WO2005003302A2 - Novel methods to modulate mast cells - Google Patents

Novel methods to modulate mast cells Download PDF

Info

Publication number
WO2005003302A2
WO2005003302A2 PCT/US2004/019400 US2004019400W WO2005003302A2 WO 2005003302 A2 WO2005003302 A2 WO 2005003302A2 US 2004019400 W US2004019400 W US 2004019400W WO 2005003302 A2 WO2005003302 A2 WO 2005003302A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
haah
antibodies
cells
cell
Prior art date
Application number
PCT/US2004/019400
Other languages
French (fr)
Other versions
WO2005003302A3 (en
Inventor
Mike Mccarthy
Laura K. Richman
Brian Mullikin
Original Assignee
Medimmune, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Medimmune, Inc. filed Critical Medimmune, Inc.
Publication of WO2005003302A2 publication Critical patent/WO2005003302A2/en
Publication of WO2005003302A3 publication Critical patent/WO2005003302A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • the present invention is based, in part, on the inventors' discovery that human aspartyl (asparaginyl)- ⁇ hydroxylase (HAAH) is strongly expressed on the surface of mast cells.
  • the present invention also provides for prophylactic and therapeutic protocols designed to prevent, manage, treat, or ameliorate respiratory conditions (e.g. asthma) or one or more symptoms thereof.
  • the present invention further provides for prophylactic and therapeutic protocols designed to prevent, treat, manage or ameliorate an inflammatory or an autoimmune disorder or cardiovascular/coronary heart disease.
  • the present invention further provides methods for the prevention, management, treatment, or amelioration of respiratory conditions (e.g.
  • asthma or one or more symptoms thereof and/or to the treatment, management or amelioration of an inflammatory or an autoimmune disorder or cardiovascular disease
  • methods comprising administering to a subject in need thereof an effective amount of one or more agents that modulate the activity ofHAAH.
  • the agent that modulates HAAH activity immunospecifically binds to HAAH on the surface of the mast cell.
  • the modulation ofHAAH activity modulates mast cells.
  • the invention also provides combination therapies for the prevention, management, treatment or amelioration of respiratory conditions (e.g. asthma) and/or to treat, manage or ameliorate an inflammatory disorder or an autoimmune disorder or cardiovascular disease.
  • the invention further provides pharmaceutical compositions and articles of manufacture for use in the prevention, management, treatment, or amelioration of respiratory conditions or one or more symptoms thereof and/or the treatment, management or amelioration an inflammatory disorder or an autoimmune disorder or cardiovascular disease.
  • Mast cells are derived from CD34+ hematopoietic progenitor cells and except for the small number of mast cells that reside in the bone marrow, maturation typically occurs in the peripheral tissues. Williams et al., 2000 J. Allergy Clin. Immunol. 105:847-859. Mast cells are ideally positioned anatomically to participate in allergic reaction at cutanious or mucosal surfaces.
  • mast cells Unlike mature basophils, mature mast cells do not normally circulate in the blood but are widely distributed throughout normal connective tissues, where they often lie adjacent to blood and lymphatic vessels, near or within nerves, and beneath epithelial surfaces that are exposed to the external environment, such as those of the respiratory and gastrointestinal systems and skin. Id. Positioned at strategic points around capillaries and small blood vessels, mast cells are important in regulating the extent of constriction or dilation of the vessels including those which make up the blood-brain barrier, the protective lining of the brain which excludes toxic materials (Theoharides, 1990, Life Sciences 46:607-17). Mast cells produce an impressively broad array of mediators and cell-cell signaling molecules.
  • TNF- ⁇ tumor necrosis factor- ⁇
  • a specific program of gene expression is also activated leading to de novo synthesis of several cytokines (L -3, L -4, IL-5, L -6, L -9, L -10, IL-13, L -14), chemokines (macrophage inflammation protein l ⁇ , monocyte chemoattractant protein 1 (MCP-1) and lymphotactin) and more TNF- ⁇ .
  • cytokines L -3, L -4, IL-5, L -6, L -9, L -10, IL-13, L -14
  • chemokines microphage inflammation protein l ⁇ , monocyte chemoattractant protein 1 (MCP-1) and lymphotactin
  • MCP-1 monocyte chemoattractant protein 1
  • lymphotactin lymphotactin
  • mediators Compounds released by mast cell stimulation, collectively called mediators, include: histamine, kinins, prostaglandin D 2, tryptase and vasoactive intestinal peptide (VIP), which are vasodilatory, as well as serotonin, prostaglandin F2-alpha and leukotrienes, which are vasoconstrictive.
  • VIP vasoactive intestinal peptide
  • cytokines, histamine, kinins and prostaglandins can cause pain directly, while enzymes which destroy proteins and phospholipids can cause tissue damage directly.
  • cytokines such as LL-6 can cause inflammation and regulate other biological responses (Galli (1993) above).
  • mast cell responses One of the strongest triggers of mast cell responses are allergens complexed to immunoglobulin- ⁇ (IgE molecules. Because of the unusually high affinity to the Fc receptor for IgE, mast cells are constantly coated with antigen-specific IgE. The crosslinking of these surface bound IgE molecules by antigen leads to activation and degranulation. Anaphylatoxins generated by activation of the pathway are also potent activators of some mast cells.
  • IgE immunoglobulin- ⁇
  • cytokines and chemokines can also activate mast cells.
  • mast cells are best known for producing the "allergic" reactions, they have also been implicated in other autoimmune and inflammatory conditions. A number of studies have shown that chronic inflammation is often associated with mast cell hyperplasia and signs of mast cell activation. See Nalent et al., 2001 Immunological Review 179:74-81.
  • mast cell activation is found particularly in antigen-induced inflammation, as in autoimmune disorders or atopic diseases, hi addition, mast cells are known for their involvement in neuroinflammatory conditions that are precipitated or exacerbated by stress (Theoharides, et al., 1996 int. J. Tissue React. 18:1). Furthermore increased numbers of activated cardiac mast cells are found in ventricles, the sinusoidal node and the fibrous plaque associated with atherosclerosis and other cardiac disease states (reviewed in Marone et al, 1995, Immunopharm. 31:1-18).
  • Allergies are disorders of the immune system in which the body reacts to innocuous substances by inducing the generation of large amounts of IgE. the presence of an allergen, IgE activates mast cells and promotes mast cell proliferation, infiltration, and/or degranulation that results in the release of histamines, leukotrienes, and cytokines which cause rhinitis, hives, redness, itchiness, watery eyes, skin rashes, bronchoconstriction (wheezing), coughing, and difficulty breathing.
  • allergens include, but are not limited to, pollens, molds, dust (e.g., dust mites and dust mite waste), animal protein (e.g., dander, urine, oil from the skin), industrial chemicals, foods, medicines, feathers, and insects (e.g., insect stings, cockroaches, and insect waste).
  • dust e.g., dust mites and dust mite waste
  • animal protein e.g., dander, urine, oil from the skin
  • industrial chemicals e.g., foods, medicines, feathers, and insects (e.g., insect stings, cockroaches, and insect waste).
  • Pollinosis commonly known as hay fever, is generally induced by wind-borne pollens, including, but not limited to tree pollens (e.g., oak, elm, maple, alder, birch, juniper, and olive), grass pollens (e.g., Bermuda, timothy, sweet vernal, orchard, and Johnson), weed pollens (e.g., Russian thistle, English plantain, and ragweed), and airborne fungal spores.
  • tree pollens e.g., oak, elm, maple, alder, birch, juniper, and olive
  • grass pollens e.g., Bermuda, timothy, sweet vernal, orchard, and Johnson
  • weed pollens e.g., Russian thistle, English plantain, and ragweed
  • Symptoms of pollinosis include itchy nose, roof of the mouth, pharynx, and eyes, sneezing, runny nose, watery eyes, headaches, anorexia, depression, coughing, insomnia, and wheezing.
  • Common therapies include administration of antihistamines, sympathomimetics, glucocorticoids, and systemic corticosteroids and allergen immunotherapy. Unfortunately, these therapies have many limitations such as unwanted side effects such as hypertension and drowsiness.
  • Anaphylaxis is an acute allergic reaction that results when the allergen reaches the circulation.
  • Common allergens are parenteral enzymes, blood products, ⁇ -lactam antibiotics, allergen immunotherapy, and insect stings.
  • Anaphylaxis is characterized by smooth muscle contraction that causes wheezing, vasodilation, pulmonary edema, and obstructive angiodema. If the reaction is prolonged, the subject may develop arrhythmias or cardiogenic shock. In severe cases, the patient may suffer from primary cardiovascular collapse without respiratory symptoms. Long-term immunotherapy is effective for preventing anaphylaxis from insect stings, but is rarely available for patients with drug or serum anaphylaxis. Immediate administration of epinephrine is the most common treatment for anaphylaxis, but may cause side effects including headache, tremulousness, nausea, and arrhythmias. Thus, new therapies for the prevention, treatment, management, and amelioration of allergic reactions are needed.
  • Asthma About 12 million people in the U.S. have asthma and it is the leading cause of hospitalization for children. Tl%e Merck Manual of Diagnosis and Therapy (17th ed., 1999). Asthma is an inflammatory disease of the lung that is characterized by airway hyperresponsiveness ("AHR"), bronchoconstriction (i.e., wheezing), eosinophilic inflammation, mucus hypersecretion, subepithelial fibrosis, and elevated IgE levels. Asthmatic attacks can be triggered by environmental triggers (e.g.
  • acarids insects, animals (e.g., cats, dogs, rabbits, mice, rats, hamsters, guinea pigs, mice, rats, and birds), fungi, air pollutants (e.g., tobacco smoke), irritant gases, fumes, vapors, aerosols, or chemicals, or pollen), exercise, or cold air.
  • air pollutants e.g., tobacco smoke
  • irritant gases e.g., irritant gases
  • Cromolyn and nedocromil are administered prophylatically to inhibit mediator release from inflammatory cells, reduce airway hyperresponsiveness, and block responses to allergens.
  • therapies available that prevent the development of asthma in subjects at increased risk of developing asthma.
  • new therapies with fewer side effects and better prophylactic and/or therapeutic efficacy are needed for asthma.
  • Respiratory infections are common infections of the upper respiratory tract (e.g., nose, ears, sinuses, and throat) and lower respiratory tract (e.g., trachea, bronchial tubes, and lungs). Symptoms of upper respiratory infection include runny or stuffy nose, irritability, restlessness, poor appetite, decreased activity level, coughing, and fever. Viral upper respiratory infections cause and/or are associated with sore throats, colds, croup, and the flu. Examples of viruses that cause upper respiratory tract infections include rhino viruses and influenza viruses A and B. Common upper respiratory bacterial infections cause and/or associated with, for example, whooping cough and strep throat.
  • Streptococcus An example of a bacterium that causes an upper respiratory tract infection is Streptococcus.
  • Clinical manifestations of a lower respiratory infection include shallow coughing that produces sputum in the lungs, fever, and difficulty breathing.
  • lower respiratory viral infections are parainfluenza virus infections ("PIN”), respiratory syncytial virus (“RSN”), human metapneumovirus (bJVTPN), and bronchiolitis.
  • PIN parainfluenza virus infections
  • RSN respiratory syncytial virus
  • bJVTPN human metapneumovirus
  • bacteria that cause lower respiratory tract infections include Streptococcus pneumoniae that causes pneumonococcal pneumonia and Mycobacterium tuberculosis that causes tuberculosis.
  • Respiratory infections caused by fungi include systemic candidiasis, blastomycosis crytococcosis, coccidioidomycosis, and aspergiUosis. Respiratory infections may be primary or secondary infections.
  • Current therapies for respiratory infections involve the administration of anti-viral agents, anti-bacterial, and anti-fungal agents for the treatment, prevention, or amelioration of viral, bacterial, and fungal respiratory infections, respectively.
  • anti-viral agents anti-bacterial, and anti-fungal agents for the treatment, prevention, or amelioration of viral, bacterial, and fungal respiratory infections, respectively.
  • infections have been proven to be refractory to therapies, or the occurrence of side effects outweighs the benefits of the administration of a therapy to a subject.
  • the use of anti-bacterial agents for treatment of bacterial respiratory infections may also produce side effects or result in resistant bacterial strains.
  • anti-fungal agents may cause renal failure or bone marrow dysfunction and may not be effective against fungal infection in patients with suppressed immune systems.
  • the infection causing microorganism e.g., virus, bacterium, or fungus
  • the infection causing microorganism may be resistant or develop resistance to the administered therapeutic agent or combination of therapeutic agents.
  • microorganisms that develop resistance to administered therapeutic agents often develop pleiotropic drug or multidrug resistance, that is, resistance to therapeutic agents that act by mechanisms different from the mechanisms of the administered agents.
  • pleiotropic drug or multidrug resistance that is, resistance to therapeutic agents that act by mechanisms different from the mechanisms of the administered agents.
  • Inflammation is a process by which the body's white blood cells and chemicals protect our bodies from infection by foreign substances, such as bacteria and viruses. It is usually characterized by pain, swelling, warmth and redness of the affected area. Chemicals known as cytokines and prostaglandins control this process, and are released in an ordered and self- limiting cascade into the blood or affected tissues. This release of chemicals increases the blood flow to the area of injury or infection, and may result in the redness and warmth. Some of the chemicals cause a leak of fluid into the tissues, resulting in swelling. This protective process may stimulate nerves and cause pain. These changes, when occurring for a limited period in the relevant area, work to the benefit of the body.
  • the immune system triggers an inflammatory response when there are no foreign substances to fight and the body's normally protective immune system causes damage to its own tissues by mistakenly attacking self.
  • autoimmune disorders that affect the body in different ways.
  • the brain is affected in individuals with multiple sclerosis
  • the gut is affected in individuals with Crohn's disease
  • the synovium, bone, cartilage of various joints are affected in individuals with rheumatoid arthritis and the skin this psoriasis.
  • the autoimmune disorder may affect only one organ or tissue type or may affect multiple organs and tissues.
  • Organs and tissues commonly affected by autoimmune disorders include red blood cells, blood vessels, connective tissues, endocrine glands (e.g., the thyroid or pancreas), muscles, joints, and skin.
  • autoimmune disorders include, but are not limited to, Hashimoto's thyroiditis, pernicious anemia, Addison's disease, type 1 diabetes, rheumatoid arthritis, psoriasis, systemic lupus erythematosus, dermatomyositis, Sjogren's syndrome, dermatomyositis, lupus erythematosus, multiple sclerosis, autoimmune inner ear disease myasthenia gravis, Reiter's syndrome, Graves disease, autoimmune hepatitis, familial adenomatous polyposis and ulcerative colitis.
  • Rheumatoid arthritis and juvenile rheumatoid arthritis are types of inflammatory arthritis.
  • Arthritis is a general term that describes inflammation in joints. Some, but not all, types of arthritis are the result of misdirected inflammation.
  • other types of arthritis associated with inflammation include the following: psoriatic arthritis, Reiter's syndrome, ankylosing spondylitis arthritis, and gouty arthritis.
  • Rheumatoid arthritis is a type of chronic arthritis that occurs in joints on both sides of the body (such as both hands, wrists or knees). This symmetry helps distinguish rheumatoid arthritis from other types of arthritis.
  • rheumatoid arthritis may occasionally affect the skin, eyes, lungs, heart, blood or nerves.
  • Rheumatoid arthritis affects about 1% of the world's population and is potentially disabling. There are approximately 2.9 million incidences of rheumatoid arthritis in the United States. Two to three times more women are affected than men. The typical age that rheumatoid arthritis occurs is between 25 and 50. Juvenile rheumatoid arthritis affects 71,000 young Americans (aged eighteen and under), affecting six times as many girls as boys.
  • Rheumatoid arthritis is an autoimmune disorder where the body's immune system improperly identifies the synovial membranes that secrete the lubricating fluid in the joints as foreign. Inflammation results, and the cartilage and tissues in and around the joints are damaged or destroyed. In severe cases, this inflammation extends to other joint tissues and surrounding cartilage, where it may erode or destroy bone and cartilage and lead to joint deformities. The body replaces damaged tissue with scar tissue, causing the normal spaces within the joints to become narrow and the bones to fuse together. Rheumatoid arthritis creates 1 stiffness, swelling, fatigue, anemia, weight loss, fever, and often, crippling pain.
  • rheumatoid arthritis Some common symptoms of rheumatoid arthritis include joint stiffness upon awakening that lasts an hour or longer; swelling in a specific finger or wrist joints; swelling in the soft tissue around the joints; and swelling on both sides of the joint. Swelling can occur with or without pain, and can worsen progressively or remain the same for years before progressing.
  • the diagnosis of rheumatoid arthritis is based on a combination of factors, including: the specific location and symmetry of painful joints, the presence of joint stiffness in the morning, the presence of bumps and nodules under the skin (rheumatoid nodules), results of X-ray tests that suggest rheumatoid arthritis, and/or positive results of a blood test called the rheumatoid factor.
  • rheumatoid arthritis Many, but not all, people with rheumatoid arthritis have the rheumatoid-factor antibody in their blood.
  • the rheumatoid factor may be present in people who do not have rheumatoid arthritis.
  • Other diseases can also cause the rheumatoid factor to be produced in the blood. That is why the diagnosis of rheumatoid arthritis is based on a combination of several factors and not just the presence of the rheumatoid factor, in the blood.
