Methods of treating tauopathy

文档序号:1258843 发布日期:2020-08-25 浏览:13次 中文

阅读说明:本技术 治疗tau病变的方法 (Methods of treating tauopathy ) 是由 I·格里斯沃尔德-普伦那 G·帕里 于 2014-11-25 设计创作,主要内容包括:本发明涉及治疗tau病变的方法。本发明提供用于在个体中通过对所述个体施用抗Tau抗体治疗tau病变(例如急性tau病变)的方法。还提供了在个体中通过对所述个体施用抗Tau抗体治疗外伤性脑损伤的方法和治疗中风的方法。(The present invention relates to methods of treating tauopathy. The invention provides methods for treating tauopathy (e.g., acute tauopathy) in an individual by administering to the individual an anti-Tau antibody. Also provided are methods of treating traumatic brain injury and methods of treating stroke in an individual by administering to the individual an anti-Tau antibody.)

1. Use of an anti-Tau antibody in the manufacture of a medicament for treating a tauopathy in a human subject, wherein the anti-Tau antibody comprises an immunoglobulin light chain variable region and an immunoglobulin heavy chain variable region, wherein the heavy chain variable region comprises the amino acid sequence set forth in SEQ ID NO:37 and the light chain variable region comprises the amino acid sequence set forth in SEQ ID NO:41, and wherein any two doses of the anti-Tau antibody are administered to the human subject within 5 days, 4 weeks, or 2 months of each other.

2. The use of claim 1, wherein the medicament is in a subcutaneous, intrathecal, or intravenous dosage form.

3. The use of claim 1 or 2, wherein the tauopathy is alzheimer's disease, amyotrophic lateral sclerosis/parkinson-dementia syndrome, dementia with silvery particles, amyloid angiopathy of the british type, cerebral amyloid angiopathy, corticobasal degeneration, creutzfeldt-jakob disease, dementia pugilistica, calcified diffuse neurofibrillary tangles, down's syndrome, frontotemporal dementia with parkinson's disease linked to chromosome 17, frontotemporal lobar degeneration, Gerstmann-Straussler-Scheinker disease, hallowden-schutz disease, inclusion body myositis, multiple system atrophy, myotonic dystrophy, niemann-pick disease type C, non-guam motor neuron disease with neurofibrillary tangles, pick's disease, postencephalitic parkinson's disease, prion protein amyloid angiopathy, prion protein, parkinson's disease, alzheimer's disease, creutzfeldt-jakob disease, parkinson's disease, and alzheimer's disease, Progressive subcortical hyperplasia, progressive supranuclear palsy, subacute sclerosing panencephalitis, dementia with tangles only, or multi-infarct dementia.

4. The use of claim 3, wherein the tauopathy is Alzheimer's disease.

5. The use of claim 3, wherein the tauopathy is progressive supranuclear palsy.

6. The use of claim 3, wherein the tauopathy is a stroke, a chronic traumatic encephalopathy, a traumatic brain injury, a concussion, a seizure, an epilepsy, or an acute plumbeous encephalopathy.

7. Use of an anti-Tau antibody in the manufacture of a medicament for treating a tauopathy in a human subject, wherein the anti-Tau antibody comprises an immunoglobulin light chain variable region and an immunoglobulin heavy chain variable region, wherein (1) the heavy chain variable region comprises the amino acid sequence shown in SEQ ID No. 36 and the light chain variable region comprises the amino acid sequence shown in SEQ ID No. 40, (2) the heavy chain variable region comprises the amino acid sequence shown in SEQ ID No. 38 and the light chain variable region comprises the amino acid sequence shown in SEQ ID No. 42, (3) the heavy chain variable region comprises the amino acid sequence shown in SEQ ID No. 39 and the light chain variable region comprises the amino acid sequence shown in SEQ ID No. 43, (4) the heavy chain variable region comprises the amino acid sequence shown in SEQ ID No. 14 and the light chain variable region comprises the amino acid sequence shown in SEQ ID No. 13, or (5) the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:16 and the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:15,

and wherein (i) the anti-Tau antibody is administered to the human subject in a single bolus injection or (ii) any two doses of the anti-Tau antibody are administered within 5 days, 4 weeks, or 2 months of each other.

Background

Microtubule-associated protein Tau is abundant in the central nervous system and is produced mainly by neurons. The main function of Tau is to stabilize microtubules. There are 6 Tau isoforms in adult brain; tau isoforms are the products of alternative splicing of a single gene.

tauopathies (tauopathies) are a class of neurodegenerative diseases that result from the pathological aggregation of Tau protein in the so-called neurofibrillary tangles (NFTs) in the brain. Some examples of tauopathies include frontotemporal dementia (FTD), Alzheimer's disease, progressive supranuclear palsy, corticobasal degeneration, and frontotemporal lobar degeneration.

There is a need in the art for methods for treating tauopathy.

Summary of The Invention

The present invention provides methods for treating tauopathies (e.g., acute tauopathies) in an individual.

Thus, in one aspect, there is provided a method of treating acute tauopathy in an individual, the method comprising administering to the individual an anti-Tau antibody in an amount effective to significantly reduce the level of free Tau in an extracellular fluid of the individual.

In one embodiment, the anti-Tau antibody is effective to significantly reduce the level of free Tau in an extracellular fluid within 72 hours of administration of the anti-Tau antibody. In another embodiment, the anti-Tau antibody is effective to significantly reduce the level of free Tau in an extracellular fluid within 48 hours of administration of the anti-Tau antibody. In another embodiment, the anti-Tau antibody is effective to significantly reduce the level of free Tau in an extracellular fluid within 36 hours of administration of the anti-Tau antibody. In another embodiment, the anti-Tau antibody is effective to significantly reduce the level of free Tau in an extracellular fluid within 24 hours of administration of the anti-Tau antibody. In another embodiment, the anti-Tau antibody is effective to significantly reduce the level of free Tau in an extracellular fluid within 12 hours of administration of the anti-Tau antibody. In another embodiment, the anti-Tau antibody is effective to significantly reduce the level of free Tau in an extracellular fluid within 8 hours of administration of the anti-Tau antibody. In another embodiment, the anti-Tau antibody is effective to significantly reduce the level of free Tau in an extracellular fluid within 7 hours of administration of the anti-Tau antibody. In another embodiment, the anti-Tau antibody is effective to significantly reduce the level of free Tau in an extracellular fluid within 4 hours of administration of the anti-Tau antibody. In another embodiment, the anti-Tau antibody is effective to significantly reduce the level of free Tau in an extracellular fluid within 2 hours of administration of the anti-Tau antibody. In another embodiment, the anti-Tau antibody is effective to significantly reduce the level of free Tau in an extracellular fluid within 1 hour of administration of the anti-Tau antibody. In another embodiment, the anti-Tau antibody is effective to significantly reduce the level of free Tau in an extracellular fluid within 30 minutes of administration of the anti-Tau antibody.

In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 10%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 15%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 20%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 25%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 30%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 35%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 40%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 45%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 50%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 55%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 60%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 65%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 70%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 75%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 80%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 85%. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid by at least about 90%.

In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid to an undetectable level. In another embodiment, the anti-Tau antibody is effective to reduce the level of free Tau in an extracellular fluid to a normal level. In another embodiment, the reduced level of free Tau is maintained for a period of at least 2 hours after administration of the anti-Tau antibody. In another embodiment, the reduced level of free Tau is maintained for a period of at least 5 hours after administration of the anti-Tau antibody. In another embodiment, the reduced level of free Tau is maintained for a period of at least 10 hours after administration of the anti-Tau antibody. In another embodiment, the reduced level of free Tau is maintained for a period of at least 24 hours following administration of the anti-Tau antibody. In another embodiment, the reduced level of free Tau is maintained for a period of at least 7 days following administration of the anti-Tau antibody. In some cases, the reduced level of free Tau is maintained for a period of at least 2 weeks following administration of the anti-Tau antibody.

In one embodiment, the extracellular fluid is plasma. In another embodiment, the extracellular fluid is cerebrospinal fluid. In another embodiment, the extracellular fluid is interstitial fluid (interstitial fluid). In another embodiment, the extracellular fluid is blood.

The anti-Tau antibody may be administered by any suitable means. For example, the anti-Tau antibody may be administered by subcutaneous administration, by intrathecal administration, or by intravenous administration.

In one embodiment, the anti-Tau antibody is administered in an amount of about 0.1mg/kg body weight to about 50mg/kg body weight. In another embodiment, the anti-Tau antibody is administered as a single bolus injection (bolus injection).

In another embodiment, multiple doses (e.g., 2, 3,4, 5,6, 7, 8, or 9 doses) of the anti-Tau antibody are administered. In one embodiment, when multiple doses of anti-Tau antibody are administered, any two doses of anti-Tau antibody are administered within 3 days or more of each other. In another embodiment, when multiple doses of anti-Tau antibody are administered, any two doses of anti-Tau antibody are administered within 5 days or more of each other. In another embodiment, when multiple doses of anti-Tau antibody are administered, any two doses of anti-Tau antibody are administered within 7 days or more of each other. In another embodiment, when multiple doses of anti-Tau antibody are administered, any two doses of anti-Tau antibody are administered within 2 weeks or more of each other. In another embodiment, when multiple doses of anti-Tau antibody are administered, any two doses of anti-Tau antibody are administered within 4 weeks or more of each other. In another embodiment, when multiple doses of anti-Tau antibody are administered, any two doses of anti-Tau antibody are administered within 2 months or more of each other.

The invention also provides a method of treating acute tauopathy in an individual, the method comprising administering to the individual an anti-Tau antibody in an amount effective to provide a minimum anti-Tau antibody concentration in the cerebrospinal fluid (CSF) of the individual. In one embodiment, the minimum anti-Tau antibody concentration in the CSF is reached within 1 hour of administration of the anti-Tau antibody. In another embodiment, the minimal anti-Tau antibody concentration in the CSF is at least 20 ng/ml. In another embodiment, the minimum anti-Tau antibody concentration in the CSF is at least 30 ng/ml. In another embodiment, the minimal anti-Tau antibody concentration in the CSF provides an anti-Tau antibody to Tau molar ratio in the CSF of at least 2: 1. In another embodiment, the minimal anti-Tau antibody concentration in the CSF provides an anti-Tau antibody to Tau molar ratio in the CSF of at least 2.5: 1.

In any of the embodiments described above or herein, the acute tauopathy can be traumatic brain injury (e.g., diffuse axonal injury (diffuse axonanal injury), concussion (concussion), contusion (contusion), Coup-controlled injury, secondary Impact Syndrome (Second Impact Syndrome), punch-through (perforation injury), infant wobble Syndrome (ShakenBaby Syndrome), and Locked In Syndrome In any of the embodiments described above or herein, the acute tauopathy can be stroke (stroke) In any of the embodiments described above or herein, the acute tauopathy can be chronic traumatic brain disease (chronic traumatic encephalopathy).

The invention also provides a method of treating traumatic brain injury in an individual, comprising administering to the individual an anti-Tau antibody in an amount effective to significantly reduce the level of free Tau in the extracellular fluid of the individual. In some cases, the antibody is administered within 48 hours of traumatic brain injury. In some cases, the antibody is administered in a single dose. In some cases, the antibody is administered in multiple doses. In some cases, the antibody is administered weekly, every 2 weeks, every 4 weeks, every 6 weeks, every 8 weeks, every 3 months, or every 6 months.

The invention also provides a method of treating stroke in an individual, comprising administering to the individual an anti-Tau antibody in an amount effective to significantly reduce the level of free Tau in the extracellular fluid of the individual. In some cases, the antibody is administered within 48 hours of the stroke. In some cases, the antibody is administered in a single dose. In some cases, the antibody is administered in multiple doses. In some cases, the antibody is administered weekly, every 2 weeks, every 4 weeks, every 6 weeks, every 8 weeks, every 3 months, or every 6 months.

The invention also provides a method of treating an ongoing tauopathy in an individual, the method comprising administering to the individual an anti-Tau antibody in an amount effective to significantly reduce the level of free Tau in an extracellular fluid of the individual for a time sufficient to reduce the level of a β in the extracellular fluid. In one embodiment, the antibody is administered in a single dose. In another embodiment, the antibody is administered in multiple doses. In another embodiment, the antibody is administered weekly, every 2 weeks, every 4 weeks, every 6 weeks, every 8 weeks, every 3 months, or every 6 months.

In any of the embodiments described above or herein, the level of a β is significantly reduced over a period of time from about 5 days to about 15 days after administration of the anti-Tau antibody.

Any suitable anti-Tau antibody can be used in the methods described herein. An exemplary anti-Tau antibody is hu-IPN002 (also known as IPN007 and IPN002 variant 2) comprising a heavy chain and a light chain having the sequences shown in SEQ ID NOS: 37 and 41, respectively; or antigen binding fragments and variants thereof. hu-IPN002 is a humanized immunoglobulin (IgG4) monoclonal antibody that binds extracellular Tau.

In one embodiment, the antibody comprises the heavy and light chain CDRs or variable regions of hu-IPN 002. Thus, in one embodiment, the antibody comprises the CDR1, CDR2, and CDR3 domains of the hu-IPN002 VH region having the sequence shown in SEQ ID NO. 37 and the CDR1, CDR2, and CDR3 domains of the hu-IPN002 VL having the sequence shown in SEQ ID NO. 41. In another embodiment, the antibody comprises heavy chain CDR1, CDR2, and CDR3 domains having the sequences shown in SEQ ID NOs 10, 11, and 12, respectively, and light chain CDR1, CDR2, and CDR3 domains having the sequences shown in SEQ ID NOs 7, 8, and 9, respectively. In another embodiment, the antibody comprises VH and/or VL regions having the amino acid sequences shown in SEQ ID NO 37 and/or SEQ ID NO 41, respectively. In another embodiment, the antibody comprises a heavy chain variable region (VH) and/or a light chain variable region (VL) encoded by the nucleic acid sequences shown in SEQ ID NO:29 and/or SEQ ID NO:33, respectively. In another embodiment, the antibody competes for binding to, and/or binds to, the same epitope on Tau as the above-described antibody. In another embodiment, the antibody has at least about 90% variable region amino acid sequence identity to an antibody described above (e.g., at least about 90%, 95%, or 99% variable region identity to SEQ ID NO:37 or SEQ ID NO: 41).

In another embodiment, the administered anti-Tau antibody specifically binds to an epitope within amino acids 1-158 of Tau of 2N 4R. In another embodiment, the administered anti-Tau antibody is capable of specifically binding to an epitope within amino acids 2-18 of Tau. In another embodiment, the administered anti-Tau antibody is capable of specifically binding to an epitope within amino acids 7-13 or within amino acids 25-30 of Tau. In another embodiment, the administered anti-Tau antibody is capable of specifically binding to an epitope within amino acids 15-24 of Tau. In another embodiment, the administered anti-Tau antibody is capable of specifically binding to an epitope within amino acids 28-126 of Tau of 2N 4R. In another embodiment, the anti-Tau antibody administered is capable of specifically binding to an epitope within amino acids 150-158 of 2N4R Tau. In another embodiment, the administered anti-Tau antibody binds a linear epitope. In another embodiment, the administered anti-Tau antibody binds to an epitope within a Tau polypeptide having at least 95% amino acid sequence identity to an eTau4 amino acid sequence set forth in figure 9.

In another embodiment, the administered anti-Tau antibody may be an anti-Tau antibody that competes for binding with another antibody comprising: a) a light chain region comprising: (i) VL CDR1 comprising the amino acid sequence of SEQ ID NO. 1 or SEQ ID NO. 7; (ii) VL CDR2 comprising the amino acid sequence of SEQ ID NO. 2 or SEQ ID NO. 8; and (iii) a VLCDR3 comprising the amino acid sequence of SEQ ID NO. 3 or SEQ ID NO. 9; and b) a heavy chain region comprising: (i) VH CDR1 comprising the amino acid sequence of SEQ ID NO.4 or SEQ ID NO. 10; (ii) VH CDR2 comprising the amino acid sequence of SEQ ID NO 5 or SEQ ID NO 11; and (iii) a VH CDR3 comprising the amino acid sequence of SEQ ID NO 6 or SEQ ID NO 12.

In another embodiment, the anti-Tau antibody is an anti-Tau antibody comprising: a) a light chain region comprising: (i) VL CDR1 comprising the amino acid sequence of SEQ ID NO. 1 or SEQ ID NO. 7; (ii) VL CDR2 comprising the amino acid sequence of SEQ ID NO. 2 or SEQ ID NO. 8; and (iii) a VL CDR3 comprising the amino acid sequence of SEQ ID NO. 3 or SEQ ID NO. 9; and b) a heavy chain region comprising: (i) VH CDR1 comprising the amino acid sequence of SEQ ID NO.4 or SEQ ID NO. 10; (ii) VH CDR2 comprising the amino acid sequence of SEQ ID NO 5 or SEQ ID NO 11; and (iii) a VH CDR3 comprising the amino acid sequence of SEQ ID NO 6 or SEQ ID NO 12.

In another embodiment, the anti-Tau antibody is an anti-Tau antibody that specifically binds to an epitope independent of phosphorylation of amino acids within the epitope. In another embodiment, the anti-Tau antibody is a humanized anti-Tau antibody.

Brief Description of Drawings

Figure 1 depicts IPN002 levels in plasma and cerebrospinal fluid (CSF) after a single injection of IPN002 into cynomolgus monkeys.

Figure 2 depicts the effect of IPN002 on Tau levels in CSF of cynomolgus monkeys treated with IPN 002.

Figure 3 depicts the effect of IPN002 on the levels of Α β protein in CSF of cynomolgus monkeys treated with IPN 002.

FIG. 4 depicts hu-IPN002 levels in serum of non-human primates treated with 5mg/kg (left panel) or 20mg/kg (right panel) of hu-IPN 002.

FIG. 5 depicts hu-IPN002 levels in CSF of non-human primates treated with hu-IPN002 at 5mg/kg (left panel) or 20mg/kg (right panel).

Figure 6 provides an overview of the pharmacokinetic data depicted in figures 4 and 5.

FIG. 7 depicts the effect of administration of hu-IPN002 antibody on the level of free Tau in CSF of non-human primates treated with 5mg/kg or 20mg/kg hu-IPN002 antibody.

FIG. 8 depicts the effect of administration of hu-IPN002 antibody on the levels of Abeta 40 in CSF of non-human primates treated with 5mg/kg or 20mg/kg hu-IPN002 antibody.

FIG. 9 provides an alignment of the amino acid sequence of 2N4R Tau (SEQ ID NO:72) with that of eTau4(SEQ ID NO: 71).

Figure 10 depicts the presence of Tau fragments in CSF from individuals likely to have chronic traumatic encephalopathy.

FIGS. 11A and 11B provide the amino acid sequences of IPN001 VH (FIG. 11A) and VL (FIG. 11B). Complementarity Determining Regions (CDRs) are shown in bold and underlined.

Fig. 12A and 12B provide the amino acid sequences of IPN002 VH (fig. 12A) and VL (fig. 12B). Complementarity Determining Regions (CDRs) are shown in bold and underlined.

Fig. 13 depicts the amino acid sequence of IPN002 VH variant 1; and nucleotide sequences encoding the amino acid sequences.

Fig. 14 depicts the amino acid sequence of IPN002 VH variant 2; and nucleotide sequences encoding the amino acid sequences.

Fig. 15 depicts the amino acid sequence of IPN002 VH variant 3; and nucleotide sequences encoding the amino acid sequences.

Fig. 16 depicts the amino acid sequence of IPN002 VH variant 4; and nucleotide sequences encoding the amino acid sequences.

Fig. 17 depicts the amino acid sequence of IPN002 vk variant 1; and nucleotide sequences encoding the amino acid sequences.

Fig. 18 depicts the amino acid sequence of IPN002 vk variant 2; and nucleotide sequences encoding the amino acid sequences.

Fig. 19 depicts the amino acid sequence of IPN002 vk variant 3; and nucleotide sequences encoding the amino acid sequences.

Fig. 20 depicts the amino acid sequence of IPN002 vk variant 4; and nucleotide sequences encoding the amino acid sequences.

FIG. 21 provides the amino acid sequences of various extracellular Tau polypeptides (SEQ ID NOS 73-78 in order of occurrence).

FIG. 22 depicts hu-IPN002 levels in serum of non-human primates treated with 5mg/kg or 20mg/kg hu-IPN002 until 56 days post-treatment.

