray (2014), doi: https: //doi.org/10.1038/nri3605) .
  • Tregs are found in low numbers in the mouse and human brain, and are observed in both the parenchyma and meningeal lymphatics (Pasciuto et al. (2020), doi: https://doi.org/10.1016/j.cell.2020.06026).
  • Tregs have been proposed to have a protective function in a variety of neuroinflammatory or neurodegenerative diseases, with protective effects observed in mouse models of MS, AD, PD, ALS and stroke, among others ⁇ Reynolds et al. (2007), doi: .https://doi.org/10.1189/jlb.0507296, B0e2e6r)s. et al.
  • Proposed therapeutic strategies to harness Tregs are typically based on either direct delivery of Tregs, through cell-based approaches, or the provision of IL-2, the key survival factor for Tregs (Pierson et al. (2013), doi: https://doi.org/10.1038/ni2649). Despite this growing recognition of protective role of IL-2 and Tregs in neuroinflammatory and neurodegenerative processes, the potential of these cells to protect the ageing brain remain unknown.
  • a pharmaceutical composition comprising IL-2 and a targeting moiety specific for a tissue or organ of a subject for use in a method of treating, preventing and/or reducing ageing, wherein said tissue or organ is the central nervous system.
  • a method of expanding a population of regulatory T cells in a tissue or organ of a subject in need thereof for treating, preventing and/or reducing ageing comprising administering to the subject the pharmaceutical composition defined herein, wherein said tissue or organ is the central nervous system
  • a method of treating, preventing and/or reducing ageing in a tissue or organ of a subject in need thereof comprising administering to the subject the pharmaceutical composition defined herein, wherein said tissue or organ is the central nervous system.
  • the ageing is neurological ageing.
  • the tissue or organ is the brain.
  • a method of reprogramming cells of a tissue or organ in a subject in need thereof to a younger transcriptional program or signature comprising administration of the pharmaceutical composition defined herein, wherein said tissue or organ is the central nervous system.
  • a method of treating, preventing and/or reducing cognitive decline comprising administering to a subject in need thereof the pharmaceutical composition defined herein.
  • PHP.GFAP-IL2 or PHP.GFAP-GFP control vector
  • Each panel shows multipletesting corrected p-values for t-tests comparing the means of the samples between treatments within each age group, and the means of the two age groups irrespective of treatment.
  • DAM disease-associated microglia
  • E Proportions of DAM cells in each sample, with multipletesting corrected p-values for t-tests comparing the means of the samples between treatments within each age group, and the means of the two age groups irrespective of treatment.
  • Each panel shows multiple-testing corrected p-values fort-tests comparing the means of the samples between treatments within each age group, and the means of the two age groups irrespective of treatment.
  • D) The sum of deviance plot summarising the proportions of age groups in the trajectory trees generated for oligodendrocytes and precursors. The plot visualises the difference of proportion of cells by age in each branch of the respective trajectory tree relative to young PHP.GFAP-GFP treated mice, illustrating which age and treatment group shows the most deviation in the cell trajectory relative to the reference.
  • FIG. 4 Partial prevention of age-dependent transcriptional trajectory in astrocytes following IL-2-treatment.
  • Astrocyte clusters were reclustered and reprojected in LIMAP, with subset annotation based on key marker expression and mapping onto known astrocyte subtypes.
  • FIG. 5 Age-induced molecular changes in resident glia are mitigated by brainspecific IL-2 delivery.
  • Two-dimensional differential expression plots illustrating the contrast for normal aging (aged PHP.GFAP-GFP mice versus young PHP.GFAP-GFP mice) to the contrast for treatment-induced changes in aged mice (aged PHP.GFAP-IL2 mice versus aged PHP.GFAP-GFP).
  • FIG. 6 Age-induced pathways in glia are reverted to the young transcriptional state by local IL-2 production. Gene set enrichment was performed based on differential expression across the comparisons for age (aged PHP.GFAP-GFP mice versus young PHP.GFAP-GFP mice), treatment in young mice (young PHP.GFAP-GFP mice versus young PHP.GFAP-GFP mice), treatment in old mice (aged PHP.GFAP-IL2 mice versus aged PHP.GEAP-GFP mice) and age in treated mice (aged PHP.GEAP-IL2 mice versus young PHP.GEAP-IL2 mice). Gene sets were taken from the GAGE library and curated to the respective KEGG pathway.
  • pathways were manually curated for relevance to glial biology and non-redundancy from the unbiased list enriched in at least one of the four respective differential expression contrasts.
  • directionality maps were generated, summarising the change in direction of gene expression for member genes relative to the expression change with age (aged PHP.GFAP-GFP mice contrasted with young PHP.GEAP-GFP mice).
  • Directionality is shown for each pathway across each of the four contrasts for A) microglia, B) oligodendrocytes and OPCs, and C) total astrocytes and the identified astrocyte subsets.
  • FIG. 7 Synthetic IL-2 delivery prevents age-induced decline in spatial memory formation without altering age-induced degeneration in mobility or novelty-seeking behaviours.
  • Wildtype mice treated with PHP.GFAP-GFP control vector at two months of age were compared to aged mice treated with PHP.GEAP-IL2 (or PHP.GEAP-GFP control vector) at 22 months of age. Two months post-treatment, behaviour was tested.
  • A) Home cage activity was assessed through infrared registration of horizontal movements of single housed mice every 30 minutes (n 10, 10, 12). Left, activity during 24 h. Dotted line indicate day/night boundaries. Right, average activity counts.
  • B) Time spent on the rod, average of 4 repeated tests of 300 seconds (n 10,9,10).
  • C) Total distance moved in an open field test in trials of 1 day intervals (n 10,10,12).
  • D) Social preference to novel stranger over empty box (n 10, 10, 12).
  • E) Light-darktest (n 10,9,10). Left, distance moved in the lighted arena. Right, latency to enter the dark zone.
  • F-l) Spatial learning in the Morris water maze (n 10,10,12).
  • a and F 2-way ANOVA repeated measures with age and treatment as the main factors
  • B-D 2-way ANOVA with age and treatment as the main factors
  • G one-sample t-test to 25% chance level.
  • a pharmaceutical composition comprising IL-2 and a targeting moiety specific for a tissue or organ of a subject for use in a method of treating, preventing and/or reducing ageing, wherein said tissue or organ is the central nervous system.
  • the pharmaceutical composition leads to the expansion of a population of regulatory T cells in the tissue or organ targeted by the targeting moiety, i.e. the central nervous system, for the treatment, prevention and/or reduction of ageing.
  • a method of expanding a population of regulatory T cells in a tissue or organ of a subject in need thereof for treating, preventing and/or reducing ageing wherein said tissue or organ is the central nervous system.
  • the method comprises administration of a pharmaceutical composition comprising IL-2 and a targeting moiety specific for said tissue or organ as described herein.
  • the methods defined herein comprise expanding a population of cells, such as a population of regulatory T cells.
  • said expanding of a population of cells, such as a population of regulatory T cells is in a tissue or organ of a subject in need thereof, such as a particular tissue or organ of interest.
  • said expanding of a population of cells is in the central nervous system (e.g. the brain).
  • references herein to the terms “expanding ", “expansion” and “ expanded” or to the phrases “expanding a population of regulatory T cells” and” expanded population of regulatory T cells” include references to populations of cells which are larger than or comprise a larger number of cells than a non-expanded population. It will thus be appreciated that such an" expanded” population comprises a larger number of cells than a population which has not been subjected to IL-2.
  • the expanded population of cells such as an expanded population of regulatory T cells, comprises a larger number of cells compared to a reference population of cells.
  • the reference population of cells may be a population of cells not subjected to or administered with IL-2.
  • the expanded population of cells comprises a larger number of cells than the population prior to any administration of IL-2.
  • the reference population of cells may be located in a different tissue or organ to the expanded population of cells.
