SERPINS FOR THE TREATMENT OF NEUROINFLAMMATORY DISEASES

Information

  • Patent Application
  • 20180050082
  • Publication Number
    20180050082
  • Date Filed
    March 09, 2016
    8 years ago
  • Date Published
    February 22, 2018
    6 years ago
Abstract
The present invention relates to the use of serpins, including A1AT, its derivatives and analogs thereof, in the prevention or treatment of neuroinflammatory diseases. In particular embodiments, the invention relates to the combination of A1AT and another anti-inflammatory therapeutic compound. The present invention further relates to methods for administering said A1AT combination.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been filed electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Mar. 3, 2016, is named ANV002W_SL.txt and is 161,319 bytes in size.


FIELD OF THE INVENTION

The present invention relates to the use of serpins, including alpha-1 antitrypsin (A1AT or AAT), its derivatives and analogs thereof, in the prevention or treatment of neuroinflammatory diseases. In particular embodiments, the invention relates to the combination of A1AT and another anti-inflammatory therapeutic compound. The present invention further relates to methods of administering said A1AT combination.


BACKGROUND OF THE INVENTION

Alpha-1 antitrypsin (A1AT) is a member of the serpin superfamily of protease inhibitors. Normally found in serum, A1AT inhibits a wide variety of proteases and has been shown to protect tissues from the enzymes secreted by activated immune cells. Alpha-1 antitrypsin (AAT) inhibits IL-8 production as well as IL-8 binding to its receptors. A1AT also inactivates elastase to decrease extracellular matrix degradation of the blood-brain barrier. A1AT inhibits macrophage production of pro-inflammatory cytokines that are upregulated in neuromyelitis optica (NMO), a neurodegenerative disease. A1AT induces Treg cells, tolerogenic dendritic cells, and anti-inflammatory cytokines. Circulating levels of A1AT vary with a normal reference range in the blood of 1.5-3.5 g/L in humans. Since it is a negative feedback molecule that downregulates immune system activity, it is used as a marker of inflammation.


Serpins inactivate enzymes such as neutrophil elastase by covalently binding to the protease in a manner that inhibits enzyme activity. During infection or acute phase response, degranulation rates of immune cells increase markedly, thus high levels of serpins are required for enzyme neutralization and to limit damage to tissue. In circumstances where there are insufficient serpin concentrations in the tissue, fibrosis or scaring of the tissue can arise.


Several genetic mutations have been identified in humans that correspond to the incorrect folding of the beta sheets and alpha helices of A1AT and render the protein non-functional. This can cause A1AT deficiency and results in hepatic cirrhosis and fibrosis throughout the body.


Blood-derived A1AT has been used clinically to rescue patients deficient in A1AT. It has also been used recently to neutralize the effects of neutrophil elastase in patients with emphysema and chronic obstructive pulmonary disease.


Work by Subramanian et al. has demonstrated that sustained expression of human A1AT in a transgenic C57BL/6 mouse background were resistant to MOG-35-55 peptide-induced experimental autoimmune encephalomyelitis. Furthermore, hA1AT expression was also characteristic of elevated CD4+FoxP3+Treg cell counts and diminished pro-inflammatory cytokine expression IL17, IL1b and IL6 and reduced CCR6 chemokine levels. (Metab. Brain Dis. 26(2):107-13 (2011), incorporated by reference herein in its entirety.)


NMO is a rare disorder that resembles multiple sclerosis in several ways but requires a different treatment regimen for disease maintenance. Symptoms of NMO include loss of vision and spinal cord function. Optical neuritis may manifest in patients as visual impairment with decreased visual acuity. Spinal cord demyelination may manifest in patients as muscle weakness, reduced sensory proprioception or loss of bladder and bowel control. During severe flares, patients may experience acute paraparesis or quadriparesis.


Recently, neutrophil elastase involvement, driven by granulocte-mediated inflammation, has been demonstrated in a mouse model of NMO. (See Mult. Scler. 18:398-408 (2012), incorporated by reference herein in its entirety.) At least two different causes are known for NMO including the presence of autoantibodies against aquaporin 4 and aberrant astrocyte activity. An increasing body of evidence also supports the contribution of Th17 cells and a role of granulocytic release at the site of inflammation.


Diagnosis of NMO is currently accepted as requiring two absolute criteria plus at least two supportive criteria. The absolute criteria are optic neuritis and acute myelitis whilst the supportive criteria are brain MRI not consistent with multiple sclerosis at disease onset, spinal cord MRI with contiguous T2-weighted signal abnormality extending over three or more vertebral segments and NMO-IgG seropositivity against aquaporin 4 antigen.


Multiple sclerosis (MS) is a heterogeneous condition consisting of recurrent and simultaneous inflammatory lesions of the spinal cord and brain causing demyelination of nerves in the central nervous system. Symptoms range widely and may be physical (motor function loss, sensory function loss, or pain) or psychological (mood alteration, depression). Several forms of MS exist including remitting and progressive forms. Specifically, forms include relapse-remitting, secondary progressive, primary progressive and progressive relapsing.


Diagnosis of MS is typically based on presenting symptoms with the most common diagnostic tools being neuroimaging, analysis of cerebrospinal fluid (CSF) and evoked potentials. In neuroimaging of patients suspected of having MS, MRI may be used to identify demyelinated areas of the brain and spinal cord. Gadolinium can be administered intravenously to highlight active inflammatory lesions. CSF can be obtained by lumbar puncture and can be used to measure CNS levels of inflammation, mainly oligoclonal bands of IgG by electrophoresis, and cytokines IL17, IL8, IL1b and T cells. Brain responses can be examined using visual and sensory evoked potentials.


Amyotrophic lateral sclerosis (ALS) is a heterogeneous condition involving neuronal cell death. Symptoms of ALS include muscle stiffness and/or muscle twitching with gradual progression to muscle weakness and wasting. Patients with ALS have difficulty speaking, swallowing and eventually breathing. There are currently no definitive diagnostic tests for ALS and the majority of clinical testing is used to rule out other diseases. Often electromyography and nerve conduction velocity testing are used. Magnetic Resonance Imaging is often indeterminate with ALS patients. The disease presents symptoms that include muscle stiffness and twitching in a single limb involving upper or lower motor neurons.


Therapeutic inhibition of granulocyte proteases by sivelestat has been proposed (WO2011100567, incorporated by reference herein in its entirety.). Due to Sivelestat's low potency and therapeutic efficacy, however, more improved treatment regimens are needed for neuroinflammatory diseases associated with significant granulocyte involvement.


SUMMARY OF THE INVENTION

The present invention relates to the use of serpins, including A1AT, its derivatives and analogs thereof, in the prevention or treatment of neuroinflammatory diseases. In particular embodiments, the invention relates to the combination of A1AT and methylprednisolone. The present invention further relates to methods administering said A1AT combination.


Thus, the invention provides a method of treating an inflammatory condition in a subject, comprising administering a serpin protein and administering methylprednisolone. In a preferred embodiment, the serpin protein and methylprednisolone are administered simultaneously.


In other embodiments, the inflammatory condition is neuromyelitis optica (NMO), multiple sclerosis (MS) or amyotrophic lateral sclerosis (ALS).


In some embodiments, the serpin protein has at least a 90% sequence identity to SEQ ID NO:1 and has alpha-1 antitrypsin (A1AT) activity. In a preferred embodiment, the serpin protein has the sequence of SEQ ID NO:1. In another embodiment, the serpin protein is encoded by a nucleic acid that has at least a 90% sequence identity to SEQ ID NO:2 and wherein said serpin protein has alpha-1 antitrypsin (A1AT) activity. In a preferred embodiment, the serpin protein is encoded by a nucleic acid that has the sequence of SEQ ID NO:2.


The invention provides a method of treating an inflammatory condition in a subject, comprising administering a nucleic acid that encodes a serpin protein and administering methylprednisolone. In some embodiments, the inflammatory condition is neuromyelitis optica (NMO), multiple sclerosis (MS), or amyotrophic lateral sclerosis (ALS).


In some embodiments, the nucleic acid encodes a protein having at least a 90% sequence identity to SEQ ID NO:1 and has alpha-1 antitrypsin (A1AT) activity. In a preferred embodiment, the nucleic acid encodes a protein having the sequence of SEQ ID NO:1. In other embodiments, the nucleic acid has at least a 90% sequence identity to SEQ ID NO:2 and the serpin protein has alpha-1 antitrypsin (A1AT) activity. In a preferred embodiment, the nucleic acid has the sequence of SEQ ID NO:2. In other embodiments, the nucleic acid is administered by a route selected from the group consisting of transfected autologous patient cells, viral vectors, and naked nucleic acid preparations.


The invention provides a method of treating an inflammatory condition in a subject, comprising increasing the expression of an endogenous serpin protein and administering methylprednisolone. In other embodiments, the inflammatory condition is neuromyelitis optica (NMO), multiple sclerosis (MS), or amyotrophic lateral sclerosis (ALS).


In some embodiments, the endogenous serpin protein has at least a 90% sequence identity to SEQ ID NO:1 and has alpha-1 antitrypsin (A1AT) activity. In a preferred embodiment, the endogenous serpin protein has the sequence of SEQ ID NO:1. In other embodiments, the endogenous serpin protein is encoded by a nucleic acid that has at least a 90% sequence identity to SEQ ID NO:2 and wherein the serpin protein has alpha-1 antitrypsin (A1AT) activity. In a preferred embodiment, the endogenous serpin protein is encoded by a nucleic acid that has the sequence of SEQ ID NO:2. In other embodiments, the increase in serpin expression is accomplished using a technology selected from the group consisting of zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9.


The invention provides a pharmaceutical composition, comprising a serpin protein and methylprednisolone. In some embodiments, the serpin protein within the pharmaceutical composition has at least a 90% sequence identity to SEQ ID NO:1 and has alpha-1 antitrypsin (A1AT) activity. In a preferred embodiment, the serpin protein has the sequence of SEQ ID NO:1. In other embodiments, the serpin protein is encoded by a nucleic acid that has at least a 90% sequence identity to SEQ ID NO:2 and wherein the serpin protein has alpha-1 antitrypsin (A1AT) activity. In a preferred embodiment, the serpin protein within the pharmaceutical composition is encoded by a nucleic acid that has the sequence of SEQ ID NO:2.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows A1AT treatment of Neuromyelitis Optica (NMO) induced by the adoptive transfer of Th17 Experimental Autoimmune Encephalomyelitis (EAE) cells to C57BL/6 mice.



FIG. 2 shows that A1AT treatment reduced the NMO disease score compared to PBS (control) and was superior to Sivelestat at all days of overt signs of disease as well as delay onset of disease.



FIG. 3 shows mean NMO disease scores are reduced by A1AT+Mpred treatment when compared to no treatment (PBS) or either treatment alone. The data represent 10 recipient mice per cohort in a blinded study and is expressed as the mean disease score+/−SEM.



FIG. 4 shows that A1AT+Mpred resulted in less body weight loss than either treatment alone. Ten recipient mice per cohort were used in this blinded study. The graph shows mean body weight as a percentage of the value measured at day 0+/−SEM.



FIG. 5A shows that A1AT alone and A1AT+Mpred decreased the inflammatory cytokine IL-17A when compared to PBS and Mpred alone.



FIG. 5B shows that A1AT alone and A1AT+Mpred decreased the inflammatory cytokine IFN-γ when compared to Mpred alone.



FIG. 5C shows that all treatments increased IL-6 compared to PBS. A1AT+Mpred, however, showed the most significant reduction.



FIG. 5D shows that IL-2 levels were higher without antigenic restimulation (0 MOG peptide) when treated with A1AT alone or A1AT+Mpred when compared to Mpred alone.





DETAILED DESCRIPTION OF THE INVENTION

The invention provides for the treatment or prevention of neuroinflammatory or neurodegenerative diseases with the combination of a serpin and another anti-inflammatory therapeutic compound. In particular embodiments, the neuroinflammatory condition is neuromyelitis optica (NMO), multiple sclerosis (MS), or amyotrophic lateral sclerosis (ALS). In other embodiments, the disease is associated with abnormal levels of Th17, CCR6 or IL8. In yet other embodiments, the Serpin is alpha-1 antitrypsin (A1AT). In other embodiments, the anti-inflammatory compound is methylprednisolone. In yet other embodiments of the invention, the therapeutic combinations disclosed herein are administered following diagnosis of a neuroinflammatory or neurodegenerative disease using a diagnostic test that measures circulating IL17, CCR6, IL8, anti-aquaporin 4 antibodies, Kir4 antibodies, neutrophil elastase or A1AT levels.


In describing and claiming one or more embodiments of the present invention, the following terminology will be used in accordance with the definitions described below:


The singular form “a”, “an”, and “the” includes plural references unless indicated otherwise.


The term “absorption” is the movement of a drug into the bloodstream. A drug needs to be introduced via some route of administration. For example, drugs of the invention may be delivered by oral, buccal, topical, dermal, inhalation, nasal, subcutaneous, intramuscular, or intravenous route or by any other route known in the pharmaceutical arts. Exemplary dosage forms include a solution, emulsion, inhalable powder, suspension, tablet, patch, capsule or other liquid.


“Amyotrophic lateral sclerosis” (ALS) as used herein includes, without limitation, a heterogeneous condition involving neuronal cell death. Symptoms of ALS include muscle stiffness and/or muscle twitching with gradual progression to muscle weakness and wasting. Patients with ALS have difficulty speaking, swallowing and eventually breathing.


A “clinician” or “medical researcher” or “veterinarian” as used herein, can include, without limitation, doctors, nurses, physician assistants, lab technicians, research scientists, clerical workers employed by the same, or any person involved in determining, diagnosing, aiding in the diagnosis or influencing the course of treatment for the individual.


An “effective amount” refers to an amount of therapeutic compound that is effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result. A “therapeutically effective amount” of a therapeutic compound may vary according to factors such as the disease state, age, sex, and weight of the individual. A therapeutically effective amount may be measured, for example, by improved survival rate, more rapid recovery, or amelioration, improvement or elimination of symptoms, or other acceptable biomarkers or surrogate markers. A “therapeutically effective amount” is also one in which any toxic or detrimental effects of the therapeutic compound are outweighed by the therapeutically beneficial effects. A “prophylactically effective amount” refers to an amount of therapeutic compound that is effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, but not necessarily, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.


“Homologs” are bioactive molecules that are similar to a reference molecule at the nucleotide sequence, peptide sequence, functional, or structural level. Homologs may include sequence derivatives that share a certain percent identity with the reference sequence. Thus, in one embodiment, homologous or derivative sequences share at least a 70 percent sequence identity. In a preferred embodiment, homologous or derivative sequences share at least an 80 or 85 percent sequence identity. In a more preferred embodiment, homologous or derivative sequences share at least an 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99 percent sequence identity. Homologous or derivative nucleic acid sequences may also be defined by their ability to remain bound to a reference nucleic acid sequence under high stringency hybridization conditions. Homologs having a structural or functional similarity to a reference molecule may be chemical derivatives of the reference molecule. Methods of detecting, generating, and screening for structural and functional homologs as well as derivatives are known in the art.


“Hybridization” generally depends on the ability of denatured DNA to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature that can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency of hybridization reactions, see Ausubel et al, Current Protocols in Molecular Biology, Wiley Interscience Publishers (1995).


An “individual,” “subject” or “patient” is a vertebrate. In certain embodiments, the vertebrate is a mammal. Mammals include, but are not limited to, primates (including human and non-human primates), rodents (e.g., mice, hamsters, guinea pigs, and rats), farm animals, sport animals, and pets (e.g. dogs and cats). In certain embodiments, a mammal is a human. A “control subject” may refer to a healthy subject who has not been diagnosed as having a disease, dysfunction, or condition that has been identified in an individual, subject, or patient. The control subject does not suffer from any sign or symptom associated with the disease, dysfunction, or condition. Alternatively, a control subject may be a sick subject that does not receive the therapeutic drug, another drug, or a lower dose of the drug.


A “medicament” is an active drug that has been manufactured for the treatment of a disease, disorder, or condition.


“Morpholinos” are synthetic molecules that are non-natural variants of natural nucleic acids that utilize a phosphorodiamidate linkage, described in U.S. Pat. No. 8,076,476, incorporated by reference herein in its entirety.


“Multiple sclerosis” (MS) as used herein can include without limitation a heterogeneous condition consisting of recurrent and simultaneous inflammatory lesions of the spinal cord and brain causing demyelination of nerves in the central nervous system.


“Neuromyelitis optica” (NMO) as used herein can include without limitation a heterogeneous condition consisting of recurrent and simultaneous inflammation and demyelination of the optic nerve (optic neuritis) and spinal cord (myelitis). At least two different causes are known for NMO including the presence of autoantibodies against aquaporin 4 and aberrant astrocyte activity. An increasing body of evidence also supports the contribution of Th17 cells and a role of granulocytic release at the site of inflammation.


“Neuromyelitis optica” (NMO) as used herein can include without limitation a heterogeneous condition consisting of acute, recurrent or chronic/progressive inflammation or demyelination of the optic nerve (optic neuritis) or spinal cord (myelitis).


“Nucleic acids” are any of a group of macromolecules, either DNA, RNA, or variants thereof, that carry genetic information that may direct cellular functions. Nucleic acids may have enzyme-like activity (for instance ribozymes) or may be used to inhibit gene expression in a subject (for instance RNAi). The nucleic acids used in the inventions described herein may be single-stranded, double-stranded, linear or circular. The inventions further incorporate the use of nucleic acid variants including, but not limited to, aptamers, PNA, Morpholino, or other non-natural variants of nucleic acids. By way of example, nucleic acids useful for the invention are described in U.S. Pat. No. 8,076,476, incorporated by reference herein in its entirety.


“Patient response” or “response” can be assessed using any endpoint indicating a benefit to the patient, including, without limitation, (1) inhibition, to some extent, of disease progression, including stabilization, slowing down and complete arrest; (2) reduction in the number of disease episodes and/or symptoms; (3) inhibition (i.e., reduction, slowing down or complete stopping) of a disease cell infiltration into adjacent peripheral organs and/or tissues; (4) inhibition (i.e. reduction, slowing down or complete stopping) of disease spread; (5) decrease of an autoimmune condition; (6) favorable change in the expression of a biomarker associated with the disorder; (7) relief, to some extent, of one or more symptoms associated with a disorder; (8) increase in the length of disease-free presentation following treatment; or (9) decreased mortality at a given point of time following treatment.


As used herein, the term “peptide” is any peptide comprising two or more amino acids. The term peptide includes short peptides (e.g., peptides comprising between 2-14 amino acids), medium length peptides (15-50) or long chain peptides (e.g., proteins). The terms peptide, medium length peptide and protein may be used interchangeably herein. As used herein, the term “peptide” is interpreted to mean a polymer composed of amino acid residues, related naturally occurring structural variants, and synthetic non-naturally occurring analogs thereof linked via peptide bonds, related naturally-occurring structural variants, and synthetic non-naturally occurring analogs thereof. Synthetic peptides can be synthesized, for example, using an automated peptide synthesizer. Peptides can also be synthesized by other means such as by cells, bacteria, yeast or other living organisms. Peptides may contain amino acids other than the 20 gene-encoded amino acids. Peptides include those modified either by natural processes, such as processing and other post-translational modifications, but also by chemical modification techniques. Such modifications are well described in basic texts and in more detailed monographs, and are well-known to those of skill in the art. Modifications can occur anywhere in a peptide, including the peptide backbone, the amino acid side chains, and the amino or carboxyl termini.


As used herein, a “pharmaceutically acceptable carrier” or “therapeutic effective carrier” is aqueous or nonaqueous (solid), for example alcoholic or oleaginous, or a mixture thereof, and can contain a surfactant, emollient, lubricant, stabilizer, dye, perfume, preservative, acid or base for adjustment of pH, a solvent, emulsifier, gelling agent, moisturizer, stabilizer, wetting agent, time release agent, humectant, or other component commonly included in a particular form of pharmaceutical composition. Pharmaceutically acceptable carriers are well known in the art and include, for example, aqueous solutions such as water or physiologically buffered saline or other solvents or vehicles such as glycols, glycerol, and oils such as olive oil or injectable organic esters. A pharmaceutically acceptable carrier can contain physiologically acceptable compounds that act, for example, to stabilize or to increase the absorption of specific modulator(s), for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients.


The term “pharmaceutical dose” or “pharmaceutical dosage form,” refers to physically discrete units suitable as unitary dosages for humans and other mammals, each unit comprising a predetermined quantity of agents in an amount calculated sufficient to produce the desired effect in association with an acceptable diluent, carrier, or vehicle of a formulation. The specifications for the unit dosage forms may depend on the particular serpin form employed, the effect to be achieved, the route of administration and the pharmacodynamics associated with the mammal.


“PNA” refers to peptide nucleic acids with a chemical structure similar to DNA or RNA. Peptide bonds are used to link the nucleotides or nucleosides together.


“Stringency” of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures.


“Stringent conditions” or “high stringency conditions”, as defined herein, can be identified by those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C.; or (3) overnight hybridization in a solution that employs 50% formamide, 5×SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5×Denhardt's solution, sonicated salmon sperm DNA (50 μl/ml), 0.1% SDS, and 10% dextran sulfate at 42° C., with a 10 minute wash at 42° C. in 0.2×SSC (sodium chloride/sodium citrate) followed by a 10 minute high-stringency wash consisting of 0.1×SSC containing EDTA at 55° C.


As used herein, “treatment” refers to clinical intervention in an attempt to alter the natural course of the individual or cell being treated, and can be performed before or during the course of clinical pathology. Desirable effects of treatment include preventing the occurrence or recurrence of a disease or a condition or symptom thereof, alleviating a condition or symptom of the disease, diminishing any direct or indirect pathological consequences of the disease, decreasing the rate of disease progression, ameliorating or palliating the disease state, and achieving remission or improved prognosis. In some embodiments, methods and compositions of the invention are useful in attempts to delay development of a disease or disorder.


It is intended that every maximum numerical limitation given throughout this specification includes every lower numerical limitation, as if such lower numerical limitations were expressly written herein. Every minimum numerical limitation given throughout this specification will include every higher numerical limitation, as if such higher numerical limitations were expressly written herein. Every numerical range given throughout this specification will include every narrower numerical range that falls within such broader numerical range, as if such narrower numerical ranges were all expressly written herein.


In some embodiments, serpins and the other anti-inflammatory therapeutic compounds work synergistically. A person of skill in the art would appreciate that the synergistic activities may be established at appropriate fixed-dose ratios for efficacy against granulocyte-associated neuroinflammatory diseases. This may be determined by varying the amounts of two agents administered to appropriate animal models of inflammatory disease. The model may reflect either an active disease (following disease onset) or an early time point representative of pre-clinical disease. The effect on disease activity or progression is measured.


In other embodiments, the effects of varying amounts of the two agents may be determined in a cellular response mediating inflammation that may be involved in the pathogenesis of the disease. In other embodiments, the effects of varying amounts of the two agents in various formulations is determined by measuring enzymatic activity in vitro.


The presence, absence or degree of associated disease pathology or inflammatory markers such as granulocyte counts (in situ or circulating), protease activity, Th17 effector cell counts, plasma cell counts, CCR6, IL8, IL17, IL23, endogenous serpin, anti-aquaporin 4 antibodies, anti-Kir4 antibody levels can be used to evaluate efficacy. Successful compositions with the appropriate determined dose are selected that reduce said inflammatory markers, ameliorate disease symptoms or are tolerated by test animals.


The invention provides that the pharmaceutical compositions disclosed herein are formulated into solid, semi-solid, pressed powder, powder, liquid, gel, suspension, emulsion, or gaseous forms. In other embodiments, the pharmaceutical compositions are formulated into liquids, syrups, concentrates, tablets, capsules, caplets, powders, rapid melts, thin strips, granules, ointments, crémes, solutions, suspensions, emulsions, suppositories, injections, inhalants, gels, crystals and aerosols.


Methods for Treating Neuroinflammatory Diseases

Individuals and other mammals at increased risk for development of a granulocyte-mediated neuroinflammatory disease, with early-stage of disease, or with established disease, may be treated with a clinically effective amount of any of the compositions disclosed herein. In some embodiments, the pharmaceutical compositions described herein prevent the development of disease, prevent the progression of disease, or to prevent the progression of the symptoms or signs of disease.