FIG. 23 depicts hu-IPN002 levels in CSF of non-human primates treated with 5mg/kg or 20mg/kg hu-IPN002 until 56 days post-treatment.

FIG. 24 depicts free Tau levels in CSF of non-human primates treated with 5mg/kg or 20mg/kg hu-IPN002 until 56 days post-treatment.

FIG. 25 depicts Abeta 40 levels in CSF of non-human primates treated with 5mg/kg or 20mg/kg hu-IPN002 until 56 days post-treatment.

FIG. 26 depicts hu-IPN002 levels in serum of non-human primates treated with hu-IPN002 at 0.5mg/kg, 2mg/kg, 5mg/kg, or 20mg/kg until 57 days post-treatment.

FIG. 27 depicts hu-IPN002 levels in CSF of non-human primates treated with hu-IPN002 at 0.5mg/kg, 2mg/kg, 5mg/kg, or 20mg/kg until 57 days post-treatment.

FIG. 28 depicts free Tau levels in CSF of non-human primates treated with hu-IPN002 at 0.5mg/kg, 2mg/kg, 5mg/kg, or 20mg/kg until 57 days post-treatment.

FIG. 29 panels A-B compare free Tau CSF levels in non-human primates treated with hu-IPN002 at 0.5mg/kg, 2mg/kg, 5mg/kg, or 20mg/kg until 57 days post-treatment.

FIG. 30 panels A-B compare A β 40 levels in CSF of nonhuman primates treated with hu-IPN002 at 0.5mg/kg, 2mg/kg, 5mg/kg, or 20mg/kg until 57 days post-treatment, which A β 40 levels were evaluated using an internal assay.

FIG. 31 panels A-B compare A β 40 levels in CSF of nonhuman primates treated with hu-IPN002 at 0.5mg/kg, 2mg/kg, 5mg/kg, or 20mg/kg until 57 days post-treatment, as assessed by Millipore assay.

FIG. 32 depicts day 1 levels of hu-IPN002 in serum of non-human primates treated with a hu-IPN002 multi-dose regimen.

FIG. 33 depicts day 29 levels of hu-IPN002 in serum of non-human primates treated with the hu-IPN002 multi-dose regimen.

FIG. 34 depicts the day 57 levels of hu-IPN002 in serum of non-human primates treated with the hu-IPN002 multi-dose regimen.

FIG. 35 depicts day 1 levels of hu-IPN002 in CSF of non-human primates treated with a hu-IPN002 multi-dose regimen.

FIG. 36 depicts day 29 levels of hu-IPN002 in CSF of non-human primates treated with a hu-IPN002 multi-dose regimen.

FIG. 37 depicts day 57 levels of hu-IPN002 in CSF of non-human primates treated with a hu-IPN002 multi-dose regimen.

FIG. 38 depicts free Tau levels in CSF of non-human primates treated with the hu-IPN002 multi-dose regimen until day 112.

FIG. 39 depicts free Tau levels in CSF of non-human primate control (group 1) treated with a hu-IPN002 multi-dose regimen of 20mg/kg on days 1, 29, and 57 (group 2) until day 169.

FIG. 40 depicts A β 40 levels in CSF of non-human primates treated with the hu-IPN002 multi-dose regimen until day 112.

FIG. 41 depicts the simulated serum and CSF concentrations of free hu-IPN002 and free eTau in humans after 10mpk IV infusion.

Figure 42 depicts predicted human plasma concentration-time profiles following a 700mg Q4W (dashed short and horizontal line) and 700mg vehicle +280mg Q4W (dashed dotted line) dosing regimen.

FIG. 43 depicts predicted human plasma eTau concentration-time profiles following a 700mg Q4W (dashed short and horizontal line) and 700mg vehicle +280mg Q4W (dashed dotted line) dosing regimen.

Definition of

The terms "antibody" and "immunoglobulin" include antibodies or immunoglobulins of any isotype, antibody fragments that retain specific binding to an antigen, including, but not limited to, Fab, Fv, scFv, and Fd fragments, chimeric antibodies, humanized antibodies, single chain antibodies, bispecific antibodies, and fusion proteins comprising an antigen-binding portion of an antibody and a non-antibody protein. The antibody may be detectably labeled, for example, with a radioisotope, an enzyme that generates a detectable product, a fluorescent protein, or the like. The antibody may further be conjugated to other moieties, such as members of a specific binding pair, e.g., biotin (a member of a biotin-avidin specific binding pair) and the like. The antibodies may also be bound to a solid support including, but not limited to, polystyrene plates or beads, and the like. The term also encompasses Fab ', Fv, F (ab')2And or other antibody fragments that retain specific binding to the antigen, and monoclonal antibodies. The antibody may be monovalent or bivalent.

As used herein, the term "humanized immunoglobulin" refers to an immunoglobulin comprising immunoglobulin portions of different origin, wherein at least one portion comprises an amino acid sequence of human origin. For example, a humanized antibody can comprise portions derived from an immunoglobulin of non-human origin (such as a mouse) and portions derived from an immunoglobulin sequence of human origin (e.g., a chimeric immunoglobulin) that are chemically linked together by conventional techniques (e.g., synthesis) or prepared as a continuous polypeptide using genetic engineering techniques (e.g., DNA encoding portions of the proteins of the chimeric antibody can be expressed to produce a continuous polypeptide chain). Another example of a humanized immunoglobulin is an immunoglobulin comprising one or more immunoglobulin chains comprising CDRs derived from an antibody of non-human origin and framework regions derived from a light and/or heavy chain of human origin (e.g., a CDR-grafted antibody with or without framework changes). The term humanized immunoglobulin also encompasses chimeric or CDR-grafted single chain antibodies. See, e.g., Cabilly et al, U.S. Pat. Nos. 4,816,567; cabilly et al, European patent No.0,125,023B1; boss et al, U.S. Pat. No.4,816,397; boss et al, European patent No.0,120,694B1; neuberger, m.s., et al, WO 86/01533; neuberger, m.s., et al, european patent No.0,194,276B1; winter, U.S. Pat. No.5,225,539; winter, european patent No.0,239,400B1; padlan, E.A. et al, European patent application No.0,519,596A1. For single chain antibodies, see also Ladner et al, U.S. Pat. No.4,946,778; huston, U.S. patent No.5,476,786; and Bird, R.E., et al, Science,242: 423-.

For example, synthetic and/or recombinant nucleic acids can be used to prepare a gene (e.g., cDNA) encoding a desired humanized chain to produce a humanized immunoglobulin. For example, PCR mutagenesis may be used to alter the DNA sequence encoding the human or humanized strand (such as a DNA template from a previously humanized variable region) to construct a Nucleic acid (e.g., DNA) sequence encoding the humanized variable region (see, e.g., Kamman, M. et al, Nucleic Acids Res.,17:5404 (1989); Sato, K. et al, cancer research,53: 851. 856 (1993); Daugherty, B.L. et al, Nucleic Acids Res.,19(9):2471-2476 (1991); and Lewis, A.P. and J.S.Crow, Gene,101:297-302 (1991)). Variants can also be readily generated using these or other suitable methods. For example, cloned variable regions can be mutagenized, and sequences encoding variants with the desired specificity can be selected (e.g., from phage libraries; see, e.g., Krebber et al, U.S. Pat. No.5,514,548; Hoogenboom et al, published 4/1/4 in WO 93/06213,1993).

An "antibody fragment" comprises a portion of an intact antibody, such as the antigen binding or variable region of an intact antibody. Examples of antibody fragments include Fab, Fab ', F (ab')2And Fv fragments; diabodies (diabodies); linear antibodies (Zapata et al, ProteinEng.8(10):1057-1062 (1995)); a single chain antibody molecule; and multispecific antibodies formed from antibody fragments. Digestion of antibodies by papain produces two identical antigen-binding fragments, each with a single antigen-binding site (called "Fab" fragments) and one residual "Fc" fragment (the name reflecting the ability to crystallize readily). Production of F (ab')2A fragment which has two antigen binding sites and is still capable of cross-linking antigens.

"Fv" is the smallest antibody fragment that contains the entire antigen recognition and binding site. This region consists of a dimer of one heavy and one light chain variable domain in tight, non-covalent association. In this configuration, the 3 CDRs of each variable domain interact to define an antigen binding site on the surface of the VH-VL dimer. Collectively, the 6 CDRs confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only 3 CDRs specific for an antigen) has the ability to recognize and bind antigen, although with lower affinity than the entire binding site.

The "Fab" fragment also contains the constant domain of the light chain and the first constant domain of the heavy chain (CH 1). Fab fragments differ from Fab' fragments by the addition of several residues at the carboxy terminus of the heavy chain CH1 domain, including one or more cysteines from the antibody hinge region. Fab '-SH is the name of Fab' herein, in which the cysteine residues of the constant domains carry a free thiol group. F (ab')2Antibody fragments were originally generated as pairs of Fab' fragments with a hinge cysteine between them. Otherization of antibody fragments is also knownAnd (4) chemical coupling.

The "light chain" of an antibody (immunoglobulin) from any vertebrate species can be assigned to one of two distinctly different types (termed kappa and lambda) based on the amino acid sequence of its constant domains. Immunoglobulins can be assigned to different classes depending on the amino acid sequence of the constant domains of their heavy chains. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these can be further divided into subclasses (isotypes), such as IgG1, IgG2, IgG3, IgG4, IgA1, and IgA 2.

"Single-chain Fv" or "sFv" antibody fragments comprise the VH and VL domains of an antibody, wherein these domains are present in a single polypeptide chain. In some embodiments, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains that enables the sFv to form the desired structure for antigen binding. For an overview of sFvs, see Pluckthun, in The Pharmacology of monoclonal antibodies, Vol.113, Rosenburg and Moore eds, Springer-Verlag, New York, pp.269-315 (1994).

The term "diabodies" refers to small antibody fragments having two antigen-binding sites, which fragments comprise a heavy chain variable domain (VH) and a light chain variable domain (VL) linked in the same polypeptide chain (VH-VL). By using linkers that are too short to allow pairing between two domains on the same chain, the domains are forced to pair with complementary domains of another chain and two antigen binding sites are created. Diabodies are described more fully in, for example, EP404,097; WO 93/11161; and Hollinger et al (1993) Proc. Natl. Acad. Sci. USA 90: 6444-.

As used herein, the term "affinity" refers to the equilibrium constant for reversible binding of two agents (e.g., an antibody and an antigen), and is expressed as the dissociation constant (Kd). The affinity can be at least 1-fold greater, at least 2-fold greater, at least 3-fold greater, at least 4-fold greater, at least 5-fold greater, at least 6-fold greater, at least 7-fold greater, at least 8-fold greater, at least 9-fold greater, at least 10-fold greater, at least 20-fold greater, at least 30-fold greater, at least 40-fold greater, at least 50-fold greater, at least 60-fold greater, at least 70-fold greater, at least 80-fold greater, at least 90-fold greater, at least 100-fold greater, or at least 1000-fold greater or more than the affinity of the antibody for an unrelated amino acid sequence. The affinity of an antibody for a target protein can be, for example, about 100 nanomolar (nM) to about 0.1nM, about 100nM to about 1 picomolar (pM), or about 100nM to about 1 femtomolar (fM) or more. As used herein, the term "avidity" refers to the resistance of a complex of two or more agents to dissociation upon dilution. The terms "immunoreactive" and "preferential binding" are used interchangeably herein with respect to antibodies and/or antigen binding fragments.

The term "association" refers to direct association between two molecules due to, for example, covalent, electrostatic, hydrophobic, and ionic and/or hydrogen bonding interactions, including interactions such as salt bridges and water bridges. An appropriate anti-Tau antibody specifically binds to an epitope within the Tau polypeptide. Non-specific binding may refer to binding at less than about 10-7Affinity binding of M, e.g. at 10-6M,10-5M,10- 4M, etc.

As used herein, the term "CDR" or "complementarity determining region" means a non-contiguous antigen binding site found within the variable regions of both heavy and light chain polypeptides. CDRs have been determined by Kabat et al, J.biol.chem.252:6609-6616 (1977); kabat et al, U.S. Dept. of Health and Human Services, "Sequences of proteins of immunological interest" (1991); chothia et al, J.mol.biol.196:901-917 (1987); and MacCallum et al, J.mol.biol.262:732-745(1996), wherein the definition includes a subset or overlap of amino acid residues when compared to each other. However, the use of either definition to refer to the CDRs of an antibody or grafted antibody or variant thereof is intended to be within the scope of the terms defined and used herein. The amino acid residues that encompass the CDRs defined by each of the above-cited references are listed in table 1 below for comparison.

Table 1: CDR definition

Kabat1 Chothia2 MacCallum3
VH CDR1 31-35 26-32 30-35
VH CDR2 50-65 53-55 47-58
VH CDR3 95-102 96-101 93-101
VL CDR1 24-34 26-32 30-36
VL CDR2 50-56 50-52 46-55
VL CDR3 89-97 91-96 89-96

1Residue numbering follows Kabat et al, supra nomenclature

2Residue numbering follows the nomenclature of Chothia et al, supra

3Residue numbering follows MacCallum et al, supra nomenclature

As used herein, the term "framework" when used in reference to an antibody variable region means all amino acid residues outside the CDR regions within the variable region of the antibody. Variable region frameworks are typically discontinuous amino acid sequences of about 100-120 amino acids in length, but are intended to refer only to those amino acids outside of the CDRs. As used herein, the term "framework region" is intended to refer to each domain of the framework separated by CDRs.

An "isolated" antibody refers to an antibody that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of their natural environment are materials that would interfere with diagnostic or therapeutic uses of the antibodies, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes. In some embodiments, the antibody will be purified (1) to greater than 90%, greater than 95%, or greater than 98% (by weight of the antibody), as determined by Lowry methods, e.g., greater than 99% (by weight), (2) to a degree sufficient to obtain at least 15 residues of the N-terminal or internal amino acid sequence by using a rotating cup sequencer (spinning cup sequencer), or (3) to homogeneity by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) using coomassie blue or silver stain under reducing or non-reducing conditions. Isolated antibodies include antibodies in situ within recombinant cells, as at least one component of the antibody's natural environment will not be present. In some cases, the isolated antibody is prepared by at least one purification step.

The term "epitope" or "antigenic determinant" means a site on an antigen to which an immunoglobulin or antibody specifically binds. Epitopes can be formed from contiguous amino acids or non-contiguous amino acids juxtaposed by tertiary folding of the protein. Epitopes formed from contiguous amino acids are typically retained upon exposure to denaturing solvents, while epitopes formed by tertiary folding are typically lost upon treatment with denaturing solvents. Epitopes typically comprise at least 3,4, 5,6, 7, 8,9, 10, 11, 12, 13, 14 or 15 amino acids in a unique spatial conformation. Methods for determining what epitope a given antibody binds (i.e., epitope mapping) are well known in the art and include, for example, immunoblot and immunoprecipitation assays, in which overlapping or contiguous peptides from Tau are tested for reactivity with a given anti-Tau antibody. Methods for determining spatial conformation of epitopes include techniques in the art and those described herein, such as X-ray crystallography and two-dimensional nuclear magnetic resonance (see, e.g., epipope Mapping Protocols in Methods in molecular Biology, volume 66, ed. g.e.morris (1996)).

Other techniques include, for example, epitope mapping methods, such as X-ray analysis of crystals of antigen-antibody complexes, which provide atomic resolution of the epitope. Other methods monitor binding of antibodies to antigen fragments or mutated antigens, where loss of binding due to modification of amino acid residues within the antigen sequence is often taken as an indication of the epitope component. Furthermore, epitope mapping can also be performed using computational combinatorial methods. These methods rely on the ability of the antibody of interest to affinity isolate specific short peptides from combinatorial phage display peptide libraries. The peptides are then used as a guide to define the epitope corresponding to the antibody used to screen the peptide library. For epitope mapping, computational algorithms have also been developed that have been shown to map discontinuous conformational epitopes.

The term "epitope mapping" means the process of identifying the molecular determinants of antibody-antigen recognition.

The term "binding to the same epitope" in reference to two or more antibodies means that the antibodies bind to the same, overlap, or encompass continuous or discontinuous segments of amino acids. The skilled artisan understands that the phrase "binds to the same epitope" does not necessarily mean that the antibody binds to exactly the same amino acid. The precise amino acids bound by an antibody may vary. For example, a first antibody may bind to an amino acid segment that is fully encompassed by the amino acid segment bound by a second antibody. In another example, the first antibody binds to one or more amino acid segments that significantly overlap with one or more segments bound by the second antibody. For purposes herein, such antibodies are considered to "bind to the same epitope".

Thus, the invention also encompasses antibodies that bind to an epitope on Tau that comprises all or part of the epitope recognized by a particular antibody described herein (e.g., the same or overlapping regions or regions between or spanning such regions).

The invention also encompasses antibodies that compete with the antibodies described herein for binding to Tau. Antibodies that compete for binding can be identified using conventional techniques. Such techniques include, for example, immunoassays that show the ability of one antibody to block the binding of another antibody to the target antigen, i.e., competitive binding assays. Competitive binding is determined in an assay in which the immunoglobulin tested inhibits specific binding of a reference antibody to a common antigen (such as Tau). Many types of competitive binding assays are known, for example: solid phase direct or indirect Radioimmunoassays (RIA), solid phase direct or indirect Enzyme Immunoassays (EIA), sandwich competition assays (see Stahli et al, Methods in enzymology 9:242 (1983)); solid phase direct biotin-avidin EIA (see Kirkland et al, J.Immunol.137:3614 (1986)); solid phase direct labeling assay, solid phase direct labeling sandwich assay (see Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Press (1988)); direct labeling of RIA using a solid phase of the I-125 label (see Morel et al, mol. Immunol.25(1):7 (1988)); solid phase direct Biotin-avidin EIA (Cheung et al, Virology 176:546 (1990)); and direct labeling of RIA (Moldenhauer et al, Scand. J. Immunol.32:77 (1990)). Typically, such assays involve the use of purified antigen bound to a solid surface or cells carrying either a test immunoglobulin without a label or a reference immunoglobulin with a label. Competitive inhibition is measured by determining the amount of label bound to a solid surface or cells in the presence of the test immunoglobulin. Typically, the test immunoglobulin is present in excess. Typically, when a competing antibody is present in excess, it will inhibit specific binding of the reference antibody to the common antigen by at least 50-55%, 55-60%, 60-65%, 65-70%, 70-75%, or more. The terms "polypeptide", "peptide" and "protein" are used interchangeably herein and refer to polymeric forms of amino acids of any length, which may include genetically encoded and non-genetically encoded amino acids, chemically or biochemically modified or derivatized amino acids, and polypeptides having a modified peptide backbone. The term includes fusion proteins, including but not limited to fusion proteins with heterologous amino acid sequences, fusions with heterologous and homologous leader sequences, with or without an N-terminal methionine residue; an immunologically labeled protein; and so on.

As used herein, the terms "treat" and "treating" refer to obtaining a desired pharmacological and/or physiological effect. The effect may be preventative in terms of completely or partially preventing a disease or symptoms thereof and/or therapeutic in terms of a partial or complete cure of a disease and/or adverse effects attributable to a disease. As used herein, "treatment" encompasses any treatment of a disease in a mammal, particularly a human, and includes: (a) preventing the occurrence of a disease in a subject, which may have a predisposition to the disease but has not yet been diagnosed as having the disease; (b) inhibiting the disease, i.e. blocking its formation; and (c) alleviating the disease, i.e., causing regression of the disease.

The terms "individual," "subject," "host," and "patient" are used interchangeably herein to refer to a mammal, including, but not limited to, a mouse (rat, mouse), a non-human primate, a human, a dog, a cat, an ungulate (e.g., horse, cow, sheep, pig, goat), and the like.

"therapeutically effective amount" or "effective amount" refers to an amount sufficient to effect such treatment of a disease when an anti-Tau antibody is administered to a mammal or other subject to treat the disease. The "therapeutically effective amount" will vary depending on the anti-Tau antibody, the disease and its severity, and the age, weight, etc., of the subject to be treated.

As used herein, the terms "fixed dose", "flat dose" and "flat-fixed dose" are used interchangeably and refer to a dose that is administered to a patient without regard to the weight of the patient or the Body Surface Area (BSA). Thus, a fixed or flat dose is not provided in a mg/kg dose, but in an absolute amount of the agent (e.g., an anti-Tau antibody).