  • the expanded population of cells, such as an expanded population of regulatory T cells is an expanded population in a tissue or organ of a subject and comprises a larger number of cells compared to a population of cells not located in said tissue or organ of interest.
  • the expanded population of cells such as an expanded population of regulatory T cells, is located in a tissue or organ separated from other tissues or organs by a barrier (such as the blood-brain barrier) and comprises a larger number of cells compared to a population of cells not located with said barrier-separated tissue or organ.
  • the expanded population of cells is in the central nervous system (e.g. the brain) and comprises a larger number of cells compared to a population of cells located in a tissue or organ other than the central nervous system (e.g. the peripheral nervous system).
  • the expanded population of cells comprises a population at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 11- fold, at least 12-fold, at least 13-fold, at least 14-fold or more larger than a population of cells which has not been subjected to or administered with IL-2.
  • the expanded population of cells comprises a population at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 11 -fold, at least 12-fold, at least 13-fold, at least 14-fold or more larger than a population of cells not located in the tissue or organ of interest.
  • the expanded population of cells is at least 2- fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 12- fold, at least 13-fold or at least 14-fold larger than a reference population, such as a population of cells in the tissue or organ of interest which has not been subjected to or administered with IL-2 or a population of cells not located in the tissue or organ of interest.
  • a reference population such as a population of cells in the tissue or organ of interest which has not been subjected to or administered with IL-2 or a population of cells not located in the tissue or organ of interest.
  • the expanded population of cells such as an expanded population of regulatory T cells, comprises a larger proportion of cells which make up a subset of the population (e.g. a larger proportion of regulatory T cells within the total population of T cells in the tissue or organ).
  • the expanded population of regulatory T cells as defined herein may be expanded in a manner which is dependent on the dose of IL-2 administered.
  • the expanded population of regulatory T cells as defined herein comprises a population which is larger than a reference population by a factor which is IL-2 dose-dependent.
  • the expanded population of regulatory T cells comprises a population of cells which have increased survival.
  • the expanded population of regulatory T cells comprises increased survival.
  • the expanded population of regulatory T cells comprises decreased, or reduced, cell death.
  • the expanded population of regulatory T cells comprise increased proliferation.
  • the expanded population of regulatory T cells is larger than a reference population (e.g. a population of regulatory T cells not subjected to or administered with IL-2 or a population of cells not located in the tissue or organ of interest) because of increased survival of the expanded population of regulatory T cells.
  • the expanded population of regulatory T cells is larger than a reference population because of decreased, or reduced, cell death in the expanded population of regulatory T cells.
  • the expanded population of regulatory T cells is larger than a reference population because of increased proliferation. In a still further embodiment, the expanded population of regulatory T cells is larger than a reference population because of a combination of one or more of increased survival, decreased/reduced cell death and increased proliferation.
  • references herein to an" expanded population such as an “expanded population of regulatory T cells” may also include a population of cells which are activated.
  • references herein to" expanding may include the activation of a population of cells, such as a population of regulatory T cells.
  • expanding also includes the expansion of an activated population of regulatory T cells, for example, a population which is already activated prior to administration of IL-2.
  • Such activation of the population of cells, such as a population of regulatory T cells may be independent of an expansion or may be concomitant with an expansion of said population.
  • the expanded population of regulatory T cells comprises activated regulatory T cells.
  • the expanded population of regulatory T cells is an activated population of regulatory T cells.
  • references herein to"expanding" or an "expanded population” do not include activating said population or an activated population of cells.
  • the expanded population of cells such as an expanded population of regulatory T cells, does not comprise an activated phenotype.
  • the expanded population of regulatory T cells does not comprise activated regulatory T cells.
  • the expanded population of regulatory T cells comprises the phenotype, such as the surface phenotype, of a population of regulatory T cells which have not been subjected to or administered with IL-2.
  • Regulatory T cells are a subpopulation of T cells that modulate the immune system, maintain tolerance and prevent autoimmune disease. They generally suppress or downregulate the activation and/or proliferation of effector T cells and have been shown to have utility in immunosuppression.
  • regulatory T cells are highly potent cells that combine multiple immunosuppressive and regenerative capabilities and there is great interest in using exogenous regulatory T cells as a cell therapy or exogenous factors which stimulate, activate or expand endogenous regulatory T cells.
  • the inventors have previously demonstrated that regulatory T cells exist in the healthy brain (see Fig. 1 of WO2021/044175, the contents of which are hereby incorporated in their entirety and Pasciuto et al. (2020)), despite the traditional view that the brain is a tissue which is isolated from the immune system (e.g. because of the blood-brain barrier), and thus may be a valid target for immunosuppressive treatment, such as anti-inflammatory treatment, in the brain.
  • the expanded population of regulatory T cells comprises an increased anti-inflammatory potential.
  • increased anti-inflammatory potential may be compared to a non-expanded population of regulatory T cells, such as a non-expanded population of regulatory T cells present in the tissue or organ, or to a population of regulatory T cells present at another location other than the tissue or organ of interest.
  • the expanded population of regulatory T cells comprises a phenotype similar to non-expanded regulatory T cells within the tissue or organ of interest or to regulatory T cells from a location other than the tissue or organ of interest.
  • phenotypes may include surface marker phenotype, transcriptomic phenotype/signature (e.g. gene expression signature), gene and/or protein expression profile and cytokine expression profile.
  • the expanded population of regulatory T cells comprises or retains the anti-inflammatory potential of a non-expanded population of regulatory T cells or the expanded population of regulatory T cells prior to expansion.
  • the expanded population of regulatory T cells comprises or retains the anti-inflammatory potential of a population of regulatory T cells from another location other than the tissue or organ of interest.
  • the pharmaceutical composition leads to a transcriptional program or signature in the cells of the tissue or organ of interest which is more similar to a younger, or less aged, cell of said tissue or organ.
  • a transcriptional program/signature similar to a younger cell from the same tissue or organ includes a transcriptomic signature which corresponds to that of a cell from an earlier point in the life cycle of the tissue or organ, or in the life cycle of the subject.
  • Such reprogramming may also be referred to as" rejuvenation" of the cells of the tissue or organ of interest.
  • the method of reprogramming comprises rejuvenating the transcriptional program of the cells of the tissue or organ.
  • Transcriptional programs and signatures associated with ageing include genes within mitochondrial pathways, proteostasis pathways, the autophagy pathway and cellular senescence pathways, such as mitochondrial dysfunction, loss of proteostasis (e.g. protein production, processing and degradation) or autophagy and increased cellular senescence.
  • the transcriptional program or signature comprises proteostasis-related pathways, autophagy-associated pathways, key signaling pathways (e.g.
  • the transcriptional program or signature comprises proteostasis-related pathways, autophagy-associated pathways and neurodegeneration-associated genes.
  • the tissue or organ is the brain.