  • the typical course of the disease is one of persistent but fluctuating joint symptoms, and after about 10 years, 90% of sufferers will show structural damage to bone and cartilage.
  • a small percentage will have a short illness that clears up completely, and another small percentage will have very severe disease with many joint deformities, and occasionally other manifestations of the disease.
  • the inflammatory process causes erosion or destruction of bone and cartilage in the joints.
  • rheumatoid arthritis there is an autoimmune cycle of persistent antigen presentation, T-cell stimulation, cytokine secretion, synovial cell activation, and joint destruction.
  • the disease has a major impact on both the individual and society, causing significant pain, impaired function and disability, as well as costing millions of dollars in healthcare expenses and lost wages. (See, for example, the NTH website and the MAID website).
  • Currently available therapy for arthritis focuses on reducing inflammation of the joints with anti-inflammatory or immunosuppressive medications.
  • the first line of treatment of any arthritis is usually anti-inflammatories, such as aspirin, ibuprofen and Cox-2 inhibitors such as celecoxib and rofecoxib.
  • "Second line drugs” include gold, methotrexate and steroids. Although these are well-established treatments for arthritis, very few patients remit on these lines of treatment alone.
  • methotrexate in combination with antibodies to cytokines or recombinant soluble receptors.
  • recombinant soluble receptors for tumor necrosis factor (TNF)- ⁇ have been used in combination with methotrexate in the treatment of arthritis.
  • Multiple sclerosis a chronic inflammatory disorder of the central nervous system, which is characterized by a breach of the blood-brain barrier, mononuclear cell infiltration of white matter and eventual demyelinization.
  • a similar autoimmune disease can be induced in susceptible rodent strains by injecting myelin components, including myelin basic protein, proteolipid protein and mylelin oligodendrocyte glycoprotein.
  • myelin components including myelin basic protein, proteolipid protein and mylelin oligodendrocyte glycoprotein.
  • Cardiac Disease Atherosclerosis is a disease-state in which the walls of the arteries become thickened and lose their elasticity, resulting in a decreased ability to pump blood to peripheral organs and withstand systemic pressure.
  • the most common form of atherosclerosis is caused by the build- up of fatty deposits in the innermost layer of the artery wall.
  • Another form of atherosclerosis disease involves the destruction of the smaller arteries, or arterioles. All forms of the disease contribute to a decreased blood flow to vital organs, which can lead to a stroke, heart attack, or kidney failure.
  • cardiovascular diseases such as atherosclerosis and for the predisposition of unstable angina, myocardial infarction and stroke include, cigarette smoking, hypertension, dyslipidemia, diabetes mellitus, sedentary lifestyle, obesity, the imbalance of the hemostatic/fibrinolytic system and a family history of premature coronary disease.
  • these risk factors explain only a portion of the documented cardiovascular disease.
  • Other factors must play a role in the etiology of the vascular events in this disease.
  • immunologic mechanisms also play a role in the pathogenesis of cardiovascular disease. For example, it has been shown that coronary inflammation may depend on activated mast cell-derived mediators (Laine et al., 1998, J. Pharm.
  • immunological cells include mast cells, while immunological mediators and messengers include cytokines (e.g.,L-4, LL-13, GM-CSF, TNFa, IL-6), chemokines, adhesion molecules, and mast cells mediators (leukotrienes, histamine). Harris et al. further report that cardiac mast cell activation can occur, and that down-regulation of systemic inflammation by anti- inflammatory therapies of the present invention lowers the risk and/or severity of atherosclerosis and cardiovascular disease.
  • cytokines e.g.,L-4, LL-13, GM-CSF, TNFa, IL-6
  • chemokines e.g., chemokines
  • adhesion molecules e.g., IL-6
  • mast cells mediators leukotrienes, histamine.
  • Harris et al. further report that cardiac mast cell activation can occur, and that down-regulation of systemic inflammation by anti- inflammatory therapies of the present invention lowers the risk and/or severity
  • HAAH human aspartyl (asparaginyl)- ⁇ hydroxylase
  • HAAH is a protein belonging to the ⁇ -ketoglutarate dependent dioxygenase family of prolyl and lysyl hydroxylases which play a key role in collagen biosynthesis. This molecule hydroxylates aspartic acid or asparagine residues in EGF-like domains of several proteins in the presence of ferrous iron. These EGF-like domains contain conserved motifs that form repetitive sequences in proteins such as clotting factors, extracellular matrix proteins, LDL receptor, NOTCH homologues or NOTCH ligand homologues.
  • the 4.3-kb cDNA encoding the human aspartyl (asparaginyl)- ⁇ hydroxylase hybridizes with 2.6 kb and 4.3 kb transcripts in transformed cells, and the deduced amino acid sequence of the larger transcript encodes a protein of about 85-kDa. Both in vitro transcription and translation and Western blot analysis also demonstrate a 56-kDa protein that may result from posttranslational cleavage of the catalytic C terminus.
  • HAAH has several conserved functional domains including Aspartyl beta- hydroxylase N-terminal and Aspartyl/ Asparaginyl beta-hydroxylase C-terminal (residues 43- 309 and 589-746) regions that are critical for catalytic activity as well as a central tetratricopeptide repeat domain (residues 421-520) which likely plays a role in mediating protein-protein interactions, i addition, HAAH contains a hydrophobic region located between residues 54 and 75 and flanked by positively charged amino acid residues at its amino-terminal side and negatively charged residues at its carboxyl-terminal side that is a signal anchor of a type ⁇ transmembrane protein (Schutze et al., 1994, EMBO J.
  • the predicted orientation of the HAAH polypeptide is that residues 1-45 of the NH 2 terminus are in the cytoplasm and the remainder of the molecule, containing the catalytic domain, in the lumen of the endoplasmic reticulum and/or on the cell surface.
  • a physiological function ofHAAH is the post-translational beta- hydroxylation of aspartic acid in vitamin K-dependent coagulation proteins.
  • the abundant expression ofHAAH in several malignant neoplasms, and the low levels ofHAAH in many normal cells indicate a role for this enzyme in malignancy.
  • the HAAH gene is also highly expressed in cytotrophoblasts, but not syncytiotrophoblasts of the placenta.
  • Cytotrophoblasts are invasive cells that mediate placental implantation.
  • the increased levels ofHAAH expression in human cholangiocarcinomas, hepatocellular carcinomas, colon cancers, and breast carcinomas were primarily associated with invasive or metastatic lesions.
  • overexpression ofHAAH does not strictly reflect increased DNA synthesis and cellular proliferation since high levels ofHAAH immunoreactivity were observed in 100 percent of cholangiocarcinomas, but not in human or experimental disease processes associated with regeneration or nonneoplastic proliferation of bile ducts.
  • HAAH overexpression and attendant high levels of beta hydroxylase activity lead to invasive growth of transformed neoplastic cells.
  • HAAH adenosine triphosphate
  • a mast cell adenosine-containing HAAH
  • ADCC antibody mediated-cellular cytotoxicity
  • apoptosis by agonizing or antagonizing HAAH to induce mast cell death.
  • the agent that modulates HAAH activity immunospecifically binds to HAAH on the surface of the mast cell.
  • the agent that modulates HAAH activity is an inhibitor of HAAH activity.
  • HAAH inhibitors of the invention include, without limitation, antibodies, peptides, small molecules, antisense molecules, inhibitory RNA, and ribozymes, which are capable of inhibiting or preventing 1) the activity of a HAAH molecule of the invention on mast cell activity or development; or 2) the binding of a HAAH molecule with other binding partners; or 3) the biological activity of a HAAH molecule of the invention; or 4) the expression of a HAAH molecule of the invention by a cell.
  • the present invention comprises methods for identifying compounds capable of depleting mast cells and/or preventing mast cell activation, wherein said compounds immunospecifically bind to HAAH on the surface of the mast cell.
  • the present invention comprises methods for identifying compounds capable of depleting mast cells and/or preventing mast cell activation, where in said compounds immunospecifically bind to HAAH on the surface of the mast cells and exhibit little or no binding to the surface of other hematopoeitic cells that are not mast cells or related cells or cell lines or derived cell lines thereof.
  • the invention also encompasses a method for identifying compounds that immunospecifically binds to HAAH and are capable of depleting mast cells and/or preventing mast cell activation, wherein said compounds are non-toxic for other hematopoeitic cells that are not mast cells or related cells or cell lines or derived cell lines thereof.
  • the present invention also provides for prophylactic and therapeutic protocols designed to prevent, manage, treat, or ameliorate respiratory conditions (e.g. asthma) or one or more symptoms thereof and to treat, manage or ameliorate an inflammatory or an autoimmune disorder or cardiac disease.
  • respiratory conditions e.g. asthma
  • the present invention provides methods for the prevention, management, treatment, or amelioration of respiratory conditions (e.g.
  • the respiratory condition or inflammatory disease being treated includes but is of not limited to asthma, chronic obstructive pulmonary disease, interstitial lung disease, chronic obstructive lung disease, chronic bronchitis, eosinophilic bronchitis, eosinophilic pneumonia, pneumonia, inflammatory bowel disease, atopic dermatitis, atopy, allergy, allergic rhinitis, idiopathic pulmonary fibrosis, scleroderma, rheumatoid arthritis and emphysema.
  • the cardiac disease is a coronary heart disease (e.g. atherosclerosis).
  • a coronary heart disease e.g. atherosclerosis.
  • FIGURE 1 hrrmunohistochemistry of normal human tissues stained with an anti- HAAH specific antibody.
  • FIGURE 2 is a Western blot of whole cell lysates. Molecular weight marker sizes are indicated and the arrow represents the position ofHAAH.
  • FIGURE 3 is FACS analysis of UT-7 cells. The cells were labeled with A) no antibody. B) Secondary antibody alone. C) Anti-HAAH and secondary antibodies.
  • FIGURE 4 is FACS analysis of KU812 cells.
  • the cells were labeled with A) anti- HAAH and secondary antibodies.
  • DETAILED DESCRIPTION OF THE INVENTION The present invention is based, in part, on the inventors' discovery that human aspartyl (asparaginyl)- ⁇ hydroxylase (HAAH) is strongly expressed on the surface of mast cells.
  • the invention provides prophylactic and therapeutic protocols for the prevention, treatment, management, or amelioration of respiratory conditions (e.g.
  • HAAH antagonists of the invention can include, but should not be construed as being limited to a chemical compound, a protein, a peptidomemetic, an antibody, a ribozyme, and an antisense nucleic acid molecule.
  • HAAH antagonists of the invention includes, but not limited to, antibodies, antibody fragments, single chain antibodies, small molecules, fusion proteins, and BiTE molecules.
  • the antagonists of the invention can modulate mast cell activity by preventing activation, degranulation or stimulation, or by inducing apoptosis, antibody mediated cell cytotoxicity (ADCC), complement mediated killing, a molecule conjugated to a toxin or drug (e.g. antibody conjugated to ricin), and/or direct killing of mast cells (e.g. small molecule).
  • ADCC antibody mediated cell cytotoxicity
  • complement mediated killing a molecule conjugated to a toxin or drug (e.g. antibody conjugated to ricin)
  • direct killing of mast cells e.g. small molecule.
  • Antibodies or fragments thereof that immunospecifically bind to HAAH and modulate mast cells are specifically preferred embodiments of the invention.
  • antibodies or fragments thereof that immunospecifically bind to the HAAH polypeptides, or a fragment thereof do not cross-react with other non-HAAH antigens.
  • an antibody or fragment thereof that immunospecifically binds to the human HAAH could also bind the ortholog of other species (i.e. mouse, rat, chimp).
  • Preferred anti-HAAH antibodies of the invention recognize epitopes present on the extracellular portion of the protein.
  • a more preferred anti-HAAH antibody of the invention will bind to the catalytic domain ofHAAH.
  • a still more preferred anti-HAAH antibody of the invention will inhibit the activity ofHAAH.
  • Another preferred anti-HAAH antibody will bind to the tetratricopeptide repeat domain ofHAAH.
  • Yet another preferred anti-HAAH antibody will bind to HAAH and prevent interaction with HAAH binding partners.
  • Antibodies or fragments that immunospecifically bind to HAAH polypeptides, or a fragment thereof can be identified, for example, by immunoassays or other techniques known to those of skill in the art.
  • Antibodies of the invention include, but are not limited to, synthetic antibodies, monoclonal antibodies, recombinantly produced antibodies, intrabodies, multispecific antibodies, human antibodies, humanized antibodies, chimeric antibodies, synthetic antibodies, single-chain Fvs (scFv), Fab fragments, F (ab') fragments, disulfide- linked Fvs (sdFv), and anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
  • scFv single-chain Fvs
  • Fab fragments fragments
  • F (ab') fragments fragments
  • disulfide- linked Fvs sdFv
  • anti-Id anti-idiotypic antibodies
  • antibodies of the present invention include immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen-binding site that immunospecifically binds to a HAAH poiypeptide (e.g., one or more complementarity determining regions (CDRs) of an anti-HAAH antibody).
  • a HAAH poiypeptide e.g., one or more complementarity determining regions (CDRs) of an anti-HAAH antibody.
  • CDRs complementarity determining regions
  • inhibitory or antagonistic antibodies or fragments that immunospecifically bind to a HAAH polypeptide, or a fragment thereof preferentially modulate mast cells, and do not significantly modulate other cells types.
  • “Modulate mast cells”, as used herein, includes but is not limited to changes in, mast cell maturation, differentiation, proliferation, activation, stimulation, migration, degranulation, as well as mast cell inhibition and depletion.
  • HAAH antagonists of the invention result in a depletion of mast cells or prevent mast cell activation.
  • the invention provides prophylactic and therapeutic protocols for the prevention treatment, management, or amelioration of respiratory conditions (e.g. asthma) or one or more symptoms thereof and/or to treat, manage or ameliorate an inflammatory or an autoimmune disorder, said protocols comprising administering to a subject in need thereof a prophylactically or therapeutically effective amount of an HAAH antibody alone or in combination with a prophylactically or therapeutically effective amount of at least one other therapy (e.g., at least one other prophylactic or therapeutic agent) other than an HAAH antibody.
  • respiratory conditions e.g. asthma
  • at least one other therapy e.g., at least one other prophylactic or therapeutic agent
  • the present invention also provides for pharmaceutical compositions, kits, and articles of manufacture comprising one or more HAAH antagonists for use in the prevention, treatment, management, or amelioration of respiratory conditions and/or an inflammatory or autoimmune disease or one or more symptoms thereof.
  • the present invention also provides for pharmaceutical compositions, kits, and articles of manufacture comprising one or more HAAH antagonists (e.g. antibodies, small molecules, fusion proteins and/or BiTE molecule) and one or more prophylactic or therapeutic agents other than HAAH antagonists for use in prevention, treatment, management, or amelioration of respiratory conditions or one or more symptoms thereof.
  • HAAH antagonists e.g. antibodies, small molecules, fusion proteins and/or BiTE molecule
  • the invention encompasses administration of antibodies (preferably monoclonal antibodies) or fragments thereof that immunospecifically bind to HAAH polypeptides and modulate mast cells.
  • Antibodies of the invention include, but are not limited to, monoclonal antibodies, synthetic antibodies, recombinantly produced antibodies, intrabodies, multispecific antibodies (including bi-specific antibodies), human antibodies, humanized antibodies, chimeric antibodies, single-chain Fvs (scFv) (including bi-specific scFvs), single chain antibodies, Fab fragments, F (ab') fragments, disulfide-linked Fvs (sdFv), and epitope-binding fragments of any of the above.
  • antibodies used in the methods of the present invention include immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that immunospecifically binds to HAAH or a fragment thereof, and is a modulator of mast cells (e.g., proliferation, differentiation, activation, degranulation, migration).
  • antibodies of the present invention preferentially bind a HAAH polypeptide epitope, and/or binds a HAAH polypeptide of the invention with a K off of less than 3 X 10 "3 s "1 .
  • the immunoglobulin molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGi, IgG 2 , IgG 3 , IgG 4 , IgAi and IgA 2 ) or subclass of immunoglobulin molecule.
  • the antibodies used in the methods of the invention maybe from any animal origin including birds and mammals (e.g., human, murine, donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken).
  • the antibodies are human or humanized monoclonal antibodies.
  • human antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from mice or other animals that express antibodies from human genes.
  • the antibodies used in the methods of the present invention may be monospecific, bispecific, trispecific or of greater multispecificity.
  • Multispecific antibodies may immunospecifically bind to different epitopes of a HAAH polypeptide or may immunospecifically bind to both a HAAH polypeptide as well a heterologous epitope, such as a heterologous polypeptide or solid support material. See, e.g., International Publication Nos.
  • the antibodies used in the methods of the invention include derivatives that are modified, i.e., by the covalent attachment of any type of molecule to the antibody.
  • the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to, specific chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc. Additionally, the derivative may contain one or more non-classical amino acids.
  • the present invention encompasses single domain antibodies, including camelized single domain antibodies (see e.g., Muyldermans et al., 2001, Trends Biochem. Sci. 26:230; Nuttall et al., 2000, Cur. Pharm. Biotech. 1:253; Reichmann and Muyldermans, 1999, J. Immunol. Meth. 231:25; International Publication Nos. WO 94/04678 and WO 94/25591; U.S. Patent No. 6,005,079; which are incorporated herein by reference in their entireties).
