WO2006136116A1 - Use of interferon-alpha in order to obtain a compound for the treatment of optical neuromyelitis - Google Patents
Use of interferon-alpha in order to obtain a compound for the treatment of optical neuromyelitis Download PDFInfo
- Publication number
- WO2006136116A1 WO2006136116A1 PCT/CU2006/000005 CU2006000005W WO2006136116A1 WO 2006136116 A1 WO2006136116 A1 WO 2006136116A1 CU 2006000005 W CU2006000005 W CU 2006000005W WO 2006136116 A1 WO2006136116 A1 WO 2006136116A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- nmo
- ifn
- patients
- treatment
- interferon
- Prior art date
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/21—Interferons [IFN]
- A61K38/212—IFN-alpha
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/18—Growth factors; Growth regulators
- A61K38/1816—Erythropoietin [EPO]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P27/00—Drugs for disorders of the senses
- A61P27/02—Ophthalmic agents
Definitions
- the present invention relates to the biological sciences, biotechnology and medical sciences, and is based on a new use of interferon alpha (IFN ⁇ ) which due to its antiviral, immunomodulatory, regulatory and preventive T-cell inductors demyelination is able to stop the progression of optic neuromyelitis disease (NMO) in the single phase form, reduce the number of outbreaks and prolong the time between relapses in patients suffering from the recurrent form of NMO.
- IFN ⁇ interferon alpha
- NMO is characterized by attacks of optic neuritis and myelitis (Devic E. Myelitis follows complications of neurite optique. (1894) BuII Med 8: 1033-1034; Devic E. Myelite aigue dorse-lombaire de neurite optique, autopsy. (1895 ) Congress Francais Medicine (Premiere Session, Lyon) 1: 434-9; Gault F. De la neuromyelite optique aigue. (Thesis) Lyon; (1894)). These clinical events can also occur frequently in Multiple Sclerosis (MS), however, both the epidemiology, clinical characteristics, laboratory, etiopathogenesis and non-response to the accepted conventional treatments for MS, allow to consider the NMO as an entity nosological independent of MS.
- MS Multiple Sclerosis
- NMO Newcastle disease virus
- MS has a prevalence between 100-200 / 100,000 inhabitants
- NMO is considered a rare disease.
- the NMO seems to be common in non-Caucasians such as Africans, Americans, Japanese and other Pacific islands. More than 6% of cases of demyelinating diseases in India are NMO (Singhal BS. Multiple sclerosis-Indian experience. (1985) Annals of the Academy of Medicine, Singapore 14: 32-6). In Cuba the prevalence is unknown but has been increasing in recent years. The role of genetic factors is unknown. There are reports of identical twins with NMO.
- HLA human leukocyte antigens
- the NMO the paraclinical measurements such as Nuclear Magnetic Resonance (NMR) of the brain and spinal cord, as well as the analysis of the cerebrospinal fluid (CSF) also differ with MS.
- NMR Nuclear Magnetic Resonance
- CSF cerebrospinal fluid
- NMO no or few lesions in the white matter are detected in the brain, however the spinal cord images show distinctive elements: the majority of patients have extensive lesions that extend longitudinally over 3 or more vertebral segments.
- patients with NMO have a pleocytosis of more than 50 leukocytes with or without the presence of neutrophils.
- the NMO tends to follow a single-phase or outbreak or relapse course (more than 70% of cases have a recurrent course) (Wingerchuk DM, Hogancamp WF, O'Brien PC, Weinshenker BG. The clinical course of optic neuromyelitis (Devic's syndrome ). (1999) Neurology 53: 1107-14).
- patients experience unilateral or bilateral optic neuritis (NO) with a simple episode of myelitis, typically, although not always within a short period of time from each other, but do not have subsequent relapses unlike patients with a relapsing course that continue to have discrete exacerbations of NO and / or myelitis.
- NO optic neuritis
- antigens released into the CNS during the destructive process can reach the peri-vascular space and be recognized there by antibodies from the circulation.
- a non-specific inflammatory reaction initiated by the deposition of circulating immune complexes may be involved.
- the classical complement pathway is activated, allowing the recruitment of activated macrophages to these peri-vascular sites where they bind via a receptor for complement components or Ig / Fc receptors.
- Activated macrophages together with eosinophils and neutrophils, locally generate cytokines, proteases and free radicals of oxygen / nitrogen which can contribute to both vascular and parenchymal damage, resulting in non-selective destruction of white and gray matter, including axons and oligodendrocytes Increased vascular permeability and edema can contribute to parenchymal damage via secondary ischemia or favor the typical central location of NMO plaques within the spinal cord (Prineas JW, McDonald Wl. Demyelinating diseases (1997) In: Graham Dl, Lantos PL , editors, Greenfield's neuropathology, 6 th ed. London: Arnold; p. 813-96).
- Activated eosinophils release proteins with basic granules such as Myelin Basic Protein (MBP), eosinophil-derived neurotoxin, cationic protein from eosinophils and eosinophil peroxidase (Kaneko M, Kita H, Gleich GJ. Eosinophil basic proteins. In: Barnes PJ, Grunstein MM, Leff AR 1 Woolcock AJ, editors. Asthma. (1997) Philadelphia: Lippincott-Raven p 593-607) . These granules have cytotoxic properties and serve as eosinophil activation markers (Venge P.
- eosinophil cationic protein ECP
- eosinophil cationic protein ECP
- Resp Med 89 1- 2
- CCR3 is the main receptor for chemokine eotaxin, a potent chemo-attractant for eosinophils.
- CCR3 is expressed primarily in human eosinophils (Ponath PD, Qin S, Post TW, Wang J, Wu L, Gerard NP, et al.
- Eosinophils are an important source of IL-4, which can cause a change in the cytokine profile from Th 1 to Th2 (Rumbley CA, Sugaya H, Zekavat SA, El Refaei M, Perrin PJ, Phillips SM. Activated eosinophils are the major source of Th2- associated cytokines in the schistosome granuloma. (1999) J Immunol 162: 1003-9). Patients with NMO often have circulating auto-antibodies with a frequency that exceeds that view in MS (Wingerchuk DM, Hogancamp WF, O'Brien PC, Weinshenker BG. The clinical course of optic neuromyelitis (Devic's syndrome).
- BHE is highly impermeable to circulating plasma proteins and leukocytes and therefore can protect the CNS from an immune reaction.
- BHE is not effective, such as spinal nerve roots, it may happen that circulating pathogenic antibodies can access the CNS via these structures and spread to the immediate vicinity.
- active lesions predominantly affect the spinal cord (Lassmann H. Comparative neuropathology of chronic experimental allergic encephalomyelitis and multiple sclerosis. (1983) committeeenr Neurol 25: 1- 135). NO in EAE also tends predominantly to occur in the retrobulbar optic nerve (Guy J, Rao NA. Acute and chronic experimental optic neuritis. Alteration ⁇ n the blood-optic nerve barrier. (1984) Arch Ophthalmol 102: 450-4).
- the permeability of the increased BHE in the spinal cord may be due to vascular properties inherent in this region where the capillaries are longer than in the brain. Therefore, with a background inflammatory process, in the presence of extremely high antibody titres, the lesions may preferentially but not exclusively affect the spinal cord and the optic nerve. This hypothesis is compatible with the observation that in late stages of NMO, lesions often spread to other CNS regions (Wingerchuk DM, Hogancamp WF 1 O'Brien PC, Weinshenker BG. The clinical course of optic neuromyelitis (Devic's syndrome) (1999) Neurology 53: 1107-14).
- Devic NMO The important activation of complement, eosinophilic infiltration and vascular fibrosis observed in cases of Devic NMO is more prominent compared to MS, and supports the role of humoral immunity in the pathogenesis of NMO (Lucchinetti CF, Mandler RN, McGavem D , Bruck W, Gleich G, Ransohoff RM 1 Trebst C 1 Weinshenker B, Wingerchuk D, Parisi JE, Lassman H. (2002) A role for humoral mechanism in the pathogenesis of Devic's optic neuromyelitis. Brain 125: 1450-61).
- NMO HLA associations of seropositive rheumathoid arthritis in a Cree and Ojibway population. (1998) J Rheumatol 25: 2319-2323) than in MS. NMO has been associated with several systemic diseases including vascular diseases of collagen, autoantibody syndromes, infections and exposures to toxic agents.
- the NMO has also been associated with infectious diseases, for example, viral, frequently the NMO has an infectious prodrome characterized by headache, myalgias and high respiratory symptoms, in few cases an infectious agent is identified, there are reports of NMO after acute infectious mononucleosis (Williamson PM. Optic neuromyelitis following infectious mononucleosis. (1975) Proc Aust Assoc Neurol 12: 153-155), Varicella-zoster (Williamson PM. Optic neuromyelitis following infectious mononucleosis. (1975) Proc Aust Assoc Neurol 12: 153-155) .
- hypothalamic dysfunction As a cause of hyperprolactinemia.
- lesions with Gadolinium were found in the hypothalamic-pituitary region in the NMR. All patients suffered from recurrent NO and myelitis that was resistant to immunosuppressive therapy and finally caused blindness and paraplegia. The oligoclonal bands in the CSF were found only in one patient.
- tissue metalloproteinase inhibitor (TIMP-1) was significantly reduced in the EMRR CSF but not in the NMO CSF (Mandler RN, Dencoff JD, Midani F, Ford CC, Ahmed W, Rosenberg GA. Matrix Metalloproteinases and tissues inhibitors of metalloproteinases n cerebrospinal fluid differ in multiple sclerosis and Devic's optic neuromyelitis. (2001) Brain 124: 493-498). These biochemical differences in the CSF may be well related to differences in the inflammatory tissue reaction in MS and NMO of Devic.
- NMO is an invalidating and aggressive inflammatory demyelinating disease where no effective treatment for this disease has been found.
- Plasmapheresis has been reported beneficial in the management of acute crises of patients with NMO with or without associated connective tissue disorders (Konttinen YT, Kinnunen E, von Bonsdorff M, Lillqvist P, lmmonen I, Bergroth V, et al. Acute transverse myelopathy successfully treated with plasmapheresis and prednisone ⁇ na patient with primary Sjodren's syndrome. (1987) Arthritis Rheum 30: 339-44; Fletchner KM, Baum K.
- Plasmapheresis in multiple sclerosis patients with di ⁇ ferent indications (2001) MuIt Scler 7 Suppl 1: S64). Plasmapheresis reduces the number of circulating autoantibodies and immune complexes (Clark WF, Dan PC, Euler HH 1 Guillevin L, Harstord J, Heer AH, et al.
- Type I IFNs have a potent antiviral effect, they are induced after the cells are infected by viruses, causing the synthesis of a large number of enzymes such as 2.5'oligoadenylate synthetase that interferes with RNA or DNA viral replication.
- This early (non-specific antigen) response is critical to limit the spread of the viral spectrum before the antigen-specific response can completely control the infection.
- mononucleosis Williamson PM. Optical neuromyelitis following infectious mononucleosis.
- Devic's optic neuromyelitis and HIV-1 infection (2000) J Neurol Neurosurg Psychiatry 68: 795-796), where the symptoms and signs of NMO they were separated by days or weeks of the viral infection by which it is compressible to associate it with a possible infectious etiology and a possible therapeutic with a drug with antiviral properties.
- Th1 / Th2 paradigm Although there are conflicting reports that question the Th1 / Th2 paradigm, recent evidence suggests that IFN alpha in humans can directly induce a Th 1 I cell differentiation which would be beneficial in NMO characterized by a Th2 response, which constitutes an additional element that justifies The rationality of the use of interferon alfa in obtaining a pharmaceutical compound for the treatment of optic neuromyelitis.
- T lymphocytes were cultured in the presence of IFN-alpha and cloned (Parronchi P, De Carli M, Manetti R, Simonelli C, Sampognaro S, Piccini MP, Macchia D, Maggi E, Del Prate G, Romagnani S.
- IL-4 and IFN alfa and IFN gamma exert opposite regulatory effects on the development of cytolytic potential by Th1 or Th2 human T cell clones. (1992) J Immunol 149: 2977-2983) or directly stimulated (Brinkmann V, Geiger T, Alkan S, Heusser Interferon alpha increases the frequency of IFN gamma-producing CD4 + T cells. (1993) J Exp Med 178: 1655-1663) via the TCR / CD3 complex, the proportion of IFN gamma producing cells (Th1 type cells) increased.
- IFN alpha to human umbilical cord leukocytes stimulated under Th2 inducing conditions (IL-4 and anti-IL-12 monoclonal antibody) resulted in the development of IFN gamma-producing Th1 cells (Rogge L, Barberis- Maino L, Biffi M, Passini N, Presky DH, Gubler U, Sinigaglia F. Selective expression of an interleukin-12 receptor component by human T helper 1 cells. (1997) J Exp Med 185: 825-832).
- Type I IFNs act directly on virgin cells inducing Th1 differentiation of CD45RA + cells stimulated with anti-CD3 (Rogge L 1 D'Ambrosio D 1 Biffi M 1 Penna G, Minetti LJ, Presky DH, Adorini L, Sinigaglia F. The role of stat4 in species-specific regulation of Th cell development by type I IFNs. (1998) J Immunol 161: 12: 6567-6574). There is evidence in vivo that the administration of type I IFNs can favor the development of human Th 1 cells. IFN gamma producing cells of patients with MS increase in number when patients are treated with IFN beta (Dayal A, Jensen MA, Lledo A, Arnason BGW.
- Th2 cytokines such as hlpereosinophilic syndrome (Zielinskl RM, Lawrence WD. Interferon alpha for the hyper-eosinophilic syndrome. (1990) Ann Int Med 113: 716-718; Butterfield JH, Gleich GJ. Interferon alpha treatment of 6
- IFN alpha induces activation of Stat4 and therefore Th1 response (Cho SS, Bacon CM 1 Sudarshan C, Rees RC, Finbloom D, Pine R, O'Shea JJ. Activation of Stat4 by IL-12 and IFN alpha. Evidence for the involvement of ligand-induced tyrosine and serine phosphorilation. (1996) J Immunol 157: 4781-4789).
- IFN alpha has been shown to induce gamma IFN.
- IFN gamma knockout mice an increase in inflammation and demyelination has been observed (Tran EH, Prince EN, Owens T. IFN-gamma shapes immune invasion of the central nervous system via regulation of chemokines. (2000) J Immunol 164 : 2759-2768) suggesting that an independent IFN gamma pathway is the one that mediates the destruction of myelin. IFN gamma prevents the accumulation of neutrophils in the CNS of patients with NMO.
- EPO Erythropoietin
- EPO has been associated with an immunomodulatory effect by demonstrating that it delays the increase in TNF-alpha levels and decreases IL-6 levels in an EAE model in Lewis rats (Agnello D., Bigini P, Villa P, Mennini T, Cerami A, Brines ML, Ghezzi P (2002) Brain Research 952, 128-134). EPO directly affects neuronal excitability in this way can also limit neuronal necrosis (Brines, ML, Ghezzi, P., Keenan, S., Agnello, D., de Lanerolle, N. C 1 Cerami, C, Itri, LM & Cerami, A. (2000) Proc. Nati Acad. Sci. USA 97, 10526-10531).
- the present invention describes the use of interferon alfa for obtaining a pharmaceutical compound that contains more specifically, recombinant interferon alfa 2b, useful in the treatment of patients with NMO, a disease for which an effective treatment has not been described so far.
- said compound was used at therapeutic doses of 3 and 10 million Ul, twice a week.
- the immunomodulatory, antiviral, remyelinating and inducing properties of regulatory T cells of the pharmaceutical compound of the present invention justify its effectiveness in the NMO.
- IFN alpha induces gamma IFN, which is corroborated in another embodiment of our invention, but the demyelination observed in the NMO does not involve gamma IFN, suggesting the participation of an independent path to gamma IFN, in fact,
- Our invention is the fact that it is a double-blind, randomized and placebo-controlled clinical trial where it is demonstrated that IFN alfa2brec achieves the reduction of outbreaks and the frequency of relapses in patients with recurrent NMO.
- the invention also includes pharmaceutical compositions comprising the combination with Erythropoietin (EPO rec).
- EPO rec Erythropoietin
- the combination of IFN alfa2brec with EPO rea showed a decrease in the degree of cerebral atrophy, as well as positively modified molecular markers involved in the pathogenesis of the disease.
- the combination showed a synergistic effect between IFN alfa2brec and EPO rec.
- the pharmaceutical compound referred to in our invention for the treatment of NMO is characterized in that it contains interferon alfa that can be modified by peguylation or fusion to other proteins while maintaining the activities referred to above.
- interferon alfa 2b or of the combination IFN alfa with EPO requires repeated administrations.
- both the pharmaceutical compound containing IFN alpha alone or modified by peeling, independently or as part of pharmaceutical compositions that combine it with Growth Factors such as Erythropoietin could be administered intramuscularly, intravenously , subcutaneous, oral, nasal and intrathecal.
- Example 1 Characteristics of the compound.