Thus, disclosed herein are methods for treating a patient with a neurodegenerative disease. Also disclosed herein are methods for treating a patient with symptoms consistent with neurodegenerative disease. Persons of skill in the art may determine preferred routes of administering the pharmaceutical compositions described herein, the corresponding dosage form, dose amount, and the dosing regimen (i.e., the frequency of dosing).


In some embodiments, the composition may be delivered in multi-dosing formats whereby the compositions are administered several times a week, once a day, twice a day, three times a day or more to achieve the appropriate therapeutic level. Other variables to consider include the specific serpin, inflammatory markers that are measured, the disease symptoms to be affected, the specific neuroinflammatory disease, the other anti-inflammatory therapeutic agent involved and its pharmacokinetic profile, and the specific individual involved.


In other embodiments, the frequency of administration may be once a month, once a week, once a day, or on an as-needed basis. Frequency of administration may be dependent on the identity and concentration of serpin in the composition or the disease risk assessment, disease severity, test results, clinician preference, or pharmaceutical formulation.


Polynucleotides Encoding Serpins

Some embodiments of the invention provide serpin compositions for use as treatment for neuroinflammatory diseases. Said compositions may be administered on a daily, weekly, monthly, yearly or on an as-needed basis to reduce symptoms of disease or to reduce disease progression. In some embodiments, the serpin is A1AT.


The invention provides nucleic acids encoding a serpin protein. The nucleic acids may be DNA molecules, RNA molecules, aptamers (single-stranded or double-stranded), DNA or RNA oligonucleotides, larger DNA molecules that are linear or circular, oligonucleotides that are used for RNA interference (RNAi), variations of DNA such as substitution of DNA/RNA hybrid molecules, synthetic DNA-like molecules such as PNA or other nucleic acid derivative molecules (see WO07/035922, incorporated by reference herein in its entirety). In another embodiment, the therapeutic compound is composed of nuclease-resistant DNA or RNA oligonucleotides. In a preferred embodiment, nuclease-resistant DNA oligonucleotides are Morpholinos, (i.e. phosphorodiamidate analogs of nucleic acids that bind to nucleic acids in a sequence-specific manner, Sarepta Therapeutics, Cambridge Mass.).


In some embodiments, the serpin nucleic acids of the invention are synthesized using methods well-known in the art. In one embodiment, the nucleic acids are generated by enzymes. In exemplary embodiments, the enzymes may include DNA polymerases, RNA polymerases, ligases, and DNA repair enzymes. In another preferred embodiment, the nucleic acids are generated by a polymerase chain reaction (PCR) protocol. See, e.g. U.S. Pat. No. 4,683,195. In other embodiments, the nucleic acids are chemically synthesized using techniques well-known in the art. Typically, solid-phase nucleic acid synthesizers are used. Exemplary chemistries include phosphodiester synthesis, phosphotriester synthesis, and phosphite triester synthesis. See, e.g., Reese, Colin B. (2005). “Oligo- and poly-nucleotides: 50 years of chemical synthesis”. Organic & Biomolecular Chemistry 3 (21): 3851. The skilled artisan would understand that any techniques for synthesizing the nucleic acids and derivatives disclosed herein may be used.


In some embodiments, the serpin compositions of the invention may include A1AT. In a preferred embodiment, a nucleic acid containing at least about a 90% sequence identity to the human gene encoding A1AT precursor protein (SEQ ID NO:1) is delivered to a patient having neuroinflammatory symptoms consistent with NMO, MS and ALS. In another preferred embodiment, the A1AT composition may include a nucleic acid containing a sequence derived from A1AT mRNA. In another preferred embodiment, mRNA encoding human A1AT precursor protein is delivered to a patient having neuroinflammatory symptoms consistent with NMO, MS and ALS. In more preferred embodiments, the invention contemplates nucleic acids that hybridize with high stringency to a nucleic acid encoding A1AT (e.g. SEQ ID NO:2). Other preferred embodiments, nucleic acids encoding serpins are delivered to an individual via a viral vector, as a naked nucleic acid, or in a transformed cell.


In some embodiments, nucleic acids encoding serpins are administered to a patient in a cell-dependent manner. In preferred embodiments, the serpins or nucleic acids encoding them are delivered using transfected autologous patient cells. In other embodiments, serpins are delivered by intrathecal, intramuscular, intravascular, subcutaneous, intracranial, intraocular injection or inhaled routes. In more preferred embodiments, the nucleic acid encodes an A1AT protein having at least about a 90% sequence identity to SEQ ID NO:1. In more preferred embodiments, the serpin is an A1AT protein encoded by a nucleic acid that hybridize with high stringency to a nucleic acid encoding A1AT (e.g. SEQ ID NO:2).


The nucleic acids of the invention encode serpins that retain serpin functional activity. In preferred embodiments, the nucleic acids of the invention encode proteins that retain A1AT functional activity.


The invention provides non-viral serpin liquid or powder formulations. In some embodiments, the serpin is A1AT. In preferred embodiments, the serpin dose range is based on the selection of serpin form and associated properties. For example, plasmid backbone, promoter strength, and size, etc. In preferred embodiments, the copy number ranges from about 500 mM to about 10 pM per dose, depending on the use. Other embodiments comprise a serpin from about 500 mM to about 1 mM per dose. Further embodiments comprise a serpin from about 500 μM to about 1 μM per dose. Yet other embodiments comprise a serpin from about 10 μM to about 10 nM per dose. Further embodiments comprise a serpin from about 800 nM to about 10 pM per dose.


The invention provides viral serpin liquid or powder formulations. In preferred embodiments, the serpin is A1AT. Serpin dose can range based on selection of virus. Generally recommended are dose ranges from about 5×109 PFU/mL to about 1×103 PFU/mL per dose, depending on the use. Some compositions may comprise serpins from about 5×109 PFU/mL to about 1×108 PFU/mL per dose. Some compositions may comprise serpins from about 0.9×108 PFU/mL to about 1×106 PFU/mL per dose. Other compositions may comprise serpins from about 0.9×106 PFU/mL to about 1×105 PFU/mL per dose. Yet other compositions may comprise serpins from about 0.9×105 PFU/mL to about 1×103 PFU/mL per dose.


Periodicity of dosing may vary based on patient needs. In some embodiments, serpins are administered on a weekly or monthly basis. One advantage of a genetic approach is that serpin levels can be sustained longer than recombinant and human-derived purified forms.


Gene therapies and viral vectors that introduce DNA, RNA, transgenes, or other nucleic acid sequences to individuals are known in the art. (See, e.g., US20160046961; US20160040150; USRE045847; US20150218585; US20150167003; and U.S. Pat. No. 9,023,646; Rosenberg et al. N Engl. J. Med. 323: 570-8 (1990); Baltimore et al., Science 348: 36-8 (2015). The foregoing references are incorporated by reference in their entirety.)


The invention contemplates providing serpins using recombinant DNA techniques that result in addition or increased expression of a serpin. Exemplary technologies include homologous recombination, knock-in, ZFNs (zinc finger nucleases), TALENs (transcription activator-like effector nucleases), CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9, and other site-specific nuclease technologies. These techniques enable double-strand DNA breaks at desired locus sites. These controlled double-strand breaks promote homologous recombination at the specific locus sites. This process relies on targeting specific sequences of nucleic acid molecules, such as chromosomes, with endonucleases that recognize and bind to the sequences and induce a double-stranded break in the nucleic acid molecule. The double-strand break is repaired either by an error-prone non-homologous end-joining (NHEJ) or by homologous recombination (HR). (See, e.g., WO2016025759, WO2015191693, US20160046961, US20160046960, US20160046952, US20160046915, and U.S. Pat. No. 9,260,752, each of which are incorporated by reference herein in their entirety.)


Serpin Proteins

The invention provides therapeutic serpin peptides as disclosed herein. In one embodiment, the Serpin is a protein that has at least a 90% sequence identity to SEQ ID NO:1 and has A1AT activity. In preferred embodiments, the Serpin has the sequence of SEQ ID NO:1.


The terms “protein” and “peptide” refer to molecules that include a string of amino acids. The amino acids in the peptides of the invention may be naturally-occurring or non-naturally-occurring. The peptides of the invention may be synthesized chemically or biologically, and can include cysteine-rich peptides, circular peptides, stapled peptides, peptides that include D- or L-amino acids and mixtures thereof, peptidomimetics, peptide-nucleic acids (PNAs), and combinations thereof.


The invention provides recombinant or synthesized serpin compositions. In preferred embodiments, recombinant serpin compositions comprise cell-derived, purified serpins. In other preferred embodiments, human serpin precursor proteins are purified from an in vitro transfected cell culture.


In some embodiments, synthetic serpins are synthesized using protein chemistry known in the art. In preferred embodiments, the synthetic proteins are synthesized using liquid-phase or solid-phase peptide synthesis techniques.


Also contemplated within the scope of embodiments described herein are serpin peptides that are branched or cyclic, with or without branching. Cyclic, branched and branched circular peptides result from post-translational natural processes and are also made by suitable synthetic methods. In some embodiments, any peptide product described herein comprises a peptide analog described above that is then covalently attached to an alkyl-glycoside surfactant moiety.


Other embodiments include serpin peptide chains that are comprised of natural and unnatural amino acids or analogs of natural amino acids. As used herein, peptide and/or protein “analogs” comprise non-natural amino acids based on natural amino acids, such as tyrosine analogs, which includes para-substituted tyrosines, ortho-substituted tyrosines, and meta-substituted tyrosines, wherein the substituent on the tyrosine comprises an acetyl group, a benzoyl group, an amino group, a hydrazine, an hydroxyamine, a thiol group, a carboxy group, a methyl group, an isopropyl group, a C2-C20 straight chain or branched hydrocarbon, a saturated or unsaturated hydrocarbon, an O-methyl group, a polyether group, a halogen, a nitro group, or the like.


Additional embodiments include serpin peptide chains having modified amino acids.


Examples include acylated amino acids at the ε-position of Lysine, amino acids with fatty acids such as octanoic, decanoic, dodecanoic, tetradecanoic, hexadecanoic, octadecanoic, 3-phenylpropanoic acids and the like, or with saturated or unsaturated alkyl chains. (Zhang, L. and Bulaj, G (2012) Curr Med Chem 19: 1602-1618, incorporated herein by reference in its entirety).


The invention further contemplates serpin peptide chains comprising natural and unnatural amino acids or analogs of natural amino acids. In some embodiments, peptide or protein “analogs” comprise non-natural amino acids based on natural amino acids, such as tyrosine analogs, which includes para-substituted tyrosines, ortho-substituted tyrosines, and meta-substituted tyrosines, wherein the substituent on the tyrosine comprises an acetyl group, a benzoyl group, an amino group, a hydrazine, an hydroxyamine, a thiol group, a carboxy group, a methyl group, an isopropyl group, a C2-C20 straight chain or branched hydrocarbon, a saturated or unsaturated hydrocarbon, an O-methyl group, a polyether group, a halogen, a nitro group, or the like. Examples of Tyr analogs include 2,4-dimethyl-tyrosine (Dmt), 2,4-diethyl-tyrosine, O-4-allyl-tyrosine, 4-propyl-tyrosine, Ca-methyl-tyrosine and the like. Examples of lysine analogs include ornithine (Orn), homo-lysine, Ca-methyl-lysine (CMeLys), and the like. Examples of phenylalanine analogs include, but are not limited to, meta-substituted phenylalanines, wherein the substituent comprises a methoxy group, a C1-C20 alkyl group, for example a methyl group, an allyl group, an acetyl group, or the like. Specific examples include, but are not limited to, 2,4,6-trimethyl-L-phenylalanine (Tmp), O-methyl-tyrosine, 3-(2-naphthyl)alanine (Nal(2)), 3-(1-naphthyl)alanine (Nal(1)), 3-methyl-phenylalanine, 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (Tic), fluorinated phenylalanines, isopropyl-phenylalanine, p-azido-phenylalanine, p-acyl-phenylalanine, p-benzoyl-phenylalanine, p-iodo-phenylalanine, p-bromophenylalanine, p-amino-phenylalanine, and isopropyl-phenylalanine, and the like.


Also contemplated within the scope of embodiments are therapeutic peptide chains containing nonstandard or unnatural amino acids known to the art, for example, C-alpha-disubstituted amino acids such as Aib, Ca-diethylglycine (Deg), aminocyclopentane-1-carboxylic acid (Ac4c), aminocyclopentane-1-carboxylic acid (Ac5c), and the like. Such amino acids frequently lead to a restrained structure, often biased toward an alpha helical structure (Kaul, R. and Balaram, P. (1999) Bioorg Med Chem 7: 105-117, incorporated herein by reference in its entirety). Additional examples of such unnatural amino acids useful in analog design are homo-arginine (Har) and the like. Substitution of reduced amide bonds in certain instances leads to improved protection from enzymatic destruction or alters receptor binding. By way of example, incorporation of a Tic-Phe dipeptide unit with a reduced amide bond between the residues (designated as Tic-F[CH2-NH]̂-Phe) reduces enzymatic degradation.


In some embodiments, modifications at the amino or carboxyl terminus may optionally be introduced into the present peptides or proteins (Nestor, J. J., Jr. (2009) Current Medicinal Chemistry 16: 4399-4418). For example, the present peptides or proteins can be truncated or acylated on the N-terminus (Gourlet, P., et al. (1998) Eur J Pharmacol 354: 105-1 1 1, Gozes, I. and Furman, S. (2003) Curr Pharm Des 9: 483-494), the contents of which is incorporated herein by reference in their entirety). Other modifications to the N-terminus of peptides or proteins, such as deletions or incorporation of D-amino acids such as D-Phe result in potent and long acting agonists or antagonists when substituted with the modifications described herein such as long chain alkyl glycosides.


Thus, the invention provides serpin compound analogs wherein the native therapeutic compound is modified by acetylation, acylation, PEGylation, ADP-ribosylation, amidation, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-link formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins, such as arginylation, and ubiquitination. See, for instance, (Nestor, J. J., Jr. (2007) Comprehensive Medicinal Chemistry II 2: 573-601, Nestor, J. J., Jr. (2009) Current Medicinal Chemistry 16: 4399-4418, Uy, R. and Wold, F. (1977) Science 198:890-6, Seifter, S. and Englard, S. (1990) Methods Enzymol 182: 626-646, Rattan, S. I., et al. (1992) Ann NY Acad Sci 663: 48-62). The foregoing references are incorporated by reference in their entirety.


Glycosylated serpin peptides may be prepared using conventional Fmoc chemistry and solid phase peptide synthesis techniques, e.g., on resin, where the desired protected glycoamino acids are prepared prior to peptide synthesis and then introduced into the peptide chain at the desired position during peptide synthesis. Thus, the therapeutic peptide polymer conjugates may be conjugated in vitro. The glycosylation may occur before deprotection. Preparation of amino acid glycosides is described in U.S. Pat. No. 5,767,254, WO 2005/097158, and Doores, K., et al., Chem. Commun., 1401-1403, 2006, which are incorporated herein by reference in their entirety. For example, alpha and beta selective glycosylations of serine and threonine residues are carried out using the Koenigs-Knorr reaction and Lemieux's in situ anomerization methodology with Schiff base intermediates. Deprotection of the Schiff base glycoside is then carried out using mildly acidic conditions or hydrogenolysis. A composition, comprising a glycosylated therapeutic peptide conjugate is made by stepwise solid phase peptide synthesis involving contacting a growing peptide chain with protected amino acids in a stepwise manner, wherein at least one of the protected amino acids is glycosylated, followed by water-soluble polymer conjugation. Such compositions may have a purity of at least 95%, at least 97%, or at least 98%, of a single species of the glycosylated and conjugated therapeutic peptide.


Monosaccharides that may by used for introduction at one or more amino acid residues of the therapeutic peptides defined and/or disclosed herein include glucose (dextrose), fructose, galactose, and ribose. Additional monosaccharides suitable for use include glyceraldehydes, dihydroxyacetone, erythrose, threose, erythrulose, arabinose, lyxose, xylose, ribulose, xylulose, allose, altrose, mannose, N-Acetylneuraminic acid, fucose, N-Acetylgalactosamine, and N-Acetylglucosamine, as well as others. Glycosides, such as mono-, di-, and trisaccharides for use in modifying a therapeutic peptide, one or more amino acid residues of the therapeutic peptides defined and/or disclosed herein include sucrose, lactose, maltose, trehalose, melibiose, and cellobiose, among others. Trisaccharides include acarbose, raffinose, and melezitose.


In other embodiments, the nucleic acids of the invention may be expressed in microorganisms. Promoters for expressing genes of interest are known in the art. In some embodiments, the expression vectors of the invention may have promoters, transcription terminators, or selectable markers. Either inducible or constitutive promoters are contemplated by the invention.


In a preferred embodiment, the nucleic acids of the invention are expressed in bacterial systems because of their low cost, high productivity, and rapid use. Thus, the nucleic acids are expressed in, for example, Bacillus brevis, Bacillus megaterium, Bacillus subtilis, Caulobacter crescentus, Escherichia coli and their derivatives. Exemplary promoters include the 1-arabinose inducible araBAD promoter (PBAD), the lac promoter, the 1-rhamnose inducible rhaP BAD promoter, the T7 RNA polymerase promoter, the trc and tac promoter, the lambda phage promoter pL, and the anhydrotetracycline-inducible tetA promoter/operator.


In some embodiments, the nucleic acids of the invention are expressed in yeast expression systems. Exemplary promoters used in yeast vectors include the promoters for 3-phosphoglycerate kinase (Hitzeman et al., J. Biol. Chem. 255:2073 ((1980)); and other glycolytic enzymes (Hess et al., J. Adv. Enzyme Res. 7:149 (1968); Holland et al., Biochemistry 17:4900 (1978)), e.g., enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyvurate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate somerase, phosphoglucose isomerase, glucokinase alcohol oxidase I (AOX1), alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, and the aforementioned glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization. Any plasmid vector containing a yeast-compatible promoter and termination sequences, with or without an origin of replication, is suitable. Yeast expression systems are commercially available, for example, from Clontech Laboratories, Inc. (Palo Alto, Calif., e.g. pYEX 4T family of vectors for S. cerevisiae), Invitrogen (Carlsbad, Calif., e.g. pPICZ series Easy Select Pichia Expression Kit) and Stratagene (La Jolla, Calif., e.g. ESP™ Yeast Protein Expression and Purification System for S. pombe and pESC vectors for S. cerevisiae).


In other embodiments, the nucleic acids of the invention are expressed in mammalian expression systems. Examples of suitable mammalian promoters for use in the invention include, for example, promoters from the following genes: ubiquitin/S27a promoter of the hamster (WO 97/15664), Simian vacuolating virus 40 (SV40) early promoter, adenovirus major late promoter, mouse metallothionein-I promoter, the long terminal repeat region of Rous Sarcoma Virus (RSV), mouse mammary tumor virus promoter (MMTV), Moloney murine leukemia virus Long Terminal repeat region, and the early promoter of human Cytomegalovirus (CMV). Examples of other heterologous mammalian promoters are the actin, immunoglobulin or heat shock promoter(s). In a preferred embodiment, a yeast alcohol oxidase promoter is used.


In additional embodiments, promoters for use in mammalian host cells can be obtained from the genomes of viruses such as polyoma virus, fowlpox virus (UK 2,211,504 published 5 Jul. 1989), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40). In further embodiments, heterologous mammalian promoters are used. Examples include the actin promoter, an immunoglobulin promoter, and heat-shock promoters. The early and late promoters of SV40 are conveniently obtained as an SV40 restriction fragment which also contains the SV40 viral origin of replication. Fiers et al., Nature 273: 113-120 (1978). The immediate early promoter of the human cytomegalovirus is conveniently obtained as a HindIII E restriction fragment. Greenaway, P. J. et al., Gene 18: 355-360 (1982). The foregoing references are incorporated by reference in their entirety.


In some embodiments, the nucleic acids of the invention are expressed in insect cell expression systems. Eukaryotic expression systems employing insect cell hosts may rely on either plasmid or baculoviral expression systems. The typical insect host cells are derived from the fall army worm (Spodoptera frugiperda). For expression of a foreign protein these cells are infected with a recombinant form of the baculovirus Autographa californica nuclear polyhedrosis virus which has the gene of interest expressed under the control of the viral polyhedrin promoter. Other insects infected by this virus include a cell line known commercially as “High 5” (Invitrogen) which is derived from the cabbage looper (Trichoplusia ni). Another baculovirus sometimes used is the Bombyx mori nuclear polyhedorsis virus which infect the silk worm (Bombyx mori). Numerous baculovirus expression systems are commercially available, for example, from Invitrogen (Bac-N-Blue™), Clontech (BacPAK™ Baculovirus Expression System), Life Technologies (BAC-TO-BAC™), Novagen (Bac Vector System™), Pharmingen and Quantum Biotechnologies). Another insect cell host is the common fruit fly, Drosophila melanogaster, for which a transient or stable plasmid based transfection kit is offered commercially by Invitrogen (The DES™ System).


In some embodiments, cells are transformed with vectors that express the nucleic acids of the invention. Transformation techniques for inserting new genetic material into eukaryotic cells, including animal and plant cells, are well known. Viral vectors may be used for inserting expression cassettes into host cell genomes. Alternatively, the vectors may be transfected into the host cells. Transfection may be accomplished by calcium phosphate precipitation, electroporation, optical transfection, protoplast fusion, impalefection, and hydrodynamic delivery.


In preferred embodiments, the serpin nucleic acids are expressed in mammalian cell lines that are well-known in the art. Exemplary mammalian cell lines include Chinese hamster ovary cells (CHO) and Vero cells. The serpins are recovered using known biochemical and biologics manufacturing techniques. (See, e.g., Lai et al., Pharmaceuticals 6:579-603 (2013), incorporated by reference herein in its entirety.)


Examplary therapeutic Serpin family members are presented in Table 1.













TABLE 1





SERPIN
SOURCE
SEQ ID NO
DEMONSTRATED EFFECTS
DOSE RANGE (per kg)







A1AT
Human
1, 2
Inhibits neutrophil elastase
1,000 mg to 0.5 ng;


(SerpinA1)


and other proteases
500 mM to 10 pM


CrmA
Cowpox
3, 4
Suppressor of IL1 and IL18
1,000 mg to 0.5 ng;



virus


300 mM to 10 pM


Serpin1 or

Arabidopsis

5, 6
Suppressor of metacaspases
2,000 mg to 10 ng;


AtSerpin1 or


and papain-like cysteine
1000 mM to 50 pM


Serpin-ZX


protease


SerpinB9
Human
7, 8
Inhibits Granzyme B
1,000 mg to 0.5 ng;






500 mM to 10 pM


SerpinA3
Human
9, 10, 11
Inhibits cathepsin G
1,000 mg to 0.5 ng;






500 mM to 10 pM


SerpinA4
Human
12-17
Inhibits kallikrein
1,000 mg to 0.5 ng;






500 mM to 10 pM


SerpinA5
Human
18, 19
Inhibits protein C, reduces
1,000 mg to 0.5 ng;





PCI in MS plaques
500 mM to 10 pM


SerpinA9
Human
20-27
Inhibits B cell activation
2,000 mg to 1.5 ng;






700 mM to 50 pM


SerpinA10
Human
28-31
Inhibits protein Z-related
1,000 mg to 0.5 ng;





protease, factors Xa and XIa
500 mM to 10 pM


SerpinA12
Human
32-35
Inhibits Kallikrein-7
1,000 mg to 0.5 ng;






500 mM to 10 pM


SerpinB1
Human
36, 37
Inhibits neutrophil elastase
1,000 mg to 0.5 ng;





and monocytes
500 mM to 10 pM


SerpinB6
Human
38-53
Inhibits cathepsin G
1,000 mg to 0.5 ng;






500 mM to 10 pM


SerpinB9
Human
54, 55
Inhibits cytotoxic granule
2,000 mg to 5 ng;





proteases such as granzyme B
1000 mM to 10 pM


Serping1
Human
56-59
C1 esterase inhibitor
1,000 mg to 0.5 ng;






500 mM to 10 pM


Serpini1
Human
60-63
Inhibits tPA, uPA and plasmin,
2,000 mg to 5 ng;


(neuroserpin)


mutated in dementia
1000 mM to 10 pM









Formulations

The invention provides that any of the serpin compositions disclosed herein are formulated into a pharmaceutical composition. In some embodiments, a serpin and an anti-inflammatory therapeutic compound are the only active ingredients. In other embodiments, they are formulated with one or more additional active ingredients. In preferred embodiments, the combinatorial pharmaceutical compositions comprise steroids such as prednisolone or prednisone, anti-inflammatory compounds such as doxycycline, tetracycline, intravenous immunoglobulin, non-steroidal anti-inflammatory drugs (NSAID) such as celecoxib, indomethacin, naproxen, ibuprofen, acetaminophen, and rofecoxib.


