[go: up one dir, main page]

MX2008006948A - Antibodies against amyloid beta 4 with glycosylated in the variable region - Google Patents

Antibodies against amyloid beta 4 with glycosylated in the variable region

Info

Publication number
MX2008006948A
MX2008006948A MXMX/A/2008/006948A MX2008006948A MX2008006948A MX 2008006948 A MX2008006948 A MX 2008006948A MX 2008006948 A MX2008006948 A MX 2008006948A MX 2008006948 A MX2008006948 A MX 2008006948A
Authority
MX
Mexico
Prior art keywords
antibody
glycosylated
antibody molecule
antibodies
seq
Prior art date
Application number
MXMX/A/2008/006948A
Other languages
Spanish (es)
Inventor
Hans Koll
Kurt Lang
Andreas Schaubmar
Hansruedi Loetscher
Walter Huber
Diana Schuhbauer
Karl Weyer
Manfred Brockhaus
Bernd Bohrmann
Original Assignee
Bernd Bohrmann
Manfred Brockhaus
F Hoffmannla Roche Ag
Walter Huber
Hans Koll
Kurt Lang
Hansruedi Loetscher
Andreas Schaubmar
Diana Schuhbauer
Karl Weyer
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bernd Bohrmann, Manfred Brockhaus, F Hoffmannla Roche Ag, Walter Huber, Hans Koll, Kurt Lang, Hansruedi Loetscher, Andreas Schaubmar, Diana Schuhbauer, Karl Weyer filed Critical Bernd Bohrmann
Publication of MX2008006948A publication Critical patent/MX2008006948A/en

Links

Abstract

The present invention relates to a purified antibody molecule preparation being characterized in that at least one antigen binding site comprises a glycosylated asparagine (Asn) in the variable region of the heavy chain (VH). More specifically, a pharmaceutical and a diagnostic composition comprising said antibody molecule and antibody mixtures are provided which is/are capable of specifically recognizing theβ-A4 peptide/Aβ4. The present invention relates in particular to a mixture of antibodies comprising one or two glycosylated antigen binding sites with a glycosylated asparagine (Asn) in the variable region of the heavy chain, i.e. mixtures of isoforms of antibodies which comprise a glycosylated Asn in the variable region of the heavy chain (VH). Also disclosed are compositions or antibody preparations comprising the specifically glycosylated antibody isoforms. Furthermore, the pharmaceutical and diagnostic uses for these antibodies are provided. The antibody isoforms may for example be used in the pharmaceutical intervention of amyloidogenesis or amyloid-plaque formation and/or in the diagnosis of the same.