- PEG-IFN Polyethylene Glycol
- Each vial contains recombinant Interferon alfa 2b in a range between 3 and 10 x10 6 Ul, 10 mg benzyl alcohol, 0.2mg polysorbate 80, 4.67mg NaCL, 2.96mg NaH 2 PO 4 ⁇ H 2 O, 14.41 mg Na 2 HPO 4 .2H 2 O and water for injection to complete 1 mL
- Each vial contains recombinant PEG-I interferon alfa 2b in a range between 180 and 360 ⁇ g, 2,617 mg tri-hydrated sodium acetate, 8 mg NaCL, 10 mg benzyl alcohol, 0.05 mg polysorbate and water for injection at complete 1 mL.
- Each vial contains recombinant Interferon alfa 2b in a range between 3 and 10 x10 6 Ul, recombinant Erythropoietin 2000 Ul, 10 mg benzyl alcohol, 0.2mg polysorbate 80, 4.67mg NaCL, 2.96mg NaH 2 PO 4 .2H 2 O, 14.41 mg Na 2 HPO 4 .2H 2 O and water for injection to complete 1 mL
- Example 2 Characteristics of the sample evaluated. Evaluation of the therapeutic effect of IFN ⁇ 2b rec in NMO.
- Example 3 Evaluation of the evolution of the outbreaks.
- Example 4 Characteristics of the sample evaluated. Evaluation of the therapeutic effect of the IFN ⁇ 2brec / EPO combination in NMO.
- the 3 patients in the first group received IFN ⁇ 2brec 3 million IM twice a week, the 3 in the second group received EPO rec SC 500U / kg twice a week and 3 in the third group, both treatments combined at the doses referred to above for a period of 1 year.
- the clinical evaluation was performed at 6 months and 1 year of NMR treatment to measure the evolution of cerebral atrophy.
- RNA and protein that included: Determination by RT-PCR in mononuclear cells isolated from peripheral blood of the gene expression for the CCR3 chemokine receptor, cationic protein of eosinophils, CD25, Foxp3, IFN gamma, IL-10 and TGF-beta and determination of serum concentrations of brain-derived neurotrophic factor (BDNF) and ciliary neurotrophic factor (CNTF) by ELISA.
- BDNF brain-derived neurotrophic factor
- CNTF ciliary neurotrophic factor
- Table 4 shows the characteristics of the patients evaluated.
- the sample was homogeneous in terms of age, sex and race. The majority of the patients were female and the black race, which is consistent with the literature reports for this disease. Table 4. Homogeneity of the sample studied.
- Example 5 Effect of the treatment with the IFN ⁇ 2brec / EPO combination on cerebral atrophy.
- T1 measurements of the degree of cerebral atrophy were performed by NMR at 6 months and 1 year of treatment. The results are reflected as the% of patients in whom there was a decrease in cerebral atrophy at the time of the evaluation with respect to the onset.
- Table 5 shows the result of the evaluation of cerebral atrophy in patients treated with IFN ⁇ 2brec, EPO rec and the IFN ⁇ 2brec / EPO combination.
- the IFN ⁇ 2brec / EPO combination reduced the degree of cerebral atrophy in 66.6% of patients a year of treatment, demonstrating a greater effect than with independent treatments.
- Example 6 Modulation of genes involved in the pathogenesis of the NMO.
- the evaluation by RT-PCR of the modulation of different genes involved in the pathogenesis of the NMO was performed one year after treatment with IFN ⁇ 2brec, EPO rec and the IFN ⁇ 2brec / EPO combination in the patients.
- the results are reflected as% of patients in whom the relative levels of the evaluated gene increase / decrease with respect to the expression of a constitutive gene.
- Table 6 shows the results where it is valid to highlight that in 100% of the patients treated with the IFN ⁇ 2brec / EPO combination, the expression of CCR3, a chemokine receptor that is expressed exclusively in eosinophils, decreased in accordance with this result also it was found that in the majority of patients treated with the combination the expression of the gene corresponding to the cationic eosinophilic protein decreased, which seems to indicate that the IFN ⁇ 2brec / EPO combination can have an effect both on the decrease of the characteristic eosinophilic infiltrate of this disease as in the decrease of the degranulation of these cells where one of the fundamental components of these granules is precisely the cationic protein of eosinophils.
- the treatment with the combination caused an increase in the expression of the IL-10 and TGF-beta gene and may indicate the participation of a mechanism of induced regulatory T cells.
- a significant fact is that 100% of the patients treated with the combination have an increase in the expression of CD25 and the Foxp3 transcriptional factor, which may indicate the participation of natural regulatory cells.
- Another relevant data obtained constitutes that 66.6% of the patients present an increase in the levels of IFN gamma, cytokine TH 1 which is beneficial in this disease that when characterized by a TH2 predominance, a polarization of the response to TH 1 would result beneficial
- Example 7 Evaluation of anti-apoptotic (BDNF) and remyelinating (CNTF) factors in patients with NMO treated with independent treatments and the IFN ⁇ 2brec / EPO combination.
- BDNF anti-apoptotic
- CNTF remyelinating
- ELISA measurements of anti-apoptotic (BDNF) and remyelinating (CNTF) factors were performed in the serum of patients with NMO at one year of treatment with IFN ⁇ 2brec, EPO rec and the IFN ⁇ 2brec / EPO combination.
- the results are expressed as% of patients in whom the serum concentrations of both factors increased / decreased at one year of treatment with respect to the values before starting treatment.
- the results are reflected in Table 7, observing how for both factors an increase in the concentrations of these factors is observed with the treatment with the combination that speaks in favor of the anti-apoptotic and remyelinating effect of this molecule.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Veterinary Medicine (AREA)
- General Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Public Health (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Gastroenterology & Hepatology (AREA)
- Immunology (AREA)
- Zoology (AREA)
- Ophthalmology & Optometry (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The invention relates to the use of interferon-alpha in order to obtain a pharmaceutical compound for the treatment of neuromyelitis optica (NMO) or Devic's syndrome. Interferon-alpha (IFN α) can be used as a treatment for NMO in both the monophase form and the relapsing form. In addition, IFN α can be combined with other growth factors such as recombinant erythropoietin (rEPO), thereby increasing the rationale behind its use in relation to said disease.
Description
USO DEL INTERFERÓN ALFA PARA LA OBTENCIÓN DE UN COMPUESTO PARA EL TRATAMIENTO DE LA NEUROMIELITIS ÓPTICA.USE OF THE ALFA INTERFERON TO OBTAIN A COMPOUND FOR THE TREATMENT OF OPTICAL NEUROMYELITIS.
Campo de Ia Técnica La presente invención se relaciona con las ciencias biológicas, Ia biotecnología y las ciencias médicas, y se basa en un nuevo uso del interferón alfa (IFN α) que por sus efectos antivirales, inmunomoduladores, inductor de células T reguladoras y preventivo de Ia desmielinización es capaz de detener Ia progresión de Ia enfermedad neuromielitis óptica (NMO) en Ia forma monofásica, disminuir el número de brotes y prolongar el tiempo entre las recaídas en pacientes que padecen Ia forma recurrente de NMO.Field of the Technique The present invention relates to the biological sciences, biotechnology and medical sciences, and is based on a new use of interferon alpha (IFN α) which due to its antiviral, immunomodulatory, regulatory and preventive T-cell inductors demyelination is able to stop the progression of optic neuromyelitis disease (NMO) in the single phase form, reduce the number of outbreaks and prolong the time between relapses in patients suffering from the recurrent form of NMO.
Arte previoPrior art
La NMO está caracterizada por ataques de neuritis óptica y de mielitis (Devic E. Myelitis subsigue compliquess de neurite optique. (1894) BuII Med 8: 1033- 1034; Devic E. Myelite aigue dorse- lombaire de neurite optique, autopsie. (1895). Congreso Francais Medicine (Premiere Session, Lyon) 1 : 434- 9; Gault F. De Ia neuromyelite optique aigue. (thesis) Lyon; (1894)). Estos eventos clínicos pueden también ocurrir frecuentemente en Ia Esclerosis Múltiple (EM), sin embargo, tanto Ia epidemiología, características clínicas, de laboratorio, etiopatogenia y Ia no respuesta a los tratamientos convencionales aceptados para Ia EM, permiten considerar a Ia NMO como una entidad nosológica independiente a Ia EM. En Ia NMO, los brotes o crisis son más agudos (a veces fulminante) y severos, afecta fundamentalmente a adultos jóvenes y ha sido reportada en Ia infancia. La edad media reportada de comienzo es más tardía que en Ia EM, entre 35 y 47 años (Mandler RN, Davis LE, Jeffery DR, Komfeld M. Devic's neuromyelitis óptica: a clinicopathological study of 8 patients. (1993) Ann Neurol 34: 162- 8), afecta fundamentalmente al sexo femenino y razas no blancas a diferencia de Ia EM que predomina en Ia raza blanca (Weinshenker BG, Sibley WA, Natural history and treatment of múltiple sclerosis. Curr Opinión neurol Neurosurg 1992, 5.203-211). La incidencia o prevalencia de NMO se desconoce. En Europa, donde Ia EM tiene una prevalencia entre 100-200 / 100 000 habitantes, Ia NMO se considera como una enfermedad rara. La NMO parece ser común en no Caucasianos tales como Africanos, Americanos,
Japoneses y otras islas del Pacífico. Más del 6% de los casos de enfermedades desmielinizantes en Ia India son NMO (Singhal BS. Múltiple sclerosis-lndian experience. (1985) Annals of the Academy of Medicine, Singapore 14: 32- 6). En Cuba Ia prevalencia se desconoce pero ha ido aumentando en los últimos años. El papel de los factores genéticos se desconoce. Existen reportes de gemelos idénticos con NMO. Ciertos alelos de antígenos leucocitarios humanos (HLA) están asociados a Ia NMO como es el HLA-DPB1*0501 mientras que en Ia EM el alelo HLA que con mayor frecuencia está asociado es el HLADR2B. En Ia NMO las mediciones paraclínicas tales como Resonancia Magnético Nuclear (RMN) de cerebro y médula espinal, así como el análisis del líquido cefalorraquídeo (LCR) también difieren con Ia EM. En NMO ninguna o pocas lesiones en Ia materia blanca se detectan en el cerebro, sin embargo las imágenes de Ia médula espinal muestran elementos distintivos: Ia mayoría de los pacientes tienen extensivas lesiones que se extienden longitudinalmente sobre 3 o más segmentos vertebrales. Además, los pacientes con NMO tienen una pleocitosis de más de 50 leucocitos con o sin presencia de neutrófilos. La NMO tiende a seguir un curso monofásico o por brotes o recaídas (más del 70% de los casos tienen un curso recidivante) (Wingerchuk DM, Hogancamp WF, O'Brien PC, Weinshenker BG. The clinical course of neuromyelitis óptica (Devic's syndrome). (1999) Neurology 53: 1107- 14). En NMO monofásica, Los pacientes experimentan una neuritis óptica (NO) unilateral o bilateral con un simple episodio de mielitis, típicamente, aunque no siempre dentro de un corto período de tiempo uno del otro, pero no tiene recaídas posteriores a diferencia de los pacientes con un curso recidivante que continúan a tener discretas exacerbaciones de NO y/o mielitis.NMO is characterized by attacks of optic neuritis and myelitis (Devic E. Myelitis follows complications of neurite optique. (1894) BuII Med 8: 1033-1034; Devic E. Myelite aigue dorse-lombaire de neurite optique, autopsy. (1895 ) Congress Francais Medicine (Premiere Session, Lyon) 1: 434-9; Gault F. De la neuromyelite optique aigue. (Thesis) Lyon; (1894)). These clinical events can also occur frequently in Multiple Sclerosis (MS), however, both the epidemiology, clinical characteristics, laboratory, etiopathogenesis and non-response to the accepted conventional treatments for MS, allow to consider the NMO as an entity nosological independent of MS. In the NMO, outbreaks or crises are more acute (sometimes fulminant) and severe, mainly affects young adults and has been reported in childhood. The reported mean age of onset is later than in MS, between 35 and 47 years (Mandler RN, Davis LE, Jeffery DR, Komfeld M. Devic's optic neuromyelitis: a clinicopathological study of 8 patients. (1993) Ann Neurol 34: 162-8), fundamentally affects the female sex and non-white races unlike the MS that predominates in the white race (Weinshenker BG, Sibley WA, Natural history and treatment of multiple sclerosis. Curr Neurol Opinion Neurosurg 1992, 5,203-211) . The incidence or prevalence of NMO is unknown. In Europe, where MS has a prevalence between 100-200 / 100,000 inhabitants, NMO is considered a rare disease. The NMO seems to be common in non-Caucasians such as Africans, Americans, Japanese and other Pacific islands. More than 6% of cases of demyelinating diseases in India are NMO (Singhal BS. Multiple sclerosis-Indian experience. (1985) Annals of the Academy of Medicine, Singapore 14: 32-6). In Cuba the prevalence is unknown but has been increasing in recent years. The role of genetic factors is unknown. There are reports of identical twins with NMO. Certain alleles of human leukocyte antigens (HLA) are associated with the NMO such as the HLA-DPB1 * 0501 while in the MS the HLA allele that is most frequently associated is the HLADR2B. In the NMO the paraclinical measurements such as Nuclear Magnetic Resonance (NMR) of the brain and spinal cord, as well as the analysis of the cerebrospinal fluid (CSF) also differ with MS. In NMO, no or few lesions in the white matter are detected in the brain, however the spinal cord images show distinctive elements: the majority of patients have extensive lesions that extend longitudinally over 3 or more vertebral segments. In addition, patients with NMO have a pleocytosis of more than 50 leukocytes with or without the presence of neutrophils. The NMO tends to follow a single-phase or outbreak or relapse course (more than 70% of cases have a recurrent course) (Wingerchuk DM, Hogancamp WF, O'Brien PC, Weinshenker BG. The clinical course of optic neuromyelitis (Devic's syndrome ). (1999) Neurology 53: 1107-14). In monophasic NMO, patients experience unilateral or bilateral optic neuritis (NO) with a simple episode of myelitis, typically, although not always within a short period of time from each other, but do not have subsequent relapses unlike patients with a relapsing course that continue to have discrete exacerbations of NO and / or myelitis.
La causa de Ia NMO se desconoce. De hecho las enfermedades autoinmunes sistémicas se han asociado con NMO sugiriendo que una única causa de esta enfermedad es poco probable. En Ia patogenia de Ia EM Ia respuesta inmune celular tiene un papel relevante, a diferencia de Ia NMO donde el componente humoral parece ser predominante.The cause of the NMO is unknown. In fact, systemic autoimmune diseases have been associated with NMO suggesting that a single cause of this disease is unlikely. In the pathogenesis of MS, the cellular immune response has an important role, unlike the NMO where the humoral component seems to be predominant.
Diferentes mecanismos se han involucrado en Ia patogénesis de Ia NMO. La pronunciada reactividad a lnmunoglobulinas (Ig) co-localizándose con activación del complemento en sitios de daño vascular sugiere que el espacio peri-vascular
puede ser el sitio primario del daño en Ia NMO. Esto puede ser debido a anticuerpos específicos a antígenos vasculares.Different mechanisms have been involved in the pathogenesis of NMO. The pronounced reactivity to lnmunoglobulins (Ig) co-localizing with complement activation at sites of vascular damage suggests that the peri-vascular space It may be the primary site of damage in the NMO. This may be due to antibodies specific to vascular antigens.