  • the cells of the tissue or organ comprise glial cells, such as microglia, astrocytes or oligodendrocytes.
  • the method comprises reprogramming the transcriptional program or signature in glial cells of the subject, such as microglia, astrocytes or oligodendrocytes, to a younger transcriptional program or signature.
  • the targeting moiety specific for the tissue or organ is specific for glial cells, such as microglia, astrocytes or oligodendrocytes.
  • references herein to the phrase" in a tissue or organ refer to a discrete location in the subject such as in a particular tissue or organ. It will be appreciated that such terms do not relate to wherein an effect is produced systemically or outside of the tissue or organ of interest, or wherein a cell type or cell population not located in the tissue or organ of interest is affected (e.g. expanded or activated).
  • the population of regulatory T cells is affected (e.g. expanded) in a particular tissue or organ, i.e. locally.
  • the population of regulatory T cells is affected (e.g. expanded) in a particular tissue or organ only.
  • the population of regulatory T cells located outside or not in the tissue or organ of interest is not affected (e.g.
  • the systemic or peripheral population of regulatory T cells is not affected (e.g. expanded).
  • the population of regulatory T cells is affected (e.g. expanded) in the central nervous system only.
  • the population of regulatory T cells is not affected (e.g. expanded) outside of the central nervous system, such as is not affected in the peripheral nervous system or systemically.
  • Tissues or organs as defined herein comprise a discrete location of the body or of an organism.
  • the tissue or organ may comprise a compartment of the body such as the central nervous system (e.g. the brain).
  • the tissue or organ is separated from other tissues or organs by a barrier, such as the blood-brain barrier.
  • the tissue or organ is the central nervous system.
  • the tissue or organ is the brain.
  • IL-2 is a key population control factor for regulatory T cells. Regulatory T cells have a naturally high turnover frequency compared to other T cells, with rapid proliferation and high apoptosis rates. IL-2 is able to increase the frequency of regulatory T cells through the induction of the anti-apoptotic protein Mcl1 , which in turn reduces the Bim-dependent apoptotic rate (Pierson et al. (2013)). Increased IL-2 levels can therefore expand the size of the regulatory T cell population (Liston and Gray (2014)). IL-2 delivery has been shown to be a potent antiinflammatory agent via the expansion of this regulatory T cell population in multiple pre-clinical studies, and optimisation of IL-2 delivery is being clinically investigated.
  • IL-2 as an anti-inflammatory mediator
  • the systemic delivery of IL-2 should, in theory, drive an increase in regulatory T cell numbers in the brain as this population is seeded by regulatory T cells in the circulation (see Pasciuto et al. (2020) and Fig. 2 of WO2021/044175).
  • a barrier such as the blood-brain barrier
  • the disclosures herein provide for the expansion of a population of regulatory T cells within a tissue or organ which, due to the presence of a barrier such as the blood-brain barrier, is difficult to achieve with systemic delivery of IL-2.
  • any dose of IL-2 sufficient to affect a population of cells present in the tissue or organ would have to be at a level high enough to give wide-spread peripheral or systemic effects.
  • the resulting wide-spread peripheral or systemic immunosuppression would be untenable to patients due to an increased risk of infection.
  • the methods described herein comprise administration of IL-2.
  • Administration will be appreciated to refer to the providing or the making available of IL-2 at a discrete location or site of the organism, such as a particular tissue or organ (i.e. the central nervous system and/or brain). Such administration will therefore be likened with the definitions of i"n a tissue or organ" as previously described herein.
  • administration of IL-2 comprises administration to or in a particular tissue or organ.
  • administration of IL-2 comprises expression of IL-2 in a particular tissue or organ (i.e. the nervous system and/or brain).
  • administration comprises expression of a gene encoding for IL-2 in a particular tissue or organ (i.e. the nervous system and/or brain).
  • the method of treating, preventing and/or reducing ageing comprises tissue- or organ-specific expression of IL-2 in said tissue or organ of the subject.
  • expression of IL-2 is not detectable outside the tissue or organ of interest, such as in the periphery.
  • expression of IL-2 is expression which is restricted to the particular tissue or organ of interest.
  • expression of IL-2 is tissue- or organ-specific expression.
  • administration or expression of IL-2 may be in more than one related tissue or organ of interest (e.g. related tissues or organs of the central nervous system).
  • administration or expression of IL-2 is in one, two, or more tissues or organs of the central nervous system, such as in the brain and the spinal cord.
  • references herein to"administration" and “expression” also refer to wherein IL-2 is provided to a population of cells in a tissue or organ.
  • Such provision of IL-2 may, in one embodiment, comprise administration of IL-2 in protein or peptide form to or in the tissue or organ of interest, i.e. locally.
  • the provision of IL-2 comprises the expression of IL-2 in the cells of the tissue or organ of interest.
  • expression of IL-2 comprises the cells of the tissue or organ of interest, such as those cells which make up said tissue or organ (e.g. neurons), expressing IL-2.
  • expression of IL-2 comprises neurons, oligodendrocytes and/or astrocytes.
  • expression of IL-2 comprises astrocytes.
  • the expression of IL-2 by/in astrocytes will be appreciated to provide several advantages: 1) astrocytes are efficient secretory cells which are widely distributed across the brain; 2) astrocytes are well represented in the spinal cord, providing the possibility of administration or expression of IL-2 in the spinal cord; 3) astrocytes demonstrate temporal and spatial numerical increases during neuroinflammatory events such as traumatic brain injury; and 4) expression of the astrocytespecific promoter GFAP is upregulated in response to injury and disease (see Pasciuto et al. (2020) and Fig. 5B of WO2021/044175).
  • expression of IL-2 comprises expression in cells other than the regulatory T cells which make up the expanded population of regulatory T cells.
  • expression of IL-2 is not in a population of regulatory T cells.
  • administration or expression of IL-2 comprises expression from the endogenous IL-2-encoding gene of cells of the tissue or organ of interest.
  • expression of IL-2 in the cells of the tissue or organ does not comprise transfection, transduction or introduction of exogenous sequence.
  • expression of IL-2 in the cells of the tissue or organ comprises tissue- or organ-specific stimulation using a compound which upregulates or"turns on" expression of the gene encoding for IL-2 only in those cells of the tissue or organ of interest. It will be appreciated that, according to this embodiment, stimulation of expression of the endogenous gene encoding IL-2 is specific and localised only to the tissue or organ of interest.
  • administration or expression of IL-2 comprises introducing into the cells of the tissue or organ (i.e. the central nervous system and/or brain) exogenous sequence encoding IL-2.
  • administration or expression of IL-2 comprises expression from an exogenous sequence.
  • administration or expression of IL-2 comprises expression from a transgene.
  • the transgene comprises a gene or an element encoding for IL-2.
  • the exogenous sequence is an IL-2 encoding sequence.
  • the transgene comprises an IL-2 encoding sequence or gene.
  • the exogenous sequence encoding IL-2 is in the form of a transgene comprising a tissue- or organ-specific promoter.
  • tissue- or organ-specific promoters are known in the art and include promoters which drive the expression of tissue- or organ-specific genes.
  • the transgene comprises a tissue- or organ-specific promoter which specifically drives expression in the tissue or organ of interest.
  • the transgene comprises a tissue- or organ-specific promoter which does not lead to expression in a tissue or organ other than the tissue or organ of interest.
  • the transgene comprises a promoter which drives expression specifically in neurons.
  • the transgene comprises a promoter which drives expression specifically in cells of the central nervous system.