Description

GLICOSILATION IN THE VARIABLE REGION The present invention relates to a preparation of purified antibody molecule characterized in that at least one antigen-binding site comprises a glycosylated asparagin (Asn) in the variable region of the heavy chain (VH). More specifically, a purified antibody molecule is provided that is capable of specifically recognizing the β-A4 / Aβ4 peptide and comprising a glycosylation in the variable region of the heavy chain (VH). The present invention relates, in particular, to a mixture of antibodies comprising one or two glycosylated antigen-binding sites with a glycosylated asparagin (Asn) in the variable region of the heavy chain, ie, mixtures of antibody isoforms which they comprise a glycosylated Asn in the variable region of the heavy chain (VH). Also disclosed are compositions or preparations of antibodies that specifically comprise isoforms of glycosylated antibodies. In addition, pharmaceutical and diagnostic uses for these antibodies are provided. The antibody isoforms can be used, for example, in the pharmaceutical intervention of amyloidogenesis or amyloid plaque formation and / or in their diagnosis. Approximately 70% of all cases of dementia are due to Alzheimer's disease that is associated with selective damage to brain regions and neural circuits critical for cognition. The disease Alzheimer's disease is characterized by neurofibrillary tangles, particularly in the pyramidal neurons of the hippocampus and numerous amyloid plaques that contain mostly a dense core of amyloid deposits and diffuse haloes. Extracellular neuritic plaques contain large amounts of a predominantly fibrilated peptide termed "β-amyloid", "A-beta", "Aβ4", "β-A4" or "Aβ", see Selkoe (1994), Ann. Rev. Cell Biol. 10, 373-403, Koo (1999), PNAS Vol. 96, p. 9989-9990, US 4,666,829 or Glenner (1984), BBRC 12, 1131. This β-amyloid is derived from the "Alzheimer's precursor protein / β-amyloid precursor protein" (APP). APPs are integral membrane glycoproteins (see Sisodia (1992), PNAS Vol. 89, page 6075) and endoproteolytically cleaved within the Aβ sequence by means of a plasma membrane protease, a-secretase (see Sisodia (1992), loc. cit.) . In addition, an additional activity of secretase, in particular, the activity of β-secretase and β-secretase, leads to the extracellular release of β-amyloid (Aβ) comprising either 39 amino acids (Aß39), 40 amino acids (Aβ 40). ), 42 amino acids (Aβ42) or 43 amino acids (Aβ43); see Sinha (1999), PNAS 96, 11094-1053; Price (1998), Science 282, 1078 to 1083; WO 00/72880 or Hardy (1997), TINS 20, 154. It should be noted that Aß has many forms that occur naturally, where human forms are referred to as the aforementioned Aß39, Aß40, Aβ41, Aβ42 and Aβ43. The most predominant form, Aß42, has the amino acid sequence (beginning with the term N): DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO: 3). In Aß41, Aß40, Aß39, the C-terminal amino acids, A, IA and VIA, respectively, are missing. In the Aβ43 form, an additional threonine residue is comprised in the C-terminus of the sequence described above (SEQ ID NO: 3). The time required to nuclear Aβ40 fibrils was significantly longer than to nuclear Aβ42 fibrils; see Koo, loe. cit. and Harper (1997), Ann. Rev. Biochem. 66, 385-407. As indicated in Wagner (1999), J. Clin. Invest. 104, 1239-1332, Aß42 is found, more frequently, associated with neuritic plaques and is considered to be more fibrillogenic in vi tro. It was also suggested that Aβ42 serves as a "seed" in polymerization dependent on the nucleation of ordered non-crystalline Aβ peptides; Jarrett (1993), Cell 93, 1055-1058. The processing of modified APP and / or the generation of extracellular plaques containing protein depositions are not only known from Alzheimer's disease, but also from subjects suffering from other neurological and / or degenerative disorders. These disorders include, inter alia, Down syndrome, hereditary cerebral hemorrhage with "Dutch type" amyloidosis, Parkinson's disease, ALS (amyotrophic lateral sclerosis), Creutzfeld Jacob's disease, HIV-related dementia and motor neuropathy So far, only limited medical intervention schemes have been described for diseases related to amyloid. For example, cholinesterase inhibitors, such as galantamine, rivastigmine or donepezil, have been described as beneficial for Alzheimer's patients with only mild to moderate disease. However, adverse events have also been reported due to the cholinergic action of these drugs. Although these cholinergic enhancement treatments produce some symptomatic benefit, the therapeutic response is not satisfactory for the majority of treated patients. It has been estimated that significant cognitive improvement occurs only in approximately 5% of treated patients and there is little evidence that the treatment significantly alters the course of this progressive disease. Consequently, there is a great clinical need for more effective treatments and, in particular, those that can stop or delay the progression of the disease. Also, more recently, NMDA receptor antagonists, such as memantine, have been used. However, adverse events have been reported due to pharmacological activity. Furthermore, such treatment with these NMDA receptor antagonists can simply be considered as a symptomatic method and not as a method that modifies the disease.
In addition, immunomodulation methods have been proposed for the treatment of disorders related to amyloid. WO 99/27944 discloses conjugates comprising parts of the Aβ peptide and carrier molecules, wherein said carrier molecule must improve the immune response. Another method of active immunization is mentioned in WO 00/72880, where fragments of Aβ are also used to induce the immune response. Also, methods of passive immunization with general anti-Aβ antibodies have been proposed in WO 99/27944 or WO 01/62801 and specific humanized antibodies directed against parts of Aβ have been described in WO 02/46237, WO 02 / 088306 and WO 02/088307. WO 00/77178 describes antibodies that bind to a transition state adopted by β-amyloid during hydrolysis. WO 03/070760 discloses antibody molecules that recognize two discontinuous amino acid sequences in the Aβ peptide. WO 03/016466 describes a humanized anti-Aβ antibody that is modified in order to avoid any potential glycosylation in its heavy chain, since a glycosylation in the variable region (s) of the antibodies has been described in Wallick (1988) J. Exp. Med. 168, 1099-1109. The technical problem underlying the present invention is to provide effective means and methods in the medical management of amyloid disorders, in particular, means and methods for the reduction of harmful amyloid plaques in patients who need a medical intervention (corresponding). In a first aspect, the present invention provides a purified antibody molecule characterized in that at least one antigen-binding site comprises a glycosylated asparagin (Asn) in the variable region of the heavy chain (VH). The purified antibody of the invention or the antibody composition provided herein is directed, in particular, against Aß and / or a fragment of Aβ. The purified antibody molecule provided herein and, in particular, the antibody composition or antibody preparation of the invention, is useful in the preparation of a pharmaceutical or diagnostic composition for the treatment, reduction or prevention of a disease associated with amyloidosis and / or formation of amyloid plaques. An example of such a disease is Alzheimer's disease. In the context of the present invention, it was surprisingly discovered that purified antibody molecules, wherein at least one antigen-binding site comprises N-linked glycosylation in the variable region of the heavy chain, is particularly useful, for example, in the reduction of amyloid plaques. Furthermore, it has been discovered in the context of the invention that the glycosylated antibodies or the antibody compositions provided herein are particularly useful and effective to cross the blood-brain barrier / hematoencephalic border in vivo illustrated by the efficient binding of the plates. This is very different from the teachings of the previous art. WO 03/016466 discloses antibodies that are specifically modified to lack an N-glycosylation site in the heavy chain and it is disclosed that glycosylation in the structure of the variable region has a negative effect on the binding affinity of the antibodies It is described in the prior art that the anti-Aβ antibody described in a deglycosylated form of the variable CDR2 region of the heavy chain has a markedly greater affinity with the synthetic Aβ peptide and purified in vi tro. Accordingly, the present invention relates to an improved purified antibody molecule or an antibody preparation, in particular, a preparation of antibody molecules that are directed against the Aβ4 / Aβ peptide (Amyloid β) and that it is highly effective in vivo. The improvement of the antibody molecule preparation / antibody preparation herein lies in the provision of purified antibody molecules comprising an N-glycosylation in at least one of their variable regions in the heavy chain, for example, in the region CDR2 of said variable region of the heavy chain. As mentioned above, this differs from the prior art according to WO 03/016466 which describes that said N-glycosylation has to be avoided in antibodies directed against, for example, Aβ. Examples of the antibody molecule of the present invention are immunoglobulin molecules, for example, IgG molecules. IgGs are characterized by comprising two light chains and two heavy chains (illustrated, for example, in Figure 14) and these molecules comprise two antigen-binding sites. Said antigen-binding sites comprise "variable regions", composed of parts of the heavy chains (VH) and parts of the light chains (VL). The antigen binding sites are formed by juxtaposition of the VH and VL domains. For general information on antibody molecules or immunoglobulin molecules see also common texts, such as Cellular and Molecular Immunology, W.B. Sounders Company (2003). In one aspect, for example, in the provision of an immunoglobulin molecule characterized in the present invention, an antibody is described wherein an antigen-binding site comprises a glycosylated asparagin (Asn) in the corresponding variable region of the heavy chain (VH ). Said antibody is referred to herein as "monoglycosylated ANTIBODY", see also Figure 14. In another aspect, an immunoglobulin molecule is provided where both antigen-binding sites comprise a glycosylated asparagine (Asn) in the variable region of the heavy chains (VH) corresponding. Bliss antibody molecule is hereinafter referred to as "double glycosylated ANTIBODY", see Figure 14. An immunoglobulin where no antigen binding site comprises a glycosylated asparagin (Asn) in the variable region of the heavy chain (VH) is referred to as "ANTIBODY" not glycosylated. " The monoglycosylated ANTIBODY, the double glycosylated ANTIBODY and the non-glycosylated ANTIBODY may comprise identical amino acid sequences or different amino acid sequences. The term "ANTIBODY" therefore comprises antibody molecules, in particular, antibody molecules produced in a recombinant manner, such as immunoglobulins. However, as described below, the phrase "ANTIBODY molecule (s)" also comprises known isoforms and modifications of immunoglobulins, such as single chain antibodies or single chain Fv fragments (secAB / scFv) or antibody structures. bispecific, and said isoforms and modifications are characterized by comprising at least one glycosylated V H region as defined herein. A specific example of said isoform or modification may be an antibody (single chain) in the VH-VL or VL-VH format, wherein said VH comprises the glycosylation as described herein. Bispecific scFvs are also contemplated, for example, in the format VH-VL-VH-VL, VL-VH-VH-VL, VH-VL-VL-VH, which comprise the glycosylation that is described herein in the CDR region -2.
In the context of the present invention, the term "ANTIBODY" is used in uppercase in order to provide better clarity. However, the term "antibody" used is also used in the context of the present application. The terms "ANTIBODY" / "ANTIBODIES" / "antibody" and "antibodies" are used interchangeably The phrases "monoglycosylated antibody" and "double glycosylated ANTIBODY" refer herein to "isoforms of glycosylated ANTIBODIES." A purified antibody molecule , characterized in that at least one antigen-binding site comprises a glycosylated asparagin (Asn) in the variable region of the heavy chain (VH), is a monoglycosylated ANTIBODY that is free or to a lesser degree associated with an isoform selected from an ANTIBODY double glycosylated and a non-glycosylated ANTIBODY, ie, a "purified monoglycosylated ANTIBODY." A double glycosylated ANTIBODY in the context of the present invention is free or to a lesser extent associated with an isoform selected from a monoglycosylated ANTIBODY and an unglycosylated ANTIBODY, that is, a "double glycosylated purified ANTIBODY." The phrase "that is free or in a minor g "indicates" the complete absence of the other respective (glycosylation) isoforms or the presence of another (glycosylated) isoform at a maximum concentration of 10%, for example, not more than 5%, for example, not more than 4% , for example, of 3% maximum, for example, 2% maximum, per example, maximum 1%, for example, 0.5% maximum, for example, 0.3% maximum, for example, 0.2% maximum. More information is provided later in the attached examples. In the context of the present invention, the phrase "monoglycosylated antibody (s)" or "monoglycosylated (S) ANTIBODY (S)" refers to antibody molecules comprising an N-glycosylation in a (VH) region of an individual antibody molecule, for example, of an immunoglobulin, for example, an IgG, for example, of an IgGl. For example, said "mono-glycosylated form" comprises a glycosylation in a variable region of the heavy chain, for example, at the position of asparagine "Asn 52" as defined below. This "glycosylated IgGl form or monoglycosylated isoform" may also comprise, as illustrated herein, glycosylation at the well conserved glycosylation site in the Fc part, for example, asparagine Asn 306 in the non-variable Fc part. The phrase "double glycosylated antibody (s)" or "double glycosylated (S) ANTIBODY (S)" in the meaning of the present invention comprises glycosylation as defined herein in both variable regions of the region of the Heavy chain (VH). Again, this "double glycosylated form" comprises a glycosylation in the variable region of both heavy chains, in particular, in the position of asparagine Asn 52 which is described below and as exemplified in the appended examples. This double glycosylated IgGl form or double glycosylated isoform "may also comprise, as illustrated herein, glycosylation at the well conserved glycosylation site in the non-variable / constant Fc part, in particular at position 306 of the immunoglobulin The accompanying Figure 14 illustrates the corresponding antibody molecules.Antibodies lacking such post-adductional modification in the variable region, for example, in both variable regions of the heavy chain (both regions (VH)) are considered, in the In the context of the present invention, a "non-glycosylated form" which does not comprise glycosylation in the variable region of the heavy chain, However, this "non-glycosylated form" can comprise (a) glycosylation (s) in the constant region (region- C) of the antibody, for example, and more commonly at the well-conserved glycosylation site of part Fc, in particular, asparagine (Asn) 306 in the non-variable / constant Fc part as defined herein, see also SEQ ID NO: 6. Asparagin (Asn) glycosylated in the variable region of the heavy chain ( VH) can be found in the complementarity determining region 2 (CDR2 region). Said glycosylated asparagine (Asn) in the variable region of the heavy chain (VH) can be found at position 52 of the variable region as defined below and is shown in SEQ ID No. 2 (or at position 52 of SEQ ID NO: 6 that it also comprises the Fc part of a heavy chain of antibodies as disclosed herein). The isoforms of ANTIBODIES may also comprise (a) additional glycosylation (s) in the constant / non-variable part of the antibody molecules, for example, in the Fc part of an IgG, for example, in the Fc part in an IgGl. Said glycosylation in the Fc part relates to a well conserved glycosylation, characterized by located on Asn306 position of the heavy chain, for example, according to the following sequence: QVELVESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSAINASGTRTYYA DSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGKGNTHKPYGYVRYFDVWGQGTLV TVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVL QSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELL GGPSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQY NSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDE LTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQ QGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO : 6). This sequence is also described herein and the CDRs, CH regions, heavy regions as well as two N-glycosylation sites (N52 and N306) are indicated: QVELVESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVS ^ .INASGTRTYYADSVKGJRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR GKGNTHKPYGYVRYFDVWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDY FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTK VDKKVEPJSCDXTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDP EVKFNWYVDGVEVHNAKTKPREEOYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPI EKTISKAKGQPREPQ TLPP TPPVLDSDGSFFLYSKLTVDK ^ (SEQ ID NO: 6). in box |: CDRl, 2, 3 underlined: CH1 in italics: double underlined joint: CH2 underlined..with., l.line ^ of together ^^ CH3 in bold N: N-linked glycosylation sites The IgG-Fc region of the antibodies of the present invention may be a homodimer composed of regions of articulation with disulfide bonds between the chains, glycosylated CH 2 domains, having oligosaccharide attached to N in asparagine 306 (Asn-306) of the CH2 and CH3 domains non-covalently paired. The glycosylation oligosaccharide in Asn-306 is of the complex biantennary type and may comprise a central heptasaccharide structure with variable addition of the sugars from external arms. The oligosaccharide influences or determines the structure and function Fc (Jefferis (1998) Immunol Rev. 163, 50-76). Effector functions, including the particular specific IgG-Fc / effector ligand interactions, are mentioned in (Jefferis (2002) Immunol Lett 82 (1-2), 57-65 and Krapp (2003) J Mol Biol. 325 (5 ), 979-89). This Asn 306 in the preserved Fc position corresponds to the "Asn-297" in the Kabat-system (Kabat (1991) Sequences of Proteins of Immunological Interest, 5th Ed., Public Health Service, National Institutes of Health, Bethesda MD). Exemplified heavy chain may be encoded by the following sequence: caggtggaattggtggaaagcggcggcggcctggtgcaaccgggcggcagcctgcgtctga gctgcgcggcctccggatttacctttagcagctatgcgatgagctgggtgcgccaagcccc tgggaagggtctcgagtgggtgagcgctattaatgcttctggtactcgtacttattatgct gattctgttaagggtcgttttaccatttcacgtgataattcgaaaaacaccctgtatctgc aaatgaacagcctgcgtgcggaagatacggccgtgtattattgcgcgcgtggtaagggtaa tactcataagccttatggttatgttcgttattttgatgtttggggccaaggcaccctggtg acggttagctcagcctccaccaagggtccatcggtcttccccctggcaccctcctccaaga gcacctctgggggcacagcggccctgggctgcctggtcaaggactacttccccgaaccggt cagtcctcaggactctactccctcagcagcgtggtgaccgtgccctccagcagcttgggca gacggtgtcgtggaactcaggcgccctgaccagcggcgtgcacaccttcccggctgtceta cccagacctacatctgcaacgtgaatcacaagcccagcaacaccaaggtggacaagaaagt tgagcccagatatcgtgcgatatcgtgcaatcttgtgacaaaactcacacatgcccaccgt gcccagcacctgaactcctggggggaccgtcagtcttcctcttccccccaaaacccaagga caceetcatgatctcccggacccctgaggtcacatgcgtggtggtggacgtgagccacgaa gaccctgaggtcaagttcaactggtacgtggacggcgtggaggtgcataatg ccaagacaa agccgcgggaggagcagtacaacagcacgtaccgggtggtcagcgtcctcaccgtcctgca ccaggactggctgaatggcaaggagtacaagtgcaaggtctccaacaaagccctcccagcc cccatcgagaaaaccatctccaaagccaaagggcagccccgagaaccacaggtgtacaece tgcccccatcccgggatgagctgaccaagaaccaggtcagcctgacctgcctggtcaaagg cttctatcccagcgacatcgccgtggagtgggagagcaatgggcagccggagaacaactac aagaccacgcctcccgtgctggactccgacggctccttcttcctctacagcaagctcaccg tggacaagagcaggtggcagcaggggaacgtcttctcatgctccgtgatgcatgaggctct gcacaaccactacacgcagaagagcctctccctgtctccgggtaaatga (SEQ ID NO: 5). Said heavy sequence may also comprise (in particular, during its recombinant production) additional sequences, such as "leader sequences". A corresponding example is encoded by the following sequence: atgaaacacctgtggttcttcctcctgctggtggcagctcccagatgggtcctgtcc (followed by) caggtggaattggtggaaagcggcggcggcctggtgcaaccgggcggcagcctgcgtctga gctgcgcggcctccggatttacctttagcagctatgcgatgagctgggtgcgccaagcccc tgggaagggtctcgagtgggtgagcgctattaatgcttctggtactcgtacttattatgct gattctgttaagggtcgttttaccatttcacgtgataattcgaaaaacaccctgtatctgc aaatgaacagcctgcgtgcggaagatacggccgtgtattattgcgcgcgtggtaagggtaa tactcataagccttatggttatgttcgttattttgatgtttggggccaaggcaccctggtg acggttagctcagcctccaccaagggtccatcggtcttccccctggcaccctcctccaaga gcacctctgggggcacagcggccctgggctgcctggtcaaggactacttccccgaaccggt cagtcctcaggactctactccctcagcagcgtggtgaccgtgccctccagcagcttgggca gacggtgtcgtggaactcaggcgccctgaccagcggcgtgcacaccttcccggctgtceta cccagacctacatctgcaacgtgaatcacaagcccagcaacaccaaggtggacaagaaagt tgagcccaaatcttgtgacaaaactcacacatgcccaccgtgcccagcacctgaactcctg gggggaccgtcagtcttcctcttccccccaaaacccaaggacaccctcatgatctcccgga cccctgaggtcacatgcgtggtggtggacgtgagccacgaagaccctgaggtc aagttcaá ctggtacgtggacggcgtggaggtgcataatgccaagacaaagccgcgggaggagcagtac aacagcacgtaccgggtggtcagcgtcctcaccgtcctgcaccaggactggctgaatggca aggagtacaagtgcaaggtctccaacaaagccctcccagcccccatcgagaaaaccatctc caaagccaaagggcagccccgagaaccacaggtgtacaecetgcccccatcccgggatgag ctgaccaagaaccaggtcagcctgacctgcctggtcaaaggcttctatcccagcgacatcg ccgtggagtgggagagcaatgggcagccggagaacaactacaagaccacgcctcccgtgct ggactccgacggctccttcttcctctacagcaagctcaccgtggacaagagcaggtggcag agagcctctccctgtctccgggtaaatga caggggaacgtcttctcatgctccgtgatgcatgaggctctgcacaaccactacacgcaga (SEQ ID NO: 25) the corresponding amino acid sequence would be: MKHLWFFLLLVAAPRWVLS (followed by) QVELVESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSAINASGTRTYYA DSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGKGNTHKPYGYVRYFDVWGQGTLV TVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVL QSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELL GGPSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQY NSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDE LTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQ QGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 26). The above sequence also comprises a "signal peptide" and said signal peptide is proteolytically cleaved by the host signal peptidase during the secretory path, during the production of the antibody molecules of the invention in host cells, such as CHO cells. Alternatively, said heavy chain can be encoded by a nucleic acid sequence that is optimized for recombinant production, as exemplified in the following sequence: 1 atggagtttg ggctgagctg ggttttcctc gttgctcttt taagaggtga 51 ttcatggaga aatagagaga ctgagtgtga gtgaacatga gtgagaaaaa 101 ctggatttgt gtggcatttt ctgataacgg tgtccttctg tttgcaggtg 151 tccagtgt followed by: ca ggtggagctg gtggagtctg ggggaggcct ggtccagcct 201 ggggggtccc tgagactctc ctgtgcagcg tctggattca ccttcagtag 251 ctatgccatg agctgggtcc gccaggctcc aggcaagggg ctcgagtggg 301 tgtccgccat aaacgccagc ggtacccgca cctactatgc agactccgtg 351 aagggccgat tcaccatctc cagagacaat tccaagaaca cgctgtatct 401 gcaaatgaac agcctgagag ccgaggacac ggctgtgtat tactgtgcga 451 gaggcaaggg gaacacccac aagccctacg gctacgtacg ctactttgac 501 gtgtggggcc aaggaaccct ggtcaccgtc tcctcaggtg agtcctcaca 551 acctctctcc tgcggccgca gcttgaagtc tgaggcagaa tcttgtccag 601 ggtctatcgg actcttgtga gaattagggg ctgacagttg atggtgacaa 651 tttcagggtc agtgactgtc tggtttctct gaggtgagac tggaatatag 701 gtcaccttga agactaaaga ggggtccagg ggcttttctg cacaggcagg 751 gaacagaatg tggaacaatg acttgaatgg ttgattcttg tgtgacacca 801 agaattggca taatgtctga gttgcccaag ggtgatctta gctagactct 851 ggggtttttg tcgggtacag aggaaaaacc cactattgtg attactatgc 901 tatggactac tggggtcaag gaacctcagt caccgtctcc tcaggtaaga 951 atggcctctc caggtcttta tttttaacct ttgttatgga gttttctgag 1001 cattgcagac taatcttgga tatttgccct gagggagccg gctgagagaa 1051 gttgggaaat aaatctgtct agggatctca gagcctttag gacagattat 1101 ctccacatct ttgaaaaact aagaatctgt gtgatggtgt tggtggagtc 1151 cctggatgat gggataggga ctttggaggc tcatttgagg gagatgctaa 1201 aacaatccta tggctggagg gatagttggg gctgtagttg gagattttca 1251 gtttttagaa tgaagtatta gctgcaatac ttcaaggacc acctctgtga 1301 caaccatttt atacagtatc caggcatagg gacaaaaagt ggagtggggc 1351 actttcttta gatttgtgag gaatgttcca cactagattg tttaaaactt 1401 catttgttgg aaggagctgt cttagtgatt gagtcaaggg agaaaggcat 1451 ctagcctcgg tctcaaaagg gtagttgctg tctagagagg tctggtggag 1501 cctgcaaaag tccagctttc aaaggaacac agaagtatgt gtatggaata 1551 ttagaagatg ttgcttttac tcttaagttg gttcctagga aaaatagtta 1601 aatactgtga ctttaaaatg tgagagggtt ttcaagtact cattttttta 1651 aatgtccaaa atttttgtca atcaatttga ggtcttgttt gtgtagaact 1701 gacattactt aaagtttaac cgaggaatgg gagtgaggct ctctcatacc 1751 ctattcagaa ctgactttta acaataataa attaagttta aaatattttt 1801 aaatgaattg agcaatgttg agttgagtca agatggccga tcagaaccgg 1851 aacacctgca gcagctggca ggaagcaggt catgtggcaa ggctatttgg 1901 ggaagggaaa ataaaaccac taggtaaact tgtagctgtg gtttgaagaa 1951 gtggttttga aacactctgt ccagccccac caaaccgaaa gtccaggctg 2001 agcaaaacac cacctgggta atttgcattt ctaaaataag ttgaggattc 2051 agccgaaact ggagaggtcc tcttttaact tattgagttc aaccttttaa 2101 ttttagcttg agtagttcta gtttccccaa acttaagttt atcgacttct 2151 aaaatgtatt tagaattcga gctcggtaca gctttctggg gcaggccagg 2201 cctgaccttg gctttggggc agggaggggg ctaaggtgag gcaggtggcg 2251 ccagcaggtg cacacccaat gcccatgagc ccagacactg gacgctgaac 2301 ctcgcggaca gttaagaacc caggggcctc tgcgcctggg cccagctctg 2351 tcccacaccg cggtcacatg gcaccacctc tcttgcagcc tccaccaagg 2401 gcccatcggt cttccccctg gcaccctcct ccaagagcac ctctgggggc 2451 acagcggccc tgggctgcct ggtcaaggac tacttccccg aaccggtgac 2501 ggtgtcgtgg aactcaggcg ccctgaccag cggcgtgcac accttcccgg 2551 ctgtcctaca gtcctcagga ctctactccc tcagcagcgt ggtgaccgtg 2601 ccctccagca gcttgggcac ccagacctac atctgcaacg tgaatcacaa 2651 gcccagcaac accaaggtgg acaagaaagt tggtgagagg ccagcacagg 2701 gagggagggt gtctgctgga agccaggctc agcgctcctg cctggacgca 2751 tcccggctat gcagccccag tccagggcag caaggcaggc cccgtctgcc 2801 tcttcacccg gagcctctgc ccgccccact catgctcagg gagagggtct 2851 tctggctttt tcccaggctc tgggcaggca caggctaggt gcccctaacc 2901 caggccctgc acacaaaggg gcaggtgctg ggctcagacc tgccaagagc 2951 catatccggg aggaccctgc ccctgaccta agcccacccc aaaggccaaa 3001 ctctccactc cctcagctcg gacaccttct ctcctcccag attccagtaa 3051 ctcccaatct tctctctgca gagcccaaat cttgtgacaa aactcacaca 3101 tgcccaccgt gcccaggtaa gccagcccag gcctcgccct ccagctcaag 3151 gcgggacagg tgccctagag tagcctgcat ccagggacag gccccagccg 3201 ggtgctgaca cgtccacctc catctcttcc tcagcacctg aactcctggg 3251 gggaccgtca gtcttcctct tccccccaaa acccaaggac accctcatga 3301 tctcccggac ccctgaggtc acatgcgtgg tggtggacgt gagccacgaa 3351 gaccctgagg tcaagttcaa ctggtacgtg gacggcgtgg aggtgcataa 3401 tgccaagaca aagccgcggg aggagcagta caacagcacg taccgtgtgg 3451 tcagcgtcct caccgtcctg caccaggact ggctgaatgg caaggagtac 3501 aagtgcaagg tctccaacaa agccctccca gcccccatcg agaaaaccat 3551 aaaggtggga ctccaaagcc gcgagggcca cccgtggggt catggacaga 3601 ggccggctcg gcccaccc tc tgccctgaga gtgaccgctg taccaacctc 3651 tgtccctaca gggcagcccc gagaaccaca ggtgtacacc ctgcccccat 3701 cccgggatga gctgaccaag aaccaggtca gcctgacctg cctggtcaaa 3751 ggcttctatc ccagcgacat cgccgtggag tgggagagca atgggcagcc 3801 ggagaacaac tacaagacca cgcctcccgt gctggactcc gacggctcct 3851 tcttcctcta cagcaagctc accgtggaca agagcaggtg gcagcagggg 3901 aacgtcttct catgctccgt gatgcatgag gctctgcaca accactacac 3951 ctctccctgt gcagaagagc ATGA ccccgggcaa (SEQ ID NO: 2. 3' The "alternative" protein sequence as shown above as SEQ ID NO: 23 comprises the same coding sequence that the first alternative, but in a slightly different genomic organization, as additional introns and a "leader sequence" / "signal sequence" slightly different. Said "leader sequence" may also comprise, as indicated above, an intron (is) (additional). The person skilled in the art is in a position to deduce, in the sequence shown here, the corresponding exon / intron structure by conventional methods. The exemplified antibody described herein may also comprise a light chain and said light chain may comprise or have the following amino acid sequence DIVLTQSPATLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPRLLIYGASSRATGVPA RFSGSGSGTDFTLTISSLEPEDFATYYCLQIYNMPITFGQGTKVEIKRTVAAPSVFIFPPS DEQLKSGTASWCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLS KADYEKHKVYACEVTHQGLSSPVTKSFNRGEC (SEQ ID NO: 8) that can be encoded by the following sequence of acids nucleic: gatatcgtgctgacccagagcccggcgaccctgagcctgtctccgggcgaacgtgcgaccc tgagctgcagagcgagccagagcgtgagcagcagctatctggcgtggtaccagcagaaacc aggtcaagcaccgcgtctattaatttatggcgcgagcagccgtgcaactggggtcccggcg cgttttagcggctctggatccggcacggattttaccctgaccattagcagcctggaacctg aagactttgcgacttattattgccttcagatttataatatgcctattacctttggccaggg tacgaaagttgaaattaaacgtacggtggctgcaccatctgtcttcatcttcccgccatct gatgagcagttgaaatctggaactgcctctgttgtgtgcctgctgaataacttctatccca gagaggccaaagtacagtggaaggtggataacgccctccaatcgggtaactcccaggagag tgtcacagagcaggacagcaaggacagcacctccacaccacacctgacgctgagc aaagcagactacgagaaacacaaagtctacgcctgcgaagtcacccatcagggcctgagct cgcccgtcacaaagagcttcaacaggggagagtgttag (SEQ ID NO: 7). Also, the "light chain" of the exemplified antibody described herein may comprise a "leader sequence" that is particularly useful in technical production. A corresponding sequence can be (Or may comprise, for example, in a vector system) the following sequence: atggtgttgcagacccaggtcttcatttctctgttgctctggatctctggtgcctacggg (followed by) gatatcgtgctgacccagagcccggcgaccctgagcctgtctccgggcgaacgtgcgaccc tgagctgcagagcgagccagagcgtgagcagcagctatctggcgtggtaccagcagaaacc aggtcaagcaccgcgtctattaatttatggcgcgagcagccgtgcaactggggtcccggcg cgttttagcggctctggatccggcacggattttaccctgaccattagcagcctggaacctg aagactttgcgacttattattgccttcagatttataatatgcctattacctttggccaggg tacgaaagttgaaattaaacgtacggtggctgcaccatctgtcttcatcttcccgccatct gatgagcagttgaaatctggaactgcctctgttgtgtgcctgctgaataacttctatccca gagaggccaaagtacagtggaaggtggataacgccctccaatcgggtaactcccaggagag tgtcacagagcaggacagcaaggacagcacctacagcctcagcagcaccctgacgctgagc aaagcagactacgagaaacacaaagtctacgcctgcgaagtcacccatcagggcctgagct cgcccgtcacaaagagcttcaacaggggagagtgttag (SEQ ID NO: 27). This sequence encodes the following amino acid sequence MVLQTQVFISLLLWISGAYG (followed by) DIVLTQSPATLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPRLLIYGASSRATGVPA RFSGSGSGTDFTLTISSLEPEDFATYYCLQIYNMPITFGQGTKVEIKRTVAAPSVFIFPPS DEQLKSGTASWCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLS KADYEKHKVYACEVTHQGLSSPVTKSFNRGEC (SEQ ID NO: 28). Alternatively, said light chain can also be encoded by a nucleic acid sequence that is optimized for recombinant production exemplified by the following sequence: 1 atggacatga gggtcctcgc tcagctcctg gggctcctgc tgctctgttt 51 cccaggtaag gatggagaac actagcagtt tactcagccc agggtgctca 101 gtactgcttt actattcagg gaaattctct tacaacatga ttaattgtgt 151 ggacatttgt ttttatgttt ccaatctcag gcgccagatg t followed by gatatcgtg 201 ttgacgcagt ctccagccac cctgtctttg tctccagggg aaagagccac 251 cctctcctgc cgggccagtc agagtgttag cagcagctac ttagcctggt 301 accagcagaa acctggccag gcgcccaggc tcctcatcta tggcgcatcc 351 agcagggcca ctggcgtgcc agccaggttc agtggcagtg ggtctgggac 401 ctcaccatca agacttcact gcagcctgga gcctgaagat ttcgcgacct 451 attactgtct gcagatttac aacatgccta tcacgttcgg ccaagggacc 501 tcaaacgtga aaggtggaaa gtagaattta aactttgcgg ccgcctagac 551 gagatttgga gtttaagtgg ggggatgagg aatgaaggaa cttcaggata 601 gaaaagggct gaagtcaagt tcagctccta aaatggatgt gggagcaaac 651 tttg Aagata aactgaatga cccagaggat gaaacagcgc agatcaaaga 701 gctctgagaa ggggcctgga gagaaggaga ctcatccgtg ttgagtttcc 751 acaagtactg tcttgagttt tgcaataaaa gtgggatagc agagttgagt 801 gagccgtagg ctgagttctc tcttttgtct cctaagtttt tatgactaca 851 aaaatcagta gtatgtcctg aaataatcat taagctgttt gaaagtatga 901 ctgcttgcca tgtagatacc atgtcttgct gaatgatcag aagaggtgtg 951 actcttattc taaaatttgt cacaaaatgt caaaatgaga gactctgtag 1001 gaacgagtcc ttgacagaca gctcaagggg tttttttcct ttgtctcatt 1051 tctacatgaa agtaaatttg aaatgatctt ttttattata agagtagaaa 1101 tacagttggg tttgaactat atgttttaat ggccacggtt ttgtaagaca 1151 tttggtcctt tgttttccca gttattactc gattgtaatt ttatatcgcc 1201 agcaatggac tgaaacggtc cgcaacctct tctttacaac tgggtgacct 1251 cgcggctgtg ccagccattt ggcgttcacc ctgccgctaa gggccatgtg 1301 aacccccgcg gtagcatccc ttgctccgcg tggaccactt tcctgaggca 1351 cagtgatagg aacagagcca ctaatctgaa gagaacagag atgtgacaga 1401 ctacactaat gtgagaaaaa caaggaaagg gtgacttatt ggagatttca 1451 gaaataaaat gcatttatta ttatattccc ttattttaat tttctattag 1501 ggaattagaa agggcataaa ctgctttatc cagtgttata ttaaaagctt 1551 aatgtatata atcttttaga ggtaaaatct acagccagca aaagtcatgg 1601 taaatattct ttgactgaac tctcactaaa ctcctctaaa ttatatgtca 1651 tattaactgg ttaaattaat ataaatttgt gacatgacct taactggtta 1701 ggtaggatat ttttcttcat gcaaaaatat gactaataat aatttagcac 1751 aaaaatattt cccaatactt taattctgtg atagaaaaat gtttaactca 1801 gctactataa tcccataatt ttgaaaacta tttattagct tttgtgtttg 1851 acccttccct agccaaaggc aactatttaa ggacccttta aaactcttga 1901 aactacttta gagtcattaa gttatttaac cacttttaat tactttaaaa 1951 tgatgtcaat tcccttttaa ctattaattt attttaaggg gggaaaggct 2001 gctcataatt ctattgtttt tcttggtaaa gaactctcag ttttcgtttt 2051 tactacctct gtcacccaag agttggcatc tcaacagagg ggactttccg 2101 agaggccatc tggcagttgc ttaagatcag aagtgaagtc tgccagttcc 2151 tcccaggcag gtggcccaga ttacagttga cctgttctgg tgtggctaaa 2201 aattgtccca tgtggttaca aaccattaga ccagggtctg atgaattgct 2251 cagaatattt ctggacaccc aaatacagac cctggcttaa ggccctgtcc 2301 atacagtagg tttagcttgg ggaagccata ctacaccaaa cagaggctaa 2351 tatcagagta ttcttggaag agacaggaga aaatgaaagc cagtttctgc 2401 tcttacctta tgtgcttgtg ttcagactcc caaacatcag gagtgtcaga 2451 taaactggtc tgaatctctg tctgaagcat ggaactgaaa agaatgtagt 2501 ttcagggaag aaaggcaata gaaggaagcc tgagaatgatcaattct 2551 aaactctgag ggggtcggat gggcca ttctttgcct aaagcattga 2601 aggtcagaaa gtttactgca agcatgcaaa gccctcagaa tggctgcaaa 2651 gagctccaac aaaacaattt agaactttat taaggaatag ggggaagcta 2701 ggaagaaact caaaacatca agattttaaa tttctt ggtctccttg 2751 ctataattat ctgggataag catgctgttt tctgtctgtc cctaacatgc 2801 tccgcaaaca cctgtgatta acacacccaa gggcagaact ttgttactta 2851 aacaccatcc tgtttgcttc tttcctcagg aactgtggct gcaccatctg 2901 tcttcatctt cccgccatct gatgagcagt tgaaatctgg aactgcctct 2951 gttgtgtgcc tgctgaataa cttctatccc agagaggcca aagtacagtg 3001 gaaggtggat acctcc aatcgggtaa ctcccaggag agtgtcacag 3051 agcaggacag caaggacagc acctacagcc tcagcagcac cctgtg 3101 agcaaagcag actg aa acacaaagtc tctgcg aagtcaccca 3151 tcagggcctg agctcgcccg tcacaaagag cttcaacagg ggagagtgtt 3201 ag (SEQ ID NO: 24) The above "sequence" for an exemplified light chain also has a slightly different genomic structure. This "alternative sequence" comprises different and / or additional introns. Accordingly, embodiments describing a "light chain" are applied herein mutatis mutandis. In the context of the present invention, the phrase "antibody molecule" refers to molecules of total immunoglobulins, for example, IgMs, IgDs, IgEs, IgAs or IgGs, such as IgG1, IgG2, IgG2b, IgG3 or IgG4, as well as parts of said immunoglobulin molecules, such as Fab fragments, Fabd fragments F (ab) fragments 2, chimeric F (ab) 2 fragments or chimeric Fab ', chimeric Fab fragments or isolated VH or CDR regions (said isolated V or CDR regions are, for example, integrated or modified in the corresponding "structure (s)") (s)). The phrase "antibody molecule" also comprises diabodies and molecules comprising an antibody Fc domain as at least one part attached vehicle / antigen-binding peptide, for example, peptibodies as described in WO 00/24782 . Accordingly, and in the context