Alternativamente, antígenos liberados dentro del SNC durante el proceso destructivo pueden llegar al espacio peri-vascular y ser reconocidos allí por anticuerpos provenientes de Ia circulación. Finalmente, puede estar involucrada una reacción inflamatoria no específica iniciada por Ia deposición de complejos inmunes circulantes. En este escenario, Ia vía clásica del complemento se activa, permitiendo el reclutamiento de macrófagos activados a estos sitios peri-vasculares donde ellos se unen vía un receptor para componentes del complemento o Ig/receptores Fc. Los macrófagos activados, conjuntamente con eosinófilos y neutrófilos, localmente generan citoquinas, proteasas y radicales libres del oxígeno/nitrógeno los cuales pueden contribuir tanto al daño vascular como al parenquimatoso, resultando en una destrucción no selectiva de Ia materia blanca y gris, incluyendo axones y oligodendrocitos. La permeabilidad vascular incrementada y el edema pueden contribuir al daño parenquimatoso vía isquemia secundaria o favorecer Ia localización central típica de las placas de NMO dentro de Ia médula espinal (Prineas JW, McDonald Wl. Demyelinating diseases (1997) In: Graham Dl, Lantos PL, editors. Greenfield's neuropathology. 6th ed. London: Arnold; p. 813- 96). Una similar localización central fue encontrada en casos severos de Encefalomielitis autoinmune experimental (EAE) inducida por Ia Proteína mielínica del oligodendrocito (MOG) y este fenómeno es probablemente debido a Ia isquemia inducida por el edema. (Lassmann H. Comparative neuropathology of chronic experimental allergic encephalomyelitis and múltiple sclerosis. (1983) Schriftenr Neurol 25: 1- 135). Nuevos antígenos liberados durante el proceso destructivo pueden amplificar Ia respuesta inmune destructiva in NMO. El complemento puede ser no específicamente activado en respuesta a necrosis tisular. Uno de las características más novedosas que se describen en Ia histopatología de las lesiones activas de NMO es Ia intensidad de Ia infiltración meníngea y perivascular de Ia médula espinal con eosinófilos y neutrófilos. Los eosinófilos activados liberan proteínas con granulos básicos tales como Proteína Básica de Ia Mielina (MBP), neurotoxina derivada de eosinófilos, proteína catiónica de
eosinófilos y peroxidasa de eosinófilos (Kaneko M, Kita H, Gleich GJ. Eosinophil basic proteins. In: Barnes PJ, Grunstein MM, Leff AR1 Woolcock AJ, editors. Asthma. (1997) Philadelphia: Lippincott-Raven p 593-607). Estos granulos tienen propiedades citotóxicas y sirven como marcadores de activación de eosinófilos (Venge P. Monitoring of asthma inflammation by serum measurements of eosinophil cationic protein (ECP): a new clinical approach to asthma management. (1995) Resp Med 89: 1- 2). Además del número de eosinófilos elevados en las lesiones de NMO, se ha confirmado Ia presencia de degranulación de eosinófilos en el tejido de Ia médula espinal de pacientes con NMO. Se han demostrado evidencias de expresión de CCR3 en lesiones de NMO. CCR3 es el principal receptor para Ia quimiocina eotaxina, un potente quimio-atrayente de eosinófilos. CCR3 es expresado primariamente en eosinófilos humanos (Ponath PD, Qin S, Post TW, Wang J, Wu L, Gerard NP, et al. Molecular cloning and characterization of human eotaxin receptor expressed selectively on eosinophils. (1996) J Exp. Med 183: 2437- 48). La señalización de eotaxina a través del CCR3 es un importante índice de reclutamiento de eosinófilos (Daugherty BL, Siciliano SJ, DeMartino JA, Malkowitz I, Sirotina A, Springer MS. Cloning, expression, and characterization of the human eosinophil eotaxin receptor. (1996) J Exp Med 183: 2349- 54). CCR3 es selectivamente expresado en células T helper 2 (Th2) y por tanto está asociado con Ia respuesta Th2 (Sallusto F, Lenig D, Mackay CR, Lanzavecchia A. Flexible programs of chemokines receptors expression on human polarized T helper 1 and 2 lymphocytes. (1998) J Exp Med 187: 875- 83). Existen evidencias morfológicas de que están aumentados en número y son funcionalmente activos y probablemente contribuyan al proceso inflamatorio destructivo en NMO. La citotoxicidad de las proteínas de los granulos de eosinófilos ha sido bien establecida (Corrigan CJ, Kay AB. T cell/eosinophils interactions in the induction of asthma. (Review) (1996) Eur Respr J Suppl 22: 72s-8s). Los eosinófilos son una fuente importante de IL-4, Ia cual puede causar un cambio en el perfil de citoquinas de Th 1 a Th2 (Rumbley CA, Sugaya H, Zekavat SA, El Refaei M, Perrin PJ, Phillips SM. Activated eosinophils are the major source of Th2- associated cytokines in the schistosome granuloma. (1999) J Immunol 162: 1003- 9).
Los pacientes con NMO a menudo tienen auto-anticuerpos circulantes con una frecuencia que excede a aquella vista en Ia EM (Wingerchuk DM, Hogancamp WF, O'Brien PC, Weinshenker BG. The clinical course of neuromyelitis óptica (Devic's syndrome). (1999) Neurology 53: 1107- 14). La patogenicidad de estos auto-anticuerpos se desconoce: ellos pueden causar daño directamente a través del reconocimiento de epítopes sobre células normales, o indirectamente a través de Ia formación de complejos inmunes que se depositan en el tejido normal y activan Ia cascada del complemento. Su presencia en Devic pueden también reflejar el mayor amplio espectro de Ia respuesta a células B. Prominente respuesta a anticuerpos puede también encontrarse con respecto a antígenos de Ia mielina endógenos, tales como el MOG. Un reciente estudio analizó Ia respuesta a anticuerpos a MOG, MBP y Proteína unidora de Calcio de Ia astroglia (SIOOb) en el suero de 4 casos de Enfermedad de Devic (Haase CG, Schmidt S. Detection of brain-specific autoantibodies to myelin oligodendrocyte glycoprotein, SiOObeta and myelin basic protein in patients with Devic's neuromyelitis óptica. (2001) Neurosci Lett 307: 131-3). Los autores reportaron una pronunciada respuesta antiMOG, específicamente al epítope 63- 87 de Ia secuencia de aminoácidos en todos los pacientes, anticuerpos antiMBP en dos pacientes y anticuerpos antiSIOOb en un paciente. En Ia NMO, Ia médula espinal y el nervio óptico son preferencialmente afectados. Es posible, aunque poco probable que estos sitios tengan un antígeno vascular o del SNC restringido o quizás pueda ser que ambos sitios tengan una vulnerabilidad particular al daño mediado por anticuerpos debido a Ia inherente debilidad de Ia barrera hemato-encefálica en estos sitios. La BHE es altamente impermeable a proteínas plasmáticas y leucocitos circulantes y por tanto puede proteger al SNC de una reacción inmunológica. Sin embargo, en algunas áreas donde Ia BHE no es efectiva, tales como las raíces de los nervios espinales, puede suceder que anticuerpos patogénicos circulantes puedan acceder al SNC vía estas estructuras y difundir a Ia vecindad inmediata. En los modelos de EAE las lesiones activas afectan predominantemente Ia médula espinal (Lassmann H. Comparative neuropathology of chronic experimental allergic encephalomyelitis and múltiple sclerosis. (1983) Schriftenr Neurol 25: 1- 135). La NO en EAE también tiende predominantemente a ocurrir en el nervio óptico retrobulbar (Guy J, Rao NA.
Acute and chronic experimental optic neuritis.Altaration ¡n the blood-optic nerve barrier. (1984) Arch Ophthalmol 102: 450- 4). Las lesiones en estos dos sitios son un reflejo del más alto grado de permeabilidad de Ia BHE en estas regiones comparado con el cerebro (Guy J1 Rao NA. Acute and chronic experimental optic neuritis.Altaration in the blood-optic nerve barrier. (1984) Arch Ophthalmol 102: 450- 4; Rao NA. Chronic experimental allergic optic neuritis. (1981) Invest Ophthalmol Vis Sci 20: 159- 72; Butter C, Baker D, O'Neill JK1 Turk JL. Mononuclear cell trafficking and plasma protein extravasation into the CNS during chronic relapsing experimental allergic encephalomyelitis in Biozzi AB/H mice. (1991) J Neurol Sci 104: 9- 12). La permeabilidad de Ia BHE incrementada en Ia médula espinal puede ser debido a propiedades vasculares inherentes a esta región donde los capilares son más largos que en el cerebro. Por tanto, con un proceso inflamatorio de fondo, en presencia de títulos extremadamente elevados de anticuerpos, las lesiones pueden preferencialmente pero no exclusivamente, afectar Ia médula espinal y el nervio óptico. Esta hipótesis es compatible con Ia observación que en estadios tardíos de NMO, las lesiones a menudo se diseminan a otras regiones del SNC (Wingerchuk DM, Hogancamp WF1 O'Brien PC, Weinshenker BG. The clinical course of neuromyelitis óptica (Devic's syndrome) (1999) Neurology 53: 1107- 14). Existe desmielinización extensa a nivel de médula espinal y estuvo asociada con cavitación, necrosis y patología axonal aguda (esferoides) tanto en Ia materia gris como en Ia materia blanca. Existe pérdida pronunciada de los oligodendrocitos dentro de las lesiones. El infiltrado inflamatorio dentro de las lesiones activas se caracterizó por pronunciada infiltración de macrófagos asociado con gran número de granulocitos y eosinófilos y raramente células T CD3+ CD8+. Existe una marcada deposición perivascular de inmunoglobulinas (principalmente IgM) y el antígeno C9neo de complemento en lesiones activas asociadas a prominente fibrosis vascular e hialinización tanto en lesiones activas como inactivas. La importante activación del complemento, infiltración eosinofílica y fibrosis vascular observada en los casos de NMO de Devic es más prominente comparada con Ia EM, y apoya el papel de Ia inmunidad humoral en Ia patogénesis de Ia NMO (Lucchinetti CF, Mandler RN, McGavem D, Bruck W, Gleich G, Ransohoff RM1 Trebst C1 Weinshenker B, Wingerchuk D, Parisi JE,
Lassman H. (2002) A role for humoral mechanism in the pathogenesis of Devic's neuromyelitis óptica. Brain 125: 1450- 61).Alternatively, antigens released into the CNS during the destructive process can reach the peri-vascular space and be recognized there by antibodies from the circulation. Finally, a non-specific inflammatory reaction initiated by the deposition of circulating immune complexes may be involved. In this scenario, the classical complement pathway is activated, allowing the recruitment of activated macrophages to these peri-vascular sites where they bind via a receptor for complement components or Ig / Fc receptors. Activated macrophages, together with eosinophils and neutrophils, locally generate cytokines, proteases and free radicals of oxygen / nitrogen which can contribute to both vascular and parenchymal damage, resulting in non-selective destruction of white and gray matter, including axons and oligodendrocytes Increased vascular permeability and edema can contribute to parenchymal damage via secondary ischemia or favor the typical central location of NMO plaques within the spinal cord (Prineas JW, McDonald Wl. Demyelinating diseases (1997) In: Graham Dl, Lantos PL , editors, Greenfield's neuropathology, 6 th ed. London: Arnold; p. 813-96). A similar central location was found in severe cases of experimental autoimmune encephalomyelitis (EAE) induced by myelinic oligodendrocyte protein (MOG) and this phenomenon is probably due to edema induced ischemia. (Lassmann H. Comparative neuropathology of chronic experimental allergic encephalomyelitis and multiple sclerosis. (1983) Schriftenr Neurol 25: 1- 135). New antigens released during the destructive process can amplify the destructive immune response in NMO. The complement may not be specifically activated in response to tissue necrosis. One of the most novel features described in the histopathology of active NMO lesions is the intensity of the meningeal and perivascular infiltration of the spinal cord with eosinophils and neutrophils. Activated eosinophils release proteins with basic granules such as Myelin Basic Protein (MBP), eosinophil-derived neurotoxin, cationic protein from eosinophils and eosinophil peroxidase (Kaneko M, Kita H, Gleich GJ. Eosinophil basic proteins. In: Barnes PJ, Grunstein MM, Leff AR 1 Woolcock AJ, editors. Asthma. (1997) Philadelphia: Lippincott-Raven p 593-607) . These granules have cytotoxic properties and serve as eosinophil activation markers (Venge P. Monitoring of asthma inflammation by serum measurements of eosinophil cationic protein (ECP): a new clinical approach to asthma management. (1995) Resp Med 89: 1- 2 ). In addition to the high number of eosinophils in NMO lesions, the presence of eosinophil degranulation in the spinal cord tissue of patients with NMO has been confirmed. Evidence of CCR3 expression has been demonstrated in NMO lesions. CCR3 is the main receptor for chemokine eotaxin, a potent chemo-attractant for eosinophils. CCR3 is expressed primarily in human eosinophils (Ponath PD, Qin S, Post TW, Wang J, Wu L, Gerard NP, et al. Molecular cloning and characterization of human eotaxin receptor expressed selectively on eosinophils. (1996) J Exp. Med 183 : 2437-48). Eotaxin signaling through CCR3 is an important index of eosinophil recruitment (Daugherty BL, Siciliano SJ, DeMartino JA, Malkowitz I, Sirotina A, Springer MS. Cloning, expression, and characterization of the human eosinophil eotaxin receptor. (1996 ) J Exp Med 183: 2349-54). CCR3 is selectively expressed in T helper 2 (Th2) cells and is therefore associated with the Th2 response (Sallusto F, Lenig D, Mackay CR, Lanzavecchia A. Flexible programs of chemokines receptors expression on human polarized T helper 1 and 2 lymphocytes. (1998) J Exp Med 187: 875-83). There is morphological evidence that they are increased in number and are functionally active and probably contribute to the destructive inflammatory process in NMO. The cytotoxicity of eosinophil granule proteins has been well established (Corrigan CJ, Kay AB. T cell / eosinophils interactions in the induction of asthma. (Review) (1996) Eur Respr J Suppl 22: 72s-8s). Eosinophils are an important source of IL-4, which can cause a change in the cytokine profile from Th 1 to Th2 (Rumbley CA, Sugaya H, Zekavat SA, El Refaei M, Perrin PJ, Phillips SM. Activated eosinophils are the major source of Th2- associated cytokines in the schistosome granuloma. (1999) J Immunol 162: 1003-9). Patients with NMO often have circulating auto-antibodies with a frequency that exceeds that view in MS (Wingerchuk DM, Hogancamp WF, O'Brien PC, Weinshenker BG. The clinical course of optic neuromyelitis (Devic's syndrome). (1999 ) Neurology 53: 1107-14). The pathogenicity of these autoantibodies is unknown: they can cause damage directly through the recognition of epitopes on normal cells, or indirectly through the formation of immune complexes that are deposited in normal tissue and activate the complement cascade. Their presence in Devic may also reflect the greater spectrum of the response to B cells. Prominent antibody response can also be found with respect to endogenous myelin antigens, such as MOG. A recent study analyzed the response to antibodies to MOG, MBP and Calcium-binding protein of astroglia (SIOOb) in the serum of 4 cases of Devic's Disease (Haase CG, Schmidt S. Detection of brain-specific autoantibodies to myelin oligodendrocyte glycoprotein , SiOObeta and myelin basic protein in patients with Devic's optic neuromyelitis. (2001) Neurosci Lett 307: 131-3). The authors reported a pronounced antiMOG response, specifically to epitope 63-87 of the amino acid sequence in all patients, antiMBP antibodies in two patients and antiSIOOb antibodies in one patient. In the NMO, the spinal cord and optic nerve are preferentially affected. It is possible, although unlikely that these sites have a restricted vascular or CNS antigen or it may be that both sites have a particular vulnerability to antibody-mediated damage due to the inherent weakness of the blood-brain barrier at these sites. BHE is highly impermeable to circulating plasma proteins and leukocytes and therefore can protect the CNS from an immune reaction. However, in some areas where BHE is not effective, such as spinal nerve roots, it may happen that circulating pathogenic antibodies can access the CNS via these structures and spread to the immediate vicinity. In EAE models, active lesions predominantly affect the spinal cord (Lassmann H. Comparative neuropathology of chronic experimental allergic encephalomyelitis and multiple sclerosis. (1983) Schriftenr Neurol 25: 1- 135). NO in EAE also tends predominantly to occur in the retrobulbar optic nerve (Guy J, Rao NA. Acute and chronic experimental optic neuritis. Alteration ¡n the blood-optic nerve barrier. (1984) Arch Ophthalmol 102: 450-4). The lesions in these two sites are a reflection of the highest degree of permeability of BHE in these regions compared to the brain (Guy J 1 Rao NA. Acute and chronic experimental optic neuritis. Alteration in the blood-optic nerve barrier. (1984 ) Arch Ophthalmol 102: 450-4; Rao NA. Chronic experimental allergic optic neuritis. (1981) Invest Ophthalmol Vis Sci 20: 159-72; Butter C, Baker D, O'Neill JK 1 Turk JL. Mononuclear cell trafficking and plasma protein extravasation into the CNS during chronic relapsing experimental allergic encephalomyelitis in Biozzi AB / H mice. (1991) J Neurol Sci 104: 9-12). The permeability of the increased BHE in the spinal cord may be due to vascular properties inherent in this region where the capillaries are longer than in the brain. Therefore, with a background inflammatory process, in the presence of extremely high antibody titres, the lesions may preferentially but not exclusively affect the spinal cord and the optic nerve. This hypothesis is compatible with the observation that in late stages of NMO, lesions often spread to other CNS regions (Wingerchuk DM, Hogancamp WF 1 O'Brien PC, Weinshenker BG. The clinical course of optic neuromyelitis (Devic's syndrome) (1999) Neurology 53: 1107-14). There is extensive demyelination at the level of the spinal cord and it was associated with cavitation, necrosis and acute axonal pathology (spheroids) in both gray matter and white matter. There is pronounced loss of oligodendrocytes within the lesions. Inflammatory infiltrate within active lesions was characterized by pronounced macrophage infiltration associated with large numbers of granulocytes and eosinophils and rarely CD3 + CD8 + T cells. There is a marked perivascular deposition of immunoglobulins (mainly IgM) and the complement C9neo antigen in active lesions associated with prominent vascular fibrosis and hyalinization in both active and inactive lesions. The important activation of complement, eosinophilic infiltration and vascular fibrosis observed in cases of Devic NMO is more prominent compared to MS, and supports the role of humoral immunity in the pathogenesis of NMO (Lucchinetti CF, Mandler RN, McGavem D , Bruck W, Gleich G, Ransohoff RM 1 Trebst C 1 Weinshenker B, Wingerchuk D, Parisi JE, Lassman H. (2002) A role for humoral mechanism in the pathogenesis of Devic's optic neuromyelitis. Brain 125: 1450-61).