  • the present invention provides single domain antibodies comprising two NH domains having the amino acid sequence of any of the NH domains of a HAAH antibody with modifications such that single domain antibodies are formed.
  • the present invention also provides single domain antibodies comprising two NH domains comprising one or more of the NH CDRs of an HAAH antibody.
  • the methods of the present invention also encompass the use of antibodies or fragments thereof that have half-lives (e.g., serum half-lives) in a mammal, preferably a human, of greater than 15 days, preferably greater than 20 days, greater than 25 days, greater than 30 days, greater than 35 days, greater than 40 days, greater than 45 days, greater than 2 months, greater than 3 months, greater than 4 months, or greater than 5 months.
  • the increased half-lives of the antibodies of the present invention or fragments thereof in a mammal, preferably a human, result in a higher serum titer of said antibodies or antibody fragments in the mammal, and thus, reduce the frequency of the administration of said antibodies or antibody fragments and/or reduces the concentration of said antibodies or antibody fragments to be administered.
  • Antibodies or fragments thereof having increased in vivo half-lives can be generated by techniques known to those of skill in the art.
  • antibodies or fragments thereof with increased in vivo half-lives can be generated by modifying (e.g., substituting, deleting or adding) amino acid residues identified as involved in the interaction between the Fc domain and the FcRn receptor (see, e.g., international Publication ⁇ os. WO 97/34631 and WO 02/060919, which are incorporated herein by reference in their entireties).
  • Antibodies or fragments thereof with increased in vivo half-lives can be generated by attaching to said antibodies or antibody fragments polymer molecules such as high molecular weight polyethyleneglycol (PEG).
  • PEG high molecular weight polyethyleneglycol
  • PEG can be attached to said antibodies or antibody fragments with or without a multifunctional linker either through site-specific conjugation of the PEG to the N- or C- terminus of said antibodies or antibody fragments or via epsilon- amino groups present on lysine residues.
  • Linear or branched polymer derivatization that results in minimal loss of biological activity will be used.
  • the degree of conjugation will be closely monitored by SDS-PAGE and mass spectrometry to ensure proper conjugation of PEG molecules to the antibodies.
  • Unreacted PEG can be separated from antibody-PEG conjugates by, e.g., size exclusion or ion-exchange chromatography.
  • the derivatives include less than 15 amino acid substitutions, less than 10 amino acid substitutions, less than 5 amino acid substitutions, less than 4 amino acid substitutions, less than 3 amino acid substitutions, or less than 2 amino acid substitutions relative to the original antibody or fragment thereof.
  • the derivatives have conservative amino acid substitutions made at one or more predicted non- essential amino acid residues.
  • the present invention further encompasses antibodies or fragments thereof that immunospecifically bind to HAAH and modulated a mast cells (e.g. inhibit mast cell activity), said antibodies or antibody fragments comprising an amino acid sequence of one or more CDRs that is at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% identical to the amino acid sequence of one or more CDRs of a HAAH antibody.
  • the determination of percent identity of two amino acid sequences can be determined by any method known to one skilled in the art, including BLAST protein searches.
  • the present invention encompasses the use of antibodies or fragments thereof that bind to HAAH recombinantly fused or chemically conjugated (including both covalent and non-covalent conjugations) to a heterologous agent.
  • the heterologous agent may be a polypeptide (or portion thereof, preferably to a polypeptide of at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90 or at least 100 amino acids), nucleic acid, small molecule (less than 1000 daltons), or inorganic or organic compound.
  • the fusion does not necessarily need to be direct, but may occur through linker sequences.
  • antibodies may be used to target heterologous polypeptides to particular cell types, either in vitro or in vivo, by fusing or conjugating the antibodies to antibodies specific for particular cell surface receptors.
  • Antibodies fused or conjugated to heterologous polypeptides may also be used in in vitro immunoassays and purification methods using methods known in the art. See e.g., International Publication WO 93/21232; EP 439,095; Naramura et al., 1994, Immunol. Lett. 39:91-99; U.S. Patent 5,474,981; Gillies et al., 1992, PNAS 89:1428-1432; and Fell et al, 1991, J. Immunol.
  • the present invention further includes compositions comprising heterologous agents fused or conjugated to antibody fragments.
  • the heterologous polypeptides may be fused or conjugated to a Fab fragment, Fd fragment; Fv fragment, F (ab) 2 fragment, or portion thereof.
  • Methods for fusing or conjugating polypeptides to antibody portions are known in the art. See, e.g., U.S. Patent Nos. 5,336,603, 5,622,929, 5,359,046, 5,349,053, 5,447,851, and 5,112,946; EP 307,434; EP 367,166; International Publication Nos.
  • WO 96/04388 and WO 91/06570 Ashkenazi et al., 1991, PNAS 88: 10535-10539; Zheng et al., 1995, J. Immunol. 154:5590-5600; and Nil et al, 1992, PNAS 89:11337- 11341 (said references incorporated by reference in their entireties). Additional fusion proteins may be generated through the techniques of gene-shuffling, motif-shuffling, exon-shuffling, and/or codon-shuffling (collectively referred to as "D ⁇ A shuffling").
  • D ⁇ A shuffling may be employed to alter the activities of antibodies of the invention or fragments thereof (e.g., antibodies or fragments thereof with higher affinities and lower dissociation rates). See, generally, U.S. Patent os. 5,605,793; 5,811,238; 5,830,721; 5,834,252; and 5,837,458, and Patten et al, 1997, Curr. Opinion Biotechnol. 8:724-33; Harayama, 1998, Trends Biotechnol. 16:76; Hansson, et al., 1999, J. Mol. Biol.
  • Antibodies or fragments thereof, or the encoded antibodies or fragments thereof may be altered by being subjected to random mutagenesis by error-prone PCR, random nucleotide insertion or other methods prior to recombination.
  • One or more portions of a polynucleotide encoding an antibody or antibody fragment, which portions immunospecifically bind to HAAH may be recombined with one or more components, motifs, sections, parts, domains, fragments, etc. of one or more heterologous agents.
  • antibodies of the present invention or fragments or variants thereof are conjugated to a marker sequence, such as a peptide, to facilitate purification.
  • the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available. As described in Gentz et al., 1989, PNAS 86:821, for instance, hexa-histidine provides for convenient purification of the fusion protein.
  • peptide tags useful for purification include, but are not limited to, the hemagglutinin "HA” tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al, 1984, Cell 37:767) and the "Flag" tag.
  • antibodies of the present invention or fragments or variants thereof are conjugated to a diagnostic or detectable agent. Such antibodies can be useful for monitoring or prognosing the development or progression of an asthma and/or autoimmune or inflammatory disease as part of a clinical testing procedure, such as determining the efficacy of a particular therapy.
  • Such diagnosis and detection can accomplished by coupling the antibody to detectable substances including, but not limited to various enzymes, such as but not limited to horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; prosthetic groups, such as but not limited to streptavidin/biotin and avidin/biotin; fluorescent materials, such as but not limited to, umbelliferone, fluorescein, fluorescein isothiocynate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; luminescent materials, such as but not limited to, luminol; bioluminescent materials, such as but not limited to, luciferase, luciferin, and aequorin; radioactive materials, such as but not limited to, bismuth ( 213 Bi), carbon ( 14 C), chromium ( 51 Cr), cobalt ( 57 Co
  • antibodies of the present invention or fragments or variants thereof are conjugated to a therapeutic agent such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters.
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to cells.
  • Examples include paclitaxel, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1- dehydrotestosterone, glucocorticoids, procaine, tetracaine, onconase, lidocaine, propranolol, puromycin, epirubicin, and cyclophosphamide and analogs or homologs thereof.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BCNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cisdichlorodiamine platinum (11) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincri
  • antibodies of the present invention or fragments or variants thereof are conjugated to a therapeutic agent or drug moiety that modifies a given biological response.
  • Therapeutic agents or drug moieties are not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, cholera toxin, or diphtheria toxin; a protein such as tumor necrosis factor, ⁇ -interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF- ⁇ , TNF- ⁇ , AIM I (see, International Publication No. WO 97/33899), AIM II (see, International Publication No. WO 97/34911), Fas Ligand (Takahashi et al, 1994, J.
  • a toxin such as abrin, ricin A, pseudomonas exotoxin, cholera toxin, or diphtheria toxin
  • a protein such as tumor necrosis factor, ⁇ -interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor
  • a thrombotic agent or an anti-angiogenic agent e.g., angiostatin or endostatin
  • a biological response modifier such as, for example, a lymphokine (e.g., interleukin-1 ("IL-1"), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), and granulocyte colony stimulating factor (“G-CSF”)), or a growth factor (e.g., growth hormone (“GH”)).
  • IL-1 interleukin-1
  • IL-2 interleukin-2
  • IL-6 interleukin-6
  • GM-CSF granulocyte macrophage colony stimulating factor
  • G-CSF granulocyte colony stimulating factor
  • GH growth hormone
  • antibodies of the present invention or fragments or variants thereof are conjugated to a therapeutic agent such as a radioactive materials or macrocyclic chelators useful for conjugating radiometal ions (see above for examples of radioactive materials).
  • a therapeutic agent such as a radioactive materials or macrocyclic chelators useful for conjugating radiometal ions (see above for examples of radioactive materials).
  • the macrocyclic chelator is 1,4,7,10- tetraazacyclododecane-N,N',N",N"-tetraacetic acid (DOTA) which can be attached to the antibody via a linker molecule.
  • linker molecules are commonly known in the art and described in Denardo et al., 1998, Clin Cancer Res. 4:2483-90; Peterson et al, 1999, Bioconjug. Chem.
  • an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described by Segal in U.S. Patent No.
  • antibodies of the invention are bispecific T cell engagers (BiTE).
  • Bispecific T cell engagers are bispecific antibodies that can redirect T cells for antigen-specific elimination of targets.
  • a BiTE molecule has an antigen-binding domain that binds to a T cell antigen (e.g. CD3) at one end of the molecule and an antigen binding domain that will bind to an antigen on the target cell.
  • a BiTE molecule was recently described in WO 99/54440, which is herein incorporated by reference.
  • This publication describes a novel single-chain multifunctional polypeptide that comprises binding sites for the CD19 and CD3 antigens (CD19xCD3).
  • This molecule was derived from two antibodies, one that binds to CD 19 on the B cell and an antibody that binds to CD3 on the T cells.
  • the variable regions of these different antibodies are linked by a polypeptide sequence, thus creating a single molecule.
  • Also described, is the linking of the variable heavy chain (NH) and light chain (NL) of a specific binding domain with a flexible linker to create a single chain, bispecific antibody.
  • an antibody or ligand that immunospecifically binds to HAAH will comprise a portion of the BiTE molecule.
  • the NH and/or NL (preferably a scFN) of an antibody that binds HAAH can be fused to an anti-CD3 binding portion such as that of the molecule described above, thus creating a BiTE molecule that targets HAAH on the mast cells.
  • other molecules that bind HAAH can comprise the BiTE molecule.
  • the BiTE molecule can comprise a molecule that binds to other T cell antigens (other than CD3).
  • ligands and/or antibodies that immunospecifically bind to T-cell antigens like CD2, CD4, CD8, CDl la, TCR, and CD28 are contemplated to be part of this invention.
  • binding domain denotes a domain comprising a three-dimensional structure capable of specifically binding to an epitope like native antibodies, free scFv fragments or one of their corresponding immunoglobulin chains, preferably the NH chain.
  • said domain can comprise the NH and/or NL domain of an antibody or an immunoglobulin chain, preferably at least the NH domain or more preferably the NH and NL domain linked by a flexible polypeptide linker (scFv).
  • said binding domain contained in the polypeptide of the invention may comprise at least one complementarity determining region (CDR) of an antibody or immunoglobulin chain recognizing an antigen on the T cell or a cellular antigen.
  • CDR complementarity determining region
  • the binding domain present in the polypeptide of the invention may not only be derived from antibodies but also from other T cell or cellular antigen binding protein, such as naturally occurring surface receptors or ligands.
  • said first and or second domain of the above- described polypeptide mimic or correspond to a NH and NL region from a natural antibody.
  • the antibody providing the binding site for the polypeptide of the invention can be, e.g., a monoclonal antibody, polyclonal antibody, chimeric antibody, humanized antibody, bispecific antibody, synthetic antibody, antibody fragment, such as Fab, Fv or scFv fragments etc., or a chemically modified derivative of any of these.
  • Antibodies may also be attached to solid supports, which are particularly useful for immunoassays or purification of the target antigen. Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • the antibodies or fragments thereof can be produced by any method known in the art for the synthesis of antibodies, in particular, by chemical synthesis or preferably, by recombinant expression techniques.
  • Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof.
  • monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed.
  • the term "monoclonal antibody” as used herein is not limited to antibodies produced through hybridoma technology.
  • the term “monoclonal antibody” refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
  • mice can be immunized with HAAH (either the full length protein or a domain thereof, e.g., the catalytic domain) and once an immune response is detected, e.g., antibodies specific for HAAH are detected in the mouse serum, the mouse spleen is harvested and splenocytes isolated. The splenocytes are then fused by well known techniques to any suitable myeloma cells, for example cells from cell line SP20 available from the ATCC. Hybridomas are selected and cloned by limited dilution.
  • HAAH either the full length protein or a domain thereof, e.g., the catalytic domain
  • Hybridoma clones are then assayed by methods known in the art for cells that secrete antibodies capable of binding a polypeptide of the invention.
  • Ascites fluid which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones.
  • Antigenic epitopes ofHAAH preferably contain a sequence of at least 4, at least 5, at least 6, at least 7, more preferably at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, and, most preferably, between about 15 to about 30 amino acids.
  • Preferred polypeptides comprising immunogenic or antigenic epitopes are at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 amino acid residues in length. These antigenic epitopes can be used to raise antibodies; including monoclonal antibodies, that specifically bind the epitope. Preferred antigenic epitopes include the antigenic epitopes disclosed herein, as well as any combination of two, three, four, five or more of these antigenic epitopes. Antigenic epitopes can be used as the target molecules in immunoassays.
  • the polypeptides comprising one or more immunogenic epitopes may be presented for eliciting an antibody response together with a carrier protein, such as an albumin, to an animal system (such as rabbit or mouse), or, if the polypeptide is of sufficient length (at least about 25 amino acids), the polypeptide may be presented without a carrier.
  • a carrier protein such as an albumin
  • immunogenic epitopes comprising as few as 8 to 10 amino acids have been shown to be sufficient to raise antibodies capable of binding to, at the very least, linear epitopes in a denatured polypeptide (e.g., in Western blotting).
  • monoclonal antibodies can be generated by culturing a hybridoma cell secreting an antibody of the invention wherein, preferably, the hybridoma is generated by fusing splenocytes isolated from a mouse immunized with HAAH or fragment thereof with myeloma cells and then screening the hybridomas resulting from the fusion for hybridoma clones that secrete an antibody able to bind HAAH.
  • Antibody fragments which recognize specific HAAH epitopes may be generated by any technique known to those of skill in the art.
  • Fab and F(ab')2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments).
  • F(ab')2 fragments contain the variable region, the light chain constant region and the CHI domain of the heavy chain.
  • the antibodies of the present invention can also be generated using various phage display methods known in the art. hi phage display methods, functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them.
  • DNA sequences encoding VH and NL domains are amplified from animal cD ⁇ A libraries (e.g., human or murine cD ⁇ A libraries of lymphoid tissues).
  • the D ⁇ A encoding the NH and NL domains are recombined together with an scFv linker by PCR and cloned into a phagemid vector (e.g., p CANTAB 6 or pComb 3 HSS).
  • the vector is electroporated in E. coli and the E. coli is infected with helper phage.
  • Phage used in these methods are typically filamentous phage including fd and Ml 3 and the VH and VL domains are usually recombinantly fused to either the phage gene m or gene NHL Phage expressing an antigen binding domain that binds to the HAAH epitope of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead.
  • Examples of phage display methods that can be used to make the antibodies of the present invention include those disclosed in Brinkman et al., 1995, J. Immunol. Methods 182:41-50; Ames et al., 1995, J. Immunol.
  • the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described below.
  • Fab, Fab' and F(ab')2 fragments can also be employed using methods known in the art such as those disclosed in International Publication No. WO 92/22324; Mullinax et al., 1992, BioTechniques 12:864; Sawai et al., 1995, AJRI 34:26; and Better et al., 1988, Science 240:1041 (said references incorporated by reference in their entireties).
  • the nucleotide sequence encoding an antibody of the invention can be obtained from sequencing hybridoma clone DNA.
  • a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA library, or a cDNA library generated from, or nucleic acid, preferably poly A+ RNA, isolated from any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody) by PCR amplification using synthetic primers that hybridize to the 3' and 5 'ends of the sequence or by cloning using an ohgonucleotide probe specific for the particular gene sequence to identify, e.g., a cDNA clone from a cDNA library that encodes the antibody.
  • a suitable source e.g., an antibody cDNA library, or a cDNA library generated from, or nucleic acid, preferably poly A+ RNA, isolated from any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody
  • Amplified nucleic acids generated by PCR may then be cloned into replicable cloning vectors using any method well known in the art.