of the present invention, the phrase "heavy chain variable region (VH)" is not limited to a variable region in a total immunoglobulin, but refers to the corresponding parts of said variable region of the heavy chain (VH), as the CRDs, either alone or in combination of CDR1, 2 and / or 3 or the corresponding "structure" of the variable region. Therefore, an antibody molecule of the present invention can also be an antibody structure comprising, as an antigen-binding site, the CDRs or at least one CDR of a given variable region of the glycosylated heavy chain (VH). Said corresponding part of said variable region of the heavy chain (VH) in the antibody structure of the invention is glycosylated as defined herein, example, it comprises a glycosylated asparagine (Asn) at the antigen-binding site. An example of said "isolated part", a variable region of the heavy chain (VH), is the CDR2 region exemplified herein comprising SEQ ID NO: 12 (or encoded by a nucleic acid sequence as shown in FIG. SEQ ID NO: 11). further, the phrase "antibody molecule" refers to modified and / or altered antibody molecules, such as chimeric, humanized or fully humanized antibodies. Said molecules of "fully humanized antibodies" are also characterized and described as "fully human" antibodies. All of these antibodies can be generated by methods known in the art. For example, through phage display technology, can recombinant antibody molecules be generated due to the use of in vitro maturation that is the use of an immunoglobulin structure? human complete subclass-1 (IgGl) as described in Knappik (2000) J Mol Biol. 296 (1), 57-86, and Rauchenberger (2003) J Biol Chem. 278 (40), 38194-205. As documented in the appended examples, the term "antibody" refers, for example, to an IgG molecule and, for example, to an IgG1. The term also refers to modified or altered monoclonal or polyclonal antibodies, as well as to recombinantly or synthetically generated / synthesized antibodies. The term also refers to intact antibodies, as well as antibody fragments / parts thereof, such as light chains and separate weights, Fab, Fab / c, Fv, Fabd F (ab ') 2. The term "antibody molecule" also comprises antibody derivatives, bifunctional antibodies and antibody structures, such as single chain Fvs (scFv) or fusion proteins to antibodies. Catalytic and / or proteolytic antibodies comprising a glycosylated V H domain, for example, a glycosylated V H -CDR as defined herein, are also contemplated. The phrase "antibody molecule" also refers to antibody structures / recombinantly produced antibody molecules which may comprise, in addition to a specific (for example, against Aß / Aß), another specificity or additional specificity. Such structures may include, without limitation, "bispecific" or "trispecific" structures. Further information is provided on the phrase "antibody molecule" of the invention below. As indicated above, a single chain antibody, a chimeric antibody or a CDR-grafted antibody, an antibody-bivalent structure, an antibody-fusion protein, a cross-linked antibody or a synthetic antibody comprising the glycosylation that has been defined herein in at least one antigen-binding site, for example, in at least one variable region of a heavy chain defined herein and glycosylated. When, for example, single-chain antibodies are produced, the "variable region of "heavy chain" as defined herein is not limited to a heavy chain per se, but means that it also refers to the corresponding parts derived from a heavy chain of a complete antibody, eg, a complete immunoglobulin, such as a These parts may be the corresponding CDRs either alone or with parts of their corresponding structure.In addition, genetic variants of the immunoglobulin genes are also contemplated in the context of the present invention.The genetic variants, for example, of the heavy G chain of immunoglobulin subclass 1 (IgGl) may comprise the alotypic markers Glm (17) or Glm (3) in the CH1 domain, or the allotypic marker Glm (l) or Glm (no 1) in the CH3 domain. , an IgGl of the allotype Gm (17) (z) and Gm (l) (a) is preferably used.The antibody molecule of the invention also comprises modified or mutant antibodies, such as the mutant IgG with improved Fc-receptor binding or uada, or complementary activation. In one embodiment, the antibody provided in the present invention is a fully humanized antibody or a "fully human" antibody. Accordingly, the antibodies of the invention can also comprise inter-cloned antibodies, ie, antibodies comprising different regions of antibodies (e.g., CDR regions) of one or more parental or optimized affinity antibodies as described herein. These inter-cloned antibodies can be antibodies in several different structures, for example, an IgG structure, for example, an IgGl (human) structure or an IgG2a or IgG2b structure. For example, said antibody structure is a mammalian, e.g., human, structure. The domains in the light and heavy chains have the same general structure and each domain comprises four regions of the structure, whose sequences are relatively conserved, joined by three hypervariable domains known as complementarity determining regions (CDR1-3). As used herein, a "human framework region" refers to a region of structure that is substantially identical (approximately 85% or more usually 90-95% or more) to the framework region of a human immunoglobulin. of natural origin. The region of the structure of an antibody (e.g., the combined regions of the structure of the light and heavy chains) serves to position and align the CDRs. The CDRs are mainly responsible for the binding of an antigen to an epitope. It should be emphasized that not only can there be present, inter-cloned antibodies described herein in a preferred (human) antibody structure., but also antibody molecules comprising CDRs of the antibodies described herein can be introduced into an immunoglobulin structure. Examples of structures include IgG1, IgG2a and IgG2b. Most preferred are human structures and IgGl structures human antibodies, such as the heavy chain of an ANTIBODY as shown, inter alia, in SEQ ID NO: 6. In one embodiment, the isoforms of ANTIBODIES may comprise in a variable region of the heavy chain a CDR1 comprising the following amino acids: GFTFSSYAMS (SEQ ID NO: 10). Said CDR1 can be encoded by the following nucleic acid sequence: ggatttacctttagcagctatgcgatgagc (SEQ ID NO: 9). The isoforms of ANTIBODIES may comprise the following CDR2 in the variable region of the heavy chain: AINASGTRTYYADSVKG (SEQ ID NO: 12) (N: N-linked glycosylation site in Asn-52 of a complete heavy chain). Said CDR2 can be encoded by the following nucleic acid sequence: gctattaatgcttctggtactcgtacttattatgctgattctgttaagggt (SEQ ID NO: 11). The N-glycosylation according to the present invention is included, for example, in said CDR2 region and is located in the corresponding Asn52 of the variable region of the heavy chain, said variable region (VH) is encoded by a nucleic acid molecule as shown in the SEQ ID NO: 1 and has an amino acid sequence as shown in SEQ ID NO: 2.
In addition, the isoforms of ANTIBODIES may comprise in their variable region of the heavy chain a CDR3 comprising the following amino acid sequence: GKGNTHKPYGYVRYFDV (SEQ ID NO: 14). Said CDR3 can be encoded by the following nucleic acid sequence: ggtaagggtaatactcataagccttatggttatgttcgttattttgatgtt (SEQ ID NO: 13). The isoforms of ANTIBODIES may comprise a light chain (L) which may be characterized by the following CDRs: CDR1: RASQSVSSSYLA (SEQ ID NO: 16) agagcgagccagagcgtgagcagcagctatctggcg (SEQ ID NO: 15) CDR2: GASSRAT (SEQ ID NO: 18) ggcgcgagcagccgtgcaact (SEQ ID NO: 17) CDR3: LQIYNMPI (SEQ ID NO: 20) cttcagatttataatatgcctatt (SEQ ID NO: 19). The isoforms of ANTIBODIES may comprise additional potential glycosylation sites (which as known in the art comprise the Asn-X-Ser / Thr motifs) in the amino acid sequence of the heavy chains of the antibody, for example, at the glycosylation site well preserved in Asn 306 in the non-variable Fc part (corresponding to "Asn297" in the Kabat-system (Kabat (1991) Sequences of Proteins of Immunological Interest, 5th Ed., Public Health Service, National Institutes of Health, Bethesda Md.: National Center for Biotechnology Information, National Library of Medicine), said heavy chains are or comprise the sequence that has been previously provided, that is, in SEQ ID NO: 6 (encoded by SEQ ID NO: 5). In one embodiment of the present invention, the isoforms of ANTIBODIES are characterized in that at least one antigen binding site comprises a glycosylated asparagin (Asn) in the variable region of the heavy chain (VH), and said VH is encoded by (a) ) a nucleic acid comprising the nucleotide sequence shown in SEQ ID NO: l CAGGTGGAATTGGTGGAAAGCGGCGGCGGCCTGGTGCAACCGGGCGGCAGCCT GCGTCTGAGCTGCGCGGCCTCCGGATTTACCTTTAGCAGCTATGCGATGAGCT GGGTGCGCCAAGCCCCTGGGAAGGGTCTCGAGTGGGTGAGCGCTATTAATGCT TCTGGTACTCGTACTTATTATGCTGATTCTGTTAAGGGTCGTTTTACCATTTC ACGTGATAATTCGAAAAACACCCTGTATCTGCAAATGAACAGCCTGCGTGCGG AAGATACGGCCGTGTATTATTGCGCGCGTGGTAAGGGTAATACTCATAAGCCT TATGGTTATGTTCGTTATTTTGATGTTTGGGGCCAAGGCACCCTGGTGACGGT TAGCTCA (SEQ ID NO: 1); (b) a nucleic acid molecule encoding a polypeptide having the amino acid sequence shown in SEQ ID NO: 2 QVELVESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSAINA SGTRTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGKGNTHKP YGYVRYFDVWGQGTLVTVSS (SEQ ID NO: 2; "N" in bold represents the Asn defined in the present in the position 52 of the variable region of the heavy chain); (c) a nucleic acid molecule that hybridizes to the nucleic acid molecule of (a) or (b) and that encodes a polypeptide that is capable of binding to the β-A4 / Aβ4 peptide shown in the following sequence of amino acids DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO: 3) or a fragment thereof comprising at least 15 amino acids; (d) a nucleic acid molecule that hybridizes to the nucleic acid molecule of (a) or (b) and that encodes a polypeptide that is capable of binding to at least two regions in the β-A4 / Aβ4 peptide such as is shown in the following amino acid sequence DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO: 3) or to at least two regions of a fragment of SEQ ID NO: 3 comprising at least 15 amino acids where said two regions in the β-A4 Aβ4 peptide or its fragment comprises the amino acids in the 3 to 6 position and in the 18 to 26 position of the SEQ ID NO: 3; or (e) a nucleic acid sequence that degenerates to a nucleic acid sequence as defined in any of (a) to (d).
The skilled artisan knows that the phrase "nucleic acid molecule that hybridizes to the nucleic acid molecule of (a) or (b) and that encodes a polypeptide that is capable of binding to at least two regions in the β-peptide -A4 / Aβ4"as used herein refers to a coding strand of a double-stranded nucleic acid molecule, wherein the non-coding strand is hybridized to the above-identified nucleic acid molecule of (a) and (b). As stated above, the purified antibody molecule comprising the glycosylation of Asn defined herein can be characterized and described, inter alia, as an antibody molecule where the variable region comprising a glycosylated Asn is comprised in a heavy chain selected from the group comprising: (a) a heavy chain polypeptide encoded by a nucleic acid molecule as shown in SEQ ID NOS: 5, 23 or 25; (b) a heavy chain polypeptide having an amino acid sequence as shown in SEQ ID NO: 6 or 26; (c) a heavy chain polypeptide encoded by a nucleic acid molecule that hybridizes to the nucleic acid molecule of (a) and that encodes a polypeptide that is capable of binding to the β-A4 / Aβ4 peptide as shown in the next amino acid sequence DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO: 3) or a fragment thereof comprising at least 15 amino acids; or (d) a heavy chain polypeptide encoded by a nucleic acid molecule that hybridizes to the nucleic acid molecule of (a) and that encodes a polypeptide that is capable of binding to at least two regions on the β-A4 peptide / Aβ4 as shown in the following amino acid sequence DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO: 3) or at least two regions of a fragment of SEQ ID NO: 3 comprising at least 15 amino acids where said two regions in the β-peptide -A4 Aβ4 or its fragment comprises the amino acids in the 3 to 6 position and in the 18 to 26 position. The above identified antibody (for example an exemplified antibody of the invention) may also comprise an L chain with the following amino acid sequence: DIVLTQSPATLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPRLLIYGASSRATGVPA RFSGSGSGTDFTLTISSLEPEDFATYYCLQIYNMPITFGQGTKVEIKRTVAAPSVFIFPPS DEQLKSGTASWCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLS KADYEKHKVYACEVTH QGLSSPVTKSFNRGEC (SEQ ID NO: 22) or an L chain, for example, encoded by the following nucleic acid sequence: gatatcgtgctgacccagagcccggcgaccctgagcctgtctccgggcgaacgtgcgaccc tgagctgcagagcgagccagagcgtgagcagcagctatctggcgtggtaccagcagaaacc aggtcaagcaccgcgtctattaatttatggcgcgagcagccgtgcaactggggtcccggcg cgttttagcggctctggatccggcacggattttaccctgaccattagcagcctggaacctg aagactttgcgacttattattgccttcagatttataatatgcctattacctttggccaggg tacgaaagttgaaattaaacgtacggtggctgcaccatctgtcttcatcttcccgccatct gatgagcagttgaaatctggaactgcctctgttgtgtgcctgctgaataacttctatccca gagaggccaaagtacagtggaaggtggataacgccctccaatcgggtaactcccaggagag tgtcacagagcaggacagcaaggacagcacctacagcctcagcagcaccctgacgctgagc aaagcagactacgagaaacacaaagtctacgcctgcgaagtcacccatcagggcctgagct cgcccgtcacaaagagcttcaacaggggagagtgttag (SEQ ID NO: 21). As mentioned above, the purified antibody molecule comprising the glycosylation of Asn defined herein in the heavy chain may also comprise a light chain selected from the group consisting of: (a) a light chain polypeptide encoded by a molecule of nucleic acid as shown in SEQ ID Nos. 7, 21, 24 or 27; (b) a light chain polypeptide having an amino acid sequence as shown in SEQ ID NO: 8, 22 or 28; (c) a light chain polypeptide encoded by a nucleic acid molecule that hybridizes to the nucleic acid molecule of (a) and that encodes a polypeptide that is capable of binding to the β-A4 / Aβ4 peptide as shown in the following sequence of amino acids DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO: 3) or a fragment thereof comprising at least 15 amino acids; or (d) a light chain polypeptide encoded by a nucleic acid molecule that hybridizes to the nucleic acid molecule of (a) and that encodes a polypeptide that is capable of binding to at least two regions on the β-A4 peptide / Aβ4 as shown in the following amino acid sequence DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO: 3) or to at least two regions of a fragment of SEQ ID NO: 3 comprising at least 15 amino acids. The term "hybridization" or "hybridizes" as used herein in the context of nucleic acid molecules / DNA sequences can refer to hybridizations under astringent or non-astringent conditions. If not specified, the conditions are preferably non-astringent. Said hybridization conditions can be established according to the conventional protocols described, for example, in Sambrook, Russell Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y. (2001); Ausubel, Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N.Y. (1989), or Higgins and Hames (Eds.) Nucleic acid hybridization, a practical approach IRL Press Oxford, Washington DC, (1985). The establishment of the conditions is within the skill of the expert and can be determined in accordance with the protocols described in the art. Therefore, detection of only specific hybridization sequences will usually require stringent hybridization and wash conditions such as O.lxSSC, 0.1% SDS at 65 ° C. Non-astringent hybridization conditions for the detection of homologous or not exactly complementary sequences can be established in 6xSSC, 1% SDS at 65 ° C. As is known, the length of the probe and the composition of the nucleic acid to be determined constitute other parameters of the hybridization conditions. Note that variations in the above conditions can be achieved through the inclusion and / or substitution of alternating blocking reagents used to suppress background in the hybridization experiments. Typical blocking reagents include Denhardt's reagent, BLOTTO, heparin, denatured salmon sperm DNA, and commercially available patented formulations. The inclusion of specific blocking reagents may require modification of the hybridization conditions that have been described above, due to problems with compatibility. The hybridizing nucleic acid molecules also comprise fragments of the molecules described above. Such fragments may represent sequences of nucleic acids that encode a molecule of non-functional antibody or a non-functional fragment thereof or for a CDR as defined herein and having a length of at least 12 nucleotides, preferably at least 15, more preferably at least 18, more preferably at least 21 nucleotides, more preferably at least 30 nucleotides, even more preferably at least 40 nucleotides and more preferably at least 60 nucleotides. In addition, nucleic acid molecules that hybridize with any of the nucleic acid molecules also include complementary fragments, derivatives and allelic variants of these molecules. Additionally, a hybridization complex refers to a complex between two nucleic acid sequences by virtue of the formation of hydrogen bonds between the complementary G and C bases and between the complementary A and T bases; These hydrogen bonds can also be stabilized by stacking interactions between bases. The two complementary nucleic acid sequences have hydrogen bonds in an antiparallel configuration. A hybridization complex can be formed in solution (eg, Cot or Rot analysis) or between a nucleic acid sequence present in the solution and another nucleic acid sequence immobilized on a solid support (e.g., membranes, filters, chips, nails or glass slides to which, for example, cells have been fixed). The terms "complementary" or "complementarity" refer to the natural union of the polynucleotides under salt conditions and temperature allowed through base pairing. For example, the sequence "A-G-T" joins the complementary sequence "T-C-A". The complementarity between two single-stranded molecules can be "partial", where only some of the nucleic acids bind, or it can be complete when there is a complete complementarity between the single-stranded molecules. The degree of complementarity between the nucleic acid strands has significant effects on the efficiency and intensity of the hybridization between nucleic acid strands. This is of particular importance in amplification reactions, which depend on the binding between the nucleic acid strands. The phrase "hybridization sequences" preferably refers to sequences exhibiting a sequence identity of at least 40%, preferably at least 50%, more preferably at least 60%, even more preferably at least 70%, with particular preference at least 80%, more particularly at least 90%, even more particularly at least 95% and more preferably at least 97% identity with the nucleic acid sequence as described above encoding an antibody molecule. In addition, the phrase "hybridization sequences" preferably refers to sequences that encode an antibody molecule having a sequence identity of at least 40%, preferably at least 50%, more preferably at least 60%, even more preferably at least 70%, particularly at least 80%, more particularly at least 90%, even more particularly at least 95% and more preferably even at least 97% identity with an amino acid sequence of the antibody molecule as described above. In accordance with the present invention, the term "identical" or "percent identity" in the context of two or more nucleic acid or amino acid sequences refers to two or more sequences or subsequences that are the same, or that have a specified percentage of amino acid or nucleotide residues that are the same (eg, 60% or 65% identity, preferably 70-95% identity, more preferably at least 95% identity), when compared and aligned for maximum correspnce in a comparison window, or in a designated region measured using a sequence comparison algorithm known in the art, or by manual alignment and visual inspection. Sequences having, for example, 60% to 95% or more of sequence identity are considered substantially identical. This definition also applies to the complement of a test sequence. Preferably, the sequence identity exists in a region having a length of at least about 15 to 25 amino acids or nucleotides, more preferably, in a region having a length of about 50 to 100 amino acids or nucleotides. Those skilled in the art will know how to determine the percent identity between the sequences using, for example, algorithms such as those based on the program.
CLUSTALW (Thompson Nucí, Acids Res. 2 (1994), 4673-4680) or FASTDB (Brutlag Comp.App.Biosci., 6 (1990), 237-245) as is known in the art. Although the FASTDB algorithm typically does not consider the eliminations or additional internal mismatches in the sequences, that is, empty, in its calculations, this can be corrected manually to avoid an overestimation of% identity. However, CLUSTALW does not take into account the gaps of the sequences in their identity calculations. The BLAST and BLAST 2.0 algorithms are also available to those skilled in the art (Altschul, Nucí Acids Res. (1997), 3389-3402; Altschul, J. Mol. Evol. 36 (1993), 290-300; Altschul, J. Mol. Biol. 215 (1990), 403-410). The BLASTN program for nucleic acid sequences uses a word length (W) of 11, an expectation (E) of 10, M = 5 as preset values., N = 4, and a comparison of both chains. For the amino acid sequences, the BLASTP program uses a word length (W) of 3 as a pre-established value, and an expectation (E) of 10. The BLOSUM62 score matrix (Henikoff Proc. Nati. Acad. Sci., USA, 89, (1989), 10915) uses alignments (B) of 50, expectations (E) of 10, M = 5, N = 4 and a comparison of both chains. In addition, the present invention also relates to nucleic acid molecules whose sequence degenerates in comparison to the sequence of the hybridizing molecule described above. When used in accordance with the present invention, the phrase "degenerating as a result of the genetic code" means that, due to the redundancy of the genetic code, different nucleotide sequences code for the same amino acid. To determine whether an amino acid residue or nucleotide residue in a given antibody sequence corresponds to a certain position in the amino acid sequence or nucleotide sequence, for example, of any of SEQ ID Nos: 1, 5, 23 and 25, the person skilled in the art can use means and methods known in the art, for example, alignments, either manually or using computer programs such as those mentioned below in connection with the definition of the term "hybridization" and degree of homology. For example, BLAST 2.0 which means basic local alignment search tool (Basic Local Alignment Search Tool) BLAST (Altschul (1997), loe. Cit.; Altschul (1993), loe. Cit .; Altschul (1990), loe. cit.) can be used to search for local sequence alignments. BLAST, as discussed above, produces alignments of both the nucleotide and amino acid sequences to determine the similarity of the sequences. Due to the local nature of the alignments, BLAST is especially useful for determining exact matches or identifying similar sequences. The fundamental unit of performance of the BLAST algorithm is the High-scoring Segment Pair (HSP). An HSP is composed of two fragments of sequences of lengths arbitrary but equal, whose alignment is locally maximum and for which the alignment score meets or exceeds a threshold or discriminatory score established by the user. The BLAST method consists of searching the HSPs between an interrogation sequence and a sequence of databases, to evaluate the statistical significance of any found match and report only those matches that satisfy the threshold of significance selected by the user. The parameter E establishes the statistically significant threshold for reporting the sequence matches of the database. E is interpreted as the superior union of the expected frequency of opportunity of occurrence of an HSP (or set of HSPs) within the context of the entire search in the database. Any sequence in the database whose match satisfies E is reported on the output device of the program. Analogous computer techniques using BLAST (Altschul (1997), loc. Cit., Altschul (1993), loc. Cit., Altschul (1990), loe. Cit.) Are used to search for identical or related molecules in the database. of nucleotides such as in GenBank or EMBL. This analysis is much faster than hybridizations based on multiple membranes. In addition, the sensitivity of a computer search can be modified to determine if any particular match is categorized as exact or similar. The basis of the search is the score of the product that is defined as follows: % sequence identity x% maximum BLAST score 100 and takes into account both the degree of similarity between two sequences and the length of the sequence match. For example, with a product score of 40, the match will be exact with an error of 1-2%, and in 70, the match will be exact. Usually similar molecules are defined by selecting those that exhibit a product score that ranges between 15 and 40, although minor scores can identify related molecules. Another example for a program capable of generating sequence alignments is the CLUSTALW computer programs (Thompson, Nucí Acids Res. 2 (1994), 4673-4680) or FASTDB (Brutlag Comp.App.Biosci. 6 (1990), 237- 245), known in the art. In one embodiment, the present invention provides isoforms of glycosylated ANTIBODIES where the glycosylation at Asn in the VH region is selected from the group consisting of (a) a sugar structure of the biantennary complex type without fucosylation in the nucleus; (b) a sugar structure of the biantennarous hybrid type; (c) a sugar structure of the oligomeric biantennary type; Y (d) a biantennary structure of any of the structures provided in the attached Figure 5 or the Figure 27 attached. In an embodiment of the antibody / antibodies of the present invention, the corresponding sugar structure does not comprise fucosylation in the core. The corresponding N-glycosylation can be composed predominantly of sugar structures of the biantenary complex type (> 75%, mainly 80-90%) without fucosylation in the nucleus and highly sialidated with up to 80% of antennas. The secondary sugar structures belong to the hybrid biantennary type and the oligomannose type (< 25%), respectively, and are also shown in the attached Figures 5 and 27. The glycosylation structures in the variable region are resistant to cleavage by the N-glycosidase F of the protein (amino acid polypeptide). In one embodiment, dominant complex biantennary sugar structures are also characterized: by containing one or two sialic acids attached to one or the other antenna or both antennas. Sialic acid is of the N-acetyl neuraminic acid type and is most likely bound at the alpha 2,3 linkage to the galactoses with 1,4-terminal beta bonds; for lack of fucosylation in the nucleus, that is, the fucose residue bound in the alpha-1,6 bond to the innermost N-acetyl-glucosamine at the reduction end of the sugar chain.
In one embodiment, hybrid sugar structures are also characterized: by containing a complex type antenna (a lactosaminyl unit (GlcNAc-Gal) attached to the sugar structure of the core) as an arm of the biantenary structure. This arm contains predominantly acid N-acetyl neuraminic bound to galactose with 1,4-terminal beta bonds; by having one and up to 3 additional trunnion subunits attached to the core sugar structure as another antenna; for lack of fucosylation in the nucleus, that is, for lacking the fucose residue bound in the alpha bond 1.6 to the innermost N-acetyl-glucosamine at the reducing end of the sugar chain. In one embodiment, sugar structures of the oligomannose type are also characterized by containing subunits of mannose 4 (Man4 -> GlcNAc2), (Man5- * GlcNAc2) or 6 (Man6? GlcNAc2) in the complete sugar structure, ie, including the 3 subunits of branched mannose present in an N-linked sugar structure in the typical nucleus; for lack of fucosylation in the nucleus, that is, for lacking the fucose residue bound in the alpha l- > 6 to the innermost N-acetyl-glucosamine at the reducing end of the sugar chain.
In another embodiment of the present invention, there is provided a composition comprising an antibody molecule characterized in that an antigen-binding site comprises a glycosylated asparagin (Asn) in the variable region of the heavy chain (VH) and an antibody molecule characterized because two antigen binding sites comprise a glycosylated asparagin (Asn) in the variable region of the heavy chain (VH), ie, a composition comprising monoglycosylated ANTIBODY and double glycosylated ANTIBODY, and hereinafter referred to as ANTIBODY COMPOSITION. The phrase "ANTIBODY COMPOSITION" also refers to compositions comprising molecules comprising at least one glycosylated V H region as defined herein or at least one glycosylated CDR of said V H region, wherein said molecules can be, inter alia, immunoglobulins or isoforms and modifications of immunoglobulins, as described above. For example, said composition may also comprise single chain antibodies (scFvs) or bispecific molecules comprising CDR regions derived from glycosylated VH. Other definitions of the ANTIBODY COMPOSITION of the present invention are provided below. The COMPOSITION OF ANTIBODIES does not comprise or simply comprises to a very low degree "non-glycosylated VH" antibody molecules, ie, antibodies that do not comprise the glycosylation defined herein in the region variable, in particular, in the variable part of the heavy chain (VH). In the context of the present invention and in particular in the context of the antibody mixtures provided herein, the phrase "does not comprise or simply comprises to a very low degree non-glycosylated antibody molecules" means that the COMPOSITION OF ANTIBODIES comprises less from 10%, for example, less than 5%, for example, less than 4%, for example, less than 3%, for example, less than 2%, for example, less than 1%, for example, less than 0.5 % or less of the / non-glycosylated isoform as described herein. Accordingly, in one embodiment the present invention provides an antibody preparation comprising monoglycosylated and / or doubly glycosylated antibodies (said glycosylation is located in the variable region of the heavy chain) and which lacks antibody molecules without glycosylation in the region variable. Again, the phrase "lacking antibody molecules without glycosylation in the variable region" refers to antibody preparations / antibody mixtures / groups of antibodies comprising at most 10%, for example, at most 5%, for example, a maximum of 4%, for example, a maximum of 3%, for example, a maximum of 2%, for example, a maximum of 1%, for example, a maximum of 0.5%, for example, a maximum of 4%, for example, maximum 3%, for example, at most 2%, for example, at most 1%, for example, at most 0.5%, for example, at most 0.3%, for example, at most 0.2% of non-glycosylated isoforms as described herein. In one embodiment, the present invention provides a composition that does not comprise more than 0.5% of isoforms of antibodies that are not glycosylated in their variable regions, for example, that are not glycosylated in the variable region of the heavy region. As indicated above, in one embodiment of the present invention, a mixture of monoglycosylated and doubly glycosylated antibodies, for example, immunoglobulins, is provided, and said mixture lacks antibody molecules without glycosylation at the variable region. Antibodies lacking such posttranslational modification in the variable region, for example, in both variable regions of the heavy chain (both regions (VH)) is considered, in the context of the present invention, a "non-glycosylated form" which does not comprise glycosylation in the variable region of the heavy chain. However, this "non-glycosylated form" can comprise (a) glycosylation (s) in the constant region (region C) of the antibody, for example, and more commonly at the well-conserved glycosylation site of the Fc part, in particular, asparagine (Asn) 306 in the non-variable / constant Fc part as defined herein. The isoforms of glycosylated ANTIBODIES themselves or as a combination of mono-glycosylated isoforms and doubly glycosylated are very useful and advantageous therapeutic antibody preparations for the treatment of Alzheimer's disease (AD) and other disorders related to amyloid such as Down syndrome, hereditary cerebral hemorrhage with "Dutch type" amyloidosis, Parkinson's disease , ASL (amyotrophic lateral sclerosis), Creutzfeld Jacob's disease, HIV-related dementia and motor neuropathy. The isoforms of glycosylated ANTIBODIES themselves or as a combination of mono-glycosylated and double-glycosylated isoforms are also unique diagnostic tools. Both glycosylated isoforms described herein show enhanced and highly effective in vivo penetration into the brain. Effective penetration into the brain and specific binding to β-amyloid plaques can be demonstrated in PS2APP mice, a mouse model for amyloidosis related to AD. In addition, improved specificity could be detected for genuine human β-amyloid plaques through immunohistochemical staining with significantly reduced non-specific viscosity. The minimum effective concentration for a consistent staining of the human β-amyloid plaques was determined at 10 ng / ml, as documented in the appended examples. As documented in the appended examples, the separation and characterization of the differently glycosylated antibodies, for example, the immunoglobulins, revealed that the glycosylation of the variable region of the heavy chain has a great influence on the binding of the antigen to the Aβ peptides, the diagnostic value, the pharmacological profile and the functional activity. Purified antibody molecules can be subjected to analytical binding studies by MS (Biacore) and binding by epitope mapping (Pepspot analysis) to soluble Aβ, aggregation of Aβ dissociation and microscopic analysis of the binding to β-amyloid plaques in vivo and in vi tro. In one embodiment of the present invention, the purified ANTIBODY or the ANTIBODY COMPOSITION is capable of specifically recognizing the β-A4 / Aβ4 peptide. Accordingly, as described herein, the phrases "purified ANTIBODY" or "ANTIBODY COMPOSITION" refers in a specific embodiment to an ANTIBODY or ANTIBODY COMPOSITION capable of specifically recognizing two regions (the N-terminal region and the central / middle part) of the Aß / Aß4. The phrase "specifically recognize" means in accordance with the present invention that the antibody molecule is capable of specifically interacting with and / or binding to at least two amino acids from each of the two ß-A4 regions as defined in I presented. Said phrase refers to the specificity of the antibody molecule, ie, to its ability to discriminate between specific regions of the β-A4 peptide defined herein and another unrelated region of the β-A4 peptide, or other protein related to the APP / peptide / test peptide (not related). Accordingly, the specificity can be determined experimentally by methods known in the art and the methods disclosed and described herein. These methods include, but are not limited to, Western blot, ELISA, RIA, ECL, IRMA and peptide scans. Said methods also comprise the determination of the KD values as illustrated, inter alia, in the appended examples. Peptide screening (Pepspot assay) is routinely employed to map linear epitopes on a polypeptide antigen. The primary sequence of the polypeptide is successively synthesized in activated cellulose with peptides that overlap each other. The recognition of certain peptides by the antibody to be tested for its ability to detect or recognize a specific