The invention provides pharmaceutical compositions formulated with pharmaceutically acceptable excipients, carriers, diluents or vehicles. In some embodiments, they are formulated in powders, liquids, gels, pastes, suspensions, emulsions, or gaseous forms. In other embodiments, they are formulated into pharmaceutically acceptable dosage forms such as: tablets, capsules, caplets, powders, granules, ointments, crémes, solutions, suspensions, emulsions, suppositories, injections, inhalants, gels, particles, or aerosols. In other embodiments, the formulations are administered as disclosed herein. In other embodiments, serpins are administered in a free form, as pharmaceutically acceptable salts, in a time-release formulation, sequentially in a discrete manner, or in combination with other pharmaceutically active compounds.


In some embodiments, the serpins of the invention are delivered to a patient by intrathecal, intramuscular, intravascular, subcutaneous, intracranial, or intraocular injection. In a preferred embodiment, the serpin is A1AT. In another preferred embodiment, the serpins are provided in liquid and powder formulations at amounts ranging from about 1,000 mg/kg to about 50 ng/kg per dose, depending on the method of administration, potency and use. Some formulations may comprise recombinant serpins from about 1,000 mg to about 50 mg per dose. Some formulations may comprise recombinant serpins from about 75 mg to about 5 mg per dose. Some formulations may comprise recombinant serpins from about 5 mg to about 100 μg per dose. Other formulations may comprise recombinant serpins from about 150 μg to about 8 μg per dose. Yet other formulations comprise recombinant serpins from about 7.5 μg to about 50 ng per dose. In preferred embodiments, the formulated serpin is A1AT.


In other embodiments, the periodicity of dosing varies based on patient needs. In preferred embodiments, the dosing schedule is approximately: multiple times per day, daily, multiple times per week, weekly, bi-weekly, monthly, every 6 weeks, every two months, every three months, every four months, every five months, every 6 months, annually, or on an as-needed basis. In a preferred embodiment, the serpin is A1AT. In a more preferred embodiment 60 mg/kg is administered weekly


In other embodiments, the serpin is a human A1AT that is, for example, Prolastin-C (Grifols USA, Los Angeles, Calif.), Aralast NP (Baxter Healthcare Corp., Westlake Village, Calif.), Glassia (Baxter Healthcare Corp., Westlake Village, Calif.) and Zemaira (CSL Behring, King of Prussia, Pa.). In other embodiments, A1AT formulations derived from human blood comprise liquid and powder formulations at amounts ranging from about 1,000 mg/kg to about 50 ng/kg per dose, depending on the method of administration, potency and use. In other embodiments, the formulations comprise Recombinant A1AT from about 1,000 mg to about 50 mg per dose. In other embodiments, formulations comprise recombinant A1AT from about 75 mg to about 5 mg per dose. In other embodiments, formulations comprise Recombinant A1AT from about 5 mg to about 100 μg per dose. In other embodiments, formulations comprise recombinant A1AT from about 150 μg to about 8 μg per dose. In other embodiments, formulations comprise Recombinant A1AT from about 7.5 μg to about 50 ng per dose.


Exemplary drug formulations of the invention include aqueous solutions, organic solutions, powder formulations, solid formulations and mixed phase formulations.


Pharmaceutical compositions of this invention comprise any of the compounds of the present invention, and pharmaceutically acceptable salts thereof, with any pharmaceutically acceptable carrier, adjuvant or vehicle. Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.


Pharmaceutically acceptable salts retain the desired biological activity of the therapeutic composition without toxic side effects. Examples of such salts are (a) acid addition salts formed with inorganic acids, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid and the like/and salts formed with organic acids such as, for example, acetic acid, trifluoroacetic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tanic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid, naphthalene disulfonic acid, polygalacturonic acid and the like; (b) base addition salts or complexes formed with polyvalent metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium, and the like; or with an organic cation formed from N,N′-dibenzylethylenediamine or ethlenediamine; or (c) combinations of (a) and (b), e.g. a zinc tannate salt and the like.


The pharmaceutical compositions of this invention may be administered by subcutaneous, transdermal, oral, parenteral, inhalation, ocular, topical, rectal, nasal, buccal (including sublingual), vaginal, or implanted reservoir modes. The pharmaceutical compositions of this invention may contain any conventional, non-toxic, pharmaceutically-acceptable carriers, adjuvants or vehicles. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrastemal, intrathecal, intralesional, and intracranial injection or infusion techniques.


Also contemplated, in some embodiments, are pharmaceutical compositions comprising as an active ingredient, therapeutic compounds described herein, or pharmaceutically acceptable salt thereof, in a mixture with a pharmaceutically acceptable, non-toxic component. As mentioned above, such compositions may be prepared for parenteral administration, particularly in the form of liquid solutions or suspensions; for oral or buccal administration, particularly in the form of tablets or capsules; for intranasal administration, particularly in the form of powders, nasal drops, evaporating solutions or aerosols; for inhalation, particularly in the form of liquid solutions or dry powders with excipients, defined broadly; for transdermal administration, particularly in the form of a skin patch or microneedle patch; and for rectal or vaginal administration, particularly in the form of a suppository.


The compositions may conveniently be administered in unit dosage form and may be prepared by any of the methods well-known in the pharmaceutical art, for example, as described in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Co., Easton, Pa. (1985), incorporated herein by reference in its entirety. Formulations for parenteral administration may contain as excipients sterile water or saline alkylene glycols such as propylene glycol, polyalkylene glycols such as polyethylene glycol, saccharides, oils of vegetable origin, hydrogenated napthalenes, serum albumin or other nanoparticles (as used in Abraxane™, American Pharmaceutical Partners, Inc. Schaumburg, Ill.), and the like. For oral administration, the formulation can be enhanced by the addition of bile salts or acylcarnitines. Formulations for nasal administration may be solid or solutions in evaporating solvents such as hydrofluorocarbons, and may contain excipients for stabilization, for example, saccharides, surfactants, submicron anhydrous alpha-lactose or dextran, or may be aqueous or oily solutions for use in the form of nasal drops or metered spray. For buccal administration, typical excipients include sugars, calcium stearate, magnesium stearate, pregelatinated starch, and the like.


Delivery of modified therapeutic compounds described herein to a subject over prolonged periods of time, for example, for periods of one week to one year, may be accomplished by a single administration of a controlled release system containing sufficient active ingredient for the desired release period. Various controlled release systems, such as monolithic or reservoir-type microcapsules, depot implants, polymeric hydrogels, osmotic pumps, vesicles, micelles, liposomes, transdermal patches, iontophoretic devices and alternative injectable dosage forms may be utilized for this purpose. Localization at the site to which delivery of the active ingredient is desired is an additional feature of some controlled release devices, which may prove beneficial in the treatment of certain disorders.


In certain embodiments for transdermal administration, delivery across the barrier of the skin would be enhanced using electrodes (e.g. iontophoresis), electroporation, or the application of short, high-voltage electrical pulses to the skin, radiofrequencies, ultrasound (e.g. sonophoresis), microprojections (e.g. microneedles), jet injectors, thermal ablation, magnetophoresis, lasers, velocity, or photomechanical waves. The drug can be included in single-layer drug-in-adhesive, multi-layer drug-in-adhesive, reservoir, matrix, or vapor style patches, or could utilize patchless technology. Delivery across the barrier of the skin could also be enhanced using encapsulation, a skin lipid fluidizer, or a hollow or solid microstructured transdermal system (MTS, such as that manufactured by 3M), jet injectors. Additives to the formulation to aid in the passage of therapeutic compounds through the skin include prodrugs, chemicals, surfactants, cell penetrating peptides, permeation enhancers, encapsulation technologies, enzymes, enzyme inhibitors, gels, nanoparticles and peptide or protein chaperones.


One form of controlled-release formulation contains the therapeutic compound or its salt dispersed or encapsulated in a slowly degrading, non-toxic, non-antigenic polymer such as copoly(lactic/glycolic) acid, as described in the pioneering work of Kent et al., U.S. Pat. No. 4,675,189, incorporated by reference herein. The compounds, or their salts, may also be formulated in cholesterol or other lipid matrix pellets, or silastomer matrix implants. Additional slow release, depot implant or injectable formulations will be apparent to the skilled artisan. See, for example, Sustained and Controlled Release Drug Delivery Systems, JR Robinson ed., Marcel Dekker Inc., New York, 1978; and Controlled Release of Biologically Active Agents, R W Baker, John Wiley & Sons, New York, 1987. The foregoing are incorporated by reference in their entirety.


An additional form of controlled-release formulation comprises a solution of biodegradable polymer, such as copoly(lactic/glycolic acid) or block copolymers of lactic acid and PEG, is a bioacceptable solvent, which is injected subcutaneously or intramuscularly to achieve a depot formulation. Mixing of the therapeutic compounds described herein with such a polymeric formulation is suitable to achieve very long duration of action formulations.


When formulated for nasal administration, the absorption across the nasal mucous membrane may be further enhanced by surfactants, such as, for example, glycocholic acid, cholic acid, taurocholic acid, ethocholic acid, deoxycholic acid, chenodeoxycholic acid, dehdryocholic acid, glycodeoxycholic acid, cycledextrins and the like in an amount in the range of between about 0.1 and 15 weight percent, between about 0.5 and 4 weight percent, or about 2 weight percent. An additional class of absorption enhancers reported to exhibit greater efficacy with decreased irritation is the class of alkyl maltosides, such as tetradecylmaltoside (Arnold, J J et al., 2004, J Pharm Sci 93: 2205-13; Ahsan, F et al., 2001, Pharm Res 18:1742-46) and references therein, all of which are hereby incorporated by reference.


The pharmaceutical compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant such as Ph. Helv or a similar alcohol.


The pharmaceutical compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspensions and solutions. In the case of tablets for oral use, carriers that are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.


The pharmaceutical compositions of this invention may also be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient that is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.


Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application. For application topically to the skin, the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. The pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topical transdermal patches are also included in this invention.


The pharmaceutical compositions of this invention may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.


When formulated for delivery by inhalation, a number of formulations offer advantages. Adsorption of the therapeutic compound to readily dispersed solids such as diketopiperazines (for example, Technosphere particles (Pfutzner, A and Forst, T, 2005, Expert Opin Drug Deliv 2:1097-1106) or similar structures gives a formulation that results in rapid initial uptake of the therapeutic compound. Lyophilized powders, especially glassy particles, containing the therapeutic compound and an excipient are useful for delivery to the lung with good bioavailability, for example, see Exubera® (inhaled insulin, Pfizer, Inc. and Aventis Pharmaceuticals Inc.) and Afrezza® (inhaled insulin, Mannkind, Corp.).


Dosage levels of between about 0.01 and about 100 mg/kg body weight per day, preferably 0.5 and about 50 mg/kg body weight per day of the active ingredient compound are useful in the prevention and treatment of disease. Such administration can be used as a chronic or acute therapy. The amount of drug that may be combined with the carrier to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. A typical preparation will contain from about 5% to about 95% active compound (w/w). Preferably, such preparations contain from about 20% to about 80% active compound.


Upon improvement of a patient's condition, a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level, treatment should cease. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.


As the skilled artisan will appreciate, lower or higher doses than those recited above may be required. Specific dosage and treatment regimens for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, gender, diet, time of administration, rate of excretion, drug combination, the severity and course of an infection, the patient's disposition to the infection and the judgment of the treating physician.


The carrier-drug conjugates described herein provide advantages to drug manufacturers and patients over unmodified drugs. Specifically, the carrier-drug conjugate or formulation will be a more potent, longer lasting, and require smaller and less frequent dosing. This translates into lowered healthcare costs and more convenient drug administration schedules for patients. The carrier-drug conjugates can also provide subcutaneous or transdermal routes of administration as alternatives to intravenous injection. These routes can be self-administered by patients and thus improve patient compliance.


In order that the invention described herein may be more fully understood, the following examples are set forth. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting this invention in any manner.


Example 1—A1AT Reduces NMO Disease in a Mouse Model

C57BL/6 donor mice were immunized via subcutaneous injection with MOG-35-55 peptide in complete Freund's Adjuvant. This induces Experimental Autoimmune Encephalomyelitis (EAE), an animal model for progressive multiple sclerosis. (Miller et al., Curr. Protoc. Immunol., Chapter Unit 15.1 (2007); incorporated herein by reference in its entirety). The mice were boosted twice via intraperitoneal injection with Pertussis toxin on day 0, 2, and 7 days post-immunization. Mice were sacrificed and spleens and lymph nodes collected and resuspended into single cell suspensions. Cells were cultured with MOG peptide and IL-23 to increase Th17 skewing and cultured for 3 days. Cells were harvested and approximately 15×10̂6 cells were adoptively transferred into recipient C57BL/6 mice via intraperitoneal injection. Starting day 5 for 2 weeks until day 18, mice were administered daily via intraperitoneal injection with 100 uL of PBS or A1AT (2 mg A1AT formulated in PBS). Clinical scores were measured each day at approximately the same time of day. Clinical scores measure ascending paralysis. A score of 1 is a limp tail. A score of 2 includes hind limb weakness. A score of 3 includes hind limb paralysis. A score of 4 includes forelimb weakness. A score of 5 is indicated by death. (See Miller et al.). A1AT showed a significant improvement when compared to PBS treatment. n=10 mice per group.


Example 2—A1AT Reduces NMO Disease in a Mouse Model

Blood-derived human A1AT was compared to Sivelestat in a murine model of NMO. A less severe disease was induced when 12×106 Th17-polarized cells were adoptively transferred i.p. into recipient C57BL/6 mice. To induce Th17-polarized cells, C57BL/6 mice were induced with MOG35-55/CFA. 11 days later, their spleen cells were harvested and restimulated in culture with MOG35-55 peptide (Miller et al., supra) for 3 days.


Mice received PBS, Sivelestat (0.5 mg), or A1AT (2 mg) as daily doses administered i.p. starting on Day 5 post-transfer until Day 18 post-transfer. There were 10 recipient mice per cohort. Error bars represent SEM. Points where A1AT compared to PBS and mean disease scores were significant (p<0.05) are indicated (*). A1AT treatment reduced the NMO disease score compared to PBS (control) and was superior to Sivelestat at all days of overt signs of disease as well as delay onset of disease (see FIG. 2).


Example 3—A1AT+Methylprednisolone Reduces NMO Disease in a Mouse Model

A more severe disease was induced by adoptively transferring a higher number of pathogenic Th17-polarized cells in order to demonstrate the additional efficacy from a combination treatment of A1AT plus methylprednisolone (Mpred) compared to individual treatments using a blinded study.


25×106 cells prepared as above were adoptively transferred i.p. into recipient C57BL/6 mice. Mice received PBS, Mpred (0.5 mg), AAT (2 mg), or both AAT+Mpred as daily doses administered i.p. starting on Day 4 post-transfer until Day 16 post-transfer. Ten recipient mice per cohort. Table 2 shows the Mean day of disease onset+/−SEM and p-value determined by the two-tailed Student's t-test; the end disease score at Day 17 post-transfer+/−SD, p-value determined by Mann-Whitney U test; and the mean maximum disease score (MMS)+/−SD, p-value by Mann-Whitney U test. The bold p-values indicate significant (p<0.05) differences between the treatments when compared to PBS (control).


The AAT+Mpred combination demonstrated superior and significant delay of disease onset, showed improved disease end scores equivalent to Mpred alone, and improved mean maximum disease score (MMS) equivalent to Mpred alone.















TABLE 2






Mean Day of







Treatment
Onset +/− SEM
p-value
End Score
p-value
MMS +/− SD
p-value







PBS
7.9 +/− 0.3

3.00 +/− 1.11

3.15 +/− 1.11



Mpred
8.5 +/− 0.4
0.2520
2.25 +/− 0.42

0.0039

2.95 +/− 0.44

0.0228



A1AT
8.4 +/− 0.4
0.3110
3.10 +/− 0.57
0.6890
3.50 +/− 0.00
0.3173


A1AT + Mpred
9.3 +/− 0.5

0.0164

2.15 +/− 1.00

0.0150

2.70 +/− 1.06

0.0447










Mean disease scores showed that A1AT+Mpred treatment reduced the disease over all days measured more than either treatment alone (see FIG. 3). AAT alone showed early efficacy compared to Mpred alone and PBS (control) on Day 5 and 6 but afterwards did not show therapeutic effect. While not being bound to theory, this may be due to anti-drug immunogenicity of the human AAT in mouse that is known to develop (70) that may not have developed in our previous less severe disease induction, and where the Mpred in combination with AAT treatment may have diminished the anti-drug immunogenicity effect.


Histological analysis showed that A1AT+Mpred and A1AT alone reduced the number of inflammatory foci compared to Mpred alone. Additionally, A1AT+Mpred similarly reduced demyelination and the number of apoptotic cells when compared to Mpred alone.















TABLE 3






Inflammatory

Demyelination

Apoptotic



Treatment
foci +/− SD
p-value
(H + E)
p-value
Cells +/− SD
p-value





















PBS
3.6 +/− 0.5

1.8 +/− 0.2

2.9 +/− 0.9



Mpred
3.0 +/− 0.9
0.2856
1.2 +/− 0.3

0.0325

1.3 +/− 0.9

0.0393



A1AT
2.0 +/− 0.5

0.0037

1.8 +/− 0.4
1.000
2.5 +/− 0.3
0.4262


A1AT + Mpred
2.5 +/− 0.6

0.0369

1.3 +/− 0.3

0.0360

1.5 +/− 0.3

0.0273










Table 3 shows histopathology scores in a blinded study. An NMO mouse model as described above was used. Spines were collected from 4 mice per treatment cohort on Day 18 and prepared for histology. Sections from cervical, thoracic and lumbar spine regions were scored and averaged. Table 3 shows the number of inflammatory foci per section >20 cells+/−SD, p-value by two-tailed Student's t-test; demyelination scoring of hematoxylin and eosin stained section+/−SD, p-value by Mann-Whitney U test; and apoptotic cell counts+/−SD, p-value by two-tailed Student's t-test. Bold formatting indicates significant (p<0.05) difference of treatment compared to PBS (control).


Body weight loss results from, and is a prognostic measurement of, autoimmune diseases such as NMO. A1AT+Mpred resulted in less body weight loss than either treatment alone (see FIG. 4). Ten recipient mice per cohort were used in this blinded study. The graph shows mean body weight as a percentage of the value measured at day 0+/−SEM. Interestingly, Mpred treatment alone appeared to cause early weight loss from Day 5 to Day 7. While not being bound to theory, this suggests the possibility that weight loss is a side effect of the steroid rather than NMO disease. At later times, the Mpred cohort weight loss was similar to PBS (control) even though Mpred had a lower disease score. Thus, Mpred toxicity side effects may be ameliorated by the AAT+Mpred combination cohort.


Example 4—A1AT+Methylprednisolone Reduces Autoimmune Cytokine Profile

Ex vivo cytokine measurements from spleen cells of Th17-polarized cell transplanted mice restimulated with MOG peptide were conducted to survey effector and memory recall responses of the immune cell compartment, primarily from T cells. Spleens were collected from 6 mice per treatment cohort on Day 18. Single cell suspensions were prepared and cultured with the indicated amounts of MOG35-55 peptide. Supernatants were collected after 3 days of culture and IL-17A, IFN-γ, IL-6, or IL-2 measured by cytokine bead assay (BD Biosciences, San Jose, Calif., Cat. No. 560485). The results in this blinded study were expressed in cytokine pg/mL+/−SEM.


Consistent with the mouse NMO model, A1AT alone and A1AT+Mpred decreased the inflammatory cytokines IL-17A compared to PBS and Mpred alone (FIG. 5A), and decreased IFN-γ when compared to Mpred alone (FIG. 5B). All treatments resulted in increased IL-6 compared to PBS (FIG. 5C). IL-2 levels were higher without antigenic restimulation (0 MOG peptide) when treated with A1AT alone or A1AT+Mpred when compared to Mpred alone (FIG. 5D). IL-2 promotes T cell proliferation but is also a key cytokine for the maintenance of regulatory T cells in vivo. TNF-α levels were similar and IL-4 levels were at the borderline limit of detection in all four cohorts (data not shown). These cytokine restimulation measurements show that AAT and Mpred treatments resulted in differential immunomodulation of NMO disease that helps explain their additional therapeutic efficacy when used in combination.