As used herein, the term "Body Surface Area (BSA) based dose" means a dose (e.g., of anti-Tau antibody) that is adjusted to the Body Surface Area (BSA) of an individual patient. The BSA based dose may be provided in mg/kg body weight. A number of calculations have been published for obtaining BSA without direct measurement, among the most widely used are the Du Bois formula (see Du Boisd, Du Bois EF (6.1916) Archives of Internal Medicine 17(6): 863-71; and Verbracken, J. et al (4.2006) Metabolism-Clinical and Experimental 55(4): 515-24). Other exemplary BSA formulas include Mosteller formula (Mosteller RD. N Engl J Med., 1987; 317:1098), Haycock formula (Haycock GB et al, J Pediatr 1978,93:62-66), Gehan and George formulas (Gehan EA, George SL, Cancer Chemother Rep 1970,54:225-235), Boyd formula (Current, JD (1998) The Internet Journal of insulation 2(2), and Boyd, Edith (1935) University of Minnesota. The Institute of Child Welfa, Monograph Series, No. x. London: Oxford University Press, Fujimoto formula (Fujimoto S et al, Nippon Eiseneis et al, 1968, Zjira 5: Zjiri et al, Zjiru 443, Zjiru 5,umschau 2010; 57:178-183). "biological samples" encompass a variety of sample types obtained from an individual and can be used in diagnostic or monitoring assays. This definition encompasses blood and other liquid samples of biological origin, solid tissue samples such as biopsy specimens or tissue cultures or cells derived therefrom and the progeny thereof. The definition also includes samples that have been manipulated in any way after being obtained, such as treatment with reagents, solubilization, or enrichment for certain components (such as polynucleotides). The term "biological sample" encompasses clinical samples and also includes cells in culture, cell supernatants, cell lysates, serum, plasma, biological fluids, and tissue samples. The term "biological sample" includes urine, saliva, cerebrospinal fluid, blood fractions such asPlasma and serum, and the like.

As used herein, the term "acute tauopathy" means a disease, disorder or condition associated with a burst of abnormally elevated Tau (e.g., elevated compared to normal, control levels of Tau) in a subject's extracellular fluid (e.g., cerebrospinal fluid (CSF), interstitial fluid (ISF), blood, or blood fraction (e.g., a blood fraction such as serum or plasma)), e.g., an elevation of Tau in extracellular fluid following damage associated with a physical perturbation to a tissue of interest of the subject's brain and/or central nervous system. Such damage is typically followed by an increase in Tau in extracellular fluid (e.g., CSF, ISF, blood and/or blood fractions (e.g., plasma)) over a relatively short period of time, e.g., weeks or months (or shorter periods of time). Examples of such damage include, but are not necessarily limited to, physical trauma (e.g., head injury) and stroke. Non-limiting examples of acute tauopathies are stroke, chronic traumatic encephalopathy, traumatic brain injury, concussion, seizures (seizurs), epilepsy (e.g., Dravet syndrome (also known as infantile Severe Myoclonic Epilepsy (SMEI)), and acute plumbeous encephalopathy.

The phrase "traumatic brain injury" (also referred to as "TBI") is a form of acquired brain injury that occurs when trauma causes damage to the brain, such as damage to the brain caused by an external force. For example, TBI can occur when the head suddenly and violently impacts an object (e.g., during a fall, car accident, sporting event, or in any number of different ways) or when an object pierces the skull and enters the brain tissue. Both types of TBI can cause injury to brain tissue (bruised), intracerebral hemorrhage, large or small lacerations in the brain, and/or nerve damage, which is caused by shear forces. The brain may also experience some secondary types of injury, such as swelling, fever, seizures, or neurological chemical imbalances. Symptoms of TBI can be mild, moderate or severe, depending on the extent of brain damage. People with mild TBI may remain conscious or may experience loss of consciousness for seconds or minutes. Other symptoms of mild TBI include headache, confusion, dizziness, blurred vision or eye fatigue, tinnitus, an unpleasant taste in the mouth, fatigue or lethargy, a change in sleep pattern, a change in behavior or mood, and difficulty in memory, concentration, attention, or thinking. Persons with moderate or severe TBI may exhibit these same symptoms, but may also have a worsening or unabated headache, repeated vomiting or nausea, convulsions or seizures, inability to wake from sleep, enlargement of one or both pupils of the eyes, unclear mouth and teeth, weakness or numbness in the limbs, loss of coordination, and increased confusion, restlessness, or agitation. Examples of TBI include, but are not limited to, diffuse axonal injury, concussion, contusion, impingement injury, secondary impact syndrome, punch-through, wobble infant syndrome, and atresia syndrome.

By "chronic tauopathy" is used herein to generally mean a condition associated with increased Tau efflorescence in the extracellular fluid of a subject, e.g., accumulation of Tau in the extracellular fluid (e.g., CSF, ISF, blood, and/or blood fractions (e.g., plasma)) over a relatively long period of time (e.g., years, e.g., decades). Chronic tauopathies include, but are not necessarily limited to: alzheimer's disease, amyotrophic lateral sclerosis/Parkinson-dementia syndrome, dementia with silvery particles, amyloid angiopathy of the British type, cerebral amyloid angiopathy, corticobasal degeneration, Creutzfeldt-Jakob disease, dementia pugilistica, diffuse neurofibrillary tangles with calcification, Down's syndrome, frontotemporal dementia (FTD), frontotemporal dementia with Parkinson's disease linked to chromosome 17, frontotemporal degeneration, Gerstmann-Straussler-Scheinker disease, Hallervorden-Scherts disease, Inclusion body myositis, multiple system atrophy, myotonic dystrophy, Niemann-Pick (Nieman-Pick) disease of the C type, non-synaptic neuron disease with neurofibrillar tangles, Pick's disease, post-parkinsonism disease, Prion protein cerebral amyloid angiopathy, progressive subcortical gliosis, progressive supranuclear palsy, subacute sclerosing panencephalitis, tangle-only dementia, and multi-infarct dementia.

Before the present invention is further described, it is to be understood that this invention is not limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.

Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.

It should be noted that, as used herein and in the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "an anti-Tau antibody" includes a plurality of such antibodies, and reference to "the tauopathy" includes reference to one or more tauopathies and equivalents thereof known to those skilled in the art, and so forth. It is further noted that the claims may be drafted with the exclusion of any optional elements. Thus, to the extent that an exclusive term, such as "alone," "only," or "negative" limitation is used in connection with a claim element is recited, such recitation is intended to serve as a basis for the prior art.

It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination. All combinations that are embodiments of the present invention are specifically embraced by the present invention and are disclosed herein just as if each and every combination were individually and specifically disclosed. In addition, all subcombinations of the various embodiments and elements thereof are also expressly contemplated and disclosed herein as if each such subcombination was individually and specifically disclosed herein.

The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such publication by virtue of prior invention. Moreover, the dates of publication provided may be different from the actual publication dates which may need to be independently confirmed.

Detailed Description

The present invention provides methods for treating tauopathy in an individual.

Methods of treating tauopathy

The present invention provides methods of treating tauopathies, such as acute tauopathies. The method generally involves administering to an individual in need thereof an effective amount of an anti-Tau antibody or a pharmaceutical composition comprising an anti-Tau antibody. In some cases, the anti-Tau antibody specifically binds to an epitope within the N-terminal region of Tau. In some cases, the anti-Tau antibody specifically binds to an epitope within an N-terminal region of extracellular Tau (etau). In some cases, the antibody is humanized. In some cases, the extracellular fluid is cerebrospinal fluid (CSF), interstitial fluid (ISF), blood, or a blood fraction (e.g., a blood fraction such as serum or plasma). In some cases, the tauopathy is an acute tauopathy, such as stroke, chronic traumatic encephalopathy, traumatic brain injury, concussion, seizure, epilepsy (e.g., Dravet syndrome, also known as infantile Severe Myoclonic Epilepsy (SMEI)), and acute plumbeous encephalopathy. As described by Gheyara et al, Tau reduction may be of therapeutic benefit in Dravet syndrome or other refractory genetic epilepsy (Ann neurol.2014Sep; 76(3): 443-56). Thus, the methods described herein may be useful for treating any acute tauopathy, including, for example, epilepsy (e.g., Dravet syndrome).

In some cases, the level of free Tau is reduced. By "free Tau" is meant a Tau polypeptide that is not bound to an anti-Tau antibody. In one embodiment, the free Tau is extracellular Tau (etau). Total Tau includes free Tau and Tau bound to an anti-Tau antibody. In some cases, the level of overall Tau is decreased. In some cases, the level of bound Tau (Tau bound to anti-Tau antibodies) in the extracellular fluid is elevated.

The invention provides a method of treating a tauopathy (e.g., an acute tauopathy) in an individual, the method comprising administering to the individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid (e.g., CSF, ISF, blood, or a blood fraction (e.g., serum or plasma)) of the individual. In some embodiments, the level of Tau (e.g., total Tau and/or free Tau) is significantly reduced within 36 hours of administration of the anti-Tau antibody. For example, in some cases, an effective amount of an anti-Tau antibody is an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid within 48 hours, 36 hours, 24 hours, 12 hours, 8 hours, 4 hours, 2 hours, 1 hour, 30 minutes, 15 minutes, or 5 minutes of administration of the anti-Tau antibody. For example, in some cases, an effective amount of an anti-Tau antibody is an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid within 5 minutes to about 10 minutes, about 10 minutes to about 15 minutes, about 15 minutes to about 30 minutes, about 30 minutes to about 1 hour, about 1 hour to about 2 hours, about 2 hours to about 4 hours, about 4 hours to about 8 hours, about 8 hours to about 12 hours, about 12 hours to about 24 hours, about 24 hours to about 36 hours, about 24 to about 48 hours, or about 36 hours to about 48 hours.

A significant reduction in the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid (e.g., CSF, ISF, blood, or blood fraction (e.g., serum or plasma)) of a subject is at least a 10% reduction, at least a 15% reduction, at least a 20% reduction, at least a 25% reduction, at least a 30% reduction, at least a 40% reduction, at least a 45% reduction, at least a 50% reduction, at least a 75% reduction, at least an 80% reduction, at least an 85% reduction, at least a 90% reduction, at least a 95% reduction, or greater than a 90% reduction. In some embodiments, the level of Tau (e.g., total Tau and/or free Tau) in the extracellular fluid is reduced to a normal, control level (e.g., about 100 pg/ml). In some embodiments, the level of Tau (e.g., total Tau and/or free Tau) in the extracellular fluid is reduced to an undetectable level. In some cases, the extracellular fluid is CSF. In other cases, the extracellular fluid is interstitial fluid (ISF). In other cases, the extracellular fluid is plasma. In other cases, the extracellular fluid is whole blood. In other cases, the extracellular fluid is serum.

The present invention provides methods of treating tauopathy (e.g., acute tauopathy) in an individual. The methods generally involve administering to the subject an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid (e.g., CSF, ISF, blood, or a blood fraction (e.g., serum or plasma)) of the subject.

A significant reduction in the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid of a subject is at least a 10% reduction, at least a 15% reduction, at least a 20% reduction, at least a 25% reduction, at least a 30% reduction, at least a 35% reduction, at least a 40% reduction, at least a 45% reduction, at least a 50% reduction, at least a 55% reduction, at least a 60% reduction, at least a 65% reduction, at least a 70% reduction, at least a 75% reduction, at least an 80% reduction, at least an 85% reduction, at least a 90% reduction, at least a 95% reduction, or greater than a 90% reduction. In some embodiments, the level of Tau (e.g., total Tau and/or free Tau) in the extracellular fluid is reduced to a normal, control level (e.g., about 100 pg/ml). In some embodiments, the level of Tau (e.g., total Tau and/or free Tau) in the extracellular fluid is reduced to an undetectable level. In some cases, the extracellular fluid is CSF. In other cases, the extracellular fluid is interstitial fluid (ISF). In other cases, the extracellular fluid is plasma. In other cases, the extracellular fluid is serum. In other cases, the extracellular fluid is whole blood.

In some cases, a method of treating a tauopathy (e.g., an acute tauopathy) of the invention involves administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid (e.g., CSF, ISF, blood, or a blood fraction (e.g., serum or plasma)) of the individual, wherein the anti-Tau antibody is effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in the extracellular fluid within 48 hours of administration of the anti-Tau antibody. For example, in some cases, a method of the invention for treating a tauopathy (e.g., an acute tauopathy) involves administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid of the individual, wherein the anti-Tau antibody is effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in the extracellular fluid within 48 hours, 36 hours, 24 hours, 12 hours, 8 hours, 4 hours, 2 hours, 1 hour, or 30 minutes (or less than 30 minutes) of administration of the anti-Tau antibody. For example, in some cases, a method of the invention for treating a tauopathy (e.g., an acute tauopathy) involves administering to an individual an anti-Tau antibody in an amount effective to significantly reduce a level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid of the individual, wherein the anti-Tau antibody is effective to significantly reduce a level of Tau (e.g., total Tau and/or free Tau) in the extracellular fluid over a period of time of about 15 minutes to about 30 minutes, about 30 minutes to about 1 hour, about 1 hour to about 2 hours, about 2 hours to about 4 hours, about 4 hours to about 8 hours, about 8 hours to about 12 hours, about 12 hours to about 24 hours, about 24 hours to about 36 hours, or about 36 hours to about 48 hours.

In some cases, a method of treating a tauopathy (e.g., an acute tauopathy) of the invention involves administering to an individual an anti-Tau antibody in an amount effective to significantly reduce a level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid (e.g., CSF, ISF, blood, or a blood fraction (e.g., serum or plasma)) of the individual, wherein the reduced level of Tau (e.g., total Tau and/or free Tau) is maintained for a period of at least 2 hours following administration of the anti-Tau antibody. For example, in some cases, a method of the invention for treating a tauopathy (e.g., an acute tauopathy) involves administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in extracellular fluid of the individual, wherein the reduced level of Tau (e.g., total Tau and/or free Tau) is maintained for a period of at least 2 hours, at least 4 hours, at least 8 hours, at least 12 hours, at least 24 hours, at least 36 hours, at least 48 hours, at least 72 hours, at least 96 hours, at least 120 hours, at least 144 hours, at least 168 hours, or greater than 168 hours after administration of the anti-Tau antibody. For example, in some cases, a method of the invention for treating a tauopathy (e.g., an acute tauopathy) involves administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in extracellular fluid of the individual, wherein the reduced level of Tau (e.g., total Tau and/or free Tau) is maintained for a period of time of about 2 hours to about 4 hours, about 4 hours to about 8 hours, about 8 hours to about 12 hours, about 12 hours to about 24 hours, about 24 hours to about 36 hours, about 36 hours to about 48 hours, about 48 hours to about 72 hours, about 72 hours to about 96 hours, about 96 hours to about 120 hours, about 120 hours to about 144 hours, about 144 hours to about 168 hours, or greater than 168 hours (e.g., 8 days, 10 days, 14 days, or greater than 14 days). In some cases, the methods of the invention for treating a tauopathy (e.g., an acute tauopathy) involve administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in the extracellular fluid of the individual, wherein the reduced level of Tau (e.g., total Tau and/or free Tau) is maintained for a period of at least 7 days, at least 10 days, at least 2 weeks, or at least 4 weeks. For example, in some cases, a method of the invention for treating a tauopathy (e.g., an acute tauopathy) involves administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in the extracellular fluid of the individual, wherein the reduced level of Tau (e.g., total Tau and/or free Tau) is maintained for a period of time of about 7 days to about 10 days, about 10 days to about 2 weeks, or about 2 weeks to about 4 weeks, or greater than 4 weeks (e.g., 3 months, 4 months, 6 months, or greater than 6 months).

In some cases, a method of treating a tauopathy (e.g., an acute tauopathy) of the invention involves administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid (e.g., CSF, ISF, blood, or a blood fraction (e.g., serum or plasma)) of the individual, wherein the reduced level of Tau (e.g., total Tau and/or free Tau) is maintained for a period of time that provides a reduction in the level of Α β in the extracellular fluid (e.g., CSF, ISF, blood, or a blood fraction (e.g., serum or plasma)). For example, in some embodiments, the level of Abeta (a β) in the extracellular fluid is significantly reduced over a period of time from about 1 day to about 25 days after administration of the anti-Tau antibody. For example, in some embodiments, the level of a β in the extracellular fluid is significantly reduced over a period of time from about 1 day to about 5 days, from about 5 days to about 10 days, from about 10 days to about 15 days, from about 15 days to about 20 days, or from about 20 days to about 25 days after administration of the anti-Tau antibody. The anti-Tau antibody may be administered to provide continuous inhibition of Α β levels over time. A β includes a β 40 and a β 42. In some cases, a β 40 levels are decreased. In some cases, a β 42 levels are decreased. In some cases, both a β 40 and a β 42 levels are reduced. A "significant decrease" in a β level is at least a 5% decrease, at least a 10% decrease, at least a 15% decrease, at least a 20% decrease, at least a 25% decrease, at least a 30% decrease, at least a 40% decrease, at least a 45% decrease, at least a 50% decrease, or greater than a 50% decrease in a β level compared to a β level without administration of the anti-Tau antibody (e.g., compared to a β level prior to administration of the anti-Tau antibody).

In some cases, the methods of treating tauopathies (e.g., acute tauopathies) of the invention involve administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid (e.g., CSF, ISF, blood, or a blood fraction (e.g., serum or plasma)) of the individual, wherein the extracellular fluid is CSF. In some cases, the methods of the invention for treating a tauopathy (e.g., an acute tauopathy) involve administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid of the individual, wherein the extracellular fluid is ISF. In some cases, the methods of the invention for treating a tauopathy (e.g., an acute tauopathy) involve administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid of the individual, wherein the extracellular fluid is plasma.

In some cases, the methods of treating tauopathy (e.g., acute tauopathy) of the present invention involve administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid (e.g., CSF, ISF, blood, or a blood fraction (e.g., serum or plasma)) of the individual, wherein the anti-Tau antibody is administered by subcutaneous administration (e.g., by subcutaneous injection). In some cases, the methods of the invention for treating a tauopathy (e.g., an acute tauopathy) involve administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in the extracellular fluid of the individual, wherein the anti-Tau antibody is administered by intrathecal administration. In some cases, the methods of the invention for treating a tauopathy (e.g., an acute tauopathy) involve administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid of the individual, wherein the anti-Tau antibody is administered by intravenous administration (e.g., by intravenous injection).

In some cases, the methods of the invention for treating a tauopathy (e.g., an acute tauopathy) involve administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid (e.g., CSF, ISF, blood, or a blood fraction (e.g., serum or plasma)) of the individual, wherein the anti-Tau antibody is administered in an amount of about 0.1mg/kg body weight to about 50mg/kg body weight. For example, in some cases, a method of the invention for treating a tauopathy (e.g., an acute tauopathy) involves administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid of the individual, wherein the anti-Tau antibody is administered in an amount from about 0.1mg/kg body weight to about 0.5mg/kg body weight, from about 0.5mg/kg body weight to about 1mg/kg body weight, from about 1mg/kg body weight to about 5mg/kg body weight, from about 5mg/kg body weight to about 10mg/kg body weight, from about 10mg/kg body weight to about 15mg/kg body weight, from about 15mg/kg body weight to about 20mg/kg body weight, from about 20mg/kg body weight to about 25mg/kg body weight, from about 25mg/kg body weight to about 30mg/kg body weight, from about 30mg/kg body weight to about 35mg/kg body weight, From about 35mg/kg body weight to about 40mg/kg body weight, from about 40mg/kg body weight to about 45mg/kg body weight, or from about 45mg/kg body weight to about 50mg/kg body weight, or more than 50mg/kg body weight.

In some cases, the anti-Tau antibody is administered at a dose of about 0.1mg/kg body weight to about 0.5mg/kg body weight, about 0.5mg/kg body weight to about 1mg/kg body weight, about 1mg/kg body weight to about 5mg/kg body weight, about 5mg/kg body weight to about 10mg/kg body weight, about 10mg/kg body weight to about 15mg/kg body weight, about 15mg/kg body weight to about 20mg/kg body weight, about 20mg/kg body weight to about 25mg/kg body weight, (ii) an amount of about 25mg/kg body weight to about 30mg/kg body weight, about 30mg/kg body weight to about 35mg/kg body weight, about 35mg/kg body weight to about 40mg/kg body weight, about 40mg/kg body weight to about 45mg/kg body weight, or about 45mg/kg body weight to about 50mg/kg body weight, or more than 50mg/kg body weight; and the anti-Tau antibody is administered in a single dose.