antigen / epitope is qualified through the development of routine color (secondary antibody with an enzyme or a dye, such as horseradish peroxidase). , 4-chloronaphthol or hydrogen peroxide), through a chemiluminescence reaction or similar means known in the art. In the case of chemiluminescence reactions, inter alia, or the use of a secondary fluorescent antibody, the reaction can be quantified. If the antibody reacts with a certain group of overlapping peptides, we can deduce the minimum sequence of amino acids that is necessary for the reaction; see them Illustrative examples provided in accordance with the present invention. The same assay can reveal two distant groups of reactive peptides, which indicate the recognition of a discontinuous epitope, ie, conformational, in the antigenic polypeptide (Geysen (1986), Mol.Immunol.23, 709-715). In addition to the Pepspot assay, a standard ELISA assay can be carried out. As demonstrated in the appended examples, the small hexapeptides can be coupled to a protein and spread on an immunoplate and reacted with the antibodies to be tested. Classification can be carried out through the development of standard color (for example, secondary antibody with horseradish peroxidase and tetramethyl benzidine with hydrogen peroxide). The reaction in certain containers is classified according to optical density, for example, at 450 nm. The typical background (= negative reaction) can be 0.1 OD, the typical positive reaction can be 1 OD. This means that the positive / negative difference (ratio) can be more than 10 times more. More information is provided in the attached examples. Further, additional quantitative methods are provided to determine the specificity and ability to "specially recognize" the two regions of the β-A4 peptide defined herein. The phrase "two regions of the β-A4 peptide" refers to two regions related, for example, to amino acids N-terminals 3 to 6 and a central / middle epitope at the position of amino acids 18 to 24 of SEQ ID NO: 3 (the β-A4 peptide). As documented in the appended examples, in particular the double glycosylated ANTIBODY A isoform provided and exemplified herein (see appended examples) detects two parts of the Aβ molecule, the first part comprises amino acids 1 to 10 in the N-terminus. and the second part comprises amino acids 17 to 26 of the central / middle part of Aß (as shown in SEQ ID NO: 3). Accordingly, in the antibody mixtures provided herein and comprising monoglycosylated and doubly glycosylated isoforms of the antibodies provided herein, the two regions may also be enlarged in a certain way, and therefore, comprise, for example, amino acids 1 to 10 (or 11 or 12) or a shorter part and amino acids 17 to 26 (or amino acids 16 to 27) or a shorter part comprised between amino acids 17 to 26, such as, for example, amino acids 19 to 26 or 20 to 26. The phrase "β-A4 peptide" in the context of the present invention refers to Aß39, Aβ41, Aβ43 previously described, in particular, to Aβ40 and Aβ42. Aβ42 is also described in SEQ ID NO: 3 attached. It should be noted that the phrase "two regions of the β-A4 peptide" also refers to an "epitope" and / or an "antigenic determinant" comprising the two regions of the β-A4 peptide or their parts defined herein. According to the present invention, said two regions of the β-A4 peptide are separated (at the level of the sequence of amino acids) in the primary structure of the β-A4 peptide for at least one amino acid, for example, by at least two amino acids, for example, by at least three amino acids, for example, by at least four amino acids, for example, by at least five amino acids, for example, by at least six amino acids. As shown herein and documented in the appended examples, antibodies / antibody molecules of the invention detect / interact with and / or bind to two regions of the β-A4 peptide as defined herein, and through it said two regions are separated (at the level of the primary structure of the amino acid sequence) by at least one amino acid, and the sequence separating said two regions / "epitopes" can comprise more than seven amino acids, more than 8 amino acids , more than 10 amino acids or even approximately 14 amino acids. The phrase "two regions of the β-A4 peptide" can also refer to a conformational epitope or a discontinuous epitope composed of said two regions or their parts; see also Geysen (1986) loe. cit. In the context of the present invention, a conformational epitope is defined by two or more discrete amino acid sequences separated in the primary sequence, which bind at the surface when the polypeptide is incorporated into the native protein (Sela, (1969) Science 166, 1365 and Laver, (1990) Cell 61, 553-6). It is contemplated that the antibody molecules of the present invention bind / interact specifically with a conformational epitope (s) composed of and / or comprising the two regions of the ß-A4 described herein or its parts as disclosed below. It is believed that the "antibody molecules" of the present invention comprise a dual simultaneous and independent specificity for (a) a length of amino acids comprising amino acids 1 through 11 (or (a) part (s) thereof) of β- A4 and (b) a length of amino acids comprising amino acids 16 to 27 (or (a) part (s) thereof) of ß-A4 (SEQ ID NO: 3). The fragments or parts of these lengths comprise at least two, in most cases at least three amino acids. Molecules of antibodies, for example, immunoglobulins, can be expressed, inter alia, in three systems: a) in transiently transfected human embryonic kidney cells containing the nuclear antigen of the Epstein barr virus (HEK 293 EBNA, Invitrogen), b) in transiently transfected Chinese hamster ovary (CHO) cells, and c) in stably transfected CHO cell lines (CHO Kl and CHO Kl SV, Lonza Biologics). The three different antibody molecules (non-glycosylated, monoglycosylated or doubly glycosylated) can be separated through specific purification steps, comprising the purification of protein A, cation exchange chromatography, as well as column separation as described. details later. In an embodiment of the invention, the antibody molecule is produced recombinantly, for example, in a CHO cell or a HEK 293 cell, preferably CHO cells. In a particular embodiment, the above-identified glycosylation patterns can be obtained after expression in CHO cells. CHO cells are known in the art and comprise, inter alia, CHO cells used in the experimental part, such as CHO Kl or CHO Kl SV cells. The HEK 293 cells commonly used are HEK 293 EBNA. The recombinant expression of the glycosylated antibody of the invention is carried out, as shown in the examples, in a eukaryotic expression system, in particular, in CHO cells. However, other eukaryotic cells can be contemplated. Eukaryotic cells comprise, for example, fungal cells or animals. Examples of fungal cells are yeast cells, for example, those of the genus Saccharomyces, for example, those of the species Saccharomyces cerevisiae. Suitable animal cells are, for example, insect cells, vertebrate cells, e.g., mammalian cells, such as, for example, NSO, MDCK, U2-OSHela, NIH3T3, MOLT-4, Jurkat, PC-12. , PC-3, IMR, NT2N, Sk-n-sh, CaSki, C33A. Human cell lines are also contemplated. These host cells, for example, CHO cells, provide post-translational modifications to the antibody molecules of the invention, which include the removal of leader peptides or signal sequences, the folding or assembly of H (heavy) and L ( light) and, what is more importantly, the glycosylation of the molecule on the correct sides, that is, in the variable region of the heavy chain. Said leader sequence or signal peptide is proteolytically cleaved by the host signal peptide during the secretory pathway during its recombinant production, for example, in CHO cells. Other suitable cell lines known in the art are obtained from depositories of cell lines, such as the American Type Culture Collection (ATCC). In accordance with the present invention, it is also contemplated that the primary cells / cell cultures may act as host cells. These cells derive, in particular, from insects (such as insects of the species Drosophila or Blatta) or mammals (such as humans, pigs, mice or rats). Said host cells can also comprise cell line cells and / or derived from them, such as neuroblastoma cell lines. Accordingly, the antibody molecule of the invention is prepared using a recombinant expression system. An example of such a system, as indicated above, is a mammalian expression system utilizing Chinese hamster ovary (CHO) cells. These can be used with the glutamine synthetase (GS) system (WO 87/04462, WO 89/01036, Bebbington, 1992, Biotechnology (N Y), 10, 169-75). This system comprises the transfection of a CHO cell with a gene encoding the GS enzyme and the desired antibody genes. Then, the CHO cells are they select and grow in a glutamine-free medium, and they are also subjected to inhibition of the GS enzyme using methionine sulfoximine (MSX). In order to survive, the cells will amplify the expression of the GS enzyme and concomitantly the expression of mAb. Another possible expression system is the CHO dhfr- system, where the CHO cells are deficient for dihydrofolate reductase (dhfr-) and dependent on thymidine and hypoxanthine for their growth. The parenteral CHO dhfr- cell line is transfected with the antibody and the dhfr gene, thus allowing the selection of CHO cell transformants of the dhfr + phenotype. The selection is carried out in the absence of thymidine and hypoxanthine. Expression of the antibody gene can be increased by amplification using methotrexate (MTX). This drug is a direct inhibitor of the dhfr enzyme and allows the isolation of resistant colonies that amplify their dhfr gene copy number and, therefore, the antibody gene enough to survive under these conditions. Purified antibody molecules, eg, immunoglobulins, can be prepared through a method comprising the following steps: (a) recombinant expression of a heterologous nucleic acid molecule encoding an antibody molecule as defined above in a mammalian cell, for example, a CHO cell or a HEK 293 cell; Y (b) purification of said recombinantly expressed antibody molecule through a method comprising the following steps: (bl) column purification of protein A; (b2) column purification by ion exchange, for example, cation exchange chromatography; and optionally (b3) column purification by size exclusion. The purification protocol may comprise other steps, such as other concentration steps, for example, diafiltration or analytical steps, for example, comprising analytical columns. The method / method may also comprise the steps of inactivating the virus and / or stages of viral removal, for example, through filtrations / nanofiltration. It is also contemplated and possible to repeat certain particular steps (for example, two steps of ion exchange chromatography can be carried out) or that certain steps are missed (for example, size exclusion chromatography). Protein A is a ligand specific to the group that binds to the Fc region of most IgGl isotypes. It is synthesized by some strains of Staphylococcus aureus and can be isolated from them and coupled to chromatographic beads. Various types of gel preparations are commercially available.
An example for a protein A column that can be used is a MabSelect column (trademark). Ideally, the column is equilibrated with 25 mM Tris / HCl, 25 mM NaCl, 5 mM EDTA, the culture supernatant is located on the column, the column is washed with 1 M Tris / HCl pH 7.2 and the antibody is elute at pH 3.2 using 100 mM acetic acid. Cation exchange chromatography exploits the interactions between positively charged groups in a stationary phase and the sample that is in the mobile phase. When a weak cation exchanger is used (eg CM Toyopearl 650®), the following chromatographic steps are used: after pre-equilibration with 100 mM acetic acid pH 4, loading of a protein A eluate and washing with 100 mM of acetic acid pH 4, the antibody is eluted and fractionated by applying the steps comprising 250 mM sodium acetate (pH 7.8-8.5) and 500 mM sodium acetate (pH 7.8-8.5). With the first step, a mixture of doubly glycosylated isoforms fraction and monoglycosylated isoform fraction is eluted and when using the second step the fraction of non-glycosylated isoforms is usually eluted. From a strong cation exchanger (for example, SP Toyopearl 650) the antibody can be eluted through salt steps: After equilibration of the column with 50 mM acetic acid pH 5.0, loading of protein A eluate with pH 4, the first elution stage is carried out using 50 mM acetic acid and 210 mM sodium chloride. Then a second elution step of 50 mM acetic acid and 350 mM sodium chloride is applied. Through the first salt stage, a mixture of doubly glycosylated isoform fraction and a monoglycosylated isoform fraction is eluted through a second salt stage, the non-glycosylated isoform is usually eluted. In addition, the antibody can also be used from a strong cation exchange column (eg, SP-Sepharose®) by means of a salt gradient: After equilibration, loading and washing of the column at pH 4.5, a salt gradient is applied from 50 mM of MES pH 5.8 to 50 mM of MES / 1 M of sodium chloride pH 5.8. Here, the double glycosylated isoform, monoglycosylated isoform and non-glycosylated isoform fractions are normally eluted separately. Then, the double glycosylated isoform fraction and the mono-glycosylated isoform fraction can be combined to produce as a result a combination of product and / or a desired antibody mixture. Further purification of the mixture of double glycosylated and monoglycosylated antibody molecules, eg, immunoglobulins, can be performed by size exclusion chromatography. An example of a useful column is the Superdex 200® column. Examples of working buffers include histidine / sodium chloride, for example, 10 mM histidine / 125 mM sodium chloride / pH 6, and phosphate buffered saline (PBS). Anion exchange chromatography in the continuous flow mode followed by a concentration / diafiltration constitutes an alternative purification step. Q Sepharose® is an example of a resin for the anion exchange step. For example, the eluate of SP chromatography can be diluted to a third with 37.5 mM Tris / HCl pH 7.9 and passed through a Q-Sepharose column pre-equilibrated with 25 mM Tris / 83 mM sodium acetate. The continuous flow is collected, adjusted to pH 5.5 and concentrated by ultrafiltration using, for example, a Hydrosart 30 kD® membrane. Then, the concentrate can be diafiltered against, for example, 10 volumes of 20 mM histidine / HCl pH 5.5. The aforementioned purification protocol also comprises an additional step (c) and an analytical chromatography step, such as the use of a Mono-S HR5 / 5 column. However, additional steps are also contemplated, such as diafiltration, for example, for the concentration of the antibody molecules. In one embodiment of the present invention, there is provided a composition, preparation of antibodies or combination of antibodies comprising antibody molecules as described herein or antibody molecules that are prepared by the method provided above. In this embodiment of the invention, said The composition comprises monoglycosylated or double glycosylated antibodies. In another embodiment, said composition comprises monoglycosylated and double glycosylated antibodies (in the variable region (s) of the heavy chain (s)) and said composition is derived from antibody molecules that are lacking of glycosylation in the variable region. In the context of this embodiment, the phrase "antibody combination" refers to a mixture of mono-glycosylated and double-glycosylated antibodies (in the variable region (s) of the heavy chain (s) ( s)) that have been isolated individually and then combined in a mixture. The mixtures of antibodies or combinations of antibodies provided herein may comprise 50% monoglycosylated antibodies and 50% double glycosylated antibodies as defined herein. However, relationships of 30/70 to 70/30 are also contemplated. But the skilled artisan also knows that other relationships are contemplated in the antibody mixtures of the present invention. For example, 10/90 or 90/10, 20/80 or 80/20 can be used, as well as 40/60 or 60/40 in the context of the present invention. As documented in the examples, a particular useful relationship in the ANTIBODY MIXTURES of the invention comprises monoglycosylated antibodies and double glycosylated antibodies as hereinbefore defined in a ratio ranging from 40/60 to 45/55.
The compositions provided herein are particularly useful in diagnosis or in a pharmaceutical composition. Accordingly, the invention provides diagnostic or pharmaceutical compositions comprising: (a) an antibody molecule as defined above, comprising an antigen-binding site with a glycosylated Asn; (b) an antibody molecule as defined above, comprising two antigen-binding sites with a glycosylated Asn; or more preferably, (c) a combination of the antibody molecules (a) and (b) The combination (c) that is provided herein, comprising the antibody molecule (s) that includes an antigen-binding site with a glycosylated Asn and the antibody molecule (s) including two antigen binding sites with a glycosylated Asn, lacks non-glycosylated isoforms (with respect to the variable region of the heavy chain). As indicated above, the phrase "lacks non-glycosylated isoforms (with respect to the variable region of the heavy chain)" refers to combinations / combinations of antibodies / antibody preparations, where less than 5%, for example, less than 4%, less than 3%, less than 2%, less than 1% or even less than 0.5% of the antibody species in said combination do not they are glycosylated in the variable region of the heavy chain. As demonstrated in the examples, said combinations / combinations of antibodies / antibody preparations can comprise almost none (less than 0.5%) non-glycosylated isoform. The percentage and / or amount of a given glycosylation isoform (as defined herein, eg, glycosylation in the variable region of the heavy chain, see inter alia Figure 14 attached) in a COMPOSITION OF ANTIBODIES is it can be easily determined by methods known in the art. These methods comprise, without limitation, mass spectrometry, ion exchange by SDS-PAGE analysis, HPLL, ELISA and the like. As shown in the appended examples, the specific and sensitive immunodecoration of genuine Alzheimer's β-amyloid plaques by the antibodies of the invention is demonstrated in vitro with immunohistochemical staining experiments using human brain tissue cryoses from patients with AD. Effective staining of β-amyloid plaques from brain slices with human anti-Aβ antibodies from patients vaccinated with Aβ was also demonstrated (Hock, 2002, Nature Medicine, 8, 1270-1275). Likewise, immunodecoration is also demonstrated in a transgenic animal model that represents the loading of human β-amyloid plaques (Richards, 2003, J. Neuroscience, 23, 8989-9003). In similar animal models it has been shown that this union of the plates leads to their clearance [clearance] and subsequently to an improvement of symptoms related to the disease, where the inclusion of Fc-dependent procedures has been treated (Bard, 2000, Nature Medicine, 6, 916-919, Wilcock, 2003, Neurobiology Disease, 15, 11-20; Wilcock, 2004, J. Neuroscience, 24, 6144-6151). In addition, it was reported that an effective binding of anti-Aβ antibodies with β-amyloid plaques correlates with a lesser progression of the disease (Hock, 2002, Nature Medicine, 8, 1270-1275; Hock, 2003, Neuron, 38; 547-554). This analysis and post-mortem analysis of human brain tissue suggests that phagocytosis of microglial cells is mechanically compromised in plaque clearance in man (? Icoll, 2003, Nature Medicine, 9, 448-452). Therefore, the antibody of the present invention or comprised, in particular, in pharmaceutical compositions is a human IgGl, which is primarily responsible for FcR-dependent procedures in humans. Immunodecoration of β-amyloid plaques effective from the antibodies of the invention / mixture of the invention suggests that the drug will be effective for passive immunization to remove the existing β-amyloid plaques and prevent the formation of β-amyloid plaques in Humans. In addition, the antibodies must preferably cross the blood-brain barrier to reach their destination. For molecules of a larger size such as human IgGs, this process is drastically diminished, so that only approximately between 0.1 and 0.2% can be achieved of the plasma concentration of an antibody in CSF. The mechanism of clearance of plaques is still subject to a series of controversial debates, which may include peripheral effects on the Aβ peptide (Dodart, 2002, Nature Neuroscience, 5: 452-457). Thus, the therapeutic antibody generated or the corresponding mixtures of the invention of monoglycosylated and double glycosylated antibodies (in the heavy chain of the variable region) of the invention also have the property of depolymerizing Aβ multimers in vi tro without including procedures Fc-dependent and binding to CSF-soluble Aβ monomers and oligomers, since the neutralization of soluble Aβ peptides or oligomeric Aβ peptides (eg, aggregation intermediates) may also contribute to the overall reduction effect of amyloid (Du, 2003, Brain, 126: 1-5). The compositions of the invention can be administered in solid or liquid form and can be presented, inter alia, in the form of (a) powder (s), (a) tablet (s), (a) solution or (a) spray (s). Said composition may comprise one or more antibodies / antibody molecules of the invention, more preferably a mixture of monoglycosylated and doubly glycosylated antibodies as provided herein. It is preferred that said pharmaceutical composition optionally comprises a pharmaceutically acceptable carrier and / or diluent. The pharmaceutical composition disclosed in the present may be particularly useful for the treatment of neurological and / or neurodegenerative disorders. Such disorders include, but are not limited to, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), hereditary cerebral hemorrhage with "Dutch-type" amyloidosis, Down syndrome, HIV dementia, Parkinson's disease and disorders. neuronal related to age. The pharmaceutical composition of the invention is contemplated, inter alia, as a potent inhibitor of amyloid plaque formation or as a potent stimulator for the depolymerization of amyloid plaques. Therefore, the present invention provides pharmaceutical compositions comprising compounds of the invention that are used for the treatment of amyloidogenic diseases / disorders. The phrase "amyloidogenic disease / disorder" includes any disease associated with or caused by the formation or deposition of amyloid fibrils and / or pathological APP proteolysis. An example of amyloidogenic disease includes, but is not limited to, Alzheimer's disease (AD), Down syndrome, dementia associated with the formation of Lewy bodies, Parkinson's disease with dementia, mild cognitive impairment, amyloid angiopathy cerebral and vascular dementia. Different amyloidogenic diseases are defined and / or characterized according to the nature of the polypeptide component of the amyloid deposits. For example, the β-amyloid protein is characteristic of amyloid deposits found in subjects with Alzheimer's disease. Examples of suitable pharmaceutical carriers, excipients and / or diluents are known in the art and include phosphate-buffered saline solutions, water, emulsions, such as oil / water emulsions, various types of wetting agents, sterile solutions, etc. The compositions comprising said carriers can be formulated by known conventional methods. Suitable carriers can comprise any material which, when combined with the specific binding agent or anti-Aβ antibody, maintains binding by high affinity to Aβ and is not reactive with the subject's immune systems including excipients, surfactants, tonicity agents and the like; see Remington's Pharmaceutical Sciences (1980) 16th edi tion, Osol, A. Ed. These pharmaceutical compositions can be administered to the subject in a suitable dose. Administration of the appropriate compositions can be effected in different ways, for example, by parenteral, subcutaneous, intraperitoneal, topical, intrabronchial, intrapulmonary and intranasal administration and, if desired for local treatment, by intralesional administration. Parenteral administrations include intraperitoneal, intramuscular, intradermal, subcutaneous, intravenous or intraarterial administration. It is preferred, in particular, that said administration be carried out by injection and / or delivery, for example, to a site in an artery of the brain or directly in brain tissue. The compositions of the invention can also be administered directly at the desired site, for example, through biolistic delivery to an external or internal target site, such as the brain. Pharmaceutical compositions comprising the glycosylated antibodies described herein are prepared by mixing the antibody having the desired degree of purity with optional physiologically acceptable carriers, excipients, stabilizers, surfactants, buffers and / or tonicity agents. Acceptable carriers, excipients and / or stabilizers are not toxic to containers at the dosages and concentrations employed, and include buffers such as phosphate, citrate and other organic acids; antioxidants that include ascorbic acid, glutathione, cysteine, methionine and citric acid; preservatives (such as ethanol, benzyl alcohol, phenol, m-cresol, p-chlor-m-cresol, methylparaben or propylparaben, benzalkonium chloride or combinations thereof); amino acids such as arginine, glycine, ornithine, lysine, histidine, glutamic acid, aspartic acid, isoleucine, leucine, alanine, phenylalanine, tyrosine, tryptophan, methionine, serine , praline and combinations of them; monosaccharides, disaccharides and other carbohydrates; Low molecular weight polypeptides (less than about residues), - proteins such as gelatin or serum albumin; chelating agents such as EDTA; sugars such as trehalose, sucrose, lactose, glucose, mannose, maltose, galactose, fructose, sorbose, raffinose, glucosamine, N-methylglucosamine (termed "Meglumine"), galactosamine and neuraminic acid; and / or nonionic surfactants such as Tween, Brij Pluronics, Triton-X or polyethylene glycol (PEG). The pharmaceutical composition can be presented in liquid form, lyophilized form or a reconstituted liquid form from a lyophilized form, where the lyophilized preparation must be reconstituted with a sterile solution before administration. The standard procedure for reconstituting a lyophilized composition is to add a volume of pure water again (typically equivalent to the volume removed during lyophilization). However, solutions comprising antibacterial agents for the production of pharmaceutical compositions for parenteral administration can also be used; see also Chen (1992) Drug Dev Ind Pharm 18, 1311-54. Examples of antibody concentrations in the pharmaceutical composition can range from about 1 mg / mL to about 200 mg / mL or from about 50 mg / mL to about 200 mg / mL, or from about 150 mg / mL to about 200 mg / mL. mL. For clarity, it is emphasized that the concentrations indicated herein refer to the concentration in a liquid or in a liquid that is accurately reconstituted from a solid form. An aqueous formulation of the antibody can be prepared in a buffered pH solution, for example, at a pH ranging from about 4.0 to about 7.0, or from about 5.0 to about 6.0, or alternatively about 5.5. Examples of buffers that are suitable for a pH within this range include phosphate buffers, histidine, citrate, succinate, acetate and other organic acid buffers. The buffer concentration can range from about 1 mM to about 100 mM, or between about 5 mM and about 50 mM, depending, for example, on the desired buffer and tonicity of the formulation. A tonicity agent may be included in the formulation of antibodies to modulate the tonicity of the formulation. Examples of tonicity agents include sodium chloride, potassium chloride, glycerin and any component of the group of amino acids, sugars, as well as combinations thereof. Preferably, the aqueous formulation is isotonic, although hypertonic or hypotonic solutions may be suitable. The term "isotonic" indicates a solution that has the same tonicity as another solution with which it is compared, such as a physiological saline solution and blood serum. The tonicity agents can be used in an amount ranging from about 5 mM to about 350 mM, in particular, in an amount ranging from 105 mM to 305 mM. A surfactant can also be added to the antibody formulation to reduce aggregation of the formulated antibody and / or minimize formation of particles in the formulation and / or reduce adsorption. Examples of surfactants include polyoxyethylene sorbitan fatty acid esters (Tween), polyoxyethylene alkyl ethers (Brij), alkylphenyl polyoxyethylene (Triton-X) ethers, polyoxyethylene-polyoxypropylene copolymer (Poloxamer, Pluronic), and sodium dodecyl sulfate (SDS) . Preferred fatty acid-polyoxyethylene sorbitan esters are polysorbate 20 (marketed under the trademark Tween 20 ™) and polysorbate 80 (marketed for the Tween 80 ™ brand). Preferred polyethylene-polypropylene copolymers are those marketed under the names Pluronic® F68 or Poloxamer 188 ™. Preferred polyoxyethylene alkyl ethers are those marketed under the Brij ™ brand. Examples of surfactant concentrations may range from about 0.001% to about 1% w / v. A lyoprotectant can also be added in order to protect the labile active ingredient (e.g., a protein) from the destabilizing conditions during the lyophilization process. For example, known lyoprotectants include sugars (including glucose and sucrose); polyols (including mannitol, sorbitol and glycerol); and amino acids (including alanine, glycine and glutamic acid). The lyoprotectants are generally used in an amount ranging from about 10 mM to 500 mM. In one embodiment, the formulation contains the above-indicated agents (ie, glycosylated antibody, surfactant, buffer, stabilizer and / or tonicity agent) and is essentially free of one or more preservatives, such as ethanol, benzyl alcohol, phenol, m -cresol, p-chlor-m-cresol, methylparaben or propylparaben, benzalkonium chloride, and combinations thereof. In another embodiment, a preservative may be included in the formation, for example, in concentrations ranging from about 0.001 to about 2% (w / v). In one embodiment, the antibody formulation of the invention is a liquid or lyophilized formulation suitable for parenteral administration which may comprise: between about 1 and about 200 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - between about 0.001 and about 1% of at least one surfactant; between about 1 and about 100 mM of a buffer; optionally between about 10 and about 500 mM of a stabilizer and / or between about 5 and about 305 mM of a tonicity agent; at a pH ranging from about 4.0 to about 7.0. In a preferred embodiment, the parenteral formulation of the invention is a liquid or lyophilized formulation comprising: between about 1 and about 200 mg / mL of the glycosylated antibodies described herein or ANTIBODY COMPOSITION, - 0.04% Tween 20 p / v, - 20 mM L-histidine, 250 mM sucrose, at a pH of 5.5. In a more preferred embodiment, the parenteral formulation according to the invention also comprises a lyophilized formulation comprising: 15 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.04% Tween 20 p / v, 20 mM L-histidine, - 250 mM sucrose, at a pH of 5.5; or 75 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.04% Tween 20 w / v, 20 mM L-histidine, 250 mM sucrose, at a pH of 5.5; or 75 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.02% Tween 20 w / v, 20 mM L-histidine, 250 mM sucrose, at a pH of 5.5; or - 75 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.04% Tween 20 p / v, 20 mM L-histidine, 250 mM trehalose, - at a pH of 5.5; or 75 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.02% Tween 20 p / v, - 20 mM L-histidine, 250 mM trehalose, at a pH of 5.5. In another more preferred embodiment, the parenteral formulation according to the invention also comprises a liquid formulation comprising: 75 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.022% Tween 20 p / v, 120 mM L-histidine, 250 125 mM sucrose, at a pH of 5.5; or 37.5 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.02% Tween 20 p / v, 10 mM L-histidine, - 125 mM sucrose, at a pH of 5.5; or 37.5 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.01% Tween 20 w / v, 10 mM L-histidine, 125 mM sucrose, at a pH of 5.5; or - 37.5 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.02% Tween 20 p / v, 10 mM L-histidine, 125 mM trehalose, - at a pH of 5.5; or 37. 5 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.01% Tween 20 p / v, 10 mM L-histidine, - 125 mM Trehalose, at a pH of 5.5; or 75 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.02% Tween 20 w / v, 20 mM L-histidine, - 250 mM Trehalose, at a pH of 5.5; or - 75 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.02% Tween 20 p / v, 20 mM L-histidine, - 250 mM Mannitol, - at a pH of 5.5; or 75 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.02% Tween 20 p / v, - 20 mM L-histidine, 140 mM sodium chloride, at a pH of 5.5; 150 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.02% Tween 20 p / v, - 20 mM L-histidine, - 250 mM Trehalose, at a pH of 5.5; or 150 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.02% Tween 20 p / v, 20 mM L-histidine, - 250 mM Mannitol, at a pH of 5.5; or 150 mg / mL of the glycosylated antibodies described herein or COMPOSITION OF ANTIBODIES, - 0.02% Tween 20 p / v, 20 mM L-histidine, - 140 mM sodium chloride, at a pH of 5.5. The phrase "glycosylated antibodies that are described herein" in the context of the exemplified formulations may comprise in the present invention the monoglycosylated antibodies defined herein, the double glycosylated antibodies defined herein, as well as mixtures thereof.
The dosage regimen will be determined by the intervening physician and clinical factors. As is known in the art of medicine, dosages for a patient depend on many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs administered concurrently. The pharmaceutically active protein material can be present in amounts ranging from 1 ng to 20 mg / kg of body weight per dose, for example, between 0.1 mg and 10 mg / kg of body weight, for example, between 0.5 and 5 mg /. kg of body weight; however, doses below or above this exemplary range are contemplated, especially considering the factors mentioned above. If the regimen is a continuous infusion, it should also range between 1 μg and 10 mg / kg of body weight per minute. The pharmaceutical compositions described herein can be formulated to be short acting, fast releasing, long acting or delayed release. Therefore, the pharmaceutical compositions may also be suitable for slow release or controlled release. Delayed release preparations can be prepared using methods known in the art. Suitable examples of delayed release preparations include semipermeable matrices of hydrophobic polymers solids containing the antibody, wherein the matrices are presented in the form of shaped articles, for example, films or microcapsules. Examples of delayed release matrices include polyesters, L-glutamic acid and ethyl-L-glutamate copolymers, non-degradable ethylene-vinyl acetate, hydrogels, polylactides, degradable lactic acid-glycolic acid copolymers, and poly-D- acid ( -) -3-hydroxybutyric. The possible loss of biological activity and possible changes in the immunogenicity of the antibodies comprised in the delayed-release preparations can be avoided by using appropriate additives, controlling the moisture content and developing specific polymer matrix compositions.