  • the nucleotide sequence of the antibody may be manipulated using methods well known in the art for the manipulation of nucleotide sequences, e.g. recombinant DNA techniques, site directed mutagenesis, PCR, etc. (see, Or example, the techniques described in Current Protocols in Molecular Biology, F.M. Ausubel et al., ed., John Wiley & Sons (Chichester, England, 1998); Molecular Cloning: A Laboratory Manual, 3nd Edition, J.
  • N H and N L nucleotide sequences are cloned and used to generate whole antibodies.
  • the PCR primers including N H or N nucleotide sequences, a restriction site, and a flanking sequence to protect the restriction site are used to amplify the N H or N L sequences in scFv.
  • the PCR amplified N H domains are cloned into vectors expressing a N H constant region, e.g., the human gamma 4 constant region
  • the PCR amplified N L domains are cloned into vectors expressing a N constant region, e.g., human kappa or lambda constant regions.
  • the N H and N L domains may also be cloned into one vector expressing the necessary constant regions.
  • the heavy chain conversion vectors and light chain conversion vectors are then co-transfected into cell lines to generate stable or transient cell lines that express full-length antibodies, e.g., IgG, using techniques known to those of skill in the art. It is specifically contemplated that for some uses, including in vivo use of antibodies in humans and in vitro detection assays, it may be preferable to use human, chimeric or humanized antibodies. Completely human antibodies are particularly desirable for therapeutic treatment of human subjects. Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See also U.S. Patent ⁇ os. 4,444,887 and 4,716,111; and International Publication ⁇ os.
  • Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes.
  • the human heavy and light chain immunoglobulin gene complexes may be introduced randomly or by homologous recombination into mouse embryonic stem cells.
  • the human variable region, constant region, and diversity region may be introduced into mouse embryonic stem cells in addition to the human heavy and light chain genes.
  • the mouse heavy and light chain immunoglobulin genes may be rendered non-functional separately or simultaneously with the introduction of human immunoglobulin loci by homologous recombination. In particular, homozygous deletion of the JH region prevents endogenous antibody production.
  • the modified embryonic stem cells are expanded and microinjected into blastocysts to produce chimeric mice. The chimeric mice are then bred to produce homozygous offspring which express human antibodies.
  • the transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a polypeptide of the invention. Monoclonal antibodies directed against the antigen can be obtained from the immunized, transgenic mice using conventional hybridoma technology.
  • the human immunoglobulin transgenes harbored by the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation.
  • Lonberg and Huszar (1995, Int. Rev. Immunol.. 13:65-93).
  • this technology for producing human antibodies and human monoclonal antibodies and protocols for producing such antibodies see, e.g., International Publication Nos. WO 98/24893, WO 96/34096, and WO 96/33735; and U.S. Patent Nos.
  • a chimeric antibody is a molecule in which different portions of the antibody are derived from different immunoglobulin molecules such as antibodies having a variable region derived from a non-human antibody and a human immunoglobulin constant region. Methods for producing chimeric antibodies are known in the art.
  • Humanized are antibodies comprising one or more CDRs from a non-human species and framework regions from a human immunoglobulin molecule can be produced using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; International Publication No. WO 91/09967; and U.S. Patent Nos.
  • a humanized antibody is an antibody or its variant or fragment thereof which is capable of binding to a predetermined antigen and which comprises a framework region having substantially the amino acid sequence of a human immunoglobulin and a CDR having substantially the amino acid sequence of a non-human immunoglobulin.
  • a humanized antibody comprises substantially all of at least one, and typically two, variable domains in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin (i.e., donor antibody) and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence.
  • a humanized antibody also comprises at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • the antibody will contain both the light chain as well as at least the variable domain of a heavy chain.
  • the antibody also may include the CHI, hinge, CH2, CH3, and CH4 regions of the heavy chain.
  • the humanized antibody can be selected from any class of immunoglobulins, including IgM, IgG, IgD, IgA and IgE, and any isotype, including IgG l5 IgG 2 , IgG 3 and IgG 4 .
  • the constant domain is a complement fixing constant domain where it is desired that the humanized antibody exhibit cytotoxic activity, and the class is typically IgG Where such cytotoxic activity is not desirable, the constant domain may be of the IgG 2 class.
  • the humanized antibody may comprise sequences from more than one class or isotype, and selecting particular constant domains to optimize desired effector functions is within the ordinary skill in the art.
  • the framework and CDR regions of a humanized antibody need not correspond precisely to the parental sequences, e.g., the donor CDR or the consensus framework maybe mutagenized by substitution, insertion or deletion of at least one residue so that the CDR or framework residue at that site does not correspond to either the consensus or the import antibody. Such mutations, however, will not be extensive. Usually, at least 75% of the humanized antibody residues will correspond to those of the parental framework region (FR) and CDR sequences, more often 90%, and most preferably greater than 95%. Humanized antibodies can be produced using variety of techniques known in the art, including but not limited to, CDR-grafting (European Patent No. EP 239,400; International Publication No. WO 91/09967; and U.S. Patent Nos.
  • framework residues in the framework regions will be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding.
  • framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Patent No.
  • the antibodies of the invention can, in turn, be utilized to generate anti- idiotype antibodies using techniques well known to those skilled in the art. (See, e.g., Greenspan & Bona, 1989, FASEB J. 7:437-444; and Nissinoff, 1991, J. Immunol. 147:2429- 2438).
  • the invention provides methods employing the use of polynucleotides comprising a nucleotide sequence encoding an antibody of the invention or a fragment thereof.
  • the antibodies of the invention can be produced by any method known in the art for the synthesis of antibodies, in particular, by chemical synthesis or preferably, by recombinant expression techniques.
  • Recombinant expression of a HAAH specific antibody of the invention, or fragment, derivative or analog thereof, e.g., a heavy or light chain of an antibody of the invention requires construction of an expression vector containing a polynucleotide that encodes the antibody.
  • the vector for the production of the antibody molecule may be produced by recombinant DNA technology using techniques well known in the art.
  • methods for preparing a protein by expressing a polynucleotide containing an antibody encoding nucleotide sequence are described herein. Methods which are well known to those skilled in the art can be used to construct expression vectors containing antibody coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.
  • the invention thus, provides replicable vectors comprising a nucleotide sequence encoding an antibody molecule of the invention, or a heavy or light chain thereof, or a heavy or light chain variable domain, operably linked to a promoter.
  • Such vectors may include the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., PCT Publication WO 86/05807; PCT Publication WO 89/01036; and U.S. Pat. No. 5,122,464) and the variable domain of the antibody may be cloned into such a vector for expression of the entire heavy or light chain.
  • the expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce a HAAH specific antibody of the invention.
  • the invention includes host cells containing a polynucleotide encoding an antibody of the invention, or a heavy or light chain thereof, operably linked to a heterologous promoter.
  • vectors encoding both the heavy and light chains may be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below.
  • the antibody expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce a substituted antibody have improved producibility.
  • host-expression vector systems may be utilized to express the antibody molecules of the invention.
  • Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences express an antibody molecule of the invention in situ.
  • the invention includes host cells containing a polynucleotide encoding an antibody of the invention, or a heavy or light chain thereof, operably linked to a heterologous promoter.
  • vectors encoding both the heavy and light chains may be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below.
  • host-expression vector systems may be utilized to express the antibody molecules of the invention.
  • Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express an antibody molecule of the invention in situ.
  • These include but are not limited to microorganisms such as bacteria (e.g., E. coli, B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMN; tobacco mosaic virus, TMN) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or
  • bacterial cells such as Escherichia coli , and more preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule.
  • mammalian cells such as Chinese hamster ovary cells (CHO)
  • CHO Chinese hamster ovary cells
  • a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., 1986, Gene 45:101; Cockett et al., 1990, Bio/Technology 8:2).
  • a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed.
  • vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable.
  • Such vectors include, but are not limited, to the E. coli expression vector pUR278 (Ruther et al., 1983, EMBO J . 2: 1791), in which the antibody coding sequence may be ligated individually into the vector in frame with the lacZ coding region so that a fusion protein is produced; pT ⁇ vectors (Inouye & friouye, 1985, Nucleic Acids Res. 13:3101-9; Nan Heeke & Schuster, 1989, J. Biol. Chem.
  • pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to a matrix glutathione-agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • Ad ⁇ PN Autographa californica nuclear polyhedrosis virus
  • the virus grows in Spodoptera frugiperda cells.
  • the antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an Ac ⁇ PN promoter (for example the polyhedrin promoter).
  • an Ac ⁇ PN promoter for example the polyhedrin promoter
  • a number of viral-based expression systems maybe utilized.
  • the antibody coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination.
  • Insertion in a non- essential region of the viral genome will result in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts, (e.g., see Logan & Shenk, 1984, Proc. Natl. Acad. Sci. USA 81:355-9).
  • Specific initiation signals may also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert.
  • These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic.
  • the efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bitter et al., 1987, Methods in Enzymol., 153:516-44).
  • a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g. glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed.
  • eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of-the gene product may be used.
  • mammalian host cells include but are not limited to CHO, VERY, BHK, HeLa, COS, MDCK, 293, 3T3, W138, and in particular, breast cancer cell lines such as, for example, BT483, Hs578T, HTB2, BT20 and T47D, and normal mammary gland cell line such as, for example, CRL7030 andHs578Bst.
  • stable expression is preferred.
  • cell lines which stably express the antibody molecule may be engineered.
  • host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • appropriate expression control elements e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.
  • engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines which express the antibody molecule.
  • Such engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the antibody molecule.
  • a number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler et al., 1977, Cell 11 :223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, 1992, Proc. Natl. Acad. Sci. USA 48:202), and adenine phosphoribosyltransferase (Lowy et al., 1980, Cell 22:817) genes can be employed in tk-, hgprt- or aprt-cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al., 1980, Proc. Natl. Acad. Sci. USA 77:357; O'Hare et al., 1981, Proc. Natl. Acad. Sci. USA 78:1527); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, 1981, Proc. Natl. Acad. Sci. USA 78:2072); neo, which confers resistance to the aminoglycoside G-418 (Wu and Wu, 1991, Biotherapy 3:87-95; Tolstoshev, 1993, Ann. Rev.
  • the host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide.
  • the two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides.
  • a single vector may be used which encodes both heavy and light chain polypeptides. hi such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, 1986, Nature 322:52; Kohler, 1980, Proc. Natl.
  • the coding sequences for the heavy and light chains may comprise cDNA or genomic DNA.
  • an antibody molecule of the invention may be purified by any method known in the art for purification of an immunoglobulin molecule, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • Polypeptides of the invention include, but are not limited to, HAAH polypeptides exemplified by GenBank Ace. No. AAB50779, including full-length polypeptides, fragments, variants or derivatives and epitopes of the full-length HAAH. Specifically contemplated are polypeptides comprising the luminal/extracellular domains ofHAAH.
  • HAAH luminal/extracellular domains include but are not limited to, the Aspartyl beta-hydroxylase N-terminal and Aspartyl/ Asparaginyl beta-hydroxylase C-terminal (residues 43-309 and 589- 746) regions that are critical for catalytic activity as well as a central tetratricopeptide repeat domain (residues 421-520).
  • Polynucleotides of the invention include, but are not limited to, HAAH polynucleotides exemplified by GenBank Ace. No. NM_004318, including full-length polynucleotides, fragments, variants or derivatives and epitopes of the full-length HAAH.
  • transcript variants (exemplified by GenBank Ace. Nos. NM_004318, NM_020164, NM_032466, NM B2467, NM_032468) which vary in protein translation, the coding of catalytic domains, and tissue expression. Variation among these transcripts impacts their functions which involve roles in the calcium storage and release process in the endoplasmic and sarcoplasmic reticulum as well as hydroxylation of aspartic acid and asparagine in epidermal growth factor-like domains of various proteins.
  • polypeptides of the present invention may include full-length polypeptides, fragments, variants or derivatives and epitopes of HAAH splice variants.
  • Polypeptides of the invention further include, but are not limited to, polypeptide antagonists ofHAAH polypeptides.
  • the present invention further relates to polynucleotides which hybridize to the hereinabove-described sequences if there is at least 70%, preferably at least 90%, and more preferably at least 95% identity between the sequences.
  • the present invention particularly relates to polynucleotides which hybridize under stringent conditions to the hereinabove- described polynucleotides.
  • polypeptide of the present invention are could be fused to other heterologous sequences. These fusion proteins can be used for a variety of applications. For example, fusion of the present polypeptides to His-tag, HA-tag, protein A, IgG domains, and maltose binding protein facilitates purification. (See EP A 394,827; Traunecker, et al., 1988, Nature 331:84-86). Similarly, fusion to IgG-1, IgG-3, and albumin increases the half life time in vivo.
  • Nuclear localization signals fused to the polypeptides of the present invention can target the protein to a specific subcellular localization, while covalent heterodimer or homodimers can increase or decrease the activity of a fusion protein. Fusion proteins can also create chimeric molecules having more than one function. Finally, fusion proteins can increase solubility and/or stability of the fused protein compared to the non-fused protein. All of the types of fusion proteins described above can be made by modifying the following protocol, which outlines the fusion of a polypeptide to an IgG molecule. Briefly, the human Fc portion of the IgG molecule can be PCR amplified.
  • primers also should have convenient restriction enzyme sites that will facilitate cloning into an expression vector, preferably a mammalian expression vector.
  • an expression vector preferably a mammalian expression vector.
  • pC4 Accession No. 209646
  • the human Fc portion can be ligated into the BamHI cloning site. Note that the 3' BamHI site should be destroyed.
  • the vector containing the human Fc portion is re-restricted with BamHI, linearizing the vector, and a polynucleotide of the present invention, isolated by the PCR, is ligated into this BamHI site.
  • the polynucleotide is cloned without a stop codon, otherwise a fusion protein will not be produced ⁇ If the naturally occurring signal sequence is used to produce the secreted protein, pC4 does not need a second signal peptide. Alternatively, if the naturally occurring signal sequence is not used, the vector can be modified to include a heterologous signal sequence. (See, e.g., WO 96/34891.) As one of skill in the art will appreciate, and as discussed above, the polypeptides of the present invention comprising an immunogenic or antigenic epitope can be fused to other polypeptide sequences.
  • polypeptides of the present invention maybe fused with the constant domain of inrmunoglobulins (IgA, IgE, IgG, IgM), or portions thereof (CHI, CH2, CH3, or any combination thereof and portions thereof) resulting in chimeric polypeptides.
  • IgA, IgE, IgG, IgM inrmunoglobulins
  • CHI constant domain of inrmunoglobulins
  • CH2, CH3 any combination thereof and portions thereof
  • antigens e.g., insulin
  • FcRn binding partner such as IgG or Fc fragments
  • IgG Fusion proteins that have a disulfide-linked dimeric structure due to the IgG portion disulfide bonds have also been found to be more efficient in binding and neutralizing other molecules than monomeric polypeptides or fragments thereof alone. See, e.g., Fountoulakis et al., 1995, J. Biochem., 270:3958-3964.
  • the present invention comprises methods for identifying compounds capable of depleting mast cells, wherein said compounds are immunospecifically binds to HAAH on the surface of the mast cell and not other hematopoeitic cells that are not mast cells or related cells or cell lines or derived cell lines thereof, comprising the steps consisting of: a) culturing mast cells in vitro in a suitable culture medium, b) adding to said culture medium at least one candidate compound to be tested and incubating said cells for a prolonged period of time, c) measuring the extent to which said compounds promote mast cells death or disrupt, interfere with, or inhibit mast cells growth, and selecting compounds for which mast cells depletion is observed, d) identifying a compound of a subset of a compound selected in step c) that promote significant death of a mast death or disrupt, interfere with, or inhibit mast cells growth, and selecting compounds for which mast cells depletion is observed.
  • the invention also encompasses to a method for identifying compounds that immunospecifically binds to HAAH and are capable of depleting mast cells, wherein said compounds are non-toxic for other hematopoeitic cells that are not mast cells or related cells or cell lines or derived cell lines thereof, comprising the step consisting of: a) providing a culture of mast cells, wherein said mast cells are selected from wild type mast cells and cell lines derived thereof, activated mutant mast cell lines, and activated wild type mast cells and cell lines derived thereof, b) contacting the culture of said cells with at least one candidate compound under conditions allowing growth and/or survival of mast cells, measuring the level of cell death in the presence of the candidate compound; and comparing the level of cell death in the presence of the candidate compound to the level of cell death in the absence of the candidate compound, wherein an increase in the level of cell death in the presence of the candidate compound is indicative of the mast cells depletion ability of the candidate compound, c) providing a culture of at least one cell other than mast cells, wherein
  • Method of modulating a mast cell includes antibody mediated cellular cytotoxicity (ADCC), apoptosis, agonizing or antagonizing HAAH to induce cells death, or recruiting a T cell as in a BiTE molecule (see supra).
  • the cell death assay can further comprise a cell proliferation assay, a cell viability assay and/or an apoptosis assay.
  • the extent of cell death can be measured by 3H thymidine incorporation, the trypan blue exclusion method, using propidium iodide or by the 5 "Cr-release assay.