Diferentes asociaciones clínicas con NMO sugieren que esta enfermedad es diferente a Ia EM. A diferencia de Ia EM1 existen numerosos reportes de enfermedad óptico-espinal asociada con enfermedades del tejido conectivo y otras enfermedades autoinmunes (April RS, Vansonnenberg E. A case of neuromyelitis óptica (Devic's syndrome) in systemic lupus erythematosus: clinico-pathologic report and review of the literature (1976) Neurology 26: 1066- 70; Kinney EL, Berdoff RL1 Rao NS, Fox LM. Devic's syndrome and systemic lupus erythematosus: a case report with necropsy. (1979) Arch Neurol 36: 643- 4; Goldman M, Herode A, Borenstein S, Zanen A. Optic neuritis, transverse myelitis, and anti-DNA antibodies nine years after thymectomy for myasthenia gravis. (1984) Arthritis Rheum 27: 701- 3; Nambu M, Hayakawa Y, lto T, Takeda Z, Sakamoto Y, Mikawa H. Hypergammaglobulinemic purpura associated with múltiple sclerosis. (1988) J Pediatric 113: 331- 3; Lindsey LW, Albers GW, Steinman L. Recurrent transverse myelitis, myasthenia gravis, and áutoantibodies. (1992) Ann Neurol 32: 407- 9; Simeon-Aznar CP, Tolosa-Vilella C, Cuenca-Luque R, Jordana-Comajuncosa R, Ordi-Ros J, Bosch-Gil JA. Transverse myelitis in systemis lupus erythematosus: two cases with magnetic resonance imaging. (1992) Br J Rheumatol 31 : 555- 8; Bonnet F, Mercie P, Morlat P, Hocke C, Vergnes C, Ellie E, et al. Devic's neuromyelitis óptica during pregnancy in a patient with systemic lupus erythematosus. (1999) Lupus 8: 244- 7; Mochizuki A, Hayashi A, Hisahara S, Shoji S. Steroid-responsive Devic's variant in Sjogren's syndrome. Neurology 2000; 54: 1391- 2). A diferencia de Ia EM, existe una predilección racial por NMO en no blancos (O'Riordan Jl, Gallagher HL, Thompson AJ, et al. Clinical, CSF, and MRI findings Devic's neuromyelitis óptica. (1996) J Neurol Neurosurg Psychiatr 60: 382- 7). Los aborígenes en Manitoba tienen un riesgo incrementado para diferentes enfermedades que han sido implicadas más en Ia patogénesis de Ia NMO incluyendo infección por Mycobacterium Tuberculosis (Arvanitakis Z, Long RL, Hershfield ES, et al. M. Tuberculosis molecular variation in CNS infection : evidence for strain -dependent neurovirulence. (1998) Neurology 50: 1827- 1832), otras causas infecciosas (Young TK. The health of native Americans: toward a bioculturalepidemiology. Toronto: Oxford University Press, (1994)),
enfermedades autoinmunes (Oen K, El-Gabalawy HS, Canvin JM, et al. HLA associations of seropositive rheumathoid artritis in a Cree and Ojibway population. (1998) J Rheumatol 25: 2319- 2323) que en Ia EM. La NMO ha sido asociada a varias enfermedades sistémicas incluyendo enfermedades vasculares del colágeno, síndromes de auto-anticuerpos, infecciones y exposiciones a agentes tóxicos.Different clinical associations with NMO suggest that this disease is different from MS. Unlike EM 1, there are numerous reports of optic-spinal disease associated with connective tissue diseases and other autoimmune diseases (April RS, Vansonnenberg E. A case of optic neuromyelitis (Devic's syndrome) in systemic lupus erythematosus: clinical-pathologic report and review of the literature (1976) Neurology 26: 1066-70; Kinney EL, Berdoff RL 1 Rao NS, Fox LM. Devic's syndrome and systemic lupus erythematosus: a case report with necropsy. (1979) Arch Neurol 36: 643-4; Goldman M, Herode A, Borenstein S, Zanen A. Optic neuritis, transverse myelitis, and anti-DNA antibodies nine years after thymectomy for myasthenia gravis. (1984) Arthritis Rheum 27: 701-3; Nambu M, Hayakawa Y, lto T , Takeda Z, Sakamoto Y, Mikawa H. Hypergammaglobulinemic purple associated with multiple sclerosis. (1988) J Pediatric 113: 331-3; Lindsey LW, Albers GW, Steinman L. Recurrent transverse myelitis, myasthenia gravis, and áutoantibodies. (1992) Ann Neurol 32: 407-9; Sim eon-Aznar CP, Tolosa-Vilella C, Cuenca-Luque R, Jordana-Comajuncosa R, Ordi-Ros J, Bosch-Gil JA. Transverse myelitis in systemis lupus erythematosus: two cases with magnetic resonance imaging. (1992) Br J Rheumatol 31: 555-8; Bonnet F, Mercie P, Morlat P, Hocke C, Vergnes C, Ellie E, et al. Devic's optic neuromyelitis during pregnancy in a patient with systemic lupus erythematosus. (1999) Lupus 8: 244-7; Mochizuki A, Hayashi A, Hisahara S, Shoji S. Steroid-responsive Devic's variant in Sjogren's syndrome. Neurology 2000; 54: 1391-2). Unlike MS, there is a racial predilection for NMOs in non-whites (O'Riordan Jl, Gallagher HL, Thompson AJ, et al. Clinical, CSF, and MRI findings Devic's optic neuromyelitis. (1996) J Neurol Neurosurg Psychiatr 60: 382-7). Aboriginal people in Manitoba have an increased risk for different diseases that have been more implicated in the pathogenesis of NMO including Mycobacterium Tuberculosis infection (Arvanitakis Z, Long RL, Hershfield ES, et al. M. Molecular tuberculosis variation in CNS infection: evidence for strain -dependent neurovirulence. (1998) Neurology 50: 1827-1832), other infectious causes (Young TK. The health of native Americans: toward a bioculturalepidemiology. Toronto: Oxford University Press, (1994)), autoimmune diseases (Oen K, El-Gabalawy HS, Canvin JM, et al. HLA associations of seropositive rheumathoid arthritis in a Cree and Ojibway population. (1998) J Rheumatol 25: 2319-2323) than in MS. NMO has been associated with several systemic diseases including vascular diseases of collagen, autoantibody syndromes, infections and exposures to toxic agents.
La NMO también se ha asociado con enfermedades infecciosas, por ejemplo, virales, frecuentemente Ia NMO tiene un pródomo infeccioso caracterizado por cefalea, mialgias y síntomas respiratorios altos, en pocos casos se identifica un agente infeccioso, existen reportes de NMO después de mononucleosis infecciosa aguda (Williamson PM. Neuromyelitis óptica following infectious mononucleosis. (1975) Proc Aust Assoc Neurol 12: 153- 155), Varicela-zoster (Williamson PM. Neuromyelitis óptica following infectious mononucleosis. (1975) Proc Aust Assoc Neurol 12: 153- 155). Existe también una alta frecuencia de alteraciones en Ia RMN en el eje hipotálamo-pituitario en pacientes con NMO y endocrinopatías asociadas. Vernant y colaboradores (Vernart JC, Cabré P, Smadja D, et al. Recurrent optic neuromyelitis with endocrinopathies: a new syndrome (1997) Neurology 48: 58- 64) describieron una serie de 8 mujeres de Martinica y Guadalupe que sufrieron de NMO y endocrinopatías, 7 de las 8 pacientes tenían una amenorrea secundaria que coincidía con exacerbaciones de NMO. Una paciente postmenopáusica y otras 2 tenían galactorrea con hiperprolactinemia. 4 pacientes tenían hipotiroidismo y una paciente tenía Diabetes insípida. Los autores reportaron que 3 pacientes que tenían hiperfagia y obesidad sufrían de disfunción hipotalámica. En una paciente, un test de estimulación de hormona liberadora de tirotropina, indicó disfunción hipotalámica como causa de hiperprolactinemia. En 3 pacientes se encontraron lesiones con Gadolinio en Ia región hipotálamo-hipofisaria en Ia RMN. Todas las pacientes sufrieron de NO recurrente y mielitis que fue resistente a Ia terapia inmunosupresora y finalmente provocó ceguera y paraplejia. Las bandas oligoclonales en el LCR se encontraron solo en una paciente. En este estudio los hallazgos neuropatológicos demostraron pronunciada necrosis con desmielinización limitada, similar a Io reportado en Ia NMO (Baudoin D, Gambarelli D, Gayraud D, et al. Devic's neuromyelitis óptica: a clinicopathological review of the literature in
connection with a case showing fatal dysautonomia. (1998) Clin Neuropathol 17: 175- 183; Filippi M, Rocca MA, Moiola L1 et al. MRI and magnetization transfer imaging changes in the brain and cervical cord of patients with Devic's neuromyelitis óptica. Neurology 1999; 53: 1705- 1710) Io cual podría explicar Ia ausencia de beneficio observado con Glucocorticoídes y del tratamiento con Interferón beta.The NMO has also been associated with infectious diseases, for example, viral, frequently the NMO has an infectious prodrome characterized by headache, myalgias and high respiratory symptoms, in few cases an infectious agent is identified, there are reports of NMO after acute infectious mononucleosis (Williamson PM. Optic neuromyelitis following infectious mononucleosis. (1975) Proc Aust Assoc Neurol 12: 153-155), Varicella-zoster (Williamson PM. Optic neuromyelitis following infectious mononucleosis. (1975) Proc Aust Assoc Neurol 12: 153-155) . There is also a high frequency of NMR alterations in the hypothalamus-pituitary axis in patients with NMO and associated endocrinopathies. Vernant et al. (Vernart JC, Cabré P, Smadja D, et al. Recurrent optic neuromyelitis with endocrinopathies: a new syndrome (1997) Neurology 48: 58-64) described a series of 8 women from Martinique and Guadeloupe who suffered from NMO and Endocrinopathies, 7 of the 8 patients had a secondary amenorrhea that coincided with exacerbations of NMO. One postmenopausal patient and 2 others had galactorrhea with hyperprolactinemia. 4 patients had hypothyroidism and one patient had diabetes insipidus. The authors reported that 3 patients who had hyperphagia and obesity suffered from hypothalamic dysfunction. In one patient, a thyrotropin-releasing hormone stimulation test indicated hypothalamic dysfunction as a cause of hyperprolactinemia. In 3 patients, lesions with Gadolinium were found in the hypothalamic-pituitary region in the NMR. All patients suffered from recurrent NO and myelitis that was resistant to immunosuppressive therapy and finally caused blindness and paraplegia. The oligoclonal bands in the CSF were found only in one patient. In this study, the neuropathological findings demonstrated pronounced necrosis with limited demyelination, similar to that reported in the NMO (Baudoin D, Gambarelli D, Gayraud D, et al. Devic's optic neuromyelitis: a clinicopathological review of the literature in connection with a case showing fatal dysautonomia. (1998) Clin Neuropathol 17: 175-183; Filippi M, Rocca MA, Moiola L 1 et al. MRI and magnetization transfer imaging changes in the brain and cervical cord of patients with Devic's optic neuromyelitis. Neurology 1999; 53: 1705-1710) Which could explain the absence of benefit observed with Glucocorticoids and treatment with Interferon beta.
Existen 4 vertientes de evidencias separadas que soportan el papel de los mecanismos humorales en Ia patogénesis NMO: Ia patología de Ia lesión, Ia similitud con una variante selectiva de EAE inducida por MOG, Ia asociación clínica con trastornos vasculares del colágeno mediado por anticuerpos y Ia respuesta a Ia plasmaféresis. Aunque Ia activación del complemento se observa en una población de pacientes con EM (Lucchinetti CF, Mandler RN, McGavern D, Bruck W, Gleich G, Ransohoff RM, Trebst C, Weinshenker B, Wingerchuk D, Parisi JE, Lassman H. A role for humoral mechanism in the pathogenesis of Devic's neuromyelitis óptica. (2002) Brain 125: 1450- 61; Storch MK, Piddlesden S, Haltia M, livanainen M, Morgan P, Lassmann H. Múltiple Sclerosis: in situ evidence for antibody-and complement-mediated demyelination. (1998) Ann Neurol 43: 465- 71), el patrón y distribución perivascular pronunciada de activación del complemento que se ha visto en NMO es único. A pesar de las diferencias en las manifestaciones clínicas, descubrimientos imagenológicos, patología y bioquímica del LCR entre NMO y EM , Ia distinción entre estas enfermedades permanece controvertida (Mandler RN, Davis LE, Jeffery DR, Kornfeld M. Devic's neuromyeliris óptica: a clinicopathological study of 8 patients. (1993) Ann Neurol 34: 162- 8; Wingerchuk DM, Hogancamp WF, O'Brien PC, Weinshenker BG. The clinical course of neuromyelitis óptica (Devic's syndrome). (1999) Neurology 53: 1107- 14). Niveles reducidos del marcador de metaloproteinasa neuro-inflamatoria MMP-9 en el LCR de pacientes con NMO apoyan Ia posibilidad de un mecanismo patogénico diferente de producción de Ia lesión, ya que MMP-9 está marcadamente elevada en el LCR de Ia EM (Mandler RN, Davis LE, Jeffery DR, Kornfeld M. Devic's neuromyeliris óptica: a clinicopathological study of 8 patients. (1993) Ann Neurol 34: 162- 8). Además, el inhibidor tisular de metaloproteinasas (TIMP-1) fue significativamente reducido en el LCR de EMRR pero no en el LCR de NMO (Mandler RN, Dencoff JD, Midani F, Ford CC, Ahmed W, Rosenberg GA. Matrix
metalloproteinases and tissues inhibitors of metalloproteinases ¡n cerebrospinal fluid differ in múltiple sclerosis and Devic's neuromyelitis óptica. (2001) Brain 124: 493- 498). Estas diferencias bioquímicas en el LCR pueden estar bien relacionadas a diferencias en Ia reacción inflamatoria tisular en EM y NMO de Devic. Sin embargo, debido al espectro completo de Ia patología de la EM, incluyendo, Ia enfermedad óptico-espinal, puede reproducirse en el modelo de rata EAE inducido por MOG usando el mismo antígeno MOG pero con regímenes diferentes de sensibilización y cepas (Storch MK, Stefferl A, Brehm U, Weissert R, Wallstrom E, Kerschensteiner M1 et al. Autoimmunity to myelin oligodendrocyte glycoprotein in rats mimics the spectrum of múltiple sclerosis pathology. (1998) Brain Pathol ; 8: 681- 94), no está claro si Ia NMO es patogeneticamente distinta de Ia EM o si es un mayor reflejo de hospederos inmunogeneticamente diferentes donde influyen los factores ambientales.There are 4 separate evidences that support the role of humoral mechanisms in NMO pathogenesis: the pathology of the lesion, the similarity with a selective variant of EAE induced by MOG, the clinical association with vascular disorders of antibody-mediated collagen and Ia response to plasmapheresis. Although complement activation is observed in a population of patients with MS (Lucchinetti CF, Mandler RN, McGavern D, Bruck W, Gleich G, Ransohoff RM, Trebst C, Weinshenker B, Wingerchuk D, Parisi JE, Lassman H. A role for humoral mechanism in the pathogenesis of Devic's optic neuromyelitis. (2002) Brain 125: 1450-61; Storch MK, Piddlesden S, Haltia M, livanainen M, Morgan P, Lassmann H. Multiple Sclerosis: in situ evidence for antibody-and complement -mediated demyelination. (1998) Ann Neurol 43: 465-71), the pattern and pronounced perivascular distribution of complement activation seen in NMO is unique. Despite the differences in clinical manifestations, imaging findings, pathology and biochemistry of CSF between NMO and MS, the distinction between these diseases remains controversial (Mandler RN, Davis LE, Jeffery DR, Kornfeld M. Devic's optic neuromyeliris: a clinicopathological study of 8 patients. (1993) Ann Neurol 34: 162-8; Wingerchuk DM, Hogancamp WF, O'Brien PC, Weinshenker BG. The clinical course of optic neuromyelitis (Devic's syndrome). (1999) Neurology 53: 1107-14) . Reduced levels of the MMP-9 neuro-inflammatory metalloproteinase marker in the CSF of patients with NMO support the possibility of a different pathogenic mechanism of lesion production, since MMP-9 is markedly elevated in the CSF of the MS (Mandler RN , Davis LE, Jeffery DR, Kornfeld M. Devic's optic neuromyeliris: a clinicopathological study of 8 patients. (1993) Ann Neurol 34: 162-8). In addition, the tissue metalloproteinase inhibitor (TIMP-1) was significantly reduced in the EMRR CSF but not in the NMO CSF (Mandler RN, Dencoff JD, Midani F, Ford CC, Ahmed W, Rosenberg GA. Matrix Metalloproteinases and tissues inhibitors of metalloproteinases n cerebrospinal fluid differ in multiple sclerosis and Devic's optic neuromyelitis. (2001) Brain 124: 493-498). These biochemical differences in the CSF may be well related to differences in the inflammatory tissue reaction in MS and NMO of Devic. However, due to the complete spectrum of the pathology of MS, including, the optic-spinal disease, it can be reproduced in the MOG-induced EAE rat model using the same MOG antigen but with different sensitization regimes and strains (Storch MK, Stefferl A, Brehm U, Weissert R, Wallstrom E, Kerschensteiner M 1 et al. Autoimmunity to myelin oligodendrocyte glycoprotein in rats mimics the spectrum of multiple sclerosis pathology. (1998) Brain Pathol; 8: 681-94), it is unclear whether The NMO is pathogenetically different from the MS or if it is a greater reflection of immunogenetically different hosts where environmental factors influence.