In some cases, the anti-Tau antibody is administered at a dose of about 0.1mg/kg body weight to about 0.5mg/kg body weight, about 0.5mg/kg body weight to about 1mg/kg body weight, about 1mg/kg body weight to about 5mg/kg body weight, about 5mg/kg body weight to about 10mg/kg body weight, about 10mg/kg body weight to about 15mg/kg body weight, about 15mg/kg body weight to about 20mg/kg body weight, about 20mg/kg body weight to about 25mg/kg body weight, (ii) an amount of about 25mg/kg body weight to about 30mg/kg body weight, about 30mg/kg body weight to about 35mg/kg body weight, about 35mg/kg body weight to about 40mg/kg body weight, about 40mg/kg body weight to about 45mg/kg body weight, or about 45mg/kg body weight to about 50mg/kg body weight, or more than 50mg/kg body weight; and the anti-Tau antibody is administered in multiple doses (2 or more doses).

In some cases, the anti-Tau antibody is administered at a dose of about 0.1mg/kg body weight to about 0.5mg/kg body weight, about 0.5mg/kg body weight to about 1mg/kg body weight, about 1mg/kg body weight to about 5mg/kg body weight, about 5mg/kg body weight to about 10mg/kg body weight, about 10mg/kg body weight to about 15mg/kg body weight, about 15mg/kg body weight to about 20mg/kg body weight, about 20mg/kg body weight to about 25mg/kg body weight, (ii) an amount of about 25mg/kg body weight to about 30mg/kg body weight, about 30mg/kg body weight to about 35mg/kg body weight, about 35mg/kg body weight to about 40mg/kg body weight, about 40mg/kg body weight to about 45mg/kg body weight, or about 45mg/kg body weight to about 50mg/kg body weight, or more than 50mg/kg body weight; and the anti-Tau antibody is administered in a plurality of doses, e.g., the anti-Tau antibody is administered 1 time per hour, 1 time per 2 hours, 1 time per 3 hours, 1 time per 4 hours, 1 time per 5 hours, 1 time per 6 hours, 1 time per 7 hours, 1 time per 8 hours, 1 time per 9 hours, 1 time per 10 hours, 1 time per 12 hours, 1 time per 24 hours, 1 time per 48 hours, 1 time per 3 days, 1 time per 4 days, 1 time per 5 days, 1 time per 6 days, 1 time per 7 days, 1 time per 2 weeks, 1 time per month, 1 time per 2 months, 1 time per 4 months, 1 time per 6 months, or 1 time per year.

In some cases, the anti-Tau antibody is administered at a dose of about 0.1mg/kg body weight to about 0.5mg/kg body weight, about 0.5mg/kg body weight to about 1mg/kg body weight, about 1mg/kg body weight to about 5mg/kg body weight, about 5mg/kg body weight to about 10mg/kg body weight, about 10mg/kg body weight to about 15mg/kg body weight, about 15mg/kg body weight to about 20mg/kg body weight, about 20mg/kg body weight to about 25mg/kg body weight, (ii) an amount of about 25mg/kg body weight to about 30mg/kg body weight, about 30mg/kg body weight to about 35mg/kg body weight, about 35mg/kg body weight to about 40mg/kg body weight, about 40mg/kg body weight to about 45mg/kg body weight, or about 45mg/kg body weight to about 50mg/kg body weight, or more than 50mg/kg body weight; and the anti-Tau antibody is administered in multiple doses, e.g., an initial dose of the anti-Tau antibody is administered within 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours, 12 hours, 24 hours, 2 days, 4 days, 1 week, 2 weeks, 4 weeks, or 2 months of damage associated with physical damage to the brain and/or central nervous system-associated tissue of the subject that results in elevated Tau levels; and the subsequent agent of anti-Tau antibody is administered at a time period of about 1 hour to about 1 year or more (e.g., about 1 hour to about 4 hours, about 4 hours to about 8 hours, about 8 hours to about 12 hours, about 12 hours to about 24 hours, about 24 hours to about 2 days, about 2 days to about 4 days, about 4 days to about 7 days, about 1 week to about 2 weeks, about 2 weeks to about 4 weeks, about 4 weeks to about 2 months, about 2 months to about 4 months, about 4 months to about 6 months, about 6 months to about 1 year, or greater than 1 year) after the administration of the initial agent of anti-Tau antibody.

In some cases, a method of treating a tauopathy (e.g., an acute tauopathy) of the invention involves administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid (e.g., CSF, ISF, blood, or a blood fraction (e.g., serum or plasma)) of the individual, wherein the anti-Tau antibody is administered in a single bolus injection (bolus injection).

In other instances, the methods of the invention for treating a tauopathy (e.g., an acute tauopathy) involve administering to an individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in an extracellular fluid (e.g., CSF, ISF, blood, or a blood fraction (e.g., serum or plasma)) of the individual, wherein the anti-Tau antibody is administered in multiple doses (e.g., 2, 3,4, 5, or more doses). When multiple doses are administered, the dosing interval may be hourly, every 2 hours, every 3 hours, every 4 hours, every 5 hours, every 6 hours, every 7 hours, every 8 hours, every 9 hours, every 10 hours, every 12 hours, every 24 hours, every 48 hours, every 3 days, every 4 days, every 5 days, every 6 days, every 7 days, and the like.

The invention provides methods of treating a tauopathy, e.g., an acute tauopathy, in an individual, wherein the methods involve administering to the individual an anti-Tau antibody in an amount effective to provide a minimal concentration of anti-Tau antibody in the cerebrospinal fluid (CSF) of the individual. In some cases, the minimum anti-Tau antibody concentration in the CSF is reached within 30 minutes of administration of the anti-Tau antibody. In some cases, the minimum anti-Tau antibody concentration in the CSF is reached within 1 hour of administration of the anti-Tau antibody. In some cases, the minimum anti-Tau antibody concentration in the CSF is achieved within 48 hours, 36 hours, 24 hours, 12 hours, 8 hours, 4 hours, 2 hours, 1 hour, or 30 minutes (or less than 30 minutes) of administration of the anti-Tau antibody. In some cases, the minimum anti-Tau antibody concentration in the CSF is achieved over a period of time of about 15 minutes to about 30 minutes, about 30 minutes to about 1 hour, about 1 hour to about 2 hours, about 2 hours to about 4 hours, about 4 hours to about 8 hours, about 8 hours to about 12 hours, about 12 hours to about 24 hours, about 24 hours to about 36 hours, or about 36 hours to about 48 hours.

In some cases, a method of the invention for treating a tauopathy (e.g., an acute tauopathy) in an individual involves administering to the individual an anti-Tau antibody in an amount effective to provide a minimum concentration of anti-Tau antibody in the cerebrospinal fluid (CSF) of the individual, wherein the minimum anti-Tau antibody concentration in the CSF is at least 20 ng/ml. For example, in some cases, a method of the invention for treating a tauopathy, e.g., an acute tauopathy, in an individual involves administering to the individual an anti-Tau antibody in an amount effective to provide a minimal anti-Tau antibody concentration in cerebrospinal fluid (CSF) of the individual, wherein the minimal anti-Tau antibody concentration in CSF is at least 20ng/ml, at least 25ng/ml, at least 30ng/ml, at least 40ng/ml, at least 50ng/ml, at least 60ng/ml, at least 75ng/ml, at least 100ng/ml, at least 125ng/ml, at least 150ng/ml, at least 175ng/ml, at least 200ng/ml, at least 250ng/ml, at least 300ng/ml, at least 350ng/ml, at least 400ng/ml, at least 450ng/ml, or a combination thereof, At least 500ng/ml, at least 550ng/ml, at least 600ng/ml, at least 650ng/ml, at least 700ng/ml, at least 750ng/ml, or at least 800 ng/ml. For example, in some cases, a method of the invention for treating a tauopathy, e.g., an acute tauopathy, in an individual involves administering to the individual an anti-Tau antibody in an amount effective to provide a minimal anti-Tau antibody concentration in cerebrospinal fluid (CSF) of the individual, wherein the minimal anti-Tau antibody concentration in CSF is about 20ng/ml to about 30ng/ml, about 30ng/ml to about 40ng/ml, about 40ng/ml to about 50ng/ml, about 50ng/ml to about 60ng/ml, about 60ng/ml to about 75ng/ml, about 75ng/ml to about 100ng/ml, about 100ng/ml to about 150ng/ml, about 150ng/ml to about 200ng/ml, about 200ng/ml to about 250ng/ml, about 250ng/ml to about 300ng/ml, about 100ng/ml, or a combination thereof, About 300ng/ml to about 350ng/ml, about 350ng/ml to about 400ng/ml, about 400ng/ml to about 450ng/ml, about 450ng/ml to about 500ng/ml, about 500ng/ml to about 550ng/ml, about 550ng/ml to about 600ng/ml, about 600ng/ml to about 700ng/ml, about 700ng/ml to about 800ng/ml, or more than 800 ng/ml.

In some cases, a method of the invention for treating a tauopathy (e.g., an acute tauopathy) in an individual involves administering to the individual an anti-Tau antibody in an amount effective to provide a minimum concentration of anti-Tau antibody in the cerebrospinal fluid (CSF) of the individual, wherein the minimum concentration of anti-Tau antibody in the CSF provides a molar anti-Tau antibody to Tau ratio in the CSF of at least 2: 1. For example, in some cases, a method of the invention for treating a tauopathy (e.g., an acute tauopathy) in an individual involves administering to the individual an anti-Tau antibody in an amount effective to provide a concentration of minimal anti-Tau antibody in cerebrospinal fluid (CSF) of the individual, wherein the minimal anti-Tau antibody concentration in CSF provides a molar ratio of anti-Tau antibody to Tau in CSF of at least 2:1, at least 2.5:1, at least 3:1, at least 3.5:1, at least 4:1, at least 4.5:1, at least 5:1, at least 6:1, at least 7:1, at least 8:1, at least 9:1, or at least 10: 1.

In some cases, the methods of the invention for treating a tauopathy (e.g., an acute tauopathy) in an individual involve administering to the individual an anti-Tau antibody in an amount effective to provide a minimal concentration of anti-Tau antibody in cerebrospinal fluid (CSF) of the individual, wherein the acute tauopathy is traumatic brain injury. In some cases, the methods of the invention for treating a tauopathy (e.g., an acute tauopathy) in an individual involve administering to the individual an anti-Tau antibody in an amount effective to provide a minimal concentration of anti-Tau antibody in cerebrospinal fluid (CSF) of the individual, wherein the acute tauopathy is a stroke.

The present invention provides methods for treating Traumatic Brain Injury (TBI) in an individual, the methods generally involving administering to the individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in the extracellular fluid of the individual. In some cases, the antibody is administered within 48 hours of traumatic brain injury. In some cases, the antibody is administered within 48 hours, 36 hours, 24 hours, 12 hours, 8 hours, 4 hours, 2 hours, 1 hour, or 30 minutes (or less than 30 minutes) of TBI.

The present invention provides methods for treating stroke in an individual, the methods generally involving administering to the individual an anti-Tau antibody in an amount effective to significantly reduce the level of Tau (e.g., total Tau and/or free Tau) in the extracellular fluid of the individual. In some cases, the antibody is administered within 48 hours of stroke. In some cases, the antibody is administered within 48 hours, 36 hours, 24 hours, 12 hours, 8 hours, 4 hours, 2 hours, 1 hour, or 30 minutes (or less than 30 minutes) of the stroke.

The amount of free Tau (e.g., free extracellular Tau (eTau)) in the extracellular fluid that is not bound to the anti-eTau antibody can be determined as follows. The amount of free Tau can be determined by a method comprising the steps of: a) contacting an immobilized antibody with an extracellular fluid (e.g., CSF, ISF, serum, blood, or plasma) sample obtained from a subject, wherein the immobilized antibody competes for binding to eTau with an anti-eTau antibody administered to the subject, and wherein the contacting is performed under conditions suitable for unbound eTau to bind to the immobilized antibody; and b) determining the amount of eTau bound to the immobilized antibody. The amount of eTau bound to the immobilized antibody is indicative of the amount of eTau not bound to the anti-Tau antibody in the sample. In some cases, the amount of eTau bound to the immobilized antibody is determined with a detectably labeled third antibody that does not compete with the immobilized antibody for binding to eTau.

Antibodies

anti-Tau antibodies (or VH/VL domains or CDRs derived from said antibodies) suitable for use in the present invention may be produced using methods well known in the art. Alternatively, art-recognized anti-Tau antibodies may be used. Antibodies that bind to the same Tau epitope and/or compete therewith for binding to Tau as any of these art-recognized antibodies may also be used.

An exemplary anti-Tau antibody is hu-IPN002 (also known as IPN007 and IPN002 variant 2) comprising a heavy chain and a light chain (having the sequences shown in SEQ ID NOS: 37 and 41, respectively), or antigen-binding fragments and variants thereof. hu-IPN002 is a humanized immunoglobulin (IgG4) monoclonal antibody that binds to extracellular Tau.

In other embodiments, the antibody comprises the heavy and light chain CDRs or variable regions of hu-IPN 002. Thus, in one embodiment, the antibody comprises the CDR1, CDR2, and CDR3 domains of the VH region of hu-IPN002 (having the sequence shown in SEQ ID NO: 37), and the CDR1, CDR2, and CDR3 domains of the VL region of hu-IPN002 (having the sequence shown in SEQ ID NO: 41). In another embodiment, the antibody comprises heavy chain CDR1, CDR2, and CDR3 domains (having the sequences shown in SEQ ID NOs: 10, 11, and 12, respectively) and light chain CDR1, CDR2, and CDR3 domains (having the sequences shown in SEQ ID NOs: 7, 8, and 9, respectively). In another embodiment, the antibody comprises VH and/or VL regions having the amino acid sequences shown in SEQ ID NO 37 and/or SEQ ID NO 41, respectively. In another embodiment, the antibody comprises a heavy chain variable region (VH) and/or a light chain variable region (VL) encoded by the nucleic acid sequences shown in SEQ ID NO:29 and/or SEQ ID NO:33, respectively. In another embodiment, the antibody competes for binding with the above antibody, and/or binds to the same epitope on Tau. In another embodiment, the antibody has at least about 90% variable region amino acid sequence identity to an antibody described above (e.g., at least about 90%, 95%, or 99% variable region identity to SEQ ID NO:37 or SEQ ID NO: 41).

In one embodiment, an antibody that binds an N-terminal region of a Tau polypeptide, and an antibody suitable for use in a method of the invention for treating a tauopathy (e.g., an acute tauopathy), is an antibody that binds an epitope of Tau within amino acids 2-176 of Tau, e.g., within amino acids 2-15, amino acids 15-24, amino acids 24-50, amino acids 2-25, amino acids 15-50, amino acids 50-75, amino acids 40-60, amino acids 75-100, amino acids 60-80, amino acids 100-125, amino acids 80-115, amino acids 125-150, amino acids 115-140, amino acids 150-176, or amino acids 140-160 of Tau. Exemplary Tau polypeptides are shown in figure 9; an antibody suitable for treating a tauopathy (e.g. an acute tauopathy) in an individual may be a humanized antibody that specifically binds to an epitope in a Tau polypeptide as shown in figure 9. Fig. 21 depicts an example of an eTau polypeptide; an antibody suitable for treating a tauopathy (e.g. an acute tauopathy) in an individual may be a humanized antibody that specifically binds to an epitope in a Tau polypeptide as shown in figure 21.

Humanized antibodies that bind to an N-terminal region of a Tau polypeptide, and humanized antibodies suitable for use in the methods of the invention for treating tauopathies (e.g., acute tauopathies), are humanized antibodies that bind to an epitope of Tau within amino acids 2-176 of Tau, e.g., within amino acids 2-15, amino acids 15-24, amino acids 24-50, amino acids 2-25, amino acids 15-50, amino acids 50-75, amino acids 40-60, amino acids 75-100, amino acids 60-80, amino acids 100-125, amino acids 80-115, amino acids 125-150, amino acids 115-140, amino acids 150-176, or amino acids 140-160. Exemplary Tau polypeptides are shown in figure 9; the antibody that binds to the N-terminal region of the Tau polypeptide, and antibodies suitable for use in the methods of the invention for treating tauopathies, may be humanized antibodies that specifically bind to an epitope in the Tau polypeptide shown in figure 9.

In some cases, antibodies that bind to the N-terminal region of a Tau polypeptide, and antibodies suitable for use in the methods of the invention for treating tauopathies (e.g., acute tauopathies), are humanized anti-Tau antibodies that bind to an epitope within amino acids 15-24 of Tau.

In some cases, an antibody that binds an N-terminal region of a Tau polypeptide, and an antibody suitable for use in a method of the invention for treating a tauopathy (e.g., an acute tauopathy), is an antibody that binds an epitope of Tau within amino acids 1-158 of Tau, e.g., amino acids 1-15, amino acids 7-13, amino acids 2-18, amino acids 15-24, amino acids 19-46, amino acids 24-50, amino acids 2-25, amino acids 25-30, amino acids 15-50, amino acids 28-126, amino acids 50-75, amino acids 40-60, amino acids 75-100, amino acids 60-80, amino acids 100-125, amino acids 80-115, amino acids 125-150, amino acids 115-140, or amino acids 150-158, wherein the amino acid numbering is based on the amino acid number of Tau of 2N4R, as shown in fig. 9. In some cases, the antibody is humanized.

In some cases, the methods of the invention involve treating a tauopathy (e.g., an acute tauopathy) by administering an anti-Tau antibody, wherein the antibody binds to an epitope comprising amino acid residues within amino acids 1-158 of Tau, wherein the amino acid numbering is based on the 2N4R Tau amino acid sequence shown in figure 9. In some cases, the administered anti-Tau antibody specifically binds Tau, wherein the epitope bound by the antibody comprises amino acid residues within amino acids 2-18 of Tau. In some cases, the administered anti-Tau antibody specifically binds Tau, wherein the epitope bound by the antibody is a linear epitope, and wherein the epitope bound by the antibody comprises amino acid residues within amino acids 2-68 of Tau. In some cases, the administered anti-Tau antibody specifically binds to a Tau4 polypeptide having at least 95%, at least 98%, at least 99%, or 100% amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 71. In some cases, the administered anti-Tau antibody specifically binds to a linear epitope within the Tau polypeptide, wherein the epitope is within amino acids 2-68 of Tau. In some cases, the administered anti-Tau antibody specifically binds to a linear epitope within the Tau polypeptide, wherein the epitope is within amino acids 15-24 of Tau. In some cases, the administered anti-Tau antibody specifically binds Tau, wherein the epitope bound by the antibody comprises amino acid residues within amino acids 7-13 of Tau, e.g., amino acid EFEVMED (SEQ ID NO: 21). In some cases, the administered anti-Tau antibody specifically binds Tau, wherein the epitope bound by the antibody comprises amino acid residues within amino acids 25-30 of Tau, such as the amino acids DQGGYT (SEQ ID NO: 22). In some cases, the administered anti-Tau antibody specifically binds Tau, wherein the epitope bound by the antibody comprises amino acid residues within amino acids 28-126 of Tau, wherein the amino acid numbering is based on the 2N4R Tau amino acid sequence shown in figure 9. In some cases, the administered anti-Tau antibody specifically binds Tau, wherein the epitope bound by the antibody comprises amino acid residues within amino acid 150-158 of Tau, wherein the amino acid numbering is based on the 2N4R Tau amino acid sequence shown in figure 9. In some cases, the administered anti-Tau antibody specifically binds Tau, wherein the epitope bound by the antibody comprises amino acid residues within amino acids 19-46 of Tau, wherein the amino acid numbering is based on the 2N4R Tau amino acid sequence shown in figure 9.