Progress can be monitored through periodic evaluation. The compositions, ie, monoglycosylated and / or doubly glycosylated antibodies of the invention or a mixture thereof, can be administered locally or systemically. It should be noted that peripherally administered antibodies can enter the central nervous system; see, inter alia, Bard (2000), Nature Med. 6, 916-919. Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, solutions, emulsions or alcoholic / aqueous suspensions, including saline and buffered media. Parenteral vehicles include a solution of sodium chloride, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer, or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives such as, for example, antimicrobials, antioxidants, chelating agents and inert gases, and the like may also be present. In addition, the pharmaceutical composition of the invention may comprise other agents according to the intended use of the pharmaceutical composition. These agents can be drugs that act on the central nervous system, such as neuroprotective factors, cholinesterase inhibitors, Ml muscarinic receptor agonists, hormones, antioxidants, inflammation inhibitors, etc. It is particularly preferred that said pharmaceutical composition comprises other agents such as, for example, neurotransmitters and / or substitution molecules for neurotransmitters, vitamin E or alpha-lipoic acid. The expert in the art, in particular, but without limitation, the biochemists, biologists, chemists, pharmacists and groups of said professionals are already in a position to work and generate the aforementioned pharmaceutical compositions. Also medical personnel expert in the art, such as intervening physicians, know how to administer the pharmaceutical compositions to a patient in need of treatment with the pharmaceutical compositions defined herein. Said administration may comprise systemic administration, for example, through infusions and / or injections. However, direct administration of the compounds and / or mixtures of compounds of the invention to the brain is also contemplated. For example, the compound or mixture of compounds or formulation of compounds can be administered by interventricular or direct intrathecal injection to the brain, preferably through a slow infusion to minimize the impact on the brain parenchyma. A slow-release implant can also be used in the brain. The use of gene therapy methods is also contemplated, for example, through the use of implanted recombinant cells that produce the antibodies that are defined in the present invention. These "recombinant cells" must be able to provide the glycosylations defined herein in the variable regions / parts of the antibodies described herein, in particular, the anti-β antibodies of the invention. However, as set forth above, an advantage of the antibodies / antibody mixtures of the present invention is their ability to cross the blood-brain barrier and bind to the amyloid plaques. The pharmaceutical compositions of the invention described infra can be used for the treatment of all types of diseases unknown up to now or related or dependent on the aggregation of pathological APP or pathological APP processing. They can be particularly useful for the treatment of Alzheimer's disease and other diseases where extracellular deposits of β-amyloid appear to play a role. They can be conveniently used in humans, although the treatment in animals is also comprised in the methods, uses and compositions described herein. In a preferred embodiment of the invention, the composition of the present invention as disclosed herein is a diagnostic composition that also optionally comprises suitable detection means. The diagnostic composition comprises at least one of the aforementioned compounds of the invention, ie, the glycosylated antibodies described herein. Said diagnostic composition may comprise the compounds of the invention, in particular, the glycosylated antibody molecules of the present invention, soluble form / liquid phase, but it is also contemplated that said compounds are attached / attached and / or bound to a solid support. The solid supports can be used in combination with the diagnostic composition as defined herein or the compounds of the present invention can be linked directly to said solid supports. These supports they are known in the art and comprise, inter alia, commercially available column materials, polystyrene beads, latex beads, magnetic beads, colloidal metal particles, chips and surfaces of glass and / or silicon, nitrocellulose strips, membranes, sheets , duracitos, containers and walls of reaction trays, plastic tubes, etc. The compound (s) of the invention, in particular, the antibodies of the present invention, can bind to many different carriers. Examples of known carriers include glass, polystyrene, polyvinyl chloride, polypropylene, polyethylene, polycarbonate, dextran, nylon, amyloses, natural or modified celluloses, polyacrylamides, agarose and magnetite. The nature of the carrier can be soluble or insoluble for the purposes of the invention. Suitable markers and methods for labeling have been identified above and are also mentioned hereinafter. Suitable methods for fixing / immobilizing said compound (s) of the invention are known and include, without limitation, ionic, hydrophobic, covalent interactions and the like. It is particularly preferred that the diagnostic composition of the invention be used for the detection and / or quantification of APP and / or APP processing products, such as β-amyloid or for the detection and / or quantification of cleavage sides of APP pathological and / or (genetically) modified.
As illustrated in the appended examples, the glycosylated antibody molecules of the invention are particularly useful as diagnostic reagents in the detection of genuine human amyloid plaques in sections of the brain of patients with Alzheimer's disease by indirect immunofluorescence. It is preferred that said compounds of the present invention, which are employed in a diagnostic composition, be detectably labeled. There are several techniques available for labeling biomolecules, which are known to those skilled in the art and which are considered within the scope of the present invention. There are many different markers and methods for marking that are known to those skilled in the art. Examples of the types of labels that can be used in the present invention include enzymes, radioisotopes, colloidal metals, fluorescent compounds, chemiluminescent compounds, and bioluminescent compounds. Commonly used markers include, inter alia, fluorochromes (such as fluorescein, rhodamine, Texas, etc.) enzymes (such as horseradish peroxidase, ß-galactosidase, alkaline phosphatase), radioactive isotopes (such as 32P or 125I), biotin, digoxigenin, colloidal metals, chemiluminescent or bioluminescent compounds (such as dioxetanes, luminol or acridinium). Marking procedures, such as covalent coupling of enzymes or groups Biotinyl, iodinations, phosphorylations, biotinylations, etc. are known to those skilled in the art. Detection methods include, without limitation, autoradiography, fluorescent microscopy, direct and indirect enzymatic reactions, etc. The detection assays commonly used include radioisotope or non-radioisotopic methods. These comprise, inter alia, Western blot, superposition assays, RIA (radioimmunoassay) and IRMA (radioimmunometric assay), EIA (enzyme immunoassay), ELISA (enzyme-linked immunosorbent assay), FIA (fluorescent immunoassay) and CLIA (chemiluminescent immunoassay). . In addition, the present invention provides the use of glycosylated antibody molecules of the invention, or an antibody molecule produced by the method of the invention, or a mixture of monoglycosylated and doubly glycosylated antibodies provided herein for the preparation of a pharmaceutical composition. or diagnostic for the prevention, treatment and / or diagnosis of a disease associated with amyloidogenesis and / or formation of amyloid plaques. It is also preferred that the compounds described herein, in particular, the antibody molecules of the invention, are employed in the prevention and / or treatment of neuropathologies associated with the processing of modified or abnormal APP and / or amyloidogenesis. The antibody molecules, for example, in the immunoglobulin (modified) format, as antibodies in an IgG structure, in particular, in an IgGl structure, or in the format of chimeric antibodies (in particular, fully humanized antibodies or complete antibodies), bispecific antibodies, single chain Fvs (scFvs) or bispecific scFvs and the like are employed in the preparation of the pharmaceutical compositions herein. However, the antibody molecules and mixtures provided herein are also useful in the diagnostic configurations as documented in the appended examples, since the antibody molecules of the invention specifically interact with / detect Aβ4 and / or deposits / amyloid plaques. Therefore, an inventive use of the compounds of the present invention is the use for the preparation of a pharmaceutical composition for a neurological disorder that needs to be reduced, for example, by disintegration of the β-amyloid plaques, by clearance (from plaques) of amyloid or by passive immunization against the formation of β-amyloid plaques. As illustrated in the appended examples, the antibody molecules of the invention are particularly useful in the prevention of Aβ aggregation and in the depolymerization of already formed amyloid aggregates. Accordingly, the glycosylated antibodies of the invention, or a mixture of monoglycosylated or doubly glycosylated antibodies as described herein, should be employed in the reduction of pathological deposits / amyloid plaques, in clearance. of amyloid plaques / plate precursors, as well as in neuronal protection. It is particularly contemplated that the antibody molecules of the invention can be employed in the in vivo prevention of amyloid plaques, as well as in the in vivo clearance of pre-existing amyloid plaques / deposits. In addition, the antibody molecules or mixtures of the invention can be used in passive immunization methods against the Aβ peptide and aggregates of Aβ, ie the β-amyloid plaques. Clearance of Aβ4 / Aβ4 deposits can be achieved, inter alia, through the medical use of antibodies of the present invention comprising an Fc part. Said Fc part of an antibody can be particularly useful in immune responses mediated by the Fc receptor, for example, the attraction of macrophages (phagocytic cells and / or microglia) and / or helper cells. For the mediation of the in-response related to the Fc part, the antibody molecule of the invention is preferably in an IgGl (human) structure. As discussed herein, the preferred subject to be treated with the antibody molecules of the invention, or mixtures of antibodies, is a human subject. Other structures, such as the IgG2a or IgG2b structures, are also contemplated for the antibody molecules of the invention. The structures of immunoglobulins in the IgG2a and IgG2b formats are particularly contemplated in scenarios with mice, for example, in scientific uses of the antibody molecules of the invention, for example, in tests in transgenic mice expressing wild type (human) or mutated APP, fragments of APP and / or Aβ4. The aforementioned diseases associated with amyloidogenesis and / or amyloid plaque formation include, without limitation, dementia, Alzheimer's disease, motor neuropathy, Parkinson's disease, ALS (amyotrophic lateral sclerosis), scrapie, HIV-related dementia, as well as also Creutzfeld-Jakob disease, hereditary cerebral hemorrhage with Dutch-type amyloidosis, Down syndrome and neuronal disorders related to age. The antibody molecules of the invention and the compositions provided herein may also be useful in the reduction or prevention of inflammatory processes related to amyloidogenesis and / or the formation of amyloid plaques. In consecuense, the present invention also provides a method for treating, preventing and / or delaying neurological and / or neurodegenerative disorders comprising the step of administering to a subject suffering said neurological and / or neurodegenerative disorder, and / or to a subject susceptible to said neurological and / or neurodegenerative disorder, an effective amount of an antibody molecule or a mixture of monoglycosylated and / or double glycosylated antibodies of the invention as provided herein and / or a composition as defined above. The treatment that is provided in the present may comprise administering the compound / compositions of the present invention alone or in the form of a co-therapeutic treatment, ie, in combination with other drugs or medicaments. The term "treatment" as used herein contemplates the administration of monoglycosylated and / or double glycosylated antibodies (or mixtures thereof) as described herein to a patient in need thereof. Said patient may be a human patient, in one embodiment a human being suffering or being susceptible to a disorder related to the processing of pathological APP. Accordingly, the term "treatment" as used herein comprises the prophylactic, as well as curative, administration of the compounds or mixtures of compounds herein. A disorder to be treated with the compounds and the composition provided herein is Alzheimer's disease. Patients who have a diagnosis of probable Alzheimer's disease based on the criteria of the National Institute of Neurological and Communicative Disorders and Emiplejia / Association for Alzheimer's Disease and Related Disorders [National Institute of Neurological and Communicative Disorders and Stroke / Alzheimer's Disease and Related Disorders Association] (NINCDS / ADRDA criteria) Mckhann et al., 1984. Medical use of the compounds and / or compositions is also contemplated in the context of the present invention. provided herein in a "therapy" scenario, for example, in the case of disorders related to APP, such as Alzheimer's disease. In this case, the therapy with approved medications is contemplated, such as memantine, donepezil, rivastigmine or galantamine. In another embodiment, the present invention provides a kit comprising at least one glycosylated antibody molecule as defined herein or the mixture of the monoglycosylated and / or double glycosylated methods of the invention as provided herein. Advantageously, the kit of the present invention also optionally comprises (a) buffer (s), storage solutions and / or reagents or remaining materials required for the performance of assays and for medical, scientific or diagnostic purposes. In addition, parts of the kit of the invention can be packaged individually in vials or bottles or in combination in multi-pack containers or units. The kit of the present invention can be used advantageously, inter alia, to carry out the method of the invention and can be used in a variety of applications mentioned herein, for example, as diagnostic kits, as search tools or medical tools Additionally, the kit of the invention may contain means for detection suitable for scientific, medical and / or diagnostic purposes. The manufacture of the kits preferably follows standard procedures that are known to those skilled in the art.
The figures show: Figure 1A to 1C: Plasmid map showing the insertion of sites for heavy and light chain sequences. Figure 2: Example of an analytical chromatogram. Figure 3: Chromatogram of a CMT column as described in the text. The double glycosylated and mono-glycosylated isoforms are eluted in the double peak 1, the non-glycosylated isoform is eluted in peak 2. Figure 4A to 4C: Complete ESI-MS analysis of IgG isoforms of ANTIBODY A. The molecular mass of the main peak is indicates in Da. A: ANTIBODY A non-glycosylated; B: monoglycosylated ANTIBODY; C: Double glycosylated ANTIBODY. Figure 5: Scheme of N-glycosylation patterns of ANTIBODIES deducted. Structures that occur only partially are indicated in parentheses. A: Complex Type; B: Hybrid type; C: Oligomanosa type; GlcNAc = N-acetylglucosamma, Mannose; Gal = galactose; It was = fucose; NeuAc = N-acetyl-neuraminic acid. Figure 6: Schematic presentation of carbohydrate structures in Asn30ß of ANTIBODY A deduced from the MS and HPAEC-PAD analysis. Structures that occur only partially are indicated in parentheses. GlcNAc = N-acetyl-glucosamine, Man = crafty Gal = galactose; It was = fucose; NeuAc = N-acetyl-neuraminic acid. Figure 7: Union of isoforms from ANTIBODY A to Aß40 f.protilled bristle (Biacore sensor chip). Antibody concentration 60 nM. Also shown is the binding curve of a mixture of all isoforms, ie, before purification, as indicated. Figure 8A and 8B: Mapping of epitopes of the COMPOSITION OF ANTIBODIES A by pepspot analysis. A) Signs Pepspot of indicated individual superimposed decapeptide points; B) densitometpco analysis of the signal intensity of unique superimposed decapeptide points. Figure 9: Depolymerization test. The COMPOSITION OF ANTIBODIES A and the isoforms of ANTIBODIES A induce the release of biotinylated Aβ from aggregated Aβ. Figure 10: COMPOSITION OF ANTIBODIES A comprising isoforms of ANTIBODIES A that capture soluble Aβ from human cerebrospinal fluid (CSF). Average of 4 samples of CSF of patients with Alzheimer's disease analyzed in groups of 2. Two immunoprecipitations followed by Western transfers by combination with quantification of Aβ captured by densitometpa of transfers Western. The highest Aß value was taken in a series of Western transfers given as 100%. Figure 11: Indirect immunofluorescence staining of human amyloid plaques with isoforms of ANTIBODIES A in vi tro. Highly sensitive and specific detection of genuine ex vivo human β-amyloid plaques after staining with 10 ng / ml of ANTIBODY A concentration. The bound ANTIBODY A was revealed by goat anti-human Cy3 (H + L) for (A) COMPOSITION OF ANTIBODIES A; (B) Double glycosylated ANTIBODY; (C) monoglycosylated ANTIBODY; Y (D) Non-glycosylated ANTIBODY. Scale bar = 80 μm. Figure 12: In vivo immunodecoration of PS2APP transgenic mouse plates with glycosylated ANTIBODY A isoforms revealed by confocal microscopy. Immunodecoration reveals the in vivo binding of isoforms of ANTIBODIES TO 3 days after a single dose of 1 mg of isoforms of ANTIBODIES A. Representative images of the distribution of the isoforms of ANTIBODIES A are shown for the isoform of double-glycosylated ANTIBODIES (A), monoglycosylated (B) and non-glycosylated (C). Scale bar = μm. Figure 13: Analysis of binding of anti-Aβ antibodies with the APP of the cell surface. The Union of antibodies with HEK293 cells transfected to human APP and non-transfected control cells was analyzed by flow cytometry. Figure 14: Scheme of non-glycosylated, mono-glycosylated and double-glycosylated antibody molecules of ANTIBODY A (immunoglobulins).
Figure 15A A 15C: Total surface area of plaques (A), total number of plaques (B) and number and distribution of plaque size (C) in the thalamus region after a 5-month treatment with the COMPOSITION OF ANTIBODIES A (comprising monoglycosylated and double glycosylated ANTIBODY), double glycosylated and mono glycosylated ANTIBODY A isoforms (20 mg / kg weekly, i.v.) or vehicle. Figure 16A to 16C: Total area of plaques (A), total number of plaques (B) and number and distribution of plaque size (C) in the region of the cortex and corpus callosum after a 5-month treatment with COMPOSITION OF ANTIBODIES A (comprising monoglycosylated and doubly glycosylated ANTIBODY), double glycosylated and mono glycosylated ANTIBODY A isoforms (20 mg / kg weekly, iv) or vehicle. Figure 17A to 17C: Total surface of the plates (A), total number of plates (B) and number and distribution of size of the plates (C) in the hippocampal region after of a 5-month treatment with the COMPOSITION OF ANTIBODIES A (comprising monoglycosylated and double glycosylated ANTIBODY), double glycosylated and mono glycosylated ANTIBODY A isoforms (20 mg / kg weekly, v.v.) or vehicle. Figure 18A to 18C: Total surface of the plates (A), total number of plates (B) and number and distribution of size of the plates (C) in the subiculum region after a 5-month treatment with the COMPOSITION OF ANTIBODIES A (comprising monoglycosylated and double glycosylated ANTIBODY), isoforms of double glycosylated and mono glycosylated ANTIBODIES (20 mg / kg weekly, v.v.) or vehicle. Figure 19: Measurement of the fluorescence intensity of the COMPOSITION OF ANTIBODIES A mmunochemistry bound to ß-amyloid plaques after the biweekly dosage of 0.1 mg / kg with 1, 2 and 4 applications i.v. to PS2APP mice. The analysis was carried out 2 weeks after the last injection. Figure 20: Measurement of the fluorescent intensity of the immunosorbent COMPOSITION OF ANTIBODIES A bound to ß-amyloid plaques after monthly dosing of 0.15 mg / kg with 2 and 3 applications i.v. to PS2APP mice. The analysis was carried out 2 weeks after the last injection.
Figure 21: Measurement of the fluorescence intensity of the COMPOSITION OF ANTIBODIES A immunostained bound to β-amyloid plaques after 4 biweekly injections of 0.05, 0.1 and 0.30 mg / kg to PS2APP mice, which suggests the binding of the plaques of Amyloid related to the dose. The analysis was carried out 2 weeks after the last injection. Figure 22: Measurement of the fluorescence intensity of the COMPOSITION OF ANTIBODIES A immunostained bound to ß-amyloid plaques after 3 monthly injections of 0.075, 0.15 and 0.45 mg / kg to PS2APP mice, which suggests the union of the plaques of Amyloid related to the dose. The analysis was carried out 2 weeks after the last injection. Figure 23: Brain sections with human AD stained against Aβ with anti-Aβ monoclonal mouse antibody (BAP-2) after 40 hours of incubation with the COMPOSITION OF ANTIBODIES A at the indicated concentrations together with living differentially-labeled primary human macrophages (0.8 million cells / ml). The results indicate a reduction in the amyloid load indicating the effect of antigen-dependent cellular phagocytosis of the COMPOSITION OF ANTIBODIES A in ß-amyloid plaques. Scale bar = 300 μm.
Figure 24A and 24B: Response to the dose of the COMPOSITION OF ANTIBODIES A in ß-amyloid plaques from brain sections with human AD incubated with 0.8 million cells / ml. (A) shows the total area of the plates and (B) the intensity of the staining. Figure 25: Fluorescent microscopy of P388D1 cells incubated with 0, 0.1, 1 and 10 μg / ml COMPOSITION OF ANTIBODIES A (A to D, respectively). Figure 26: Quantitative measurement of the dose response of the COMPOSITION OF ANTIBODIES A using conjugated fluroperlas of Aß and P3881D1 cells (shown in relative fluorescent units, RFU). Two independent experiments indicate a considerable range of effectiveness for the COMPOSITION OF ANTIBODIES A. Figure 27: Table showing different glycan structures of ANTIBODY A in the constant region of the heavy chain (Asn 306, first two columns) and in the variable region of the heavy chain (Asn 52, third and fourth columns) ).
EXAMPLES The following non-limiting examples illustrate the invention.
Example 1: Generation of ANTIBODY A through cloning techniques In accordance with the present invention, an IgGl molecule was generated by common cloning techniques. ANTIBODY A is found in its coding sequence and in its expressed amino acid sequence characterized by its variable region of the heavy chain (VH). The corresponding example of a heavy chain encoded by a DNA sequence is as follows: caggtggaattggtggaaagcggcggcggcctggtgcaaccgggcggcagcctgcgtctga gctgcgcggcctccggatttacctttagcagctatgcgatgagctgggtgcgccaagcccc tgggaagggtctcgagtgggtgagcgctattaatgcttctggtactcgtacttattatgct gattctgttaagggtcgttttaccatttcacgtgataattcgaaaaacaccctgtatctgc aaatgaacagcctgcgtgcggaagatacggccgtgtattattgcgcgcgtggtaagggtaa tactcataagccttatggttatgttcgttattttgatgtttggggccaaggcaccctggtg acggttagctcagcctccaccaagggtccatcggtcttccccctggcaccctcctccaaga gcacctctgggggcacagcggccctgggctgcctggtcaaggactacttccccgaaccggt cagtcctcaggactctactccctcagcagcgtggtgaccgtgccctccagcagcttgggca gacggtgtcgtggaactcaggcgccctgaccagcggcgtgcacaccttcccggctgtceta cccagacctacatctgcaacgtgaatcacaagcccagcaacaccaaggtggacaagaaagt tgagcccagatatcgtgcgatatcgtgcaatcttgtgacaaaactcacacatgcccaccgt caceetcatgatctcccggacccctgaggtcacatgcgtggtggtggacgtgagccacgaa gcccagcacctgaactcctggggggaccgtcagtcttcctcttccccccaaaacccaagga gaccctgaggtcaagttcaactggtacgtggacggcgtggaggtgcataatgccaagacaa agccgcgggaggagcagtacaacagcacgtaccgggtggtcagcgtcctcaccgtcctgca ccaggactggctgaatggcaaggagtacaagtgcaaggtctccaacaaagccctcccagcc cccatcgagaaaaccatctccaaagccaaagggcagccccgagaaccacaggtgtacaece tgcccccatcccgggatgagctgaccaagaaccaggtcagcctgacctgcctggtcaaagg cttctatcccagcgacatcgccgtggagtgggagagcaatgggcagccggagaacaactac aagaccacgcctcccgtgctggactccgacggctccttcttcctctacagcaagctcaccg tggacaagagcaggtggcagcaggggaacgtcttctcatgctccgtgatgcatgaggctct gcacaaccactacacgcagaagagcctctccctgtctccgggtaaatga (SEQ ID NO: 5). and codes for the following immunoglobulin H chain: QVELVESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSAINASGTRTYYA DSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGKGNTHKPYGYVRYFDVWGQGTLV TVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVL QSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELL GGPSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQY NSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDE LTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQ QGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 6). The same heavy chain may also be encoded by a sequence comprising a "leader sequence" as further shown in the following sequence: atgaaacacctgtggttcttcctcctgctggtggcagctcccagatgggtcctgtcc caggtggaattggtggaaagcggcggcggcctggtgcaaccgggcggcagcctgcgtctga gctgcgcggcctccggatttacctttagcagctatgcgatgagctgggtgcgccaagcccc tgggaagggtctcgagtgggtgagcgctattaatgcttctggtactcgtacttattatgct gattctgttaagggtcgttttaccatttcacgtgataattcgaaaaacaccctgtatctgc aaatgaacagcctgcgtgcggaagatacggccgtgtattattgcgcgcgtggtaagggtaa tactcataagccttatggttatgttcgttattttgatgtttggggccaaggcaccctggtg acggttagctcagcctccaccaagggtccatcggtcttccccctggcaccctcctccaaga gcacctctgggggcacagcggccctgggctgcctggtcaaggactacttccccgaaccggt gacggtgtcgtggaactcaggcgccctgaccagcggcgtgcacaccttcccggctgtceta cagtcctcaggactctactccctcagcagcgtggtgaccgtgccctccagcagcttgggca cccagacctacatctgcaacgtgaatcacaagcccagcaacaccaaggtggacaagaaagt tgagcccaaatcttgtgacaaaactcacacatgcccaccgtgcccagcacctgaactcctg gggggaccgtcagtcttcctcttccccccaaaacccaaggacaccctcatgatctcccgga cccctgaggtcacatgcgtggtggtggacgtgagccacgaagaccctgaggtcaagttcaa aacagcacgtaccgggtggtcagcgtcctcaccgtcctgcaccaggactggctgaatggca ctggtacgtggacggcgtggaggtgcataatgccaagacaaagccgcgggaggagcagtac aggagtacaagtgcaaggtctccaacaaagccctcccagcccccatcgagaaaaccatctc caaagccaaagggcagccccgagaaccacaggtgtacaecetgcccccatcccggga gag ctgaccaagaaccaggtcagcctgacctgcctggtcaaaggcttctatcccagcgacatcg ccgtggagtgggagagcaatgggcagccggagaacaactacaagaccacgcctcccgtgct ggactccgacggctccttcttcctctacagcaagctcaccgtggacaagagcaggtggcag agagcctctccctgtctccgggtaaatga caggggaacgtcttctcatgctccgtgatgcatgaggctctgcacaaccactacacgcaga (SEQ ID NO: 25). The corresponding amino acid sequence would be: MKHLWFFLLLVAAPRWVLS QVELVESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSAINASGTRTYYA DSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGKGNTHKPYGYVRYFDVWGQGTLV TVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVL QSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELL GGPSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQY NSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDE LTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQ QGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 26). In a similar way, the light chain of ANTIBODY A is encoded by the following nucleotide sequence: gatatcgtgctgacccagagcccggcgaccctgagcctgtctccgggcgaacgtgcgaccc tgagctgcagagcgagccagagcgtgagcagcagctatctggcgtggtaccagcagaaacc aggtcaagcaccgcgtctattaatttatggcgcgagcagccgtgcaactggggtcccggcg cgttttagcggctctggatccggcacggattttaccctgaccattagcagcctggaacctg aagactttgcgacttattattgccttcagatttataatatgcctattacctttggccaggg tacgaaagttgaaattaaacgtacggtggctgcaccatctgtcttcatcttcccgccatct gatgagcagttgaaatctggaactgcctctgttgtgtgcctgctgaataacttctatccca gagaggccaaagtacagtggaaggtggataacgccctccaatcgggtaactcccaggagag tgtcacagagcaggacagcaaggacagcacctacagcctcagcagcaccctgacgctgagc aaagcagactacgagaaacacaaagtctacgcctgcgaagtcacccatcagggcctgagct cgcccgtcacaaagagcttcaacaggggagagtgttag (SEQ ID NO: 7). and encoding the following amino acid sequence (L-chain): DIVLTQSPATLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPRLLIYGASSRATGVPA RFSGSGSGTDFTLTISSLEPEDFATYYCLQIYNMPITFGQGTKVEIKRTVAAPSVFIFPPS DEQLKSGTASWCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLS KADYEKHKVYACEVTHQGLSSPVTKSFNRGEC (SEQ ID NO: 8). Again also here, Can employ a "leader sequence" and the corresponding sequences would be: atggtgttgcagacccaggtcttcatttctctgttgctctggatctctggtgcctacggg gatatcgtgctgacccagagcccggcgaccctgagcctgtctccgggcgaacgtgcgaccc tgagctgcagagcgagccagagcgtgagcagcagctatctggcgtggtaccagcagaaacc aggtcaagcaccgcgtctattaatttatggcgcgagcagccgtgcaactggggtcccggcg cgttttagcggctctggatccggcacggattttaccctgaccattagcagcctggaacctg aagactttgcgacttattattgccttcagatttataatatgcctattacctttggccaggg tacgaaagttgaaattaaacgtacggtggctgcaccatctgtcttcatcttcccgccatct gatgagcagttgaaatctggaactgcctctgttgtgtgcctgctgaataacttctatccca gagaggccaaagtacagtggaaggtggataacgccctccaatcgggtaactcccaggagag tgtcacagagcaggacagcaaggacagcacctacagcctcagcagcaccctgacgctgagc aaagcagactacgagaaacacaaagtctacgcctgcgaagtcacccatcagggcctgagct cgcccgtcacaaagagcttcaacaggggagagtgttag (SEQ ID NO: 27). This sequence encodes the following amino acid sequence: MVLQTQVFISLLLWISGAYG DIVLTQSPATLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPRLLIYGASSRATGVPA RFSGSGSGTDFTLTISSLEPEDFATYYCLQIYNMPITFGQGTKVEIKRTVAAPSVFIFPPS DEQLKSGTASWCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLS KADYEKHKVYACEVTHQGLSSPVTKSFNRGEC (SEQ ID NO: 28). These prior sequences are known from MAB31 as disclosed in 03/070760. However, the heavy and light chains of the exemplified ANTIBODY A may also be encoded by the sequences listed below: a) the heavy chain gaaatagagagactgagtgtgagtgaacatgagtgagaaaaactggatttgtgtggca atggagtttgggctgagctgggttttcctcgttgctcttttaagaggtgattcatgga ttttctgataacggtgtccttctgtttgcaggtgtccagtgtcaggtggagctggtgg agtctgggggaggcctggtccagcctggggggtccctgagactctcctgtgcagcgtc tggattcacettcagtagetatgccatgagctgggtccgccaggctccaggcaagggg ctcgagtgggtgtccgccataaacgccagcggtacccgcacctactatgcagactccg tgaagggccgattcaccatctccagagacaattccaagaacacgctgtatctgcaaat gaacagcctgagagccgaggacacggctgtgtattactgtgcgagaggcaaggggaac acccacaagccctacggctacgtacgctactttgacgtgtggggccaaggaaccctgg tcaccgtctcctcaggtgagtcctcacaacctctctcctgcggccgcagcttgaagtc tgaggcagaatcttgtccagggtctatcggactcttgtgagaattaggggctgacagt tgatggtgacaatttcagggtcagtgactgtctggtttctctgaggtgagactggaat ataggtcaccttgaagactaaagaggggtccaggggcttttctgcacaggcagggaac agaatgtggaacaatgacttgaatggttgattcttgtgtgacaccaagaattggcata atgtctgagttgcccaagggtgatcttagctagactctggggtttttgtcgggtacag aggaaaaacccactattgtgattactatgctatggactactggggtcaaggaacctca gtcaccgtctcctcaggtaagaatggcctctccaggtctttatttttaacctttgtta tggagttttctgagcattgcagactaatcttggatatttgccctgagggagccggctg agagaagttgggaaataaatctgtctagggatctcagagcctttaggacagattatct ccacatctttgaaaaactaagaatctgtgtgatggtgttggtggagtccctggatgat gggatagggactttggaggctcatttgagggagatgctaaaacaatcetatggctgga atacttcaaggaccacctctgtgacaaccattttatacagtatccaggcatagggaca gggatagttggggctgtagttggagattttcagtttttagaatgaagtattagctgca aaaagtggagtggggcactttctttagatttgtgaggaatgttccacactagattgtt taaaacttcatttgttggaaggagctgtcttagtgattgagtcaagggagaaaggcat ctagcctcggtctcaaaagggtagttgctgtctagagaggtctggtggagcctgcaaa agtccagctttcaaaggaacacagaagtatgtgtatggaatattagaagatgttgc tt ttactcttaagttggttcctaggaaaaatagttaaatactgtgactttaaaatgtgag agggttttcaagtactcatttttttaaatgtccaaaatttttgtcaatcaatttgagg tcttgtttgtgtagaactgacattacttaaagtttaaccgaggaatgggagtgaggct ctctcataccctattcagaactgacttttaacaataataaattaagtttaaaatattt ttaaatgaattgagcaatgttgagttgagtcaagatggccgatcagaaccggaacacc tgcagcagctggcaggaagcaggtcatgtggcaaggctatttggggaagggaaaataa aaccactaggtaaacttgtagctgtggtttgaagaagtggttttgaaacactctgtcc agccccaccaaaccgaaagtccaggctgagcaaaacaccacctgggtaatttgcattt ctaaaataagttgaggattcagccgaaactggagaggtcctcttttaacttattgagt tcaaccttttaattttagcttgagtagttctagtttccccaaacttaagtttatcgac ttctaaaatgtatttagaattcgagctcggtacagctttctggggcaggccaggcctg accttggctttggggcagggagggggctaaggtgaggcaggtggcgccagcaggtgca cacccaatgcccatgagcccagacactggacgctgaacctcgcggacagttaagaacc caggggcctctgcgcctgggcccagctctgtcccacaccgcggtcacatggcaccacc tctcttgcagcctccaccaagggcccatcggtcttccccctggcaccctcctccaaga gcacctctgggggcacagcggccctgggctgcctggtcaaggactacttccccgaacc ggtgacggtgtcgtggaactcaggcgccctgaccagcggcgtgcacaccttcccggct gtcctacagtcctcaggactctactccctcagcagcgtggtgaccgtgccctccagca gcttgggcacccagacctacatctgcaacgtgaatcacaagcccagcaacaccaaggt ggacaagaaagttggtgagaggccagcacagggagggagggtgtctgctggaagccag gctcagcgctcctgcctggacgcatcccggctatgcagccccagtccagggcagcaag gcaggccccgtctgcctcttcacccggagcctctgcccgccccactcatgctcaggga gagggtcttctggctttttcccaggctctgggcaggcacaggctaggtgcccctaacc caggccctgcacacaaaggggcaggtgctgggctcagacctgccaagagccatatccg ggaggaccctgcccctgacctaagcccaccccaaaggccaaactctccactccctcag ctcggacaccttctctcctcccagattccagtaactcccaatcttctctctgcagagc ccaaatcttgtgacaaaactcacacatgcccaccgtgcccaggtaagccagcccag gc ctcgccctccagctcaaggcgggacaggtgccctagagtagcctgcatccagggacag gccccagccgggtgctgacacgtccacctccatctcttcctcagcacctgaactcctg gggggaccgtcagtcttcctcttccccccaaaacccaaggacaccctcatgatctccc ggacccctgaggtcacatgcgtggtggtggacgtgagccacgaagaccctgaggtcaá gttcaactggtacgtggacggcgtggaggtgcataatgccaagacaaagccgcgggag gagcagtacaacagcacgtaccgtgtggtcagcgtcctcaccgtcctgcaccaggact ggctgaatggcaaggagtacaagtgcaaggtctccaacaaagccctcccagcccccat cgagaaaaccatctccaaagccaaaggtgggacccgtggggtgcgagggccacatgga cagaggccggctcggcccaccctctgccctgagagtgaccgctgtaccaacctctgtc cctacagggcagccccgagaaccacaggtgtacaecetgcccccatcccgggatgagc tgaccaagaaccaggtcagcctgacctgcctggtcaaaggcttctatcccagcgacat cgccgtggagtgggagagcaatgggcagccggagaacaactacaagaccacgcctccc gtgctggactccgacggctccttcttcctctacagcaagctcaccgtggacaagagca ggtggcagcaggggaacgtcttctcatgctccgtgatgcatgaggctctgcacaacca ctacacgcagaagagcctctccctgtccccgggcaaatga (SEQ ID NO: 23) light chain atggacatgagggtcctcgctcagctcctggggctcctgctgctctgtttcccagg taaggatggagaacactagcagtttactcagcccagggtgctcagtactgctttac tattcagggaaattctcttacaacatgattaattgtgtggacatttgtttttatgt ttccaatctcaggcgccagatgtgatatcgtgttgacgcagtctccagccaccctg tctttgtctccaggggaaagagccaccctctcctgccgggccagtcagagtgttag cagcagctacttagcctggtaccagcagaaacctggccaggcgcccaggctcctca tctatggcgcatccagcagggccactggcgtgccagccaggttcagtggcagtggg tctgggacagacttcactctcaccatcagcagcctggagcctgaagatttcgcgac ctattactgtctgcagatttacaacatgcctatcacgttcggccaagggaccaagg tggaaatcaaacgtgagtagaatttaaactttgcggccgcctagacgtttaagtgg gagatttggaggggatgaggaatgaaggaacttcaggatagaaaagggctgaagtc aagttcagctcctaaaatggatgtgggagcaaactttgaagataaactgaatgacc cagag gatgaaacagcgcagatcaaagaggggcctggagctctgagaagagaagga gactcatccgtgttgagtttccacaagtactgtcttgagttttgcaataaaagtgg gatagcagagttgagtgagccgtaggctgagttctctcttttgtctcctaagtttt tatgactacaaaaatcagtagtatgtcctgaaataatcattaagctgtttgaaagt atgactgcttgccatgtagataccatgtcttgctgaatgatcagaagaggtgtgac tcttattctaaaatttgtcacaaaatgtcaaaatgagagactctgtaggaacgagt ccttgacagacagctcaaggggtttttttcctttgtctcatttctacatgaaagta aatttgaaatgatcttttttattataagagtagaaatacagttgggtttgaactat atgttttaatggccacggttttgtaagacatttggtcctttgttttcccagttatt actcgattgtaattttatatcgccagcaatggactgaaacggtccgcaacctcttc tttacaactgggtgacctcgcggctgtgccagccatttggcgttcaccctgccgct aagggccatgtgaacccccgcggtagcatcccttgctccgcgtggaccactttcct gaggcacagtgataggaacagagccactaatctgaagagaacagagatgtgacaga ctacactaatgtgagaaaaacaaggaaagggtgacttattggagatttcagaaata aaatgcatttattattatattcccttattttaattttctattagggaattagaaag ggcataaactgctttatccagtgttatattaaaagcttaatgtatataatctttta gaggtaaaatctacagccagcaaaagtcatggtaaatattctttgactgaactctc actaaactcctctaaattatatgtcatattaactggttaaattaatataaatttgt gacatgaccttaactggttaggtaggatatttttcttcatgcaaaaatatgactaa taataatttagcacaaaaatatttcccaatactttaattctgtgatagaaaaatgt ttaactcagctactataatcccataattttgaaaactatttattagcttttgtgtt tgacccttccctagccaaaggcaactatttaaggaccctttaaaactcttgaaact actttagagtcattaagttatttaaccacttttaattactttaaaatgatgtcaat tcccttttaactattaatttattttaaggggggaaaggctgctcataattctattg tttttcttggtaaagaactctcagttttcgtttttactacctctgtcacccaagag ttggcatctcaacagaggggactttccgagaggccatctggcagttgcttaagatc agaagtgaagtctgccagttcctcccaggca ggtggcccagattacagttgacctg ttctggtgtggctaaaaattgtcccatgtggttacaaaccattagaccagggtctg atgaattgctcagaatatttctggacacccaaatacagaccctggcttaaggccct gtccatacagtaggtttagcttggctacaccaaaggaagccatacagaggctaata tcagagtattcttggaagagacaggagaaaatgaaagccagtttctgctcttacct tatgtgcttgtgttcagactcccaaacatcaggagtgtcagataaactggtctgaa tctctgtctgaagcatggaactgaaaagaatgtagtttcagggaagaaaggcaata gaaggaagcctgagaatacggatcaattctaaactctgagggggtcggatgacgtg gccattctttgcctaaagcattgagtttactgcaaggtcagaaaagcatgcaaagc cctcagaatggctgcaaagagctccaacaaaacaatttagaactttattaaggaat agggggaagctaggaagaaactcaaaacatcaagattttaaatacgcttcttggtc tccttgctataattatctgggataagcatgctgttttctgtctgtccctaacatgc cctgtgattatccgcaaacaacacacccaagggcagaactttgttacttaaacacc atcetgtttgcttctttcctcaggaactgtggctgcaccatctgtcttcatcttce cgccatctgatgagcagttgaaatctggaactgcctctgttgtgtgcctgctgaat aacttctatcccagagaggccaaagtacagtggaaggtggataacgccctccaatc gggtaactcccaggagagtgtcacagagcaggacagcaaggacagcacctacagcc tcagcagcaccctgacgctgagcaaagcagactacgagaaacacaaagtctacgcc tgcgaagtcacccatcagggcctgagctcgcccgtcacaaagagcttcaacagggg agagtgttag (SEQ ID NO: 24).