NMO es una enfermedad desmielinizante inflamatoria invalidante y agresiva donde no se ha encontrado un tratamiento efectivo para esta enfermedad.NMO is an invalidating and aggressive inflammatory demyelinating disease where no effective treatment for this disease has been found.
Todas las recomendaciones terapéuticas representan experiencias anécdotales de una pequeña serie de casos no controlada. En Ia forma monofásica de NMO y por el índice de eventos y crisis en Ia NMO recurrente, Ia principal etapa en Ia terapia es el tratamiento de los ataques agudos, prevención médica de las complicaciones y Ia rehabilitación.All therapeutic recommendations represent anecdotal experiences of a small series of uncontrolled cases. In the single-phase form of NMO and by the index of events and crises in the recurrent NMO, the main stage in therapy is the treatment of acute attacks, medical prevention of complications and rehabilitation.
La mayoría de los pacientes que presentan NMO con exacerbaciones reciben tratamiento corticosteroide intravenoso, por ejemplo, 1000 mg de metilprepnisolona (MP) al día por 5 días consecutivos. La plasmaféresis ha sido reportada beneficiosa en el manejo de las crisis agudas de los pacientes con NMO con o sin trastornos del tejido conectivo asociado (Konttinen YT, Kinnunen E, von Bonsdorff M, Lillqvist P, lmmonen I, Bergroth V, et al. Acute transverse myelopathy successfully treated with plasmapheresis and prednisone ¡n a patient with primary Sjodren's syndrome. (1987) Arthritis Rheum 30: 339- 44; Fletchner KM, Baum K. Mixed connective tissue disease: recurrent episodes oh optic neuropathy and transverse myelopathy. Successful treatment with plamapheresis. (1994) J Neurol Sci 126: 146- 8; Weinshenker BG, O'Brien PC, Petterson TM, Noseworthy JH, Lucchinetti CF, Didick DW, et al. A randomized trial of plasma exchangein acute central nervous system inflammatory demyelinating disease (1999) Ann Neurol
46: 878- 86; Biliciler S1 Uygucgil H, Saip S, Altintas A, Soysal T, Ozdemir SE, et al. Plasmapheresis in múltiple sclerosis patients with diíferent indications (2001) MuIt Scler 7 Suppl 1: S64). La plasmaféresis reduce el número de auto- anticuerpos circulantes y complejos inmunes (Clark WF, Dan PC, Euler HH1 Guillevin L, Harstord J, Heer AH, et al. Plasmapheresis and subsequent pulse cyclophosphamide versus pulse cyclophosphamide alone in severe lupus: desing of the LPSG trial. (1991) J Clin Apheresis 6: 40-7J Io cual puede explicar cierta mejoría en algunos pacientes NMO. La inmunoglobulina intravenosa también ha sido usada anecdóticamente. La terapia preventiva se requiere para pacientes con enfermedad recidivante. La mayoría de las pacientes con NMO de Norteamérica reciben tratamiento conMost patients with NMO with exacerbations receive intravenous corticosteroid therapy, for example, 1000 mg of methylprepnisolone (MP) per day for 5 consecutive days. Plasmapheresis has been reported beneficial in the management of acute crises of patients with NMO with or without associated connective tissue disorders (Konttinen YT, Kinnunen E, von Bonsdorff M, Lillqvist P, lmmonen I, Bergroth V, et al. Acute transverse myelopathy successfully treated with plasmapheresis and prednisone ¡na patient with primary Sjodren's syndrome. (1987) Arthritis Rheum 30: 339-44; Fletchner KM, Baum K. Mixed connective tissue disease: recurrent episodes oh optic neuropathy and transverse myelopathy. Successful treatment with plamapheresis. (1994) J Neurol Sci 126: 146-8; Weinshenker BG, O'Brien PC, Petterson TM, Noseworthy JH, Lucchinetti CF, Didick DW, et al. A randomized trial of plasma exchangein acute central nervous system inflammatory demyelinating disease (1999) Ann Neurol 46: 878-86; Biliciler S 1 Uygucgil H, Saip S, Altintas A, Soysal T, Ozdemir SE, et al. Plasmapheresis in multiple sclerosis patients with diíferent indications (2001) MuIt Scler 7 Suppl 1: S64). Plasmapheresis reduces the number of circulating autoantibodies and immune complexes (Clark WF, Dan PC, Euler HH 1 Guillevin L, Harstord J, Heer AH, et al. Plasmapheresis and subsequent pulse cyclophosphamide versus pulse cyclophosphamide alone in severe lupus: desing of the LPSG trial (1991) J Clin Apheresis 6: 40-7J Which may explain some improvement in some NMO patients Intravenous immunoglobulin has also been used anecdotally Preventive therapy is required for patients with recurrent disease. NMO patients in North America receive treatment with
Interferón Beta parenteral pero basado en el hecho que no existe una experiencia controlada, creen que este tratamiento es inefectivo.Parenteral beta interferon but based on the fact that there is no controlled experience, they believe that this treatment is ineffective.
En el único estudio de tratamiento prospectivo publicado. Mandler et al encontraron que 7 pacientes NMO nuevamente diagnosticados se estabilizaron por al menos 18 meses con un régimen de Azatioprina y Prepnisona oral (Mandler RN, Ahmed W, Dencoff JE. Devic's neuromyelitis óptica : a prospective study of seven patients treated with prednisone and azathioprine . (1998) Neurology 51: 1219- 20). Existen evidencias que sugieren un efecto beneficioso de los IFNs tipo I en Ia NMO. Los IFN tipo I tienen un potente efecto antiviral, se inducen después que las células son infectadas por virus, causando Ia síntesis de un gran número de enzimas como Ia 2'5'oligoadenilato sintetasa que interfiere en Ia replicación viral RNA o DNA. Esta respuesta temprana (no específica de antígeno) es crítica para limitar Ia extensión del espectro viral antes que Ia respuesta específica para antígeno pueda controlar completamente Ia infección. Existen evidencias para una fuerte asociación de Ia NMO seguida a una infección viral, mononucleosis (Williamson PM. Neuromyelitis óptica following infectious mononucleosis. (1975) Proc Aust Assoc Neurol 12: 153- 155), varicela zoster (Doutlik S, Sblova O, Kryl R, Novak M. (Neuromyelitis óptica as a parainfectious complication of varicella). (1975) Cesk Neurol Neurochir 38: 238- 242), infecciones por HIV-1 (Blanche P, Díaz E, Gombert B, Sicard D, Rivoal O, Brezin A. Devic's neuromyelitis óptica and HIV-1 infection (2000) J Neurol Neurosurg Psychiatry 68: 795- 796), donde los síntomas y signos de NMO
estuvieron separados por días o semanas de Ia infección viral por Io que es compresible asociarla a una posible etiología infecciosa y una posible terapéutica con un fármaco con propiedades antivirales. Por otra parte, se reporta que el IFN alfa sinergiza con Ia IL-10 a inducir Ia diferenciación a células T reguladoras (Tr1), sugiriendo que vía Ia producción de grandes cantidades de IFN alfa, las células dendríticas 2 (DC2) pueden ser criticas para Ia inducción de Tr1 in vivo, por tanto, bajo una infección viral, las células DC2 pueden simultáneamente alertar a Ia respuesta inmune innata y adaptativa para producir IFNs tipo I. (Roncarolo MG1 Levings MK1 Traversari C. Differentiation of T Regulatory cells by immature dendritic cells. (2001) J Exp Med 193: F5-F9).In the only prospective treatment study published. Mandler et al found that 7 newly diagnosed NMO patients were stabilized for at least 18 months with an oral Azathioprine and Prepnisone regimen (Mandler RN, Ahmed W, Dencoff JE. Devic's optic neuromyelitis: a prospective study of seven patients treated with prednisone and azathioprine . (1998) Neurology 51: 1219-20). There is evidence to suggest a beneficial effect of type I IFNs in the NMO. Type I IFNs have a potent antiviral effect, they are induced after the cells are infected by viruses, causing the synthesis of a large number of enzymes such as 2.5'oligoadenylate synthetase that interferes with RNA or DNA viral replication. This early (non-specific antigen) response is critical to limit the spread of the viral spectrum before the antigen-specific response can completely control the infection. There is evidence for a strong association of NMO followed by a viral infection, mononucleosis (Williamson PM. Optical neuromyelitis following infectious mononucleosis. (1975) Proc Aust Assoc Neurol 12: 153-155), chickenpox zoster (Doutlik S, Sblova O, Kryl R, Novak M. (Optical neuromyelitis as a parainfectious complication of varicella). (1975) Cesk Neurol Neurochir 38: 238-242), HIV-1 infections (Blanche P, Diaz E, Gombert B, Sicard D, Rivoal O, Brezin A. Devic's optic neuromyelitis and HIV-1 infection (2000) J Neurol Neurosurg Psychiatry 68: 795-796), where the symptoms and signs of NMO they were separated by days or weeks of the viral infection by which it is compressible to associate it with a possible infectious etiology and a possible therapeutic with a drug with antiviral properties. On the other hand, it is reported that IFN alpha synergizes with IL-10 to induce differentiation to regulatory T cells (Tr1), suggesting that via the production of large amounts of IFN alpha, dendritic cells 2 (DC2) can be critical for induction of Tr1 in vivo, therefore, under a viral infection, DC2 cells can simultaneously alert the innate and adaptive immune response to produce type I IFNs (Roncarolo MG 1 Levings MK 1 Traversari C. Differentiation of T Regulatory cells by immature dendritic cells. (2001) J Exp Med 193: F5-F9).
Aunque existen reportes contradictorios que cuestionan el paradigma Th1/Th2, recientes evidencias sugieren el IFN alfa en humanos puede directamente inducir una diferenciación celular Th 1 Io cual sería beneficioso en NMO que se caracteriza por una respuesta Th2, Io cual constituye un elemento adicional que justifica Ia racionalidad del uso de interferón alfa en Ia obtención de un compuesto farmacéutico para el tratamiento de Ia neuromielitis óptica. Cuando los linfocitos T fueron cultivados en presencia de IFN-alfa y clonados (Parronchi P, De Carli M, Manetti R, Simonelli C, Sampognaro S, Piccini MP, Macchia D, Maggi E, Del Prate G, Romagnani S. IL-4 and IFN alfa and IFN gamma exert opposite regulatory effects on the development of cytolytic potential by Th1 or Th2 human T cell clones. (1992) J Immunol 149: 2977- 2983) o directamente estimulados (Brinkmann V, Geiger T, Alkan S, Heusser CH. interferón alpha increases the frequency of IFN gamma-producing CD4+ T cells. (1993) J Exp Med 178: 1655- 1663) vía el complejo TCR/CD3, Ia proporción de células productoras de IFN gamma (células tipo Th1) incrementó. Además, se demostró que Ia adición de IFN alfa a leucocitos del cordón umbilical humano estimulados bajo condiciones inductoras Th2 (IL-4 y anticuerpo monoclonal anti IL-12) resultó en el desarrollo de células Th1 productoras de IFN gamma (Rogge L, Barberis-Maino L, Biffi M, Passini N, Presky DH, Gubler U, Sinigaglia F. Selective expression of an interleukin-12 receptor component by human T helper 1 cells. (1997) J Exp Med 185: 825- 832). Los IFNs tipo I actúan directamente sobre células vírgenes induciendo diferenciación Th1 de células CD45RA+ estimuladas con anti-CD3 (Rogge L1 D'Ambrosio D1 Biffi M1 Penna G, Minetti LJ, Presky DH, Adorini L, Sinigaglia F. The role of stat4 in species-specific
regulation of Th cell development by type I IFNs. (1998) J Immunol 161 : 12: 6567- 6574). Existen evidencias in vivo de que Ia administración de los IFNs tipo I pueden favorecer el desarrollo de células Th 1 humanas. Las células productoras de IFN gamma de los pacientes con EM aumentan en número cuando los pacientes son tratados con IFN beta (Dayal A, Jensen MA, Lledo A, Arnason BGW. interferón-gamma-secreting cells ¡n múltiple sclerosis patients treated with interferón beta-1b. (1995) Neurology 45: 2173- 2177). La terapia con IFN alfa recombinante se ha usado en enfermedades consideradas ser principalmente mediadas por Ia sobre-expresión de citoquinas Th2 tales como el síndrome hlpereosinofílico (Zielinskl RM, Lawrence WD. interferón alpha for the hyper-eosinophilic syndrome. (1990) Ann Int Med 113: 716-718; Butterfield JH, Gleich GJ. interferón alpha treatment of 6 patients with the idiopathic hypereosinophilic syndrome. (1994) Ann Int Med 121 : 648- 653) y dermatitis atópica (Mackie RM. interferón alpha for atopic dermatitis. (1990) Lancet 335: 1282- 1283). El IFN alfa induce activación de Stat4 y por consiguiente respuesta Th1(Cho SS, Bacon CM1 Sudarshan C, Rees RC, Finbloom D, Pine R, O'Shea JJ. Activation of Stat4 by IL-12 and IFN alpha. Evidence for the involvement of ligand-induced tyrosine and serine phosphorilation. (1996) J Immunol 157: 4781- 4789). En Ia patogenia de Ia NMO al igual que en Ia EM, Ia desmielinización juega un papel patológico importante y se ha observado que el IFN gamma no está implicado en Ia desmielinización. Se ha demostrado que el IFN alfa induce IFN gamma. En los ratones knockout de IFN gamma se ha observado un incremento de Ia inflamación y Ia desmielinización (Tran EH, Prince EN, Owens T. IFN- gamma shapes immune invasión of the central nervous system vía regulation of chemokines. (2000) J Immunol 164: 2759- 2768) sugiriendo que una vía independiente de IFN gamma es Ia que media Ia destrucción de Ia mielina. El IFN gamma previene Ia acumulación de neutrófilos en el SNC de los pacientes con NMO. Dos recientes reportes (Tran EH, Prince EN, Owens T. IFN-gamma shapes immune invasión of the central nervous system vía regulation of chemokines. (2000) J Immunol 164: 2759- 2768; Willenborg DO, Fordham S, Bernard CC, Cowden WB, Ramshaw IA. IFN-gamma plays a critical down- regulatory role in the induction and effector phase of myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis. (1996) J Immunol 157:
3223- 3227) han demostrado que en los ratones_knockout de IFN gamma o su receptor hay acumulación de gran número de neutrófilos y que estos ratones expresan quimiocinas en el SNC que atraen neutrófilos. De acuerdo a las argumentaciones expresadas anteriormente sería provechoso para el tratamiento de Ia NMO Ia utilización de un compuesto farmacéutico caracterizado por contener componentes con actividad remielinizante, inmunomoduladora y/o inductora de células T reguladoras. Otra de las proteínas cuyas propiedades pudieran explicar una acción beneficiosa en el SNC es Ia Eritropoyetina (EPO), Ia observación de que Ia EPO y su receptor se expresan en el tejido cerebral de humanos y roedores (Juul, S. E., Anderson, D. K., Li, Y. & Christensen, R. D. (1998) Pediatr. Res. 43, 40-49; Marti, H. H., Wenger, R. H., Rivas, L A., Straumann, U., Digicaylioglu, M., Henn, V., Yonekawa, Y., Bauer, C. & Gassmann, M. (1996) Eur. J. Neurosci. 8, 666-676; Sirén, A.-L, Knerlich, F., Poser, W., Gleiter, C, Brück, W. & Ehrenreich, H. (2001) Acta Neuropathol) así como por cultivos neuronales (Morishita, E., Masuda, S., Nagao, M., Yasuda, Y. & Sasaki, R. (1997) Neuroscience 76, 105-116; Konishi, Y., Chui, D. H., Hirose, H., Kunishita, T. & Tabira, T. (1993) Brain Res. 609, 29-35; Lewczuk, P., Hasselblatt, M., Kamrowski-Kruck, H., Heyer, A., Unzicker, C, Siren, A. L. & Ehrenreich, H. (2000) NeuroReport 11 , 3485-3488) y astrocitos (Lewczuk, P., Hasselblatt, M., Kamrowski-Kruck, H., Heyer, A., Unzicker, C, Siren, A. L. & Ehrenreich, H. (2000) NeuroReport 11 , 3485-3488; Marti, H. H., Wenger, R. H., Rivas, L. A., Straumann, U., Digicaylioglu, M., Henn, V., Yonekawa, Y., Bauer, C. & Gassmann, M. (1996) Eur. J. Neurosci. 8, 666-676) y que tiene efectos sobre las neuronas (Konishi, Y., Chui, D. H., Hirose, H., Kunishita, T. & Tabira, T. (1993) Brain Res. 609, 29-35) expandiendo sus efectos biológicos más allá que Ia hematopoyesis podrían explicar algunos de sus beneficios teniendo en cuenta la patogenia de Ia NMO. El efecto neutoprotector ha sido bien documentado en cultivos celulares (Bemaudin, M., Bellail, A., Marti, H. H., Yvon, A., Vivien, D., Duchatelle, I., Mackenzie, E. T. & Petit, E. (2000) GHa 30, 271-278; Lewczuk, P., Hasselblatt, M., Kamrowski-Kruck, H., Heyer, A., Unzicker, C, Siren, A. L. & Ehrenreich, H. (2000) NeuroReport 11 , 3485-3488; Morishita, E., Masuda, S., Nagao, M., Yasuda, Y. & Sasaki, R. (1997) Neuroscience 76, 105-116) y en modelos de infarto en roedores (Bernaudin, M., Marti, H. H., Roussel, S.,
Divoux, D., Nouvelot, A., MacKenzie, E. T. & Petit, E. (1999) J. Cereb. Blood Flow Metab. 19, 643-651 ; Brines, M. L., Ghezzi, P., Keenan, S., Agnello, D., de Lanerolle, N. C1 Cerami, C1 Itri, L. M. & Cerami, A. (2000) Proc. Nati. Acad. Sci. USA 97, 10526-10531 ; Sadamoto, Y., Igase, K., Sakanaka, M., Sato, K., Otsuka, H., Sakaki, S., Masuda, S. & Sasaki, R. (1998) Biochem. Biophys. Res. Commun. 253, 26-32; Sakanaka, M., Wen, T. C1 Matsuda, S., Masuda, S., Morishita, E., Nagao, M. & Sasaki, R. (1998) Proc. Nati. Acad. Sci. USA 95, 4635-4640). Existen evidencias que cascadas de señalización que han sido bien caracterizadas en líneas celulares hematopoyéticas (IhIe, J. N. (1995) Nature (London) 377, 591-594) son funcionales en neuronas y pueden ser moduladas por EPO. El efecto anti-apoptótico del BDNF donde están involucradas las cascadas de señalización referidas anteriormente (Hetmán, M., Kanning, K., Cavanaugh, J. E. & Xia, Z. (1999) J. Biol. Chem. 274, 22569- 22580). Los efectos antioxidantes (Chattopadhyay, A., Choudhury, T. D., Bandyopadhyay, D. & Datta, A. G. (2000) Biochem. Pharmacol. 59, 419-425), angiogénico (Ribatti, D., Presta, M., Vacca, A., Ria, R., Giuliani, R., Dell'Era, P., Nico, B., Roncali, L. & Dammacco, F. (1999) Blood 93, 2627-2636) y neurotrófico (Campana, W. M., Misasi, R. & O'Brien, J. S. (1998) Int. J. Mol. Med. 1 , 235-241) enfatizan el potencial regenerativo de esta molécula, además del efecto neuroprotector. El efecto antinflamatorio ya que disminuye el infiltrado inflamatorio inducido por trauma cortical en ratones y mejora Ia EAE por Io que es considerada una citoquina antinflamatoria: Un efecto directo antinflamatorio de EPO a nivel de citoquinas no puede excluirse ya que ha sido reportado que EPO disminuye Ia producción ex vivo de TNF-alfa y aumenta Ia producción de IL-10 en cultivo de células de pacientes hemodializados (Bryl, E., Mysliwska, J., Debska-Slizien, A., Rachon, D., Bullo, B., Lizakowski, S., Mysliwski, A. & Rutkowski, B. (1998) Artif. Organs 22, 177-181). A Ia EPO se Ie ha asociado un efecto inmunomodulador al demostrarse que retarda el incremento de los niveles de TNF-alfa y disminuye los niveles de IL-6 en un modelo de EAE en ratas Lewis ( Agnello D., Bigini P, Villa P, Mennini T, Cerami A, Brines ML, Ghezzi P (2002) Brain Research 952, 128- 134). EPO directamente afecta Ie excitabilidad neuronal de esta manera también puede limitar Ia necrosis neuronal (Brines, M. L., Ghezzi, P., Keenan, S., Agnello, D., de Lanerolle, N.