In some cases, the methods of the invention involve treating a tauopathy (e.g., an acute tauopathy) by administering an antibody that specifically binds to extracellular Tau (etau), wherein the antibody binds to an epitope comprising amino acid residues within amino acids 1-158 of Tau, wherein the amino acid numbering is based on the 2N4R Tau amino acid sequence shown in figure 9. In some cases, the administered anti-Tau antibody specifically binds eTau, wherein the epitope bound by the antibody comprises amino acid residues within amino acids 2-18 of eTau. In some cases, the administered anti-Tau antibody specifically binds eTau, wherein the epitope bound by the antibody is a linear epitope, and wherein the epitope bound by the antibody comprises amino acid residues within amino acids 2-68 of eTau. In some cases, the administered anti-Tau antibody specifically binds an eTau4 polypeptide having at least 95%, at least 98%, at least 99%, or 100% amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 71. In some cases, the administered anti-Tau antibody specifically binds a linear epitope within eTau4 polypeptide, wherein the epitope is within amino acids 2-68 of eTau 4. In some cases, the administered anti-Tau antibody specifically binds a linear epitope within eTau4 polypeptide, wherein the epitope is within amino acids 15-24 of eTau 4. In some cases, the administered anti-Tau antibody specifically binds eTau, wherein the epitope bound by the antibody comprises amino acid residues within amino acids 7-13 of eTau, e.g., amino acid EFEVMED (SEQ ID NO: 21). In some cases, the administered anti-Tau antibody specifically binds eTau, wherein the epitope bound by the antibody comprises amino acid residues within amino acids 25-30 of eTau, such as the amino acids DQGGYT (SEQ ID NO: 22). In some cases, the administered anti-Tau antibody specifically binds eTau, wherein the epitope bound by the antibody comprises amino acid residues within amino acids 28-126 of eTau, wherein the amino acid numbering is based on the 2N4R Tau amino acid sequence shown in figure 9. In some cases, the administered anti-Tau antibody specifically binds eTau, wherein the epitope bound by the antibody comprises amino acid residues within amino acid 150-158 of eTau, wherein the amino acid numbering is based on the 2N4R Tau amino acid sequence shown in figure 9. In some cases, the administered anti-Tau antibody specifically binds eTau, wherein the epitope bound by the antibody comprises amino acid residues within amino acids 19-46 of eTau, wherein the amino acid numbering is based on the 2N4R Tau amino acid sequence shown in figure 9.

The present invention provides methods of treating tauopathy (e.g., acute tauopathy) in an individual. The method generally involves administering to an individual: a) an effective amount of an antibody (e.g., a monoclonal antibody), which may optionally be a humanized antibody, that binds to the N-terminal region of the Tau polypeptide; or b) a pharmaceutical composition comprising said humanized antibody.

An antibody (optionally a humanized antibody, e.g. a monoclonal antibody) that binds to an N-terminal region of a Tau polypeptide and is suitable for use in a method of the invention for treating a tauopathy, e.g. an acute tauopathy, is an antibody that binds to an epitope of Tau within amino acids 1-158 of Tau, e.g. amino acids 1-15, 7-13, 2-18, 15-24, 19-46, 24-50, 2-25, 25-30, 15-50, 28-126, 50-75, 40-60, 75-100, 60-80, 100-125, 80-115, 125-150, 115-140, or 150-158 of Tau, wherein the amino acid numbering is based on the amino acid number of 2N4R Tau, as shown in fig. 9. In some cases, the antibody is humanized.

In some cases, an antibody that binds to the N-terminal region of a Tau polypeptide and is suitable for use in a method of the invention for treating a tauopathy (e.g., an acute tauopathy) is a humanized anti-Tau antibody of the invention. In some cases, the antibody is a humanized antibody that binds to an epitope (e.g., a linear epitope) within amino acids 15-24 of Tau.

In some cases, a method of treating a tauopathy (e.g., an acute tauopathy) in an individual involves administering to the individual an effective amount of an anti-Tau antibody that does not require the presence of a 2N insert (2N insert) of Tau to bind to Tau. In some cases, the epitope recognized by an anti-Tau antibody suitable for use in the methods of the invention for treating tauopathies is not within the 2N insert of Tau. The 2N insert of Tau comprises amino acids 45-102 of the 2N4R amino acid sequence shown in figure 9.

In some cases, an anti-Tau antibody that binds to the N-terminal region of a Tau polypeptide and that is suitable for use in a method of the invention for treating a tauopathy (e.g., an acute tauopathy) specifically binds to Tau, wherein the epitope to which the antibody binds comprises amino acid residues within amino acids 2-68 of Tau. In some cases, an anti-Tau antibody that binds to the N-terminal region of a Tau polypeptide and is suitable for use in a method of the invention for treating a tauopathy (e.g., an acute tauopathy) specifically binds to extracellular Tau (eTau), wherein the epitope to which the antibody binds comprises amino acid residues within amino acids 2-68 of eTau. In some cases, an anti-Tau antibody that binds an N-terminal region of a Tau polypeptide and that is suitable for use in a method of the invention for treating a tauopathy (e.g., an acute tauopathy) specifically binds eTau, wherein the epitope bound by the antibody is a linear epitope, and wherein the epitope bound by the antibody comprises amino acid residues within amino acids 2-68 of eTau. In some cases, an anti-Tau antibody that binds to an N-terminal region of a Tau polypeptide and that is suitable for use in a method of the invention for treating a tauopathy (e.g., an acute tauopathy) specifically binds to an eTau4 polypeptide having at least 95%, at least 98%, at least 99%, or 100% amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO: 48. In some cases, an anti-Tau antibody that binds to the N-terminal region of a Tau polypeptide and that is suitable for use in a method of the invention for treating a tauopathy (e.g., an acute tauopathy) specifically binds to a linear epitope within eTau4 polypeptide, wherein the epitope is within amino acids 2-68 of eTau 4. In any of the above embodiments, the antibody may be humanized.

In some cases, an antibody that binds to the N-terminal region of a Tau polypeptide and is suitable for use in a method of the invention for treating a tauopathy (e.g. an acute tauopathy), wherein the antibody binds to an epitope comprising amino acid residues within amino acids 1 to 158 of Tau, wherein the amino acid numbering is based on the 2N4R form of Tau, for example as shown in figure 9. In some of these embodiments, the antibody is humanized. In some of these embodiments, the epitope is a linear epitope.

In some cases, an antibody that binds to the N-terminal region of a Tau polypeptide and that is suitable for use in a method of the invention for treating a tauopathy (e.g. an acute tauopathy) specifically binds to Tau, wherein the epitope to which the antibody binds comprises amino acid residues within amino acids 2-18 of Tau, wherein the amino acid numbering is based on the 2N4R form of Tau, for example as shown in figure 9. In some of these embodiments, the antibody is humanized. In some of these embodiments, the epitope is a linear epitope.

In some cases, an antibody that binds to the N-terminal region of a Tau polypeptide and that is suitable for use in a method of the invention for treating a tauopathy (e.g. an acute tauopathy) specifically binds to Tau, wherein the epitope to which the antibody binds comprises amino acid residues within amino acids 7-13 of Tau, wherein the amino acid numbering is based on the 2N4R form of Tau, for example as shown in figure 9. In some of these embodiments, the antibody is humanized. In some of these embodiments, the epitope is a linear epitope.

In some cases, an antibody that binds to the N-terminal region of a Tau polypeptide and that is suitable for use in a method of the invention for treating a tauopathy (e.g. an acute tauopathy) specifically binds to Tau, wherein the epitope to which the antibody binds comprises amino acid residues within amino acids 25-30 of Tau, wherein the amino acid numbering is based on the 2N4R form of Tau, for example as shown in figure 9. In some of these embodiments, the antibody is humanized. In some of these embodiments, the epitope is a linear epitope.

In some cases, an antibody that binds to the N-terminal region of a Tau polypeptide and that is suitable for use in a method of the invention for treating a tauopathy (e.g. an acute tauopathy) specifically binds to Tau, wherein the epitope to which the antibody binds comprises amino acid residues within amino acids 28-126 of Tau, wherein the amino acid numbering is based on the 2N4R form of Tau, for example as shown in figure 9. In some of these embodiments, the antibody is humanized. In some of these embodiments, the epitope is a linear epitope.

In some cases, an antibody that binds to the N-terminal region of a Tau polypeptide and is suitable for use in a method of the invention for treating a tauopathy (e.g. an acute tauopathy) specifically binds to Tau, wherein the epitope bound by the antibody comprises amino acid residues within amino acid 150-158 of Tau, wherein the amino acid numbering is based on the 2N4R form of Tau, for example as shown in figure 9. In some of these embodiments, the antibody is humanized. In some of these embodiments, the epitope is a linear epitope.

In some instances, anti-Tau antibodies suitable for use in the methods of the invention are anti-Tau antibodies that specifically bind to an epitope within the N-terminal region of a Tau polypeptide (e.g., a linear epitope within the amino-terminal (N-terminal) portion of Tau, such as amino acids 1-25 of Tau, amino acids 1-18 of Tau, amino acids 9-18 of Tau (where amino acids 1-18 of Tau are: MAEPRQEFEVMEDHAGTY; SEQ ID NO:23), amino acids 15-44 of Tau, amino acids 13-24 of Tau, or amino acids 15-24 of Tau (where amino acids 15-24 of Tau are an epitope within: AGTYGLGDRK (SEQ ID NO: 24)).

In some cases, a humanized monoclonal antibody suitable for use in the methods of the invention specifically binds to an epitope within amino acids 15-24 of a Tau polypeptide. In some cases, the epitope does not comprise a phosphorylated amino acid. In some cases, the epitope does not comprise a nitrated amino acid. In some cases, the epitope comprises a phosphorylated amino acid, a nitrated amino acid, or both a phosphorylated amino acid and a nitrated amino acid.

In some cases, antibodies suitable for use in the methods of the invention are humanized. Humanization of the framework regions reduces the risk of the antibody eliciting a human anti-mouse antibody (HAMA) response in humans. The HAMA response in a particular patient or during a clinical trial can be monitored using art-recognized methods of determining immune response. Immunogenicity assessments can be made on patients administered humanized antibodies at the beginning or throughout the course of administration of the treatment. For example, by detecting antibodies to the humanized therapeutic agent in a serum sample taken from the patientHAMA responses, which are detected using methods known to those skilled in the art, including surface plasmon resonance (BIACORE) and/or solid phase enzyme-linked immunosorbent assay (ELISA) assays. In many cases, suitable humanized anti-Tau antibodies elicit substantially no HAMA response in a human subject. In some cases, suitable humanized anti-Tau antibodies have reduced potential immunogenicity, e.g., with CD8+EpiScreen by depleted peripheral blood mononuclear cellsTMThe determined values were measured. In some cases, suitable humanized anti-Tau antibodies exhibit a Stimulation Index (Stimulation Index) of less than 2.0.

Some amino acids from the framework residues of the human variable region were selected for substitution based on their possible effect on CDR conformation and/or on antigen binding. The unnatural juxtaposition of murine CDR regions to human variable framework regions can result in unnatural conformational constraints that result in a loss of binding affinity (unless it is corrected by substitution of certain amino acid residues).

The choice of amino acid residues for substitution can be determined in part by computer modeling. Computer hardware and software for generating three-dimensional images of immunoglobulin molecules are known in the art. Typically, molecular models are provided starting with the interpretation of the structure of an immunoglobulin chain or domain thereof. The amino acid sequence similarity of the chain to be modeled and the chain or domain that resolves the three-dimensional structure is compared, and the chain or domain that exhibits the greatest sequence similarity is selected as the starting point for constructing the molecular model. Chains or domains with 50% sequence identity are selected for modeling, for example those with at least 60%, 70%, 80%, 90% or more than 90% sequence identity or more. The starting structures of the resolution are modified to allow for differences between the actual amino acids in the immunoglobulin chain or domain being modeled and those in the starting structures. The modified structure is then assembled into a complex immunoglobulin. Finally, the model was refined by energy minimization and by verifying that all atoms were within the right distance of each other and that the bond length and angle were within chemically acceptable limits.

CDRs and framework regions are defined by Kabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md.,1987 and 1991). An alternative structural definition is provided by Chothia et al, J.mol.biol.196:901 (1987); nature 342:878 (1989); and J.mol.biol.186:651(1989) (collectively referred to as "Chothia"). When framework residues as defined by Kabat above constitute structural loop residues as defined by Chothia above, amino acids present in mouse antibodies can be selected for substitution in humanized antibodies. Residues that "border a CDR region" include amino acid residues in positions that directly border one or more CDRs in the primary sequence of the humanized immunoglobulin chain, e.g., directly border a CDR as defined by Kabat or a CDR as defined by Chothia (see, e.g., Chothia and Lesk JMB 196:901 (1987)). These amino acids are most likely to interact with amino acids in the CDRs and, if selected from acceptors, distort donor CDRs and reduce affinity. Furthermore, contiguous amino acids may interact directly with the antigen (Amit et al, Science,233:747(1986)), and these amino acids may need to be selected from donors to maintain all antigen contacts that provide affinity in the original antibody.

Antibodies suitable for use in the methods of the invention may comprise humanized light chain framework regions; a humanized heavy chain framework region, wherein the isolated antibody competes for binding to an epitope in an N-terminal region of a Tau polypeptide with another antibody comprising: a) a light chain region comprising: (i) VL CDR1 comprising the amino acid sequence of SEQ ID NO. 1 or SEQ ID NO. 7; (ii) VL CDR2 comprising the amino acid sequence of SEQ ID NO. 2 or SEQ ID NO. 8; and (iii) a VL CDR3 comprising the amino acid sequence of SEQ ID NO. 3 or SEQ ID NO. 9; and b) a heavy chain region comprising: (i) VH CDR1 comprising the amino acid sequence of SEQ ID NO.4 or SEQ ID NO. 10; (ii) VH CDR2 comprising the amino acid sequence of SEQ ID NO 5 or SEQ ID NO 11; and (iii) a VH CDR3 comprising the amino acid sequence of SEQ ID NO 6 or SEQ ID NO 12. In some cases, the light chain region and the heavy chain region are present in an isolated polypeptide. In other cases, the light chain region and the heavy chain region are present in a single polypeptide. The isolated antibody comprises a heavy chain comprising a constant region of isotype IgG1, IgG2, IgG3, or IgG 4. In other cases, the antibody is an Fv, scFv, Fab, F (ab ')2, or Fab'. The antibody may comprise a non-peptidic synthetic polymer covalently linked, for example when the synthetic polymer is a poly (ethylene glycol) polymer. In some cases, the isolated antibody is fused directly or via a linker to a carrier molecule that is a peptide or protein that helps it cross the blood brain barrier. In some cases, the epitope to which the isolated antibody binds is within amino acids 15-24 of the Tau polypeptide. Isolated antibody humanized light chain framework regions may comprise 1, 2, 3,4, 5,6, 7, 8,9, or 10 amino acid substitutions as set forth in table 3. Isolated antibody humanized heavy chain framework regions comprise 1, 2, 3,4, 5,6, 7, 8,9, 10, 11, or 12 amino acid substitutions as set forth in table 2.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise: a) a light chain region comprising: i) 1, 2 or 3 Complementarity Determining Regions (CDRs) of the IPN001 antibody, wherein the CDRs are as defined by Kabat (see, e.g., table 1 above; and Kabat et al, U.S. Dept. of Health and Human Services, "Sequences of proteins of immunological interest" (1991).

In some embodiments, an antibody suitable for use in the methods of the invention may comprise: a) a light chain region comprising: i) 1, 2 or 3 VL CDRs of the IPN001 antibody; and ii) a humanized light chain framework region; and b) a heavy chain region comprising: i) 1, 2 or 3 VH CDRs of the IPN001 antibody; and ii) a humanized heavy chain framework region; wherein the VH and VL CDRs are as defined by Kabat (see, e.g., Table 1 above; and Kabat et al, U.S. Dept. of Health and Human Services, "Sequences of proteins of immunological interest" (1991)). In some of these embodiments, the anti-Tau antibody comprises a humanized VH and/or VL framework region.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise: a) a light chain region comprising: i) 1, 2 or 3 VL CDRs of the IPN001 antibody; and ii) a humanized light chain framework region; and b) a heavy chain region comprising: i) 1, 2 or 3 VH CDRs of the IPN001 antibody; and ii) a humanized heavy chain framework region; wherein the VH and VL CDRs are as defined by Chothia (see, e.g., Table 1 above; and Chothia et al, J.mol.biol.196:901-917 (1987)).

In some embodiments, an antibody suitable for use in the methods of the invention may comprise: a) a light chain region comprising: i) 1, 2 or 3 VL CDRs of IPN002 antibody; and ii) a humanized light chain framework region; and b) a heavy chain region comprising: i) 1, 2 or 3 VH CDRs of IPN002 antibody; and ii) a humanized heavy chain framework region; wherein the VH and VL CDRs are as defined by Kabat (see, e.g., Table 1 above; and Kabat et al, U.S. Dept. of Health and Human Services, "Sequences of proteins of immunological interest" (1991)).

In some embodiments, an antibody suitable for use in the methods of the invention may comprise: a) a light chain region comprising: i) 1, 2 or 3 VL CDRs of IPN002 antibody; and ii) a humanized light chain framework region; and b) a heavy chain region comprising: i) 1, 2 or 3 VH CDRs of IPN002 antibody; and ii) a humanized heavy chain framework region; wherein the VH and VL CDRs are as defined by Chothia (see, e.g., Table 1 above; and Chothia et al, J.mol.biol.196:901-917 (1987)).

In some embodiments, an antibody suitable for use in the methods of the invention may comprise: a) a light chain region comprising: i) 1, 2 or 3 CDRs selected from SEQ ID NO 1, 2 and 3; and ii) a humanized light chain framework region; and b) a heavy chain region comprising: i) 1, 2 or 3 CDRs selected from SEQ ID NO 4,5 and 6; and ii) a humanized heavy chain framework region.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise: a) a light chain region comprising: i) 1, 2 or 3 CDRs selected from SEQ ID NO 7, 8 and 9; and ii) a humanized light chain framework region; and b) a heavy chain region comprising: i) 1, 2 or 3 CDRs selected from SEQ ID NO 10, 11 and 12; and ii) a humanized heavy chain framework region.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise: a) a light chain region comprising: (i) VL CDR1 comprising the amino acid sequence of SEQ ID NO. 1 or SEQ ID NO. 7; (ii) VL CDR2 comprising the amino acid sequence of SEQ ID NO. 2 or SEQ ID NO. 8; (iii) a VL CDR3 comprising the amino acid sequence of SEQ ID NO. 3 or SEQ ID NO. 9; and (iv) a humanized light chain framework region; and b) a heavy chain region comprising: (i) VH CDR1 comprising the amino acid sequence of SEQ ID NO.4 or SEQ ID NO. 10; (ii) a VH CDR2 comprising the amino acid sequence of SEQ ID NO 5 or SEQ ID NO 11; (iii) VH CDR3 comprising the amino acid sequence of SEQ ID NO 6 or SEQ ID NO 12; and (iv) a humanized heavy chain framework region.

In some embodiments, an antibody suitable for use in the methods of the invention comprises a heavy chain variable region comprising 1, 2, or 3 heavy chain CDRs having an amino acid sequence selected from one or more of SEQ ID NOs 4,5, and 6, and 1, 2, 3, or 4 humanized FR regions. For example, in some embodiments, a suitable antibody comprises a heavy chain variable region comprising, in order from N-terminus to C-terminus: humanized heavy chain FR 1; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 4; humanized heavy chain FR 2; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 5; humanized heavy chain FR 3; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 6; and humanized heavy chain FR 4.

In some embodiments, an antibody suitable for use in the methods of the invention comprises 1, 2, or 3 light chain CDRs having a polypeptide sequence selected from one or more of SEQ ID NOs 1, 2, and 3; and 1, 2, 3 or 4 humanized FR regions. For example, in some embodiments, a suitable antibody comprises a light chain variable region comprising, in order from N-terminus to C-terminus: humanized light chain FR 1; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 1; humanized light chain FR 2; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 2; humanized light chain FR 3; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 3; and humanized light chain FR 4.

In some embodiments, an antibody suitable for use in the methods of the invention comprises 1, 2, or 3 heavy chain CDRs having an amino acid sequence selected from one or more of SEQ ID NOs 10, 11, and 12; and 1, 2, 3 or 4 humanized FR regions. For example, in some embodiments, a suitable antibody comprises a heavy chain variable region comprising, in order from N-terminus to C-terminus: humanized heavy chain FR 1; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 10; humanized heavy chain FR 2; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 11; humanized heavy chain FR 3; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 12; and humanized heavy chain FR 4.

