MXPA00000840A - Activated t-cells and their uses - Google Patents

Activated t-cells and their uses

Info

Publication number
MXPA00000840A
MXPA00000840A MXPA/A/2000/000840A MXPA00000840A MXPA00000840A MX PA00000840 A MXPA00000840 A MX PA00000840A MX PA00000840 A MXPA00000840 A MX PA00000840A MX PA00000840 A MXPA00000840 A MX PA00000840A
Authority
MX
Mexico
Prior art keywords
cells
activated
disease
injury
cns
Prior art date
Application number
MXPA/A/2000/000840A
Other languages
Spanish (es)
Inventor
Michal Eisenbachschwartz
Irun R Cohen
David L Hirschberg
Original Assignee
Irun R Cohen
Michal Eisenbachschwartz
David L Hirschberg
Yeda Research And Development Co Ltd
Filing date
Publication date
Application filed by Irun R Cohen, Michal Eisenbachschwartz, David L Hirschberg, Yeda Research And Development Co Ltd filed Critical Irun R Cohen
Publication of MXPA00000840A publication Critical patent/MXPA00000840A/en

Links

Abstract

The present invention relates to compositions and methods for the treatment or diagnosis of injury of the central nervous system (CNS). In particular, the invention relates to compositions comprising activated T-cells which are used to deliver (a) a diagnostic substance or (b) a therapeutic substance to a site of damage of the CNS caused by injury or disease. The invention also relates to pharmaceutical compositions comprising antiself T-cells that recognize antigens present in a greater concentration in the CNS compared to the circulation and methods of use thereof to prevent or inhibit degeneration of nerves within the CNS. The invention also relates to pharmaceutical compositions comprising an antigen (or derivative thereof) present in a greater concentration in the CNS compared to the circulation (NS-specific antigen or derivative) and methods of use thereof to prevent or inhibit degeneration of nerves within the CNS. The substance-delivering activated T-cell composition of the present invention may be administered alone or in combination with NS-specific T-cells or NS-specific antigen or in combination with NS-specific T-cells and NS-specific antigen.