  • the extent of cell death can be determined by a test of intracellular esterase activity, and a test of plasma membrane integrity, preferably using fluorescent calcein and ethidium homodimer-1. These tests are described in J. Neurosci 15, 5389 (1995), in J. Cell Sci. 106, 685 (1993). Detailed protocols are given in the Molecular Probes Catalogue product number L-3224 (Live/Dead® Kits) incorporated herein by reference. Basically, calcein AM is the cell-permeant esterase substrate, which is nonfluorescent until converted by enzymatic activity to highly fluorescent calcein. It remains within living cells exhibiting an intense green fluorescence. Ethidium homodimer-1 fluorescence is enhanced upon binding nucleic acids.
  • the extent of cell death is determined by discriminating between living and dead cells using DiOC 18 and propidium iodide. Protocols are described in details in the Molecular Probes Catalogue product number L-7010 (Live/Dead® Kits) incorporated herein by reference.
  • cell death can be determined using the Caspase activity test. Caspase is a key player in the activation of apoptosis.
  • the Molecular probe kit E-13183 (EnzCheck Caspase-3 Assay kit®, Molecular Probe) is particularly useful for testing Jurkat cells. Phosphatidyl exposure can also be used in this regard. This method has been employed in Dan S, et al, 1998, Cell Death Differ. 5:710-5.
  • cell death can be determined using the Mitochondrial membrane depolarization test using the JC-1 or JC-9 cationic dyes of Molecular Probe, which have been described as a useful indicator in HL-60 cells.
  • MTS tetrazolium Cell Titer96 Aqueous; Promega, Madison, Wis.
  • the invention encompasses fluorometric assays of cell viability and cytotoxicity using a fluorescence microscope, a fluorometer, a fluorescence microplate reader and/or a flow cytometer.
  • fluorometric assays of cell viability and cytotoxicity using a fluorescence microscope, a fluorometer, a fluorescence microplate reader and/or a flow cytometer.
  • the above methods are not meant to be an exhaustive list of all mast cell proliferation/death assays, but just an example assays that be utilized.
  • the present invention comprises methods for identifying compounds capable of preventing mast cell activation, wherein said compounds are immunospecifically binds to HAAH on the surface of the mast cell and not other hematopoeitic cells that are not mast cells or related cells or cell lines or derived cell lines thereof, comprising the steps consisting of: a) culturing mast cells in vitro in a suitable culture medium, b) adding to said culture medium at least one candidate compound to be tested and incubating said cells for a prolonged period of time, c) adding to the culture medium at least one compound known to promote mast cell activation, d) measuring the extent to which said compounds prevent mast cell activation and selecting compounds for which inhibition of mast cell activation is observed, e) identifying a compound of a subset of a compound selected in step d) that prevent significant activation of a mast, and selecting compounds for which inhibition of mast cells activation is observed.
  • Mast cell activation can be monitored by examining the amount of degranulation. Degranulation leads to the release of preformed mediators of inflammation including, histamine, JL-3, IL-5 and/or TNF. Thus, one can monitor the levels of one or more of these mediators released from a treated mast cell and compare it to a control treated mast cell.
  • the present invention provides methods for preventing, managing, treating, or ameliorating respiratory conditions comprising administering to a subject in need thereof one or more HAAH antagonists alone or in combination with one or more therapies (e.g., one or more prophylactic or therapeutic agents) other than an HAAH antagonist.
  • the present invention also provides compositions comprising one or more HAAH antagonists and one or more prophylactic or therapeutic agents other than HAAH antagonists and methods of preventing, managing, treating, or ameliorating respiratory conditions or one or more symptoms thereof utilizing said compositions.
  • Therapeutic or prophylactic agents include, but are not limited to, small molecules, synthetic drugs, peptides, polypeptides, proteins, nucleic acids (e.g., DNA and RNA nucleotides including, but not limited to, antisense nucleotide sequences, triple helices, RNAi, and nucleotide sequences encoding biologically active proteins, polypeptides or peptides) antibodies, synthetic or natural inorganic molecules, mimetic agents, and synthetic or natural organic molecules. Any therapy which is known to be useful, or which has been used or is currently being used for the prevention, management, treatment, or amelioration of respiratory conditions or one or more symptoms thereof can be used in combination with an HAAH antagonist in accordance with the invention described herein.
  • nucleic acids e.g., DNA and RNA nucleotides including, but not limited to, antisense nucleotide sequences, triple helices, RNAi, and nucleotide sequences encoding biologically active proteins, polypeptides
  • agents include, but are not limited to, immunomodulatory agents, anti-inflammatory agents (e.g., adrenocorticoids, corticosteroids (e.g., beclomethasone, budesonide, flunisolide, fluticasone, triamcinolone, methlyprednisolone, prednisolone, prednisone, hydrocortisone), glucocorticoids, steroids, non-steriodal anti-inflammatory drugs (e.g., aspirin, ibuprofen, diclofenac, and COX-2 inhibitors), leukotreine antagonists (e.g., montelukast, methyl xanthines, zafirlukast, and zileuton), beta2-agonists (e.g., albuterol, biterol, fenoterol, isoetharie, metaproterenol, pirbuterol, salbutamol, terbutalin formoterol, sal
  • Immunomodulatory agents can affect one or more or all aspects of the immune response in a subject. Aspects of the immune response include, but are not limited to, the inflammatory response, the complement cascade, leukocyte and lymphocyte differentiation, proliferation, and/or effector function, monocyte and/or basophil counts, and the cellular communication among cells of the immune system. In certain embodiments of the invention, an immunomodulatory agent modulates one aspect of the immune response.
  • an immunomodulatory agent modulates more than one aspect of the immune response, hi a preferred embodiment of the invention, the administration of an immunomodulatory agent to a subject inhibits or reduces one or more aspects of the subject's immune response capabilities. In an alternative embodiment of the invention, the immunomodulatory agent enhances one or more aspects of a subject's immune response.
  • an immunomodulatory agent is not an LL-9 antagonist. In certain embodiments, an immunomodulatory agent is not an anti- inflammatory agent. In other embodiments, an immunomodulatory agent is a chemotherapeutic agent. In yet other embodiments, an immunomodulatory agent an agent other than a chemotherapeutic agent.
  • immunomodulatory agents include, but are not limited to, proteinaceous agents such as cytokines, peptide mimetics, and antibodies (e.g., human, humanized, chimeric, monoclonal, polyclonal, Fvs, ScFvs, Fab or F(ab)2 fragments or epitope binding fragments), nucleic acid molecules (e.g., antisense nucleic acid molecules and triple helices), small molecules, organic compounds, and inorganic compounds.
  • proteinaceous agents such as cytokines, peptide mimetics, and antibodies (e.g., human, humanized, chimeric, monoclonal, polyclonal, Fvs, ScFvs, Fab or F(ab)2 fragments or epitope binding fragments)
  • nucleic acid molecules e.g., antisense nucleic acid molecules and triple helices
  • small molecules organic compounds, and inorganic compounds.
  • immunomodulatory agents include, but are not limited to, methotrexate, leflunomide, cyclophosphamide, cytoxan, Immuran, cyclosporine A, minocycline, azathioprine, antibiotics (e.g., FK506 (tacrolimus)), methylprednisolone (MP), corticosteroids, steroids, mycophenolate mofetil, rapamycin (sirolimus), mizoribine, deoxyspergualin, brequinar, malononitriloamindes (e.g., leflunamide), T cell receptor modulators, cytokine receptor modulators, and modulators mast cell modulators.
  • T cell receptor modulators include, but are not limited to, anti-T cell receptor antibodies (e.g., anti-CD4 antibodies (e.g., cM-T412 (Boeringer), IDEC-CE9.1® (IDEC and SKB), mAB 4162W94, Orthoclone and OKTcdr4a (Janssen-Cilag)), anti-CD3 antibodies (e.g., Nuvion (Product Design Labs), OKT3 (Johnson & Johnson), or Rituxan (IDEC)), anti-CD5 antibodies (e.g., an anti-CD5 ricin-linked immunoconjugate), anti-CD7 antibodies (e.g., CHH-380 (Novartis)), anti-CD8 antibodies, anti-CD40 ligand monoclonal antibodies (e.g., HOEC-131 (IDEC)), anti-CD52 antibodies (e.g., CAMPATH IH (Ilex)), anti- CD2 antibodies (e.g., MEDI-507
  • anti-CDl la antibodies e.g., Xanelim (Genentech)
  • anti-B7 antibodies e.g., TDEC-114 (IDEC)
  • CTLA4-immunoglobulin e.g., CTLA4-immunoglobulin
  • LFA-3TTP Biogen, International Publication No. WO 93/08656 and U.S. Patent No. 6,162,432.
  • cytokine receptor modulators include, but are not limited to, soluble cytokine receptors (e.g., the extracellular domain of a TNF- ⁇ receptor or a fragment thereof, the extracellular domain of an LL-l ⁇ receptor or a fragment thereof, and the extracellular domain of an LL-6 receptor or a fragment thereof), cytokines or fragments thereof (e.g., interleukin LL-2, IL-3, LL-4, IL-5, IL-6, LL-7, LL-8, IL-9, IL-10, LL-11, IL-12, IL-13, IL-15, IL-23, TNF- ⁇ , TNF- ⁇ , interferon (IFN)- ⁇ , IFN- ⁇ , IFN- ⁇ , and GM-CSF), anti-cytokine receptor antibodies (e.g., anti-IFN receptor antibodies, anti-LL-2 receptor antibodies (e.g., Zenapax (Protein Design Labs)), anti-IL-3 receptor antibodies, anti-JL-4 receptor antibodies,
  • a cytokine receptor modulator is LL-3, LL-4, LL-10, or a fragment thereof.
  • a cytokine receptor modulator is an anti-LL-l ⁇ antibody, anti-IL-6 antibody, anti-IL-12 receptor antibody, or anti-TNF- ⁇ antibody.
  • a cytokine receptor modulator is the extracellular domain of a TNF- ⁇ receptor or a fragment thereof.
  • a cytokine receptor modulator is not a TNF- ⁇ antagonist.
  • a cytokine receptor modulator is a mast cell modulator. In an alternative embodiment, a cytokine receptor modulator is not a mast cell modulator.
  • mast cell modulators include, but are not limited to stem cell factor (c-kit receptor ligand) inhibitor (e.g., mAb 7H6, mAb 8H7a, pAb 1337, FK506, CsA, dexamthasone, and fluconcinonide), c-kit receptor inhibitor (e.g., STI 571 (formerly known as CGP 57148B)), mast cell protease inhibitor (e.g., GW-45, GW-58, wortmannin, LY 294002, calphostin C, cytochalasin D, genistein, KT5926, staurosproine, and lactoferrin), relaxin ("RLX”), IgE antagonist (e.g., antibodies rhuMAb-E25 omalizumab, HMK-12 and 6HD5, and mAB Hu-901), LL-3 antagonist, LL-4 antagonists, LL-10 antagonists, and TGF- beta.
  • stem cell factor c-kit
  • An immunomodulatory agent may be selected to interfere with the interactions between the T helper subsets (TH1 or TH2) and B cells to inhibit neutralizing antibody formation.
  • Antibodies that interfere with or block the interactions necessary for the activation of B cells by TH (T helper) cells, and thus block the production of neutralizing antibodies, are useful as immunomodulatory agents in the methods of the invention.
  • B cell activation by T cells requires certain interactions to occur (Durie et al., 1994, Immunol. Today, 15:406-10), such as the binding of CD40 ligand on the T helper cell to the CD40 antigen on the B cell, and the binding of the CD28 and/or CTLA4 ligands on the T cell to the B7 antigen on the B cell.
  • CD40 ligand (CD40L)-CD40 interaction is a desirable point to block the immune response because of its broad activity in both T helper cell activation and function as well as the absence of redundancy in its signaling pathway.
  • CD40L CD40 ligand
  • the interaction of CD40L with CD40 is transiently blocked at the time of administration of one or more of the immunomodulatory agents. This can be accomplished by treating with an agent which blocks the CD40 ligand on the TH cell and interferes with the normal binding of CD40 ligand on the T helper cell with the CD40 antigen on the B cell.
  • An antibody to CD40 ligand (anti-CD40L) (available from Bristol-Myers Squibb Co; see, e.g., European patent application 555,880) or a soluble CD40 molecule can be selected and used as an immunomodulatory agent in accordance with the methods of the invention.
  • An immunomodulatory agent may be selected to inhibit the interaction between TH1 cells and cytotoxic T lymphocytes ("CTLs") to reduce the occurrence of CTL-mediated killing.
  • An immunomodulatory agent may be selected to alter (e.g., inhibit or suppress) the proliferation, differentiation, activity and/or function of the CD4 + and/or CD8 + T cells.
  • antibodies specific for T cells can be used as immunomodulatory agents to deplete, or alter the proliferation, differentiation, activity and/or function of CD4 + and/or CD8 + T cells.
  • an immunomodulatory agent that reduces or depletes T cells preferably memory T cells
  • an immunomodulatory agent that inactivates CD8 + T cells is administered to a subject with respiratory conditions in accordance with the methods of the invention.
  • anti-CD8 antibodies are used to reduce or deplete CD8 + T cells.
  • an immunomodulatory agent which reduces or inhibits one or more biological activities (e.g., the differentiation, proliferation, and/or effector functions) of TH0, TH1, and/or TH2 subsets of CD4 + T helper cells is administered to a subject with respiratory conditions in accordance with the methods of the invention.
  • one or more biological activities e.g., the differentiation, proliferation, and/or effector functions
  • LL-4 enhances antigen-specific activity of TH2 cells at the expense of the TH1 cell function (see, e.g., Yokota et al, 1986 Proc. Natl. Acad. Sci., USA, 83:5894-5898; and U.S. Pat. No. 5,017,691).
  • immunomodulatory agents that affect the biological activity (e.g., proliferation, differentiation, and/or effector functions) of T-helper cells (in particular, TH1 and/or TH2 cells) include, but are not limited to, LL-2, LL-4, LL-5, LL-6, LL-10, IL-12, LL-13, LL-15, JL-23, and interferon (FFN)- ⁇ .
  • an immunomodulatory agent administered to a subject with respiratory conditions in accordance with the methods of the invention is a cytokine that prevents antigen presentation.
  • an immunomodulatory agent used in the methods of the invention is LL-10.
  • EL- 10 also reduces or inhibits macrophage action, which involves bacterial elimination.
  • An immunomodulatory agent may be selected to reduce or inhibit the activation, degranulation, proliferation, and/or infiltration of mast cells, hi certain embodiments, the immunomodulatory agent interferes with the interactions between mast cells and mast cell activating agents, including, but not limited to stem cell factors (c-kit ligands), IgE, IL-4, environmental irritants, and infectious agents.
  • the immunomodulatory agent reduces or inhibits the response of mast cells to environmental irritants such as, but not limited to pollen, dust mites, tobacco smoke, and/or pet dander.
  • the immunomodulatory agent reduces or inhibits the response of mast cells to infectious agents, such as viruses, bacteria, and fungi.
  • stem cell factor c-kit receptor ligand
  • mAb 7H6, mAb 8H7a, and pAb 1337 see Mendiaz et al., 1996, Eur J Biochem 293:842-49
  • FK506 and CsA Ito et al., 1999 Arch DermatolRes 291:275-83
  • c-kit receptor inhibitors e.g., STI 571 (formerly known as CGP 57148B) (see Heinrich et al., 2000, Blood 96:925-32)
  • mast cell protease inhibitors e.g., GW-45 and GW-58 (see Temkin et al., 2002, J Immunol 169:2662-9)
  • wortmannin LY 294002
  • calphostin C calphostin C
  • cytochalasin D see Nosseller et al., 1997, Mol Biol Cell 8:909-22
  • genistein, KT5926, and staurosproine see ⁇ agai et al., 1995, Biochem Biophys Res Commun 208:576-81
  • lactoferrin see He et al., 2003 Biochem Pharmacol 65:1007-15
  • relaxin RLX
  • LL-9 antagonists that can be utilized in combination with the invention, but are not limited to, proteinaceous agents (e.g., proteins, polypeptides, peptides, fusion proteins, antibodies, and antibody fragments), nucleic acid molecules (e.g., DL-9 antisense nucleic acid molecules, triple helices, RNAi, or nucleic acid molecules encoding proteinaceous agents), organic molecules, inorganic molecules, small organic molecules, drugs, and small inorganic molecules that block, inhibit, reduce or neutralize a pathologic cellular or humoral phenotype associated with or resulting from LL-9 expression and/or activity (e.g., decreases the secretion of mucin, the differentiation of LL-9 expressing cells into a mucin-secreting cell, the secretion of inflammatory agents, the proliferation, migration, and increase in volume of cells (e.g., immune and smooth muscle cells), the secretion of extracellular matrix molecules or matrix metalloproteinases and/or the binding of
  • proteinaceous agents
  • an LL-9 antagonist is an antibody or fragment thereof that immunospecifically binds to an LL-9 polypeptide.
  • an LL-9 antagonist is an antibody or fragment thereof that immunospecifically binds to an LL-9R or a subunit thereof. See, e.g. co-pending application 60/462,307, incorporated in its entirety by reference.
  • proteins, polypeptides or peptides (including antibodies) that are utilized as immunomodulatory agents are derived from the same species as the recipient of the proteins, polypeptides or peptides so as to reduce the likelihood of an immune response to those proteins, polypeptides or peptides.
  • the proteins, polypeptides, or peptides that are utilized as immunomodulatory agents are human or humanized.