Exemplary Sequences










Sequence ID No. 1



AlAT (Human) Protein



MPSSVSWGIL LLAGLCCLVP VSLAEDPQGD AAQKTDTSHH DQDHPTFNKI





TPNLAEFAFS LYRQLAHQSN STNIFFSPVS IATAFAMLSL GTKADTHDEI





LEGLNFNLTE IPEAQIHEGF QELLRTLNQP DSQLQLTTGN GLFLSEGLKL





VDKFLEDVKK LYHSEAFTVN FGDTEEAKKQ INDYVEKGTQ GKIVDLVKEL





DRDTVFALVN YIFFKGKWER PFEVKDTEEE DFHVDQVTTV KVPMMKRLGM





FNIQHCKKLS SWVLLMKYLG NATAIFFLPD EGKLQHLENE LTHDIITKFL





ENEDRRSASL HLPKLSITGT YDLKSVLGQL GITKVFSNGA DLSGVTEEAP





LKLSKAVHKA VLTIDEKGTE AAGAMFLEAI PMSIPPEVKF NKPFVFLMIE





QNTKSPLFMG KVVNPTQK





SEQ ID No. 2



AlAT (Human) cDNA



ATGCCGTCTTCTGTCTCGTGGGGCATCCTCCTGCTGGCAGGCCTGTGCTGCCT





GGTCCCTGTCTCCCTGGCTGAGGATCCCCAGGGAGATGCTGCCCAGAAGACA





GATACATCCCACCATGATCAGGATCACCCAACCTTCAACAAGATCACCCCCA





ACCTGGCTGAGTTCGCCTTCAGCCTATACCGCCAGCTGGCACACCAGTCCAAC





AGCACCAATATCTTCTTCTCCCCAGTGAGCATCGCTACAGCCTTTGCAATGCT





CTCCCTGGGGACCAAGGCTGACACTCACGATGAAATCCTGGAGGGCCTGAAT





TTCAACCTCACGGAGATTCCGGAGGCTCAGATCCATGAAGGCTTCCAGGAAC





TCCTCCGTACCCTCAACCAGCCAGACAGCCAGCTCCAGCTGACCACCGGCAA





TGGCCTGTTCCTCAGCGAGGGCCTGAAGCTAGTGGATAAGTTTTTGGAGGATG





TTAAAAAGTTGTACCACTCAGAAGCCTTCACTGTCAACTTCGGGGACACCGA





AGAGGCCAAGAAACAGATCAACGATTACGTGGAGAAGGGTACTCAAGGGAA





AATTGTGGATTTGGTCAAGGAGCTTGACAGAGACACAGTTTTTGCTCTGGTGA





ATTACATCTTCTTTAAAGGCAAATGGGAGAGACCCTTTGAAGTCAAGGACAC





CGAGGAAGAGGACTTCCACGTGGACCAGGTGACCACCGTGAAGGTGCCTATG





ATGAAGCGTTTAGGCATGTTTAACATCCAGCACTGTAAGAAGCTGTCCAGCTG





GGTGCTGCTGATGAAATACCTGGGCAATGCCACCGCCATCTTCTTCCTGCCTG





ATGAGGGGAAACTACAGCACCTGGAAAATGAACTCACCCACGATATCATCAC





CAAGTTCCTGGAAAATGAAGACAGAAGGTCTGCCAGCTTACATTTACCCAAA





CTGTCCATTACTGGAACCTATGATCTGAAGAGCGTCCTGGGTCAACTGGGCAT





CACTAAGGTCTTCAGCAATGGGGCTGACCTCTCCGGGGTCACAGAGGAGGCA





CCCCTGAAGCTCTCCAAGGCCGTGCATAAGGCTGTGCTGACCATCGACGAGA





AAGGGACTGAAGCTGCTGGGGCCATGTTTTTAGAGGCCATACCCATGTCTATC





CCCCCCGAGGTCAAGTTCAACAAACCCTTTGTCTTCTTAATGATTGAACAAAA





TACCAAGTCTCCCCTCTTCATGGGAAAAGTGGTGAATCCCACCCAAAAATAA





SEQ ID No. 3



CrmA (Cowpox virus) Protein



MDIFREIASSMKGENVFISPPSISSVLTILYYGANGSTAEQLSKYVEKEADKNKDDI





SFKSMNKVYGRYSAVFKDSFLRKIGDNFQTVDFTDCRTVDAINKCVDIFTEGKIN





PLLDEPLSPDTCLLAISAVYFKAKWLMPFEKEFTSDYPFYVSPTEMVDVSMMSM





YGEAFNHASVKESFGNFSIIELPYVGDTSMVVILPDNIDGLESIEQNLTDTNFKKW





CDSMDAMFIDVHIPKFKVTGSYNLVDALVKLGLTEVFGSTGDYSNMCNSDVSVD





AMIHKTYIDVNEEYTEAAAATCALVADCASTVTNEFCADHPFIYVIRHVDGKILF





VGRYCSPTTN





SEQ ID No. 4



CrmA (Cowpox virus) cDNA



ATGGATATCTTCAGGG AAATCGCATC TTCTATGAAA GGAGAGAATG





TATTCATTTC TCCACCGTCAATCTCGTCAG TATTGACAAT ACTGTATTAT





GGAGCTAATG GATCCACTGC TGAACAGCTATCAAAATATG TAGAAAAGGA





GGCGGACAAG AATAAGGATG ATATCTCATT CAAGTCCATGAATAAAGTAT





ATGGGCGATA TTCTGCAGTG TTTAAAGATT CCTTTTTGAG AAAAATTGGA





GATAATTTCC AAACTGTTGA CTTCACTGAT TGTCGCACTG TAGATGCGAT





CAACAAGTGTGTTGATATCT TCACTGAGGG GAAAATTAAT CCACTATTGG





ATGAACCATT GTCTCCAGATACCTGTCTCC TAGCAATTAG TGCCGTATAC





TTTAAAGCAA AATGGTTGAT GCCATTTGAAAAGGAATTTA CCAGTGATTA





TCCCTTTTAC GTATCTCCAA CGGAAATGGT AGATGTAAGTATGATGTCTA





TGTACGGCGA GGCATTTAAT CACGCATCTG TAAAAGAATC ATTCGGCAAC





TTTTCAATCA TAGAACTGCC ATATGTTGGA GATACTAGTA TGGTGGTAAT





TCTTCCAGACAATATTGATG GACTAGAATC CATAGAACAA AATCTAACAG





ATACAAATTT TAAGAAATGGTGTGACTCTA TGGATGCTAT GTTTATCGAT





GTGCACATTC CCAAGTTTAA GGTAACAGGCTCGTATAATC TGGTGGATGC





GCTAGTAAAG TTGGGACTGA CAGAGGTGTT CGGTTCAACTGGAGATTATA





GCAATATGTG TAATTCAGAT GTGAGTGTCG ACGCTATGAT CCACAAAACG





TATATAGATG TCAATGAAGA GTATACAGAA GCAGCTGCAG CAACTTGTGC





GCTGGTGGCAGACTGTGCAT CAACAGTTAC AAATGAGTTC TGTGCAGATC





ATCCGTTCAT CTATGTGATTAGGCATGTCG ATGGCAAAAT TCTTTTCGTT





GGTAGATATT GCTCTCCAAC AACTAATTAA





SEQ ID No. 5



Serpin1 (Arabidopsis) Protein



MDVRESISLQ NQVSMNLAKH VITTVSQNSN VIFSPASINV VLSIIAAGSA





GATKDQILSFLKFSSTDQLN SFSSEIVSAV LADGSANGGP KLSVANGAWI





DKSLSFKPSF KQLLEDSYKAASNQADFQSK AVEVIAEVNS WAEKETNGLI





TEVLPEGSAD SMTKLIFANA LYFKGTWNEKFDESLTQEGE FHLLDGNKVT





APFMTSKKKQ YVSAYDGFKV LGLPYLQGQD KRQFSMYFYLPDANNGLSDL





LDKIVSTPGF LDNHIPRRQV KVREFKIPKF KFSFGFDASN VLKGLGLTSP





FSGEEGLTEM VESPEMGKNL CVSNIFHKAC IEVNEEGTEA AAASAGVIKL





RGLLMEEDEIDFVADHPFLL VVTENITGVV LFIGQVVDPL H





SEQ ID No. 6



Serpin1 (Arabidopsis) DNA



CGTCTTCTCC TAAACCCAGC AAATTCGTTT ACCAGTCATC ACCACCACAA





CCTCCGGCGA AAATGGACGT GCGTGAATCA ATCTCACTGC AAAACCAAGT





CTCCATGAAT CTCGCAAAAC ACGTAATCAC CACCGTCTCT CAAAACTCCA





ACGTCATCTT CTCACCGGCT TCAATCAACG TCGTACTCAG TATAATCGCC





GCTGGATCCG CCGGCGCTAC CAAAGATCAG ATCCTCTCGT TTCTCAAATT





CTCTTCCACT GATCAACTTA ATTCATTCTC TTCCGAAATC GTCTCCGCTG





TTCTCGCTGA CGGTAGTGCT AACGGTGGTC CTAAGCTCTC GGTGGCTAAT





GGCGCCTGGA TCGATAAGTC TCTCTCCTTT AAACCTTCCT TTAAACAGCT





CTTGGAAGAT TCGTATAAAG CTGCTTCGAA TCAAGCTGAT TTTCAATCGA





AGGCTGTGGA GGTGATTGCT GAAGTGAATT CATGGGCTGA AAAGGAGACA





AATGGTCTCA TCACTGAGGT TCTTCCAGAA GGATCAGCTG ATAGTATGAC





CAAACTGATA TTTGCAAATG CATTGTACTT CAAGGGAACA TGGAACGAGA





AATTCGATGA GTCGTTAACA CAAGAAGGCG AGTTTCACCT TCTTGACGGT





AACAAAGTGA CTGCACCATT CATGACCAGC AAGAAGAAAC AATACGTAAG





TGCTTACGAT GGTTTCAAAG TTTTGGGACT TCCTTACTTA CAAGGACAGG





ATAAGCGACA ATTCTCCATG TACTTTTATC TTCCCGATGC AAACAACGGA





CTGTCTGATC TTCTGGACAA AATAGTTTCC ACTCCTGGGT TCTTAGACAA





CCACATCCCA CGCAGACAAG TTAAAGTCCG CGAATTCAAG ATTCCAAAGT





TTAAATTCTC TTTCGGGTTC GATGCTTCAA ATGTTTTAAA AGGATTGGGA





CTGACTTCGC CTTTCAGCGG TGAAGAAGGT TTAACTGAGA TGGTTGAATC





TCCTGAGATG GGGAAGAATC TATGCGTATC GAACATTTTC CATAAAGCGT





GTATCGAAGT GAATGAAGAA GGAACAGAAG CTGCAGCTGC ATCAGCTGGA





GTTATAAAGC TAAGAGGATT GCTTATGGAG GAAGATGAAA TAGATTTTGT





TGCAGACCAT CCGTTTCTAT TGGTGGTCAC AGAGAACATA ACAGGAGTGG





TTCTGTTCAT TGGCCAAGTT GTTGATCCGT TGCATTAATC TAAAGCTAAT





GTGGAAGTTT TTGGTTTTAC TTAAAATAAA TGAGTCATTG GTTTTGAGGA





CTCATCTTTA TGTAACATCC TTTGTCTTGA CTCTTTGATG TGTGTAAGAA





TAATAGTGAT ACATAACAGC TTTTCTTCTG TATTTGGATC ACATGTACTG





AACTGATAGA CATACATACA TGTGATGCAT CTTAATGATT CACTGT





SEQ ID No. 7



SerpinB9 (Human) protein



METLSNASGTFAIRLLKILCQDNPSHNVFCSPVSISSALAMVLLGAKGNTATQMA





QALSLNTEEDIHRAFQSLLTEVNKAGTQYLLRTANRLFGEKTCQFLSTFKESCLQF





YHAELKELSFIRAAEESRKHINTWVSKKTEGKIEELLPGSSIDAETRLVLVNAIYFK





GKWNEPFDETYTREMPFKINQEEQRPVQMMYQEATFKLAHVGEVRAQLLELPY





ARKELSLLVLLPDDGVELSTVEKSLTFEKLTAWTKPDCMKSTEVEVLLPKFKLQE





DYDMESVLRHLGIVDAFQQGKADLSAMSAERDLCLSKFVHKSFVEVNEEGTEAA





AASSCFVVAECCMESGPRFCADHPFLFFIRHNRANSILFCGRFSSP





SEQ ID No. 8



SerpinB9 (Human) cDNA



ATGGAAACTC TTTCTAATGC AAGTGGTACT TTTGCCATAC GCCTTTTAAA





GATACTGTGTCAAGATAACC CTTCGCACAA CGTGTTCTGT TCTCCTGTGA





GCATCTCCTC TGCCCTGGCCATGGTTCTCC TAGGGGCAAA GGGAAACACC





GCAACCCAGA TGGCCCAGGC ACTGTCTTTAAACACAGAGG AAGACATTCA





TCGGGCTTTC CAGTCGCTTC TCACTGAAGT GAACAAGGCTGGCACACAGT





ACCTGCTGAG AACGGCCAAC AGGCTCTTTG GAGAGAAAAC TTGTCAGTTC





CTCTCAACGT TTAAGGAATC CTGTCTTCAA TTCTACCATG CTGAGCTGAA





GGAGCTTTCCTTTATCAGAG CTGCAGAAGA GTCCAGGAAA CACATCAACA





CCTGGGTCTC AAAAAAGACCGAAGGTAAAA TTGAAGAGTT GTTGCCGGGT





AGCTCAATTG ATGCAGAAAC CAGGCTGGTTCTTGTCAATG CCATCTACTT





CAAAGGAAAG TGGAATGAAC CGTTTGACGA AACATACACAAGGGAAATGC





CCTTTAAAAT AAACCAGGAG GAGCAAAGGC CAGTGCAGAT GATGTATCAG





GAGGCCACGT TTAAGCTCGC CCACGTGGGC GAGGTGCGCG CGCAGCTGCT





GGAGCTGCCCTACGCCAGGA AGGAGCTGAG CCTGCTGGTG CTGCTGCCTG





ACGACGGCGT GGAGCTCAGCAAGAGTACTG AGGTTGAAGT TCTCCTTCCA





AAATTTAAAC TACAAGAGGA TTATGACATGGAATCTGTGC TTCGGCATTT





GGGAATTGTT GATGCCTTCC AACAGGGCAA GGCTGACTTGTCGGCAATGT





CAGCGGAGAG AGACCTGTGT CTGTCCAAGT TCGTGCACAA GAGTTTTGTG





GAGGTGAATG AAGAAGGCAC CGAGGCAGCG GCAGCGTCGA GCTGCTTTGT





AGTTGCAGAGTGCTGCATGG AATCTGGCCC CAGGTTCTGT GCTGACCACC





CTTTCCTTTT CTTCATCAGGCACAACAGAG CCAACAGCAT TCTGTTCTGT





GGCAGGTTCT CATCGCCA





SEQ ID No. 9



SerpinA3 (Human) Protein



MERMLPLLTL GLLAAGFCPA VLCHPNSPLD EENLTQENQD RGTHVDLGLA





SANVDFAFSLYKQSPRWSIR LCLMYLRRAQ KHLLPQQSKS PSFLH





SEQ ID No. 10



SerpinA3 (Human) Protein Precursor



MERMLPLLALGLLAAGFCPAVLCHPNSPLDEENLTQENQDRGTHVDLGLASANV





DFAFSLYKQLVLKAPDKNVIFSPLSISTALAFLSLGAHNTTLTEILKGLKFNLTETS





EAEIHQSFQHLLRTLNQSSDELQLSMGNAMFVKEQLSLLDRFTEDAKRLYGSEAF





ATDFQDSAAAKKLINDYVKNGTRGKITDLIKDLDSQTMMVLVNYIFFKAKWEMP





FDPQDTHQSRFYLSKKKWVMVPMMSLHHLTIPYFRDEELSCTVVELKYTGNASA





LFILPDQDKMEEVEAMLLPETLKRWRDSLEFREIGELYLPKFSISRDYNLNDILLQ





LGIEEAFTSKADLSGITGARNLAVSQVVHKAVLDVFEEGTEASAATAVKITLLSA





LVETRTIVRFNRPFLMIIVPTDTQNIFFMSKVTNPKQA





SEQ ID No. 11



SerpinA3 (Human) cDNA precursor



ATGGAGAGAAT GTTACCTCTC CTGGCTCTGG GGCTCTTGGC GGCTGGGTTC





TGCCCTGCTG TCCTCTGCCA CCCTAACAGC CCACTTGACG AGGAGAATCT





GACCCAGGAG AACCAAGACC GAGGGACACA CGTGGACCTC GGATTAGCCT





CCGCCAACGT GGACTTCGCT TTCAGCCTGT ACAAGCAGTT AGTCCTGAAG





GCCCCTGATA AGAATGTCAT CTTCTCCCCA CTGAGCATCT CCACCGCCTT





GGCCTTCCTG TCTCTGGGGG CCCATAATAC CACCCTGACA GAGATTCTCA





AAGGCCTCAA GTTCAACCTC ACGGAGACTT CTGAGGCAGA AATTCACCAG





AGCTTCCAGC ACCTCCTGCG CACCCTCAAT CAGTCCAGCG ATGAGCTGCA





GCTGAGTATG GGAAATGCCA TGTTTGTCAA AGAGCAACTC AGTCTGCTGG





ACAGGTTCAC GGAGGATGCC AAGAGGCTGT ATGGCTCCGA GGCCTTTGCC





ACTGACTTTC AGGACTCAGC TGCAGCTAAG AAGCTCATCA ACGACTACGT





GAAGAATGGA ACTAGGGGGA AAATCACAGA TCTGATCAAG GACCTTGACT





CGCAGACAAT GATGGTCCTG GTGAATTACA TCTTCTTTAA AGCCAAATGG





GAGATGCCCT TTGACCCCCA AGATACTCAT CAGTCAAGGT TCTACTTGAG





CAAGAAAAAG TGGGTAATGG TGCCCATGAT GAGTTTGCAT CACCTGACTA





TACCTTACTT CCGGGACGAG GAGCTGTCCT GCACCGTGGT GGAGCTGAAG





TACACAGGCA ATGCCAGCGC ACTCTTCATC CTCCCTGATC AAGACAAGAT





GGAGGAAGTG GAAGCCATGC TGCTCCCAGA GACCCTGAAG CGGTGGAGAG





ACTCTCTGGA GTTCAGAGAG ATAGGTGAGC TCTACCTGCC AAAGTTTTCC





ATCTCGAGGG ACTATAACCT GAACGACATA CTTCTCCAGC TGGGCATTGA





GGAAGCCTTC ACCAGCAAGG CTGACCTGTC AGGGATCACA GGGGCCAGGA





ACCTAGCAGT CTCCCAGGTG GTCCATAAGG CTGTGCTTGA TGTATTTGAG





GAGGGCACAG AAGCATCTGC TGCCACAGCA GTCAAAATCA CCCTCCTTTC





TGCATTAGTG GAGACAAGGA CCATTGTGCG TTTCAACAGG CCCTTCCTGA





TGATCATTGT CCCTACAGAC ACCCAGAACA TCTTCTTCAT GAGCAAAGTC





ACCAATCCCA AGCAAGCCTA G





SEQ ID No. 12



SerpinA4 transcript variant 3 (Human) Protein



MHLIDYLLLLLVGLLALSHGQLHVEHDGESCSNSSHQQILETGEGSPSLKIAPANA





DFAFRFYYLIASETPGKNIFFSPLSISAAYAMLSLGACSHSRSQILEGLGFNLTELSE





SDVHRGFQHLLHTLNLPGHGLETRVGSALFLSHNLKFLAKFLNDTMAVYEAKLF





HTNFYDTVGTIQLINDHVKKETRGKIVDLVSELKKDVLMVLVNYIYFKALWEKP





FISSRTTPKDFYVDENTTVRVPMMLQDQEHHWYLHDRYLPCSVLRMDYKGDAT





VFFILPNQGKMREIEEVLTPEMLMRWNNLLRKRNFYKKLELHLPKFSISGSYVLD





QILPRLGFTDLFSKWADLSGITKQQKLEASKSFHKATLDVDEAGTEAAAATSFAI





KFFSAQTNRHILRFNRPFLVVIFSTSTQSVLFLGKVVDPTKP





SEQ ID No. 13



SerpinA4 transcript variant 3 (Human) cDNA



ATGCATCT TATCGACTAC CTGCTCCTCC TGCTGGTTGG ACTACTGGCC





CTTTCTCATG GCCAGCTGCA CGTTGAGCAT GATGGTGAGA GTTGCAGTAA





CAGCTCCCAC CAGCAGATTC TGGAGACAGG TGAGGGCTCC CCCAGCCTCA





AGATAGCCCC TGCCAATGCT GACTTTGCCT TCCGCTTCTA CTACCTGATC





GCTTCGGAGA CCCCGGGGAA GAACATCTTT TTCTCCCCGC TGAGCATCTC





GGCGGCCTAC GCCATGCTTT CCCTGGGGGC CTGCTCACAC AGCCGCAGCC





AGATCCTTGA GGGCCTGGGC TTCAACCTCA CCGAGCTGTC TGAGTCCGAT





GTCCATAGGG GCTTCCAGCA CCTCCTGCAC ACTCTCAACC TCCCCGGCCA





TGGGCTGGAA ACACGCGTGG GCAGTGCTCT GTTCCTGAGC CACAACCTGA





AGTTCCTTGC AAAATTCCTG AATGACACCA TGGCCGTCTA TGAGGCTAAA





CTCTTCCACA CCAACTTCTA CGACACTGTG GGCACAATCC AGCTTATCAA





CGACCACGTC AAGAAGGAAA CTCGAGGGAA GATTGTGGAT TTGGTCAGTG





AGCTCAAGAA GGACGTCTTG ATGGTGCTGG TGAATTACAT TTACTTCAAA





GCCCTGTGGG AGAAACCATT CATTTCCTCA AGGACCACTC CCAAAGACTT





CTATGTTGAT GAGAACACAA CAGTCCGGGT GCCCATGATG CTGCAGGACC





AGGAGCATCA CTGGTATCTT CATGACAGAT ACTTGCCCTG CTCGGTGCTA





CGGATGGATT ACAAAGGAGA CGCAACCGTG TTTTTCATTC TCCCTAACCA





AGGCAAAATG AGGGAGATTG AAGAGGTTCT GACTCCAGAG ATGCTAATGA





GGTGGAACAA CTTGTTGCGG AAGAGGAATT TTTACAAGAA GCTAGAGTTG





CATCTTCCCA AGTTCTCCAT TTCTGGCTCC TATGTATTAG ATCAGATTTT





GCCCAGGCTG GGCTTCACGG ATCTGTTCTC CAAGTGGGCT GACTTATCCG





GCATCACCAA ACAGCAAAAA CTGGAGGCAT CCAAAAGTTT CCACAAGGCC





ACCTTGGACG TGGATGAGGC TGGCACCGAG GCTGCAGCAG CCACCAGCTT





CGCGATCAAA TTCTTCTCTG CCCAGACCAA TCGCCACATC CTGCGATTCA





ACCGGCCCTT CCTTGTGGTG ATCTTTTCCA CCAGCACCCA GAGTGTCCTC





TTTCTGGGCA AGGTCGTCGA CCCCACGAAA CCATAG





SEQ ID No. 14



SerpinA4 transcript variant 2 (Human) Protein



MHLIDYLLLLLVGLLALSHGQLHVEHDGESCSNSSHQQILETGEGSPSLKIAPANA





DFAFRFYYLIASETPGKNIFFSPLSISAAYAMLSLGACSHSRSQILEGLGFNLTELSE





SDVHRGFQHLLHTLNLPGHGLETRVGSALFLSHNLKFLAKFLNDTMAVYEAKLF





HTNFYDTVGTIQLINDHVKKETRGKIVDLVSELKKDVLMVLVNYIYFKALWEKP





FISSRTTPKDFYVDENTTVRVPMMLQDQEHHWYLHDRYLPCSVLRMDYKGDAT





VFFILPNQGKMREIEEVLTPEMLMRWNNLLRKRNFYKKLELHLPKFSISGSYVLD





QILPRLGFTDLFSKWADLSGITKQQKLEASKSFHKATLDVDEAGTEAAAATSFAI





KFFSAQTNRHILRFNRPFLVVIFSTSTQSVLFLGKVVDPTKP





SEQ ID No. 15



SerpinA4 transcript variant 2 (Human) cDNA



ATGCATCT TATCGACTAC CTGCTCCTCC TGCTGGTTGG ACTACTGGCC





CTTTCTCATG GCCAGCTGCA CGTTGAGCAT GATGGTGAGA GTTGCAGTAA





CAGCTCCCAC CAGCAGATTC TGGAGACAGG TGAGGGCTCC CCCAGCCTCA





AGATAGCCCC TGCCAATGCT GACTTTGCCT TCCGCTTCTA CTACCTGATC





GCTTCGGAGA CCCCGGGGAA GAACATCTTT TTCTCCCCGC TGAGCATCTC





GGCGGCCTAC GCCATGCTTT CCCTGGGGGC CTGCTCACAC AGCCGCAGCC





AGATCCTTGA GGGCCTGGGC TTCAACCTCA CCGAGCTGTC TGAGTCCGAT





GTCCATAGGG GCTTCCAGCA CCTCCTGCAC ACTCTCAACC TCCCCGGCCA





TGGGCTGGAA ACACGCGTGG GCAGTGCTCT GTTCCTGAGC CACAACCTGA





AGTTCCTTGC AAAATTCCTG AATGACACCA TGGCCGTCTA TGAGGCTAAA





CTCTTCCACA CCAACTTCTA CGACACTGTG GGCACAATCC AGCTTATCAA





CGACCACGTC AAGAAGGAAA CTCGAGGGAA GATTGTGGAT TTGGTCAGTG





AGCTCAAGAA GGACGTCTTG ATGGTGCTGG TGAATTACAT TTACTTCAAA