Example 1: Construction of vector The sequences for ANTIBODY A originated from the 2nd round of maturation after the primary selection of the MorphoSys HuCAL library, a synthetic phage display library. DNA for ANTIBODY A was originally provided in pMorph vectors of MorphoSys, Germany, and corresponds to the vectors expressing Fab in Figure 2 of WO 03/070760, appendix p. 6/43. In constructing the vector for the purposes of the present invention, vectors pEE6.1 and pEE 14.4 (both commercially available from Lonza Biologics) are encoded to obtain a structure with both chains in a vector, see Figure 1; see WO 87/04462 or WO 89/01036. The following cloning strategy was applied: The Ig kappa chain was isolated from MS-Roche vector # 7.9H7_Ig_kappa chain (as described in WO 03/070760) by PCR with the primer ACGTAAGCTTGCCGCCACCATGGTGTTGCAG (sense, HindIII, SEQ ID NO: 29) and the primer ACGTGAATTCCTAACACTCTCCCCTGTT ( antisense, EcoRI; SEQ ID NO: 30), inserted in pCR 2.1 TA mole and the insert was completely sequenced. The Ig kappa chain insert was removed from pCR Topo 2.1 by the Hindi / EcoRI digest and ligated to the vector pEE14.4 as a Hindi / EcoRI insert. The heavy gamma 1 Ig chain was cloned from the vector MS-Roche # 7.9H7_IgGl by PCR with the primer ACGTAAGCTTGCCGCCACCATGAAACACCTG (sense, HindIII, SEQ ID NO: 31) and the primer ACGTGAATTCTCATTTACCCGGAGACAG (antisense, EcoRI; SEQ ID NO: 32), inserted in pCR 2.1 Topo TA and the insert was completely sequenced. The heavy chain IgG 1 Ig insert was removed from pCR Topo 2.1 by the HindIII / EcoRI digest and ligated to the pEE 6.4 vector as a HindIII / EcoRI insert. The heavy chain expression cassette was removed from pEE 6.4 IgGl by the Notl / Sall digest and the isolated fragment was inserted into pEE 14.4 kappa digested by Sall / Notl which resulted in the final double gene structure pEE 14.4 mAb-31. Example 1.2: Transfection of CHO cells and expression of ANTIBODY A The transfections were carried out according to standard protocols. The CHO Kl host cell line is obtained from the working cell bank (WCB) of Lonza Biologics # 028-W2 (Lonza, 2002, 1-179) and the host cell line CHO Kl SV was obtained from the master cell bank (MCB) of Lonza Biologics # 269 -M (Lonza, 2003, 1-87). The adherent CHO Kl cells of WCB # 028-W2 were transfected with the pEE 14.4 Mab31 vector containing both kappa light chain and heavy chain genes by liposomal transfection (Fugene, Roche Diagnostics). Transfection isolates in DMEM, GS supplement (both JRH Biosciences), 10% dialyzed FCS (PAA Laboratories, CoS # R0-CEP 2001-083-Rev 00) and 50 μm methionine sulphoximine (MSX from Sigma) were selected. Two weeks later, the colonies were harvested and transferred to 96-well plates and subjected to ELISA testing for antibody production. Four colonies with higher ANTIBODY expression were cloned by serial limited dilution to obtain cultures obtained from a single cell, of which 82 clones were obtained after one week and expanded. One of these clones was selected as having the highest specific production rate of 48 pg / cell / day in an adherent state. Then it was subcloned by limited dilution to obtain the good producers that express ANTIBODY A with a high stability (Pu, (1998) Mol Biotechnol, 10, 17-25). Additionally, a suspension variant of CHO Kl cells, the CHO Kl SV cells of MCB # 269-M, was transfected with the vector pEE 14.4 MAb31 by electroporation. The transfectants were selected as before and the resulting clones were subjected to a cloning of a single cell limiting the dilution, which resulted in several clones of high producers of ANTIBODY A. Example 1.3. Adaptation of clones expressing ANTIBODY A to a suspension culture The optimal growth properties of the CHO Kl clones were determined in a DHI medium with different protein hydrolysates: the cells were finally adapted to the DHI medium without glutamine (Invitrogen), which is a mixture of DMEM, Ham 's F12 and IMDM in the respective proportions of 1: 2: 2 (v: v: v) (Schlaeger and Schumpp, 1992, JI munol Methods, 146, 111-20) with the following modifications : soy and rice hydrolyzate: 0. 2% HyPep 1510 soybeans and 0. 2% HyPep 5603 rice (Kerry Bioscience), 0.03% Pluronic F68 (Invitrogen), 25μM MSX (Sigma) and 5% dialysed FCS (PAA Laboratories). The concentration of FCS gradually decreased until the cells grew exponentially in a serum free DHI medium. The primary seed banks in the serum free DHI medium were frozen for several recombinant cell clones. The CHO Kl SV clones were adapted from DMEM containing 10% dialyzed FCS to a suspension culture in chemically defined CD-CHO medium with 25 μM MSX (Gibco-Invitrogen) in a two-step procedure (Lonza) . The cell banks were created in CD-CHO. Optionally, it was possible to use any other free means of serum and protein free for suspension culture and as a basis for antibody expression. Example 2: Production of ANTIBODY A Production of ANTIBODY A (fermentation by fed batch) CHO clones were prepared for fermentation from concentrated cultures in shake flasks or centrifuge cultures as follows: A cryovial was de-frozen from the clones in the respective culture medium containing 25 μM of MSX in a 100 ml shake flask or centrifuge with a nominal volume of 50-75 ml. Then, the cells were expanded in consecutive fractions of 1: 5 x to arrive at a concentrated culture of 400-500 ml volume in the shake flasks or centrifuges. The cells used for the inoculation of fermentors were obtained from these concentrated cultures up to 90 days after thawing. The sowing train consists of a 2x 1000 ml stage in 2L shake flasks or centrifuges, followed by the inoculation of a 10 L fermenter as another container.
Alternatively, the 10 L fermenter acted as batch container fed or as inoculated for the 100 L fed batch fermenter. MSX was present in the culture medium for selection until inoculation of the 10 L fermenter, where it was excluded. Fermentation process: Day 0: Start with 3-4xl05 / ml of cells (fraction 1: 4-1: 5 of the seed culture) Day 2-3: start of feeding, the density of the cells must be higher than 1.5xl06 / ml. Feeding: Continuous feeding or in the form of bolus at 2% per day. The composition of isoforms of ANTIBODY A was monitored throughout the fermentation by ion exchange chromatography (see below). Day 14-18: When the viability of the cells began to decrease (50%) and the expected titers were achieved, the cell supernatant was harvested by centrifugation and / or filtration and sterilized on a filter. It was aseptically stored and then processed as described in the next section. The fermentation was carried out in accordance with standard protocols; see, for example, Werner, (1993), Arzneimittelforschung, 43, 1242-9 or Rendall, (2003).
Proceedings of the 18th ESACT meeting, 11-14 May 2003, 1, 701-704).
Example 3: Purification of ANTIBODY A The purification process was based on three chromatographic steps and one diafiltration step: affinity chromatography with Protein A, cation exchange chromatography, anion exchange chromatography and diafiltration with a 100 kD membrane. The types of gels and the column sizes were 1 1 MabSelect (GE Healthcare, Art. 17-5199, column diameter 9 cm, bed length 18 +/- 2 cm), 0.4 1 CM-Toyopearl 650M (Toso Bioscience , Art. 007972, small ion capacity = 85 microequivalents / ml, diameter 5.0 cm, bed length 20 +/- 2 cm), 1.3 1 Q-Sepharose FF (GE Healthcare, Art. 17-0510-04), diameter 9 cm, bed length 20 +/- 2 cm. The columns worked at room temperature. The fractions were stored at 2-8 ° C. The detection was at 280 nm. A Biomax 100 ultrafiltration module with an area of 0.1 m2 (Millipore Corp. Art. P2B100A01) was used for concentration and diafiltration. Chromatography of Protein A The following solutions were prepared using purified water: Solution A (equilibration buffer): 25 mM Tris, 25 mM NaCl, 5 mM EDTA, regulated at pH 7.1 + / 0.1 by HCl Solution B (wash buffer 1 ): 100 M acetic acid regulated at pH 4.5 +/- 0.1 by NaOH Solution C (elution buffer): 100 mM acetic acid regulated at pH 3.2 +/- 0.1 by NaOH Solution D (wash buffer 2): 100 mM acetic acid, 75 mM NaCl, pH 3 +/- 0.1 Solution E: (regeneration buffer): 2 M guanidinium hydrochloride, 100 mM Tris, regulated at pH 7.5 +/- 0.1 by HCl Solution F (storage buffer): 200 mM benzyl alcohol, 100 mM acetic acid, regulated at pH 5.0 +/- 0.1. The column was first equilibrated with 3 bed volumes of solution A. Then, it was loaded with the clarified cell culture supernatant (45 1, 386 mg / l of antibody) was washed with 5 bed volumes of solution A, washed with 3 bed volumes of solution B, was eluted with 3.5 bed volumes of solution C and the eluate was collected, washed with 3 column volumes of solution D, and regenerated with 2 column volumes of solution E, equilibrated with 3 bed volumes of buffer A, and washed with bed volumes of buffer F for storage. A linear flow rate of 100 cm / h was used for all the chromatographic steps. The column loading was 17.4 g of antibody / 1 Mabselect gel and the yield of the total isoform mixture was 96%. Viral inactivation The following solution was prepared using purified water: Solution G (buffer): 2M sodium acetate The pH of the protein A eluate was regulated to a pH ranging from 3.5 to 3.7 by the addition of concentrated acetic acid or 2 M of sodium acetate (solution G). It stirred for 15 minutes and then it was adjusted to pH 4 +/- 0.1 by adding 2 M sodium acetate (solution G). Cation exchange chromatography The following solutions were prepared using purified water: Solution H (equilibrium buffer): 100 mM acetic acid regulated at pH 4.0 +/- 0.1 by NaOH, Solution I (elution buffer 1): 250 mM Sodium acetate without regulating the pH, pH 7.8-8.5, Solution J (elution buffer 2): 500 mM sodium acetate without regulating the pH, pH 7.8-8.5, Solution K (regeneration solution): 0.5 M hydroxide of sodium, Solution L (storage buffer): 0.01 M sodium hydroxide. The column was first regenerated with 2 bed volumes of the K solution and then equilibrated with 5 bed volumes of the H solution. Then, it was loaded with an amount of the protein A eluate and washed with 1 volume of solution bed. H. Enclosed, eluted with 6 volumes of solution bed I. In this step, a mixture of double glycosylated and mono-glycosylated isoforms was eluted. In the next step, 3 bed volumes of solution J were used to elute non-glycosylated isoforms. After use, the column was regenerated with 2 bed volumes of K solution, stored for 24 hours in this buffer and then washed again with 2 bed volumes of solution K. To store it, it was washed with 3 bed volumes of solution L. An example of a chromatogram is shown in Fig. 3. Fractions of the chromatography were analyzed by IEX analytic as described below. A linear flow rate of 100 cm / h was used for all the chromatographic steps. The column loading was 14.3 g of CM-Toyopearl 650 M antibody / 1, and the yield was 79% for the mixture of double glycosylated and mono-glycosylated isoforms and 6.2% for non-glycosylated isoforms. Continuous flow chromatography using Q-Sepharose FF The following solutions were prepared in purified water: Solution M (dilution buffer): 37.5 mM Tris, regulated at pH 7.9 +/- 0.1 acetic acid, Solution N (buffer): 2M Tris, Solution O (equilibrium buffer): 83 mM sodium acetate, 25 mM Tris, pH 7.5 +/- 0.1, Solution P (regeneration buffer 1): 0.5 M NaOH / l M NaCl, Solution Q (buffer of regeneration 2): 0.2 M acetic acid / 1 M NaCl, Solution R (storage buffer): 0.01 M NaOH.
The eluate of the CMT (acid) column was first diluted to 1: 3 with the M solution and then regulated to pH 7.5 with the N solution. The column was first equilibrated with 2 bed volumes of the O solution and then the diluted eluate of the CMT column was processed on the column and the continuous flow was collected. The product was removed by washing the column with the O solution until the absorption at 280 nm was less than 0.1 (continuous flow collected). The column was regenerated with 1.5 bed volumes of P solution, stored for 1 hour and then regenerated with another 1.5 bed volumes of P solution. The column was then regenerated with 2 bed volumes of Q solution and washed with 3 bed volumes of solution R and was stored. A linear flow rate of 100 cm / h was used for all the chromatographic steps. The loading of the column was 3.5 g of antibody / 1 Sepharose FF, and the yield was 91% for the mixture of double glycosylated and mono-glycosylated isoforms. Diafiltration The following solution was prepared using purified water: Solution S (diafiltration buffer): 20 mM Histidine, regulated at pH 5.5 by HCl. A filter holder Pellicon 2 (Millipore Corp.) was equipped with 1 ultrafiltration module type Biomax 100 (Millipore Corp., area = 0.1 m2, Art. P2B100A01). It was used a WATSON-MARLOW 501 U pump equipped with a slicona pipe for pumping. The system was rinsed with buffer O and then 3.8 liters (1.1 g antibody / 1) of the continuous flow of QS chromatography (regulated to pH 5.5 by concentrated acetic acid) were concentrated at 250-300 ml after 1 ha 4- 11 ° C. Then, a diafiltration (V = const.) was performed (4-11 °) against 3 liters of buffer S (approximately 10 volumes). Finally, the product was filtered in sterile form using a Millipac 20 filter (Millipore Corp.). The performance of the ultrafiltration / diafiltration stage was 91%. The concentration of the product was 15 mg / ml. The product could be frozen at -70 ° C. Analytical IEX method for the analysis of fractions Column: Mono-S HR 5/5 (GE Healthcare, Art. 17-0547-01) Buffer 1: 50 mM morpholinoethanesulfonic acid, regulated at pH 5.8 by sodium hydroxide Buffer 2: 50 mM morpholinoethanesulfonic acid, 1 M NaCl regulated at pH 5.8 by sodium hydroxide Flow rate: 1 ml / min Detection: 280 nm Sample charge: 36-72 μ Gradient: Time% buffer 2 0 min 0 1 0 25 63 27 63 28 O 35 O Fig. 2 presents an example of a chromatogram, yields Example 4: Characterization of isotypes of ANTIBODY A by SDS-PAGE The SDS-PAGE analysis was carried out using standard protocols with 4-12% Bis-Tris gel gradient NuPage (Invitrogen) and MARK12 marker (Invitrogen) as control. 1-3 ug of purified Protein A supernatants from fermentations (Prod 01, 02, 03) or centrifuge cultures (the other lanes) were loaded for each container. Analysis under reducing conditions resulted in a single band for peak 1 (double glycosylated ANTIBODY), a double band for peak 2 (monoglycosylated ANTIBODY) and a single band for peak 3 (non-glycosylated ANTIBODY A) in the molecular weight range of chains heavy. The molecular weights of the two bands of peak 2 corresponded to the molecular weights of peak 1 and peak 2, respectively. Similar results were obtained using various expression systems such as: transient transfection in HEK 293 EBNA cells, transient transfection in CHO cells and stable expression in CHO cells. Example 5: Characterization of isotypes of ANTIBODY A by mass spectrometry (MS) analysis A complete antibody mass profile of all isotypes of ANTIBODY A was determined by mass spectroscopy with electrospray ionization (ESI-MS). For this, samples of ANTIBODY A were prepared under non-reducing conditions. Samples were desalted in 2% formic acid and 40% acetonitrile by G25 gel filtration and used for ESI-MS analysis on a Q-Tof2 instrument or Waters LCT mass spectrometer. A separation by molecular mass is obtained with a difference of 1623 between the non-glycosylated ANTIBODY A and the monoglycosylated ANTIBODY A. The expected mass for the non-glycosylated ANTIBODY A of the amino acid sequence is 145.987 Da, which corresponds optimally with the experimentally determined mass of 145.979 Da. In a similar fashion, glycosylated and doubly glycosylated monocyte glycoprotein isoforms differ by 1624 Da, as indicated in Figure 4. The differences observed in the Molecular masses are compatible with the N-glycosylation patterns that are described in detail below. Example 6: Asn-52 glycosylation structure of ANTIBODY A Asn52 is part of the aaa-aaa-Asn-Ala-Ser-aaa-aaa sequence of the variable part of the heavy chain, corresponding to the consensus sequence of N- glycosylation Asn-aaa-Ser / Thr. The N-linked glycosylation of Asn52 was confirmed by mapping tryptic peptides of the ANTIBODY A isoforms and mass spectrometric evaluation of the HC / T4 peptide containing Asn52. In the triptych peptide maps of the non-glycosylated ANTIBODY, only one peptide appears that corresponds, according to the mass, to the non-glycosylated HC / T4 peptide, which indicates that Asn52 was not glycosylated, whereas in the monoglycosylated ANTIBODY A or doubly glycosylated peptides were detected, according to the mass, to the sugar structures linked to N containing HC / T4. To further confirm the glycosylation of the consensus sequence in the HC / T4 heavy chain tryptic peptide, the glycosylated HC / T4 peptide was isolated from the peptide maps of the glycosylated ANTIBODY A isoforms and analyzed by MALDI mass spectrometry before and after incubation with N-glycosidase. Before treatment with N-glycosidase F, masses were obtained that corresponded to HC / T4 peptide that contained structures of N-linked sugars. However, the mass of HC / T4 peptide treated with N- glycosidase F corresponded to the expected mass for non-glycosylated HC / T4 + 1 Da, as expected in the case of removing a sugar chain from asparagine by N-glycosidase F (Asn to Asp conversion). The presence of N-acetyl-neuraminic acids in the structures of sugars bound to Asn52 indicates, in addition, the presence of structures of hybrid and complex type sugars linked to N. For this, the isoforms of ANTIBODY A were treated with N-glycosidase F, which removes the N-sugar in Asn306, but not in Asn52, and with or without neuraminidase, and analyzed after separation of HC and LC by denaturation, reduction and desalination. The masses obtained for HC of both methods differed by approximately 291 Da or 582 Da corresponding to one or 2 sialic acids. From this, it is also concluded that the N-linked sugars of the complex and / or hybrid type were bound to Asn52. This glycosylation of Asn-52, an N-glycosylation, is composed predominantly of sugar structures of the biantennary complex type (>; 75%; mainly 80-90%) without fucosylation in the nucleus and highly sialidated with up to 80% of the complex type antenna containing N-acetyl-neuraminic acids. The secondary sugar structures belong to the hybrid biantennary type and the oligomannose type (< 25%), respectively (Figure 5 or Figure 27). Resistance to cleavage by the N-glycosidase F of intact ANTIBODY A was common to all the glycosylation structures of Asn52.
Example 7: Asn306 glycosylation structure of ANTIBODY A As indicated above, ANTIBODY A contained asparagine 306 (Asn306) in the Fc part of the heavy chain (HC) an antibody-like glycosylation composed of a chain of biantennary complex oligosaccharides. It is known that antibodies contain different isoforms of said complex biantennary oligosaccharide chain, which vary in the degree of terminal galactosylation, sialiation and in the degree of fucosylation of the nucleus. Furthermore, it is known that the degree of non-fucosylation of the nucleus in the sugar chain located in Fc is important for the in vivo efficacy of antibodies, since it has been accepted that the degree of fucosylation of the nucleus modulates effector functions of the antibodies. For ANTIBODY A, typical principal variations of antibodies were found (Routier (1997), Glyoconjugate 14 (2), 201-207, Raju (2003), BioProcess International, 44-52) in the sugar chains located in Fc together with Asn306 with respect to terminal galactosylation and core glycosylation. The heterogeneity in the degree of terminal galactosylation (G0: Gl: G2 structures) was determined in approximately 35-40% of G0 structures, approximately 45% Gl structures and approximately 15-20% of G2 structures (for the schematic demonstration of the structures see Figure 6 or Figure 27).
The content of Fc sugar structures that lack fucosylation, that is, that they lack the fucose unit attached to the innermost N-acetyl-glucosamine of the central sugar structure, is probably important for an antibody, since the presence or absence of this fucose unit can modulate the binding of the antibody to the Fc receptors of the effector cells, thus influencing the activity of these cells. For ANTIBODY A, the relative content of sugar chain isoforms lacking nucleus fucosylation in Asn 306 was determined by two different methods, as described below: A) Mass glycosylated HC mass spectrometry: Samples were denatured of ANTIBODY A and were reduced to a light chain (LC) and glycosylated HC in the presence of 6 M guanidine hydrochloride and 250 mM TCEP. The reduced samples were desalted in 2% formic acid and 40% acetonitrile and used for the ESI-MS analysis in a Q-Tof2 instrument or Waters LCT mass spectrometer. From the obtained m / z spectrum, the individual oligosaccharide isoforms were calculated by the peak height of glycosylated HCs containing the individual oligosaccharide isoforms of selected individual m / z states. For the calculation of the content of sugar structures lacking in fucosylation, the height of the fucose peak of the core lacking structure G0 (GO-Fuc) was related to the sum of G0 + (GO-Fuc).
The respective carbohydrate structures were assigned according to the differences of the masses obtained for glycosylated HC and for HC, whose oligosaccharide structures were removed by incubation with N-glycosidase F before the MS analysis in the control experiments. B) Chromatic analysis of oligosaccharides released by HPEAC-PAD: The samples of ANTIBODY A were incubated with N-glycosidase F in sodium phosphate buffer at pH 7.2 to release the oligosaccharide chains of Asn306 (the sugar structures in Asn52 do not they were removed from the undenatured antibody intact under the conditions used). The liberated sugar chains were separated from the ANTIBODY A protein by centrifugation filtration and analyzed on a Carbo Pac PA200 column from Dionex in a BioLC system., using a gradient of sodium acetate at a strong alkaline pH (pH 13). The column used was able to resolve the chains of non-fucosylated oligosaccharides of the fucosylated ones. The assignment of the individual peaks obtained to the respective carbohydrate structures was carried out by comparing the retention times with those of the appropriate oligosaccharide standards analyzed in the Carbo Pac PA200 column and by determining the molar mass of the separated and collected peaks. by MALDI mass spectrometry, respectively. For the calculation of the relative content of structures of sugar that lack fucosylation in the nucleus, the sum of the% area of all the structures lacking fucose in the nucleus was formed. Analysis of several lots (combinations of double glycosylated and monoglycosylated ANTIBODY A isoforms) and purified ANTIBODY A isoforms, respectively, revealed that the content of non-fucosylated Asn306-linked oligosaccharide chains ranged from ~ 14% to 27% (measured by MS) and between 6% and 26% (measured by HPAEC-PAD), respectively. Example 8: Determination of KD values for the COMPOSITION OF ANTIBODY A and isoforms (for example, non-glycosylated, monoglycosylated or double glycosylated antibody of the invention) that bind to the Aßl-40 and Aßl-42 in vi tro fibers by resonance of superficial plasmons (SPR) The binding of the fibrillated ANTIBODY A to Aβ was measured online by surface plasmon resonance (SPR) and the affinities of the molecular interactions were determined as follows: the Biacore2000 and Biacore3000 instruments were used for these measurements. The Aßl-40 and Aßl-42 fibers were generated in vi tro by inoculation of synthetic peptides at a concentration of 200 μg / ml in 10 mM sodium acetate buffer (pH 4.0) for three days at 37 ° C. Microscopic electron analysis confirmed a fibrillated structure for both peptides, and Aßl-40 showed predominantly short fibers (<1 micron) and predominantly long Aßl-42 fibers (> 1 micron). It was assumed that these fibers represented aggregated Aβ peptides in brain with human AD much better than mixtures defined for the disease of amorphous aggregates and unstructured precipitates. The fibers were diluted 1:10 and directly coupled to CM5 as described in the manufacturer's Instructions Manual (BIApplication Handbook, AB version, Biacore AB, Uppsala, 1998). The coupling procedure included an activation step, during which the carboxylic acid groups on the surface were transferred to chemically reactive succinimide ester groups by contacting the surface with an aqueous mixture of N-hydroxysuccinimide and 1-ethyl-hydrochloride. l- (3-diaminopropyl) -carbodiimide, and an immobilization step, during which the activated surface came in contact with the fibers dissolved in 10 mM acetate buffer (pH 4.5) at 200-350 resonance units (1 unit resonance (UR) corresponds approximately to a surface load of 1 picogram / mm2). The fiber-laden surface then came into contact with the antibody solutions in the concentration range of 200 nM > C > 0.15 nM. The typical curves of time-dependent responses (= sensograms) monitored during the association phase (during contact with buffer) and the dissociation phase (after contact with buffer) are shown in Figure 7. The KD values for the junction The fibers of Aßl-40 and Aßl-42 of the isoforms of ANTIBODY A are provided in the table that appears later. In summary, KD values were calculated by Scatchard type analysis using concentration-dependent equilibrium binding responses. These equilibrium binding constants were obtained in two ways. Due to the very slow process of association with a low concentration of antibodies, the contact intervals to reach equilibrium were very long (Figure 7). However, these contact intervals could be performed on Biacore instruments and the experimental responses to equilibrium were subjected to a Scatchard analysis. Equilibrium binding data was also obtained by extrapolating shorter time-dependent infinity association curves. Then, these theoretically obtained equilibrium bond levels were used again for the determination of the KD values. Regardless of the way to determine the equilibrium sensory responses, curvilinear Scatchard plots were obtained. From the curvilinear Scatchard plot, a higher (bivalent) and lower (monovalent) affinity interaction was obtained for the ANTIBODY A isoforms of the second affinity maturation cycle. These two affinities represent the upper and lower KD values of the range indicated in the following table: The above table presents the KD values of the low affinity (monovalent) complex and the high affinity (bivalent) complex formed by the interaction of the isoforms. of ANTIBODY A and Aßl-40 fibrils determined by surface plasmon resonance. Certain KD values are provided using extrapolated equilibrium responses (marked as "extrapolation") and determined KD values using experimentally determined equilibrium responses (marked "experimental"). Extrapolated values were determined at least six times and a standard deviation is provided. The KD values based on the experimental and extrapolated equilibrium sensor responses are equal within the limits given by these standard deviations. Example 9: Mapping of epitopes of the ANTIBODY COMPOSITION A and isoforms (eg, non-glycosylated, monoglycosylated or double glycosylated antibody of the invention) by Pepspot analysis with decapeptides An epitope (antigenic determinant) can be linear or conformational. The dual specificity of the epitope that is described herein was defined by reactivity of the antibody with two non-sequential linear peptides. The epitope mapping methods used to define the specific recognition of epitopes are based on ELISA technology with conjugates of hexapeptides spread on microplates or in Pepspot technology. This latter technology allows the detection and quantification of the antibody by protocols that are commonly known for the Western Transfer of proteins to PVDF membranes.
Applied epitope mapping technologies are designed to specifically detect linear epitopes, whereas they can not be applied to map more spatially complex epitopes such as discontinuous or conformational epitopes. The available techniques for the mapping of conformational or discontinuous epitopes, such as the exploration of domains and arrangements of combinatorial peptides, require extensive peptides of up to 36 amino acids. (domains) or combined peptides, each composed of 12 amino acids. Therefore, the applied techniques are considered specific for linear epitopes, not including that the conformational epitopes, either discontinuous or discontinuously distributed epitopes, are involved. In conclusion, the data presented show that the two regions within the Aβ peptide defined herein look like independent linear epitopes recognized simultaneously on the basis of the dual and unique epitope specificity of the antibodies investigated in individual hexameric or decameric Aβ peptides. . The following amino acid sequence comprising Aβ (1-42) was divided into 43 superimposed decapeptides with a frame shift of 1 amino acid. The numbers refer to the essential amino acids of the Aßl-40 sequence that must be present in the decapeptide for optimal binding of the antibody.
ISEVKM ^ -DAEF RHDSGYEVHH QKLVFFAEDV GSNKGAIIGL MVGGWI 2ATV IV (SEQ ID NO: 4). Accordingly, DAEF RHDSGYEVHH QKLVFFAEDV GSNKGAIIGL MVGGWIA (SEQ ID NO: 3) represents amino acids 1 to 42 of the Aβ4 / β-A4 peptide. The 43 decapeptides were synthesized with N-terminal acetylation and C-terminal covalent attachment to the cellulose sheet ("Pepspot") by a commercial supplier (Jerini BioTools, Berlin). The cellulose sheet was incubated for 2 hours on an oscillating platform with monoclonal antibody (1 μg / ml) in blocking buffer (50 mM Tris'HCl, 140 mM NaCl, 5 mM NaEDTA, 0.05% NP40 (Fluka), 0.25% gelatin (Sigma), 1% fraction of bovine serum albumin V (Sigma), pH 7.4). The sheet was washed 3 times for 3 minutes on an oscillating platform with TBS (10 mM Tris, HCl, 150 mM NaCl, pH 7.5). Then, it was pressed on filter paper, moistened with cathode buffer (25 mM Tris base, 40 mM 6-aminohexane acid, 0.01% SDS, 20% methanol) and transferred to a semi-dry transfer stack with the side of the peptide facing the PVDF membrane (Biorad) of equal size. The semi-dry transfer stack was composed of freshly moistened filter papers (Whatman NO.3) slightly larger than the peptide sheet: 3 papers moistened with cathode buffer the peptide sheet a PVDF membrane sheet moistened with methanol 3 papers moistened with anode buffer 1 (30 mM base Tris, 20% methanol) 3 papers moistened with 2 anode buffer (0.3 mM Tris base, 20% methanol). The transfer was effected at a cathode-anode current density of 0.8 mA / cm2 for one hour, which was sufficient to elute the antibody completely from the cellulose sheet and transfer it to the PVDF membrane. The PVDF membrane was immersed in blocking buffer for 10 minutes. Goat antihuman IgG (H + L) with IRdye800 fluorochrome (Rockland code # 609-132-123) was added at a 1: 10,000 dilution in Odyssey blocking buffer (Li-Cor) and then diluted to 1: 1 with PBS, 0.05% Tween20. The membrane was incubated on an oscillating platform for 1 hour. Was washed 3x10 minutes with TBST (TBS with 0.005% Tween20). The membrane was dried and screened for fluorescence at 800 nm using a long wavelength fluorescence scanner (Odyssey) as shown in Figure 8. Accurate assignment of reactive sites to the antibody was achieved by labeling the PVDF membrane through a needle puncture. The epitopes of the antibody in question were defined as the minimum amino acid sequence in reactive peptides. The intensity of the fluorescence was integrated at each point and recorded as the relative fluorescence unit (RFU). For comparison, two mouse monoclonal antibodies were analyzed (BAP-1 which is equivalent to the 6E10 antibody (Kim (1998)) with specificity for the N-terminal domain, and BAP-44 which is equivalent for the 4G8 antibody (Kim ( 1998)) with specificity for the middle domain) in the same manner, except that anti-mouse Ig was used instead of anti-human Ig for detection. It should be noted that maturation of the affinity and conversion of the monovalent Fab fragments into full-length IgGl antibodies usually result in some widening of the epitope recognition sequence as indicated by the Pepspot and ELISA analyzes. This may be related to the recruitment of more contact points in the area of antibody-antigen interaction as a result of affinity maturation or with a stronger binding to the minimal epitope, so that weak interactions with amino acids can also be detected. adjacent. The latter may be the case when Aβ-derived peptides were placed on a probe with full-length IgG antibodies. As illustrated in the table below, the recognition sequences of the N-terminal and central epitopes were extended up to three amino acids when comparing the mother Fabs and the corresponding fully mature IgG antibodies. However, it should be noted that the decapeptides were modified by covalent attachment at the C-terminal amino acid and, therefore, this amino acid may not be readily accessible to the full-length antibody due to a spherical hindrance. If this were the case, the C-terminal amino acid does not contribute significantly to the epitope recognition sequence, and in the Pepspot analysis that is used in the present invention should be considered a potential reduction of the minimum recognition sequence by an amino acid at the C-terminus.
The above table refers to the Pepspot analysis of full-length IgG antibodies to decapeptides on a cellulose sheet. The numbers refer to the position of the amino acid in the sequence Aßl-40 that must be present in the decapeptide for the binding of the antibody. Another extension to the epitope is indicated in parentheses to facilitate the flanking amino acids that are required to achieve maximum binding. Example 10: Depolymerization assay employing ANTIBODY A isoforms (eg, non-glycosylated, monoglycosylated and doubly glycosylated antibody of the invention) that induces the release of biotinylated Aβ from aggregated Aβ The experimental setup to evaluate the potential of the ANTIBODY isoforms A to induce the dissociation of Aβ aggregates was as follows: First, biotinylated Aßl-40 was incorporated into preformed Aßl-40 / Aßl-42 fibers before treatment with the ANTIBODY A isoforms. The release of biotinylated Aβ was measured using an assay that employs streptavidin-POD conjugate as described below. When incubated in aqueous buffer for several days, Synthetic Aß spontaneously aggregates and forms fibrillated structures that are similar to those observed in amyloid deposits in the brains of patients with Alzheimer's disease. The following in vi tro assay is suitable for measuring the incorporation or release of biotinylated Aβ into preformed Aβ aggregates to analyze the potential Aß neutralizing of anti-Aβ antibodies and other Aβ binding proteins such as albumin (Bohrmann (1999) J. Biol. Chem. 274, 15990-15995). The isoforms of ANTIBODY A induced the depolymerization of aggregated Aβ measured through the release of incorporated biotinylated Aßl-40.