Description

T CELLS ACTIVATED AND THEIR USES 1. FIELD OF THE INVENTION The present invention relates to compositions and methods for the treatment or diagnosis of central nervous system (CNS) injury. In one embodiment, activated T cells are used to deliver (a) a diagnostic substance to detect a site of injury or disease, or (b) a therapeutic substance to alleviate the effect of a disease or injury such as, for example, promoting axonal regeneration or preventing or inhibiting degeneration caused by injury or disease. In a preferred embodiment, activated T cells that are used to deliver a substance do not recognize an antigen of the nervous system (SN). More preferably, the activated T cells supplying substances recognize a non-self antigen (e.g., ovalbumin). 2. BACKGROUND OF THE INVENTION CNS damage may be the result of a traumatic injury, such as penetrating trauma or blunt trauma, or a disease or disorder including, but not limited to, Alzheimer's disease, Parkinson's disease, multiple sclerosis, Huntington's disease, amyotrophic sclerosis lateral (ALS) and ischemia.
Consecutive to traumatic injuries of the peripheral nervous system (SNP), there is an invasion of monocytes derived from blood, as well as activation of microglia within the SNP (Stoll et al., 1989, Neurosci., 9: 2327-35; Gordon, 1991, Int. Rev. Cytol, 125: 203-44, Perry and Gordon, 1988, Trends Neurosci., 11: 273-277, Jordan and Thomas, 1988, Brain macrophages: Question or origin and Interrelationships, 13: 165 -178; Griffin et al., 1990, Ann Neurol., 27: 8, Giulian et al., 1989, J. Neurosci., 9: 4416-29, Giulian, 1987, J. Neurosci. Res., 18: 155- 171; de Groot et al., 1989, 179: 314-27; and Bauer et al., J. Neurosci. Res., 38: 365-75). In contrast, invasion of blood derived monocytes is delayed and more limited in scope in traumatic CNS lesions (Perry and Gordon, 1991, Int.Rev.Cytol, 125: 203-44; Anderson et al., 1991, Immunol. Lett., 30: 177-81; and Perry et al., 1987, J. Exp. Med., 165: 1218-1223). In addition, the duration of the events associated with the acute phase of the lesion, although less pronounced, is prolonged in the CNS, compared to the SNP. For example, several weeks after the injury, numerous macrophages and activated microglia cells are found in the CNS, while only a few are detected in SNP nerves at that time after the injury (Perry et al., 1987, J. Exp. Med., 165: 1218-1223; Lunn et al., 1990, Neuroscience 35: 157-165). Mammalian CNS neurons do not undergo spontaneous regeneration after injury. In this way, an injury to the CNS causes permanent impairment of motor and sensory functions. In contrast, SNP neurons have a much greater ability to regenerate. Studies in which allogeneic macrophages are incubated with a stimulant (eg, a nerve segment) and subsequently administered in the CNS of a mammal at or near the site of injury, have shown that regeneration of impaired sensory or motor function occurs. (PCT publication WO 97/09885 and Spiegler et al., 1996, FASEB J. 10: 1296). Another tragic consequence of CNS injury is that the primary lesion is often formed by a degenerative process that results in a secondary loss, over time, of adjacent neurons that were not damaged by the initial injury. It has been suggested that secondary degeneration results from diffusion of toxic chemical compounds produced by damaged neurons (Mclntosh, 1993, J. Neurotrauma 10: 215, Lynch and Dawson, 1994, Curr Opin. Neurol 7: 510, Smith and others, 1995, New Horiz 3: 562, Faden, 1996, Pharmacol, Toxicol., 78:12, Faden, 1996, JAMA, 276: 569). Popovitch et al. Have shown that CNS trauma such as spinal injury triggers a systemic response to self epitopes such as myelin basic proteins (MBP) (Popovitch et al., 1996, J. Neurosci. Res., 45: 349). It has been shown that activated T cells that recognize a self antigen, as well as activated T cells that recognize a non-self antigen, enter the parenchyma of the CNS. Only T cells capable of recognizing a CNS antigen appear to persist in nervous tissue (Hickey et al., 1991, J. Neurosci Res. 28, 254-60). Although activated T cells that recognize a self antigen (non-self T cells) apparently persist in nervous tissue, the use of activated T cells that recognize a non-self antigen (non-self T cells) for administration to an individual has advantages such as absence of risk of induction of autoimmune disease. In addition, the use of activated non-self T cells eliminates the need to activate autologous or syngenic T cells; therefore, non-self T cells can be activated and stored for use in any individual. T cells reactive to CNS white matter antigens, such as myelin basic protein (MBP), can induce experimental autoimmune paralytic disease (EAE) paralytic disease within several days of their inoculation in unaffected recipient rats (Ben Nun and others, 1981, Eur. J. Immunol., 11, 195-9). Studies have suggested a role for anti-MBP T cells in human multiple sclerosis disease (Ota et al., 1990, Nature 346, 183-7, Martin, 1997, J. Neural, Transm. Suppl. 49, 53-67;; Sun, 1993, Acta Neurol, Scand. Suppl 142: 1-56). Despite their pathogenic potential, anti-MBP T cell clones are present in the immune systems of healthy subjects (Burns et al., 1983, Cell Immunol 81, 435-40, Schiuesener and Wekerle 1985, J. Immunol. 135, 3128-33). However, little is known about the possible functions of non-self T cells.
The citation or identification of any reference should not be considered as an admission that said reference is available as a prior art to the present invention. 3. BRIEF DESCRIPTION OF THE INVENTION The present invention is directed to methods and compositions for the treatment or diagnosis of central nervous system (CNS) injury. The present invention provides methods for delivering a therapeutic or detectable substance to a site of CNS injury or disease, which comprises administering an effective amount of activated T cells containing or expressing a therapeutic or detectable substance to a mammal, wherein the It is effective to detect, diagnose or monitor a site of CNS injury or disease, or is effective in alleviating the effects of a CNS injury or disease. Activated T cells used to deliver a substance preferably do not recognize a specific antigen of the nervous system (SN specific antigen); more preferably, the activated T cells recognize a non-self antigen. "Activated T cells", as used herein, include (i) T cells that have been activated by (a) exposure to a cognate antigen or derivative thereof, or (b) exposure to an appropriate mitogen such as a lectin (for example, concanavalin A (Con A) or phytohemagglutinin (PHA)), and (i) the progeny of said activated T cells. As used herein, a cognate antigen is an antigen that is specifically recognized by the T cell antigen receptor of a T cell that has been previously exposed to the antigen. As used herein, a derivative of an antigen is an amino acid fragment or variant (e.g., an insertion, substitution and / or deletion derivative) of the corresponding total-length antigen, as long as the fragment or variant of the amino acid is capable of exhibiting one or more functional activities of the corresponding total length antigen. Such functional activities include, but are not limited to, antigenicity (ability to bind (or compete with the antigen to bind) to an antigen-specific antibody), immunogenicity (ability to generate antibodies which bind antigen), and ability to interact with T cells, resulting in an activation comparable to that obtained using the corresponding total length antigen. 4. BRIEF DESCRIPTION OF THE FIGURES Figure 1 shows low power epifluorescence micrographs of optic nerve after controlled compression injury in rats treated with T cell clones primed with MBP. See text, section 6, for experimental details. Figure 2 shows high-power micrographs of the optic nerve injury site shown in Figure 1, showing the high concentration of injected cells located at the site of injury.
Figure 3 shows a section in series through the non-injured optic nerve. Figure 4 is a graphical representation of the number of T cells at the site of injury of two different T cell clones primed with MBP or ovalbumin (OVA) antigen (TMBP OR TOVA, respectively) after injury, at various time intervals after the same happens. TMBP and TOVA cells were injected into animals at the time of optic nerve compression, and ipsilateral and contralateral nerves were then removed and prepared for microscopy on days 3, 7, 14 and 21. The figure shows that the T cells accumulated in the site of injury, regardless of the antigen with which they were primed (each result is an average of 5 different experiments, the bar shows the standard deviation). See text, section 6, for experimental details. Figure 5 shows the accumulation of T cells primed with MBP or OVA (TMBP or TOVA, respectively) measured immunochemically using antibodies to T cell receptors. A comparison of the number of accumulated cells in injured optic nerve (ON) and the nerve is illustrated. optical not injured. See text, section 6, for experimental details. Figure 6 shows the accumulation of T cells in injured and uninjured optic nerve after several treatment protocols. Cells MBP specific T (TMBP) were injected immediately after nerve injury (injection of TMBP cells = 0) or 14 days after injury (injection of TMBP cells = 14). Their accumulation in the optic nerve was analyzed 7 days (excision of the nerve - day 7) or 21 days (excision of the nerve = day 21) after the injury. Figure 7 shows the accumulation of T cells in the injured optic nerve one week after the injury. See text, section 6, for experimental details. Anti-MBP or anti-OVA or anti-hsp60 T cell lines were developed, maintained and activated by incubation with MBP of the spinal cords of guinea pigs, or with OVA (Sigma), or with peptide 51-70 of MBP , respectively, in the presence of irradiated syngenic thymic cells (2000 rad). See text, section 7, for experimental details. Activated T cells (1x107 cells) of the anti-MBP or anti-OVA or PBS lines were injected intraperitoneally into adult Lewis rats immediately after injury by unilateral optic nerve compression. Seven days after the injury, the optic nerves were removed, cryosected and analyzed immunohistochemically to detect the presence of marked T cells. The bars show the average total number of T cells counted in 2 or 3 sections of each nerve. Each group contained 3 or 4 rats.