GCCCTGTGGG AGAAACCATT CATTTCCTCA AGGACCACTC CCAAAGACTT





CTATGTTGAT GAGAACACAA CAGTCCGGGT GCCCATGATG CTGCAGGACC





AGGAGCATCA CTGGTATCTT CATGACAGAT ACTTGCCCTG CTCGGTGCTA





CGGATGGATT ACAAAGGAGA CGCAACCGTG TTTTTCATTC TCCCTAACCA





AGGCAAAATG AGGGAGATTG AAGAGGTTCT GACTCCAGAG ATGCTAATGA





GGTGGAACAA CTTGTTGCGG AAGAGGAATT TTTACAAGAA GCTAGAGTTG





CATCTTCCCA AGTTCTCCAT TTCTGGCTCC TATGTATTAG ATCAGATTTT





GCCCAGGCTG GGCTTCACGG ATCTGTTCTC CAAGTGGGCT GACTTATCCG





GCATCACCAA ACAGCAAAAA CTGGAGGCAT CCAAAAGTTT CCACAAGGCC





ACCTTGGACG TGGATGAGGC TGGCACCGAG GCTGCAGCAG CCACCAGCTT





CGCGATCAAA TTCTTCTCTG CCCAGACCAA TCGCCACATC CTGCGATTCA





ACCGGCCCTT CCTTGTGGTG ATCTTTTCCA CCAGCACCCA GAGTGTCCTC





TTTCTGGGCA AGGTCGTCGA CCCCACGAAA CCATAG





SEQ ID No. 16



SerpinA4 transcript variant 1 (Human) Protein



MPGDPEKPPPGTHSWYRAALTEGQGLLAANPGLRVQRMHLIDYLLLLLVGLLAL





SHGQLHVEHDGESCSNSSHQQILETGEGSPSLKIAPANADFAFRFYYLIASETPGK





NIFFSPLSISAAYAMLSLGACSHSRSQILEGLGFNLTELSESDVHRGFQHLLHTLNL





PGHGLETRVGSALFLSHNLKFLAKFLNDTMAVYEAKLFHTNFYDTVGTIQLINDH





VKKETRGKIVDLVSELKKDVLMVLVNYIYFKALWEKPFISSRTTPKDFYVDENT





TVRVPMMLQDQEHHWYLHDRYLPCSVLRMDYKGDATVFFILPNQGKMREIEEV





LTPEMLMRWNNLLRKRNFYKKLELHLPKFSISGSYVLDQILPRLGFTDLFSKWAD





LSGITKQQKLEASKSFHKATLDVDEAGTEAAAATSFAIKFFSAQTNRHILRFNRPF





LVVIFSTSTQSVLFLGKVVDPTKP





SEQ ID No. 17



SerpinA4 transcript variant 1 (Human) cDNA



ATGC CCGGAGACCC AGAAAAGCCT CCCCCAGGGA CACACAGCTG





GTACAGGGCG GCACTGACTG AGGGCCAAGG TCTTCTGGCT GCCAATCCAG





GCCTGAGAGT GCAGAGGATG CATCTTATCG ACTACCTGCT CCTCCTGCTG





GTTGGACTAC TGGCCCTTTC TCATGGCCAG CTGCACGTTG AGCATGATGG





TGAGAGTTGC AGTAACAGCT CCCACCAGCA GATTCTGGAG ACAGGTGAGG





GCTCCCCCAG CCTCAAGATA GCCCCTGCCA ATGCTGACTT TGCCTTCCGC





TTCTACTACC TGATCGCTTC GGAGACCCCG GGGAAGAACA TCTTTTTCTC





CCCGCTGAGC ATCTCGGCGGCCTACGCCAT GCTTTCCCTG GGGGCCTGCT





CACACAGCCG CAGCCAGATC CTTGAGGGCCTGGGCTTCAA CCTCACCGAG





CTGTCTGAGT CCGATGTCCA TAGGGGCTTC CAGCACCTCCTGCACACTCT





CAACCTCCCC GGCCATGGGC TGGAAACACG CGTGGGCAGT GCTCTGTTCC





TGAGCCACAA CCTGAAGTTC CTTGCAAAAT TCCTGAATGA CACCATGGCC





GTCTATGAGGCTAAACTCTT CCACACCAAC TTCTACGACA CTGTGGGCAC





AATCCAGCTT ATCAACGACCACGTCAAGAA GGAAACTCGA GGGAAGATTG





TGGATTTGGT CAGTGAGCTC AAGAAGGACGTCTTGATGGT GCTGGTGAAT





TACATTTACT TCAAAGCCCT GTGGGAGAAA CCATTCATTTCCTCAAGGAC





CACTCCCAAA GACTTCTATG TTGATGAGAA CACAACAGTC CGGGTGCCCA





TGATGCTGCA GGACCAGGAG CATCACTGGT ATCTTCATGA CAGATACTTG





CCCTGCTCGGTGCTACGGAT GGATTACAAA GGAGACGCAA CCGTGTTTTT





CATTCTCCCT AACCAAGGCAAAATGAGGGA GATTGAAGAG GTTCTGACTC





CAGAGATGCT AATGAGGTGG AACAACTTGTTGCGGAAGAG GAATTTTTAC





AAGAAGCTAG AGTTGCATCT TCCCAAGTTC TCCATTTCTGGCTCCTATGT





ATTAGATCAG ATTTTGCCCA GGCTGGGCTT CACGGATCTG TTCTCCAAGT





GGGCTGACTT ATCCGGCATC ACCAAACAGC AAAAACTGGA GGCATCCAAA





AGTTTCCACAAGGCCACCTT GGACGTGGAT GAGGCTGGCA CCGAGGCTGC





AGCAGCCACC AGCTTCGCGATCAAATTCTT CTCTGCCCAG ACCAATCGCC





ACATCCTGCG ATTCAACCGG CCCTTCCTTGTGGTGATCTT TTCCACCAGC





ACCCAGAGTG TCCTCTTTCT GGGCAAGGTC GTCGACCCCACGAAACCATA G





SEQ ID No. 18



SerpinA5 (Human) Protein



MQLFLLLCLVLLSPQGASLHRHHPREMKKRVEDLHVGATVAPSSRRDFTFDLYR





ALASAAPSQSIFFSPVSISMSLAMLSLGAGSSTKMQILEGLGLNLQKSSEKELHRG





FQQLLQELNQPRDGFQLSLGNALFTDLVVDLQDTFVSAMKTLYLADTFPTNFRD





SAGAMKQINDYVAKQTKGKIVDLLKNLDSNAVVIMVNYIFFKAKWETSFNHKG





TQEQDFYVTSETVVRVPMMSREDQYHYLLDRNLSCRVVGVPYQGNATALFILPS





EGKMQQVENGLSEKTLRKWLKMFKKRQLELYLPKFSIEGSYQLEKVLPSLGISNV





FTSHADLSGISNHSNIQVSEMVHKAVVEVDESGTRAAAATGTIFTFRSARLNSQR





LVFNRPFLMFIVDNNILFLGKVNRP





SEQ ID No. 19



SerpinA5 (Human) cDNA



ATGCAGCTCTTCCTC CTCTTGTGCC TGGTGCTTCT CAGCCCTCAG





GGGGCCTCCC TTCACCGCCACCACCCCCGG GAGATGAAGA AGAGAGTCGA





GGACCTCCAT GTAGGTGCCA CGGTGGCCCCCAGCAGCAGA AGGGACTTTA





CCTTTGACCT CTACAGGGCC TTGGCTTCCG CTGCCCCCAGCCAGAGCATC





TTCTTCTCCC CTGTGAGCAT CTCCATGAGC CTGGCCATGC TCTCCCTGGG





GGCTGGGTCC AGCACAAAGA TGCAGATCCT GGAGGGCCTG GGCCTCAACC





TCCAGAAAAGCTCAGAGAAG GAGCTGCACA GAGGCTTTCA GCAGCTCCTT





CAGGAACTCA ACCAGCCCAGAGATGGCTTC CAGCTGAGCC TCGGCAATGC





CCTTTTCACC GACCTGGTGG TAGACCTGCAGGACACCTTC GTAAGTGCCA





TGAAGACGCT GTACCTGGCA GACACTTTCC CTACCAACTTTAGGGACTCT





GCAGGGGCCA TGAAGCAGAT CAATGATTAT GTGGCAAAGC AAACGAAGGG





CAAGATTGTG GACTTGCTTA AGAACCTCGA TAGCAATGCG GTCGTGATCA





TGGTGAATTACATCTTCTTT AAAGCTAAGT GGGAGACAAG CTTCAACCAC





AAAGGCACCC AAGAGCAAGACTTCTACGTG ACCTCGGAGA CTGTGGTGCG





GGTACCCATG ATGAGCCGCG AGGATCAGTATCACTACCTC CTGGACCGGA





ACCTCTCCTG CAGGGTGGTG GGGGTCCCCT ACCAAGGCAATGCCACGGCT





TTGTTCATTC TCCCCAGTGA GGGAAAGATG CAGCAGGTGG AGAATGGACT





GAGTGAGAAA ACGCTGAGGA AGTGGCTTAA GATGTTCAAA AAGAGGCAGC





TCGAGCTTTACCTTCCCAAA TTCTCCATTG AGGGCTCCTA TCAGCTGGAG





AAAGTCCTCC CCAGTCTGGGGATCAGTAAC GTCTTCACCT CCCATGCTGA





TCTGTCCGGC ATCAGCAACC ACTCAAATATCCAGGTGTCT GAGATGGTGC





ACAAAGCTGT GGTGGAGGTG GACGAGTCGG GAACCAGAGCAGCGGCAGCC





ACGGGGACAA TATTCACTTT CAGGTCGGCC CGCCTGAACT CTCAGAGGCT





AGTGTTCAAC AGGCCCTTTC TGATGTTCAT TGTGGATAAC AACATCCTCT





TCCTTGGCAAAGTGAACCGC CCCTGA





SEQ ID No. 20



SerpinA9 transcript variant 4 (Human) Protein



MGSALFVKKELQLQANFLGNVKRLYEAEVFSTDFSNPSIAQARINSHVKKKTQG





KVVDIIQGLDLLTAMVLVNHIFFKAKWEKPFHPEYTRKNFPFLVGEQVTVHVPM





MHQKEQFAFGVDTELNCFVLQMDYKGDAVAFFVLPSKGKMRQLEQALSARTL





RKWSHSLQKRWIEVFIPRFSISASYNLETILPKMGIQNVFDKNADFSGIAKRDSLQ





VSKATHKAVLDVSEEGTEATAATTTKFIVRSKDGPSYFTVSFNRTFLMMITNKAT





DGILFLGKVENPTKS





SEQ ID No. 21



SerpinA9 transcript variant 4 (Human) cDNA



ATGGG AAGTGCCCTC TTCGTCAAGA AGGAGCTGCA GCTGCAGGCA





AATTTCTTGG GCAATGTCAA GAGGCTGTAT GAAGCAGAAG TCTTTTCTAC





AGATTTCTCCAACCCCTCCA TTGCCCAGGC GAGGATCAAC AGCCATGTGA





AAAAGAAGAC CCAAGGGAAGGTTGTAGACA TAATCCAAGG CCTTGACCTT





CTGACGGCCA TGGTTCTGGT GAACCACATTTTCTTTAAAG CCAAGTGGGA





GAAGCCCTTT CACCCTGAAT ATACAAGAAA GAACTTCCCATTCCTGGTGG





GCGAGCAGGT CACTGTGCAT GTCCCCATGA TGCACCAGAA AGAGCAGTTC





GCTTTTGGGG TGGATACAGA GCTGAACTGC TTTGTGCTGC AGATGGATTA





CAAGGGAGATGCCGTGGCCT TCTTTGTCCT CCCTAGCAAG GGCAAGATGA





GGCAACTGGA ACAGGCCTTGTCAGCCAGAA CACTGAGAAA GTGGAGCCAC





TCACTCCAGA AAAGGTGGAT AGAGGTGTTCATCCCCAGAT TTTCCATTTC





TGCCTCCTAC AATCTGGAAA CCATCCTCCC GAAGATGGGCATCCAAAATG





TCTTTGACAA AAATGCTGAT TTTTCTGGAA TTGCAAAGAG AGACTCCCTG





CAGGTTTCTA AAGCAACCCA CAAGGCTGTG CTGGATGTCA GTGAAGAGGG





CACTGAGGCCACAGCAGCTA CCACCACCAA GTTCATAGTC CGATCGAAGG





ATGGCCCCTC TTACTTCACTGTCTCCTTCA ATAGGACCTT CCTGATGATG





ATTACAAATA AAGCCACAGA CGGTATTCTCTTTCTAGGGA AAGTGGAAAA





TCCCACTAAA TCCTAG





SEQ ID No. 22



SerpinA9 transcript variant 3 (Human) Protein



MRSAGGRGEIKVRRELQPSKQVSGLTNHARTGQEKRNLQRLVLETPSQNIFFSPV





SVSTSLAMLSLGAHSVTKTQILQGLGFNLTHTPESAIHQGFQHLVHSLTVPSKDLT





LKMGSALFVKKELQLQANFLGNVKRLYEAEVFSTDFSNPSIAQARINSHVKKKT





QGKVVDIIQGLDLLTAMVLVNHIFFKAKWEKPFHPEYTRKNFPFLVGEQVTVH





VPMMHQKEQFAFGVDTELNCFVLQMDYKGDAVAFFVLPSKGKMRQLEQALSA





RTLRKWSHSLQKRWIEVFIPRFSISASYNLETILPKMGIQNVFDKNADFSGIAKRD





SLQVSKATHKAVLDVSEEGTEATAATTTKFIVRSKDGPSYFTVSFNRTFLMMITN





KATDGILFLGKVENPTKS





SEQ ID No. 23



SerpinA9 transcript variant 3 (Human) cDNA



ATGAGATCAGCTGGAGGGAGAGGAGAGATTA AAGTGAGGAGAGAGCTACAA





CCAAGTAAGC AAGTGTCAGG GCTCACCAAC CATGCAAGGA CAGGGCAGGA





GAAGAGGAAC CTGCAAAGGC TGGTTTTGGA GACCCCGAGT CAGAACATCT





TCTTCTCCCCTGTGAGTGTC TCCACTTCCC TGGCCATGCT CTCCCTTGGG





GCCCACTCAG TCACCAAGACCCAGATTCTC CAGGGCCTGG GCTTCAACCT





CACACACACA CCAGAGTCTG CCATCCACCAGGGCTTCCAG CACCTGGTTC





ACTCACTGAC TGTTCCCAGC AAAGACCTGA CCTTGAAGATGGGAAGTGCC





CTCTTCGTCA AGAAGGAGCT GCAGCTGCAG GCAAATTTCT TGGGCAATGT





CAAGAGGCTG TATGAAGCAG AAGTCTTTTC TACAGATTTC TCCAACCCCT





CCATTGCCCAGGCGAGGATC AACAGCCATG TGAAAAAGAA GACCCAAGGG





AAGGTTGTAG ACATAATCCAAGGCCTTGAC CTTCTGACGG CCATGGTTCT





GGTGAACCAC ATTTTCTTTA AAGCCAAGTGGGAGAAGCCC TTTCACCCTG





AATATACAAG AAAGAACTTC CCATTCCTGG TGGGCGAGCAGGTCACTGTG





CATGTCCCCA TGATGCACCA GAAAGAGCAG TTCGCTTTTG GGGTGGATAC





AGAGCTGAAC TGCTTTGTGC TGCAGATGGA TTACAAGGGA GATGCCGTGG





CCTTCTTTGTCCTCCCTAGC AAGGGCAAGA TGAGGCAACT GGAACAGGCC





TTGTCAGCCA GAACACTGAGAAAGTGGAGC CACTCACTCC AGAAAAGGTG





GATAGAGGTG TTCATCCCCA GATTTTCCATTTCTGCCTCC TACAATCTGG





AAACCATCCT CCCGAAGATG GGCATCCAAA ATGTCTTTGACAAAAATGCT





GATTTTTCTG GAATTGCAAA GAGAGACTCC CTGCAGGTTT CTAAAGCAAC





CCACAAGGCT GTGCTGGATG TCAGTGAAGA GGGCACTGAG GCCACAGCAG





CTACCACCACCAAGTTCATA GTCCGATCGA AGGATGGCCC CTCTTACTTC





ACTGTCTCCT TCAATAGGACCTTCCTGATG ATGATTACAA ATAAAGCCAC





AGACGGTATT CTCTTTCTAG GGAAAGTGGAAAATCCCACT AAATCCTAG





SEQ ID No. 24



SerpinA9 transcript variant 2 (Human) Protein



MQGQGRRRGTCKDIFCSKMASYLYGVLFAVGLCAPIYCVSPANAPSAYPRPSST





KSTPASQVYSLNTDFAFRLYRRLVLETPSQNIFFSPARINSHVKKKTQGKVVDIIQ





GLDLLTAMVLVNHIFFKAKWEKPFHPEYTRKNFPFLVGEQVTVHVPMMHQKEQ





FAFGVDTELNCFVLQMDYKGDAVAFFVLPSKGKMRQLEQALSARTLRKWSHSL





QKRWIEVFIPRFSISASYNLETILPKMGIQNVFDKNADFSGIAKRDSLQVSKATHK





AVLDVSEEGTEATAATTTKFIVRSKDGPSYFTVSFNRTFLMMITNKATDGILFLGK





VENPTKS





SEQ ID No. 25



SerpinA9 transcript variant 2 (Human) cDNA



ATGCAAGGA CAGGGCAGGAGAAGAGGAAC CTGCAAAGAC ATATTTTGTT





CCAAAATGGC ATCTTACCTT TATGGAGTACTCTTTGCTGT TGGCCTCTGT





GCTCCAATCT ACTGTGTGTC CCCGGCCAAT GCCCCCAGTGCATACCCCCG





CCCTTCCTCC ACAAAGAGCA CCCCTGCCTC ACAGGTGTAT TCCCTCAACA





CCGACTTTGC CTTCCGCCTA TACCGCAGGC TGGTTTTGGA GACCCCGAGT





CAGAACATCTTCTTCTCCCC TGCGAGGATC AACAGCCATG TGAAAAAGAA





GACCCAAGGG AAGGTTGTAGACATAATCCA AGGCCTTGAC CTTCTGACGG





CCATGGTTCT GGTGAACCAC ATTTTCTTTAAAGCCAAGTG GGAGAAGCCC





TTTCACCCTG AATATACAAG AAAGAACTTC CCATTCCTGGTGGGCGAGCA





GGTCACTGTG CATGTCCCCA TGATGCACCA GAAAGAGCAG TTCGCTTTTG





GGGTGGATAC AGAGCTGAAC TGCTTTGTGC TGCAGATGGA TTACAAGGGA





GATGCCGTGGCCTTCTTTGT CCTCCCTAGC AAGGGCAAGA TGAGGCAACT





GGAACAGGCC TTGTCAGCCAGAACACTGAG AAAGTGGAGC CACTCACTCC





AGAAAAGGTG GATAGAGGTG TTCATCCCCAGATTTTCCAT TTCTGCCTCC





TACAATCTGG AAACCATCCT CCCGAAGATG GGCATCCAAAATGTCTTTGA





CAAAAATGCT GATTTTTCTG GAATTGCAAA GAGAGACTCC CTGCAGGTTT





CTAAAGCAAC CCACAAGGCT GTGCTGGATG TCAGTGAAGA GGGCACTGAG





GCCACAGCAGCTACCACCAC CAAGTTCATA GTCCGATCGA AGGATGGCCC





CTCTTACTTC ACTGTCTCCTTCAATAGGAC CTTCCTGATG ATGATTACAA





ATAAAGCCAC AGACGGTATT CTCTTTCTAGGGAAAGTGGA AAATCCCACT





AAATCCTAG





SEQ ID No. 26



SerpinA9 transcript variant 1 (Human) Protein



MQGQGRRRGTCKDIFCSKMASYLYGVLFAVGLCAPIYCVSPANAPSAYPRPSST





KSTPASQVYSLNTDFAFRLYRRLVLETPSQNIFFSPVSVSTSLAMLSLGAHSVTKT





QILQGLGFNLTHTPESAIHQGFQHLVHSLTVPSKDLTLKMGSALFVKKELQLQAN





FLGNVKRLYEAEVFSTDFSNPSIAQARINSHVKKKTQGKVVDIIQGLDLLTAMVL





VNHIFFKAKWEKPFHPEYTRKNFPFLVGEQVTVHVPMMHQKEQFAFGVDTELN





CFVLQMDYKGDAVAFFVLPSKGKMRQLEQALSARTLRKWSHSLQKRWIEVFIPR





FSISASYNLETILPKMGIQNVFDKNADFSGIAKRDSLQVSKATHKAVLDVSEEGTE





ATAATTTKFIVRSKDGPSYFTVSFNRTFLMMITNKATDGILFLGKVENPTKS





SEQ ID No. 27



SerpinA9 transcript variant 1 (Human) cDNA



ATGCAAGGA CAGGGCAGGAGAAGAGGAAC CTGCAAAGAC ATATTTTGTT





CCAAAATGGC ATCTTACCTT TATGGAGTACTCTTTGCTGT TGGCCTCTGT





GCTCCAATCT ACTGTGTGTC CCCGGCCAAT GCCCCCAGTGCATACCCCCG





CCCTTCCTCC ACAAAGAGCA CCCCTGCCTC ACAGGTGTAT TCCCTCAACA





CCGACTTTGC CTTCCGCCTA TACCGCAGGC TGGTTTTGGA GACCCCGAGT





CAGAACATCTTCTTCTCCCC TGTGAGTGTC TCCACTTCCC TGGCCATGCT





CTCCCTTGGG GCCCACTCAGTCACCAAGAC CCAGATTCTC CAGGGCCTGG





GCTTCAACCT CACACACACA CCAGAGTCTGCCATCCACCA GGGCTTCCAG





CACCTGGTTC ACTCACTGAC TGTTCCCAGC AAAGACCTGACCTTGAAGAT





GGGAAGTGCC CTCTTCGTCA AGAAGGAGCT GCAGCTGCAG GCAAATTTCT





TGGGCAATGT CAAGAGGCTG TATGAAGCAG AAGTCTTTTC TACAGATTTC





TCCAACCCCTCCATTGCCCA GGCGAGGATC AACAGCCATG TGAAAAAGAA





GACCCAAGGG AAGGTTGTAGACATAATCCA AGGCCTTGAC CTTCTGACGG





CCATGGTTCT GGTGAACCAC ATTTTCTTTAAAGCCAAGTG GGAGAAGCCC





TTTCACCCTG AATATACAAG AAAGAACTTC CCATTCCTGGTGGGCGAGCA





GGTCACTGTG CATGTCCCCA TGATGCACCA GAAAGAGCAG TTCGCTTTTG





GGGTGGATAC AGAGCTGAAC TGCTTTGTGC TGCAGATGGA TTACAAGGGA





GATGCCGTGGCCTTCTTTGT CCTCCCTAGC AAGGGCAAGA TGAGGCAACT





GGAACAGGCC TTGTCAGCCAGAACACTGAG AAAGTGGAGC CACTCACTCC





AGAAAAGGTG GATAGAGGTG TTCATCCCCAGATTTTCCAT TTCTGCCTCC





TACAATCTGG AAACCATCCT CCCGAAGATG GGCATCCAAAATGTCTTTGA





CAAAAATGCT GATTTTTCTG GAATTGCAAA GAGAGACTCC CTGCAGGTTT





CTAAAGCAAC CCACAAGGCT GTGCTGGATG TCAGTGAAGA GGGCACTGAG





GCCACAGCAGCTACCACCAC CAAGTTCATA GTCCGATCGA AGGATGGCCC





CTCTTACTTC ACTGTCTCCTTCAATAGGAC CTTCCTGATG ATGATTACAA





ATAAAGCCAC AGACGGTATT CTCTTTCTAGGGAAAGTGGA AAATCCCACT





AAATCCTAG





SEQ ID No. 28



SerpinA10 transcript variant 1 (Human) Protein



MKVVPSLLLSVLLAQVWLVPGLAPSPQSPETPAPQNQTSRVVQAPKEEEEDEQE





ASEEKASEEEKAWLMASRQQLAKETSNFGFSLLRKISMRHDGNMVFSPFGMSLA





MTGLMLGATGPTETQIKRGLHLQALKPTKPGLLPSLFKGLRETLSRNLELGLTQG





SFAFIHKDFDVKETFFNLSKRYFDTECVPMNFRNASQAKRLMNHYINKETRGKIP





KLFDEINPETKLILVDYILFKGKWLTPFDPVFTEVDTFHLDKYKTIKVPMMYGAG





KFASTFDKNFRCHVLKLPYQGNATMLVVLMEKMGDHLALEDYLTTDLVETWLR





NMKTRNMEVFFPKFKLDQKYEMHELLRQMGIRRIFSPFADLSELSATGRNLQVS





RVLQRTVIEVDERGTEAVAGILSEITAYSMPPVIKVDRPFHFMIYEETSGMLLFLG





RVVNPTLL





SEQ ID No. 29



SerpinA10 transcript variant 1 (Human) cDNA



ATGA AGGTGGTGCCAAGTCTCCTG CTCTCCGTCC TCCTGGCACA





GGTGTGGCTG GTACCCGGCT TGGCCCCCAGTCCTCAGTCG CCAGAGACCC





CAGCCCCTCA GAACCAGACC AGCAGGGTAG TGCAGGCTCCCAAGGAGGAA





GAGGAAGATG AGCAGGAGGC CAGCGAGGAG AAGGCCAGTAGGAAGAGAA





AGCCTGGCTG ATGGCCAGCA GGCAGCAGCT TGCCAAGGAG ACTTCAAACT





AAGATCTCCA TGAGGCACGA TGGCAACATG





GTCTTCTCTC CATTTGGCATGTCCTTGGCC ATGACAGGCT TGATGCTGGG





GGCCACAGGG CCGACTGAAA CCCAGATCAAGAGAGGGCTC CACTTGCAGG





CCCTGAAGCC CACCAAGCCC GGGCTCCTGC CTTCCCTCTTTAAGGGACTC