Experimental procedure: NUNC Maxisorb microtiter plates (MTP) were coated with a 1: 1 mixture of Aßl-40 and Aßl-42 (2 μM each, 10 μl per container) at 37 ° C for three days. Under these conditions, highly aggregated fibrillated Aß was adsorbed and immobilized on the surface of each container. Then, the coating solution was removed and the plates were dried at room temperature for 2-4 hours. The dried plates could be stored at -20 ° C. For the incorporation of biotinylated Aβ, the coated plates were incubated with 200 μl / 20 nM container of biotinylated Aßl-40 in TBS containing 0.05% NaN3 at 37 ° C overnight. After washing the plate with 3 x 300 μl / container of T-PBS, antibodies serially diluted in TBS containing 0.05% NaN3 were added and incubated at 37 ° C for 3 hours. The plate was washed and analyzed for the presence of biotinylated Aßl-40. After washing 3 x with 300 μl of T-PBS, a streptavidin-POD conjugate (Roche Molecular Biochemicals), diluted 1: 1000 in T-PBS, was added. contained 1% BSA (100 μl / container) and incubated at room temperature for 2 hours. The vessels were washed 3x with T-PBS and 100 μl / container of freshly prepared tetramethylbenzidine (TMB) solution was added. [Preparation of the TMB solution: 10 ml of 30 mM citric acid pH 4.1 (regulated with KOH) + 0.5 ml of TMB (12 mg of TMB in 1 ml of acetone + 9 ml of methanol) + 0.01 ml of 35% H202 .] The reaction was stopped by adding 100 μl / 1 N H2SO vessel and the absorbance was read at 450 nm in a microtiter plate reader. As documented in Figure 9, the isoforms of ANTIBODY A induced the dissociation of added Aβ measured through the release of incorporated biotinylated Aßl-40. The isoforms of ANTIBODY A and the mouse monoclonal antibody BAP-1 were similarly active (Figure 9), whereas antibodies BAP-2, BAP-17 and 4G8 were clearly less effective at releasing biotinylated Aβ from the immobilized Aβ mass ( the data is not shown). BAP-1 could be clearly differentiated from the glycosylated ANTIBODY A isoforms by its reactivity with the total length APP of the cell surface. Antibodies such as BAP-1 with such properties are not useful for therapeutic applications since potential autoimmune reactions can be induced. It is interesting to note that BAP-2, despite its specificity for the amino acid residue 4-6 that was exposed in the aggregated Aβ, has a significantly lower activity in this assay, which indicates that not all specific antibodies of the N term are a priori equally effective in releasing Aβ from preformed aggregates. The relatively low efficacy of BAP-17 (specific for the C term) and 4G8 (specific for amino acid residues 16-24) in this assay is due to the cryptic nature of these two epitopes in aggregated Aβ. The BSA at the concentrations used herein had no effect on the aggregated Aβ. The monoglycosylated isoform had a greater capacity compared to the double glycosylated isoform to depolymerize the aggregated Aβ peptide in vi tro that may also be relevant in vivo. Example 11: COMPOSITION OF ANTIBODY A comprising isoforms (monoglycosylated and double glycosylated antibody of the invention) capturing soluble Aβ from human cerebrospinal fluid (CSF) The ability to capture soluble Aβ from human CSF samples was determined by immunoprecipitation (IP) and analysis by Western transfer (WB) semiquantitative. Experimental procedure: The immunoprecipitation of human CFS samples was carried out according to the following scheme: 70 ul of human CSF 20 uL of incubation buffer (50 mM Tris, 140 mM NaCl, 5 mM EDTA, 0.05% NP -40, 1% BSA, 0.25% gelatin, 0.25% milk powder, pH 7.2 ul of ANTIBODY A isoforms of concentrated solutions (1000-10 ug / ml) 100 ul. The solution was maintained for one hour at 4 ° C. 40 ul of G Sepharose beads (Amersham Biosciences # 17-0618-01, washed with PBS, 50% aqueous suspension) were added and incubated for 2 hours at 4 ° C on a rotator. After centrifugation at 500 g for 3 minutes at 4 ° C, the supernatant was removed and 200 μl of PBS was added to the beads, transferred to Millipore tubes of 0.45 μm (Millipore # UFC30HVNB) and centrifuged at 500 g for 3 minutes at 4 ° C. 200 ul of additional PBS was added to the beads, vortexed and centrifuged at 2000 g for 3 minutes at 4 ° C. 45 ul of sample buffer lxNuPage was added with DTT and kept for 10 minutes at 70 ° C followed by centrifugation at 2000 g for 3 minutes at 4 ° C. For SDS-PAGE, 18 ul of protein G eluate was applied to the NuPage 10% gel Bis-Tris gel together with Aβ? _42 (Bachem) as internal standard directly in the sample buffer as standard and passed through a MES buffer system . The gel was transferred to an extra Hybond C membrane (semi-dry Novex system), dry membrane 3 'at room temperature. The membrane was transferred to pre-warmed PBS and heated in the microwave for 3 minutes at 600 W. Blocking occurred for 1 hour with a SuperBlock solution.
(Pierce) and an additional blocking for 1 hour with 5% milk powder (Bio Rad) in T-PBS (0.1% Tween20 in PBS). Incubation occurred overnight with anti-Aβ W02 Antibody antibody (1: 1500-1: 2000 from The Genetics, Inc., Zürich, Switzerland) at 4 ° C on a rotator, followed by 3x washing with T-PBS for 5 minutes. minutes and incubation for 2 hours at RT with anti-mouse IgG-HRP (Dako) 1: 5000 in T-PBS. To another 3x wash with T-PBS for 5 minutes followed by incubation with LumiLight Plus for 5 minutes at RT. Western blots were digitized and analyzed by densometry with an Alpha Innotech Digital Camera System. As documented in Figure 10, the COMPOSITION OF ANTIBODY A (comprising isoforms of monoglycosylated and doubly glycosylated ANTIBODY) effectively linked to soluble Aβ in human CSF as demonstrated by immunoprecipitation and Western blot experiments. Notably, in this assay, monoglycosylated ANTIBODY A was more effective in capturing soluble Aβ than double glycosylated ANTIBODY (Figure 10).
Example 12: Immunostaining in vi tro of human amyloid plaques by COMPOSITION OF ANTIBODY A and isoforms (eg, unglycosylated, monoglycosylated or doubly glycosylated antibody of the invention) The glycosylated ANTIBODY A isoforms were tested for capacity to stain genuine human β-amyloid plaques obtained from sections of the Brain of patients with severe Alzheimer's disease by immunohistochemistry analysis using indirect immunofluorescence. Specific and sensitive staining of genuine human β-amyloid plaques was demonstrated. Cryostat sections of unfixed tissue from the temporal cortex obtained post-mortem from a patient who had been diagnosed with Alzheimer's disease were labeled with indirect immunofluorescence. An incubation of two successive stages was used to detect ligated ANTIBODY A isoforms, revealed by goat antihuman IgG (H + L) purified by affinity conjugated with Cy3 (# 109-165-003, lot 49353, Jackson Immuno Reseairch ). The controls included unrelated human IgGl antibodies (Sigma) and the secondary antibody alone, and all of them gave negative results. All types of ß-amyloid plaques were sensitive and specifically detected and consistently revealed at a concentration of ANTIBODY A of 10 ng / ml (Figure 11). Specific and sensitive staining of genuine human β-amyloid plaques is demonstrated for glycosylated ANTIBODY A isoforms at a concentration of up to 1 ug / ml. At a concentration of 10 ug / ml a background staining was observed, more prominent with the isoform of non-glycosylated ANTIBODY. The non-glycosylated isoform had a considerable non-specific viscosity observed on the surface of the glass slides and almost all the tissue components that were exposed after in vi tro sectioning. This appeared to be due to non-specific binding comprising ionic and / or hydrophobic interactions. Example 13: Decoration of β-amyloid plaques by ANTIBODY A in a mouse model of Alzheimer's disease. Glycosylated ANTIBODY A isoforms were tested in a single-dose study in PS2APP double transgenic mice (Richards (2003), J. Neuroscience, 23, 8989-9003) to determine its ability to immunoblot β-amyloid plaques in vivo. The glycosylated ANTIBODY A isoforms were administered at a dose of 1 mg / mouse and after 3 days the animals were perfused with a phosphate buffered saline solution and the brains were frozen in dry ice and prepared for cryosection. Both glycosylated isoforms demonstrated improved and highly effective penetration into the brain in vivo (compared to the non-glycosylated form). Effective penetration into the brain and specific binding to β-amyloid plaques was demonstrated in PS2APP mice, a mouse model for AD-related amyloidosis. The presence of antibodies bound to β-amyloid plaques was determined using unfixed cryostat sections either by indirect immunofluorescence individually labeled with goat antihuman IgG (H + L) conjugate with Cy3 (# 109-165-003, Jackson Immuno Research) shown in Figure 12 or followed by counterstaining with BAP-2-Alexa488 immunoconjugate to visualize the position and distribution of all types of ß-amyloid plaques present in the tissue. An immunofluorescent staining method was used to detect bound ANTIBODY A. After adhesion to previously cooled glass slides, the sections were hydrated in PBS and treated with 100% acetone previously cooled at -20 ° C for 2 minutes. Washing with PBS was performed twice for two minutes. Blocking of non-specific binding sites was performed either with PBS containing 1% BSA or by sequential incubation in Ultra V Block (LabVision) for 5 minutes followed by a wash with PBS and incubation in a powder blocking solution ( BioGenex) with 10% normal sheep serum for 20 minutes. After washing with PBS with 10% normal sheep serum, the slides were incubated with affinity purified goat antihuman IgG (H + L) conjugated with Cy3 (# 109-165-003, lot 49353, Jackson Immuno Research) at a concentration of 15 μg / ml for 1 hour at room temperature. A counterstain was performed to determine the amyloid plaques by incubation with BAP-2, a mouse monoclonal antibody against Ab conjugated to Alexa 488 at a concentration of 0.5 μg / ml for 1 hour at room temperature. The autofluorescence of lipofuscin was tempered by incubation in 4 mM of CuS04 in 50 mM of ammonium acetate. After rinsing the slides in bidistilled water and washing them with 2 x 500 μl / PBS slide, the slides were embedded with a fluorescence mounting medium (S3023 Dako). Imaging was performed using confocal laser microscopy and image processing for the quantitative analysis of colocalizations using the IMARIS and COLOCALIZATION software (Bitplane, Switzerland). After a single dose of 1 mg per mouse, glycosylated ANTIBODY A isoforms were found to penetrate the blood-brain barrier and effectively immunodecorated / ligated all β-amyloid plaques after three days in vivo. Representative images are indicated in Figure 12. This differs clearly from the non-glycosylated form that is not detectable in the amyloid plaques.
Example 14: Investigation of the binding of isotype isoforms of ANTIBODY A with amyloid precursor protein (APP) expressed on the surface of HEK293 cells: The flow cytometry method is known in the art. The relative fluorescence units (for example, FL1-H) measured by flow cytometry indicated the binding of the respective antibody to the surface of the cell. A variation of fluorescence in HEP293 cells transfected to APP compared to non-transfected HEK293 cells indicated undesired reaction with APP of the cell surface. As an example, antibodies BAP-1 and BAP-2 against the N-terminal domain exhibited a significant variation of the FL-1 signal in HEK293 / APP (Figure 13, thick line, right panel) compared to the non-transfected HEK293 cells (Figure 13, dotted lines, right panel). In a similar manner, the BAP-44 antibody (specific for the medium A-beta epitope) exhibited a similar size variation. In contrast, all of the ANTIBODY A isoforms (Figure 13 left panel) (specific for the N-terminal and middle A-beta epitopes) did not demonstrate significant variation in fluorescence. The non-transfected HEK293 cells had a higher basal fluorescence than the cells transfected to APP, due to different properties of cell size and surface. An instrument was used FACScan in combination with the Cellquest Pro Software package (both by Becton Dickinson). The ANTIBODY A isoforms were free of reactivity to the cell surface APP (Figure 13). Example 15: Morphometric analysis of plaque deposition of Aβ in a mouse model of Alzheimer's disease The ability of the COMPOSITION OF ANTIBODY A or the isoforms of ANTIBODY A to decrease amyloidosis in vivo was studied in several regions of the brain ( thalamus, neocortex, hippocampus and subiculum), using computer-assisted quantitative analysis of the brains of PS2APP mice that received a five-month treatment with the ANTIBODY COMPOSITION A or the ANTIBODY A isoforms.
Therefore, the PS2APP transgenic male mice were injected i.v. with COMPOSITION OF ANTIBODY A or isoforms of ANTIBODY A and vehicle. Seventy-five PS2APP mice of 5-6 months of age were divided into five groups (A-E), composed of 15 mice each. Beginning on day 0, each mouse received 0.1 mL of vehicle (0 mg / kg) or preparations of ANTIBODY A (20 mg / kg) by i.v. by bolus through the vein of the tail. Groups A, B, C, D and E of the PS2APP mice received vehicle (histidine-buffered saline), ANTIBODY COMPOSITION A comprising monoglycosylated ANTIBODY A and double glycosylated ANTIBODY A and lacking unglycosylated ANTIBODY A as previously defined, double glycosylated ANTIBODY, monoglycosylated ANTIBODY and non-glycosylated ANTIBODY, respectively. Immunotolerance was induced against human anti-Aβ antibodies administered by injecting anti-CD4 antibody (hybridoma clone GK 1.5 commercially available from ATCC). Antibody antibody monitoring indicated that animals treated with antibody only developed a moderate immune response after more than 16 weeks of treatment and that detectable antibodies have low affinity or occur only in low amounts (data not shown). After 5 months of treatment, the mice were sacrificed. The unfixed brains were sectioned sagittally, including the thalamic area, hippocampal formation and cortical. From each hemisphere of the brain, 50 sections were prepared as follows: Starting at the lateral level -1.92, 5 consecutive series of 5x 10 μm and 5x 20 μm sections were obtained. There was no gap between consecutive sections, which resulted in a total tissue usage of 750 μm. Therefore, the series of sections ends approximately at the lateral level 1.20 (Paxinos and Franklin, 2003). For the quantitative morphometric analysis, 10 sections were used. Sections were stained with double glycosylated ANTIBODY A isoform at a concentration of 5 μg / ml to determine the amyloid deposits. Staining against Aβ using mouse monoclonal antibody (BAP-2) conjugated with fluorophore Alexa-488 at 5 μg / ml showed comparable results, although with intracellular and significant background staining of neurons that interfered with the image processing routine that was describe later. For detection, a goat anti-human IgG (GAH) purified by affinity (H + L) conjugated with Cy3 (# 109-165-003) was applied, lot 49353, Jackson Immuno Research) at a concentration of 15 μg / ml for 1 hour at room temperature. After washing with 2 x 500 μl of PBS / slide, the slides were embedded with fluorescence mounting medium (S3023, Dako). The images were acquired using a GenePix Personal 4100A microarray scanner (Axon Instruments, now Molecular Devices, CA, USA). The load was measured and the number of β-amyloid plaques using two parameters, that is, the percentage of area covered by β-amyloid plaques and the number of β-amyloid plaques, using a morphometric method without margin of error by means of assisted image analysis by computer. The quantification of the charge and the number of plates was made with the MCID M7 Elite software (Imaging Research Inc., St. Catherines / Notary, Canada). The scanned images were enhanced with a detail extractor filter followed by an objective Accent filter. Then, the resulting image was binarized, adjusting the threshold according to the intensity of staining. Artifacts, blood vessels and edge effects were identified in the original reference image and then removed from the binarized image. The regions of interest were marked in the reference image. For the final quantification, we then measured the area of these regions and the area occupied by the plates, as well as the number of plates, in the binarized image. The individual pixels were ignored. Calculations were made with the common spreadsheet software (Microsoft Excel, Redmond / WA, USA). The size of the plates was separated into 11 groups ranging from < 100 and > 1000 μm2. A statistical evaluation was performed using a two-tailed heteroscedastic test. For comparison and statistical evaluation, the baseline of amyloidosis (pathology of β-amyloid plaques) was determined at the beginning of the study with a cohort (15 animals) of untreated 6-month-old PS2APP mice. The results are described in Figures 15 to 18 with levels of significance (*: p = 0.05; **: p = O.Ol; ***: p = 0.001). The reduction of amyloid plaques was more pronounced in the thalamus region (Figure 15). The average reduction in the surface area of ß-amyloid plaques was determined for the groups treated with antibody: 64% for the ANTIBODY A COMPOSITION, 70% for the double glycosylated ANTIBODY, 81% for the mono-glycosylated ANTIBODY and 44% for the ANTIBODY A non-glycosylated. The average reduction in the total number of β-amyloid plaques was 70% for ANTIBODY COMPOSITION A, 78% for double glycosylated ANTIBODY, 82% for monoglycosylated ANTIBODY A and 36% for non-glycosylated ANTIBODY. Note that the significance for the non-glycosylated ANTIBODY A was low and the observed variations were considerable. The reduction of the amyloid plaques in the neocortical region together with the corpus callosum is described in Figure 16. The average reduction of the total surface area of the β-amyloid plaque was determined for the antibody-treated groups: 19% for the COMPOSITION OF ANTIBODY A, 27% for the double glycosylated ANTIBODY, 30% for the monoglycosylated ANTIBODY A and 10% for the non-glycosylated ANTIBODY. It was found that the average reduction in the total number of β-amyloid plaques was 40% for the COMPOSITION OF ANTIBODY A, 46% for the double glycosylated ANTIBODY, 42% for the monoglycosylated ANTIBODY A and 11% for the non-glycosylated ANTIBODY. The reduction of amyloid plaques in the entire hippocampal region is described in Figure 17. The average reduction of the total surface area of β-amyloid plaques was determined for the groups treated with antibody: 12% for the COMPOSITION OF ANTIBODY A, 24% for the double glycosylated ANTIBODY, 24% for the monoglycosylated ANTIBODY A and 6% for the non-glycosylated ANTIBODY. The average reduction in the total number of ß-amyloid plaques was 36% for ANTIBODY COMPOSITION A, 46% for the double glycosylated ANTIBODY, 37% for the monoglycosylated ANTIBODY A and 3% for the non-glycosylated ANTIBODY. The reduction of amyloid plaques in the subiculum, a region of high susceptibility to amyloidosis, is shown in Figure 18. The average reduction in the total surface area of the β-amyloid plaques was determined for the antibody-treated groups: 2% for the COMPOSITION OF ANTIBODY A, 12% for the double glycosylated ANTIBODY, 5% for the monoglycosylated ANTIBODY A and 1% for the non-glycosylated ANTIBODY. It was found that the average reduction in the total number of β-amyloid plaques was 22% for ANTIBODY COMPOSITION A, 36% for double glycosylated ANTIBODY, 13% for monoglycosylated ANTIBODY A and 1% for ANTIBODY A not glycosylated The COMPOSITION OF ANTIBODY A and the isoforms of N- Main glycosylation (double glycosylated ANTIBODY and monoglycosylated ANTIBODY A) demonstrated a comparable efficacy to decrease the β-amyloid plaque burden and the number of plaques. The reduction in plaque burden was more pronounced and statistically significant in regions with low to moderate amyloidosis. In general, it was found that the reduction in the number of β-amyloid plaques was statistically significant after treatment with ANTIBODY COMPOSITION A and both comprised glycosylated isoforms Asn52 of ANTIBODY A in all measured regions of the brain. In contrast, only a minor effect on the number of β-amyloid plaques in the thalamus was found and no significant effect on the number of β-amyloid plaques was found in other regions of the brain investigated after isoform treatment. Non-glycosylated antibody A, which is excluded from the COMPOSITION OF ANTIBODY A after the purification detailed in the invention. We also investigated the potency of the clearance of the plates in relation to the size of the plates. In general, it was found that the effectiveness of the human anti-Aβ antibodies tested was more pronounced for the clearance of small β-amyloid plaques. This was observed in all regions of the brain (Figures 15 C, 16 C, 17 C, 17 C and 18 C). On the contrary, there was only a minimal or non-significant trend observed for the non-glycosylated isoform of ANTIBODY A.
The comparative analysis of ANTIBODY A and the major glycosylation isoforms of Asn52 demonstrated a capacity comparable to a lower plate load, while the non-glycosylated isoform has no significant effect on plaque depletion. Example 16: Pharmacokinetics of in vivo binding of ANTIBODY COMPOSITION A to β-amyloid plaques Two dosing frequencies were compared in order to investigate the binding kinetics of ANTIBODY COMPOSITION A, comprising monoglycosylated ANTIBODY A and ANTIBODY A doubly glycosylated, and lacking ANTI-glycosylated ANTIBODY A as defined above. Therefore, i.v. to transgenic PS2APP male mice with COMPOSITION OF ANTIBODY A through the tail vein either 4 times in biweekly intervals at 0.05, 0.1 and 0.3 mg / kg or 3 times with a monthly interval with 0.075, 0.15 and 0.45 mg / kg. For comparison, 0.1 mg / kg was administered once and twice with biweekly intervals and 0.15 mg / kg with monthly interval. After administration, all mice were sacrificed two weeks after the last dosage. Unsecreted PS2APP brain tissue was prepared for sagittal sectioning between ~ 1.92 and 1.2 mm laterally according to Paxinos and Franklin, including thalamic, cortical and hippocampal areas. The brains were sectioned at 40 μm using cryostat.
An ex vivo immunostaining method was used by immunofluorescence to detect bound ANTIBODY A COMPOSITION antibodies. Therefore, the brains were sectioned and incubated with the detection antibody, an affinity purified goat antihuman (GAH) IgG (H + L) conjugated with Cy3 (# 109-165-003, lot 49353, Jackson Immuno Research ) at a concentration of 15 μg / ml for 1 hour at room temperature. A counterstain was performed for ß-amyloid plaques by incubation with BAP-2, a mouse monoclonal antibody against Aβ conjugated with the fluorophore Alexa488 at a concentration of 0.5 μg / ml for 1 hour at room temperature. Images were recorded in the occipital cortex near the cerebellum using a Leica TCS SP2 AOBS confocal laser scanning microscope, as described above. Computer-assisted image processing was performed using the IMARIS software (Bitplane, Switzerland). The plate images were first selected using the "crop" function of the software for lower doses, except for the two higher doses of 0.3 and 0.45 mg / kg that required a different acquisition configuration for the linear signal recording. The SURPASS function was used to select positive voxels after the threshold (T) as a reading for GAH-Cy3 bound at the site of β-amyloid plaques. The threshold settings were 19 and 12 for lower and higher dose groups, respectively. As a control for Specificity of ß-amyloid plaques, after double labeling the images of GAH-Cy3 stains were compared with images of plates stained with mouse monoclonal BAP2 conjugated to Alexa488 and recorded in a different channel. A descriptive statistic was made for the quantitative description of all the images with the MeasurementPro IMARIS software module. The mean voxel fluorescence intensity (MVI) values of selected β-amyloid plaques were determined in the low dose groups or the total imaging signal in the high dose groups. The baseline MVI (B) is due to the instrumental noise, the diffusion signal in the tissue and the autofluorescence of lipofuscin. From the background correction, B was determined by measuring the average signal intensity in areas separated from the β-amyloid plaques and subsequently subtracted from the LVM of all the measured images (MVI - B = S). The intensity values of the signal (S) that resembled the average intensities or plates were obtained from 3 to 4 images of each brain section per mouse and dose group. For comparison, the signal intensities were normalized to a reference sample obtained from a previous study. As a reference, PS2APP mouse brain sections were used after a single dose administration of 0.25 mg / kg. The final point of the measurement was one week after dosing.
All values of the measured intensity were normalized to the average intensity in the β-amyloid plaques obtained after a single dose administration of 0.25 mg / kg of ANTIBODY COMPOSITION A, which was measured one week after dosing (see the following table). The normalized values for the relative fluorescence intensity of the immunopositive β-amyloid plaques were obtained by CLSM after immunostaining and measurement of the average signal intensities of 3 animals per dose group. Plates without ANTIBODY A derived from ANTIBODY COMPOSITION A were observed only in the lower dose groups, most likely due to the limited or partial occupation of ANTIBODY A derived from ANTIBODY COMPOSITION A on the surface of the plate, which It may have been lost during the sectioning process. Therefore, only immunopositive plates are included for the comparative analysis. The mean relative fluorescence intensity per dose group after i.v. (BOLUS) of the COMPOSITION OF ANTIBODY A in PS2APP transgenic mice is indicated in the following table: • "• The experimental values represent the intensity values normalized to the value obtained from a single dose of 0.25 mg / kg after 1 week.
Figure 19 shows the binding of ANTIBODY COMPOSITION A in relation to the number of successive biweekly doses of 0.1 mg / kg. After two applications, the average intensity was increased, although the extent of immunostaining varies considerably and, therefore, did not have significance. After 4 injections, the β-amyloid plaques were immunostained in a more homogeneous manner, but the average intensity only increased slightly. In general, the data for the biweekly application clearly indicate a trend of greater bonding of the plates that correlates with the number of applications. Figure 20 shows the binding of ANTIBODY COMPOSITION A in relation to the number of successive monthly doses of 0.15 mg / kg. Interestingly, comparable levels are obtained after 2 and 3 applications. This was not necessarily expected and may indicate the initiation of premature effects that contribute to the time-dependent differences in the clearance mechanism, such as the delayed activation of microglia cells. The binding efficacy of the ANTIBODY COMPOSITION A in relation to the administered dose is indicated in Figures 21 and 22. The biweekly doses of 0.05, 0.1 and 0.3 mg / kg (Figure 21) and the monthly doses of 0.075, 0.15 and 0.45 mg / kg (Figure 22) clearly demonstrated a relationship with the dose. It is also evident that the response is not linear and that additional factors such as an activation Temporarily delayed microglia cells may contribute to the observed non-linearity. Therefore, it can be concluded that the COMPOSITION OF ANTIBODY A that binds to mouse Aβ plaques is related to the dose with indications that multiple doses are cumulative. Example 17: Analysis of antigen-dependent cellular phagocytosis To determine the phagocytic effect mediated by ANTIBODY COMPOSITION A, authentic Aβ plaques of brain slices with AD were pre-incubated with different concentrations of the ANTIBODY COMPOSITION A comprising monoglycosylated ANTIBODY A and Double glycosylated ANTIBODY and which is free of non-glycosylated ANTIBODY A, as defined above, and exposed to live human primary monocytes. Sections of unfixed AD human brain tissue from the region of the occipital cortex were prepared from a case of severe AD (Braak stage IV). Before adding the live cells, the sections were rehydrated with PBS for 5 minutes. The antibodies of the ANTIBODY COMPOSITION A were applied by incubation in defined concentrations in PBS for 1 hour. After washing with PBS, live cells were added. Primary human monocytes pretimulated at 0.8 and 1.5 x 106 / ml in RPMI 1640 medium (Gibco # 61870-044) were used with a 1% antibiotic solution of a concentrated solution containing . 000 U / ml penicillin and 10,000 mg / ml streptomycin (Gibco # 15140-122) and incubated at 37 ° C with 5% carbon dioxide for 2 to 4 days. Methods for the preparation of pre-stimulated human primary monocytes are known in the art, for example, through the use of stimulatory factors, such as macrophage colony stimulating factor (M-CSF). After incubation, the culture medium was gently removed and the sections were maintained by chemical fixation with 2% formaldehyde in PBS for 10 minutes. Staining of the charge of residual Aβ plates was carried out by incubation with BAP-2, a mouse monoclonal antibody against Aβ conjugated with fluorophore Alexa488 (Molecular Probes: A-20181, monoclonal antibody labeling kit), in a concentration of 10 mg / ml for 1 hour at room temperature. The quantification of plaque removal was determined by measuring the immunofluorescence of the residual stained Aß plaques. The images were recorded in a Leica TCS SP2 AOBS confocal laser scanning microscope. An optical layer was recorded at an excitation wavelength of 488 nm with an orifice configuration of 4 Airy using an HCX PL FL 20x / 0.40 correction lens, except in an experiment where a HCPL Fluorar objective was used instead 10x / 0.30 in an orifice configuration of 3. The instrument configurations were kept constant for all the images to allow a comparison quantitative relational. Specifically, the laser power, acquisition and separation (offset) were adjusted to allow monitoring of the signal strength within the dynamic range. For each concentration of ANTIBODY COMPOSITION, the gray matter regions were recorded in comparable positions from consecutive sections in order to minimize fluctuations possibly arising from anatomical differences in plate loading. The potential competitive binding of ANTIBODY COMPOSITION A and detection antibody BAP-2 was measured in the absence of cells at all concentrations of ANTIBODY COMPOSITION A. An unrelated human IgGl antibody (Serotec, PHP010) was used as additional control. The analysis of the images was performed using the IMARIS software (Bitplane, Switzerland). The isosurface of positive pixels of BAP-2 representing BAP-2 objects attached to the plates was created by means of the intensity threshold. The surface area and total fluorescence intensity values were calculated using the "isosurface function" of the SurpassPro software module. Data were expressed as the average staining area and the total staining values obtained from 5 gray matter regions of a section of the brain. The baseline of the signal is composed of instrumental noise and it was found that the diffusion signal in the tissue was negligible and, therefore, was not subtracted from the total intensity signal.
The qualitative effect of ANTIBODY A COMPOSITION was visualized by decreasing Aβ plate staining, which indicates a greater phagocytosis of the Aβ plaques of human AD brain sections as shown in Figure 23. Immunohistochemistry revealed a clearly visible reduction of Aß plates that can be stained, after preincubation with 100 ng / ml ANTIBODY COMPOSITION A after 40 hours. The effect is very pronounced and in concentrations of ANTIBODY COMPOSITION A of 1 and 5 mg / ml. Aß plaques are substantially eliminated more and more by cellular phagocytosis and only a few Aß plaques remain at a concentration of 5 mg / ml. In Figure 24, a quantitative measurement based on the immunoreactivity signals expressed as area and intensity of the same experiment is shown. Alternatively, the phagocytic effect mediated by ANTIBODY COMPOSITION A was determined using fluorescent polystyrene beads conjugated with Aβ. Therefore, the fluorescent beads (3 mm, Fluoresbrite carboxy YG, Polysciences Inc.) were coupled to the Aβ. Briefly, the beads were washed 2x by suspension and centrifugation in coupling buffer (50 mM buffer MES, pH 5.2, 1% DMSO). The pellet (approximately 10 μl) was suspended in 200 μl of coupling buffer and activated by the addition of 20 μl of a 20% solution of EDC (ethyl diaminopropyl carbodiimide, Pierce) in coupling buffer. The immediate addition of 20 μg of Aβ (1-40) or Aβ (1-42) (in 0.1% of ammonium hydroxide, Bachem) initiated the coupling reaction. After an overnight incubation the beads were washed with 3x 0.5 ml of 10 mM Tris. HCl pH 8.0 and 3x 0.5 ml of storage buffer (10 mm Tris, HCl pH 8.0, 0.05% BSA, 0.05% NaN3). The 1% suspension was stored at 4 ° C until use. As a negative control, Fluoresbrite carboxy NYO beads (red fluorescence) were coupled with the amino acid Aβ all-D (1-40) (in 0.1% ammonium hydroxide, Bachem). Mouse monocytes / macrophages (cell line P388D1) grew in C24 clear tissue culture or C96 black microplate groups at approximately 50% confluency. The culture medium was IMEM with 5% FBS, glutamine and antibiotics. To block non-specific purification receptors, 10 ml of fucoidan were added (Fluka, 10 mg / ml in water) at a culture volume of 200 ml and incubated for 2 hours. The COMPOSITION OF ANTIBODY A in serial dilutions and preincubated for 30 minutes. The suspension of fluorescent Aβ beads (20 μl) was added and incubated for 3 hours to allow phagocytosis. The adherent cells were vigorously washed lx with ice-cold EDTA and 2x with PBS to remove adhering agglutinates from the cell surface. The residual beads were monitored by visual inspection on a Zeiss Axiovert 405 or for quantification using a microplate fluorimeter (Fluoroscan, Labsystems) with filter configurations of 444 nm (Exc) and 485 nm (Em).
The qualitative effect of ANTIBODY COMPOSITION A on phagocytosis of synthetic Aβ aggregates coupled to fluorescent fluoroperlas by P388D1 is shown in Figure 25. The quantitative determination of the dose response of ANTIBODY COMPOSITION A is indicated in the Figure 26. Two independent experiments revealed a range that ranged between 30 and 200 ng / ml for EC50 and between 10 and 60 ng / ml for MEC. The variability observed is probably caused by differences in incubation stoichiometry, that is, the ratio of the beads to the cells. The observed decrease in phagocytosis of beads above a concentration > 200 ng / ml is due to the interaction of monovalent antibodies with a limited antigen. Therefore, we can conclude that the COMPOSITION OF ANTIBODY A effectively induces phagocytosis of Aβ plaques of brain tissue sections with AD in a dose-dependent manner.