. DETAILED DESCRIPTION OF THE INVENTION In the practice of the invention, compositions comprising activated T cells are used to deliver (a) a diagnostic substance, or (b) a therapeutic substance to a site of CNS injury or disease in a mammal. In general, the T cells of the present invention are T cells that recognize an antigen not normally present or present in small amounts in the circulation. Such antigens include, but are not limited to, SN-specific antigens, cryptic antigens or "non-self" antigens (ie, antigens not normally present in an individual). Non-self antigens may be, without limitation, viral, bacterial, etc., including tissue-specific antigens of a different species or individual. In one embodiment, T cells are activated in vitro by exposure to an antigen, and administered to a mammal. The present invention provides methods for delivering a therapeutic or detectable substance to a site of CNS injury or disease, which comprises administering an effective amount of activated T cells containing or expressing a therapeutic or detectable substance to a mammal, wherein the amount It is effective to detect, diagnose or monitor a site of CNS injury or disease, or is effective in alleviating the effects of a CNS injury or disease. The present invention provides methods for the delivery of substances to a site of CNS injury or disease, which comprises the administration of activated T cells. In the practice of the present invention, the activated T cells supplying the substances may optionally be administered in combination with (a) non-specific T cells specific for the SN, or (b) an SN-specific antigen (or derivative thereof), or (a) and (b). If desired, the methods of the present invention may optionally be combined concurrently with one or more of the following steps: (a) administration, in the CNS, of mononuclear phagocytes (preferably cultured monocytes) that have been stimulated to increase your ability to promote axonal regeneration; (b) administration, in the CNS, of a neurotrophic factor such as fibroblast growth factor; and (c) administering a therapeutic anti-inflammatory substance (i.e., an anti-inflammatory steroid such as dexamethasone or methylprednisolone, or a non-steroidal anti-inflammatory agent or drug, such as aspirin, indomethacin, ibuprofen, fenoprofen, ketoprofen or haproxen, or a peptide anti-inflammatory, such as Thr-Lys-Pro (TKP)). . 1 Substance Delivery Described herein are activated T cells capable of delivering a therapeutic or diagnostic substance to a site of CNS injury or disease. In a preferred embodiment, the activated T cells do not recognize an SN-specific antigen; more preferably, the activated T cells recognize a non-self antigen. In one embodiment, said activated T cells can be used as part of a diagnostic technique to detect a site of damage in the CNS, caused by injury or disease. In another embodiment, activated T cells can be used as part of a therapeutic regimen to alleviate the effects of CNS injury or disease, by promoting axonal regeneration or by inhibiting or preventing CNS degeneration. . 1.1 Therapeutic and diagnostic compositions Activated T cells of the present invention can be used for the delivery of various therapeutic and detectable substances to a site of injury or disease within the CNS. In a preferred embodiment, the activated T cells of the present invention are activated by exposure to an antigen that is not specific for SN, more preferably by exposure to a non-self antigen. The detectable substances can be used to detect, diagnose or monitor a site of CNS injury or disease. In one embodiment, T cells are allogeneic T cells, for example, a preparation of pooled T cells obtained from a blood bank. The use of allogeneic T cells is applicable to various treatments comprising limited administrations including, but not limited to, delivery of T cells to a site of CNS injury for diagnostic purposes; for an individual acute administration or therapy of a dose, etc. In another embodiment, T cells are syngeneic T cells, preferably autologous T cells (ie, from the same individual).
T cells can be isolated and purified according to methods known in the art (Mor and Cohen, 1995, J. Immunol., 155: 3693-3699). For an illustrative example, see section 6.1. For use in the diagnostic methods of the invention, T cells that are preferably located at a site of CNS injury or disease can be detectably labeled. T cells can be detectably labeled with a contrast agent that includes, without limitation, metals such as gold particles, gadolinium complexes, etc. Alternatively, T cells can be detectably labeled with a radioisotope including, but not limited to: 125 Iodo, 131 Iodo or 99mtecnecio. T cells can also be detectably labeled using a fluorescent emitting metal such as 152 Eu, or others of the lanthanide series. Methods for labeling T cells in detectable form can be, for example, those described in Harlow and Lane (Harlow, E. and Lane, D., 1988, "Antibodies: a Laboratory Manual", Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York) and Current Protocols in Immunology (Current Protocols in Immunology, 1997, Eds., Coligan et al., John Wiley &Sons, Inc., NIH), citations that are incorporated herein in their entirety as a reference. . Marking of T cells with metal particles can be achieved by incubating the cells in a suspension comprising the metal particles, wherein the T cells spontaneously include said particles in the cytosol of the cell. Said substances can also be introduced into the cells by various electrophoretic techniques (Current Protocols in Immunology, 1997, Eds. Coligan et al., Jonh Wiley &Sons, Inc., NIH). Metals that emit fluorescence or radioactive metals can be attached to the T cells using metal chelating agents such as diethylenetriaminepentaacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA). The labeling of T cells with a radioisotope can be achieved by incubating the cells with a radioactive metabolic precursor. The presence of labeled active T cells can be detected in the patient using methods known in the art for in vivo screening. These methods depend on the type of brand used. Those skilled in the art will be able to determine the appropriate method for detecting a particular brand. The methods and devices that can be used in the diagnostic methods of the invention include, but are not limited to: computed tomography (CT), full-body scanning such as positron emission tomography (PET), magnetic resonance imaging (MRI), sonography, surgical instrument for radiation response (Thurston and others, US patent 5,441, 050) and scanning instrument for fluorescence response. After being labeled, the T cells of the present invention are activated. T cells can be activated by exposing the cells to one or more of a variety of natural and synthetic antigens and epitopes including, but not limited to, lipopolysaccharide (LPS), myelin basic protein (MBP), myelin / oligodendrocyte glycoprotein (MOG), myelin proteolytic protein (PLP), myelin-associated protein (MAG), S-100, β-amyloid, Thy-1 or neurotransmitter receptors. Preferably, the T cells are activated by an antigen that is not specific for the SN, more preferably, by a non-self antigen. During activation of T cells ex vivo, T cells can be activated by culturing them in a medium to which at least one suitable growth promoter factor was added. Growth promoting factors suitable for this purpose include, without limitation, cytokines, for example, tumor necrosis factor a (TNF-a), interleukin-2 (IL-2) and interleukin-4 (IL-4). In one embodiment, activated T-cards produce an endogenous substance that alleviates the effects of CNS injury or disease. In another embodiment, activated T cells endogenously produce a substance that stimulates other cells including, but not limited to, transforming growth factor β (TGF-β), nerve growth factor (NGF), neurotrophic factor 3 (NT -3), 4/5 neurotrophic factor (NT-4/5), brain-derived neurotrophic factor (BDNF), interferon-d (IFN-d) and interleukin-6 (IL-6), where the other direct cells or indirectly, alleviate the effects of injury or illness. In another embodiment, T cells can be genetically engineered in vitro to insert a nucleotide sequence therein as described in Kramer et al., 1955, Nature Medicine, 1 (11): 1162-1166. The nucleotide sequence is under the control of elements necessary for the processes of transcription and translation, so that a biologically active protein encoded by the nucleotide sequence can be expressed continuously or induced to be expressed as a result of exposure of the T cells. to a microenvironment of a type present at the site of injury. Due to the inherent degeneracy of the genetic code, other nucleotide sequences that encode substantially for the same amino acid sequence or a functionally equivalent amino acid sequence of a protein are within the scope of the invention. Preferably, the expression product of said nucleotide sequence is a secretory protein. Recombinant T cells that contain a coding sequence and that express a biologically active gene product can be identified by at least four general procedures: (a) DNA-DNA or DNA-RNA hybridization; (b) presence or absence of "marker" gene functions; (c) evaluation of the level of transcription measured by the expression of messenger RNA transcripts in the cell; and (d) detection of the product encoded by the nucleotide sequence, measured by immunoassay or by its biological activity. In the first method, the presence of the coding sequence inserted into the expression vector can be detected by DNA-DNA or DNA-RNA hybridization using probes comprising nucleotide sequences that are homologous to the coding sequence or portions or derivatives of the same. In the second procedure, the recombinant expression system can be identified and selected based on the presence or absence of certain functions of the "marker" gene (eg, thymidine kinase activity, antibiotic resistance, methotrexate resistance, transformation phenotype). , body formation of baculovirus occlusion, etc.). For example, if the coding sequence is inserted into a marker gene sequence of a vector, recombinant cells containing the coding sequence can be identified by the absence of the marker gene function. Alternatively, a marker gene may be placed one after the other with a sequence under the control of the same promoter or a different promoter used to control the expression of the coding sequence. Expression of the label in response to induction or selection indicates expression of the coding sequence. In the third procedure, the transcription activity of a nucleotide sequence can be evaluated by hybridization tests. For example, RNA can be isolated and analyzed by Northern blot using a probe having sequence homology with a transcribed coding sequence or non-coding sequence, or particular portions thereof. Alternatively, the total nucleic acid of the host cell can be extracted and quantitatively tested for hybridization with said probes.