  • one or more immunomodulatory agents are administered in combination with an HAAH antagonist to a subject with respiratory conditions so as to transiently reduce or inhibit one or more aspects of the immune response.
  • Such a transient inhibition or reduction of one or more aspects of the immune system can last for hours, days, weeks, or months.
  • the transient inhibition or reduction in one or more aspects of the immune response lasts for a few hours (e.g., 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, 14 hours, 16 hours, 18 hours, 24 hours, 36 hours, or 48 hours), a few days (e.g., 3 days, 4 days, 5 days, 6 days, 7 days, or 14 days), or a few weeks (e.g., 3 weeks, 4 weeks, 5 weeks or 6 weeks).
  • the transient reduction or inhibition of one or more aspects of the immune response enhances the prophylactic and/or therapeutic effect(s) of an HAAH antagonist.
  • Nucleic acid molecules encoding proteins, polypeptides, or peptides with immunomodulatory activity or proteins, polypeptides, or peptides with immunomodulatory activity can be administered to a subject with respiratory conditions in accordance with the methods of the invention.
  • nucleic acid molecules encoding derivatives, analogs, or fragments of proteins, polypeptides, or peptides with immunomodulatory activity, or derivatives, analogs, or fragments of proteins, polypeptides, or peptides with immunomodulatory activity can be administered to a subject with a respiratory infection in accordance with the methods of the invention.
  • such derivatives, analogs, and fragments retain the immunomodulatory activity of the full-length, wild type protein, polypeptide, or peptide.
  • agents that are commercially available and known to function as immunomodulatory agents are used in the methods of the invention.
  • the immunomodulatory activity of an agent can be determined in vitro and/or in vivo by any technique well known to one skilled in the art, including, e.g., by CTL assays, proliferation assays, and immunoassays (e.g. ELISAs) for the expression of particular proteins such as co-stimulatory molecules and cytokines.
  • anti-inflammatory agent Any anti-inflammatory agent, including agents useful in therapies for inflammatory disorders, well known to one of skill in the art can be used in the compositions and methods of the invention.
  • anti-inflammatory agents include non-steroidal anti-inflammatory drugs (NSALDs), steroidal anti-inflammatory drugs, anticholinergics (e.g., atropine sulfate, atropine methylnitrate, and ipratropium bromide (ATROVENTTM)), beta2- agonists (e.g., abuterol (VENTOLINTM and PROVENTILTM), bitolterol (TORNALATETM), levalbuterol (XOPONEXTM), metaproterenol (ALUPENTTM), pirbuterol (MAXAIRTM), terbutlaine (BRETHAIRETM and BRETHINETM), albuterol (PROVENTILTM, REPETABSTM, and VOLMAXTM), formoterol (FORADIL AEROLIZERTM), and salmeterol (
  • NSALDs include, but are not limited to, aspirin, ibuprofen, celecoxib (CELEBREXTM), diclofenac (VOLTARENTM), etodolac (LODLNETM), fenoprofen (NALFONTM), indomethacin (INDOCINTM), ketoralac (TORADOLTM), oxaprozin (DAYPROTM), nabumentone (RELAFENTM), sulindac (CLLNORILTM), tolmentin (TOLECTINTM), rofecoxib (NIOXXTM), naproxen (ALEVETM, ⁇ APROSY ⁇ TM), ketoprofen (ACTRO ⁇ TM) and nabumetone (RELAFE ⁇ TM).
  • Such ⁇ SAIDs function by inhibiting a cyclooxgenase enzyme (e.g., COX-1 and/or COX-2).
  • steroidal anti-inflammatory drugs include, but are not limited to, glucocorticoids, dexamethasone (DECADRONTM), corticosteroids (e.g., methylprednisolone (MEDROLTM)), cortisone, hydrocortisone, prednisone (PREDNISONETM and DELTASONETM), prednisolone (PRELONETM and PEDIAPREDTM), triamcinolone, azulfidine, and inhibitors of eicosanoids (e.g., prostaglandins, thromboxanes, and leukotrienes (see Table 2, infra, for non-limiting examples of leukotriene and typical dosgages of such agents)).
  • eicosanoids e.g., prostaglandins, thromboxanes
  • anti-inflammatory molecular antagonists include, but are not limited to, antagonists of HMGB1 and antagonists of one or more chitinase-like molecules.
  • the anti-inflammatory agent is an agent useful in the prevention, management, treatment, and/or amelioration of asthma or one or more symptoms thereof.
  • Non-limiting examples of such agents include adrenergic stimulants (e.g., catecholamines (e.g., epinephrine, isoproterenol, and isoetharine), resorcinols (e.g., metaproterenol, terbutaline, and fenoterol), and saligenins (e.g., salbutamol)), adrenocorticoids, blucocorticoids, corticosteroids (e.g., beclomethadonse, budesonide, flunisolide, fluticasone, triamcinolone, methylprednisolone, prednisolone, and prednisone), other steroids, beta2-agonists (e.g., albtuerol, bitolterol, fenoterol, isoetharine, metaproterenol, pirbuterol, salbutamol, terbutaline, formoterol
  • C3 a receptor antagonists including antibodies
  • immunosuppressant agents e.g., methotrexate and gold salts
  • mast cell stabilizers e.g., cromolyn sodium (INTALTM) and nedocromil sodium (TILADETM)
  • mucolytic agents e.g., acetylcysteine
  • the anti-inflammatory agent is a leukotriene modifier (e.g., montelukast (SINGULAIRTM), zafirlukast (ACCOLATETM), pranlukast (ONONTM), or zileuton (ZYFLOTM) (see Table 1)).
  • a leukotriene modifier e.g., montelukast (SINGULAIRTM), zafirlukast (ACCOLATETM), pranlukast (ONONTM), or zileuton (ZYFLOTM) (see Table 1)).
  • the anti-inflammatory agent is an agent useful in preventing, treating, managing, or ameliorating allergies or one or more symptoms thereof.
  • Non-limiting examples of such agents include antimediator drugs (e.g., antihistamine, see Table 2 for non- limiting examples of antihistamine and typical dosages of such agents), corticosteroids, decongestants, sympathomimetic drugs (e.g., ⁇ -adrenergic and ⁇ -adrenergic drugs), TNX901 (Leung et al, 2003, NEnglJMed 348:986-93), IgE antagonists (e.g., antibodies rhuMAb- E25 omalizumab (see Finn et al, 2003, J Allergy Clin Immuno 111:278-84; Corren et al, 2003, J Allergy Clin Immuno 11:87-90; Busse and Neaville, 2001, Curr Opin Allergy Clin Immuno 1:105-8; and Tang and Powell, 2001, EurJPediatr 160: 696-704), HMK-12 and 6HD5 (see Miyajim
  • Anti- Viral Agents Any anti- viral agent well-known to one of skill in the art for the treatment, prevention, management, or amelioration of respiratory conditions or a symptom thereof (e.g., viral respiratory infection) can be used in the compositions and methods of the invention.
  • anti-viral agents include proteins, polypeptides, peptides, fusion proteins antibodies, nucleic acid molecules, organic molecules, inorganic molecules, and small molecules that inhibit or reduce the attachment of a virus to its receptor, the internalization of a virus into a cell, the replication of a virus, or release of virus from a cell
  • antiviral agents include, but are not limited to, nucleoside analogs (e.g., zidovudine, acyclovir, gangcyclovir, vidarabine, idoxuridine, trifluridine, and ribavirin), foscarnet, amantadine, rimantadine, saquinavir, indinavir, ritonavir, alpha-interferons and other interferons, and AZT.
  • nucleoside analogs e.g., zidovudine, acyclovir, gangcyclovir, vidarabine, idoxuridine, trifluridine, and
  • the anti- viral agent is an antibody agent that is immunospecific for a viral antigen.
  • viral antigen includes, but is not limited to, any viral peptide, polypeptide and protein (e.g., RSV F glycoprotein, RSV G glycoprotein, influenza virus neuraminidase, influenza virus hemagglutinin, and herpes simplex virus glycoprotein (e.g., gB, gC, gD, and gE)) that is capable of eliciting an immune response.
  • Antibodies useful in this invention for prevention, management, treatment, and/or amelioration of a viral infectious disease include, but are not limited to, antibodies against antigens of pathogenic viruses, including as examples and not by limitation: adenovirdiae (e.g., mastadenovirus and aviadenovirus), herpesviridae (e.g., herpes simplex virus 1, herpes simplex virus 2, herpes simplex virus 5, and herpes simplex virus 6), leviviridae (e.g., levivirus, enterobacteria phase MS2, allolevirus), poxviridae (e.g., chordopoxvirinae, parapoxvims, avipoxvirus, capripoxvirus, leporiipoxvirus, suipoxvirus, moUuscipoxvirus, and entomopoxvirinae), papovaviridae (e.g., polyomavirus and papilloma
  • human immunodeficiency virus 1 and human immunodeficiency virus 2), spumavirus flaviviridae (e.g., hepatitis C virus), hepadnaviridae (e.g., hepatitis B virus), togaviridae (e.g., alphavirus (e.g., Sindbis virus) and rubivirus (e.g., rubella virus)), rhabdo viridae (e.g., vesiculovirus, lyssavirus, ephemera virus, cytorhabdovirus, and necleorhabdovirus), arenaviridae (e.g., arenavirus, lymphocytic choriomeningitis virus, Ippy virus, and lassa virus), and coronaviridae (e.g., coronavirus and torovirus).
  • flaviviridae e.g., hepatitis C virus
  • hepadnaviridae e.
  • antibodies available useful for the prevention, management, treatment, and/or amelioration of a viral infectious disease include, but are not limited to, PRO542 (Progenies) which is a CD4 fusion antibody useful for the treatment of HIN infection and SY ⁇ AGIS® (Medlmmune, Inc.; International Publication No. WO 02/43660) which is a humanized antibody useful for treatment of RSV.
  • the anti- viral agent used in the compositions and methods of the invention inhibits or reduces a pulmonary or respiratory virus infection, inhibits or reduces the replication of a virus that causes a pulmonary or respiratory infection, or inhibits or reduces the spread of a virus that causes a pulmonary or respiratory infection to other cells or subjects.
  • the anti-viral agent used in the compositions and methods of the invention inhibits or reduces infection by RSV, hMPV, or PLV, inhibits or reduces the replication of RSV, hMPV, or PLV, or inhibits or reduces the spread of RSV, hMPV, or PIV to other cells or subjects.
  • nucleoside analogs such as zidovudine, acyclovir, gangcyclovir, vidarabine, idoxuridine, trifluridine, and ribavirin, as well as foscarnet, amantadine, rimantadine, saquinavir, indinavir, ritonavir, and the alpha-interferons.
  • nucleoside analogs such as zidovudine, acyclovir, gangcyclovir, vidarabine, idoxuridine, trifluridine, and ribavirin
  • foscarnet such as zidovudine, acyclovir, gangcyclovir, vidarabine, idoxuridine, trifluridine, and ribavirin
  • foscarnet amantadine, rimantadine, saquinavir, indinavir, ritonavir
  • alpha-interferons See U.S. Provisional Patent Application
  • the viral infection is RSV and the anti-viral antigen is an antibody that immunospecifically binds to an antigen of RSV.
  • the anti-RSV-antigen antibody immunospecifically binds to an RSV antigen of the Group A of RSV.
  • the anti-RSV-antigen antibody immunospecifically binds to an RSV antigen of the Group B of RSV.
  • the anti-RSV antigen antibody immunospecifically binds to an antigen of RSV of one Group and cross reacts with the analogous antigen of the other Group.
  • the anti-RSV-antigen antibody immunospecifically binds to a RSV nucleoprotein, RSV phosphoprotein, RSV matrix protein, RSV small hydrophobic protein, RSV RNA-dependent RNA polymerase, RSV F protein, and/or RSV G protein
  • the anti-RSV- antigen antibody binds to allelic variants of a RSV nucleoprotein, a RSV nucleocapsid protein, a RSV phosphoprotein, a RSV matrix protein, a RSV attachment glycoprotein, a RSV fusion glycoprotein, a RSV nucleocapsid protein, a RSV matrix protein, a RSV small hydrophobic protein, a RSV RNA-dependent RNA polymerase, a RSV F protein, a RSV L protein, a RSV P protein, and/or a RSV G protein.
  • SYNAGIS® (Palivizumab) is a humanized monoclonal antibody presently used for the prevention of RSV infection in pediatric patients.
  • an antibody to be used in accordance with the methods of the present invention is SYNAGIS® or an antibody-binding fragment thereof (e.g., a fragment containing one or more complementarity determining regions (CDRs) and preferably, the variable domain of SYNAGIS®).
  • CDRs complementarity determining regions
  • the amino acid sequence of SYNAGIS® is disclosed, e.g., in Johnson et al, 1997, J. Infectious Disease 176:1215-24, and U.S. Patent No.
  • WO 02/43660 entitled “Methods of Administering/Dosing Anti-RSV Antibodies for Prophylaxis and Treatment", by Young et al, which are incorporated herein by reference in their entireties.
  • One or more antibodies or antigen-binding fragments thereof that immunospecifically bind to a RSV antigen comprise a Fc domain with a higher affinity for the FcRn receptor than the Fc domain of SYNAGIS® (Palivizumab) can also be used in accordance with the invention.
  • Such antibodies are described in U.S. Patent Application No.: 10/020,354, filed December 12, 2001, which is incorporated herein by reference in its entireties.
  • the anti-RSV-antigen antibody A4B4; P12f2 P12f4; Plld4; Ale9; A12a6; A13c4; A17d4; A4B4; 1X-493L1; FR H3-3F4; M3H9; Y10H6; DG; AFFF; AFFF(l); 6H8; L1-7E5; L2-15B10; A13al 1; Alh5; A4B4(1);A4B4-F52S; or A4B4L1FR-S28R can be used in accordance with the invention.
  • the anti-RSV-antigen antibodies are the anti-RSV-antigen antibodies of or are prepared by the methods of U.S.
  • Anti- viral therapies and their dosages, routes of administration and recommended usage are known in the art and have been described in such literature as the Physician 's Desk Reference (57th ed., 2003). Additional information on respiratory viral infections is available in Cecil Textbook of Medicine (18th ed., 1988).
  • Anti-Bacterial Agents Anti-bacterial agents and therapies well-known to one of skill in the art for the prevention, treatment, management, or amelioration of respiratory conditions or one or more symptoms thereof (e.g., a bacterial respiratory infection) can be used in the compositions and methods of the invention.
  • Non-limiting examples of anti-bacterial agents include proteins, polypeptides, peptides, fusion proteins, antibodies, nucleic acid molecules, organic molecules, inorganic molecules, and small molecules that inhibit or reduce a bacterial infection, inhibit or reduce the replication of bacteria, or inhibit or reduce the spread of bacteria to other cells or subjects.
  • anti-bacterial agents include, but are not limited to, antibiotics such as penicillin, cephalosporin, imipenem, axtreonam, vancomycin, cycloserine, bacitracin, chloramphenicol, erythromycin, clindamycin, tetracycline, streptomycin, tobramycin, gentamicin, amikacin, kanamycin, neomycin, spectinomycin, trimethoprim, norfloxacin, rifampin, polymyxin, amphotericin B, nystatin, ketocanazole, isoniazid, metronidazole, and pentamidine.
  • antibiotics such as penicillin, cephalosporin, imipenem, axtreonam, vancomycin, cycloserine, bacitracin, chloramphenicol, erythromycin, clindamycin, tetracycline, streptomycin, tobramycin, gentamicin,
  • the anti-bacterial agent is an agent that inhibits or reduces a pulmonary or respiratory bacterial infection, inhibits or reduces the replication of a bacteria that causes a pulmonary or respiratory infection, or inhibits or reduces the spread of a bacteria that causes a pulmonary or respiratory infection to other cells or subjects.
  • the pulmonary or respiratory bacterial infection is a mycoplasma infection (e.g., pharyngitis, tracheobronchitis, and pneumonia)
  • the anti-bacterial agent is preferably a tetracycline, erythromycin, or spectinomycin.
  • the anti-bacterial agent is preferably rifampcin, isonaizid, pyranzinamide, ethambutol, and streptomycin.
  • the anti-bacterial agent is preferably penicillin, first, second, or third generation cephalosporin (e.g., cefaclor, cefadroxil, cephalexin, or cephazolin), erythomycin, clindamycin, an aminoglycoside (e.g., gentamicin, tobramycin, or amikacine), or a monolactam (e.g., aztreonam).
  • the anti-bacterial agent is preferably penicillin, an aminoglycoside, or a second or third generation cephalosporin.
  • Anti-bacterial therapies and their dosages, routes of administration and recommended usage are known in the art and have been described in such literature as the Physician 's Desk Reference (57th ed., 2003), Cecil Textbook of Medicine (18th ed., 1988), and Tlie Merk Manual of Diagnosis and Tlierapy (17th ed. 1999).
  • Anti-fungal agents and therapies well known to one of skill in the art for prevention, management, treatment, and/or amelioration of respiratory conditions or one or more symptoms thereof can be used in the compositions and methods of the invention.
  • Non-limiting examples of anti-fungal agents include proteins, polypeptides, peptides, fusion proteins, antibodies, nucleic acid molecules, organic molecules, inorganic molecules, and small molecules that inhibit or reduce fungal infection, inhibit or reduce the replication of fungi, or inhibit or reduce the spread of fungi to other subjects.