Claims (30)

    CLAIMS A purified antibody molecule, characterized in that at least one antigen-binding site of said antibody molecule comprises a glycosylated asparagin (Asn) in the variable region of the heavy chain (VH). The purified antibody molecule according to claim 1, characterized in that said asparagine
  1. (Asn) glycosylated in the variable region of the heavy chain (VH) is found in the CDR2 region. The purified antibody molecule according to claim 1 or 2, characterized in that asparagine is
  2. (Asn) glycosylated in the variable region of the heavy chain (VH) is at position 52 of SEQ ID NO: 2 or at position 52 of SEQ ID NO: 6. The antibody molecule purified according to any of claims 1 to 3, characterized in that said antibody molecule is capable of specifically recognizing the β-A4 / Aβ4 peptide. The purified antibody molecule according to claim 4, characterized in that said β-A4 / Aβ4 peptide has the following sequence DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO:
  3. 3) or a part of at least 15 amino acids of said sequence. The antibody molecule according to any of claims 1 to 5, characterized in that at least one antigen-binding site of said antibody molecule comprises glycosylated asparagin (Asn) in the variable region of the heavy chain (VH) and said VH is coded by: (A) a nucleic acid molecule comprising the nucleotide sequence shown in SEQ ID N0: 1 CAGGTGGAATTGGTGGAAAGCGGCGGCGGCCTGGTGCAACCGGGCGGCAGCCTGCG TCTGAGCTGCGCGGCCTCCGGATTTACCTTTAGCAGCTATGCGATGAGCTGGGTGC GCCAAGCCCCTGGGAAGGGTCTCGAGTGGGTGAGCGCTATTAATGCTTCTGGTACT CGTACTTATTATGCTGATTCTGTTAAGGGTCGTTTTACCATTTCACGTGATAATTC GAAAAACACCCTGTATCTGCAAATGAACAGCCTGCGTGCGGAAGATACGGCCGTGT ATTATTGCGCGCGTGGTAAGGGTAATACTCATAAGCCTTATGGTTATGTTCGTTAT TTTGATGTTTGGGGCCAAGGCACCCTGGTGACGGTTAGCTCA (b) a nucleic acid molecule encoding a polypeptide having the amino acid sequence is shown in SEQ ID NO: 2 QVELVESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSAINASGT RTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGKGNTHKPYGYVRY FDVWGQGTLVTVSS (SEQ ID NO: 2;); (c) a nucleic acid molecule that hybridizes to the nucleic acid molecule of (a) or (b) and that encodes a polypeptide that is capable of binding to the β-A4 / Aβ4 peptide shown in the following sequence of amino acids DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO: 3) or is capable of binding to a fragment thereof comprising at least 15 amino acids; (d) a nucleic acid molecule that hybridizes to the nucleic acid molecule of (a) or (b) and that encodes a polypeptide that is capable of binding to at least two regions in the β-A4 / Aβ4 peptide such as is shown in the following amino acid sequence DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO: 3) or to at least two regions of a fragment of SEQ ID NO: 3 comprising at least 15 amino acids, where said two regions in the peptide β-A4 Aβ4 or the fragment thereof comprises the amino acids in the 3 to 6 position and in the 18 to 26 position; or (e) a nucleic acid sequence that degenerates to a nucleic acid sequence as defined in any of (a) to (d). The antibody molecule according to any of claims 1 to 5, characterized in that the variable region comprising a glycosylated asparagine (Asn) is comprised in the heavy chain selected from the group comprising: (a) a coded heavy chain polypeptide for a nucleic acid molecule as shown in SEQ ID NOS: 5, 23 or 25; (b) a heavy chain polypeptide having an amino acid sequence as shown in SEQ ID NO: 6 or 26; (c) a heavy chain polypeptide encoded by a nucleic acid molecule that hybridizes to the nucleic acid molecule defined in (a) and that encodes a polypeptide that is capable of binding to the β-A4 / Aβ4 peptide as shown in the following amino acid sequence DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO: 3) or a fragment thereof comprising at least 15 amino acids; or (d) a heavy chain polypeptide encoded by a nucleic acid molecule that hybridizes to the nucleic acid molecule defined in (a) and that encodes a polypeptide that is capable of binding to at least two regions in the β-peptide. A4 / Aβ4 as shown in the following amino acid sequence DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO: 3) or at least two regions of a fragment of SEQ ID NO: 3 comprising at least 15 amino acids where said two regions in the peptide β-A4 Aβ4 or its fragment comprises the amino acids in the 3 to 6 position and in the 18 to 26 position. 8. The antibody molecule according to any of claims 1 to 7, characterized in that two antigen-binding sites comprise glycosylated Asn in the variable region of the heavy chain (VH). 9. The antibody molecule according to claim 8, characterized in that the CDR2 regions of the heavy chains (VH) comprise the glycosylated Asn. 10. The antibody molecule according to any of claims 1 to 9, characterized in that said Asn is an Asn at position 52 of SEQ ID NO: 2 or is Asn at position 52 of SEQ ID NO: 6. 11. The antibody molecule according to any of claims 1 to 10, characterized in that said glycosylation at the Asn in the VH region is selected from the group consisting of (a) a sugar structure of the biantennary complex type; (b) a sugar structure of the biantennarous hybrid type; (c) a sugar structure of the oligomeric biantennary type; and (d) a biantennary structure of any of the structures provided in Figure 5 or Figure 27. 12. The antibody molecule according to claim 11, characterized in that said sugar structure does not comprise fucosylation of the nucleus. 13. The antibody molecule according to any of claims 2 to 12, characterized in that said CDR2 region has an amino acid sequence that is shown in SEQ ID NO: 12. 14. The antibody molecule according to any of claims 1 to 13, which is produced recombinantly. 15. The antibody molecule according to any of claims 1 to 14, which is produced in a CHO cell. 16. The antibody molecule according to claim 15, characterized in that said CHO cell is CHO Kl or CHO Kl SV. 17. A method for the preparation of an antibody molecule according to any of claims 1 to 16 comprising the following steps: (a) recombinant expression of a heterologous nucleic acid molecule encoding an antibody molecule as defined in any of claims 1 to 13 in a mammalian cultured cell; (b) purification of said recombinantly expressed antibody molecule through a method comprising the following steps: (bl) column purification of protein A; (b2) column purification by ion exchange; Y (b3) column purification by size exclusion. 18. The method according to claim 17, characterized in that said column purification by ion exchange comprises a cation exchange chromatography. 19. The method according to claim 17 or 18, which also comprises as an additional step (c) an analytical chromatography and / or another concentration step. 20. A composition comprising an antibody molecule characterized in that an antigen binding site of said antibody molecule comprises an asparagine (Asn) glycosylated in the variable region of the heavy chain (VH), and an antibody molecule characterized in that two antigen-binding sites of said antibody molecule comprise a glycosylated asparagin (Asn) in the variable region of the heavy chain (VH) and comprising less than 5% of an antibody molecule characterized in that no antigen-binding site of said antibody molecule comprises a glycosylated asparagin (Asn) in the variable region of the heavy chain (VH). 21. A composition comprising an antibody molecule according to any of claims 1 to 16 or an antibody molecule prepared by the method according to any of claims 17 to 19. 22. The composition according to claim 20 or 21, which is a diagnostic or pharmaceutical composition. 23. Use of an antibody molecule according to any of claims 1 to 16 or the composition according to any of claims 20 to 22 for the preparation of a medicament for the prevention and / or treatment of a disease associated with amyloidogenesis and / or amyloid plaque formation. 24. Use of an antibody molecule according to any of claims 1 to 16 or a composition according to any of claims 20 to 22 for the preparation of a diagnostic kit for the detection of an amyloidogenesis-associated disease and / or formation of amyloid plaques. 25. Use of an antibody molecule according to any of claims 1 to 16 or a composition according to any of claims 20 to 22 for the preparation of a medicament for the disintegration of β-amyloid plaques. 26. Use of an antibody molecule according to any of claims 1 to 16 or a composition according to any of claims 20 to 22 for the preparation of a pharmaceutical composition for passive immunization against the formation of β-amyloid plaques. 27. Use of an antibody molecule according to any of claims 1 to 16 or a composition according to any of claims 20 to 22 for the preparation of a pharmaceutical composition for the preventive treatment against a disease associated with amyloidogenesis and / or formation of amyloid plaques. 28. The use according to claim 27, characterized in that the pre-existing plaques or the β-amyloid aggregation intermediates are reduced. 29. Use of an antibody molecule according to any of claims 1 to 16 or a composition according to any of claims 20 to 22 for the preparation of a diagnostic kit for the diagnosis of an amyloidogenesis-associated disease and / or formation of amyloid plaques in a patient or for the diagnosis of a patient's susceptibility to the development of a disease associated with amyloidogenesis and / or formation of amyloid plaques. 30. Use according to any of claims 23, 24 or 27 to 29, characterized in that said disease is dementia, Alzheimer's disease, motor neuropathy, Down syndrome, Creutzfeld-Jacob disease, hereditary cerebral hemorrhage with amyloidosis of type Dutch, dementia associated with the formation of Lewy bodies, Parkinson's disease, HIV-related dementia, ALS or neuronal disorders related to age. A kit comprising an antibody molecule according to any one of claims 1 to 16 or prepared by the method according to any of claims 17 to 19 or a composition as defined in claims 20 to 22.
MXMX/A/2008/006948A 2006-12-12 2008-05-29 Antibodies against amyloid beta 4 with glycosylated in the variable region MX2008006948A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP05027090.9 2005-12-12

Publications (1)

Publication Number Publication Date
MX2008006948A true MX2008006948A (en) 2008-09-02

Family

ID=

Similar Documents

Publication Publication Date Title
US9272031B2 (en) Antibodies against amyloid beta 4 with glycosylation in the variable region
KR101068289B1 (en) Antibodies directed against amyloid-beta peptides and methods of use thereof
US7956173B2 (en) Nucleic acids coding for humanized antibodies for binding sphingosine-1-phosphate
US8444970B2 (en) Compositions and methods for treating ocular diseases and conditions
JP2025094219A (en) Anti-tdp-43 binding molecules and uses thereof
HK1201046A1 (en) Methods of treating ophthalmic diseases
MX2008006948A (en) Antibodies against amyloid beta 4 with glycosylated in the variable region
US20140186339A1 (en) Compositions and methods for treating ocular diseases and conditions
HK1176075B (en) Antibody glycosylation in the variable region
HK1130066B (en) Antibody glycosylation in the variable region
WO2024184494A1 (en) Anti-tdp-43 binding molecules and uses thereof
HK1142613A (en) Methods of treating ophthalmic diseases
HK1142613B (en) Methods of treating ophthalmic diseases