In the fourth method, the levels of a protein product can be evaluated immunologically, for example, by Western blots, immunoassays such as radioimmunoprecipitation, enzyme-linked immunoassays, or the like. T cells can be stably transfected with said nucleotide sequences, or they can be transiently transfected. Transient transfection may be applicable for single-dose acute therapeutic regimens. Said nucleotide sequences may code for several substances including, without limitation, therapeutic substances, enzymes that catalyze a therapeutic substance; a regulatory product that stimulates the expression of a therapeutic substance in T cells, etc. Examples include: nucleotide sequences encoding neurotrophic factors such as NGF; nucleotide sequences that code for enzymes that play a role in nerve regeneration of the CNS, such as the enzyme transglutaminase; nucleotide sequences that code for enzymes that catalyze the production of a neurotransmitter, for example, enzymes involved in the catalysis of acetylcholine or dopamine, etc. As a result, T cells that are located at the site of CNS injury or disease produce and secrete the necessary substances at the site. As will be apparent to those skilled in the art, T cells can be preserved, for example, by cryopreservation, before or after culture.
Cryopreservation agents that can be used include, but are not limited to, dimethyl sulfoxide (DMSO) (Lovelock and Bishop, 1959, Nature 183: 1394-1395; Ashwood-Smith, 1961, Nature 190: 1204-1205), glycerol, polyvinylpyrrolidone (Rinfret, 1960, Ann. NY Acad. Sci. 85: 576), polyethylene glycol (Sloviter and Ravdin, 1962, Nature 196: 548), albumin, dextran, sucrose, ethylene glycol, i-erythritol, D-ribitol, D-mannitol (Rowe et al., 1962, Fed. Proc. 21: 157), D-sorbitol, i-inositol, D-lactose, choline chloride (Bender and others, 1960, J. Appl. Physiol. 15: 520), amino acids (Phan The Tran and Bender, 1960, Exp. Cell. Res. 20: 651, methanol, acetamide, glycerol monoacetate (Lovelock, 1954, Biochem. J. 56: 265), inorganic salts (Phan The Tran and Bender, 1960, Proc. Soc. Exp. Biol. Med. 104: 388; Phan The Tran and Bender, 1961, in Radiobiology, minutes of sessions of the Third Australian Conference on Radiobiology, llbery, P.L.T., ed., Butterworth, London, p. 59), and DMSO combined with hydroxyethyl starch and human serum albumin (Zaroulis and Leiderman, 1980, Cryobiology 17: 311-317). A controlled cooling speed is critical. Different cryoprotective agents (Rapatz et al., 1968, Cryobiology 5 (1): 18-25) and different types of cells have different optimal cooling rates. See, for example, Rowe and Rinfret, 1962, Blood 20: 636; Rowe, 1966, Cryobiology 3 \: 12-18; Lewis et al., 1967, Transfusion 7 (1): 17-32; and Mazur, 1970, Science 168: 939-949, for effects of the cooling rate on the survival of the cells and on their transplantation potential. The heat of the melting phase, where the water turns to ice, should be minimal. The cooling process can be carried out by using, for example, a programmable freezing device or a methanol immersion method. Programmable freezing devices allow to determine optimal cooling speeds and facilitate standard reproducible cooling. Programmable speed controlled freezers such as Cryomed or Planar, allow to modulate the freezing regime up to the desired cooling speed curve. After freezing them completely, the cells can be rapidly transferred to a long-term cryogenic storage vessel. In one embodiment, samples can be stored cryogenically in mechanical freezers, such as freezers that maintain a temperature of about -80 ° C to about -20 ° C. In a preferred embodiment, the samples can be stored cryogenically in liquid nitrogen (-196 ° C) or its vapor. Said storage is greatly facilitated by the availability of highly efficient liquid nitrogen refrigerators, which resemble large Thermos containers with extremely low internal insulation and vacuum, so that heat leakage and nitrogen losses are maintained at an absolute minimum. Considerations and methods for long-term manipulation, cryopreservation and storage of T cells can be found, for example, in the following references, incorporated herein by reference: Gorin, 1986, Clinics in Haematology 15 (1): 19- 48; Bone-Marrow Conservation, Culture and Transplantation, minutes of a panel session, Moscow, July 22-26, 1968, International Atomic Energy Agency, Vienna, pp. 107-186. Other methods of cryopreservation of viable cells, or modifications thereof, are available and conceivable for use, as is the case, for example, of cold metal mirroring techniques. See Livesey and Linner, 1987, Nature 327: 255; Linner et al., 1986, J. Histochem. Cytochem. 34 (9): 1123-1135; see also patent of E.U.A. No. 4,199,022 by Senken et al., Patent of E.U.A. No. 3,753,357 by Schwartz, and patent of E.U.A. No. 4,559,298 by Fahy. Preferably, the frozen cells are thawed rapidly (for example, in a water bath maintained at 37-41 ° C), and cooled immediately after thawing. It may be convenient to treat the cells to prevent cell aggregation after thawing. To prevent aggregation, various methods can be used including, but not limited to, the addition before or after the freezing of DNase (Spitzer et al., 1980, Cancer 45: 3075-3085), citrate and low molecular weight dextran, hydroxyethyl starch (Stiff et al., 1983, Cryobiology 20: 17-24), or acid dextrose citrate (Zaroulis and Leiderman, 1980, Cryobiology 17: 311-37), etc. The cryoprotective agent, if toxic to humans, should be removed before the therapeutic use of thawed T cells. One way to remove the cryoprotective agent is by dilution to a negligible concentration. Once the frozen T cells have been thawed and recovered, they are used to promote axonal regeneration as described herein with respect to unfrozen T cells. . 1.2 Uses The compositions and methods of the present invention comprising activated substance-supplying T cells are useful for treating or detecting a site of damage in the CNS caused by injury or disease. Methods to detect a site of injury or disease of the CNS in a mammal comprises: (a) administering to a mammal an effective amount of labeled activated T cells; and (b) detecting in the mammal labeled activated T cells that have accumulated in said site of injury or disease, in which case step (b) is carried out after a sufficient interval to allow said labeled activated T cells administered to step (a) accumulate in said site of injury or illness. For use in the therapeutic methods of the invention, activated T cells can be used to deliver substances to alleviate the effects of injury or disease, for example, by promoting axonal regeneration, or by inhibiting or preventing the degeneration of the CNS. Such substances include, without limitation, growth factors that promote nerve regeneration, such as nerve growth factor (NGF); substances missing from the site of injury, for example, neurotransmitters such as acetylcholine, dopamine; anti-inflammatory substances, etc. In addition, activated T cells can endogenously produce a substance that has a therapeutic effect on CNS injury including, without limitation, interleukins and growth factors. In a preferred embodiment, the activated T cells do not recognize an SN-specific antigen; more preferably, the labeled activated T cells recognize a non-self antigen. The injury or illness can be located in any portion of the CNS, including the brain, spinal cord or optic nerve. An example of such an injury or illness is trauma, including blunt trauma, penetrating trauma, and sustained trauma during a neurosurgical intervention or other procedure. Another example of such injury or disease is stroke, including hemorrhagic stroke and ischemic stroke. Other examples of disease are Alzheimer's disease, multiple sclerosis, Huntington's disease, amyotrophic lateral sclerosis (ALS), Creutzfeldt-Jakob disease and Parkinson's disease. Yet another example of such injury or disease is optic nerve injury that accompanies glaucoma or optic neuropathy. Still other examples of CNS injury or disease will be apparent to those skilled in the art from this description, and are encompassed by the present invention. The compositions and methods of the present invention are useful for treating CNS injury or disease that result in axonal damage whether or not the subject also suffers from another disease of the central or peripheral nervous system, such as a neurological disease of genetic, metabolic or toxic origin. , nutritional, infectious or autoimmune. . 2 Formulations and administration Pharmaceutical compositions for use in accordance with the present invention can be formulated in conventional manner using one or more physiologically acceptable carriers or excipients. The vehicles must be "acceptable" in the sense of being compatible with the other ingredients of the composition and not harmful to those who receive them. The term "vehicle" refers to a diluent, adjuvant, excipient or vehicle with which the therapeutic composition is administered. Methods of administration include, but are not limited to, parenteral (eg, intravenous, intraperitoneal, intramuscular, subcutaneous) and mucosal (eg, oral, nasal, buccal, vaginal, rectal or intraocular routes). The compositions can be formulated for parenteral administration by injection, for example, by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, for example, in ampules or in multi-dose containers. The compositions can take the form of a suspension in aqueous vehicles.