  • the transgene comprises a promoter which drives expression in the central nervous system but not in the peripheral nervous system. In one embodiment, the transgene comprises a promoter which drives expression specifically in the brain. In a particular embodiment, the transgene comprises a promoter which drives expression specifically in astrocytes. In a further embodiment, the transgene comprises a GFAP promoter. In a yet further embodiment, the transgene comprises a minimal GFAP promoter. In alternative embodiments, the transgene comprises a PLP or CaMKIla promoter.
  • administration or expression of IL-2 comprises a transgene which comprises an element which promotes or induces the expression of IL-2 in the presence of an exogenous compound.
  • elements which promote or induce expression are known in the art and include, for example, tetracycline (Tet)-inducible systems.
  • Tet-inducible systems provide reversible control of transcription and utilise a tetracycline-controlled transactivator (tTA) which binds tetracycline operator (TetO) sequences contained in a tetracycline response element (TRE) placed upstream of the gene/coding region of interest (and its promoter, such as a tissue-specific promoter). They may either be TetOff or TetOn systems.
  • the TetOff system of inducible expression uses a tTA protein created by fusing the tetracycline repressor (TetR), found in Escherichia coli bacteria, with the activation domain of another protein, VP16, found in the Herpes Simplex Virus.
  • TetR tetracycline repressor
  • VP16 tetracycline repressor
  • the resulting tTA is able to bind TetO sequences within the TRE in the absence of tetracycline and promote expression of the downstream gene/coding region. In the presence of tetracycline, tTA binding to the TetO sequences is prevented, resulting in reduced gene expression.
  • TetOn system also known as the rtTA-dependent system
  • TetTA uses a reverse Tet repressor (rTetR) to create a reverse tetracycline-controlled transactivator (rtTA) protein which relies on the presence of tetracycline to promote expression. Therefore, rtTA only binds to TetO sequences within the TRE and promotes expression in the presence of tetracycline.
  • TetOn systems include, but are not limited to, TetOn Advanced, TetOn 3G and the T-REx system from Life Technologies.
  • Derivatives and analogues of tetracycline may be used with either the TetOff or TetOn systems and include, without limitation, doxycycline and minocycline (e.g. minomycin).
  • doxycycline and minocycline e.g. minomycin
  • Such derivatives/analogues will be appreciated to provide significant advantages compared to tetracycline such as increased stability in the case of doxycycline and/or the ability to cross the blood-brain barrier in the case of minocycline (Chtarto et al. 2003, doi: htps://doi.org/10.1016/i.neulet.2003.08.067).
  • the exogenous sequence encoding IL-2 such as the transgene comprising a tissue- or organ-specific promoter, further comprises a tetracycline response element (TRE).
  • TRE tetracycline response element
  • administration or expression of IL-2 is tetracycline-dependent or tetracycline-inducible.
  • administration or expression of IL-2 comprises introducing into the cells of the tissue or organ exogenous sequence encoding a reverse tetracycline-controlled transactivator (rtTA).
  • rtTA reverse tetracycline-controlled transactivator
  • the exogenous sequence encoding an rtTA comprises a tissue- or organ-specific promoter, i.e.
  • the exogenous sequence encoding an rtTA comprises a promoter specific for the nervous system, such as the central nervous system (e.g. the brain).
  • expression of the rtTA-encoding sequence is under the control of a promoter specific for the nervous system, such as the central nervous system (e.g. the brain).
  • the exogenous sequence encoding an rtTA comprises a promoter which drives expression specifically in astrocytes, such as a GFAP promoter or a minimal GFAP promoter.
  • Such an rtTA-encoding exogenous sequence may be a separate sequence to the exogenous sequence encoding IL-2, e.g. it may be separate from the IL-2 transgene comprising a tissue- or organ-specific promoter.
  • such an rtTA-encoding exogenous sequence may be comprised together with the IL-2-encoding sequence, e.g. it may be comprised in the same transgene.
  • administration or expression of IL-2 comprises a TetOn system. It will therefore be appreciated that in one embodiment, administration or expression of IL-2 comprises the administration of tetracycline or a derivative/analogue of tetracycline, such as doxycycline or minocycline. In a particular embodiment, administration or expression of IL-2 comprises administration of minocycline, such as administration of minomycin.
  • tetracycline-dependent or tetracycline-inducible administration or expression of IL-2 provides another level of control and allows the administration or expression of IL-2 to be ‘switched' on or off. Such switching will be appreciated to be advantageous in the methods described herein by allowing the expansion of a population of regulatory T cells in a tissue or organ to be temporally controlled.
  • expression of IL-2 may be switched ‘on' by administering tetracycline or a derivative/analogue thereof when inflammation of the central nervous system, such as neuroinflammation and/or inflammation of the brain, is detected/diagnosed.
  • IL-2 may then be switched ‘off' by removal of tetracycline or a derivative/analogue thereof when inflammation, such as neuroinflammation, is no longer detected or has reduced.
  • Said use of tetracycline-dependent or tetracycline-inducible administration or expression of IL-2 further provides dose-dependent IL-2 administration of expression.
  • the level and/or amount of IL-2 administration or expression may be altered and/or titrated in the tissue or organ to depend on the level and/or amount of inflammation, such as neuroinflammation, in the tissue or organ.
  • expression of IL-2 may be switched ‘on' by administering a particular dose of tetracycline or a derivative/analogue thereof when inflammation of the central nervous system, such as neuroinflammation and/or inflammation of the brain, is detected/diagnosed and said dose may be increased if the inflammation persists. Similarly, said dose may be decreased if the inflammation decreases following initial administration of tetracycline or a derivative/analogue thereof.
  • the transgene as defined herein is introduced into the cells of the tissue or organ of interest by transduction, such as transduction using a virus or viral vector.
  • the transduction uses an adeno-associated virus.
  • administration of IL-2 comprises transduction, such as viral transduction.
  • administration of IL-2 comprises adeno-associated virus transduction.
  • transduction of the transgene as defined herein utilises a viral vector which specifically targets or infects the cells of the tissue or organ of interest.
  • transduction of the transgene as defined herein specifically targets or infects the cells of the tissue or organ of interest.
  • transduction using a viral vector of the transgene as defined herein does not target or infect a population of regulatory T cells.
  • transduction of the transgene as defined herein comprises a viral vector which is capable of accessing the tissue or organ of interest and is capable of crossing a barrier which separates the tissue or organ of interest from other tissues, organs or the rest of the organism.
  • transduction comprises a viral vector capable of specifically targeting or infecting the nervous system.
  • transduction comprises a viral vector capable of targeting or infecting the central nervous system.
  • transduction comprises a viral vector capable of targeting or infecting the brain.
  • transduction comprises a viral vector capable of crossing the blood-brain barrier.
  • transduction comprises a blood-brain barriercrossing adeno-associated virus.
  • transduction comprises a neurotropic virus or viral vector.
  • the viral vector is a neurotropic virus or viral vector. Examples of neurotropic viruses and viral vectors capable of crossing the blood-brain barrier include, but are not limited to, AAVrh.8, AAVrhIO and AAV9 as well as its variants and derivatives (e.g. AAVhu68 and PHP.B).
  • the transgene as defined herein is comprised in a viral vector, such as a neurotropic virus or viral vector and/or an adeno-associated virus vector.
  • transduction comprises the adeno-associated virus variant AAV9 and its derivatives, such as PHP.B.
  • transduction comprises a PHP.B viral vector.
  • the transgene as defined herein is comprised in a PHP.B viral vector.
  • the transduction and/or the viral vector comprises PHP.B-GFAP-IL2, which is the PHP.B derivative of AAV9 comprising a transgene which contains an IL-2 encoding sequence and the astrocyte-specific promoter, GFAP.
  • Viral vectors may be used to integrate the target sequence, such as a transgene, into the host cell genome, such as the genome of a cell of the tissue or organ of interest.