In some embodiments, an antibody suitable for use in the methods of the invention comprises 1, 2, or 3 light chain CDRs having a polypeptide sequence selected from one or more of SEQ ID NOs 7, 8, and 9; and 1, 2, 3 or 4 humanized FR regions. For example, in some embodiments, a suitable antibody comprises a light chain variable region comprising, in order from N-terminus to C-terminus: humanized light chain FR 1; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 7; humanized light chain FR 2; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 8; humanized light chain FR 3; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 9; and humanized light chain FR 4.

The VH and VL amino acid sequences of IPN001 are shown in fig. 11A and 11B. CDRs (as defined by Kabat) are shown in bold text and underlined. The VH and VL amino acid sequences of IPN002 are shown in fig. 12A and 12B. CDRs (as defined by Kabat) are shown in bold text and underlined.

SEQ ID NOS: 1-12 are shown below:

RSSQTILHSNGNTYLE(SEQ ID NO:1);

KVSKRFS(SEQ ID NO:2);

FQGSLVPWA(SEQ ID NO:3);

SYGMS(SEQ ID NO:4);

TISSSGSRTYFPDSVKG(SEQ ID NO:5);

TWDGAMDY(SEQ ID NO:6);

KSSQSIVHSNGNTYLE(SEQ ID NO:7);

KVSNRFS(SEQ ID NO:8);

FQGSLVPWA(SEQ ID NO:9);

KYGMS(SEQ ID NO:10);

TISSSGSRTYYPDSVKG(SEQ ID NO:11);

SWDGAMDY(SEQ ID NO:12)。

in some embodiments, an antibody suitable for use in the methods of the invention may comprise a light chain variable region comprising an amino acid sequence that is 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the sequence shown in fig. 11B and shown in SEQ ID No. 13.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a heavy chain variable region comprising an amino acid sequence that is 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the sequence shown in fig. 11A and shown in SEQ ID No. 14.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a light chain variable region comprising an amino acid sequence that is 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the sequence shown in fig. 12B and shown in SEQ ID No. 15.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a heavy chain variable region comprising an amino acid sequence that is 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the sequence shown in fig. 12A and shown in SEQ ID No. 16.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a heavy chain variable region comprising an amino acid sequence that is 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the sequence set forth in fig. 13 (VH variant 1).

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a heavy chain variable region comprising an amino acid sequence that is 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the sequence shown in fig. 14 (VH variant 2).

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a heavy chain variable region comprising an amino acid sequence that is 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the sequence set forth in fig. 15 (VH variant 3).

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a heavy chain variable region comprising an amino acid sequence that is 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the sequence set forth in fig. 16 (VH variant 4).

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a light chain variable region comprising an amino acid sequence that is 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the sequence set forth in fig. 17 (Vk variant 1).

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a light chain variable region comprising an amino acid sequence that is 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the sequence set forth in fig. 18 (Vk variant 2).

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a light chain variable region comprising an amino acid sequence that is 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the sequence set forth in fig. 19 (Vk variant 3).

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a light chain variable region comprising an amino acid sequence that is 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the sequence set forth in fig. 20 (Vk variant 4).

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a heavy chain variable region comprising 1, 2, 3,4, 5,6, 7, 8,9, 10, 11, or 12 Framework (FR) amino acid substitutions relative to the FR amino acid sequence of the IPN002 parent antibody as shown in table 2.

Table 2: VH variants

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a heavy chain variable region comprising a H → Q substitution at amino acid position 3 in VH FR1 and/or a K → R substitution at amino acid position 19 in VH FR 1.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a heavy chain variable region comprising a T → a substitution at amino acid position 40 in VH FR2 and/or a D → G substitution at amino acid position 42 in VH FR2 and/or a R → G substitution at amino acid position 44 in VH FR 2.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a heavy chain variable region comprising a Q → R substitution at amino acid position 66 in VH FR3 and/or an S → N substitution at amino acid position 83 in VH FR3 and/or an L → S substitution at amino acid position 85 in VH FR3 and/or a K → R substitution at amino acid position 86 in VH FR3 and/or an S → a substitution at amino acid position 87 in VHFR3 and/or an S → a substitution at amino acid position 93 in VH FR 3.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a heavy chain variable region comprising an S → T substitution at amino acid position 108 in VH FR 4.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a VH region comprising, in order from N-terminus to C-terminus: EVX1 LVESGGALVKPGSLRLSCAASGFSFS (SEQ ID NO: 25); VHCDR1 as shown in fig. 12A; WVRQAPGKGLEWVA (SEQ ID NO: 26); a VH CDR2 as shown in fig. 12A; RFTISRDNAKNTLYLQMX2SX3X4X5EDTAMYYCX6I (SEQ ID NO: 27); a VH CDR3 as shown in fig. 12A; WGQGTX7VTVSS (SEQ ID NO:44), wherein X1 is H or Q; x2 is S or N; x3 is S or L; x4 is K or R; x5 is S or A; x6 is S or A; and X7 is S or T.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a light chain variable region comprising 1, 2, 3,4, 5,6, 7, 8,9, or 10 Framework (FR) amino acid substitutions relative to the FR amino acid sequence of the IPN002 parent antibody as shown in table 3.

Table 3: vk variants

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a light chain variable region comprising a L → V substitution at amino acid position 3 in VL FR1 and/or a T → S substitution at amino acid position 7 in VL FR1 and/or a S → T substitution at amino acid position 14 in VLFR1 and/or a D → Q substitution at amino acid position 17 in VL FR1 and/or a Q → P substitution at amino acid position 18 in VL FR 1.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a light chain variable region comprising a K → Q substitution at amino acid position 45 in VL FR2 and/or a V → I substitution at amino acid position 48 in VL FR 2.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a light chain variable region comprising a L → V substitution at amino acid position 83 in VL FR3 and/or a T → V substitution at amino acid position 85 in VL FR 3.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a light chain variable region comprising the L → V substitution at amino acid position 104 in VL FR 4.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a VL region comprising, in order from N-terminus to C-terminus: DVX1 MTQSPLSSVTLGQPASISC (SEQ ID NO: 45); a VL CDR1 as shown in fig. 12B; WYLQKPGQSPQLLX2Y (SEQ ID NO: 46); a VL CDR2 as shown in fig. 12B; GVPRFSGSGSGTDFTTLKISRVEAEDVGX 3YYC (SEQ ID NO: 47); a VL CDR3 as shown in fig. 12B; FGGGTKVEIK (SEQ ID NO: 48); wherein X1 is L or V; x2 is V or I; and X3 is T or V.

In some embodiments, an antibody suitable for use in the methods of the invention comprises:

a) VH variant 1 comprising the amino acid sequence set forth in fig. 13; and Vk variant 1 comprising the amino acid sequence set forth in fig. 17;

b) VH variant 1 comprising the amino acid sequence set forth in fig. 13; and Vk variant 2 comprising the amino acid sequence set forth in fig. 18;

c) VH variant 1 comprising the amino acid sequence set forth in fig. 13; and Vk variant 3 comprising the amino acid sequence set forth in fig. 19;

d) VH variant 1 comprising the amino acid sequence set forth in fig. 13; and Vk variant 4 comprising the amino acid sequence set forth in fig. 20;

e) VH variant 2 comprising the amino acid sequence set forth in fig. 14; and Vk variant 1 comprising the amino acid sequence set forth in fig. 17;

f) VH variant 2 comprising the amino acid sequence set forth in fig. 14; and Vk variant 2 comprising the amino acid sequence set forth in fig. 18;

g) VH variant 2 comprising the amino acid sequence set forth in fig. 14; and Vk variant 3 comprising the amino acid sequence set forth in fig. 19;

h) VH variant 2 comprising the amino acid sequence set forth in fig. 14; and Vk variant 4 comprising the amino acid sequence set forth in fig. 20;

i) VH variant 3 comprising the amino acid sequence shown in figure 15; and Vk variant 1 comprising the amino acid sequence set forth in fig. 17;

j) VH variant 3 comprising the amino acid sequence shown in figure 15; and Vk variant 2 comprising the amino acid sequence set forth in fig. 18;

k) VH variant 3 comprising the amino acid sequence shown in figure 15; and Vk variant 3 comprising the amino acid sequence set forth in fig. 19;

l) VH variant 3 comprising the amino acid sequence shown in figure 15; and Vk variant 4 comprising the amino acid sequence set forth in fig. 20;

m) VH variant 4 comprising the amino acid sequence shown in figure 16; and Vk variant 1 comprising the amino acid sequence set forth in fig. 17;

n) VH variant 4 comprising the amino acid sequence shown in figure 16; and Vk variant 2 comprising the amino acid sequence set forth in fig. 18;

o) VH variant 4 comprising the amino acid sequence shown in fig. 16; and Vk variant 3 comprising the amino acid sequence set forth in fig. 19; or

p) VH variant 4 comprising the amino acid sequence shown in figure 16; and Vk variant 4 comprising the amino acid sequence set forth in fig. 20.

In some embodiments, an antibody suitable for use in the methods of the invention comprises an anti-Tau heavy chain CDR and an anti-Tau light chain CDR in a single polypeptide chain, e.g., in some embodiments, a suitable antibody is an scFv. In some embodiments, a suitable antibody comprises, in order from N-terminus to C-terminus: a first amino acid sequence of about 5 amino acids to about 25 amino acids in length; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 1; a second amino acid sequence of about 5 amino acids to about 25 amino acids in length; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 2; a third amino acid sequence of about 5 amino acids to about 25 amino acids in length; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 3; a fourth amino acid sequence of about 5 amino acids to about 25 amino acids in length; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 4; a fifth amino acid sequence of about 5 amino acids to about 25 amino acids in length; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 5; a sixth amino acid sequence of about 5 amino acids to about 25 amino acids in length; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 6; and a seventh amino acid sequence from about 5 amino acids to about 25 amino acids in length.

In some embodiments, an antibody suitable for use in the methods of the invention comprises, in order from N-terminus to C-terminus: a light chain FR1 region; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 1; a light chain FR2 region; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 2; a light chain FR3 region; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 3; optionally a light chain FR4 region; a joint region; optionally a heavy chain FR1 region; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 4; a heavy chain FR2 region; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 5; a heavy chain FR3 region; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 6; and a heavy chain FR4 region. In some of these embodiments, one or more of the FR regions are humanized FR regions. In some of these embodiments, each of the FR regions is a humanized FR region. The linker region may be about 5 amino acids to about 50 amino acids in length, for example about 5aa to about 10aa, about 10aa to about 15aa, about 15aa to about 20aa, about 20aa to about 25aa, about 25aa to about 30aa, about 30aa to about 35aa, about 35aa to about 40aa, about 40aa to about 45aa, or about 45aa to about 50aa in length.

In some embodiments, an antibody suitable for use in the methods of the invention comprises, in order from N-terminus to C-terminus: a heavy chain FR1 region; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 4; a heavy chain FR2 region; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 5; a heavy chain FR3 region; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 6; optionally a heavy chain FR4 region; a joint region; optionally a light chain FR1 region; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 1; a light chain FR2 region; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 2; a light chain FR3 region; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 3; and a light chain FR4 region. In some of these embodiments, one or more of the FR regions are humanized FR regions. In some of these embodiments, each of the FR regions is a humanized FR region. The linker region may be about 5 amino acids to about 50 amino acids in length, for example about 5aa to about 10aa, about 10aa to about 15aa, about 15aa to about 20aa, about 20aa to about 25aa, about 25aa to about 30aa, about 30aa to about 35aa, about 35aa to about 40aa, about 40aa to about 45aa, or about 45aa to about 50aa in length.

In some embodiments, an antibody suitable for use in the methods of the invention comprises, in order from N-terminus to C-terminus: a light chain FR1 region; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 7; a light chain FR2 region; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 8; a light chain FR3 region; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 9; optionally a light chain FR4 region; a joint region; optionally a heavy chain FR1 region; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 10; a heavy chain FR2 region; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 11; a heavy chain FR3 region; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 12; and a heavy chain FR4 region. In some of these embodiments, one or more of the FR regions are humanized FR regions. In some of these embodiments, each of the FR regions is a humanized FR region. The linker region may be about 5 amino acids to about 50 amino acids in length, for example about 5aa to about 10aa, about 10aa to about 15aa, about 15aa to about 20aa, about 20aa to about 25aa, about 25aa to about 30aa, about 30aa to about 35aa, about 35aa to about 40aa, about 40aa to about 45aa, or about 45aa to about 50aa in length.

In some embodiments, an antibody suitable for use in the methods of the invention comprises, in order from N-terminus to C-terminus: a heavy chain FR1 region; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 10; a heavy chain FR2 region; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 11; a heavy chain FR3 region; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 12; optionally a heavy chain FR4 region; a joint region; optionally a light chain FR1 region; CDR1 comprising the amino acid sequence set forth in SEQ ID NO. 7; a light chain FR2 region; CDR2 comprising the amino acid sequence set forth in SEQ ID NO. 8; a light chain FR3 region; CDR3 comprising the amino acid sequence set forth in SEQ ID NO. 9; and a light chain FR4 region. In some of these embodiments, one or more of the FR regions are humanized FR regions. In some of these embodiments, each of the FR regions is a humanized FR region. The linker region may be about 5 amino acids to about 50 amino acids in length, for example about 5aa to about 10aa, about 10aa to about 15aa, about 15aa to about 20aa, about 20aa to about 25aa, about 25aa to about 30aa, about 30aa to about 35aa, about 35aa to about 40aa, about 40aa to about 45aa, or about 45aa to about 50aa in length.

Suitable linkers for use in antibodies include "flexible linkers". The linker molecules, if present, are typically long enough to allow some flexible movement between the linking regions. The linker molecule is typically about 6 to 50 atoms in length. The linker molecule can also be, for example, an aryl acetylene, a glycol oligomer containing 2-10 monomer units, a diamine, a diacid, an amino acid, or a combination thereof. Other linker molecules capable of binding to the polypeptide may also be used according to the invention.

Suitable linkers can be readily selected and can be any of a variety of suitable lengths, such as 1 amino acid (e.g., Gly) to 20 amino acids, 2 amino acids to 15 amino acids, 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids, or 7 amino acids to 8 amino acids. And may be 1, 2, 3,4, 5,6, or 7 amino acids.

Exemplary flexible linkers include glycine polymer (G) n, glycine-serine polymers (includingSuch as (GS) n, (GSGGS) n (SEQ ID NO:49) and (GGGS) n (SEQ ID NO:50), where n is an integer of at least 1), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers known in the art. Of interest are glycine and glycine-serine polymers, both of which are relatively bulky and therefore can act as neutral chains between the components. Of particular interest are glycine polymers, since glycine is even significantly more proximal than alanineSpace, and is much less restricted than residues with longer side chains (see Scheraga, Rev. computational chem.11173-142 (1992)). Exemplary flexible linkers include, but are not limited to, GGSG (SEQ ID NO:51), GGSGG (SEQ ID NO:52), GSGSGSG (SEQ ID NO:53), GSGGG (SEQ ID NO:54), GGGSG (SEQ ID NO:55), GSSSG (SEQ ID NO:56), and the like. The ordinarily skilled artisan will appreciate that the design of the peptide conjugated to any of the elements described above may include a linker that is flexible in whole or in part, such that the linker can include a flexible linker as well as one or more moieties that impart a less flexible structure.

In some embodiments, an antibody suitable for use in the methods of the invention is an antibody fragment, Fv, scFv, Fab, F (ab ')2, or Fab'. Accordingly, the invention provides an isolated antibody, wherein the antibody is an Fv, scFv, Fab, F (ab ')2, or Fab', and wherein the antibody competes for binding to an epitope within an N-terminal region of a Tau polypeptide with another antibody comprising: a) a light chain region comprising: (i) VL CDR1 comprising the amino acid sequence of SEQ ID NO. 1 or SEQ ID NO. 7; (ii) VL CDR2 comprising the amino acid sequence of SEQ ID NO. 2 or SEQ ID NO. 8; and (iii) a VL CDR3 comprising the amino acid sequence of SEQ ID NO. 3 or SEQ ID NO. 9; and b) a heavy chain region comprising: (i) VH CDR1 comprising the amino acid sequence of SEQ ID NO.4 or SEQ ID NO. 10; (ii) VH CDR2 comprising the amino acid sequence of SEQ ID NO 5 or SEQ ID NO 11; and (iii) a VH CDR3 comprising the amino acid sequence of SEQ ID NO 6 or SEQ ID NO 12. In some of these embodiments, the isolated antibody comprises 1, 2, 3, or 4 humanized VL framework regions, as described above. In some of these embodiments, the isolated antibody comprises 1, 2, 3, or 4 humanized VH framework regions, as described above.

In some embodiments, an antibody suitable for use in the methods of the invention is an scFv antibody. In some embodiments, the anti-Tau antibody of the invention comprises a scFv multimer. For example, in some embodiments, a suitable antibody is a scFv dimer (e.g., comprising two scFv in series (scFv2)), a scFv trimer (e.g., comprising three scFv in series (scFv3)), a scFv tetramer (e.g., comprising four scFv in series (scFv4)), or a multimer of more than four scFv (e.g., in series). The scFv monomers may be connected in tandem by a linker that is from about 2 amino acids to about 10 amino acids (aa) in length, for example, 2aa, 3aa, 4aa, 5aa, 6aa, 7aa, 8aa, 9aa, or 10aa in length. Suitable linkers include, for example, (Gly) x, where x is an integer from 2 to 10. Other suitable linkers are those discussed above. In some embodiments, each of the scFv monomers in the scFv multimer is humanized, as described above.

In some embodiments, antibodies suitable for use in the methods of the invention comprise a constant region (e.g., an Fc region) of an immunoglobulin. The Fc region (if present) may be a human Fc region. If present, the antibody may contain both light and heavy chain constant regions. Suitable heavy chain constant regions include the CH1, hinge, CH2, CH3, and CH4 regions. Antibodies described herein include antibodies with constant regions of all types, including IgM, IgG, IgD, IgA, and IgE, as well as any isotype, including IgG1, IgG2, IgG3, and IgG 4. An example of a suitable heavy chain Fc region is the human isotype IgG1 Fc. In some cases, the heavy chain region is of isotype IgG 4. In some of these embodiments, the hinge region comprises an S241P substitution. See, e.g., Angal et al (1993) mol.Immunol.30: 105. The light chain constant region may be lambda or kappa. Suitable antibodies (e.g., humanized antibodies) can comprise sequences from more than one class or isotype. Antibodies can be expressed as tetramers containing two light chains and two heavy chains, as isolated heavy chains, light chains, as Fab, Fab 'F (ab')2, and Fv, or as single chain antibodies, wherein the heavy and light chain variable domains are linked by a spacer.

In some embodiments, an antibody suitable for use in the methods of the invention comprises a human light chain constant region and a human heavy chain constant region, and wherein the isolated antibody competes for binding to an epitope within an N-terminal region of a Tau polypeptide with another antibody comprising: a) a light chain region comprising: (i) VL CDR1 comprising the amino acid sequence of SEQ ID NO. 1 or SEQ ID NO. 7; (ii) VL CDR2 comprising the amino acid sequence of SEQ ID NO. 2 or SEQ ID NO. 8; and (iii) a VL CDR3 comprising the amino acid sequence of SEQ ID NO. 3 or SEQ ID NO. 9; and b) a heavy chain region comprising: (i) VH CDR1 comprising the amino acid sequence of SEQ ID NO.4 or SEQ ID NO. 10; (ii) a VH CDR2 comprising the amino acid sequence of SEQ ID NO 5 or SEQ ID NO 11; and (iii) a VH CDR3 comprising the amino acid sequence of SEQ ID NO 6 or SEQ ID NO 12. In some of these embodiments, the isolated antibody comprises 1, 2, 3, or 4 humanized VL framework regions, as described above. In some of these embodiments, the isolated antibody comprises 1, 2, 3, or 4 humanized VH framework regions, as described above.

In some embodiments, an antibody suitable for use in the methods of the invention may comprise a free thiol (-SH) group at the carboxy terminus, where the free thiol group can be used to attach the antibody to a second polypeptide (e.g., another antibody, including a suitable antibody), a scaffold, a carrier, and the like.