In a preferred embodiment, the compositions comprising activated substance-supplying T cells are formulated in accordance with routine procedures as pharmaceutical compositions adapted for intravenous or intraperitoneal administration to humans. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous pH buffer. When necessary, the composition may also include a solubilizing agent and a local anesthetic such as lidocaine to relieve pain at the site of injection. Usually, the ingredients are supplied separately or mixed together. When the composition will be administered by infusion, it can be supplied with an infusion container containing sterile pharmaceutical grade saline or water. When the composition is administered by injection, a sterile water or saline injection can be provided for injection, so that the ingredients can be mixed prior to administration. The invention also provides a package or pharmaceutical equipment comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. In a preferred embodiment, the pharmaceutical compositions of the invention are administered to a mammal shortly after injury or detection of a degenerative CNS lesion. In one embodiment, the administration of activated substance-supplying T cells can be performed as a single dose, or can be repeated, preferably at 2 week intervals, and then at successively longer intervals once a month, once per week. quarter, once every six months, etc. The course of treatment may last several months, several years and occasionally also throughout the life of the individual, depending on the condition or disease that is being treated. In the case of CNS injury, treatment can vary from several days to months or even years, until the condition has stabilized and there is only a limited or no risk of developing secondary degeneration. In chronic human diseases or conditions such as Alzheimer's disease or Parkinson's disease, therapeutic treatment in accordance with the present invention may be for life. As will be apparent to those skilled in the art, the therapeutic effect is sometimes dependent on the condition or disease to be treated, the age and health condition of the individual, other physical parameters (e.g., sex, weight, etc.). of the individual, as well as several other factors, for example, if the individual is taking other drugs, etc. The optimal dose of the therapeutic compositions comprising activated T cells of the invention is proportional to the number of nerve fibers affected by the CNS lesion or disease at the site being treated. In a preferred embodiment, the dose ranges from about 5 x 106 to about 107 to treat a lesion affecting approximately 105 nerve fibers, such as a complete cross section of the optic nerve of a rat, and ranges from about 107 to about 108. to treat an injury that affects approximately 105 nerve fibers, such as a complete cross section of a human optic nerve. As will be apparent to those skilled in the art, the dose of T cells can be increased or decreased proportionally to the number of nerve fibers affected in the injury or site of damage being treated. 6. EXAMPLE Accumulation of activated T cells in damaged CNS 6. 1 Materials and methods 6. 1.1 Animals Female Lewis rats were obtained from Harlan Olac (Bicester, United Kingdom), grouped by age (8 to 12 weeks), and housed four per cage at a controlled light and temperature site. 6. 1.2 Proteins Used for Activation of T Cells Myelin basic protein (MBP) from spinal cord of guinea pigs was prepared as described previously (Ben-Nun et al., Cited above (1982)). Chicken ovalbumin (OVA) was purchased from Sigma (Israel). H37RA from heat-inactivated Mycobacterium tuberculosis (M. tuberculosis) and incomplete Freund's adjuvant (IFA) were purchased from Difco Laboratories (Detroit, Ml, USA). 6. 1.3 Media The T cell proliferation medium contained the following: Dulbecco's modified Eagle's medium (DMEM, Biological Industries, Israel), supplemented with 2mM L-glutamine (L-Glu, Sigma, USA), 5x10-mercaptoethanol "9M (2-ME, Sigma), penicillin (100 IU / ml, Biological Industries), streptomycin (100 μg / ml, Biological Industries), sodium pyruvate (1 mM, Biological Industries), non-essential amino acids (1 ml / 100 ml; Biological Industries) and autologous rat serum at 1% (vol / vol) (Mor et al., Clin. Invest., 85: 1594 1990).) The propagation medium contained: DMEM, 2-ME, L- Glu, sodium pyruvate, non-essential amino acids and antibiotics at the same concentration used previously, and also 10% fetal calf serum (FCS) and 10% T cell growth factor (TCGF) obtained from the supernatant of spleen cells stimulated by concanavalin A (Mor et al., cited above, 1990). 6. 1.4 Antibodies MBP was prepared from the spinal cord of guinea pigs as described (Hirshfeld et al., 1970, FEBS Lett.7: 317). OVA was acquired from Sigma (St. Louis, Missouri). P51-70 of the rat 18.5 kDa MBP isoform (sequence: APKRGSGKDSHTRTTHYG) SEQ ID NO: 1 and p277 of human hsp60 (sequence: VLGGGCALLRCPALDSLTPANED) SEQ ID NO: 2 were synthesized (Elias et al., 1991, Proc. Nati, Acad. Sci. USA 88, 3088-91), using the 9-fluorenylmethoxycarbonyl technique with an automatic multiple peptide synthesizer (AMS 422, ABIMED, Langenfeld, Germany). The purity of the peptides was analyzed by HPLC and amino acid composition. 6. 1.5 T cell lines T cell lines were obtained by draining lymph node cells obtained from Lewis rats immunized with an antigen. The antigen was dissolved in PBS (1 mg / ml), and emulsified with an equal volume of incomplete Freund's adjuvant (Difco Laboratories, Detroit, Michigan) supplemented with 4 mg / ml of Mycobacterium tuberculosis (Difco). The emulsion (0.1 ml) was injected into the bearing of the hind legs of the rats. Ten days later, the antigen was injected, the rats were sacrificed and the draining lymph nodes were surgically removed and dissociated. The cells were washed and activated with the antigen (10 μg / ml) in proliferation medium containing modified Dulbecco's Eagle's medium (DMEM) supplemented with L-glutamine (2mM), 2-mercaptoethanol (5x10"5M), sodium pyruvate (1 mM), penicillin (100 IU / ml), streptomycin (100 μg / ml), non-essential amino acids (1 ml / 100 ml) and autologous rat serum at 1% (volume / volume). hours at 37 ° C, 90% relative humidity and CO2 at 7%, the cells were transferred to propagation medium containing additionally fetal calf serum (FCS) at 10% (volume / volume) and T cell growth factor to 10% derived from the supernatant of spleen cells stimulated by concanavalin A. The cells were grown in propagation medium for 4 to 10 days before being re-exposed to the antigen (10 μg / ml) in the presence of irradiated thymus cells (2000 rad) (107 cells / ml) in proliferation medium, the T cell lines were ex pandidas through repeated re-exposure and propagation. 6. 1.6 T cell labeling T cells were washed and suspended in Hoechst 33342 dye at 10.7 μm (Molecular Probes, USA) for 10 minutes at 37 ° C. The cells were washed twice with 50 ml volumes of PBS, and resuspended at 5 x 106 cells / ml on ice until their injection. 6. 1.7 Compression injury of the rat optic nerve Compression injuries were produced as described previously (Hirschberg et al., 1994, J. Neuroimmunol 50: 9-16). For a short time, the rats were deeply anesthetized by i.p. of xylazine (10 mg / kg; Rompun) and ketamine (50 mg / kg; Velalar). Under a binocular operating microscope, lateral cantectomy was performed in the right eye, and the conjunctiva was cut laterally to the cornea. After separating the muscles of the retractor bulbs, the optic nerve was exposed intraorbitally by forceful dissection. A moderate compression injury to the optic nerve occurred at 2 mm from the eye, using a calibrated transverse action forceps (Duvdevani et al., Instructure Neurology and Neuroscience, 2:31, 1990). The contralateral nerve was left intact and was used as a control. 6. 1.8 Nerve Sectioning At specified time points, rats were euthanized by prolonged anesthesia with ether, and their optic nerves were removed surgically, immersed in Tissue-Tek (Miles Inc., USA), and frozen in liquid nitrogen cooled in Isopentane (BDH, United Kingdom). The nerves were then transferred to dry ice and stored at -70 ° C until sectioned. Longitudinal nerve sections maintained in cryostat (20 μm thick), were placed on glass slides coated with gelatin (4 sections per slide), and frozen at -20 ° C until they were observed or prepared for fluorescence staining. 6. 1.9 Analysis of T cell data in nerve sections Severed nerves were prepared and sectioned in several periods after injury. The counting of nuclei labeled with Hoechst dye or cells immunostained in each section was made using a fluorescence microscope. For each time point, the count of five sections was made, and the figures were averaged. 6. 1.10 Immunostaining of nerve sections Longitudinal nerve sections maintained in cryostat (from μm thick), were placed on gelatin-coated glass slides, and frozen until preparation for fluorescence staining. The sections were thawed and fixed in ethanol for 10 minutes at room temperature, washed twice with bidistilled water (ddH2O), and incubated for 3 minutes in PBS containing 0.05% polyoxyethylene sorbitan monolaurate (Tween-20; Sigma, USA). Then, they were incubated overnight at 4 ° C with a mouse monoclonal antibody directed against rat macrophages (ED1; 1: 400; Serotec, UK), and antibody against rat fibrillar acidic cell protein (GFAP; : 100; BioMakor), all diluted in PBS containing 3% FCS. T cell staining was carried out by incubating the nerve sections for 1 hour at room temperature with a mouse monoclonal antibody directed against rat T cell receptor (TCR) (1: 100, Hunig et al., J. Exp. Med., 169: 73. 1989) in PBS containing 3% FCS and 2% BSA. After three washes with PBS containing 0.05% Tween-20, the sections were incubated with F (ab ') 2 goat anti-mouse conjugated with fluorescein isothiocyanate (FITC, BioMakor) or rhodamine tetramethyl isothiocyanate (TRITC, BioMakor) at a dilution of 1: 100 and 1: 50, respectively, for 1 hour at room temperature. Then, they were washed with PBS containing Tween-20 and treated with glycerol containing 1,4-diazobicyclo- (2,2,2) octane (Sigma), to inhibit the fluorescence extinction. Sections were observed with a Zeiss universal fluorescence microscope using filters that detect TRITC, FITC and Hoechst dyes (Blaugrund et al., Exp. Neurol., 118: 105, 1992, Blaugrund et al., Brain Res., 574: 244, 1992). 6. 2 Results 6. 