  • transduction comprises integration of the transgene as defined herein into the genome of a cell of the tissue or organ of interest such that long-term expression of the transgene in the tissue or organ is achieved.
  • Viral vectors such as neurotropic viruses or viral vectors and adeno-associated viral vectors, may also be used to enable stable or long-term expression without integration of the target sequence into the host cell genome.
  • the transgene and/or target sequence are stably maintained outside the host cell genome.
  • references herein to a" virus” and/or " viral vector” include a virus which is non-lytic or lysogenic. Such viruses will be appreciated to achieve infection of a cell, such as a cell of the tissue or organ of interest, or introduction of a transgene into a cell without death or destruction of said cell.
  • combination of a virus or viral vector which specifically targets or infects cells of the tissue- or organ of interest e.g. a neurotropic virus or viral vector
  • a promoter which drives expression specifically in cells of the tissue or organ of interest provides exceptional specificity. Such specificity provides a so-called ‘dual lock', restricting both the cells into which the transgene is targeted or infected and in which cells the transgene is expressed.
  • the combination of a tissue- or organ-specific viral vector and tissue- or organ-specific promoter as defined herein provides that only those cells of the tissue or organ of interest comprise the transgene as defined herein and only those cells of the tissue or organ of interest are capable of expressing said transgene.
  • tissue- or organ-specific viral vector and tissue- or organ-specific promoter as defined herein provides that only those cells of the tissue or organ of interest comprise an IL-2-encoding gene and only those cells of the tissue or organ of interest are capable of expressing said gene.
  • tissue- or organ-specific viral vector and tissue- or organ-specific promoter as defined herein together with an inducible element provides that only those cells of the tissue or organ of interest comprise the transgene as defined herein and only those cells of the tissue or organ of interest are capable of expressing said transgene when an activator of the inducible element is administered (e.g. tetracycline, doxycycline or minocycline/minomycin).
  • an activator of the inducible element e.g. tetracycline, doxycycline or minocycline/minomycin.
  • tissue- or organ-specific viral vector and tissue- or organspecific promoter as defined herein together with an inducible element provides that only those cells of the tissue or organ of interest comprise an IL-2-encoding gene and only those cells of the tissue or organ of interest are capable of expressing said gene when an activator of the inducible element is administered (e.g. tetracycline, doxycycline or minocycline/minomycin).
  • an activator of the inducible element e.g. tetracycline, doxycycline or minocycline/minomycin.
  • said combination provides that only those cells of the tissue or organ of interest comprise an inducible IL-2-encoding gene and only those cells of the tissue or organ of interest are capable of expressing a reverse tetracycline-controlled transactivator (rtTA) which leads to the expression of IL-2 when an activator of the inducible element is administered (e.g. tetracycline, doxycycline or minocycline/minomycin).
  • rtTA reverse tetracycline-controlled transactivator
  • Administration of IL-2 as defined herein may further comprise administration of IL-2 directly to the tissue or organ of interest.
  • direct administration include injection directly into the tissue or organ of interest, such as by intracranial injection, or utilise a suitable delivery device.
  • delivery devices are known in the art and, according to the present disclosures, allow for the controlled and/or sustained administration of IL-2 for the duration of treatment (e.g. chronically or for duration of treatment of an acute inflammatory disease or disorder).
  • the duration of IL-2 administration as defined herein can be altered to depend on the treatment and the characteristics of the ageing condition or disease to be treated, prevented or reduced by the pharmaceutical compositions and methods described herein (e.g. the neurological ageing-related disease or disorder).
  • administration of IL-2 may be chronic.
  • administration of IL-2 may be for the duration of treatment for the disease or disorder, such as in the treatment of an acute neurological ageing-related disease or disorder.
  • the duration of administration or expression of IL-2 depends on the disease or disorder to be treated or on the duration of the treatment.
  • administration or expression of IL-2 is acute.
  • administration or expression of IL-2 is chronic.
  • IL-2 and a targeting moiety specific for a tissue or organ may be combined or co-administered. Therefore, the administration of IL-2 may comprise expression of IL-2 in the tissue or organ of interest as defined herein (e.g. tissue- or organ-specific expression) and can be combined with a targeting moiety specific for the tissue or organ of the subject. Furthermore, administration of IL-2 may comprise administration of IL-2 in protein or peptide form and can be combined with a targeting moiety specific for the tissue or organ of the subject.
  • targeting moiety refers to any moiety that provides for the tissue- or organ-specific administration or expression of IL-2 as defined herein. Furthermore, said targeting moiety will be appreciated to provide for the localised administration or expression of IL-2 as defined herein.
  • the methods defined herein comprise administration of a targeting moiety specific for the tissue or organ of the subject, wherein said tissue or organ is the central nervous system.
  • the targeting moiety specific for the tissue or organ of the subject localises IL-2 in or to the tissue or organ of interest (i.e. the central nervous system).
  • the targeting moiety specific for the tissue or organ of the subject localises IL-2 only in or to the tissue or organ of interest.
  • the targeting moiety specific for the tissue or organ of the subject prevents localisation of IL-2 to other tissues or organs other than the tissue or organ of interest, or localises IL-2 away from tissues or organs other than the tissue or organ of interest.
  • the targeting moiety provides for expression of IL-2 in the tissue or organ of interest.
  • the targeting moiety specific for the tissue or organ of the subject provides for expression of IL-2 only in the tissue or organ of interest.
  • Such references herein to "in the tissue or organ of interest” further include wherein said effect is in the cells which make up said tissue or organ (e.g. neurons and/or astrocytes).
  • the targeting moiety specific for the tissue or organ of the subject is a virus or viral vector as defined herein.
  • said virus or viral vector specifically targets or infects the tissue or organ of interest or specifically targets or infects cells of the tissue or organ of interest (i.e. the central nervous system).
  • said targeting moiety specific for the tissue or organ of interest which is a virus or viral vector does not target or infect cells in other tissues or organs other than the tissue or organ of interest, or target or infect cells which make up a tissue or organ other than the tissue or organ of interest.
  • said targeting moiety specific for the tissue or organ as defined herein does not target or infect a population of regulatory T cells.
  • the targeting moiety specific for the tissue or organ of a subject as defined herein comprises a virus or viral vector which is capable of accessing the tissue or organ of interest and is capable of crossing a barrier which separates the tissue or organ of interest from other tissues, organs or the rest of the subject.
  • the targeting moiety specific for a tissue or organ comprises a virus or viral vector capable of specifically targeting or infecting the nervous system, such as a neurotropic virus or viral vector.
  • the targeting moiety specific for a tissue or organ comprises a virus or viral vector capable of targeting or infecting the central nervous system.
  • the targeting moiety specific for a tissue or organ comprises a virus or viral vector capable of targeting or infecting the brain.
  • the targeting moiety specific for a tissue or organ comprises a virus or viral vector capable of crossing the blood-brain barrier.
  • the targeting moiety specific for a tissue or organ comprises a blood-brain barrier-crossing adeno- associated virus.
  • the targeting moiety specific for a tissue or organ comprises a neurotropic virus or viral vector.
  • the targeting moiety is selected from a neurotropic virus or viral vector, such as AAVrh.8, AAVrhIO or AAV9 and variants and derivatives (e.g. AAVhu68 and PHP.B).