  • anti-fungal agents include, but are not limited to, azole drugs (e.g., miconazole, ketoconazole (NIZORAL®), caspofungin acetate (CANCIDAS®), imidazole, triazoles (e.g., fluconazole (DIFLUCAN®)), and itraconazole (SPORANOX®)), polyene (e.g., nystatin, amphotericin B (FUNGIZONE®), amphotericin B lipid complex (“ABLC”)(ABELCET®), amphotericin B colloidal dispersion (“ABCD”)(AMPHOTEC®), liposomal amphotericin B (AMBISONE®)), potassium iodide (KI), pyrimidine (e.g., flucytosine (ANCOBON®)), and voriconazole (VFEND®).
  • azole drugs e.g., miconazole, ketoconazole (NIZORAL®), caspofungin
  • the anti-fungal agent is an agent that inhibits or reduces a respiratory fungal infection, inhibits or reduces the replication of a fungus that causes a pulmonary or respiratory infection, or inhibits or reduces the spread of a fungus that causes a pulmonary or respiratory infection to other subjects.
  • the anti-fungal agent is preferably itraconazole, amphotericin B, fluconazole, or ketoconazole.
  • the anti-fungal agent is preferably amphotericin B, liposomal amphotericin B, itraconazole, or fluconazole.
  • the anti-fungal agent is preferably amphotericin B, itraconazole, fluconazole, or ketoconazole.
  • the anti-fungal agent is preferably fluconazole or amphotericin B.
  • the anti-fungal agent is preferably amphotericin B, fluconazole, or combination of the two agents.
  • the anti-fungal agent is preferably itraconazole, fluconazole, or flucytosine.
  • the anti-fungal agent is preferably amphotericin B or liposomal amphotericin B.
  • the anti-fungal agent is preferably itraconazole ore miconazole.
  • Anti-fungal therapies and their dosages, routes of administration, and recommended usage are known in the art and have been described in such literature as Dodds et al, 2000, Pharmacotherapy 20:1335-55, the Physician 's Desk Reference (57th ed., 2003) and the Merk Manual of Diagnosis and Therapy (17th ed., 1999).
  • compositions for the treatment, prophylaxis, and amelioration of one or more symptoms associated with an autoimmune or inflammatory disorder comprises one or more HAAH antagonists.
  • a composition comprises one or more nucleic acid molecules encoding one or more HAAH antagonists, h another embodiment, a composition comprises one or more HAAH antagonists and one or more prophylactic or therapeutic agents other than HAAH antagonists, said prophylactic or therapeutic agents known to be useful for, or having been or currently being used in the prevention, treatment or amelioration of one or more symptoms associated an autoimmune or inflammatory disorder.
  • a composition comprises one or more nucleic acid molecules encoding one or more HAAH antagonists and one or more prophylactic or therapeutic agents other than HAAH antagonists, said prophylactic or therapeutic agents known to be useful for, or having been or currently being used in the prevention, treatment or amelioration of one or more symptoms associated an autoimmune or inflammatory disorder.
  • a composition comprises one or more HAAH antagonists and one or more nucleic acid molecules encoding one or more prophylactic or therapeutic agents other than HAAH antagonists, said prophylactic or therapeutic agents known to be useful for, or having been or currently being used in the prevention, treatment or amelioration of one or more symptoms associated an autoimmune or inflammatory disorder.
  • a composition comprises one or more nucleic acid molecules encoding one or more HAAH antagonists and one or more nucleic acid molecules encoding one or more prophylactic or therapeutic agents other than HAAH antagonists, said prophylactic or therapeutic agents known to be useful for, or having been or currently being used in the prevention, treatment or amelioration of one or more symptoms associated an autoimmune or inflammatory disorder.
  • a composition comprises a one or more HAAH antagonists and one or more immunomodulatory agents.
  • the immunomodulatory agents can be an ⁇ antagonists (e.g. VITAXLNTM), T cell antagonists (e.g.
  • a composition comprises small molecules the modulate the immune system, like methotrexate.
  • a composition comprises HAAH antagonist and one or more CD2 antagonists.
  • a composition comprises a HAAH agonist or more integrin ⁇ 3 antagonists and one or more CD2 binding molecules.
  • a composition comprises HAAH agonist or an antigen-binding fragment thereof and one or more CD2 binding molecules.
  • a composition comprises a HAAH agonist, VITAXLNTM or an antigen-binding fragment thereof and MEDI-507 or an antigen- binding fragment thereof.
  • HAAH Tissue Expression of HAAH It has been reported that HAAH is abundantly expressed in several malignant neoplasms, while low levels ofHAAH expression are seen in most normal tissues and cell types. Examination of the levels and distribution ofHAAH protein in tissues yielded the unexpected finding that HAAH is highly expressed in the tissue-resident mast cells of numerous normal human tissues. Normal human small intestine, skeletal muscle and heart tissue was stained with a monoclonal antibody specific for HAAH. In all tissue examined the tissue-resident mast cells stain intensely (see Figure 1). Some minimal nonspecific background staining is seen in the submucosal plexus of the small intestine and the cardiac myocytes.
  • a microtome was used to cut tissue sections at 4-5 microns. The sections were floated on a water bath and picked up onto slides. Slides containing paraffin sections were deparaffinized through xylene and alcohol, rehydrated, and then subjected to the steam method of target retrieval (DAKO reagent #S1700). Antibody titration experiments were conducted with anti HAAH mAb 15C7 to establish dilutions that would result in minimal background and maximal detection of signal. Serial dilutions were performed at 1:50, 1:100, 1:250, 1:400, 1:500 and 1:600.
  • the serial dilutions study demonstrated the highest signal-to-noise ratios at dilutions of 1 :400 and 1 :600 on paraffin-embedded, formalin fixed tissues after deparafrmization and rehydration. These dilutions were selected for the study. 15C7 mAb was used as the primary antibody, and the principal detection system consisted of a Vector anti-mouse secondary (BA-2000) and a Vector ABC-AP kit (AK-5000) with a Vector red substrate kit (SK-5100), which was used to produce a fuschia-colored deposit. Tissues were also stained with positive control antibodies (CD31 and vimentin) to ensure that the tissue antigens were preserved and accessible for immunohistochemical analysis.
  • BA-2000 Vector anti-mouse secondary
  • AK-5000 Vector ABC-AP kit
  • SK-5100 Vector red substrate kit
  • Tissues were also stained with positive control antibodies (CD31 and vimentin) to ensure that the tissue antigens were preserved and accessible for immunohisto
  • HAAH Expression in Mast Cell Tissue Culture Cell Lines To expand the observation that tissue-resident mast cells stain intensely with an anti- HAAH specific antibody four cell lines (H460, a human lung cancer cell line known to express HAAH on the cell surface; UT-7, a human mast cell precursor cell line, KU812, another human mast cell precursor cell line, and RBL, a rat mast cell line) were probed with a monoclonal antibody directed against the catalytic domain ofHAAH (15C7).
  • HAAH Human aspartyl/aspariginal ⁇ hydroxylase
  • KU812 cells by FACS analysis the levels appear similar for all four cell lines (see below).
  • the commonly used mast cell-precursor cell line (UT-7) was analyzed by real-time PCR to quantitate HAAH gene expression and by FACS to examine HAAH cell- surface protein expression.
  • Real-time PCR analysis demonstrated that the level ofHAAH message was at least 100-fold more abundant than the level of message encoding LL-4, a cytokine known to be expressed by mast cells (see Table 4).
  • FACS analysis demonstrated that a large number of UT-7 cells stain positive for HAAH when labeled with the HAAH specific monoclonal antibody 15C7 (see Figure 3).
  • the 15C7-stained cells clearly show at least two populations, probably due to the fact that UT-7 cells are not a clonal cell line, but are composed of sub-populations of cells at different stages of differentiation.
  • Several other cells lines including KU812, another human mast cell precursor cell line (Figure 3), Mel 9 and RBL-23H cells (mouse and rat mast cell-precursor cell lines respectively)(data not shown) were also examined by FACS analysis and are highly positive for cell-surface HAAH expression.
  • the 15C7-stained KU812 cells are a more homogenous population reflective of the homogeneity of the cell line (Figure 4).
  • FACS Analysis Cells were harvested with trypsin-free dissociation buffer, spun down and resuspended in FACS buffer (PBS, 2%FCS, 0.2% NaN3). Viable cell concentration was determined by counting typan blue-excluding cells in a hemacytometer. Cells were transferred to eppendorf tubes at a concentration of 200,000 cells in 50 ⁇ l FACS buffer. 3 tubes were set up per cell line, labelled -1,-2; -1, +15C7. 1 ⁇ l of 1 mg/ml human IgG was added to each tube, to block non-specifc binding to Fc-receptor, and incubated at room temperature for 10 minutes.
  • Human Aspartyl Asparaginyl ?-Hydroxylase or HAAH, is a highly conserved enzyme which hydroxylates aspartic acid or asparagine residues in EGF-like domains of several proteins in the presence of ferrous iron. These EGF-like domains contain conserved motifs that form repetitive sequences in proteins such as clotting factors, extracellular matrix proteins, LDL receptor, NOTCH homologues or NOTCH ligand homologues.
  • EGF-like sequences play an important role in protein-protein interactions, as shown by mutations in EGF-like domains of fibrillin that cause Marfan's syndrome or factor LX, which produces hemophilia B (Downing et al, 1996, Cell, 85: 597-605; Nishimura et al, 1993, J. Biol. Chem., 268: 24041-24046).
  • EGF-like domain containing protein are also recognized as cancer transformation-associated proteins.
  • Over-expression of HAAH is strongly associated with the progression and invasiveness of multiple cancers so it was unexpected that tissue-associated mast cells in normal tissues would show intense staining with the HAAH specific monoclonal antibody 15C7.
  • mast cell directed therapies Given the prominent role of mast cells in numerous immune responses the presence of HAAH on the surface of mast cells provides an excellent target for mast cell directed therapies. Little to no staining ofHAAH was seen on B- and T-cells and more limited staining was seen on other immune cells. Thus, targeting cell surface HAAH allows for highly directed therapies that more specifically target the mature mast cell. Such mast cell directed therapies would be useful for the treatment of a large number of disorders including inflammatory or autoimmune disorders or cardiovascular disease.
  • the disclosures of each and every patent, patent application, and publication cited herein are hereby incorporated by reference in their entirety.

Abstract

The present invention relates to agents that modulate HAAH activity by immunospecifically binding to HAAH on the surface of the mast cell. More specifically, this invention provides HAAH antagonists that modulate mast cells. Also provided are methods for the prevention, management, treatment, or amelioration of respiratory conditions, an inflammatory or autoimmune disorder or cardiovascular disease, said methods comprising administering to a subject in need thereof an effective amount of one or more agents that modulate the activity of HAAH.

Description

NOVEL METHODS TO MODULATE MAST CELLS
FIELD OF THE INVENTION The present invention is based, in part, on the inventors' discovery that human aspartyl (asparaginyl)-β hydroxylase (HAAH) is strongly expressed on the surface of mast cells. The present invention also provides for prophylactic and therapeutic protocols designed to prevent, manage, treat, or ameliorate respiratory conditions (e.g. asthma) or one or more symptoms thereof. The present invention further provides for prophylactic and therapeutic protocols designed to prevent, treat, manage or ameliorate an inflammatory or an autoimmune disorder or cardiovascular/coronary heart disease. The present invention further provides methods for the prevention, management, treatment, or amelioration of respiratory conditions (e.g. asthma) or one or more symptoms thereof and/or to the treatment, management or amelioration of an inflammatory or an autoimmune disorder or cardiovascular disease, said methods comprising administering to a subject in need thereof an effective amount of one or more agents that modulate the activity ofHAAH. In one specific embodiment the agent that modulates HAAH activity immunospecifically binds to HAAH on the surface of the mast cell. In another specific embodiment, the modulation ofHAAH activity modulates mast cells. The invention also provides combination therapies for the prevention, management, treatment or amelioration of respiratory conditions (e.g. asthma) and/or to treat, manage or ameliorate an inflammatory disorder or an autoimmune disorder or cardiovascular disease. The invention further provides pharmaceutical compositions and articles of manufacture for use in the prevention, management, treatment, or amelioration of respiratory conditions or one or more symptoms thereof and/or the treatment, management or amelioration an inflammatory disorder or an autoimmune disorder or cardiovascular disease. BACKGROUND OF THE INVENTION Mast cells are derived from CD34+ hematopoietic progenitor cells and except for the small number of mast cells that reside in the bone marrow, maturation typically occurs in the peripheral tissues. Williams et al., 2000 J. Allergy Clin. Immunol. 105:847-859. Mast cells are ideally positioned anatomically to participate in allergic reaction at cutanious or mucosal surfaces. Unlike mature basophils, mature mast cells do not normally circulate in the blood but are widely distributed throughout normal connective tissues, where they often lie adjacent to blood and lymphatic vessels, near or within nerves, and beneath epithelial surfaces that are exposed to the external environment, such as those of the respiratory and gastrointestinal systems and skin. Id. Positioned at strategic points around capillaries and small blood vessels, mast cells are important in regulating the extent of constriction or dilation of the vessels including those which make up the blood-brain barrier, the protective lining of the brain which excludes toxic materials (Theoharides, 1990, Life Sciences 46:607-17). Mast cells produce an impressively broad array of mediators and cell-cell signaling molecules. Many of these mediators, including histamine, numerous specific proteases and tumor necrosis factor-α (TNF-α), are released by triggered exocytosis from rich intracellular stores of secretory granules. Benoist et al., 2002 Nature 420:875-878. On activation, mast cells rapidly synthesize bioactive metabolites of arachidonic acid, prostaglandins and leukotrienes. A specific program of gene expression is also activated leading to de novo synthesis of several cytokines (L -3, L -4, IL-5, L -6, L -9, L -10, IL-13, L -14), chemokines (macrophage inflammation protein lα, monocyte chemoattractant protein 1 (MCP-1) and lymphotactin) and more TNF-α. This second- wave response comes after the immediate hypersensitivity reactions, which it amplifies. Id. Compounds released by mast cell stimulation, collectively called mediators, include: histamine, kinins, prostaglandin D 2, tryptase and vasoactive intestinal peptide (VIP), which are vasodilatory, as well as serotonin, prostaglandin F2-alpha and leukotrienes, which are vasoconstrictive. In addition, cytokines, histamine, kinins and prostaglandins can cause pain directly, while enzymes which destroy proteins and phospholipids can cause tissue damage directly. Finally, cytokines such as LL-6 can cause inflammation and regulate other biological responses (Galli (1993) above). Histamine, kinins, tryptase and NIP are preformed and are stored in granules; prostaglandins and cytokines are synthesized after activation of the cell and the mechanism of their secretion is not well understood. Several triggers can elicit these mast cell responses. One of the strongest triggers of mast cell responses are allergens complexed to immunoglobulin-ε (IgE molecules. Because of the unusually high affinity to the Fc receptor for IgE, mast cells are constantly coated with antigen-specific IgE. The crosslinking of these surface bound IgE molecules by antigen leads to activation and degranulation. Anaphylatoxins generated by activation of the pathway are also potent activators of some mast cells. Bacterial microbes can trigger mast cells through Toll-like receptors, endowing them with the broad pattern recognition capability of the Tolllike receptor system. Some cytokines and chemokines can also activate mast cells. Although mast cells are best known for producing the "allergic" reactions, they have also been implicated in other autoimmune and inflammatory conditions. A number of studies have shown that chronic inflammation is often associated with mast cell hyperplasia and signs of mast cell activation. See Nalent et al., 2001 Immunological Review 179:74-81. Mast cell activation is found particularly in antigen-induced inflammation, as in autoimmune disorders or atopic diseases, hi addition, mast cells are known for their involvement in neuroinflammatory conditions that are precipitated or exacerbated by stress (Theoharides, et al., 1996 int. J. Tissue React. 18:1). Furthermore increased numbers of activated cardiac mast cells are found in ventricles, the sinusoidal node and the fibrous plaque associated with atherosclerosis and other cardiac disease states (reviewed in Marone et al, 1995, Immunopharm. 31:1-18).