2.1 Accumulation of activated T cells T-cell clones primed for MBP (TMBP), were activated with MBP for 2 days before being labeled with Hoechst dye, and injected i.p. in animals at the time of injury. At 3, 7, 14 and 21 days after the injury, the nerves were excised, cryosected and analyzed microscopically to detect the presence of labeled T cells. TMBP cells were detected in the damaged optic nerves on day 3, and accumulated to a maximum on day 14 (Figure 1). Large groups of TMBP cells were observed at the site of injury, and fewer individual cells were observed proximally and distally (Fig. 29). Four weeks after the injury, the labeled T cells were still detectable in optic nerves in the process of degeneration. No T cells were found in the untreated optic nerves (Figure 3), untreated sciatic nerve or injured sciatic nerve at any time after the injury. Occasionally, marked T cells were found in capillaries and in connective tissue, but they were not concentrated or located in any specific area. T cells that were not previously stimulated with antigen did not accumulate in any of the nerves, including damaged nerves. The accumulation of TMBP cells in the injured CNS, but not in the injured SNP, suggests that there could be some specific interaction between the primed T cells and the CNS tissue from which the MBP antigen was originally derived. To determine whether the injured CNS interacted with T cells in general, or specifically with T cells primed with a CNS antigen, the previous experiments were repeated using a clone that responds to chicken ovalbumin (TOVA) - The rats were injected with a clone of labeled TOVA previously stimulated with ovalbumin (OVA) using the same protocol as in the case of TMBP-cells. The labeled TOVA cells accumulated in the injured optic nerve, and the accumulation pattern was similar to that of the TMBP-cells. the counting of TOVA and TMBP cells marked on longitudinal sections of the optic nerve prepared 3, 7, 14 and 21 days after the lesion. No significant difference was observed in the number of TMBP and TOVA cells in the injured optic nerve (Figure 4), indicating that the specificity of the antigen has little to do with the accumulation of T cells in the sites of CNS injury. TMBP cells were detectable a bit earlier than TOVA cells at the site of optic nerve injury, so the specificity of the antigen may play a role in the process, but this is not sufficient to explain the large accumulation of TOVA cells at the site of injury.
Figure 5 shows the accumulation of immunocytochemically measured T cells using antibodies to T cell receptors. This detection technique excludes the possibility that the observed labeling is due to the phagocytic cells that had phagocytosed the previously labeled T cells shown in the figure 1. The graph shows a marked elevation in the accumulation of T cells after injury, regardless of whether the systematically injected T cells are specific for an epitope of their own (MBP) or for a non-self epitope (OVA). Figure 6 shows that the accumulation of T cells depends on the lesion, and not on the rupture of the blood-brain barrier. T cells specific for MBP or OVA were injected two weeks after the injury, and their accumulation was analyzed 1 week later, namely 21 days after the primary lesion. Its accumulation was compared to that of the injected T cells immediately after the lesion, and was detected 7 or 21 days later. It seems that the time elapsed between the injury and the injection of T cells, which is a factor in the closure of the blood-brain barrier, is not an important factor in the accumulation of T cells. 7. EXAMPLE Uses of activated T cells 7. 1 Materials and methods Animals, proteins used for the stimulation of T cells, media, compression injury of the rat optic nerve, sectioning of nerves, immunostaining of nerve sections, and data analysis of T cells in nerve sections, are described in section 6, cited above. 7. 1.1 Establishment of T cell lines with active experimental allergic encephalomyelitis (EAE) MBP and OVA were dissolved in PBS (1 mg / ml), and emulsified with an equal volume of incomplete Freund's adjuvant (IFA) supplemented with 4 mg / ml of M. tuberculosis. The rats were immunized subcutaneously in the bearing of the hind legs with 0.1 ml of the emulsion. On day 9 (1 to 3 days before the clinical onset of the disease), the animals were euthanized, and the draining lymph nodes were surgically excised and dissociated under sterile conditions. The cells were washed and placed in proliferation medium with irradiated thymocytes (2000 rads) and 10 μg / ml of MBP, OVA or M. tuberculosis for 3 days. Then, the cells were washed and placed in a medium of propagation for 5 to 10 days, at which time they were again exposed to irradiated peptides and thymocytes in proliferation medium. The T cell lines were expanded by re-exposure and propagation, and tested for specificity in a proliferation test of antigen-specific T cells. The lines were expanded, and the supply materials were frozen in liquid nitrogen. The cells were thawed and stimulated once before their use in experiments. 7. 1.2 Passive transfer of T cell lines T cell lines were activated by in vitro restimulation with their own antigen (10 μg / ml) in proliferation medium. After incubation for 48 to 72 hours at 37 ° C, 90% relative humidity and CO2 at 7.5%, the cells were washed. Viable cells were isolated on Percoll and suspended in PBS. The animals were injected i.p. with 10 x 10 6 cells / ml. The control animals were injected i.p with 1 ml of PBS. 7. 1.3 Compression injury of the rat sciatic nerve Under deep anesthesia as described in section 6.1.5, the sciatic nerve was exposed, and a similar compression injury occurred. At the end of the intervention, the skin was sutured. 7. 1.4 Retrograde Marking of RGCs The optic nerve was exposed, without damaging the retinal blood flow. Solid crystals of the dye, 4- (4- (dikecylamine) styryl) -m-methyl-pyridinium (4-Di-10-Asp) iodide (Molecular Probes, Europe BV), 1 to 2 mm from the edge were deposited distal from the site of injury. Similarly, untreated optic nerves were marked at approximately the same distance from the eyeball. 5 days after the application of the dye, the retinas were excised under deep anesthesia, mounted uniformly in 4% paraformaldehyde solution, and the retinal ganglion cell (RGC) count was determined by fluorescence microscopy. 7. 1.5 Evaluation of the effects of injected T cells The effect of the injected T cells on the number of surviving optic nerve fibers was monitored by retrograde labeling of RGCs (see above) immediately after the lesion to assess primary degeneration, and two weeks later to evaluate secondary degeneration. 5 days after the application of the dye (4-Di-10-Asp), the retinas were excised, mounted intact, and their RGCs counted. The count was done in 5 randomly selected fields in each retina (all located at approximately the same distance from the optical disk). In all cases, the dye was applied 2 ml distally to the site of the previous insertion. Using this elongation approach, only the RGCs whose axons were still viable could be marked. The RGCs in each group of injured nerves treated only with PBS were injected with TMBP or TOVA- cells. The results were expressed as a percentage of axons, compared to those that survived the first shock (42% of the axons remained after the first shock). ). 7. 1.6 Clinical evaluation of EAE Clinical disease was evaluated every 1 to 2 days according to the following neurological scale: 0, without abnormality; 1, atony of the tail; 2, paralysis of the hind leg; 3, paralysis that extends to the thoracic spine; 4, paralysis of the front leg; 5 moribund state. 7. 2 Results 7. 2.1 Accumulation of ATCs The injured optic nerve was analyzed for the accumulation of T cells. As shown in Figure 7, in the non-injured optic nerves of control rats injected with phosphate buffered saline (PBS), no T cells could be detected. Small but significant numbers of T cells were observed in the non-injured optic nerves of rats injected with anti-MBP T cells (primed against a peptide comprising amino acids 51-70 of MBP, "p51-70" , which is known to be capable of inducing experimental autoimmune encephalomyelitis (EAE) under these experimental conditions), but not of rats injected with anti-OVA T cells. The compression injury of the optic nerve was accompanied by a small but significant accumulation of endogenous T cells, possibly reflecting a response to self antigens triggered by the lesion. In the injured optic nerves, the accumulation of T cells increased significantly 5 to 6 times) in rats injected with anti-OVA, anti-hsp60 or anti-MBP T cells. These observations confirmed the previous finding that axonal CNS injury is accompanied by the accumulation of endogenous T cells, and that this accumulation is increased by the systemic injection of activated T cells regardless of their specific antigenic character. 8. Discussion The results of the experiments described in sections 6 and 7 show that activated T cells accumulate at the site of CNS injury. In addition, the results also show that the accumulation of T cells at the site of injury is a non-specific process, ie, T cells that accumulate at the site of injury include T cells that are activated by exposure to an antigen present in the site of injury, as well as T cells that are activated by an antigen not normally present in the individual. The results of the experiments described in section 7 demonstrate that the beneficial effects of T cells in alleviating damage due to CNS injury are associated with a specific antigen of the SN, as illustrated by MBP. More specifically, the administration of non-recombinant T cells that were activated by exposure to an antigen causing autoimmune disease (T BP), rather than aggravating the lesion, led to a significant degree of protection from secondary degeneration. In this way, activation of T cells by exposure to a fragment of an SN-specific antigen, was beneficial in limiting the diffusion of the lesion in the CNS. The present findings show that secondary degeneration can be inhibited by the transfer, in the individual, of T cells that recognize a specific antigen of the SN, which is present at the site of injury. In addition, the studies described in sections 8 and 9 show that activation of T cells by administering an immunogenic antigen (eg, MBP) or an immunogenic epitope of an antigen (eg, MOG p35-55), can be used to prevent or inhibit secondary degeneration of the CNS following the injury. The present apation claims the priority benefits of Israeli patent apation IL 124550, filed May 19, 1998, the disclosure of which is hereby incorporated by reference in its entirety. The present invention is not limited in scope by the exemied embodiments, which are considered as illustrations of singular aspects of the invention. In addition, various modifications of the invention, in addition to those shown and described herein, will be apparent to those skilled in the art from the foregoing description and the accompanying drawings. It is intended that said modifications be within the scope of the appended claims. All publications cited herein are incorporated herein by reference in their entirety.