  • the targeting moiety specific for a tissue or organ comprises the adeno-associated virus variant PHP.B.
  • the transgene as defined herein is comprised in a targeting moiety specific for a tissue or organ, such as an adeno-associated virus vector, which is comprised within an adeno-associated virus as defined herein.
  • the transgene as defined herein is comprised in a neurotropic virus or viral vector, such as a PHP.B viral vector.
  • the transgene which contains an IL-2 encoding sequence and the astrocyte-specific promoter, GFAP or minimal GFAP is comprised in the AAV9 derivative PHP.B virus/viral vector and the virus/viral vector is PHP.B-GFAP-IL2.
  • the method for expanding a population of regulatory T cells in a tissue or organ in vivo comprises administration of IL-2 as described herein.
  • the method for expanding a population of regulatory T cells in a tissue or organ in vivo comprises administration of a targeting moiety specific for the tissue or organ of a subject in vivo.
  • the method for expanding a population of regulatory T cells in a tissue or organ in vivo comprises administration of a pharmaceutical composition as described herein.
  • the administration of IL-2 which may comprise expression of IL-2, is combined with a targeting moiety specific for a tissue or organ in vivo.
  • the method for expanding a population of regulatory T cells in a tissue or organ in vivo comprises a virus or viral vector which comprises an IL-2-encoding gene.
  • said virus or viral vector is capable of targeting or infecting a tissue or organ of interest (i.e. the central nervous system).
  • said virus or viral vector capable of targeting or infecting a tissue or organ of interest specifically targets or infects cells of a tissue or organ of interest.
  • the method for expanding a population of regulatory T cells in a tissue or organ in vivo comprises a virus or viral vector which comprises a tissue- or organ-specific promoter.
  • the method for expanding a population of regulatory T cells in a tissue or organ in vivo comprises administration of a targeting moiety specific for the tissue or organ of interest, wherein said targeting moiety is a virus or viral vector which crosses the blood-brain barrier as defined herein.
  • the method for expanding a population of regulatory T cells in a tissue or organ in vivo comprises administration of a targeting moiety specific for the tissue or organ of interest, wherein said targeting moiety is specific for the central nervous system, such as the brain.
  • the targeting moiety specific for a tissue or organ of interest is specific for astrocytes.
  • the method for expanding a population of regulatory T cells in a tissue or organ in vivo comprises administration of a neurotropic virus or viral vector containing the transgene as defined herein, such as administration of PHP.B-GFAP-IL2.
  • a population of regulatory T cells expanded according to or obtained by the methods or the pharmaceutical composition e.g. by administration of said pharmaceutical composition
  • an expanded population of regulatory T cells which have been expanded in a tissue or organ of a subject by administration of IL-2 and a targeting moiety specific for said tissue or organ for use in the treatment, prevention and/or reduction of ageing, wherein said tissue or organ is the central nervous system (e.g. the brain).
  • the methods of treating, preventing and/or reducing ageing as described herein comprise the expanded population of regulatory T cells in the central nervous system (e.g. in the brain) as described herein.
  • a pharmaceutical composition comprising IL-2 and a targeting moiety specific for a tissue or organ of a subject, wherein said tissue or organ is the central nervous system.
  • the pharmaceutical composition comprises IL-2 which promotes the expansion of a population of regulatory T cells.
  • the pharmaceutical composition comprises a targeting moiety specific for a tissue or organ of a subject (i.e. the central nervous system, such as the brain).
  • the targeting moiety specific for a tissue or organ of a subject is a virus or viral vector which specifically targets or infects cells of the tissue or organ and drives tissue- or organ-specific expression of IL-2 as described herein.
  • a pharmaceutical composition comprising a tissue- or organ-specific viral vector which expands a population of regulatory T cells in said tissue or organ of the subject (i.e. the central nervous system).
  • the pharmaceutical composition expands a population of regulatory T cells specifically or locally in a tissue or organ of interest in a subject.
  • the pharmaceutical composition as defined herein comprises a targeting moiety capable of crossing a barrier which separates a tissue or organ of interest from other tissues or organs or from the rest of the organism.
  • the pharmaceutical composition as defined herein comprises a blood-brain barrier crossing virus or viral vector, such as an adeno-associated virus and/or a neurotropic virus or viral vector.
  • the pharmaceutical composition as defined herein comprises the adeno-associated virus variant AAV9 or its derivatives, such as PHP.B.
  • the viral vector comprised in the pharmaceutical composition as defined herein comprises a gene, such as a transgene, which encodes for IL-2.
  • the transgene comprised in the viral vector of the pharmaceutical composition further comprises a tissue- or organ-specific promoter as defined herein.
  • the pharmaceutical composition as defined herein comprises a tissue- or organ-specific virus or viral vector capable of targeting or infecting cells of the tissue or organ of interest (i.e. the central nervous system), comprising an IL-2-encoding gene, expression of which is driven by a tissue- or organ-specific promoter.
  • the pharmaceutical composition as defined herein comprises a viral vector, such as an adeno-associated virus (e.g. AAV9 or its derivatives, such as PHP.B), which specifically targets or infects neurons or the nervous system, such as the brain, (i.e. a neurotropic virus or viral vector) which comprises an IL-2-encoding gene, expression of which is driven by a tissue- or organ-specific promoter.
  • the pharmaceutical composition as defined herein comprises the adeno-associated virus AAV9, which comprises an IL-2- encoding gene, expression of which is driven locally in a neuron/astrocyte or in the nervous system by a GFAP promoter or a minimal GFAP promoter.
  • the adeno-associated virus is a derivative of AAV9, such as PHP.B.
  • the pharmaceutical composition comprises PHP.B-GFAP-IL2.
  • the pharmaceutical composition in addition to a tissue- or organ-specific virus or viral vector as defined herein, further comprises one or more pharmaceutically acceptable excipients.
  • the present pharmaceutical compositions will be utilised with pharmacologically appropriate excipients or carriers.
  • these excipients or carriers include aqueous or alcoholic/aqueous solutions, emulsions or suspensions, including saline and/or buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride and lactated Ringer's.
  • Suitable physiologically-acceptable adjuvants if necessary to keep a composition comprising the targeting moiety specific for a tissue or organ as defined herein in a discrete location (e.g.
  • Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishers, such as those based on Ringer's dextrose. Preservatives and other additives, such as antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack (1982) Remington's Pharmaceutical Sciences, 16 th Edition).
  • the method of expanding a population of regulatory T cells, pharmaceutical compositions and methods of treatment described herein will find particular utility in the treatment and/or amelioration of diseases or disorders mediated by inflammation and/or in the reduction of inflammation. It will be further appreciated that a population of regulatory T cells expanded according to the methods and disclosures presented herein will also find utility in the treatment and/or amelioration of diseases or disorders mediated by inflammation and/or in the reduction of inflammation.
  • the methods and pharmaceutical compositions described herein surprisingly also find utility in treating, preventing and/or reducing ageing or treating a disease or disorder related to ageing, wherein the ageing is neurological ageing and the neurological ageing or disease/disorder (e.g.
  • a neurodegenerative disease/disorder is considered to be non-inflammatory.
  • a method for expanding a population of regulatory T cells in a tissue or organ of a subject for use in the treatment, prevention and/or reduction of ageing, wherein said tissue or organ is the central nervous system In another aspect, there is provided a method for expanding a population of regulatory T cells in a tissue or organ of a subject for use in the treatment of a disease or disorder related to ageing, wherein said tissue or organ is the central nervous system.