C1 Cerami, C, Itri, L. M. & Cerami, A. (2000) Proc. Nati. Acad. Sci. USA 97, 10526-10531). Tiene un efecto citoprotector ya que diferentes grupos han evidenciado que EPO protege cultivos neuronales contra Ia toxicidad por glutamato (Bernaudin, M., Bellail, A., Marti, H. H., Yvon, A., Vivien, D., Duchatelle, I., Mackenzie, E. T. & Petit, E. (2000) Glia 30, 271-278; Morishita, E., Masuda, S., Nagao, M., Yasuda, Y. & Sasaki, R. (1997) Neuroscience 76, 105-116) y reduce el daño neuronal isquémico y Ia disfunción neurológica en modelos de infarto en roedores (Morishita, E., Masuda, S., Nagao, M., Yasuda, Y. & Sasaki, R. (1997) Neuroscience 76, 105-116; Bernaudin, M., Marti, H. H., Roussel, S., Divoux, D., Nouvelot, A., MacKenzie, E. T. & Petit, E. (1999) J. Cereb. Blood Flow Metab. 19, 643-651; Brines, M. L., Ghezzi, P., Keenan, S., Agnello, D., de Lanerolle, N. C1 Cerami, C1 Itri, L. M. & Cerami, A. (2000) Proc. Nati. Acad. Sci. USA 97, 10526-10531). La EPO administrada sistémicamente atraviesa Ia BHE (Cerami A, Brines ML, Ghezzi P, Cerami CJ. Effects of epoetin alfa on central nervous system. Semin Oncol 2001 Apr;28(2 Suppl 8):66-70).Although there are conflicting reports that question the Th1 / Th2 paradigm, recent evidence suggests that IFN alpha in humans can directly induce a Th 1 I cell differentiation which would be beneficial in NMO characterized by a Th2 response, which constitutes an additional element that justifies The rationality of the use of interferon alfa in obtaining a pharmaceutical compound for the treatment of optic neuromyelitis. When T lymphocytes were cultured in the presence of IFN-alpha and cloned (Parronchi P, De Carli M, Manetti R, Simonelli C, Sampognaro S, Piccini MP, Macchia D, Maggi E, Del Prate G, Romagnani S. IL-4 and IFN alfa and IFN gamma exert opposite regulatory effects on the development of cytolytic potential by Th1 or Th2 human T cell clones. (1992) J Immunol 149: 2977-2983) or directly stimulated (Brinkmann V, Geiger T, Alkan S, Heusser Interferon alpha increases the frequency of IFN gamma-producing CD4 + T cells. (1993) J Exp Med 178: 1655-1663) via the TCR / CD3 complex, the proportion of IFN gamma producing cells (Th1 type cells) increased. In addition, it was demonstrated that the addition of IFN alpha to human umbilical cord leukocytes stimulated under Th2 inducing conditions (IL-4 and anti-IL-12 monoclonal antibody) resulted in the development of IFN gamma-producing Th1 cells (Rogge L, Barberis- Maino L, Biffi M, Passini N, Presky DH, Gubler U, Sinigaglia F. Selective expression of an interleukin-12 receptor component by human T helper 1 cells. (1997) J Exp Med 185: 825-832). Type I IFNs act directly on virgin cells inducing Th1 differentiation of CD45RA + cells stimulated with anti-CD3 (Rogge L 1 D'Ambrosio D 1 Biffi M 1 Penna G, Minetti LJ, Presky DH, Adorini L, Sinigaglia F. The role of stat4 in species-specific regulation of Th cell development by type I IFNs. (1998) J Immunol 161: 12: 6567-6574). There is evidence in vivo that the administration of type I IFNs can favor the development of human Th 1 cells. IFN gamma producing cells of patients with MS increase in number when patients are treated with IFN beta (Dayal A, Jensen MA, Lledo A, Arnason BGW. Interferon-gamma-secreting cells in multiple sclerosis patients treated with interferon beta -1b. (1995) Neurology 45: 2173-2177). Therapy with recombinant IFN alpha has been used in diseases considered to be mainly mediated by the over-expression of Th2 cytokines such as hlpereosinophilic syndrome (Zielinskl RM, Lawrence WD. Interferon alpha for the hyper-eosinophilic syndrome. (1990) Ann Int Med 113: 716-718; Butterfield JH, Gleich GJ. Interferon alpha treatment of 6 patients with the idiopathic hypereosinophilic syndrome. (1994) Ann Int Med 121: 648-653) and atopic dermatitis (Mackie RM. Interferon alpha for atopic dermatitis. ( 1990) Lancet 335: 1282-1283). IFN alpha induces activation of Stat4 and therefore Th1 response (Cho SS, Bacon CM 1 Sudarshan C, Rees RC, Finbloom D, Pine R, O'Shea JJ. Activation of Stat4 by IL-12 and IFN alpha. Evidence for the involvement of ligand-induced tyrosine and serine phosphorilation. (1996) J Immunol 157: 4781-4789). In the pathogenesis of the NMO as in the case of MS, demyelination plays an important pathological role and it has been observed that gamma IFN is not involved in demyelination. IFN alpha has been shown to induce gamma IFN. In IFN gamma knockout mice, an increase in inflammation and demyelination has been observed (Tran EH, Prince EN, Owens T. IFN-gamma shapes immune invasion of the central nervous system via regulation of chemokines. (2000) J Immunol 164 : 2759-2768) suggesting that an independent IFN gamma pathway is the one that mediates the destruction of myelin. IFN gamma prevents the accumulation of neutrophils in the CNS of patients with NMO. Two recent reports (Tran EH, Prince EN, Owens T. IFN-gamma shapes immune invasion of the central nervous system via regulation of chemokines. (2000) J Immunol 164: 2759-2768; Willenborg DO, Fordham S, Bernard CC, Cowden WB, Ramshaw IA. IFN-gamma plays a critical down- regulatory role in the induction and effector phase of myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis. (1996) J Immunol 157: 3223-3227) have shown that large numbers of neutrophils accumulate in the IFN gamma_nockout mice or their receptor and that these mice express chemokines in the CNS that attract neutrophils. According to the arguments expressed above, the use of a pharmaceutical compound characterized by containing components with remyelinating, immunomodulating and / or inducing regulatory T cell activity would be helpful for the treatment of the NMO. Another protein whose properties could explain a beneficial action in the CNS is Erythropoietin (EPO), the observation that EPO and its receptor are expressed in the brain tissue of humans and rodents (Juul, SE, Anderson, DK, Li , Y. & Christensen, RD (1998) Pediatr. Res. 43, 40-49; Marti, HH, Wenger, RH, Rivas, L A., Straumann, U., Digicaylioglu, M., Henn, V., Yonekawa , Y., Bauer, C. & Gassmann, M. (1996) Eur. J. Neurosci. 8, 666-676; Sirén, A.-L, Knerlich, F., Poser, W., Gleiter, C, Brück , W. & Ehrenreich, H. (2001) Acta Neuropathol) as well as by neuronal cultures (Morishita, E., Masuda, S., Nagao, M., Yasuda, Y. & Sasaki, R. (1997) Neuroscience 76, 105-116; Konishi, Y., Chui, DH, Hirose, H., Kunishita, T. & Tabira, T. (1993) Brain Res. 609, 29-35; Lewczuk, P., Hasselblatt, M., Kamrowski -Kruck, H., Heyer, A., Unzicker, C, Siren, AL & Ehrenreich, H. (2000) NeuroReport 11, 3485-3488) and astrocytes (Lewczuk, P., Hasselblatt, M., Kamrowski-Kruck, H., Heyer, A., A zicker, C, Siren, AL & Ehrenreich, H. (2000) NeuroReport 11, 3485-3488; Marti, HH, Wenger, RH, Rivas, LA, Straumann, U., Digicaylioglu, M., Henn, V., Yonekawa, Y., Bauer, C. & Gassmann, M. (1996) Eur. J. Neurosci. 8, 666-676) and which has effects on neurons (Konishi, Y., Chui, DH, Hirose, H., Kunishita, T. & Tabira, T. (1993) Brain Res. 609, 29-35) expanding its biological effects beyond that hematopoiesis could explain some of its benefits taking into account the pathogenesis of NMO. The neutral protective effect has been well documented in cell cultures (Bemaudin, M., Bellail, A., Marti, HH, Yvon, A., Vivien, D., Duchatelle, I., Mackenzie, ET & Petit, E. (2000 ) GHa 30, 271-278; Lewczuk, P., Hasselblatt, M., Kamrowski-Kruck, H., Heyer, A., Unzicker, C, Siren, AL & Ehrenreich, H. (2000) NeuroReport 11, 3485- 3488; Morishita, E., Masuda, S., Nagao, M., Yasuda, Y. & Sasaki, R. (1997) Neuroscience 76, 105-116) and in rodent infarction models (Bernaudin, M., Marti , HH, Roussel, S., Divoux, D., Nouvelot, A., MacKenzie, ET & Petit, E. (1999) J. Cereb. Blood Flow Metab. 19, 643-651; Brines, ML, Ghezzi, P., Keenan, S., Agnello, D., de Lanerolle, N. C 1 Cerami, C 1 Itri, LM & Cerami, A. (2000) Proc. Nati Acad. Sci. USA 97, 10526-10531; Sadamoto, Y., Igase, K., Sakanaka, M., Sato, K., Otsuka, H., Sakaki, S., Masuda, S. & Sasaki, R. (1998) Biochem. Biophys Res. Commun. 253, 26-32; Sakanaka, M., Wen, T. C 1 Matsuda, S., Masuda, S., Morishita, E., Nagao, M. & Sasaki, R. (1998) Proc. Nati Acad. Sci. USA 95, 4635-4640). There is evidence that signaling cascades that have been well characterized in hematopoietic cell lines (IhIe, JN (1995) Nature (London) 377, 591-594) are functional in neurons and can be modulated by EPO. The anti-apoptotic effect of BDNF where the signaling cascades referred to above are involved (Hetmán, M., Kanning, K., Cavanaugh, JE & Xia, Z. (1999) J. Biol. Chem. 274, 22569-22580) . Antioxidant effects (Chattopadhyay, A., Choudhury, TD, Bandyopadhyay, D. & Datta, AG (2000) Biochem. Pharmacol. 59, 419-425), angiogenic (Ribatti, D., Presta, M., Vacca, A ., Ria, R., Giuliani, R., Dell'Era, P., Nico, B., Roncali, L. & Dammacco, F. (1999) Blood 93, 2627-2636) and neurotrophic (Campana, WM, Misasi, R. &O'Brien, JS (1998) Int. J. Mol. Med. 1, 235-241) emphasize the regenerative potential of this molecule, in addition to the neuroprotective effect. The anti-inflammatory effect since it reduces the inflammatory infiltrate induced by cortical trauma in mice and improves EAE by what is considered an anti-inflammatory cytokine: A direct anti-inflammatory effect of EPO at the cytokine level cannot be excluded since EPO has been reported to decrease ex vivo production of TNF-alpha and increases the production of IL-10 in cell culture of hemodialyzed patients (Bryl, E., Mysliwska, J., Debska-Slizien, A., Rachon, D., Bullo, B., Lizakowski, S., Mysliwski, A. & Rutkowski, B. (1998) Artif. Organs 22, 177-181). An EPO has been associated with an immunomodulatory effect by demonstrating that it delays the increase in TNF-alpha levels and decreases IL-6 levels in an EAE model in Lewis rats (Agnello D., Bigini P, Villa P, Mennini T, Cerami A, Brines ML, Ghezzi P (2002) Brain Research 952, 128-134). EPO directly affects neuronal excitability in this way can also limit neuronal necrosis (Brines, ML, Ghezzi, P., Keenan, S., Agnello, D., de Lanerolle, N. C 1 Cerami, C, Itri, LM & Cerami, A. (2000) Proc. Nati Acad. Sci. USA 97, 10526-10531). It has a cytoprotective effect since different groups have shown that EPO protects neuronal cultures against glutamate toxicity (Bernaudin, M., Bellail, A., Marti, HH, Yvon, A., Vivien, D., Duchatelle, I., Mackenzie, ET & Petit, E. (2000) Glia 30, 271-278; Morishita, E., Masuda, S., Nagao, M., Yasuda, Y. & Sasaki, R. (1997) Neuroscience 76, 105- 116) and reduces ischemic neuronal damage and neurological dysfunction in rodent infarction models (Morishita, E., Masuda, S., Nagao, M., Yasuda, Y. & Sasaki, R. (1997) Neuroscience 76, 105 -116; Bernaudin, M., Marti, HH, Roussel, S., Divoux, D., Nouvelot, A., MacKenzie, ET & Petit, E. (1999) J. Cereb. Blood Flow Metab. 19, 643- 651; Brines, ML, Ghezzi, P., Keenan, S., Agnello, D., de Lanerolle, N. C 1 Cerami, C 1 Itri, LM & Cerami, A. (2000) Proc. Nati. Acad. Sci USA 97, 10526-10531). Systemically administered EPO crosses the BHE (Cerami A, Brines ML, Ghezzi P, Cerami CJ. Effects of epoetin alfa on central nervous system. Semin Oncol 2001 Apr; 28 (2 Suppl 8): 66-70).
DESCRIPCIÓN DETALLADA DE LA INVENCIÓNDETAILED DESCRIPTION OF THE INVENTION
La presente invención describe el uso del interferón alfa para Ia obtención de un compuesto farmacéutico que contiene más específicamente, interferón alfa 2b recombinante, útil en el tratamiento de pacientes con NMO, enfermedad para Ia cual no se ha descrito hasta el momento un tratamiento efectivo.The present invention describes the use of interferon alfa for obtaining a pharmaceutical compound that contains more specifically, recombinant interferon alfa 2b, useful in the treatment of patients with NMO, a disease for which an effective treatment has not been described so far.