Claims (25)

NOVELTY OF THE INVENTION CLAIMS
1. - A recombinant T cell comprising a promoter operably linked to a nucleotide sequence coding for a protein that alleviates the effects of a lesion or disease of the central nervous system (CNS), characterized in that the recombinant T cells do not recognize a specific antigen of the nervous system (specific to the SN).
2. The recombinant T cell according to claim 1, further characterized in that the protein is a therapeutic substance or an enzyme that catalyzes the production of said therapeutic substance or a regulatory product, which induces the production of a therapeutic substance.
3. The use of an active ingredient comprising the recombinant T cell according to claim 1, for the preparation of a pharmaceutical composition for the treatment of a CNS lesion or disease in a mammal.
4. The use according to claim 3, wherein said mammal is a human.
5. The use according to claim 3, wherein said recombinant T cell is produced using an autologous T cell.
6. A method for detecting a site of injury or disease of the central nervous system (CNS) in a mammal, characterized in that it comprises: a) administering to a mammal an effective amount of labeled activated T cells that do not recognize a specific antigen of the nervous system ( specific SN); and b) detecting in the mammal the labeled activated T cells that have accumulated in said site of injury or disease, wherein step (b) is carried out after a sufficient interval to allow said labeled activated T cells administered in the step (a) accumulate in said site of injury or illness.
7. The method according to claim 6, further characterized in that the mammal is a human.
8. The method according to claim 6, further characterized in that the activated T cells recognize a non-self antigen.
9. The method according to claim 6, further characterized in that the labeled activated T cells are labeled with a radioisotope, a contrast agent or a metal that emits fluorescence.
10. The method according to claim 6, further characterized in that the activated T cells are administered intravenously, intraperitoneally, intramuscularly or subcutaneously.
11. - The method according to claim 6, further characterized in that said cells are autologous.
12. A pharmaceutical composition comprising isolated activated T cells that do not recognize a specific antigen of the nervous system (SN specific), for use to deliver a therapeutic or detectable substance, wherein the activated T cells contain a therapeutic substance or express a recombinant substance having a therapeutic effect, or containing a detectable substance added exogenously when administered in vivo to a mammal; and a pharmaceutically acceptable vehicle.
13. The pharmaceutical composition according to claim 12, further characterized in that said activated T cells are generated by exposing the T cells to a non-self antigen or a mitogen.
14. The pharmaceutical composition according to claim 12, further characterized in that said activated T cells endogenously produce said therapeutic substance.
15. The pharmaceutical composition according to claim 12, further characterized in that said activated T cells are T cells that have been genetically engineered to express said therapeutic substance or to express an enzyme that catalyzes the production of said therapeutic substance or a regulatory product. , that induces the production of said therapeutic substance.
16. - The pharmaceutical composition according to claim 12, further characterized in that said activated T cells express said therapeutic substance or enzyme or regulatory product, under the control of elements necessary for transcription or translation continuously or after induction.
17. The pharmaceutical composition according to claim 12, further characterized in that said activated T cells are used to treat a lesion comprising blunt trauma, penetrating trauma, sustained trauma during a neurosurgical intervention, hemorrhagic stroke or ischemic stroke.
18. The pharmaceutical composition according to claim 12, further characterized in that said activated T cells are used to treat a disease selected from the group consisting of Alzheimer's disease, Parkinson's disease, multiple sclerosis, glaucoma, optic nerve injury that it accompanies optic neuropathy, Huntington's disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease.
19. The use of an active ingredient comprising activated T cells that do not recognize a specific antigen of the nervous system (specific for the SN), for the preparation of a pharmaceutical composition for the treatment or detection of a human condition or disease of the SNC.
20. The use according to claim 19, wherein said activated T cells are generated by exposing the T cells to a non-self antigen or a mitogen.
21. The use according to claim 19, wherein said activated T cells endogenously produce said therapeutic substance.
22. The use according to claim 19, wherein said activated T cells are T cells that have been genetically engineered to express said therapeutic substance or to express an enzyme that catalyzes the production of said therapeutic substance or to express a regulatory product. , that induces the production of said therapeutic substance.
23. The use according to claim 19, wherein said activated T cells express said therapeutic substance or enzyme or regulatory product, under the control of elements necessary for transcription or translation continuously or after induction.
24. The use according to claim 19, wherein said activated T cells are used to treat a lesion comprising blunt trauma, penetrating trauma, sustained trauma during a neurosurgical intervention, hemorrhagic stroke or ischemic stroke.
25. The use according to claim 19, wherein said activated T cells are used to treat a disease selected from the group consisting of Alzheimer's disease, Parkinson's disease, multiple sclerosis, glaucoma, optic nerve injury that accompanies optic neuropathy, Huntington's disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease.
MXPA/A/2000/000840A 2000-01-24 Activated t-cells and their uses MXPA00000840A (en)