  • the ageing is neurological ageing.
  • the pharmaceutical composition as defined herein for use in the treatment of a disease or disorder related to ageing wherein said tissue or organ is the central nervous system.
  • the pharmaceutical composition as defined herein for use in the treatment of a disease or disorder related to neurological ageing such as a neurodegenerative disease or disorder, wherein said tissue or organ is the central nervous system.
  • a method of treating, preventing and/or reducing ageing comprising administering to a subject in need thereof the pharmaceutical composition as defined herein.
  • the method of treating, preventing and/or reducing ageing comprises the method of reprogramming cells of the tissue or organ as defined herein, such as the method of rejuvenating the cells of the tissue or organ (i.e. the central nervous system).
  • the method is for treating a disease or disorder related to neurological ageing.
  • the ageing is neurological ageing.
  • a method of treating a disease or disorder related to neurological ageing such as a neurodegenerative disease or disorder, wherein said tissue or organ is the central nervous system.
  • a population of expanded regulatory T cells in a tissue or organ of a subject produced according to the methods defined herein or by administration of the pharmaceutical composition defined herein for use in the treatment, prevention and/or reduction of ageing, wherein said tissue or organ is the central nervous system.
  • the ageing is neurological ageing.
  • the population of expanded regulatory T cells is for use in the treatment of a disease or disorder related to neurological ageing, such as a neurodegenerative disease or disorder.
  • diseases or disorders may include neuroinflammatory conditions or neurodegenerative diseases or disorders which are considered to be non-inflammatory.
  • the expanded population of regulatory T cells in a tissue or organ of a subject produced according to the methods defined herein has been expanded by administration of IL-2 and a targeting moiety specific for said tissue or organ.
  • the population of expanded regulatory T cells in a tissue or organ of a subject produced according to the methods defined herein has been expanded by tissue- or organ-specific expression of IL-2 as defined herein.
  • the population of expanded regulatory T cells in a tissue or organ of a subject has been expanded by tissue- or organ-specific expression of IL-2 promoted or induced by an inducible element, such as a tetracycline-inducible element.
  • the population of expanded regulatory T cells has been expanded by administration of the pharmaceutical composition as defined herein.
  • the methods defined herein comprise administering a virus or viral vector comprising a gene encoding IL-2 as defined herein to a subject in need thereof.
  • the methods defined herein comprise administering to a subject in need thereof a virus or viral vector which specifically targets or infects a tissue or organ affected by neurological ageing, such as a disease or disorder related to neurological ageing (i.e. the central nervous system).
  • the methods defined herein further comprise administering to a subject in need thereof a virus or viral vector comprising a gene encoding IL-2, expression of which is driven by a tissue- or organ-specific promoter.
  • the methods defined herein comprises administering to a subject in need thereof a virus or viral vector comprising a gene encoding IL-2, expression of which is driven by a tissue- or organ-specific promoter and an inducible element, such as a tetracycline-inducible element.
  • the methods comprise administering to a subject a virus or viral vector comprising a gene encoding IL-2, expression of which is driven by an inducible element, such as a tetracyclineinducible element, under the control of a tissue- or organ-specific promoter.
  • the methods defined herein comprise administering to a subject in need thereof a neurotropic virus comprising a gene encoding IL-2, expression of which is driven by a tissue- or organ-specific promoter, such as administering PHP.B-GFAP-IL2.
  • said subject in need thereof is an aged individual.
  • the ageing is neurological ageing.
  • the subject in need thereof is suffering from a disease or disorder related to neurological ageing.
  • the subject is suffering from a neurodegenerative disease or disorder. Therefore in another embodiment, the neurological ageing or the disease or disorder related to neurological ageing is a neurodegenerative disease or disorder.
  • the neurodegenerative disease or disorder is considered to be noninflammatory.
  • the neurodegenerative disease or disorder comprises cognitive decline.
  • a method of treating, preventing and/or reducing cognitive decline comprising administering to a subject in need thereof the pharmaceutical composition defined herein.
  • the method of treating, preventing and/or reducing cognitive decline comprises the methods of expanding a population of regulatory T cells in the central nervous system of a subject as defined herein.
  • the method of treating, preventing and/or reducing cognitive decline comprises administering IL-2, such as tissue- or organ-specific expression of IL-2 in the central nervous system as described herein.
  • Example 1 Age Drives a Change in the Cellular Composition of Brain-Resident glia
  • a key limitation to proposed regulatory T cell (Treg)- or IL-2-based therapies is the peripheral impact of most proposed strategies, with the potential for undesirable peripheral immunosuppression.
  • the use of brain-targeted delivery opens the door to potential utilisation for neuropathologies, while preserving the integrity of the peripheral immune system.
  • One way to achieve brain-specific expression of IL-2 is through the gene-delivery vector PHP.GFAP-IL2. This approach results in the restricted production of IL-2 in the brain, and an accompanying expansion of brain-resident Tregs (WO2021/044175).
  • PHP.GFAP-IL2 treatment is protective against neuroinflammation in the context of traumatic brain injury, stroke and experimental autoimmune encephalitis (WO2021/044175).
  • Reclustering of microglia resulted in tightly clustered cells ( Figure 1C), while reclustering of oligodendrocytes led to a clear separation of oligodendrocyte precursors (OPCs) and oligodendrocytes ( Figure 1D). Reclustering of astrocytes, by contrast, produced a clear transcriptional separation of Bergmann glia, Cerebellar astrocytes, Olfactory astrocytes, striatal astrocytes, telencephalon astrocytes and non-telencephalon astrocytes ( Figure 1E), based on key markers and dataset integration.
  • Example 2 Brain-Specific IL-2 Delivery Mitigates Age-Induced Molecular Changes in Resident Glia
  • the effect at the gene set level was then determined, where accumulated minor changes in a molecular pathway can be assessed.
  • Gene set enrichment was performed on each glial cell type, comparing the effects of age and treatment.
  • key pathways modified by ageing in microglia were those of proteostasis-related pathways, autophagy-associated pathways, key signalling pathways (including Ras, PI3K and mTOR pathways), neurodegeneration-associated genes and the cellular senescence pathway.
  • Oligodendrocytes and astrocytes underwent a similar transcriptional change with age, albeit with fewer pathways significantly altered, with altered expression of proteostasis pathways, autophagy-associated pathways and neurodegeneration-associated genes.
  • Example 3 Brain-Specific IL-2 Gene Delivery Provides Partial Protection for Cognitive Decline in Learned Behaviour
  • Aged mice also demonstrated decline in the social novelty-seeking behaviour of the sociability test (Figure 7D) and the anxiety-testing of the light-dark test (Figure 7E), again with no effect of IL-2-treatment. Finally, the mice were tested in the Morris water maze. Aged mice demonstrated poor performance in the Morris water maze in two aspects. First, aged mice had a large reduction in swim velocity during the test (Figure 7F), a phenotype likely to be derived from physical rather than cognitive decline. Second, aged mice demonstrated a reduced preference for the target quadrant after 10 days of training (Figure 7G), a phenotype reflecting reduced cognitive capacity for spatial memory formation.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Zoology (AREA)
  • Public Health (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Wood Science & Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Neurosurgery (AREA)
  • Microbiology (AREA)
  • Neurology (AREA)
  • Biophysics (AREA)
  • Hospice & Palliative Care (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Psychiatry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Mycology (AREA)
  • Toxicology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