En una realización particular dicho compuesto se utilizo a las dosis terapéuticas de 3 y 10 millones de Ul, 2 veces a Ia semana.In a particular embodiment, said compound was used at therapeutic doses of 3 and 10 million Ul, twice a week.
Las propiedades inmunomoduladoras, antivirales, remielinizantes e inductoras de células T reguladoras del compuesto farmacéutico de Ia presente invención justifican su efectividad en Ia NMO.The immunomodulatory, antiviral, remyelinating and inducing properties of regulatory T cells of the pharmaceutical compound of the present invention justify its effectiveness in the NMO.
Los resultados obtenidos en nuestra invención, sugieren que el IFN alfa en humanos puede directamente inducir una diferenciación celular Th1 avalando su efecto inmunomodulador, ya que en Ia mayoría de los pacientes tratados con el compuesto farmacéutico se observó un aumento de Ia expresión del gen de IFN gamma y una disminución de IL-4, Io cual sería beneficioso en NMO que se caracteriza por una respuesta Th2.
Se ha demostrado que el IFN alfa induce IFN gamma, Io cual es corroborado en otra realización de nuestra invención, pero Ia desmielinización observada en Ia NMO no está implicado el IFN gamma, sugiriendo Ia participación de una vía independiente al IFN gamma, de hecho, en nuestra invención encontramos niveles elevados después de Ia terapia con IFN alfa 2b recombinante del CNTF, marcador de remielinización, Io cual avala el efecto remielinizante del compuesto farmacéutico.The results obtained in our invention suggest that IFN alpha in humans can directly induce a Th1 cell differentiation endorsing its immunomodulatory effect, since in the majority of patients treated with the pharmaceutical compound an increase in IFN gene expression was observed. gamma and a decrease in IL-4, which would be beneficial in NMO characterized by a Th2 response. It has been shown that IFN alpha induces gamma IFN, which is corroborated in another embodiment of our invention, but the demyelination observed in the NMO does not involve gamma IFN, suggesting the participation of an independent path to gamma IFN, in fact, In our invention we find high levels after the CNTF recombinant IFN alpha 2b therapy, remyelination marker, which supports the remyelinating effect of the pharmaceutical compound.
En nuestra invención se describe un mayor número de pacientes donde existió un incremento de marcadores de células T reguladoras naturales (CD25 y Foxp3) e inducidas (IL-10 y TGF-β) después del tratamiento con IFN alfa 2b recombinante, efecto del IFN alfa que no se había demostrado previamente en ausencia de sinergismo con otra molécula.In our invention, a greater number of patients are described where there was an increase in natural and induced regulatory (CD25 and Foxp3) T-cell markers (IL-10 and TGF-β) after treatment with recombinant IFN alpha 2b, effect of IFN alpha that had not previously been demonstrated in the absence of synergism with another molecule.
Estas propiedades del compuesto farmacéutico justifican sus capacidades terapéuticas en NMO. La mayoría de las recomendaciones terapéuticas para NMO representan experiencias anecdotales de una pequeña serie de casos no controlada, los reportes de uso de IFN beta en NMO son poco alentadores, por ejemplo, Mirsattari et al, 2001 reportaron un simple caso de autopsia, caracterizado por necrosis e infiltrado eosinofílico dentro de Ia lesión de Ia medula espinal Io cual atribuyeron al tratamiento crónico con IFN beta. Recientemente el grupo de Saida et al, 2005 reportaron un estudio con IFN beta con algunos resultados favorables pero tiene el inconveniente que no es un estudio a doble ciegas como el referido en nuestra invención.These properties of the pharmaceutical compound justify its therapeutic capabilities in NMO. Most therapeutic recommendations for NMO represent anecdotal experiences of a small uncontrolled case series, reports of use of IFN beta in NMO are not very encouraging, for example, Mirsattari et al, 2001 reported a simple case of autopsy, characterized by Eosinophilic necrosis and infiltrate within the spinal cord injury, which they attributed to chronic treatment with IFN beta. Recently the group of Saida et al, 2005 reported a study with IFN beta with some favorable results but it has the disadvantage that it is not a double blind study like the one referred to in our invention.
Las diferencias entre algunas actividades biológicas de los IFNs alfa y beta, así como las divergencias desde el punto de vista etiopatogénico entre Ia NMO y Ia EM, refutan el hecho de que Ia demostración del efecto del IFN beta hacen obvio el efecto terapéutico del IFN alfa Io cual debe ser demostrado como hemos descrito en nuestra invención.The differences between some biological activities of the alpha and beta IFNs, as well as the divergences from the etiopathogenic point of view between the NMO and the MS, refute the fact that the demonstration of the effect of the IFN beta makes the therapeutic effect of the IFN alpha obvious Which should be demonstrated as we have described in our invention.
Nuestra invención tiene como novedad el hecho de ser un Ensayo Clínico a doble ciegas, aleatorizado y controlado con placebo donde se demuestra que el IFN alfa2brec logra Ia disminución de los brotes y Ia frecuencia de las recaídas en pacientes con NMO recidivante.Our invention is the fact that it is a double-blind, randomized and placebo-controlled clinical trial where it is demonstrated that IFN alfa2brec achieves the reduction of outbreaks and the frequency of relapses in patients with recurrent NMO.
La invención también incluye composiciones farmacéuticas que comprenden Ia combinación con Eritropoyetina (EPO rec).
En nuestra invención Ia combinación de IFN alfa2brec con EPO rea, mostró disminuir ei grado de atrofia cerebral, así como modificó positivamente marcadores moleculares involucrados en Ia patogenia de Ia enfermedad. En nuestra invención Ia combinación mostró un efecto sinérgico entre IFN alfa2brec y EPO rec. con relación a eventos inmunomoduladores e inductores de células T reguladoras, reflejados por Ia proporción de pacientes que elevaron los niveles de expresión CD25, Foxp3, IL-10 y TGF-β demostrando mejoría del contexto autoinmune que caracteriza Ia enfermedad, por otra parte, Ia combinación mostró ser útil en acelerar los procesos de neurogénesis a través de sus propiedades neurotróficas, anti-apoptóticas y remielinizantes, reflejado en el efecto sinérgico de los 2 componentes de Ia combinación respecto a los principios independientes sobre el aumento del marcador anti-apoptótico o BDNF y el marcador de remielinización CNTF. El compuesto farmacéutico referido en nuestra invención para el tratamiento de Ia NMO se caracteriza porque contiene interferón alfa que puede ser modificado por peguilación o fusión a otras proteínas manteniendo las actividades referidas anteriormente.The invention also includes pharmaceutical compositions comprising the combination with Erythropoietin (EPO rec). In our invention, the combination of IFN alfa2brec with EPO rea, showed a decrease in the degree of cerebral atrophy, as well as positively modified molecular markers involved in the pathogenesis of the disease. In our invention, the combination showed a synergistic effect between IFN alfa2brec and EPO rec. in relation to immunomodulatory events and inducers of regulatory T cells, reflected by the proportion of patients who raised the levels of expression CD25, Foxp3, IL-10 and TGF-β demonstrating improvement of the autoimmune context that characterizes the disease, on the other hand, Ia combination was shown to be useful in accelerating neurogenesis processes through its neurotrophic, anti-apoptotic and remyelinating properties, reflected in the synergistic effect of the 2 components of the combination with respect to the independent principles on the increase of the anti-apoptotic marker or BDNF and the remyelination marker CNTF. The pharmaceutical compound referred to in our invention for the treatment of NMO is characterized in that it contains interferon alfa that can be modified by peguylation or fusion to other proteins while maintaining the activities referred to above.
La administración terapéutica del interferón alfa 2b o de Ia combinación IFN alfa con EPO requiere de administraciones repetidas. Conforme a Io descrito en Ia presente invención, tanto el compuesto farmacéutico que contiene IFN alfa solo o modificado por peguilacion, de manera independiente o formando parte de composiciones farmacéuticas que Io combinan con Factores de Crecimiento como Ia Eritropoyetina, podrían administrarse por vía intramuscular, intravenosa, subcutánea, oral, nasal e intratecal.The therapeutic administration of interferon alfa 2b or of the combination IFN alfa with EPO requires repeated administrations. According to what is described in the present invention, both the pharmaceutical compound containing IFN alpha alone or modified by peeling, independently or as part of pharmaceutical compositions that combine it with Growth Factors such as Erythropoietin, could be administered intramuscularly, intravenously , subcutaneous, oral, nasal and intrathecal.
EJEMPLOS.EXAMPLES
Ejemplo 1 : Características del compuesto.Example 1: Characteristics of the compound.
Se trata de un compuesto farmacéutico que contiene Interferón alfa 2b recombinante producido en Escherichia CoIi, donde las características de Ia formulación final se exponen en Ia Tabla 1.1, dicho compuesto además puede ser modificado por adición de Polietilenglicol (PEG-IFN) Tabla 1.2 y puede contener Factores de crecimiento como Eritropoyetina recombinante producida
en células CHO (células de ovario de hámster chínese), cuyas características de Ia formulación final se exponen en Ia Tabla 1.3. Tabla 1.1It is a pharmaceutical compound containing recombinant Interferon alpha 2b produced in Escherichia CoIi, where the characteristics of the final formulation are set forth in Table 1.1, said compound can also be modified by adding Polyethylene Glycol (PEG-IFN) Table 1.2 and can contain growth factors such as recombinant erythropoietin produced in CHO cells (chinesse hamster ovary cells), whose characteristics of the final formulation are set forth in Table 1.3. Table 1.1
Interferón alfa 2b recombinanteRecombinant interferon alpha 2b
Cada vial contiene Interferón alfa 2b recombinante en un rango entre 3 y 10 x106 Ul, 10 mg alcohol de bencilo, 0,2mg polisorbato 80, 4,67mg NaCL, 2,96mg NaH2PO4^H2O, 14,41 mg Na2HPO4.2H2O y agua para inyección a completar 1 mLEach vial contains recombinant Interferon alfa 2b in a range between 3 and 10 x10 6 Ul, 10 mg benzyl alcohol, 0.2mg polysorbate 80, 4.67mg NaCL, 2.96mg NaH 2 PO 4 ^ H 2 O, 14.41 mg Na 2 HPO 4 .2H 2 O and water for injection to complete 1 mL
Tabla 1.2Table 1.2
PEG-IFNPEG-IFN
Cada vial contiene PEG-I nterferón alfa 2b recombinante en un rango entre 180 y 360 μg, 2,617 mg acetato de sodio tri-hidratado, 8 mg NaCL, 10 mg de alcohol de bencilo, 0,05 mg de polisorbato y agua para inyección a completar 1 mL.Each vial contains recombinant PEG-I interferon alfa 2b in a range between 180 and 360 μg, 2,617 mg tri-hydrated sodium acetate, 8 mg NaCL, 10 mg benzyl alcohol, 0.05 mg polysorbate and water for injection at complete 1 mL.
Tabla 1.3Table 1.3
Interferón alfa 2b recombinante + Eritropoyetina recombinanteRecombinant interferon alfa 2b + recombinant Erythropoietin
Cada vial contiene Interferón alfa 2b recombinante en un rango entre 3 y 10 x106 Ul, Eritropoyetina recombinante 2000 Ul, 10 mg alcohol de bencilo, 0,2mg polisorbato 80, 4,67mg NaCL, 2,96mg NaH2PO4.2H2O, 14,41 mg Na2HPO4.2H2O y agua para inyección a completar 1 mLEach vial contains recombinant Interferon alfa 2b in a range between 3 and 10 x10 6 Ul, recombinant Erythropoietin 2000 Ul, 10 mg benzyl alcohol, 0.2mg polysorbate 80, 4.67mg NaCL, 2.96mg NaH 2 PO 4 .2H 2 O, 14.41 mg Na 2 HPO 4 .2H 2 O and water for injection to complete 1 mL
Ejemplo 2: Características de Ia muestra evaluada. Evaluación del efecto terapéutico del IFN α2b rec en NMO.Example 2: Characteristics of the sample evaluated. Evaluation of the therapeutic effect of IFN α2b rec in NMO.
Para evaluar el efecto terapéutico IFN α2brec se realizó un Ensayo Clínico Fase III aleatorizado, a doble ciegas y controlado con placebo que incluyó 18 pacientes que presentaban Ia forma recurrente de NMO según los criterios de Wingerchuck et al. Los pacientes recibieron de forma aleatorizada placebo, IFN α2brec 3 millones y 10 millones administrado por vía intramuscular, 2 veces por semana durante 2 años. La evaluación clínica se realizó a través de las Escalas de Scripps y EDSS realizándose Ia evaluación de Ia proporción de pacientes libres de brotes y Ia comparación del número de brotes entre los pacientes en tratamiento y los mismos pacientes los 2 años previos al inicio del tratamiento.
Como se muestra en Ia Tabla 2 Ia muestra era homogénea en cuanto a edad, sexo y raza. El hecho de que todos los pacientes eran del sexo femenino (100% en cada grupo) está en concordancia con los reportes de Ia literatura que refieren una mayor incidencia de Ia NMO en las mujeres. Además, Ia distribución uniforme de los diferentes grupos raciales en Ia muestra demostrándose un predominio de Ia enfermedad en Ia razas no blancas (75% en el placebo, 62.5% en el grupo de IFN α2brec 3 millones y 50% en el grupo de IFN α2brec 10 millones) también concuerda con los reportes de Ia literatura al respecto. Tabla 2. Homogeneidad de Ia muestra estudiada.To evaluate the therapeutic effect of IFN α2brec, a randomized, double-blind, placebo-controlled Phase III Clinical Trial was carried out that included 18 patients presenting with the recurrent form of NMO according to the criteria of Wingerchuck et al. Patients received randomized placebo, IFN α2brec 3 million and 10 million administered intramuscularly, twice a week for 2 years. The clinical evaluation was carried out through the Scripps and EDSS Scales, making the evaluation of the proportion of outbreak-free patients and the comparison of the number of outbreaks between patients under treatment and the same patients the 2 years prior to the start of treatment. As shown in Table 2, the sample was homogeneous in terms of age, sex and race. The fact that all patients were female (100% in each group) is in accordance with the reports in the literature that report a higher incidence of NMO in women. In addition, the uniform distribution of the different racial groups in the sample demonstrating a predominance of the disease in non-white races (75% in the placebo, 62.5% in the IFN α2brec 3 million group and 50% in the IFN α2brec group 10 million) also agrees with the literature reports in this regard. Table 2. Homogeneity of the sample studied.
Ejemplo 3: Evaluación de Ia evolución de los brotes.Example 3: Evaluation of the evolution of the outbreaks.
Se consideraron brotes al empeoramiento y/o aparición de nuevos síntomas y/o signos neurológicos que persistieron por más de 24 horas. Se midió Ia proporción de pacientes de los diferentes grupos (placebo, IFN α2brec 3 y 10 millones) que tuvieron brotes durante 2 años.Outbreaks were considered as worsening and / or the appearance of new symptoms and / or neurological signs that persisted for more than 24 hours. The proportion of patients from the different groups (placebo, IFN α2brec 3 and 10 million) who had outbreaks for 2 years was measured.
Se realizaron 2 evaluaciones (al año y 2 años de tratamiento) y se compararon con el número de brotes que presentaron los pacientes los 2 años previos al tratamiento.
Como se observa en Ia Tabla 3, los resultados muestran que el 66.7% de los pacientes tratados con IFN α2brec 10 millones estuvieron libres de brotes durante los 2 años evaluados. Se encontró diferencia estadísticamente significativa entre el total del número de brotes de los pacientes que recibieron placebo y aquellos tratados con IFN α2brec (p=0.013) respecto al número de brotes que tuvieron los 2 años previos, estratificando, dicha diferencia se observó entre el grupo placebo y el grupo tratado con IFN α2brec 10 millones ( p=0,026).Two evaluations were performed (per year and 2 years of treatment) and compared with the number of outbreaks that the patients presented the 2 years prior to treatment. As observed in Table 3, the results show that 66.7% of the patients treated with IFN α2brec 10 million were free of outbreaks during the 2 years evaluated. A statistically significant difference was found between the total number of outbreaks of patients receiving placebo and those treated with IFN α2brec (p = 0.013) with respect to the number of outbreaks that had the previous 2 years, stratifying, this difference was observed between the group placebo and the group treated with IFN α2brec 10 million (p = 0.026).
Tabla 3. Efecto del tratamiento con IFN α2brec sobre Ia evolución del número de brotes en pacientes con NMO.Table 3. Effect of the treatment with IFN α2brec on the evolution of the number of outbreaks in patients with NMO.
Ejemplo 4: Características de Ia muestra evaluada. Evaluación del efecto terapéutico de Ia combinación IFN α2brec / EPO en NMO.Example 4: Characteristics of the sample evaluated. Evaluation of the therapeutic effect of the IFN α2brec / EPO combination in NMO.