AGAGAGACCC TCTCCCGCAA CCTGGAACTG GGCCTCACAC AGGGGAGTTT





TGCCTTCATC CACAAGGATT TTGATGTCAA AGAGACTTTC TTCAATTTAT





CCAAGAGGTATTTTGATACA GAGTGCGTGC CTATGAATTT TCGCAATGCC





TCACAGGCCA AAAGGCTCATGAATCATTAC ATTAACAAAG AGACTCGGGG





GAAAATTCCC AAACTGTTTG ATGAGATTAATCCTGAAACC AAATTAATTC





TTGTGGATTA CATCTTGTTC AAAGGGAAAT GGTTGACCCCATTTGACCCT





GTCTTCACCG AAGTCGACAC TTTCCACCTG GACAAGTACA AGACCATTAA





GGTGCCCATG ATGTACGGTG CAGGCAAGTT TGCCTCCACC TTTGACAAGA





ATTTTCGTTGTCATGTCCTC AAACTGCCCT ACCAAGGAAA TGCCACCATG





CTGGTGGTCC TCATGGAGAAAATGGGTGAC CACCTCGCCC TTGAAGACTA





CCTGACCACA GACTTGGTGG AGACATGGCTCAGAAACATG AAAACCAGAA





ACATGGAAGT TTTCTTTCCG AAGTTCAAGC TAGATCAGAAGTATGAGATG





CATGAGCTGC TTAGGCAGAT GGGAATCAGA AGAATCTTCT CACCCTTTGC





TGACCTTAGT GAACTCTCAG CTACTGGAAG AAATCTCCAA GTATCCAGGG





TTTTACAAAGAACAGTGATT GAAGTTGATG AAAGGGGCAC TGAGGCAGTG





GCAGGAATCT TGTCAGAAATTACTGCTTAT TCCATGCCTC CTGTCATCAA





AGTGGACCGG CCATTTCATT TCATGATCTATGAAGAAACC TCTGGAATGC





TTCTGTTTCT GGGCAGGGTG GTGAATCCGA CTCTCCTATAA





SEQ ID No. 30



SerpinA10 transcript variant 2 (Human) Protein



MKVVPSLLLSVLLAQVWLVPGLAPSPQSPETPAPQNQTSRVVQAPKEEEEDEQE





ASEEKASEEEKAWLMASRQQLAKETSNFGFSLLRKISMRHDGNMVFSPFGMSLA





MTGLMLGATGPTETQIKRGLHLQALKPTKPGLLPSLFKGLRETLSRNLELGLTQG





SFAFIHKDFDVKETFFNLSKRYFDTECVPMNFRNASQAKRLMNHYINKETRGKIP





KLFDEINPETKLILVDYILFKGKWLTPFDPVFTEVDTFHLDKYKTIKVPMMYGAG





KFASTFDKNFRCHVLKLPYQGNATMLVVLMEKMGDHLALEDYLTTDLVETWLR





NMKTRNMEVFFPKFKLDQKYEMHELLRQMGIRRIFSPFADLSELSATGRNLQVS





RVLQRTVIEVDERGTEAVAGILSEITAYSMPPVIKVDRPFHFMIYEETSGMLLFLG





RVVNPTLL





SEQ ID No. 31



SerpinA10 transcript variant 2 (Human) cDNA



ATGAAGG TGGTGCCAAG TCTCCTGCTC TCCGTCCTCC TGGCACAGGT





GTGGCTGGTA CCCGGCTTGG CCCCCAGTCC TCAGTCGCCA GAGACCCCAG





CCCCTCAGAACCAGACCAGC AGGGTAGTGC AGGCTCCCAA GGAGGAAGAG





GAAGATGAGC AGGAGGCCAGCGAGGAGAAG GCCAGTGAGGAAGAGAAAGC





CTGGCTGATG GCCAGCAGGC AGCAGCTTGCCAAGGAGACT TCAAACTTCG





GATTCAGCCT GCTGCGAAAG ATCTCCATGA GGCACGATGGCAACATGGTC





TTCTCTCCAT TTGGCATGTC CTTGGCCATG ACAGGCTTGA TGCTGGGGGC





CACAGGGCCG ACTGAAACCC AGATCAAGAG AGGGCTCCAC TTGCAGGCCC





TGAAGCCCACCAAGCCCGGG CTCCTGCCTT CCCTCTTTAA GGGACTCAGA





GAGACCCTCT CCCGCAACCTGGAACTGGGC CTCACACAGG GGAGTTTTGC





CTTCATCCAC AAGGATTTTG ATGTCAAAGAGACTTTCTTC AATTTATCCA





AGAGGTATTT TGATACAGAG TGCGTGCCTA TGAATTTTCGCAATGCCTCA





CAGGCCAAAA GGCTCATGAA TCATTACATT AACAAAGAGA CTCGGGGGAA





AATTCCCAAA CTGTTTGATG AGATTAATCC TGAAACCAAA TTAATTCTTG





TGGATTACATCTTGTTCAAA GGGAAATGGT TGACCCCATT TGACCCTGTC





TTCACCGAAG TCGACACTTTCCACCTGGAC AAGTACAAGA CCATTAAGGT





GCCCATGATG TACGGTGCAG GCAAGTTTGCCTCCACCTTT GACAAGAATT





TTCGTTGTCA TGTCCTCAAA CTGCCCTACC AAGGAAATGCCACCATGCTG





GTGGTCCTCA TGGAGAAAAT GGGTGACCAC CTCGCCCTTG AAGACTACCT





GACCACAGAC TTGGTGGAGA CATGGCTCAG AAACATGAAA ACCAGAAACA





TGGAAGTTTTCTTTCCGAAG TTCAAGCTAG ATCAGAAGTA TGAGATGCAT





GAGCTGCTTA GGCAGATGGGAATCAGAAGA ATCTTCTCAC CCTTTGCTGA





CCTTAGTGAA CTCTCAGCTA CTGGAAGAAATCTCCAAGTA TCCAGGGTTT





TACAAAGAAC AGTGATTGAA GTTGATGAAA GGGGCACTGAGGCAGTGGCA





GGAATCTTGT CAGAAATTAC TGCTTATTCC ATGCCTCCTG TCATCAAAGT





GGACCGGCCA TTTCATTTCA TGATCTATGA AGAAACCTCT GGAATGCTTC





TGTTTCTGGGCAGGGTGGTG AATCCGACTC TCCTATAA





SEQ ID No. 32



SerpinA12 transcript variant 1 (Human) Protein



MNPTLGLAIFLAVLLTVKGLLKPSFSPRNYKALSEVQGWKQRMAAKELARQNM





DLGFKLLKKLAFYNPGRNIFLSPLSISTAFSMLCLGAQDSTLDEIKQGFNFRKMPE





KDLHEGFHYIIHELTQKTQDLKLSIGNTLFIDQRLQPQRKFLEDAKNFYSAETILTN





FQNLEMAQKQINDFISQKTHGKINNLIENIDPGTVMLLANYIFFRARWKHEFDPN





VTKEEDFFLEKNSSVKVPMMFRSGIYQVGYDDKLSCTILEIPYQKNITAIFILPD





EGKLKHLEKGLQVDTFSRWKTLLSRRVVDVSVPRLHMTGTFDLKKTLSYIGVSKI





FEEHGDLTKIAPHRSLKVGEAVHKAELKMDERGTEGAAGTGAQTLPMETPLVVK





IDKPYLLLIYSEKIPSVLFLGKIVNPIGK





SEQ ID No. 33



SerpinA12 transcript variant 1 (Human) cDNA



ATGA ACCCCACACT AGGCCTGGCC ATTTTTCTGG CTGTTCTCCT





CACGGTGAAA GGTCTTCTAA AGCCGAGCTT CTCACCAAGG AATTATAAAG





CTTTGAGCGAGGTCCAAGGA TGGAAGCAAA GGATGGCAGC CAAGGAGCTT





GCAAGGCAGA ACATGGACTTAGGCTTTAAG CTGCTCAAGA AGCTGGCCTT





TTACAACCCT GGCAGGAACA TCTTCCTATCCCCCTTGAGC ATCTCTACAG





CTTTCTCCAT GCTGTGCCTG GGTGCCCAGG ACAGCACCCTGGACGAGATC





AAGCAGGGGT TCAACTTCAG AAAGATGCCA GAAAAAGATC TTCATGAGGG





CTTCCATTAC ATCATCCACG AGCTGACCCA GAAGACCCAG GACCTCAAAC





TGAGCATTGGGAACACGCTG TTCATTGACC AGAGGCTGCA GCCACAGCGT





AAGTTTTTGG AAGATGCCAAGAACTTTTAC AGTGCCGAAA CCATCCTTAC





CAACTTTCAG AATTTGGAAA TGGCTCAGAAGCAGATCAAT GACTTTATCA





GTCAAAAAAC CCATGGGAAA ATTAACAACC TGATCGAGAATATAGACCCC





GGCACTGTGA TGCTTCTTGC AAATTATATT TTCTTTCGAG CCAGGTGGAA





ACATGAGTTT GATCCAAATG TAACTAAAGA GGAAGATTTC TTTCTGGAGA





AAAACAGTTCAGTCAAGGTG CCCATGATGT TCCGTAGTGG CATATACCAA





GTTGGCTATG ACGATAAGCTCTCTTGCACC ATCCTGGAAA TACCCTACCA





GAAAAATATC ACAGCCATCT TCATCCTTCCTGATGAGGGC AAGCTGAAGC





ACTTGGAGAA GGGATTGCAG GTGGACACTT TCTCCAGATGGAAAACATTA





CTGTCACGCA GGGTCGTAGA CGTGTCTGTA CCCAGACTCC ACATGACGGG





CACCTTCGAC CTGAAGAAGA CTCTCTCCTA CATAGGTGTC TCCAAAATCT





TTGAGGAACATGGTGATCTC ACCAAGATCG CCCCTCATCG CAGCCTGAAA





GTGGGCGAGG CTGTGCACAAGGCTGAGCTG AAGATGGATG AGAGGGGTAC





GGAAGGGGCC GCTGGCACCG GAGCACAGACTCTGCCCATG GAGACACCAC





TCGTCGTCAA GATAGACAAA CCCTATCTGC TGCTGATTTACAGCGAGAAA





ATACCTTCCG TGCTCTTCCT GGGAAAGATT GTTAACCCTA TTGGAAAATA





A





SEQ ID No. 34



SerpinA12 transcript variant 2 (Human) Protein



MNPTLGLAIFLAVLLTVKGLLKPSFSPRNYKALSEVQGWKQRMAAKELARQNM





DLGFKLLKKLAFYNPGRNIFLSPLSISTAFSMLCLGAQDSTLDEIKQGFNFRKMPE





KDLHEGFHYIIHELTQKTQDLKLSIGNTLFIDQRLQPQRKFLEDAKNFYSAETILTN





FQNLEMAQKQINDFISQKTHGKINNLIENIDPGTVMLLANYIFFRARWKHEFDPN





VTKEEDFFLEKNSSVKVPMMFRSGIYQVGYDDKLSCTILEIPYQKNITAIFILPDEG





KLKHLEKGLQVDTFSRWKTLLSRRVVDVSVPRLHMTGTFDLKKTLSYIGVSKIFE





EHGDLTKIAPHRSLKVGEAVHKAELKMDERGTEGAAGTGAQTLPMETPLVVKID





KPYLLLIYSEKIPSVLFLGKIVNPIGK





SEQ ID No. 35



SerpinA12 transcript variant 2 (Human) cDNA



ATGAACC CCACACTAGGCCTGGCCATT TTTCTGGCTG TTCTCCTCAC





GGTGAAAGGT CTTCTAAAGC CGAGCTTCTCACCAAGGAAT TATAAAGCTT





TGAGCGAGGT CCAAGGATGG AAGCAAAGGA TGGCAGCCAAGGAGCTTGCA





AGGCAGAACA TGGACTTAGG CTTTAAGCTG CTCAAGAAGC TGGCCTTTTA





CAACCCTGGC AGGAACATCT TCCTATCCCC CTTGAGCATC TCTACAGCTT





TCTCCATGCTGTGCCTGGGT GCCCAGGACA GCACCCTGGA CGAGATCAAG





CAGGGGTTCA ACTTCAGAAAGATGCCAGAA AAAGATCTTC ATGAGGGCTT





CCATTACATC ATCCACGAGC TGACCCAGAAGACCCAGGAC CTCAAACTGA





GCATTGGGAA CACGCTGTTC ATTGACCAGA GGCTGCAGCCACAGCGTAAG





TTTTTGGAAG ATGCCAAGAA CTTTTACAGT GCCGAAACCA TCCTTACCAA





CTTTCAGAAT TTGGAAATGG CTCAGAAGCA GATCAATGAC TTTATCAGTC





AAAAAACCCATGGGAAAATT AACAACCTGA TCGAGAATAT AGACCCCGGC





ACTGTGATGC TTCTTGCAAATTATATTTTC TTTCGAGCCA GGTGGAAACA





TGAGTTTGAT CCAAATGTAA CTAAAGAGGAAGATTTCTTT CTGGAGAAAA





ACAGTTCAGT CAAGGTGCCC ATGATGTTCC GTAGTGGCATATACCAAGTT





GGCTATGACG ATAAGCTCTC TTGCACCATC CTGGAAATAC CCTACCAGAA





AAATATCACA GCCATCTTCA TCCTTCCTGA TGAGGGCAAG CTGAAGCACT





TGGAGAAGGGATTGCAGGTG GACACTTTCT CCAGATGGAA AACATTACTG





TCACGCAGGG TCGTAGACGTGTCTGTACCC AGACTCCACA TGACGGGCAC





CTTCGACCTG AAGAAGACTC TCTCCTACATAGGTGTCTCC AAAATCTTTG





AGGAACATGG TGATCTCACC AAGATCGCCC CTCATCGCAGCCTGAAAGTG





GGCGAGGCTG TGCACAAGGC TGAGCTGAAG ATGGATGAGA GGGGTACGGA





AGGGGCCGCT GGCACCGGAG CACAGACTCT GCCCATGGAG ACACCACTCG





TCGTCAAGATAGACAAACCC TATCTGCTGC TGATTTACAG CGAGAAAATA





CCTTCCGTGC TCTTCCTGGGAAAGATTGTT AACCCTATTG GAAAATAA





SEQ ID No. 36



SerpinB1 (Human) Protein



MEQLSSANTRFALDLFLALSENNPAGNIFISPFSISSAMAMVFLGTRGNTAAQLSK





TFHFNTVEEVHSRFQSLNADINKRGASYILKLANRLYGEKTYNFLPEFLVSTQKT





YGADLASVDFQHASEDARKTINQWVKGQTEGKIPELLASGMVDNMTKLVLVNA





IYFKGNWKDKFMKEATTNAPFRLNKKDRKTVKMMYQKKKFAYGYIEDLKCRV





LELPYQGEELSMVILLPDDIEDESTGLKKIEEQLTLEKLHEWTKPENLDFIEVNVSL





PRFKLEESYTLNSDLARLGVQDLFNSSKADLSGMSGARDIFISKIVHKSFVEVNEE





GTEAAAATAGIATFCMLMPEENFTADHPFLFFIRHNSSGSILFLGRFSSP





SEQ ID No. 37



SerpinB1 (Human) cDNA



ATGG AGCAGCTGAG CTCAGCAAAC ACCCGCTTCG CCTTGGACCT





GTTCCTGGCGTTGAGTGAGA ACAATCCGGC TGGAAACATC TTCATCTCTC





CCTTCAGCAT TTCATCTGCTATGGCCATGG TTTTTCTGGG GACCAGAGGT





AACACGGCAG CACAGCTGTC CAAGACTTTCCATTTCAACA CGGTTGAAGA





GGTTCATTCA AGATTCCAGA GTCTGAATGC TGATATCAACAAACGTGGAG





CGTCTTATAT TCTGAAACTT GCTAATAGAT TATATGGAGA GAAAACTTAC





AATTTCCTTC CTGAGTTCTT GGTTTCGACT CAGAAAACAT ATGGTGCTGA





CCTGGCCAGTGTGGATTTTC AGCATGCCTC TGAAGATGCA AGGAAGACCA





TAAACCAGTG GGTCAAAGGACAGACAGAAG GAAAAATTCC GGAACTGTTG





GCTTCGGGCA TGGTTGATAA CATGACCAAACTTGTGCTAG TAAATGCCAT





CTATTTCAAG GGAAACTGGA AGGATAAATT CATGAAAGAAGCCACGACGA





ATGCACCATT CAGATTGAAT AAGAAAGACA GAAAAACTGT GAAAATGATG





TATCAGAAGA AAAAATTTGC ATATGGCTAC ATCGAGGACC TTAAGTGCCG





TGTGCTGGAACTGCCTTACC AAGGCGAGGA GCTCAGCATG GTCATCCTGC





TGCCGGATGA CATTGAGGACGAGTCCACGG GCCTGAAGAA GATTGAGGAA





CAGTTGACTT TGGAAAAGTT GCATGAGTGGACTAAACCTG AGAATCTCGA





TTTCATTGAA GTTAATGTCA GCTTGCCCAG GTTCAAACTGGAAGAGAGTT





ACACTCTCAA CTCCGACCTC GCCCGCCTAG GTGTGCAGGA TCTCTTTAAC





AGTAGCAAGG CTGATCTGTC TGGCATGTCA GGAGCCAGAG ATATTTTTAT





ATCAAAAATTGTCCACAAGT CATTTGTGGA AGTGAATGAA GAGGGAACAG





AGGCGGCAGC TGCCACAGCAGGCATCGCAA CTTTCTGCAT GTTGATGCCC





GAAGAAAATT TCACTGCCGA CCATCCATTCCTTTTCTTTA TTCGGCATAA





TTCCTCAGGT AGCATCCTAT TCTTGGGGAG ATTTTCTTCCCCTTAG





SEQ ID No. 38



SerpinB6 variant 1 (Human) Protein



MDVLAEANGTFALNLLKTLGKDNSKNVFFSPMSMSCALAMVYMGAKGNTAAQ





MAQILSFNKSGGGGDIHQGFQSLLTEVNKTGTQYLLRMANRLFGEKSCDFLSSFR





DSCQKFYQAEMEELDFISAVEKSRKHINTWVAEKTEGKIAELLSPGSVDPLTRLV





LVNAVYFRGNWDEQFDKENTEERLFKVSKNEEKPVQMMFKQSTFKKTYIGEIFT





QILVLPYVGKELNMIIMLPDETTDLRTVEKELTYEKFVEWTRLDMMDEEEVEVSL





PRFKLEESYDMESVLRNLGMTDAFELGKADFSGMSQTDLSLSKVVHKSFVEVNE





EGTEAAAATAAIMMMRCARFVPRFCADHPFLFFIQHSKTNGILFCGRFSSP





SEQ ID No. 39



SerpinB6 variant 1 (Human) cDNA



ATGGATG TTCTCGCAGAAGCAAATGGC ACCTTTGCCT TAAACCTTTT





GAAAACGCTG GGTAAAGACA ACTCGAAGAATGTGTTTTTC TCACCCATGA





GCATGTCCTG TGCCCTGGCC ATGGTCTACA TGGGGGCAAAGGGAAACACC





GCTGCACAGA TGGCCCAGAT ACTTTCTTTC AATAAAAGTG GCGGTGGTGG





AGACATCCAC CAGGGCTTCC AGTCTCTTCT CACCGAAGTG AACAAGACTG





GCACGCAGTACTTGCTTAGG ATGGCCAACA GGCTCTTTGG GGAAAAGTCT





TGTGATTTCC TCTCATCTTTTAGAGATTCC TGCCAAAAAT TCTACCAAGC





AGAGATGGAG GAGCTTGACT TTATCAGCGCCGTAGAGAAG TCCAGAAAAC





ACATAAACAC CTGGGTAGCT GAAAAGACAG AAGGTAAAATTGCGGAGTTG





CTCTCTCCGG GCTCAGTGGA TCCATTGACA AGGCTGGTTC TGGTGAATGC





TGTCTATTTC AGAGGAAACT GGGATGAACA GTTTGACAAG GAGAACACCG





AGGAGAGACTGTTTAAAGTC AGCAAGAATG AGGAGAAACC TGTGCAAATG





ATGTTTAAGC AATCTACTTTTAAGAAGACC TATATAGGAG AAATATTTAC





CCAAATCTTG GTGCTTCCAT ATGTTGGCAAGGAACTGAAT ATGATCATCA





TGCTTCCGGA CGAGACCACT GACTTGAGAA CGGTGGAGAAAGAACTCACT





TACGAGAAGT TCGTAGAATG GACGAGGCTG GACATGATGG ATGAAGAGGA





GGTGGAAGTG TCCCTCCCGC GGTTTAAACT AGAGGAAAGC TACGACATGG





AGAGTGTCCTGCGCAACCTG GGCATGACTG ATGCCTTCGA GCTGGGCAAG





GCAGACTTCT CTGGAATGTCCCAGACAGAC CTGTCTCTGT CCAAGGTCGT





GCACAAGTCT TTTGTGGAGG TCAATGAGGAAGGCACGGAG GCTGCAGCCG





CCACAGCTGC CATCATGATG ATGCGGTGTG CCAGATTCGTCCCCCGCTTC





TGCGCCGACC ACCCCTTCCT TTTCTTCATC CAGCACAGCA AGACCAACGG





GATTCTCTTC TGCGGCCGCT TTTCCTCTCC GTGA





SEQ ID No. 40



SerpinB6 variant 2 (Human) Protein



MSAIMDVLAEANGTFALNLLKTLGKDNSKNVFFSPMSMSCALAMVYMGAKGN





TAAQMAQILSFNKSGGGGDIHQGFQSLLTEVNKTGTQYLLRMANRLFGEKSCDF





LSSFRDSCQKFYQAEMEELDFISAVEKSRKHINTWVAEKTEGKIAELLSPGSVD





PLTRLVLVNAVYFRGNWDEQFDKENTEERLFKVSKNEEKPVQMMFKQSTFKKT





YIGEIFTQILVLPYVGKELNMIIMLPDETTDLRTVEKELTYEKFVEWTRLDMMDE





EEVEVSLPRFKLEESYDMESVLRNLGMTDAFELGKADFSGMSQTDLSLSKVVHK





SFVEVNEEGTEAAAATAAIMMMRCARFVPRFCADHPFLFFIQHSKTNGILFCGRF





SSP





SEQ ID No. 41



SerpinB6 variant 2 (Human) cDNA



ATGT CTGCCATCATGGATGTTCTC GCAGAAGCAA ATGGCAC CTT





TGCCTTAAAC CTTTTGAAAA CGCTGGGTAAAGACAACTCG AAGAATGTGT





TTTTCTCACC CATGAGCATG TCCTGTGCCC TGGCCATGGTCTACATGGGG





GCAAAGGGAA ACACCGCTGC ACAGATGGCC CAGATACTTT CTTTCAATAA





AAGTGGCGGT GGTGGAGACA TCCACCAGGG CTTCCAGTCT CTTCTCACCG





AAGTGAACAAGACTGGCACG CAGTACTTGC TTAGGATGGC CAACAGGCTC





TTTGGGGAAA AGTCTTGTGATTTCCTCTCA TCTTTTAGAG ATTCCTGCCA





AAAATTCTAC CAAGCAGAGA TGGAGGAGCTTGACTTTATC AGCGCCGTAG





AGAAGTCCAG AAAACACATA AACACCTGGG TAGCTGAAAAGACAGAAGGT





AAAATTGCGG AGTTGCTCTC TCCGGGCTCA GTGGATCCAT TGACAAGGCT





GGTTCTGGTG AATGCTGTCT ATTTCAGAGG AAACTGGGAT GAACAGTTTG





ACAAGGAGAACACCGAGGAG AGACTGTTTA AAGTCAGCAA GAATGAGGAG





AAACCTGTGC AAATGATGTTTAAGCAATCT ACTTTTAAGA AGACCTATAT





AGGAGAAATA TTTACCCAAA TCTTGGTGCTTCCATATGTT GGCAAGGAAC





TGAATATGAT CATCATGCTT CCGGACGAGA CCACTGACTTGAGAACGGTG





GAGAAAGAAC TCACTTACGA GAAGTTCGTA GAATGGACGA GGCTGGACAT





GATGGATGAA GAGGAGGTGG AAGTGTCCCT CCCGCGGTTT AAACTAGAGG





AAAGCTACGACATGGAGAGT GTCCTGCGCA ACCTGGGCAT GACTGATGCC





TTCGAGCTGG GCAAGGCAGACTTCTCTGGA ATGTCCCAGA CAGACCTGTC





TCTGTCCAAG GTCGTGCACA AGTCTTTTGTGGAGGTCAAT GAGGAAGGCA





CGGAGGCTGC AGCCGCCACA GCTGCCATCA TGATGATGCGGTGTGCCAGA





TTCGTCCCCC GCTTCTGCGC CGACCACCCC TTCCTTTTCT TCATCCAGCA





CAGCAAGACC AACGGGATTC TCTTCTGCGG CCGCTTTTCC TCTCCGTGA





SEQ ID No. 42



SerpinB6 variant 3 (Human) Protein



MGAAQSLPGHRSAIMDVLAEANGTFALNLLKTLGKDNSKNVFFSPMSMSCALA





MVYMGAKGNTAAQMAQILSFNKSGGGGDIHQGFQSLLTEVNKTGTQYLL





RMANRLFGEKSCDFLSSFRDSCQKFYQAEMEELDFISAVEKSRKHINTWVAEKTE





GKIAELLSPGSVDPLTRLVLVNAVYFRGNWDEQFDKENTEERLFKVSKNEEKPV





QMMFKQSTFKKTYIGEIFTQILVLPYVGKELNMIIMLPDETTDLRTVEKELTYEKF





VEWTRLDMMDEEEVEVSLPRFKLEESYDMESVLRNLGMTDAFELGKADFSGMS





QTDLSLSKVVHKSFVEVNEEGTEAAAATAAIMMMRCARFVPRFCADHPFLFFIQ





HSKTNGILFCGRFSSP





SEQ ID No. 43



SerpinB6 variant 3 (Human) cDNA