L'invention concerne une composition pharmaceutique destinée à être utilisée dans une méthode de traitement, de prévention et/ou de réduction du vieillissement, cette composition comprenant de l'IL-2 et une fraction de ciblage spécifique d'un tissu ou d'un organe d'un sujet, tel que le cerveau, et le vieillissement étant un vieillissement neurologique. La présente invention concerne également des méthodes de traitement, de prévention et/ou de réduction du vieillissement (par exemple, du vieillissement neurologique) ou du déclin cognitif, ces méthodes comprenant l'administration d'une composition pharmaceutique telle que définie dans la description.
PCT/GB2022/053228 2021-12-14 2022-12-14 Nouvelles méthode et composition comprenant de l'il-2 et une fraction de ciblage spécifique d'un tissu ou d'un organe WO2023111560A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB2118073.2 2021-12-14
GBGB2118073.2A GB202118073D0 (en) 2021-12-14 2021-12-14 Novel method and composition comprising IL-2 and a tissue- or organ-specific targeting moiety

Publications (1)

Publication Number Publication Date
WO2023111560A1 true WO2023111560A1 (fr) 2023-06-22

Family

ID=80079968

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2022/053228 WO2023111560A1 (fr) 2021-12-14 2022-12-14 Nouvelles méthode et composition comprenant de l'il-2 et une fraction de ciblage spécifique d'un tissu ou d'un organe

Country Status (2)

Country Link
GB (1) GB202118073D0 (fr)
WO (1) WO2023111560A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001083716A2 (fr) * 2000-05-03 2001-11-08 Neurotech S.A. Lignees immortalisees de cellules encephaliques endotheliales et applications therapeutiques de ces dernieres
WO2017060510A1 (fr) * 2015-10-09 2017-04-13 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes et compositions pharmaceutiques pour le traitement de la maladie d'alzheimer
WO2021044175A1 (fr) 2019-09-06 2021-03-11 Babraham Institute Nouvelle méthode

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001083716A2 (fr) * 2000-05-03 2001-11-08 Neurotech S.A. Lignees immortalisees de cellules encephaliques endotheliales et applications therapeutiques de ces dernieres
WO2017060510A1 (fr) * 2015-10-09 2017-04-13 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes et compositions pharmaceutiques pour le traitement de la maladie d'alzheimer
WO2021044175A1 (fr) 2019-09-06 2021-03-11 Babraham Institute Nouvelle méthode

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MACK: "Remington's Pharmaceutical Sciences", 1982
SANDRO ALVES ET AL, BRAIN, 20 December 2016 (2016-12-20), GB, pages aww330, XP055685364, ISSN: 0006-8950, DOI: 10.1093/brain/aww330 *

Also Published As

Publication number Publication date
GB202118073D0 (en) 2022-01-26

Similar Documents

Publication Publication Date Title
Chen et al. Interleukin-17A: the key cytokine in neurodegenerative diseases
Kam et al. Microglia and astrocyte dysfunction in parkinson's disease
Harms et al. Peripheral monocyte entry is required for alpha-Synuclein induced inflammation and Neurodegeneration in a model of Parkinson disease
Yi et al. Astrocytes in multiple sclerosis and experimental autoimmune encephalomyelitis: Star-shaped cells illuminating the darkness of CNS autoimmunity
Gaudet et al. miR-155 deletion in mice overcomes neuron-intrinsic and neuron-extrinsic barriers to spinal cord repair
Taylor et al. Neuroinflammation and oxidative stress: co-conspirators in the pathology of Parkinson’s disease
Goldmann et al. Role of microglia in CNS autoimmunity
De et al. CSF1 overexpression has pleiotropic effects on microglia in vivo
Miron et al. Cells of the oligodendroglial lineage, myelination, and remyelination
Grote et al. Regulators of adult neurogenesis in the healthy and diseased brain.
Amor et al. White matter microglia heterogeneity in the CNS
Piri et al. Heat shock proteins in the retina: focus on HSP70 and alpha crystallins in ganglion cell survival
Hilton et al. An active vesicle priming machinery suppresses axon regeneration upon adult CNS injury
Lockrow et al. Age-related neurodegeneration and memory loss in down syndrome
Au et al. Recent advances in the study of bipolar/rod-shaped microglia and their roles in neurodegeneration
Au et al. Neuroinflammation, microglia and implications for retinal ganglion cell survival and axon regeneration in traumatic optic neuropathy
Della Sala et al. Synaptic plasticity and signaling in Rett syndrome
Mehterov et al. Interactions among brain-derived neurotrophic factor and neuroimmune pathways are key components of the major psychiatric disorders
Ahmed et al. Innate immune system activation and neuroinflammation in Down syndrome and neurodegeneration: therapeutic targets or partners?
Zhang et al. SIRT1 inactivation switches reactive astrocytes to an antiinflammatory phenotype in CNS autoimmunity
US20220220180A1 (en) Novel method
Lemaitre et al. Molecular and cognitive signatures of ageing partially restored through synthetic delivery of IL2 to the brain
Andoh et al. Microglia in animal models of autism spectrum disorders
Prakash Developmental pathways linked to the vulnerability of adult midbrain dopaminergic neurons to neurodegeneration
WO2023111560A1 (fr) Nouvelles méthode et composition comprenant de l'il-2 et une fraction de ciblage spécifique d'un tissu ou d'un organe

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22830917

Country of ref document: EP

Kind code of ref document: A1