In some embodiments, antibodies suitable for use in the methods of the invention comprise one or more non-naturally occurring amino acids. In some embodiments, the non-naturally encoded amino acid comprises a carbonyl group, an acetyl group, an aminooxy (aminooxy) group, a hydrazine group, a hydrazide group, a semicarbazide group, an azide group, or an alkynyl group. Suitable non-naturally occurring amino acids are described, for example, in U.S. patent No.7,632,924. The introduction of the non-naturally occurring amino acid can provide for attachment to a polymer, a second polypeptide, a scaffold, and the like. For example, a suitable antibody attached to a water-soluble polymer can be made by reacting a water-soluble polymer comprising a carbonyl group (e.g., PEG) with an antibody, wherein the antibody comprises a non-naturally encoded amino acid comprising an aminooxy, hydrazine, hydrazide or semicarbazide group. As another example, a suitable antibody attached to a water-soluble polymer can be made by reacting a suitable antibody comprising an alkyne-containing amino acid with a water-soluble polymer (e.g., PEG) comprising an azide-containing moiety; in some embodiments, the azide or alkynyl group is linked to the PEG molecule through an amide linkage. By "non-naturally encoded amino acid" is meant an amino acid that is not the 20 common amino acids or pyrrolysine or selenocysteine. Other terms that may be used interchangeably with the term "non-naturally encoded amino acid" are "non-natural amino acid", "unnatural amino acid", "non-naturally occurring amino acid", and various hyphenated and non-hyphenated versions thereof. The term "non-naturally encoded amino acid" also includes, but is not limited to, amino acids that are formed by modification (e.g., post-translational modification) of naturally encoded amino acids (including, but not limited to, the 20 common amino acids or pyrrolysine and selenocysteine), but which are not themselves naturally integrated into a growing polypeptide chain by the translational complex. Examples of such non-naturally occurring amino acids include, but are not limited to, N-acetylglucosaminyl-L-serine, N-acetylglucosaminyl-L-threonine, and O-phosphotyrosine.

In some embodiments, an antibody suitable for use in the methods of the invention is linked (e.g., covalently linked) to a polymer (e.g., a polymer other than a polypeptide). Suitable polymers include, for example, biocompatible polymers, and water-soluble biocompatible polymers. Suitable polymers include synthetic polymers and naturally occurring polymers. Suitable polymers include, for example, substituted or unsubstituted linear or branched polyalkylene, polyalkenylene or polyoxyalkylene polymers or branched or unbranched polysaccharides, for example homopolysaccharides or heteropolysaccharides. Suitable polymers include, for example, ethylene vinyl alcohol copolymers (typically commensurate with the common name EVOH or EVAL); poly (butyl methacrylate); poly (hydroxyvalerate); poly (L-lactic acid); polycaprolactone; poly (lactide-co-glycolide); poly (hydroxybutyrate); poly (hydroxybutyrate-co-valerate); polydioxanone (polydioxanone); a polyorthoester; a polyanhydride; poly (glycolic acid); poly (D, L-lactic acid); poly (glycolic acid-co-trimethylene carbonate); polyphosphates (polyphosphasters); polyphosphate urethane; poly (amino acids); cyanoacrylate esters; poly (trimethylene carbonate); poly (imino carbonates); copoly (ether-esters) (e.g., poly (ethylene oxide) -poly (lactic acid) (PEO/PLA) copolymer); polyalkylene oxalates; polyphosphazenes; biomolecules such as fibrin, fibrinogen, cellulose, starch, collagen and hyaluronic acid; polyurethanes; silicones; polyesters; polyolefins; polyisobutylene and ethylene-alpha olefin copolymers (ethylene-alphaolephin copolymers); acrylic polymers and copolymers; vinyl halide polymers and copolymers, such as polyvinyl chloride; polyvinyl ethers such as polyvinyl methyl ether; polyvinylidene halides, such as polyvinylidene fluoride and polyvinylidene chloride; polyacrylonitrile; polyvinyl ketones; polyvinyl aromatics such as polystyrene; polyvinyl esters such as polyvinyl acetate; copolymers between vinyl monomers or with olefins, such as ethylene-methyl methacrylate (ethyl methacrylate) copolymers, acrylonitrile-styrene copolymers, ABS resins, and ethylene-vinyl acetate copolymers; polyamides, such as Nylon 66(Nylon 66) and polycaprolactam; an alkyd resin; polycarbonates; polyoxymethylene; polyimides; polyethers; an epoxy resin; polyurethanes; artificial silk; rayon-triacetate; cellulose; cellulose acetate; cellulose butyrate; cellulose acetate butyrate; cellophane; cellulose nitrate; cellulose propionate; cellulose ethers; amorphous Teflon (Teflon); poly (ethylene glycol); and carboxymethyl cellulose.

Suitable synthetic polymers include unsubstituted and substituted linear or branched poly (ethylene glycol), poly (propylene glycol), poly (vinyl alcohol), and derivatives thereof, for example substituted poly (ethylene glycols) such as methoxy poly (ethylene glycol), and derivatives thereof. Suitable naturally occurring polymers include, for example, albumin, amylose, dextran, glycogen, and derivatives thereof.

Suitable polymers may have an average molecular weight in the range 500Da to 50000Da, for example 5000Da to 40000Da, or 25000 to 40000 Da. For example, in some embodiments, when a suitable antibody comprises a poly (ethylene glycol) (PEG) or methoxy poly (ethylene glycol) polymer, the PEG or methoxy poly (ethylene glycol) polymer can have a molecular weight ranging from about 0.5 kilodaltons (kDa) to 1kDa, about 1kDa to 5kDa, 5kDa to 10kDa, 10kDa to 25kDa, 25kDa to 40kDa, or 40kDa to 60 kDa.

As described above, in some embodiments, a suitable antibody is covalently attached to the PEG polymer. In some embodiments, the scFv multimer is covalently attached to the PEG polymer. See, e.g., Albrecht et al (2006) j.immunol.methods 310: 100. Methods and reagents suitable for pegylation of proteins are well known in the art and can be found, for example, in U.S. patent No.5,849,860. PEG suitable for conjugation to proteins is typically soluble in water at room temperature and has the general formula R (O-CH2-CH2) nO-R, where R is hydrogen or a protecting group such as an alkyl or alkanol group, and where n is an integer from 1 to 1000. Wherein R is a protecting group, typically having from 1 to 8 carbons.

The PEG conjugated to the antibody may be linear. PEG conjugated to the protein may also be branched. Branched PEG derivatives such as those described in U.S. Pat. No.5,643,575, "star-PEG's" and multi-armed PEG's such as those described in Shearwater Polymers, Inc. catalog "polyethylene glycol derivatives 1997-. Star PEGs are described in the art, including, for example, in U.S. patent No.6,046,305.

In some embodiments, antibodies suitable for use in the methods of the invention may be glycosylated, for example suitable antibodies may comprise a covalently linked carbohydrate or polysaccharide moiety. Glycosylation of antibodies is usually N-linked or O-linked. N-linked means that the sugar moiety is attached to the side chain of an asparagine residue. The tripeptide sequences asparagine-X-serine and asparagine-X-threonine (where X is any amino acid except proline) are recognition sequences for enzymatic attachment of the carbohydrate module to an asparagine side chain. Thus, the presence of any of these tripeptide sequences in a polypeptide creates potential glycosylation sites. O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.

The addition of glycosylation sites to the antibody is readily accomplished by altering the amino acid sequence (such that it contains one or more of the tripeptide sequences described above) (for N-linked glycosylation sites). Modifications (for O-linked glycosylation sites) may also be made by adding, or substituting, one or more serine or threonine residues to the original antibody sequence. Similarly, glycosylation sites can be removed by amino acid modifications within the glycosylation site of the antigen.

In some embodiments, suitable antibodies will comprise "radiopaque" markers, such as markers that can be readily visualized, for example, with x-rays. Radiopaque materials are well known to those skilled in the art. The most common radiopaque materials include iodide, bromide, or barium salts. Other radiopaque materials are also known and include, but are not limited to, organobismuth derivatives (see, e.g., U.S. patent No.5,939,045), radiopaque polyureas (see, e.g., U.S. patent No.5,346,981), organobismuth compositions (see, e.g., U.S. patent No.5,256,334), radiopaque barium polymer complexes (see, e.g., U.S. patent No.4,866,132), and the like.

Suitable antibodies can be covalently linked to a second moiety (e.g., a lipid, a polypeptide other than an antibody, a synthetic polymer, a sugar, etc.) using, for example, glutaraldehyde, homobifunctional (homobifunctional) cross-linkers, or heterobifunctional cross-linkers. Glutaraldehyde crosslinks the polypeptide through its amino moiety. Homobifunctional crosslinkers (e.g., homobifunctional imidates, homobifunctional N-hydroxysuccinimide (NHS) esters, or homobifunctional mercapto-reactive crosslinkers) contain two or more identical reactive modules and can be used in a one-step reaction procedure in which the crosslinker is added to a solution containing a mixture of polypeptides to be linked. Homobifunctional NHS esters and imidates crosslink the amine containing polypeptide. At slightly alkaline pH, the imidate reacts only with primary amines to form iminamides (imidoamides), while the overall charge of the crosslinked polypeptide is unaffected. Homobifunctional thiol-reactive crosslinkers include bismaleimide hexane (BMH), 1, 5-difluoro-2, 4-dinitrobenzene (DFDNB), and 1, 4-bis- (3',2' -pyridyldithio) propylaminobutane (1,4-di- (3',2' -pyridyldithiohi) propioamido butane, DPDPDPPB).

The heterobifunctional crosslinking agent has two or more different reactive modules (e.g., an amine-reactive module and a thiol-reactive module) and is crosslinked with one of the polypeptides through the amine or thiol-reactive module, and then reacted with another polypeptide through the unreacted module. Multiple heterobifunctional haloacetyl (haloacetyl) crosslinkers are useful, as are pyridyl disulfide crosslinkers. Carbodiimides are typical examples of heterobifunctional crosslinking reagents used to couple carboxyl groups to amines, which form amide bonds.

In some embodiments, a suitable antibody comprises a detectable label. Suitable detectable labels include any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Suitable include, but are not limited to, magnetic beads (e.g., Dynabeads)TM) Fluorescent dyes (e.g., fluorescein isothiocyanate, Texas Red, rhodamine, Green fluorescent protein, Red fluorescent protein, yellow fluorescent protein, etc.), radioactive labels (e.g., fluorescent protein, etc.)3H、125I、35S、14C. Or32P), enzymes (e.g., horseradish peroxidase, alkaline phosphatase, luciferase, and other labels commonly used in enzyme-linked immunosorbent assays (ELISA), and colorimetric labels such as colloidal gold or colored glass or plastic (e.g., polystyrene, polypropylene, latex, etc.) beads.

In some embodiments, a suitable antibody comprises a contrast agent or a radioisotope, wherein the contrast agent or radioisotope is suitable for use in imaging, such as an imaging procedure performed on a human. Non-limiting examples of labels include radioisotopes such as 1231I (iodine), 18F (fluorine), 99Tc (technetium), 111In (indium), and 67Ga (gallium), and contrast agents such as gadolinium (Gd), dysprosium, and iron. Radioactive Gd isotopes (153Gd) are also useful and suitable for imaging procedures in non-human mammals. Suitable antibodies can be labeled using standard techniques. For example, a suitable antibody may be iodinated with chloramine-T or 1,3,4, 6-tetrachloro-3 α,6 α -diphenylglycoluril. For fluorination, fluorine is added to the anti-Tau antibody during synthesis by a fluoride ion displacement reaction. Protein synthesis with such radioisotopes is reviewed in Muller-Gartner, H., TIBTech.,16:122-130(1998) and Saji, H., Crit. Rev. Ther. drug Carrier Syst.,16(2):209-244 (1999). Suitable antibodies may also be labeled with contrast agents by standard techniques. For example, a suitable antibody may be labelled with Gd by conjugating a low molecular Gd chelate to the antibody, for example Gd diethylenetriaminepentaacetic acid (GdDTPA) or Gd tetraazacyclododecane tetraacetic acid (Gd tetraazacyclododecaneetacetic, GdDOTA). See Caravan et al, chem.Rev.99:2293-2352(1999) and Lauffer et al, J.Magn.Reson.imaging,3:11-16 (1985). Suitable antibodies may be labeled with Gd by, for example, conjugating a polylysine-Gd chelate to the antibody. See, e.g., Curtet et al, invest, radio, 33(10):752-761 (1998). Alternatively, suitable antibodies may be labeled with Gd by incubating paramagnetic polymeric liposomes (which include Gd chelator lipids) with avidin and a biotinylated antibody. See, e.g., Sipkins et al, Nature Med.,4: 623-.

Suitable fluorescent proteins that can be linked to suitable antibodies include, but are not limited to, green fluorescent protein from Aequoria victoria or mutants or derivatives thereof, such as those described in U.S. patent No.6,066,476; 6,020,192, respectively; 5,985,577, respectively; 5,976,796, respectively; 5,968,750, respectively; 5,968,738; 5,958,713, respectively; 5,919,445, respectively; 5,874,304; e.g., enhanced GFP, many such GFP commercially available, e.g., from Clontech, inc; a red fluorescent protein; a yellow fluorescent protein; any of a variety of fluorescent and colored proteins from the species Corallium japonicum (Anthozoan), such as described in Matz et al (1999) Nature Biotechnol.17: 969-973; and so on.

In some embodiments, the antibody is linked (e.g., covalently or non-covalently) to a fusion partner, e.g., a ligand; an epitope tag; a peptide; proteins other than antibodies; and so on. Suitable fusion partners include those that confer enhanced in vivo stability (e.g., enhanced serum half-life); provide a purification facility (e.g., (His) n, e.g., 6His (SEQ ID NO:57), etc.); providing for secretion of the fusion protein from the cell; providing epitope tags (e.g., GST, hemagglutinin (HA; e.g., YPYDVPDYA; SEQ ID NO:58), FLAG (e.g., DYKDDDDK; SEQ ID NO:59), c-myc (e.g., EQKLISEEDL; SEQ ID NO:60), etc.); providing a detectable signal (e.g., an enzyme that produces a detectable product (e.g., β -galactosidase, luciferase) or a protein that is itself detectable, e.g., green fluorescent protein, red fluorescent protein, yellow fluorescent protein, etc.); peptides and polypeptides that multimerize (e.g., a multimerization domain such as the Fc portion of an immunoglobulin), and the like, are provided.

The fusion may also comprise an affinity domain comprising a peptide sequence (e.g., one immobilized on a solid support) that is capable of interacting with a binding partner, which is useful for identification and purification. When fused to a protein, a single amino acid (e.g., histidine) in series can be used for one-step purification of the fusion protein by high affinity binding to a resin column, such as nickel sepharose. Exemplary affinity domains include His5(HHHHH) (SEQ ID NO:61), HisX6 (HHHHHHH) (SEQ ID NO:57), C-myc (EQKLISEEDL) (SEQ ID NO:60), flag (DYKDDDDK) (SEQ ID NO:59), Streptag (WSHPHPQFEK) (SEQ ID NO:62), prothrombin, such as an HA tag (YPYDVPDYA; SEQ ID NO:58), glutathione-S-transferase (GST), thioredoxin, cellulose binding domain, RYIRS (SEQ ID NO:63), Phe-His-His-Thr (SEQ ID NO:64), chitin binding domain, S-peptide, T7 peptide, SH2 domain, C-terminal RNA tag, WEAAAREACCRECCARA (SEQ ID NO:65), metal binding domains, such as zinc binding domains or calcium binding domains such as those of calcium binding proteins, e.g. calmodulin, calpain, C-B, myosin, light chain myosin, C-chain myosin, C-B-myosin, C-C, C, Recoverin, S-regulatory protein, cone protein, VILIP, calcineurin, hippocampal calcin (hippocalcin), frequenin (frequenin), kallikrein (caltractin), calpain large subunit, S100 protein, parvalbumin, calbindin D9K, calbindin D28K, and calretinin, intein (inteins), biotin, streptavidin, MyoD, leucine zipper sequence, and maltose binding protein.

In some embodiments, a suitable antibody is fused to a polypeptide that binds to an endogenous Blood Brain Barrier (BBB) receptor. Linking a suitable antibody to a polypeptide that binds to an endogenous BBB receptor facilitates crossing the BBB, for example in a method of treatment of the invention involving administration of a suitable antibody to an individual in need thereof (see below). Suitable polypeptides that bind to an endogenous BBB receptor include antibodies, e.g., monoclonal antibodies, or antigen-binding fragments thereof, that specifically bind to an endogenous BBB receptor. Suitable endogenous BBB receptors include, but are not limited to, insulin receptor, transferrin receptor, leptin receptor, lipoprotein receptor, and insulin-like growth factor receptor. See, for example, U.S. patent publication No. 2009/0156498.

For example, a suitable anti-Tau antibody may be a bispecific antibody comprising a first antigen-binding portion that specifically binds to an epitope within a Tau polypeptide; and a second antigen-binding moiety that binds to an endogenous BBB receptor. For example, in some instances, a suitable anti-Tau antibody is a bispecific antibody comprising a first antigen-binding portion that specifically binds to an epitope within a Tau polypeptide; and a second antigen-binding moiety that binds to transferrin receptor.

For example, a suitable anti-Tau antibody can be fused to a peptide that facilitates crossing the BBB, the peptide being about 15 amino acids to about 25 amino acids in length and comprising an amino acid sequence having at least about 85% amino acid sequence identity to one of the following peptides: angiopep-1 (TFFYGGCRGKRNNFKTEEY; SEQ ID NO: 66); angiopep-2 (TFFYGGSRGKRNNFKTEEY; SEQ ID NO: 67); cys-Angiopep-2 (CTFFYGGSRGKRNNFKTEEY; SEQ ID NO: 68); angiopep-2-cys (TFFYGGSRGKRNNFKTEEYC; SEQ ID NO: 69); and an aprotinin fragment (TFVYGGCRAKRNNFKS; SEQ ID NO: 70). See U.S. patent publication nos. 2011/0288011; and 2009/0016959. Peptides that facilitate crossing the BBB can be fused to the N-terminus of the anti-Tau light chain region, the C-terminus of the anti-Tau light chain region, the N-terminus of the anti-Tau heavy chain region, the C-terminus of the anti-Tau heavy chain region, the N-terminus of the anti-Tau single chain antibody, the C-terminus of the anti-Tau single chain antibody, and the like.

In some embodiments, suitable antibodies comprise polyamine modifications. Polyamine modification of suitable antibodies enhances the penetration of the modified antibody at the BBB. Suitable antibodies can be modified with naturally occurring or synthetic polyamines. See, for example, U.S. patent No.5,670,477. Useful naturally occurring polyamines include putrescine, spermidine, spermine, 1, 3-diaminopropane, norspermidine (norspermidine), symmetric homopspermidine, pyrenamine, pyrospermine, nitrohalopentamine, and canavanine. Putrescine, spermidine, and spermine are particularly useful. Synthetic polyamines consist of the empirical formula CXHYNZ, which may be cyclic or acyclic, branched or unbranched, and further comprises a hydrocarbon chain of 3 to 12 carbon atoms of a 1-6NR or n (R)2 module, wherein R is H, (C1-C4) alkyl, phenyl, or benzyl. The polyamine can be attached to the antibody using any standard crosslinking method.

In some embodiments, a suitable antibody is modified to comprise a sugar moiety, wherein the sugar moiety may be covalently linked to the antibody. In some embodiments, a suitable antibody is modified to comprise a lipid moiety, wherein the lipid moiety may be covalently linked to the antibody. Suitable lipid moieties include: for example, N-fatty acyl groups such as N-lauroyl, N-oleoyl, and the like; aliphatic amines such as dodecylamine, oleylamine, etc.; C3-C16 long chain fatty lipids; and so on. See, e.g., U.S. patent No.6,638,513). In some embodiments, suitable antibodies are incorporated into liposomes.

Combination therapy

The anti-Tau antibody can be administered to an individual in need thereof alone (e.g., monotherapy) or in combination therapy with one or more additional therapeutic agents. For example, an anti-Tau antibody can be administered in combination therapy with one or more additional therapeutic agents for the treatment of stroke, or for the treatment of TBI.