ATGGGGGCG GCGCAGAGCCTCCCGGGCCA CAGGTCTGCC ATCATGGATG





TTCTCGCAGA AGCAAATGGC ACCTTTGCCTTAAACCTTTT GAAAACGCTG





GGTAAAGACA ACTCGAAGAA TGTGTTTTTC TCACCCATGAGCATGTCCTG





TGCCCTGGCC ATGGTCTACA TGGGGGCAAA GGGAAACACC GCTGCACAGA





TGGCCCAGAT ACTTTCTTTC AATAAAAGTG GCGGTGGTGG AGACATCCAC





CAGGGCTTCCAGTCTCTTCT CACCGAAGTG AACAAGACTG GCACGCAGTA





CTTGCTTAGG ATGGCCAACAGGCTCTTTGG GGAAAAGTCT TGTGATTTCC





TCTCATCTTT TAGAGATTCC TGCCAAAAATTCTACCAAGC AGAGATGGAG





GAGCTTGACT TTATCAGCGC CGTAGAGAAG TCCAGAAAACACATAAACAC





CTGGGTAGCT GAAAAGACAG AAGGTAAAAT TGCGGAGTTG CTCTCTCCGG





GCTCAGTGGA TCCATTGACA AGGCTGGTTC TGGTGAATGC TGTCTATTTC





AGAGGAAACTGGGATGAACA GTTTGACAAG GAGAACACCG AGGAGAGACT





GTTTAAAGTC AGCAAGAATGAGGAGAAACC TGTGCAAATG ATGTTTAAGC





AATCTACTTT TAAGAAGACC TATATAGGAGAAATATTTAC CCAAATCTTG





GTGCTTCCAT ATGTTGGCAA GGAACTGAAT ATGATCATCATGCTTCCGGA





CGAGACCACT GACTTGAGAA CGGTGGAGAA AGAACTCACT TACGAGAAGT





TCGTAGAATG GACGAGGCTG GACATGATGG ATGAAGAGGA GGTGGAAGTG





TCCCTCCCGCGGTTTAAACT AGAGGAAAGC TACGACATGG AGAGTGTCCT





GCGCAACCTG GGCATGACTGATGCCTTCGA GCTGGGCAAG GCAGACTTCT





CTGGAATGTC CCAGACAGAC CTGTCTCTGTCCAAGGTCGT GCACAAGTCT





TTTGTGGAGG TCAATGAGGA AGGCACGGAG GCTGCAGCCGCCACAGCTGC





CATCATGATG ATGCGGTGTG CCAGATTCGT CCCCCGCTTC TGCGCCGACC





ACCCCTTCCT TTTCTTCATC CAGCACAGCA AGACCAACGG GATTCTCTTC





TGCGGCCGCTTTTCCTCTCC GTGA





SEQ ID No. 44



SerpinB6 variant 4 (Human) Protein



MSSRQRGNFNYKLAFKSAIMDVLAEANGTFALNLLKTLGKDNSKNVFFSPMSMS





CALAMVYMGAKGNTAAQMAQILSFNKSGGGGDIHQGFQSLLTEVNKTG





TQYLLRMANRLFGEKSCDFLSSFRDSCQKFYQAEMEELDFISAVEKSRKHINTWV





AEKTEGKIAELLSPGSVDPLTRLVLVNAVYFRGNWDEQFDKENTEERLFKVSKN





EEKPVQMMFKQSTFKKTYIGEIFTQILVLPYVGKELNMIIMLPDETTDLRTVEKEL





TYEKFVEWTRLDMMDEEEVEVSLPRFKLEESYDMESVLRNLGMTDAFELGKAD





FSGMSQTDLSLSKVVHKSFVEVNEEGTEAAAATAAIMMMRCARFVPRFCADHPF





LFFIQHSKTNGILFCGRFSSP





SEQ ID No. 45



SerpinB6 variant 4 (Human) cDNA



ATGTCT TCAAGGCAAA GAGGAAACTT TAACTACAAA TTGGCATTTA





AGTCTGCCATCATGGATGTT CTCGCAGAAG CAAATGGCAC CTTTGCCTTA





AACCTTTTGA AAACGCTGGGTAAAGACAAC TCGAAGAATG TGTTTTTCTC





ACCCATGAGC ATGTCCTGTG CCCTGGCCATGGTCTACATG GGGGCAAAGG





GAAACACCGC TGCACAGATG GCCCAGATAC TTTCTTTCAATAAAAGTGGC





GGTGGTGGAG ACATCCACCA GGGCTTCCAG TCTCTTCTCA CCGAAGTGAA





CAAGACTGGC ACGCAGTACT TGCTTAGGAT GGCCAACAGG CTCTTTGGGG





AAAAGTCTTGTGATTTCCTC TCATCTTTTA GAGATTCCTG CCAAAAATTC





TACCAAGCAG AGATGGAGGAGCTTGACTTT ATCAGCGCCG TAGAGAAGTC





CAGAAAACAC ATAAACACCT GGGTAGCTGAAAAGACAGAA GGTAAAATTG





CGGAGTTGCT CTCTCCGGGC TCAGTGGATC CATTGACAAGGCTGGTTCTG





GTGAATGCTG TCTATTTCAG AGGAAACTGG GATGAACAGT TTGACAAGGA





GAACACCGAG GAGAGACTGT TTAAAGTCAG CAAGAATGAG GAGAAACCTG





TGCAAATGATGTTTAAGCAA TCTACTTTTA AGAAGACCTA TATAGGAGAA





ATATTTACCC AAATCTTGGTGCTTCCATAT GTTGGCAAGG AACTGAATAT





GATCATCATG CTTCCGGACG AGACCACTGACTTGAGAACG GTGGAGAAAG





AACTCACTTA CGAGAAGTTC GTAGAATGGA CGAGGCTGGACATGATGGAT





GAAGAGGAGG TGGAAGTGTC CCTCCCGCGG TTTAAACTAG AGGAAAGCTA





CGACATGGAG AGTGTCCTGC GCAACCTGGG CATGACTGAT GCCTTCGAGC





TGGGCAAGGCAGACTTCTCT GGAATGTCCC AGACAGACCT GTCTCTGTCC





AAGGTCGTGC ACAAGTCTTTTGTGGAGGTC AATGAGGAAG GCACGGAGGC





TGCAGCCGCC ACAGCTGCCA TCATGATGATGCGGTGTGCC AGATTCGTCC





CCCGCTTCTG CGCCGACCAC CCCTTCCTTT TCTTCATCCAGCACAGCAAG





ACCAACGGGA TTCTCTTCTG CGGCCGCTTT TCCTCTCCGT GA





SEQ ID No. 46



SerpinB6 variant 5 (Human) Protein



MDVLAEANGTFALNLLKTLGKDNSKNVFFSPMSMSCALAMVYMGAKGNTAAQ





MAQILSFNKSGGGGDIHQGFQSLLTEVNKTGTQYLLRMANRLFGEKSCDFLSSFR





DSCQKFYQAEMEELDFISAVEKSRKHINTWVAEKTEGKIAELLSPGSVDPLTR





LVLVNAVYFRGNWDEQFDKENTEERLFKVSKNEEKPVQMMFKQSTFKKTYIGEI





FTQILVLPYVGKELNMIIMLPDETTDLRTVEKELTYEKFVEWTRLDMMDEEEVEV





SLPRFKLEESYDMESVLRNLGMTDAFELGKADFSGMSQTDLSLSKVVHKSFVEV





NEEGTEAAAATAAIMMMRCARFVPRFCADHPFLFFIQHSKTNGILFCGRFSSP





SEQ ID No. 47



SerpinB6 variant 5 (Human) cDNA



ATGGA TGTTCTCGCA GAAGCAAATG GCACCTTTGCCTTAAACCTT





TTGAAAACGC TGGGTAAAGA CAACTCGAAG AATGTGTTTT TCTCACCCAT





GAGCATGTCC TGTGCCCTGG CCATGGTCTA CATGGGGGCA AAGGGAAACA





CCGCTGCACAGATGGCCCAG ATACTTTCTT TCAATAAAAG TGGCGGTGGT





GGAGACATCC ACCAGGGCTTCCAGTCTCTT CTCACCGAAG TGAACAAGAC





TGGCACGCAG TACTTGCTTA GGATGGCCAACAGGCTCTTT GGGGAAAAGT





CTTGTGATTT CCTCTCATCT TTTAGAGATT CCTGCCAAAAATTCTACCAA





GCAGAGATGG AGGAGCTTGA CTTTATCAGC GCCGTAGAGA AGTCCAGAAA





ACACATAAAC ACCTGGGTAG CTGAAAAGAC AGAAGGTAAA ATTGCGGAGT





TGCTCTCTCCGGGCTCAGTG GATCCATTGA CAAGGCTGGT TCTGGTGAAT





GCTGTCTATT TCAGAGGAAACTGGGATGAA CAGTTTGACA AGGAGAACAC





CGAGGAGAGA CTGTTTAAAG TCAGCAAGAA





TGAGGAGAAA CCTGTGCAAA TGATGTTTAA GCAATCTACT TTTAAGAAGA





CCTATATAGGAGAAATATTT ACCCAAATCT TGGTGCTTCC ATATGTTGGC





AAGGAACTGA ATATGATCATCATGCTTCCG GACGAGACCA CTGACTTGAG





AACGGTGGAG AAAGAACTCA CTTACGAGAAGTTCGTAGAA TGGACGAGGC





TGGACATGAT GGATGAAGAG GAGGTGGAAG TGTCCCTCCCGCGGTTTAAA





CTAGAGGAAA GCTACGACAT GGAGAGTGTC CTGCGCAACC TGGGCATGAC





TGATGCCTTC GAGCTGGGCA AGGCAGACTT CTCTGGAATG TCCCAGACAG





ACCTGTCTCTGTCCAAGGTC GTGCACAAGT CTTTTGTGGA GGTCAATGAG





GAAGGCACGG AGGCTGCAGCCGCCACAGCT GCCATCATGA TGATGCGGTG





TGCCAGATTC GTCCCCCGCT TCTGCGCCGACCACCCCTTC CTTTTCTTCA





TCCAGCACAG CAAGACCAAC GGGATTCTCT TCTGCGGCCGCTTTTCCTCT





CCGTGA





SEQ ID No. 48



SerpinB6 variant 6 (Human) Protein



MDVLAEANGTFALNLLKTLGKDNSKNVFFSPMSMSCALAMVYMGAKGNTAAQ





MAQILSFNKSGGGGDIHQGFQSLLTEVNKTGTQYLLRMANRLFGEKSCDF





LSSFRDSCQKFYQAEMEELDFISAVEKSRKHINTWVAEKTEGKIAELLSPGSVDPL





TRLVLVNAVYFRGNWDEQFDKENTEERLFKVSKNEEKPVQMMFKQSTFKKTYI





GEIFTQILVLPYVGKELNMIIMLPDETTDLRTVEKELTYEKFVEWTRLDMMDEEE





VEVSLPRFKLEESYDMESVLRNLGMTDAFELGKADFSGMSQTDLSLSKVVHKSF





VEVNEEGTEAAAATAAIMMMRCARFVPRFCADHPFLFFIQHSKTNGILFCGRFSS





P





SEQ ID No. 49



SerpinB6 variant 6 (Human) cDNA



ATG GATGTTCTCGCAGAAGCAAA TGGCACCTTT GCCTTAAACC





TTTTGAAAAC GCTGGGTAAA GACAACTCGAAGAATGTGTT TTTCTCACCC





ATGAGCATGT CCTGTGCCCT GGCCATGGTC TACATGGGGGCAAAGGGAAA





CACCGCTGCA CAGATGGCCC AGATACTTTC TTTCAATAAA AGTGGCGGTG





GTGGAGACAT CCACCAGGGC TTCCAGTCTC TTCTCACCGA AGTGAACAAG





ACTGGCACGCAGTACTTGCT TAGGATGGCC AACAGGCTCT TTGGGGAAAA





GTCTTGTGAT TTCCTCTCATCTTTTAGAGA TTCCTGCCAA AAATTCTACC





AAGCAGAGAT GGAGGAGCTT GACTTTATCAGCGCCGTAGA GAAGTCCAGA





AAACACATAA ACACCTGGGT AGCTGAAAAG ACAGAAGGTAAAATTGCGGA





GTTGCTCTCT CCGGGCTCAG TGGATCCATT GACAAGGCTG GTTCTGGTGA





ATGCTGTCTA TTTCAGAGGA AACTGGGATG AACAGTTTGA CAAGGAGAAC





ACCGAGGAGAGACTGTTTAA AGTCAGCAAG AATGAGGAGA AACCTGTGCA





AATGATGTTT AAGCAATCTACTTTTAAGAA GACCTATATA GGAGAAATAT





TTACCCAAAT CTTGGTGCTT CCATATGTTGGCAAGGAACT GAATATGATC





ATCATGCTTC CGGACGAGAC CACTGACTTG AGAACGGTGGAGAAAGAACT





CACTTACGAG AAGTTCGTAG AATGGACGAG GCTGGACATG ATGGATGAAG





AGGAGGTGGA AGTGTCCCTC CCGCGGTTTA AACTAGAGGA AAGCTACGAC





ATGGAGAGTGTCCTGCGCAA CCTGGGCATG ACTGATGCCT TCGAGCTGGG





CAAGGCAGAC TTCTCTGGAATGTCCCAGAC AGACCTGTCT CTGTCCAAGG





TCGTGCACAA GTCTTTTGTG GAGGTCAATGAGGAAGGCAC GGAGGCTGCA





GCCGCCACAG CTGCCATCAT GATGATGCGG TGTGCCAGATTCGTCCCCCG





CTTCTGCGCC GACCACCCCT TCCTTTTCTT CATCCAGCAC AGCAAGACCA





ACGGGATTCT CTTCTGCGGC CGCTTTTCCT CTCCGTGA





SEQ ID No. 50



SerpinB6 variant 7 (Human) Protein



MDVLAEANGTFALNLLKTLGKDNSKNVFFSPMSMSCALAMVYMGAKGNTAAQ





MAQILSFNKSGGGGDIHQGFQSLLTEVNKTGTQYLLRMANRLFGEKSCDF





LSSFRDSCQKFYQAEMEELDFISAVEKSRKHINTWVAEKTEGKIAELLSPGSVDPL





TRLVLVNAVYFRGNWDEQFDKENTEERLFKVSKNEEKPVQMMFKQSTFKKTYI





GEIFTQILVLPYVGKELNMIIMLPDETTDLRTVEKELTYEKFVEWTRLDMMDEEE





VEVSLPRFKLEESYDMESVLRNLGMTDAFELGKADFSGMSQTDLSLSKVVHKSF





VEVNEEGTEAAAATAAIMMMRCARFVPRFCADHPFLFFIQHSKTNGILFCGRFSS





P





SEQ ID No. 51



SerpinB6 variant 7 (Human) cDNA



AT GGATGTTCTC GCAGAAGCAA ATGGCACCTTTGCCTTAAAC





CTTTTGAAAA CGCTGGGTAA AGACAACTCG AAGAATGTGT





TTTTCTCACCCATGAGCATG TCCTGTGCCC TGGCCATGGT CTACATGGGG





GCAAAGGGAA ACACCGCTGCACAGATGGCC CAGATACTTT CTTTCAATAA





AAGTGGCGGT GGTGGAGACA TCCACCAGGGCTTCCAGTCT CTTCTCACCG





AAGTGAACAA GACTGGCACG CAGTACTTGC TTAGGATGGCCAACAGGCTC





TTTGGGGAAA AGTCTTGTGA TTTCCTCTCA TCTTTTAGAG ATTCCTGCCA





AAAATTCTAC CAAGCAGAGA TGGAGGAGCT TGACTTTATC AGCGCCGTAG





AGAAGTCCAGAAAACACATA AACACCTGGG TAGCTGAAAA GACAGAAGGT





AAAATTGCGG AGTTGCTCTCTCCGGGCTCA GTGGATCCAT TGACAAGGCT





GGTTCTGGTG AATGCTGTCT ATTTCAGAGGAAACTGGGAT GAACAGTTTG





ACAAGGAGAA CACCGAGGAG AGACTGTTTA AAGTCAGCAAGAATGAGGAG





AAACCTGTGC AAATGATGTT TAAGCAATCT ACTTTTAAGA AGACCTATAT





AGGAGAAATA TTTACCCAAA TCTTGGTGCT TCCATATGTT GGCAAGGAAC





TGAATATGATCATCATGCTT CCGGACGAGA CCACTGACTT GAGAACGGTG





GAGAAAGAAC TCACTTACGAGAAGTTCGTA GAATGGACGA GGCTGGACAT





GATGGATGAA GAGGAGGTGG AAGTGTCCCTCCCGCGGTTT AAACTAGAGG





AAAGCTACGA CATGGAGAGT GTCCTGCGCA ACCTGGGCATGACTGATGCC





TTCGAGCTGG GCAAGGCAGA CTTCTCTGGA ATGTCCCAGA CAGACCTGTC





TCTGTCCAAG GTCGTGCACA AGTCTTTTGT GGAGGTCAAT GAGGAAGGCA





CGGAGGCTGCAGCCGCCACA GCTGCCATCA TGATGATGCG GTGTGCCAGA





TTCGTCCCCC GCTTCTGCGCCGACCACCCC TTCCTTTTCT TCATCCAGCA





CAGCAAGACC AACGGGATTC TCTTCTGCGGCCGCTTTTCC TCTCCGTGA





SEQ ID No. 52



SerpinB6 variant 8 (Human) Protein



MDVLAEANGTFALNLLKTLGKDNSKNVFFSPMSMSCALAMVYMGAKGNTAAQ





MAQILSFNKSGGGGDIHQGFQSLLTEVNKTGTQYLLRMANRLFGEKSCDF





LSSFRDSCQKFYQAEMEELDFISAVEKSRKHINTWVAEKTEGKIAELLSPGSVDPL





TRLVLVNAVYFRGNWDEQFDKENTEERLFKVSKNEEKPVQMMFKQSTFKKTYI





GEIFTQILVLPYVGKELNMIIMLPDETTDLRTVEKELTYEKFVEWTRLDMMDEEE





VEVSLPRFKLEESYDMESVLRNLGMTDAFELGKADFSGMSQTDLSLSKVVHKSF





VEVNEEGTEAAAATAAIMMMRCARFVPRFCADHPFLFFIQHSKTNGILFCGRFSS





P





SEQ ID No. 53



SerpinB6 variant 8 (Human) cDNA



ATGGAT GTTCTCGCAGAAGCAAATGG CACCTTTGCC TTAAACCTTT





TGAAAACGCT GGGTAAAGAC AACTCGAAGAATGTGTTTTT CTCACCCATG





AGCATGTCCT GTGCCCTGGC CATGGTCTAC ATGGGGGCAAAGGGAAACAC





CGCTGCACAG ATGGCCCAGA TACTTTCTTT CAATAAAAGT GGCGGTGGTG





GAGACATCCA CCAGGGCTTC CAGTCTCTTC TCACCGAAGT GAACAAGACT





GGCACGCAGTACTTGCTTAG GATGGCCAAC AGGCTCTTTG GGGAAAAGTC





TTGTGATTTC CTCTCATCTTTTAGAGATTC CTGCCAAAAA TTCTACCAAG





CAGAGATGGA GGAGCTTGAC TTTATCAGCGCCGTAGAGAA GTCCAGAAAA





CACATAAACA CCTGGGTAGC TGAAAAGACA GAAGGTAAAA





TTGCGGAGTT GCTCTCTCCG GGCTCAGTGG ATCCATTGAC AAGGCTGGTT





CTGGTGAATGCTGTCTATTT CAGAGGAAAC TGGGATGAAC AGTTTGACAA





GGAGAACACC GAGGAGAGACTGTTTAAAGT CAGCAAGAAT GAGGAGAAAC





CTGTGCAAAT GATGTTTAAG CAATCTACTTTTAAGAAGAC CTATATAGGA





GAAATATTTA CCCAAATCTT GGTGCTTCCA TATGTTGGCAAGGAACTGAA





TATGATCATC ATGCTTCCGG ACGAGACCAC TGACTTGAGA ACGGTGGAGA





AAGAACTCAC TTACGAGAAG TTCGTAGAAT GGACGAGGCT GGACATGATG





GATGAAGAGGAGGTGGAAGT GTCCCTCCCG CGGTTTAAAC TAGAGGAAAG





CTACGACATG GAGAGTGTCCTGCGCAACCT GGGCATGACT GATGCCTTCG





AGCTGGGCAA GGCAGACTTC TCTGGAATGTCCCAGACAGA CCTGTCTCTG





TCCAAGGTCG TGCACAAGTC TTTTGTGGAG GTCAATGAGGAAGGCACGGA





GGCTGCAGCC GCCACAGCTG CCATCATGAT GATGCGGTGT





GCCAGATTCGTCCCCCGCTT CTGCGCCGAC CACCCCTTCC TTTTCTTCAT





CCAGCACAGC AAGACCAACGGGATTCTCTT CTGCGGCCGC TTTTCCTCTC





CGTGA





SEQ ID No. 54



SerpinB9 (Human) Protein



METLSNASGTFAIRLLKILCQDNPSHNVFCSPVSISSALAMVLLGAKGNTATQMA





QALSLNTEEDIHRAFQSLLTEVNKAGTQYLLRTANRLFGEKTCQFLSTFKESCLQF





YHAELKELSFIRAAEESRKHINTWVSKKTEGKIEELLPGSSIDAETRLVLVNAIYFK





GKWNEPFDETYTREMPFKINQEEQRPVQMMYQEATFKLAHVGEVRAQLLE





LPYARKELSLLVLLPDDGVELSTVEKSLTFEKLTAWTKPDCMKSTEVEVLLPKFK





LQEDVDMESVLRHLGIVDAFQQGKADLSAMSAERDLCLSKFVHKSFVEVNEEGT





EAAAASSCFVVAECCMESGPRFCADHPFLFFIRHNRANSILFCGRFSSP





SEQ ID No. 55



SerpinB9 (Human) cDNA



ATGGAAAC TCTTTCTAAT GCAAGTGGTA CTTTTGCCAT ACGCCTTTTA





AAGATACTGTGTCAAGATAA CCCTTCGCAC AACGTGTTCT GTTCTCCTGT





GAGCATCTCC TCTGCCCTGGCCATGGTTCT CCTAGGGGCA AAGGGAAACA





CCGCAACCCA GATGGCCCAG GCACTGTCTTTAAACACAGA GGAAGACATT





CATCGGGCTT TCCAGTCGCT TCTCACTGAA GTGAACAAGGCTGGCACACA





GTACCTGCTG AGAACGGCCA ACAGGCTCTT TGGAGAGAAA ACTTGTCAGT





TCCTCTCAAC GTTTAAGGAA TCCTGTCTTC AATTCTACCA TGCTGAGCTG





AAGGAGCTTTCCTTTATCAG AGCTGCAGAA GAGTCCAGGA AACACATCAA





CACCTGGGTC TCAAAAAAGACCGAAGGTAA AATTGAAGAG TTGTTGCCGG





GTAGCTCAAT TGATGCAGAA ACCAGGCTGGTTCTTGTCAA TGCCATCTAC





TTCAAAGGAA AGTGGAATGA ACCGTTTGAC GAAACATACACAAGGGAAAT





GCCCTTTAAA ATAAACCAGG AGGAGCAAAG GCCAGTGCAG ATGATGTATC





AGGAGGCCAC GTTTAAGCTC GCCCACGTGG GCGAGGTGCG CGCGCAGCTG





CTGGAGCTGCCCTACGCCAG GAAGGAGCTG AGCCTGCTGG TGCTGCTGCC





TGACGACGGC GTGGAGCTCAGCACGGTGGA AAAAAGTCTC ACTTTTGAGA





AACTCACAGC CTGGACCAAG CCAGACTGTATGAAGAGTAC TGAGGTTGAA





GTTCTCCTTC CAAAATTTAA ACTACAAGAG GATTATGACATGGAATCTGT





GCTTCGGCAT TTGGGAATTG TTGATGCCTT CCAACAGGGC AAGGCTGACT





TGTCGGCAAT GTCAGCGGAG AGAGACCTGT GTCTGTCCAA GTTCGTGCAC





AAGAGTTTTGTGGAGGTGAA TGAAGAAGGC ACCGAGGCAG CGGCAGCGTC





GAGCTGCTTT GTAGTTGCAGAGTGCTGCAT GGAATCTGGC CCCAGGTTCT





GTGCTGACCA CCCTTTCCTT TTCTTCATCAGGCACAACAG AGCCAACAGC





ATTCTGTTCT GTGGCAGGTT CTCATCGCCA TAA





SEQ ID No. 56



Serping1 variant 1 (Human) Protein



MASRLTLLTLLLLLLAGDRASSNPNATSSSSQDPESLQDRGEGKVATTVISKMLF





VEPILEVSSLPTTNSTTNSATKITANTTDEPTTQPTTEPTTQPTIQPTQPTTQLPTDSP





TQPTTGSFCPGPVTLCSDLESHSTEAVLGDALVDFSLKLYHAFSAMKKVETNMA





FSPFSIASLLTQVLLGAGENTKTNLESILSYPKDFTCVHQALKGFTTKGVTSVSQIF





HSPDLAIRDTFVNASRTLYSSSPRVLSNNSDANLELINTWVAKNTNNKISRLLDSL





PSDTRLVLLNAIYLSAKWKTTFDPKKTRMEPFHFKNSVIKVPMMNSKKYPVAH





FIDQTLKAKVGQLQLSHNLSLVILVPQNLKHRLEDMEQALSPSVFKAIMEKLEMS





KFQPTLLTLPRIKVTTSQDMLSIMEKLEFFDFSYDLNLCGLTEDPDLQVSAMQHQ





TVLELTETGVEAAAASAISVARTLLVFEVQQPFLFVLWDQQHKFPVFMGRVYDP





RA





SEQ ID No. 57



Serping1 variant 1 (Human) cDNA



ATGGCCTCC AGGCTGACCC TGCTGACCCT CCTGCTGCTG CTGCTGGCTG





GGGATAGAGC CTCCTCAAAT CCAAATGCTA CCAGCTCCAG CTCCCAGGAT





CCAGAGAGTTTGCAAGACAG AGGCGAAGGG AAGGTCGCAA CAACAGTTAT





CTCCAAGATG CTATTCGTTGAACCCATCCT GGAGGTTTCC AGCTTGCCGA