Publications (1)

Publication Number Publication Date
MXPA00000840A true MXPA00000840A (en) 2001-11-21

Family

ID=

Similar Documents

Publication Publication Date Title
JP4328050B2 (en) Use of Copolymer 1, related peptides and polypeptides and T cells treated with them for neuroprotective therapy
JP4328090B2 (en) Use of Copolymer 1, Related Peptides and Polypeptides and T Cells Treated With them in Protective Therapy of Neurons
US7560102B2 (en) Method for reducing neuronal degeneration so as to ameliorate the effects of injury or disease
US20120135016A1 (en) Induction of neurogenesis and stem cell therapy in combination with copolymer 1
WO1999060021A2 (en) Use of activated t cells, nervous system-specific antigens for treating disorders of the nevrous system
US6835711B2 (en) Use of poly-Glu,Tyr for neuroprotective therapy
AU2002345323A1 (en) Use of poly-Glu, Tyr for neuroprotective therapy
JP2006503808A (en) Antigen-presenting cells for neuroprotection and nerve regeneration
EP1039929A1 (en) Activated t-cells and their uses
MXPA00000840A (en) Activated t-cells and their uses
JP2005504716A (en) Pharmaceutical composition comprising a modified CNS-derived peptide for promoting nerve regeneration and preventing neurodegeneration
AU768763B2 (en) Activated T cells, nervous system-specific antigens and their uses
AU755872B2 (en) Activated T-cells and their uses
EP2195650B1 (en) Methods of treating tumors in immune-privileged sites
US20100272787A1 (en) Amethod of treating neurodegenerative diseases
MXPA00011385A (en) Activated t cells, nervous system-specific antigens and their uses