Para evaluar el efecto terapéutico de Ia combinación IFN α2brec / EPO se seleccionaron 9 pacientes con NMO según los criterios de Wingerchuck et al., 3 se trataron con IFN α2brec, 3 con EPO rec y 3 con Ia combinación IFN α2brec / EPO.To evaluate the therapeutic effect of the IFN α2brec / EPO combination, 9 patients with NMO were selected according to the criteria of Wingerchuck et al., 3 were treated with IFN α2brec, 3 with rec EPO and 3 with the IFN α2brec / EPO combination.
Los 3 pacientes del primer grupo recibieron IFN α2brec 3 millones IM 2 veces a Ia semana, los 3 del segundo grupo recibieron EPO rec SC 500U/kg 2 veces a Ia semana y los 3 del tercer grupo, ambos tratamientos combinados a las dosis referidas anteriormente por un período de 1 año. La evaluación clínica se realizó a los 6 meses y 1 año de tratamiento por RMN para medir Ia evolución de Ia atrofia cerebral. Al inicio y al año con los diferentes tratamientos, se Ie realizaron determinaciones moleculares a nivel de ARN y proteína que incluyeron: Determinación por RT-PCR en células mononucleares aisladas de sangre periférica de Ia expresión del gen para el receptor de quimiocinas CCR3, proteína catiónica de eosinófilos, CD25, Foxp3, IFN gamma, IL-10 y TGF-beta y determinación de las concentraciones séricas del Factor neurotrófico derivado del cerebro (BDNF) y el Factor neurotrófico ciliar (CNTF) por ELISA.The 3 patients in the first group received IFN α2brec 3 million IM twice a week, the 3 in the second group received EPO rec SC 500U / kg twice a week and 3 in the third group, both treatments combined at the doses referred to above for a period of 1 year. The clinical evaluation was performed at 6 months and 1 year of NMR treatment to measure the evolution of cerebral atrophy. At the beginning and after one year with the different treatments, molecular determinations were made at the level of RNA and protein that included: Determination by RT-PCR in mononuclear cells isolated from peripheral blood of the gene expression for the CCR3 chemokine receptor, cationic protein of eosinophils, CD25, Foxp3, IFN gamma, IL-10 and TGF-beta and determination of serum concentrations of brain-derived neurotrophic factor (BDNF) and ciliary neurotrophic factor (CNTF) by ELISA.
En Ia Tabla 4 se muestra las características de los pacientes evaluados. La muestra era homogénea en cuanto a edad, sexo y raza. La mayoría de los pacientes eran del sexo femenino y Ia raza negra Io cual concuerda con los reportes de la literatura para esta enfermedad. Tabla 4. Homogeneidad de Ia muestra estudiada.
Table 4 shows the characteristics of the patients evaluated. The sample was homogeneous in terms of age, sex and race. The majority of the patients were female and the black race, which is consistent with the literature reports for this disease. Table 4. Homogeneity of the sample studied.
Ejemplo 5: Efecto del tratamiento con Ia combinación IFN α2brec / EPO sobre Ia atrofia cerebral.Example 5: Effect of the treatment with the IFN α2brec / EPO combination on cerebral atrophy.
Para medir el grado de atrofia cerebral de realizaron mediciones en T1 del grado de atrofia cerebral por RMN a los 6 meses y 1 año de tratamiento. Los resultados se reflejan como el % de pacientes en los que hubo una disminución de Ia atrofia cerebral en el momento de Ia evaluación respecto al inicio.To measure the degree of cerebral atrophy, T1 measurements of the degree of cerebral atrophy were performed by NMR at 6 months and 1 year of treatment. The results are reflected as the% of patients in whom there was a decrease in cerebral atrophy at the time of the evaluation with respect to the onset.
En Ia Tabla 5 se expone el resultado de Ia evaluación de Ia atrofia cerebral en los pacientes tratados con Ia IFN α2brec, EPO rec y Ia combinación IFN α2brec / EPO. Como se observa Ia combinación IFN α2brec / EPO disminuyo el grado de atrofia cerebral en el 66,6% de los pacientes al año de tratamiento demostrando un efecto mayor que con los tratamientos independientes.Table 5 shows the result of the evaluation of cerebral atrophy in patients treated with IFN α2brec, EPO rec and the IFN α2brec / EPO combination. As can be seen, the IFN α2brec / EPO combination reduced the degree of cerebral atrophy in 66.6% of patients a year of treatment, demonstrating a greater effect than with independent treatments.
Tabla 5. Evaluación del grado de atrofia cerebral en pacientes con NMO.Table 5. Evaluation of the degree of cerebral atrophy in patients with NMO.
Ejemplo 6: Modulación de genes involucrados en Ia patogenia de Ia NMO. Example 6: Modulation of genes involved in the pathogenesis of the NMO.
Se realizó Ia evaluación por RT-PCR de Ia modulación de diferentes genes involucrados en Ia patogenia de Ia NMO al año de tratamiento con IFN α2brec, EPO rec y Ia combinación IFN α2brec / EPO en los pacientes. Los resultados se reflejan como % de pacientes en los que aumenta/disminuye los niveles relativos del gen evaluado respecto a Ia expresión de un gen constitutivo.The evaluation by RT-PCR of the modulation of different genes involved in the pathogenesis of the NMO was performed one year after treatment with IFN α2brec, EPO rec and the IFN α2brec / EPO combination in the patients. The results are reflected as% of patients in whom the relative levels of the evaluated gene increase / decrease with respect to the expression of a constitutive gene.
En Ia Tabla 6 se reflejan los resultados donde es válido destacar que en el 100% de los pacientes tratados con Ia combinación IFN α2brec / EPO disminuyó Ia expresión del CCR3, receptor de quimiocinas que se expresa exclusivamente en eosinófilos, en concordancia con este resultado también se encontró que en Ia mayoría de los pacientes tratados con Ia combinación disminuyó Ia expresión del gen correspondiente a Ia proteína catiónica de eosinófilos Io que parece indicar que Ia combinación IFN α2brec / EPO puede ejercer un efecto tanto en Ia disminución del infiltrado eosinofílico característico de esta enfermedad como en Ia disminución de Ia degranulación de estas células donde uno de los componentes fundamentales de estos granulos es precisamente Ia proteína catiónica de eosinófilos. Por otra parte nos llama Ia atención que el tratamiento con Ia combinación provocó un aumento de Ia expresión del gen de IL-10 y TGF-beta pudiendo indicar Ia participación de un mecanismo de células T reguladoras inducidas. Un dato significativo resulta que el 100% de los pacientes tratados con Ia combinación presentan un aumento de la expresión del CD25 y del factor transcripcional Foxp3 pudiendo indicar participación de células reguladoras naturales. Otro dato relevante obtenido Io constituye que el 66.6% de los pacientes presentan un incremento de los niveles de IFN gamma, citoquina TH 1 lo cual es beneficioso en esta enfermedad que al caracterizarse por un predominio TH2, una polarización de Ia respuesta a TH 1 resultaría beneficiosa.Table 6 shows the results where it is valid to highlight that in 100% of the patients treated with the IFN α2brec / EPO combination, the expression of CCR3, a chemokine receptor that is expressed exclusively in eosinophils, decreased in accordance with this result also it was found that in the majority of patients treated with the combination the expression of the gene corresponding to the cationic eosinophilic protein decreased, which seems to indicate that the IFN α2brec / EPO combination can have an effect both on the decrease of the characteristic eosinophilic infiltrate of this disease as in the decrease of the degranulation of these cells where one of the fundamental components of these granules is precisely the cationic protein of eosinophils. On the other hand, it is our attention that the treatment with the combination caused an increase in the expression of the IL-10 and TGF-beta gene and may indicate the participation of a mechanism of induced regulatory T cells. A significant fact is that 100% of the patients treated with the combination have an increase in the expression of CD25 and the Foxp3 transcriptional factor, which may indicate the participation of natural regulatory cells. Another relevant data obtained constitutes that 66.6% of the patients present an increase in the levels of IFN gamma, cytokine TH 1 which is beneficial in this disease that when characterized by a TH2 predominance, a polarization of the response to TH 1 would result beneficial
Tabla 6, Efecto de los diferentes tratamientos y Ia combinación IFN α2brec / EPO sobre Ia modulación de algunos genes involucrados en Ia patogenia de Ia NMO.
Table 6, Effect of the different treatments and the IFN α2brec / EPO combination on the modulation of some genes involved in the pathogenesis of the NMO.
Ejemplo 7: Evaluación de los factores anti-apoptóticos (BDNF) y remielinizantes (CNTF) en pacientes con NMO tratados con los tratamientos independientes y Ia combinación IFN α2brec / EPO.Example 7: Evaluation of anti-apoptotic (BDNF) and remyelinating (CNTF) factors in patients with NMO treated with independent treatments and the IFN α2brec / EPO combination.
Se realizaron las mediciones por ELISA de los factores anti-apoptóticos (BDNF) y remielinizantes (CNTF) en el suero de los pacientes con NMO al año de tratamiento con IFN α2brec, EPO rec y Ia combinación IFN α2brec / EPO. Los resultados se expresan como % de pacientes en los que aumentó/disminuyó las concentraciones séricas de ambos factores al año de tratamiento respecto a los valores antes de iniciar el tratamiento.
Los resultados se reflejan en Ia Tabla 7 observándose como para ambos factores se observa un aumento de las concentraciones de dichos factores con el tratamiento con Ia combinación Io que habla a favor del efecto anti-apoptótico y remielinizante de esta molécula.ELISA measurements of anti-apoptotic (BDNF) and remyelinating (CNTF) factors were performed in the serum of patients with NMO at one year of treatment with IFN α2brec, EPO rec and the IFN α2brec / EPO combination. The results are expressed as% of patients in whom the serum concentrations of both factors increased / decreased at one year of treatment with respect to the values before starting treatment. The results are reflected in Table 7, observing how for both factors an increase in the concentrations of these factors is observed with the treatment with the combination that speaks in favor of the anti-apoptotic and remyelinating effect of this molecule.
Tabla 7. Efecto de los tratamientos independientes y de Ia combinación IFN α2brec / EPO sobre el BDNF y CNTF en pacientes con NMO.Table 7. Effect of independent treatments and the IFN α2brec / EPO combination on BDNF and CNTF in patients with NMO.
Claims
REINVIDICACIONESREDEMPTIONS
1- Uso del interferón alfa para Ia obtención de un compuesto farmacéutico para el tratamiento de Ia neuromielitis óptica.1- Use of interferon alfa to obtain a pharmaceutical compound for the treatment of optic neuromyelitis.
2- Uso de acuerdo con Ia reivindicación 1 , caracterizado porque el interferón es el interferón alfa 2b.2- Use according to claim 1, characterized in that the interferon is interferon alfa 2b.
3- Uso de acuerdo con las reivindicaciones 1 y 2, caracterizado porque el interferón se encuentra en el compuesto a una concentración entre 3 - 10 millones de Ul.3- Use according to claims 1 and 2, characterized in that the interferon is in the compound at a concentration between 3-10 million Ul.
4- Uso de acuerdo con las reivindicaciones 1 , 2 y 3, caracterizado porque el interferón puede ser modificado por peguilación o fusión a otras proteínas.4- Use according to claims 1, 2 and 3, characterized in that the interferon can be modified by peguylation or fusion to other proteins.
5- Uso de acuerdo con las reivindicaciones 1 , 2, 3 y 4, caracterizado porque se coadministran componentes con actividad remielinizante, inmunomoduladora y/o inductora de células T reguladoras.5- Use according to claims 1, 2, 3 and 4, characterized in that components with remyelinating, immunomodulating and / or inducing activity of regulatory T cells are co-administered.
6- Uso de acuerdo con las reivindicaciones anteriores, caracterizado porque coadministra Eritropoyetina.6- Use according to the preceding claims, characterized in that co-administration Erythropoietin.
7- Uso de acuerdo con las reivindicaciones 1 , 2, 3, 4, 5 y 6 caracterizado porque el compuesto farmacéutico se administra por vía intramuscular, intravenosa, subcutánea, oral, nasal e intratecal.7- Use according to claims 1, 2, 3, 4, 5 and 6 characterized in that the pharmaceutical compound is administered intramuscularly, intravenously, subcutaneously, orally, nasally and intrathecally.
8- Compuesto farmacéutico para el tratamiento de Ia neuromielitis óptica de acuerdo con Ia reivindicación 1 , caracterizado porque contiene interferón alfa.
8- Pharmaceutical compound for the treatment of optic neuromyelitis according to claim 1, characterized in that it contains interferon alfa.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CU2005-0127 | 2005-06-24 | ||
CU20050127 | 2005-06-24 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2006136116A1 true WO2006136116A1 (en) | 2006-12-28 |
Family
ID=36930260
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CU2006/000005 WO2006136116A1 (en) | 2005-06-24 | 2006-06-23 | Use of interferon-alpha in order to obtain a compound for the treatment of optical neuromyelitis |
Country Status (2)
Country | Link |
---|---|
AR (1) | AR054513A1 (en) |
WO (1) | WO2006136116A1 (en) |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1993006856A1 (en) * | 1991-10-11 | 1993-04-15 | Mark Cedric Gillies | Treating ophthalmic fibrosis using interferon-alpha |
JPH06271478A (en) * | 1993-03-23 | 1994-09-27 | Otsuka Pharmaceut Co Ltd | Agent for treating dry eye |
-
2006
- 2006-06-23 AR ARP060102706A patent/AR054513A1/en not_active Application Discontinuation
- 2006-06-23 WO PCT/CU2006/000005 patent/WO2006136116A1/en active Application Filing
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1993006856A1 (en) * | 1991-10-11 | 1993-04-15 | Mark Cedric Gillies | Treating ophthalmic fibrosis using interferon-alpha |
JPH06271478A (en) * | 1993-03-23 | 1994-09-27 | Otsuka Pharmaceut Co Ltd | Agent for treating dry eye |
Non-Patent Citations (2)
Title |
---|
DATABASE WPI Section Ch Week 199443, Derwent World Patents Index; Class B04, AN 1994-347064, XP002402334 * |
NOSEWORTHY JOHN H: "Treatment of multiple sclerosis and related disorders: What's new in the past 2 years?", 2003, CLINICAL NEUROPHARMACOLOGY, VOL. 26, NR. 1, PAGE(S) 28-37, PHILADELPHIA, ISSN: 0362-5664, XP009071976 * |
Also Published As
Publication number | Publication date |
---|---|
AR054513A1 (en) | 2007-06-27 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2018206705B2 (en) | Use of low dose IL-2 for treating autoimmune - related or inflammatory disorders | |
US10765723B2 (en) | Use of low dose IL-2 for treating autoimmune—related or inflammatory disorders | |
Liu et al. | Chemokine receptor CXCR3: an unexpected enigma | |
Vieira et al. | Mice lacking the TNF receptor p55 fail to resolve lesions caused by infection with Leishmania major, but control parasite replication. | |
JP2020040976A (en) | Reducing systemic regulatory t cell levels or activity for treatment of disease and injury of cns | |
US8017392B2 (en) | Hox11+, CD45− cells and methods of organ regeneration using the same | |
JP2010132660A5 (en) | ||
WO2013022882A1 (en) | Combination therapy for treatment of inflammatory demyelinating disease | |
RU2411042C2 (en) | Compositions and methods for treating severe acute respiratory syndrome (sars) | |
Kimura et al. | Injury and subsequent regeneration of muscles for activation of local innate immunity to facilitate the development and relapse of autoimmune myositis in C57BL/6 mice | |
US10933049B2 (en) | Mobilizing agents and uses therefor | |
Fox | Sjogren’s syndrome: evolving therapies | |
CA2936065C (en) | Methods and compositions used in treating inflammatory and autoimmune diseases | |
KR20190071785A (en) | Methods, compositions and dosage regimens for the treatment or prevention of interferon gamma related indications | |
WO2006136116A1 (en) | Use of interferon-alpha in order to obtain a compound for the treatment of optical neuromyelitis | |
Boster et al. | The potential role of B cell-targeted therapies in multiple sclerosis | |
US12030950B2 (en) | Treatment of multiple sclerosis and neuromyelitis optica | |
Glaubitz et al. | Update on myositis therapy: from today’s standards to tomorrow’s possibilities | |
Polman et al. | Interferons | |
US20210002372A1 (en) | Methods and compositions for treating natalizumab-associated progressive multifocal encephalopathy | |
ES2967696T3 (en) | Epidermal growth factor (EGF) and variants thereof for treating or preventing inflammatory demyelinating disorders | |
Sá | Dissecting pentraxin-3 role in the modulation of Multiple Sclerosis | |
Dionisio-Santos | Ameliorating Alzheimer’s Disease Tau Pathology Through Immunomodulation: A Novel Role for Interleukin 4 and Glatiramer Acetate | |
Syage | Characterizing the Roles of Microglia in Disease and Repair in the CNS using a Viral Model of Multiple Sclerosis | |
Croxford et al. | Back to the future for multiple sclerosis therapy: focus on current and emerging disease-modifying therapeutic strategies |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
DPE2 | Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101) | ||
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
NENP | Non-entry into the national phase |
Ref country code: DE |
|
WWW | Wipo information: withdrawn in national office |
Country of ref document: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 06753159 Country of ref document: EP Kind code of ref document: A1 |