CAACCAACTC AACAACCAAT TCAGCCACCAAAATAACAGC TAATACCACT





GATGAACCCA CCACACAACC CACCACAGAG CCCACCACCCAACCCACCAT





CCAACCCACC CAACCAACTA CCCAGCTCCC AACAGATTCT CCTACCCAGC





CCACTACTGG GTCCTTCTGC CCAGGACCTG TTACTCTCTG CTCTGACTTG





GAGAGTCATTCAACAGAGGC CGTGTTGGGG GATGCTTTGG TAGATTTCTC





CCTGAAGCTC TACCACGCCTTCTCAGCAAT GAAGAAGGTG GAGACCAACA





TGGCCTTTTC CCCATTCAGC ATCGCCAGCCTCCTTACCCA GGTCCTGCTC





GGGGCTGGGG AGAACACCAA AACAAACCTG GAGAGCATCC





TCTCTTACCC CAAGGACTTC ACCTGTGTCC ACCAGGCCCT GAAGGGCTTC





ACGACCAAAGGTGTCACCTC AGTCTCTCAG ATCTTCCACA GCCCAGACCT





GGCCATAAGG GACACCTTTGTGAATGCCTC TCGGACCCTG TACAGCAGCA





GCCCCAGAGT CCTAAGCAAC AACAGTGACGCCAACTTGGA GCTCATCAAC





ACCTGGGTGG CCAAGAACAC CAACAACAAG ATCAGCCGGCTGCTAGACAG





TCTGCCCTCC GATACCCGCC TTGTCCTCCT CAATGCTATC TACCTGAGTG





CCAAGTGGAA GACAACATTT GATCCCAAGA AAACCAGAAT GGAACCCTTT





CACTTCAAAAACTCAGTTAT AAAAGTGCCC ATGATGAATA GCAAGAAGTA





CCCTGTGGCC CATTTCATTGACCAAACTTT GAAAGCCAAG GTGGGGCAGC





TGCAGCTCTC CCACAATCTG AGTTTGGTGATCCTGGTACC CCAGAACCTG





AAACATCGTC TTGAAGACAT GGAACAGGCT CTCAGCCCTTCTGTTTTCAA





GGCCATCATG GAGAAACTGG AGATGTCCAA GTTCCAGCCC ACTCTCCTAA





CACTACCCCG CATCAAAGTG ACGACCAGCC AGGATATGCT CTCAATCATG





GAGAAATTGGAATTCTTCGA TTTTTCTTAT GACCTTAACC TGTGTGGGCT





GACAGAGGAC CCAGATCTTCAGGTTTCTGC GATGCAGCAC CAGACAGTGC





TGGAACTGAC AGAGACTGGG GTGGAGGCGGCTGCAGCCTC CGCCATCTCT





GTGGCCCGCA CCCTGCTGGT CTTTGAAGTG CAGCAGCCCTTCCTCTTCGT





GCTCTGGGAC CAGCAGCACA AGTTCCCTGT CTTCATGGGG CGAGTATATG





ACCCCAGGGC CTGA





SEQ ID No. 58



Serping1 variant 2 (Human) Protein



MASRLTLLTLLLLLLAGDRASSNPNATSSSSQDPESLQDRGEGKVATTVISKMLF





VEPILEVSSLPTTNSTTNSATKITANTTDEPTTQPTTEPTTQPTIQPTQPTTQLPTDSP





TQPTTGSFCPGPVTLCSDLESHSTEAVLGDALVDFSLKLYHAFSAMKKVETNMA





FSPFSIASLLTQVLLGAGENTKTNLESILSYPKDFTCVHQALKGFTTKGVTSVSQIF





HSPDLAIRDTFVNASRTLYSSSPRVLSNNSDANLELINTWVAKNTNNKISRLLDSL





PSDTRLVLLNAIYLSAKWKTTFDPKKTRMEPFHFKNSVIKVPMMNSKKYPVAH





FIDQTLKAKVGQLQLSHNLSLVILVPQNLKHRLEDMEQALSPSVFKAIMEKLEMS





KFQPTLLTLPRIKVTTSQDMLSIMEKLEFFDFSYDLNLCGLTEDPDLQVSAMQHQ





TVLELTETGVEAAAASAISVARTLLVFEVQQPFLFVLWDQQHKFPVFMGRVYDP





RA





SEQ ID No. 59



Serping1 variant 2 (Human) cDNA



A TGGCCTCCAG GCTGACCCTGCTGACCCTCC TGCTGCTGCT GCTGGCTGGG





GATAGAGCCT CCTCAAATCC AAATGCTACCAGCTCCAGCT CCCAGGATCC





AGAGAGTTTG CAAGACAGAG GCGAAGGGAA GGTCGCAACAACAGTTATCT





CCAAGATGCT ATTCGTTGAA CCCATCCTGG AGGTTTCCAG CTTGCCGACA





ACCAACTCAA CAACCAATTC AGCCACCAAA ATAACAGCTA ATACCACTGA





TGAACCCACCACACAACCCA CCACAGAGCC CACCACCCAA CCCACCATCC





AACCCACCCA ACCAACTACCCAGCTCCCAA CAGATTCTCC TACCCAGCCC





ACTACTGGGT CCTTCTGCCC AGGACCTGTTACTCTCTGCT CTGACTTGGA





GAGTCATTCA ACAGAGGCCG TGTTGGGGGA TGCTTTGGTAGATTTCTCCC





TGAAGCTCTA CCACGCCTTC TCAGCAATGA AGAAGGTGGA GACCAACATG





GCCTTTTCCC CATTCAGCAT CGCCAGCCTC CTTACCCAGG TCCTGCTCGG





GGCTGGGGAGAACACCAAAA CAAACCTGGA GAGCATCCTC TCTTACCCCA





AGGACTTCAC CTGTGTCCACCAGGCCCTGA AGGGCTTCAC GACCAAAGGT





GTCACCTCAG TCTCTCAGAT CTTCCACAGCCCAGACCTGG CCATAAGGGA





CACCTTTGTG AATGCCTCTC GGACCCTGTA CAGCAGCAGCCCCAGAGTCC





TAAGCAACAA CAGTGACGCC AACTTGGAGC TCATCAACAC CTGGGTGGCC





AAGAACACCA ACAACAAGAT CAGCCGGCTG CTAGACAGTC TGCCCTCCGA





TACCCGCCTTGTCCTCCTCA ATGCTATCTA CCTGAGTGCC AAGTGGAAGA





CAACATTTGA TCCCAAGAAAACCAGAATGG AACCCTTTCA CTTCAAAAAC





TCAGTTATAA AAGTGCCCAT GATGAATAGCAAGAAGTACC CTGTGGCCCA





TTTCATTGAC CAAACTTTGA AAGCCAAGGT GGGGCAGCTGCAGCTCTCCC





ACAATCTGAG TTTGGTGATC CTGGTACCCC AGAACCTGAA ACATCGTCTT





GAAGACATGG AACAGGCTCT CAGCCCTTCT GTTTTCAAGG CCATCATGGA





GAAACTGGAGATGTCCAAGT TCCAGCCCAC TCTCCTAACA CTACCCCGCA





TCAAAGTGAC GACCAGCCAGGATATGCTCT CAATCATGGA GAAATTGGAA





TTCTTCGATT TTTCTTATGA CCTTAACCTGTGTGGGCTGA CAGAGGACCC





AGATCTTCAG GTTTCTGCGA TGCAGCACCA GACAGTGCTGGAACTGACAG





AGACTGGGGT GGAGGCGGCT GCAGCCTCCG CCATCTCTGT GGCCCGCACC





CTGCTGGTCT TTGAAGTGCA GCAGCCCTTC CTCTTCGTGC TCTGGGACCA





GCAGCACAAGTTCCCTGTCT TCATGGGGCG AGTATATGAC CCCAGGGCCT





GA





SEQ ID No. 60



Serpini1 variant 1 (Human) Protein



MAFLGLFSLLVLQSMATGATFPEEAIADLSVNMYNRLRATGEDENILFSPLSIALA





MGMMELGAQGSTQKEIRHSMGYDSLKNGEEFSFLKEFSNMVTAKESQYVMKIA





NSLFVQNGFHVNEEFLQMMKKYFNAAVNHVDFSQNVAVANYINKWVENNTN





NLVKDLVSPRDFDAATYLALINAVYFKGNWKSQFRPENTRTFSFTKDDESEVQIP





MMYQQGEFYYGEFSDGSNEAGGIYQVLEIPYEGDEISMMLVLSRQEVPLATLEPL





VKAQLVEEWANSVKKQKVEVYLPRFTVEQEIDLKDVLKALGITEIFIKDANLTGL





SDNKEIFLSKAIHKSFLEVNEEGSEAAAVSGMIAISRMAVLYPQVIVDHPFFFLIRN





RRTGTILFMGRVMHPETMNTSGHDFEEL





SEQ ID No. 61



Serpini1 variant 1 (Human) cDNA



ATGGCTTTC CTTGGACTCT TCTCTTTGCT GGTTCTGCAA AGTATGGCTA





CAGGGGCCAC TTTCCCTGAG GAAGCCATTG CTGACTTGTC AGTGAATATG





TATAATCGTCTTAGAGCCAC TGGTGAAGAT GAAAATATTC TCTTCTCTCC





ATTGAGTATT GCTCTTGCAATGGGAATGAT GGAACTTGGG GCCCAAGGAT





CTACCCAGAA AGAAATCCGC CACTCAATGGGATATGACAG CCTAAAAAAT





GGTGAAGAAT TTTCTTTCTT GAAGGAGTTT TCAAACATGGTAACTGCTAA





AGAGAGCCAA TATGTGATGA AAATTGCCAA TTCCTTGTTT GTGCAAAATG





GATTTCATGT CAATGAGGAG TTTTTGCAAA TGATGAAAAA ATATTTTAAT





GCAGCAGTAAATCATGTGGA CTTCAGTCAA AATGTAGCCG TGGCCAACTA





CATCAATAAG TGGGTGGAGAATAACACAAA CAATCTGGTG AAAGATTTGG





TATCCCCAAG GGATTTTGAT GCTGCCACTTATCTGGCCCT CATTAATGCT





GTCTATTTCA AGGGGAACTG GAAGTCGCAG TTTAGGCCTGAAAATACTAG





AACCTTTTCT TTCACTAAAG ATGATGAAAG TGAAGTCCAA ATTCCAATGA





TGTATCAGCA AGGAGAATTT TATTATGGGG AATTTAGTGA TGGCTCCAAT





GAAGCTGGTGGTATCTACCA AGTCCTAGAA ATACCATATG AAGGAGATGA





AATAAGCATG ATGCTGGTGCTGTCCAGACA GGAAGTTCCT CTTGCTACTC





TGGAGCCATT AGTCAAAGCA CAGCTGGTTGAAGAATGGGC AAACTCTGTG





AAGAAGCAAA AAGTAGAAGT ATACCTGCCC AGGTTCACAGTGGAACAGGA





AATTGATTTA AAAGATGTTT TGAAGGCTCT TGGAATAACT GAAATTTTCA





TCAAAGATGC AAATTTGACA GGCCTCTCTG ATAATAAGGA GATTTTTCTT





TCCAAAGCAATTCACAAGTC CTTCCTAGAG GTTAATGAAG AAGGCTCAGA





AGCTGCTGCT GTCTCAGGAATGATTGCAAT TAGTAGGATG GCTGTGCTGT





ATCCTCAAGT TATTGTCGAC CATCCATTTTTCTTTCTTAT CAGAAACAGG





AGAACTGGTA CAATTCTATT CATGGGACGA GTCATGCATCCTGAAACAAT





GAACACAAGT GGACATGATT TCGAAGAACT TTAA





SEQ ID No. 62



Serpini1 variant 2 (Human) Protein



MAFLGLFSLLVLQSMATGATFPEEAIADLSVNMYNRLRATGEDENILFSPLSIALA





MGMMELGAQGSTQKEIRHSMGYDSLKNGEEFSFLKEFSNMVTAKESQYVMKIA





NSLFVQNGFHVNEEFLQMMKKYFNAAVNHVDFSQNVAVANYINKWVENNTN





NLVKDLVSPRDFDAATYLALINAVYFKGNWKSQFRPENTRTFSFTKDDESEVQIP





MMYQQGEFYYGEFSDGSNEAGGIYQVLEIPYEGDEISMMLVLSRQEVPLATLEPL





VKAQLVEEWANSVKKQKVEVYLPRFTVEQEIDLKDVLKALGITEIFIKDANLTGL





SDNKEIFLSKAIHKSFLEVNEEGSEAAAVSGMIAISRMAVLYPQVIVDHPFFFLIRN





RRTGTILFMGRVMHPETMNTSGHDFEEL





SEQ ID No. 63



Serpini1 variant 2 (Human) cDNA



AT GGCTTTCCTT GGACTCTTCT CTTTGCTGGT TCTGCAAAGTATGGCTACAG





GGGCCACTTT CCCTGAGGAA GCCATTGCTG ACTTGTCAGT GAATATGTAT





AATCGTCTTA GAGCCACTGG TGAAGATGAA AATATTCTCT TCTCTCCATT





GAGTATTGCTCTTGCAATGG GAATGATGGA ACTTGGGGCC CAAGGATCTA





CCCAGAAAGA AATCCGCCACTCAATGGGAT ATGACAGCCT AAAAAATGGT





GAAGAATTTT CTTTCTTGAA GGAGTTTTCAAACATGGTAA CTGCTAAAGA





GAGCCAATAT GTGATGAAAA TTGCCAATTC CTTGTTTGTGCAAAATGGAT





TTCATGTCAA TGAGGAGTTT TTGCAAATGA TGAAAAAATA TTTTAATGCA





GCAGTAAATC ATGTGGACTT CAGTCAAAAT GTAGCCGTGG CCAACTACAT





CAATAAGTGGGTGGAGAATA ACACAAACAA TCTGGTGAAA GATTTGGTAT





CCCCAAGGGA TTTTGATGCTGCCACTTATC TGGCCCTCAT TAATGCTGTC





TATTTCAAGG GGAACTGGAA GTCGCAGTTTAGGCCTGAAA ATACTAGAAC





CTTTTCTTTC ACTAAAGATG ATGAAAGTGA AGTCCAAATTCCAATGATGT





ATCAGCAAGG AGAATTTTAT TATGGGGAAT TTAGTGATGG CTCCAATGAA





GCTGGTGGTA TCTACCAAGT CCTAGAAATA CCATATGAAG GAGATGAAAT





AAGCATGATGCTGGTGCTGT CCAGACAGGA AGTTCCTCTT GCTACTCTGG





AGCCATTAGT CAAAGCACAGCTGGTTGAAG AATGGGCAAA CTCTGTGAAG





AAGCAAAAAG TAGAAGTATA CCTGCCCAGGTTCACAGTGG AACAGGAAAT





TGATTTAAAA GATGTTTTGA AGGCTCTTGG AATAACTGAAATTTTCATCA





AAGATGCAAA TTTGACAGGC CTCTCTGATA ATAAGGAGAT





TTTTCTTTCCAAAGCAATTC ACAAGTCCTT CCTAGAGGTT AATGAAGAAG





GCTCAGAAGC TGCTGCTGTCTCAGGAATGA TTGCAATTAG TAGGATGGCT





GTGCTGTATC CTCAAGTTAT TGTCGACCATCCATTTTTCT TTCTTATCAG





AAACAGGAGA ACTGGTACAA TTCTATTCAT GGGACGAGTCATGCATCCTG





AAACAATGAA CACAAGTGGA CATGATTTCG AAGAACTTTA A






All publications and patent documents disclosed or referred to herein are incorporated by reference in their entirety. The foregoing description has been presented only for purposes of illustration and description. This description is not intended to limit the invention to the precise form disclosed. It is intended that the scope of the invention be defined by the claims appended hereto.

Claims
  • 1. A method of treating an inflammatory condition in a subject, comprising administering a serpin protein and administering methylprednisolone.
  • 2. The method of claim 1, wherein said inflammatory condition is neuromyelitis optica (NMO).
  • 3. The method of claim 1, wherein said inflammatory condition is multiple sclerosis (MS).
  • 4. The method of claim 3, wherein said MS is progressive.
  • 5. The method of claim 1, wherein said inflammatory condition is Amyotrophic lateral sclerosis (ALS).
  • 6. The method of claim 1, wherein said serpin protein has at least a 90% sequence identity to SEQ ID NO:1 and has alpha-1 antitrypsin (A1AT) activity.
  • 7. The method of claim 6, wherein said serpin protein has the sequence of SEQ ID NO:1.
  • 8. The method of claim 1, wherein said serpin protein is encoded by a nucleic acid that has at least a 90% sequence identity to SEQ ID NO:2 and wherein said serpin protein has alpha-1 antitrypsin (A1AT) activity.
  • 9. The method of claim 8 wherein said serpin protein is encoded by a nucleic acid that has the sequence of SEQ ID NO:2.
  • 10. The method of claim 1, wherein said serpin protein and methylprednisolone are administered simultaneously.
  • 11. A method of treating an inflammatory condition in a subject, comprising administering a nucleic acid that encodes a serpin protein and administering methylprednisolone.
  • 12. The method of claim 11, wherein said inflammatory condition is neuromyelitis optica (NMO).
  • 13. The method of claim 11, wherein said inflammatory condition is multiple sclerosis (MS).
  • 14. The method of claim 13, wherein said MS is progressive.
  • 15. The method of claim 11, wherein said inflammatory condition is Amyotrophic lateral sclerosis (ALS).
  • 16. The method of claim 11, wherein said nucleic acid encodes a protein having at least a 90% sequence identity to SEQ ID NO:1 and has alpha-1 antitrypsin (A1AT) activity.
  • 17. The method of claim 16, wherein said nucleic acid encodes a protein having the sequence of SEQ ID NO:1.
  • 18. The method of claim 11, wherein said nucleic acid that has at least a 90% sequence identity to SEQ ID NO:2 and wherein said serpin protein has alpha-1 antitrypsin (A1AT) activity.
  • 19. The method of claim 18, wherein said nucleic acid has the sequence of SEQ ID NO:2.
  • 20. The method of claim 11, wherein said nucleic acid is administered by a route selected from the group consisting of transfected autologous patient cells, viral vectors, naked nucleic acid preparations, homologous recombination, knock-in, zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR).
  • 21. A method of treating an inflammatory condition in a subject, comprising increasing the expression of an endogenous serpin protein and administering methylprednisolone.
  • 22. The method of claim 21, wherein said inflammatory condition is neuromyelitis optica (NMO).
  • 23. The method of claim 21, wherein said inflammatory condition is multiple sclerosis (MS).
  • 24. The method of claim 23, wherein said MS is progressive.
  • 25. The method of claim 21, wherein said inflammatory condition is Amyotrophic lateral sclerosis (ALS).
  • 26. The method of claim 21, wherein said endogenous serpin protein has at least a 90% sequence identity to SEQ ID NO:1 and has alpha-1 antitrypsin (A1AT) activity.
  • 27. The method of claim 21, wherein said endogenous serpin protein has the sequence of SEQ ID NO:1.
  • 28. The method of claim 21, wherein said endogenous serpin protein is encoded by a nucleic acid that has at least a 90% sequence identity to SEQ ID NO:2 and wherein said serpin protein has alpha-1 antitrypsin (A1AT) activity.
  • 29. The method of claim 28, wherein said endogenous serpin protein is encoded by a nucleic acid that has the sequence of SEQ ID NO:2.
  • 30. The method of claim 21, wherein said increase in said serpin expression is accomplished using a technology selected from the group consisting of zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9.
  • 31. A pharmaceutical composition, comprising a serpin protein and methylprednisolone.
  • 32. The pharmaceutical composition of claim 31, wherein said serpin protein has at least a 90% sequence identity to SEQ ID NO:1 and has alpha-1 antitrypsin (A1AT) activity.
  • 33. The pharmaceutical composition of claim 32, wherein said serpin protein has the sequence of SEQ ID NO:1.
  • 34. The pharmaceutical composition of claim 31, wherein said serpin protein is encoded by a nucleic acid that has at least a 90% sequence identity to SEQ ID NO:2 and wherein said serpin protein has alpha-1 antitrypsin (A1AT) activity.
  • 35. The pharmaceutical composition of claim 34, wherein said serpin protein is encoded by a nucleic acid that has the sequence of SEQ ID NO:2.
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 62/131,073, filed on Mar. 10, 2015, incorporated herein by reference in its entirety.

PCT Information
Filing Document Filing Date Country Kind
PCT/US16/21412 3/9/2016 WO 00
Provisional Applications (1)
Number Date Country
62131073 Mar 2015 US