The term "in combination with … …" as used herein means a use wherein, for example, a first compound is administered throughout the administration of a second compound; wherein the period of time of administration of the first compound overlaps with the administration of the second compound, for example wherein administration of the first compound is initiated before administration of the second compound and administration of the first compound is terminated before administration of the second compound is terminated; wherein administration of the second compound is initiated prior to administration of the first compound and administration of the second compound is terminated prior to termination of administration of the first compound; wherein administration of the first compound is initiated before administration of the second compound is initiated and administration of the second compound is terminated before administration of the first compound is terminated; wherein administration of the second compound is initiated before administration of the first compound is initiated and administration of the first compound is terminated before administration of the second compound is terminated. Likewise, "combination" may also refer to a regimen involving the administration of two or more compounds. As used herein, "in combination with … …" may also refer to the administration of two or more compounds, which may be administered in the same or different formulations, by the same or different routes, and in the same or different dosage form types.

The individual to be treated

Individuals suitable for treatment with an anti-Tau antibody include individuals who have been diagnosed with a tauopathy (e.g., an acute tauopathy); individuals at greater risk of developing tauopathies than the general population (e.g., individuals with genetic susceptibility to developing tauopathies); military personnel; and so on. In some cases, the subject is a human and is aged from less than 10 to 10 years old; from 10 years to about 15 years old; from about 15 years to about 20 years old; or from about 20 to about 30 years of age. In some cases, the individual is an adult. In some cases, the adult age is from about 20 to about 30 years; 30 years old or older; 40 years or older, 50 years or older, 60 years or older, 70 years or older, or 80 years or older. For example, the adult may be 40 to 50 years old, 50 to 60 years old, 60 to 70 years old, or over 70 years old. In some cases, the individual is an individual having TBI. In some cases, the subject is a subject having a stroke.

Formulation

In the methods of the invention, the anti-Tau antibody may be administered to the host in any convenient manner that results in the desired therapeutic or diagnostic effect. Thus, the agents can be incorporated into a variety of formulations for therapeutic administration. More specifically, the anti-Tau antibody may be formulated in a pharmaceutical composition by combination with a suitable, pharmaceutically acceptable carrier or diluent, and may be formulated in preparations in solid, semi-solid, liquid or gaseous form, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants and aerosols.

In a pharmaceutical dosage form, the anti-Tau antibody may be administered in the form of a pharmaceutically acceptable salt thereof, or it may also be administered alone or in appropriate combination and combination with other pharmaceutically active compounds. The following methods and excipients are exemplary only, and in no way limiting.

For oral formulations, the anti-Tau antibody may be used alone or in combination with suitable additives, to make tablets, powders, granules or capsules, for example with conventional additives such as lactose, mannose, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia (acacia), corn starch or gelatin; with disintegrating agents, such as corn starch, potato starch or sodium carboxymethyl cellulose; with lubricating agents, such as talc or magnesium stearate; and, if desired, diluents, buffers, wetting agents, preservatives, and flavoring agents.

The anti-Tau antibody can be formulated in a preparation for injection by dissolving, suspending or emulsifying it in an aqueous or non-aqueous solvent such as a vegetable oil or other similar oil, a synthetic fatty acid glyceride, a higher fatty acid ester or an ester of propylene glycol; and if desired, with conventional additives such as solubilizers, isotonicity agents, suspending agents, emulsifiers, stabilizers and preservatives.

The pharmaceutical composition comprising the anti-Tau antibody is prepared by mixing the antibody with the desired purity with optional physiologically acceptable carriers, excipients, stabilizers, surfactants, buffers and/or tonicity agents. Acceptable carriers, excipients, and/or stabilizers are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid, glutathione, cysteine, methionine and citric acid; preservatives (e.g., ethanol, benzyl alcohol, phenol, m-cresol, p-chloro-m-cresol, methyl or propyl p-hydroxybenzoate (parabens), benzalkonium chloride (benzalkonium chloride), and combinations thereof); amino acids such as arginine, glycine, ornithine, lysine, histidine, glutamic acid, aspartic acid, isoleucine, leucine, alanine, phenylalanine, tyrosine, tryptophan, methionine, serine, proline and combinations thereof; monosaccharides, disaccharides, and other carbohydrates; low molecular weight (less than about 10 residues) polypeptides; 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-formylglucosamine, galactosamine, and neuraminic acid; and/or a non-ionic surfactant such as tween, Brij Pluronics, Triton-X, or polyethylene glycol (PEG).

The pharmaceutical composition may be in liquid form, lyophilized form, or reconstituted liquid form from a lyophilized form, wherein the lyophilized preparation is reconstituted with a sterile solution prior to administration. Standard procedures for reconstitution of lyophilized compositions are replenishing a volume of purified water (usually an equal volume to that removed during lyophilization); however, solutions containing antibacterial agents may be used to produce pharmaceutical compositions for parenteral administration; see also Chen (1992) Drug Dev Ind Pharm 18,1311-54.

Exemplary antibody concentrations in pharmaceutical compositions can range from about 1mg/mL to about 200mg/mL or from about 50mg/mL to about 200mg/mL, or from about 150mg/mL to about 200 mg/mL.

Aqueous formulations of the antibodies can be prepared in a pH buffered solution, e.g., at a pH in the range of about 4.0 to about 7.0, or about 5.0 to about 6.0, or about 5.5. Examples of buffers suitable for a pH in this range include phosphate, histidine, citrate, succinate, acetate buffers and other organic acid buffers. The buffer concentration may be from about 1mM to about 100mM, or from about 5mM to about 50mM, depending on, for example, the buffer and the desired tonicity of the solution.

Tonicity agents may be included in the antibody formulation to adjust the tonicity of the formulation. Exemplary tonicity agents include sodium chloride, potassium chloride, glycerin and any member of the amino acid group, sugars and combinations thereof. In some embodiments, the aqueous formulation is isotonic, although hypertonic or hypotonic solutions may be suitable. The term "isotonic" means that the solution has the same tonicity as some other solution to which it is compared, such as saline or serum. Tonicity agents may be used in amounts of about 5mM to about 350mM, for example 100mM to 350 nM.

Surfactants may also be added to the antibody formulation to reduce aggregation of the formulated antibody and/or to minimize particle formation in the formulation and/or to reduce adsorption. Exemplary surfactants include polyoxyethylene sorbitan fatty acid esters (tweens), polyoxyethylene alkyl ethers (Brij), alkylphenyl polyoxyethylene ethers (Triton-X), polyoxyethylene-polyoxypropylene copolymers (poloxamers, pluronics), and Sodium Dodecyl Sulfate (SDS). An example of a suitable polyoxyethylene sorbitan fatty acid ester is polysorbate 20 (as Tween 20)TMSold under the trademark Tween 80) and polysorbate 80 (sold as Tween 80)TMSold under the trademark trade mark). Examples of suitable polyethylene-polypropylene copolymers areF68 or Poloxamer 188TMThose sold under the trade name. An example of a suitable polyoxyethylene alkyl ether is BrijTMThose sold under the trademark bazaar (r). Exemplary surfactant concentrations range from about 0.001% to about 1% w/v.

Lyoprotectants may also be added to protect unstable active ingredients (e.g., proteins) against destabilizing environments during lyophilization. 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 lyoprotectant may be included in an amount of about 10mM to 500 nM.

In some embodiments, suitable formulations comprise an anti-Tau antibody, and one or more of the above-specified agents (e.g., surfactants, buffers, stabilizers, tonicity agents), and are substantially free of one or more preservatives, such as ethanol, benzyl alcohol, phenol, m-cresol, p-chloro-m-cresol, methyl or propyl p-hydroxybenzoate, benzalkonium chloride, and combinations thereof. In other embodiments, preservatives are included in the formulations at a concentration ranging, for example, from about 0.001 to about 2% (w/v).

For example, a suitable formulation may be a liquid or lyophilized formulation suitable for parenteral administration and can comprise: about 1mg/mL to about 200mg/mL of an anti-Tau antibody; from about 0.001% to about 1% of at least one surfactant; about 1mM to about 100mM of a buffering agent; optionally about 10mM to about 500mM of a stabilizer; and about 5mM to about 305mM of a tonicity agent; and has a pH of about 4.0 to about 7.0.

As another example, a suitable parenteral formulation is a liquid or lyophilized formulation comprising: about 1mg/mL to about 200mg/mL of an anti-Tau antibody; 0.04% Tween20 w/v; 20mM L-histidine; and 250mM sucrose; and has a pH of 5.5.

As another example, suitable parenteral formulations include lyophilized formulations comprising: 1)15mg/mL of an anti-Tau antibody; 0.04% Tween20 w/v; 20mM L-histidine; and 250mM sucrose; and has a pH of 5.5; or 2)75mg/mL of an anti-Tau antibody; 0.04% Tween20 w/v; 20mM L-histidine; and 250mM sucrose; and has a pH of 5.5; or 3)75mg/mL of an anti-Tau antibody; 0.02% Tween20 w/v; 20mM L-histidine; and 250mM sucrose; and has a pH of 5.5; or 4)75mg/mL of an anti-Tau antibody; 0.04% Tween20 w/v; 20mM L-histidine; and 250mM trehalose; and has a pH of 5.5; or 6)75mg/mL of an anti-Tau antibody; 0.02% Tween20 w/v; 20mM L-histidine; and 250mM trehalose; and has a pH of 5.5.

As another example, a suitable parenteral formulation is a liquid formulation comprising: 1)7.5mg/mL of an anti-Tau antibody; 0.022% Tween20 w/v; 120mM L-histidine; and 250125 mM sucrose; and has a pH of 5.5; or 2)37.5mg/mL of an anti-Tau antibody; 0.02% Tween20 w/v; 10mM L-histidine; and 125mM sucrose; and has a pH of 5.5; or 3)37.5mg/mL of an anti-Tau antibody; 0.01% Tween20 w/v; 10mM L-histidine; and 125mM sucrose; and has a pH of 5.5; or 4)37.5mg/mL of an anti-Tau antibody; 0.02% Tween20 w/v; 10mM L-histidine; 125mM trehalose; and has a pH of 5.5; or 5)37.5mg/mL of an anti-Tau antibody; 0.01% Tween20 w/v; 10mM L-histidine; and 125mM trehalose; and has a pH of 5.5; or 6)5mg/mL of an anti-Tau antibody; 0.02% Tween20 w/v; 20mM L-histidine; and 250mM trehalose; and has a pH of 5.5; or 7)75mg/mL of an anti-Tau antibody; 0.02% Tween20 w/v; 20mM L-histidine; and 250mM mannitol; and has a pH of 5.5; or 8)75mg/mL of an anti-Tau antibody; 0.02% Tween20 w/v; 20mM Lhistidine; and 140mM sodium chloride; and has a pH of 5.5; or 9)150mg/mL of an anti-Tau antibody; 0.02% Tween20 w/v; 20mM L-histidine; and 250mM trehalose; and has a pH of 5.5; or 10)150mg/mL of an anti-Tau antibody; 0.02% Tween20 w/v; 20mM L-histidine; and 250mM mannitol; and has a pH of 5.5; or 11)150mg/mL of an anti-Tau antibody; 0.02% Tween20 w/v; 20mM L-histidine; and 140mM sodium chloride and having a pH of 5.5; or 12)10mg/mL of an anti-Tau antibody; 0.01% Tween20 w/v; 20mM L-histidine; and 40mM sodium chloride; and has a pH of 5.5.

The anti-Tau antibody may be used in an aerosol formulation for administration by inhalation. The anti-Tau antibody may be formulated in a pressurized gas acceptable for sealing and pressurization, such as dichlorodifluoromethane, propane, nitrogen, and the like.

In addition, the anti-Tau antibody can be formulated into suppositories by mixing with various bases such as an emulsifying base or a water-soluble base. The anti-Tau antibody may be administered rectally by suppository. The suppository may contain media such as cocoa butter, carbowaxes (carbowaxes), and polyethylene glycols, which melt at body temperature and solidify at room temperature.

Unit dosage forms for oral or rectal administration (e.g., syrups, elixirs and suspensions) may be provided wherein each dosage unit, e.g., teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition containing one or more inhibitors. Similarly, a unit dosage form for injection or intravenous administration may comprise the anti-Tau antibody in a composition in the form of a solution in sterile water, physiological saline, or other pharmaceutically acceptable carrier.

The term "unit dosage form" as used herein means physically discrete units suitable as unitary dosages for human or animal subjects, each unit containing a predetermined quantity of an anti-Tau antibody of the invention, in an amount sufficient to produce the desired effect, in association with a pharmaceutically acceptable diluent, carrier or vehicle. The specification of anti-Tau antibodies may depend on the particular antibody employed and the effect to be achieved, as well as the pharmacodynamics associated with each antibody in the host.

Other modes of administration may also be used in the methods of the invention. For example, suitable antibodies can be formulated in suppositories, and in some cases, in aerosol and intranasal compositions. For suppositories, the vehicle composition will include conventional binders and carriers such as polyalkylene glycols, or triglycerides. Such suppositories may be formed from mixtures containing the active ingredient in the range of about 0.5% to about 10% (w/w), for example about 1% to about 2%.

Intranasal formulations typically contain mediators that neither cause irritation of the nasal mucosa nor significantly interfere with ciliary function. Diluents such as water, aqueous saline or other known materials may be employed. Nasal formulations may also contain preservatives such as, but not limited to, chlorobutanol and benzalkonium chloride. A surfactant may be present to enhance the absorption of the antibody by the nasal mucosa.

The anti-Tau antibody may be administered as an injectable formulation. Typically, injectable compositions are prepared as liquid solutions or suspensions; solid forms suitable for solution or suspension in a liquid medium are also prepared prior to injection. The preparation may also be emulsified or the antibody encapsulated in a liposome medium.

Suitable excipient media are, for example, water, saline, dextrose, glycerol, ethanol, and the like, and combinations thereof. Furthermore, the medium may contain minor amounts of additional substances, such as wetting or emulsifying agents or pH buffering agents, if desired. The actual methods of making such dosage forms are known or will be apparent to those skilled in the art. See, for example, Remington's pharmaceutical Sciences, Mack Publishing Company, Easton, Pennsylvania, 17 th edition, 1985. In any case, the composition or formulation to be administered will contain an amount of anti-Tau antibody sufficient to achieve the desired state in the subject being treated.

Pharmaceutically acceptable excipients, such as vehicles, adjuvants, carriers or diluents, are readily available to the public. In addition, pharmaceutically acceptable additional substances, such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizing agents, wetting agents and the like, are readily available to the public.

In some embodiments, the anti-Tau antibody is formulated in a controlled release formulation. Sustained release preparations may be prepared by methods well known in the art. Suitable examples of sustained release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, copolymers of L-glutamic acid and ethyl-L-glutamine, non-degradable ethylene-vinyl acetate, hydrogels, polylactic acid, degradable lactic acid-glycolic acid copolymers, and poly-D- (-) -3-hydroxybutyric acid. Possible loss of biological activity and possible changes in immunogenicity of the antibodies contained in the sustained release preparations can be avoided by using appropriate additives, by controlling the moisture content and by developing specific polymer matrix compositions.

Controlled release within the scope of the present invention may be taken to mean any of a variety of sustained release dosage forms. For the purposes of the present invention, the following terms may be considered to be substantially equivalent to controlled release: continuous release, controlled release, delayed release, sustained release, gradual release, extended release, programmed release, extended release, proportional release, delayed release, depot, lag, slow release, intermittent release, sustained release, time coat, time release, delayed action, extended action, time-stratified action (layered-time action), prolonged action, delayed action, repeat action, delayed action, sustained action drug, and extended release. Further discussion of these terms can be found in Lesczek Krowczynski, Extended-ReleaseDosage Forms,1987(CRC Press, Inc.).

A wide variety of controlled release technologies cover a very wide range of pharmaceutical dosage forms. Controlled release techniques include, but are not limited to, physical systems and chemical systems.

Physical systems include, but are not limited to, reservoir systems with rate controlling membranes, such as microencapsulation, macroencapsulation (macroencapsulation), and membrane systems; reservoir systems without rate controlling membranes, such as hollow fibers, ultra microporous cellulose triacetate, and porous polymeric substances and foams; monolayer systems, including those that are physically dissolved in a non-porous, polymeric, or elastomeric matrix (e.g., non-erodible, environmental factor leaching, and degradable), and those that are physically dispersed in a non-porous, polymeric, or elastomeric matrix (e.g., non-erodible, environmental factor leaching, and degradable); a sheet structure comprising a reservoir layer that is chemically similar or dissimilar to the outer control layer; and other physical methods such as osmotic pumping, or adsorption onto ion exchange resins.

Chemical systems include, but are not limited to, chemical attack (e.g., heterogeneous or homogeneous attack) on the polymer matrix, or biological attack (e.g., heterogeneous or homogeneous attack) on the polymer matrix. Additional discussion of the types of Controlled Release systems can be found in Agis F.Kydonieus, Controlled Release Technologies, Methods, fashion and applications,1980(CRC Press, Inc.).

There are many controlled release pharmaceutical formulations developed for oral administration. They include, but are not limited to, gastrointestinal delivery systems that control osmotic pressure; a gastrointestinal delivery system that controls hydrodynamic pressure; a gastrointestinal delivery system for controlling membrane permeation, comprising a gastrointestinal delivery system for controlling microporous membrane permeation; gastric juice resistant intestine-targeted controlled release gastrointestinal delivery systems; a gastrointestinal delivery system to control gel diffusion; and a controlled ion-exchange gastrointestinal delivery system comprising a cationic and an anionic drug. Additional information on controlled release Drug Delivery Systems can be found in Yie w. chien, Novel Drug Delivery Systems,1992(Marcel Dekker, Inc.).

Treatment regimens

In one aspect, a method of treating a tauopathy (e.g., an acute tauopathy) in an individual is provided, the method comprising administering to the individual an anti-Tau antibody.

Thus, in one embodiment, the anti-Tau antibody dose per mg/kg body weight is calculated. However, in another embodiment, the dose of anti-Tau antibody is a flat-fixed dose, which is fixed, regardless of the weight of the patient. In some embodiments, the dosage regimen is adjusted to provide the optimal desired response (e.g., an effective response).

In another embodiment, the dose of the anti-Tau antibody is varied over time. For example, the anti-Tau antibody may be administered initially at a high dose and may decrease over time. In another embodiment, the anti-Tau antibody is administered initially at a low dose and increases over time.

In another embodiment, the amount of anti-Tau antibody administered per dose is constant. In another embodiment, the amount of antibody administered varies with each dose. For example, the maintenance (or subsequent) dose of the antibody may be higher than or the same as the loading dose of the first administration. In another embodiment, the maintenance dose of the antibody may be lower than or the same as the loading dose.

In one embodiment, the anti-Tau antibody is administered at a dose of 2, 3,4, 5,6, 7, 8,9, 10, 11, or 12 mg/kg. In one embodiment, the anti-Tau antibody is administered at a dose of 10 mg/kg. In one embodiment, the anti-Tau antibody is administered at a dose of 4 mg/kg. In one embodiment, the anti-Tau antibody is administered 1 time. In one embodiment, the anti-Tau antibody is administered for more than 1 dose.

In other embodiments, the anti-Tau antibody is administered 1 time per week, 1 time every 2 or 3 weeks, 1 time per month, as long as clinical benefit is observed, or for example, until a complete response or unmanageable toxicity.

In another embodiment, the anti-Tau antibody is administered as a first line therapy (e.g., initial or first treatment). In another embodiment, the anti-Tau antibody is administered as a second line therapy (e.g., after the initial or first treatment, including after relapse and/or at the failure of the first treatment).

The following examples are illustrative only and should not be taken as limiting the scope of the invention in any way, as many variations and equivalents will occur to those skilled in the art upon reading the present disclosure.

The contents of all references, Genbank entries, patents and published patent applications cited throughout this document are expressly incorporated herein by reference.

Examples

The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention, and are not intended to limit the scope of what the inventors regard as their invention nor are they intended to represent that the experiments below are all or the only experiments that have been performed. Work has been carried out to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.), but some experimental error and deviation should be accounted for. Unless otherwise indicated, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees celsius, and pressure is at or near atmospheric. Standard abbreviations may be used, such as bp, base pair; kb, kilobases; pl, picoliter; s or sec, seconds; min, min; h or hr, hours; aa, an amino acid; kb, kilobases; bp, base pair; nt, nucleotide; i.m., intramuscularly (di); i.p., intraperitoneally (ground); s.c., subcutaneous (earth); and so on.

106页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:一种高效靶向抗肿瘤药物纳米载体系统及其应用

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!