Respiratory Conditions
Allergies Allergies are disorders of the immune system in which the body reacts to innocuous substances by inducing the generation of large amounts of IgE. the presence of an allergen, IgE activates mast cells and promotes mast cell proliferation, infiltration, and/or degranulation that results in the release of histamines, leukotrienes, and cytokines which cause rhinitis, hives, redness, itchiness, watery eyes, skin rashes, bronchoconstriction (wheezing), coughing, and difficulty breathing. Common allergens include, but are not limited to, pollens, molds, dust (e.g., dust mites and dust mite waste), animal protein (e.g., dander, urine, oil from the skin), industrial chemicals, foods, medicines, feathers, and insects (e.g., insect stings, cockroaches, and insect waste). Pollinosis, commonly known as hay fever, is generally induced by wind-borne pollens, including, but not limited to tree pollens (e.g., oak, elm, maple, alder, birch, juniper, and olive), grass pollens (e.g., Bermuda, timothy, sweet vernal, orchard, and Johnson), weed pollens (e.g., Russian thistle, English plantain, and ragweed), and airborne fungal spores. Symptoms of pollinosis include itchy nose, roof of the mouth, pharynx, and eyes, sneezing, runny nose, watery eyes, headaches, anorexia, depression, coughing, insomnia, and wheezing. Common therapies include administration of antihistamines, sympathomimetics, glucocorticoids, and systemic corticosteroids and allergen immunotherapy. Unfortunately, these therapies have many limitations such as unwanted side effects such as hypertension and drowsiness. Anaphylaxis is an acute allergic reaction that results when the allergen reaches the circulation. Common allergens are parenteral enzymes, blood products, β-lactam antibiotics, allergen immunotherapy, and insect stings. Anaphylaxis is characterized by smooth muscle contraction that causes wheezing, vasodilation, pulmonary edema, and obstructive angiodema. If the reaction is prolonged, the subject may develop arrhythmias or cardiogenic shock. In severe cases, the patient may suffer from primary cardiovascular collapse without respiratory symptoms. Long-term immunotherapy is effective for preventing anaphylaxis from insect stings, but is rarely available for patients with drug or serum anaphylaxis. Immediate administration of epinephrine is the most common treatment for anaphylaxis, but may cause side effects including headache, tremulousness, nausea, and arrhythmias. Thus, new therapies for the prevention, treatment, management, and amelioration of allergic reactions are needed.
Asthma About 12 million people in the U.S. have asthma and it is the leading cause of hospitalization for children. Tl%e Merck Manual of Diagnosis and Therapy (17th ed., 1999). Asthma is an inflammatory disease of the lung that is characterized by airway hyperresponsiveness ("AHR"), bronchoconstriction (i.e., wheezing), eosinophilic inflammation, mucus hypersecretion, subepithelial fibrosis, and elevated IgE levels. Asthmatic attacks can be triggered by environmental triggers (e.g. acarids, insects, animals (e.g., cats, dogs, rabbits, mice, rats, hamsters, guinea pigs, mice, rats, and birds), fungi, air pollutants (e.g., tobacco smoke), irritant gases, fumes, vapors, aerosols, or chemicals, or pollen), exercise, or cold air. The cause(s) of asthma is unknown. However, it has been speculated that family history of asthma (London et al., 2001, Epidemiology 12:577-83), early exposure to allergens, such as dust mites, tobacco smoke, and cockroaches (Melen et al., 2001, Allergy 56:646-52), and respiratory infections (Wenzel et al., 2002, Am JMed,
112:672-33 and Lin et al., 2001, JMicrobiol Immuno Infect, 34:259-64) may increase the risk of developing asthma. Current therapies are mainly aimed at managing asthma and include the administration of β-adrenergic drugs (e.g. epinephrine and isoproterenol), theophylline, anticholinergic drugs (e.g., atropine and ipratorpium bromide), corticosteroids, and leukotriene modifiers. These therapies are associated with side effects such as drug interactions, dry mouth, blurred vision, growth suppression in children, and osteoporosis in menopausal women. Cromolyn and nedocromil are administered prophylatically to inhibit mediator release from inflammatory cells, reduce airway hyperresponsiveness, and block responses to allergens. However, there are no current therapies available that prevent the development of asthma in subjects at increased risk of developing asthma. Thus, new therapies with fewer side effects and better prophylactic and/or therapeutic efficacy are needed for asthma.
Respiratory Infections Respiratory infections are common infections of the upper respiratory tract (e.g., nose, ears, sinuses, and throat) and lower respiratory tract (e.g., trachea, bronchial tubes, and lungs). Symptoms of upper respiratory infection include runny or stuffy nose, irritability, restlessness, poor appetite, decreased activity level, coughing, and fever. Viral upper respiratory infections cause and/or are associated with sore throats, colds, croup, and the flu. Examples of viruses that cause upper respiratory tract infections include rhino viruses and influenza viruses A and B. Common upper respiratory bacterial infections cause and/or associated with, for example, whooping cough and strep throat. An example of a bacterium that causes an upper respiratory tract infection is Streptococcus. Clinical manifestations of a lower respiratory infection include shallow coughing that produces sputum in the lungs, fever, and difficulty breathing. Examples of lower respiratory viral infections are parainfluenza virus infections ("PIN"), respiratory syncytial virus ("RSN"), human metapneumovirus (bJVTPN), and bronchiolitis. Examples of bacteria that cause lower respiratory tract infections include Streptococcus pneumoniae that causes pneumonococcal pneumonia and Mycobacterium tuberculosis that causes tuberculosis.
Respiratory infections caused by fungi include systemic candidiasis, blastomycosis crytococcosis, coccidioidomycosis, and aspergiUosis. Respiratory infections may be primary or secondary infections. Current therapies for respiratory infections involve the administration of anti-viral agents, anti-bacterial, and anti-fungal agents for the treatment, prevention, or amelioration of viral, bacterial, and fungal respiratory infections, respectively. Unfortunately, in regard to certain infections, there are no therapies available, infections have been proven to be refractory to therapies, or the occurrence of side effects outweighs the benefits of the administration of a therapy to a subject. The use of anti-bacterial agents for treatment of bacterial respiratory infections may also produce side effects or result in resistant bacterial strains. The administration of anti-fungal agents may cause renal failure or bone marrow dysfunction and may not be effective against fungal infection in patients with suppressed immune systems. Additionally, the infection causing microorganism (e.g., virus, bacterium, or fungus) may be resistant or develop resistance to the administered therapeutic agent or combination of therapeutic agents. In fact, microorganisms that develop resistance to administered therapeutic agents often develop pleiotropic drug or multidrug resistance, that is, resistance to therapeutic agents that act by mechanisms different from the mechanisms of the administered agents. Thus, as a result of drug resistance, many infections prove refractory to a wide array of standard treatment protocols. Therefore, new therapies for the treatment, prevention, management, or amelioration of respiratory infections and symptoms thereof are needed.
Autoimmune And Inflammation Inflammation is a process by which the body's white blood cells and chemicals protect our bodies from infection by foreign substances, such as bacteria and viruses. It is usually characterized by pain, swelling, warmth and redness of the affected area. Chemicals known as cytokines and prostaglandins control this process, and are released in an ordered and self- limiting cascade into the blood or affected tissues. This release of chemicals increases the blood flow to the area of injury or infection, and may result in the redness and warmth. Some of the chemicals cause a leak of fluid into the tissues, resulting in swelling. This protective process may stimulate nerves and cause pain. These changes, when occurring for a limited period in the relevant area, work to the benefit of the body. In autoimmune and/or inflammatory disorders, the immune system triggers an inflammatory response when there are no foreign substances to fight and the body's normally protective immune system causes damage to its own tissues by mistakenly attacking self. There are many different autoimmune disorders that affect the body in different ways. For example, the brain is affected in individuals with multiple sclerosis, the gut is affected in individuals with Crohn's disease, and the synovium, bone, cartilage of various joints are affected in individuals with rheumatoid arthritis and the skin this psoriasis. As autoimmune disorders progress destruction of one or more types of body tissues, abnormal growth of an organ, or changes in organ function may result. The autoimmune disorder may affect only one organ or tissue type or may affect multiple organs and tissues. Organs and tissues commonly affected by autoimmune disorders include red blood cells, blood vessels, connective tissues, endocrine glands (e.g., the thyroid or pancreas), muscles, joints, and skin. Examples of autoimmune disorders include, but are not limited to, Hashimoto's thyroiditis, pernicious anemia, Addison's disease, type 1 diabetes, rheumatoid arthritis, psoriasis, systemic lupus erythematosus, dermatomyositis, Sjogren's syndrome, dermatomyositis, lupus erythematosus, multiple sclerosis, autoimmune inner ear disease myasthenia gravis, Reiter's syndrome, Graves disease, autoimmune hepatitis, familial adenomatous polyposis and ulcerative colitis. Rheumatoid arthritis (RA) and juvenile rheumatoid arthritis are types of inflammatory arthritis. Arthritis is a general term that describes inflammation in joints. Some, but not all, types of arthritis are the result of misdirected inflammation. Besides rheumatoid arthritis, other types of arthritis associated with inflammation include the following: psoriatic arthritis, Reiter's syndrome, ankylosing spondylitis arthritis, and gouty arthritis. Rheumatoid arthritis is a type of chronic arthritis that occurs in joints on both sides of the body (such as both hands, wrists or knees). This symmetry helps distinguish rheumatoid arthritis from other types of arthritis. In addition to affecting the joints, rheumatoid arthritis may occasionally affect the skin, eyes, lungs, heart, blood or nerves. Rheumatoid arthritis affects about 1% of the world's population and is potentially disabling. There are approximately 2.9 million incidences of rheumatoid arthritis in the United States. Two to three times more women are affected than men. The typical age that rheumatoid arthritis occurs is between 25 and 50. Juvenile rheumatoid arthritis affects 71,000 young Americans (aged eighteen and under), affecting six times as many girls as boys. Rheumatoid arthritis is an autoimmune disorder where the body's immune system improperly identifies the synovial membranes that secrete the lubricating fluid in the joints as foreign. Inflammation results, and the cartilage and tissues in and around the joints are damaged or destroyed. In severe cases, this inflammation extends to other joint tissues and surrounding cartilage, where it may erode or destroy bone and cartilage and lead to joint deformities. The body replaces damaged tissue with scar tissue, causing the normal spaces within the joints to become narrow and the bones to fuse together. Rheumatoid arthritis creates1 stiffness, swelling, fatigue, anemia, weight loss, fever, and often, crippling pain. Some common symptoms of rheumatoid arthritis include joint stiffness upon awakening that lasts an hour or longer; swelling in a specific finger or wrist joints; swelling in the soft tissue around the joints; and swelling on both sides of the joint. Swelling can occur with or without pain, and can worsen progressively or remain the same for years before progressing. The diagnosis of rheumatoid arthritis is based on a combination of factors, including: the specific location and symmetry of painful joints, the presence of joint stiffness in the morning, the presence of bumps and nodules under the skin (rheumatoid nodules), results of X-ray tests that suggest rheumatoid arthritis, and/or positive results of a blood test called the rheumatoid factor. Many, but not all, people with rheumatoid arthritis have the rheumatoid-factor antibody in their blood. The rheumatoid factor may be present in people who do not have rheumatoid arthritis. Other diseases can also cause the rheumatoid factor to be produced in the blood. That is why the diagnosis of rheumatoid arthritis is based on a combination of several factors and not just the presence of the rheumatoid factor, in the blood. The typical course of the disease is one of persistent but fluctuating joint symptoms, and after about 10 years, 90% of sufferers will show structural damage to bone and cartilage. A small percentage will have a short illness that clears up completely, and another small percentage will have very severe disease with many joint deformities, and occasionally other manifestations of the disease. The inflammatory process causes erosion or destruction of bone and cartilage in the joints. In rheumatoid arthritis, there is an autoimmune cycle of persistent antigen presentation, T-cell stimulation, cytokine secretion, synovial cell activation, and joint destruction. The disease has a major impact on both the individual and society, causing significant pain, impaired function and disability, as well as costing millions of dollars in healthcare expenses and lost wages. (See, for example, the NTH website and the MAID website). Currently available therapy for arthritis focuses on reducing inflammation of the joints with anti-inflammatory or immunosuppressive medications. The first line of treatment of any arthritis is usually anti-inflammatories, such as aspirin, ibuprofen and Cox-2 inhibitors such as celecoxib and rofecoxib. "Second line drugs" include gold, methotrexate and steroids. Although these are well-established treatments for arthritis, very few patients remit on these lines of treatment alone. Recent advances in the understanding of the pathogenesis of rheumatoid arthritis have led to the use of methotrexate in combination with antibodies to cytokines or recombinant soluble receptors. For example, recombinant soluble receptors for tumor necrosis factor (TNF)-α have been used in combination with methotrexate in the treatment of arthritis. However, only about 50% of the patients treated with a combination of methotrexate and anti-TNF-α agents such as recombinant soluble receptors for TNF-α show clinically significant improvement. Many patients remain refractory despite treatment. Difficult treatment issues still remain for patients with rheumatoid arthritis. Many current treatments have a high incidence of side effects or cannot completely prevent disease progression. So far, no treatment is ideal, and there is no cure. Novel therapeutics are needed that more effectively treat rheumatoid arthritis and other autoimmune disorders. As noted supra, a potential role for mast cells in RA has been highlighted. Lee et al., 2002, Science 297:1689-1962 reported that mice that lacked mast cells are resistant to inflammatory and erosive arthritis induced by arthritogenic serum. Therefore it has been proposed that that the mast cell is a cellular link among the autoantibodies, the complement network, and inflammatory mediators that give rise to erosive arthritis. Woolley, 2003, N EnglJMed 34S: 1709-1711. The role of mast cells in autoimmune disorders initiated studies looking into the role of mast cells in multiple sclerosis and its animal model experimental allergic encephalomyelitis (EAE). See Steinmanm et al., 2001, Nature Immunol. 2:762-764. Multiple sclerosis a chronic inflammatory disorder of the central nervous system, which is characterized by a breach of the blood-brain barrier, mononuclear cell infiltration of white matter and eventual demyelinization. A similar autoimmune disease can be induced in susceptible rodent strains by injecting myelin components, including myelin basic protein, proteolipid protein and mylelin oligodendrocyte glycoprotein. Until recently it was thought that mast cells were not involved with multiple sclerosis. However, Secor et al. (2000, J. Exp. Med. 191 :813-822) showed that mice lacking mast cells developed EAE later and less severely than do control mice in response to injection with mylelin oligodendrocyte glycoprotein. Complementation of the mice with immature mast cells derived in vitro restored typical EAE susceptibility.
Cardiac Disease Atherosclerosis is a disease-state in which the walls of the arteries become thickened and lose their elasticity, resulting in a decreased ability to pump blood to peripheral organs and withstand systemic pressure. The most common form of atherosclerosis is caused by the build- up of fatty deposits in the innermost layer of the artery wall. Another form of atherosclerosis disease involves the destruction of the smaller arteries, or arterioles. All forms of the disease contribute to a decreased blood flow to vital organs, which can lead to a stroke, heart attack, or kidney failure. Traditional risk factors implicated in the development and progression of cardiovascular diseases such as atherosclerosis and for the predisposition of unstable angina, myocardial infarction and stroke include, cigarette smoking, hypertension, dyslipidemia, diabetes mellitus, sedentary lifestyle, obesity, the imbalance of the hemostatic/fibrinolytic system and a family history of premature coronary disease. However, these risk factors explain only a portion of the documented cardiovascular disease. Other factors must play a role in the etiology of the vascular events in this disease. Recent studies have suggested that immunologic mechanisms also play a role in the pathogenesis of cardiovascular disease. For example, it has been shown that coronary inflammation may depend on activated mast cell-derived mediators (Laine et al., 1998, J. Pharm. Exp. Therap. 287:307). Ixi addition, acute stress also activates cardiac mast cells leading to the release of inflammatory components (Pang et al., 1998, J. Pharm. Exp. Therap. 287:307; Huang et al., 2002, Cardiovasc Res. 55:13-5). Furthermore, Harris et al. (in PCT application WO02/067938) found that common immunological cells, mediators and/or messengers which are responsible for disease expression in allergy also subsequently increases the risk and/or severity of cardiovascular disease. These immunological cells include mast cells, while immunological mediators and messengers include cytokines (e.g.,L-4, LL-13, GM-CSF, TNFa, IL-6), chemokines, adhesion molecules, and mast cells mediators (leukotrienes, histamine). Harris et al. further report that cardiac mast cell activation can occur, and that down-regulation of systemic inflammation by anti- inflammatory therapies of the present invention lowers the risk and/or severity of atherosclerosis and cardiovascular disease. Thus, the increasing knowledge of the pathogenesis of atherosclerosis suggests that prevention of cardiovascular disease will involve not only the correction of the above risk factors, but also the direct pharmacological control of atherogenic processes occurring in the arterial wall (Ross, R., 1993, Nature, 362:801-809). The immunologic responses to allergic and or inflammatory conditions which are implicated in increased risk of cardiovascular diseases indicate that mast cells are prime targets in the development of effective therapies to treat, reduce or prevent a cardiovascular disease in patients at risk.
HAAH The present invention is based, in part, on the inventors' discovery that human aspartyl (asparaginyl)-β hydroxylase (HAAH) is strongly expressed on the surface of mast cells. HAAH is a protein belonging to the α-ketoglutarate dependent dioxygenase family of prolyl and lysyl hydroxylases which play a key role in collagen biosynthesis. This molecule hydroxylates aspartic acid or asparagine residues in EGF-like domains of several proteins in the presence of ferrous iron. These EGF-like domains contain conserved motifs that form repetitive sequences in proteins such as clotting factors, extracellular matrix proteins, LDL receptor, NOTCH homologues or NOTCH ligand homologues. The 4.3-kb cDNA encoding the human aspartyl (asparaginyl)-β hydroxylase hybridizes with 2.6 kb and 4.3 kb transcripts in transformed cells, and the deduced amino acid sequence of the larger transcript encodes a protein of about 85-kDa. Both in vitro transcription and translation and Western blot analysis also demonstrate a 56-kDa protein that may result from posttranslational cleavage of the catalytic C terminus.
HAAH has several conserved functional domains including Aspartyl beta- hydroxylase N-terminal and Aspartyl/ Asparaginyl beta-hydroxylase C-terminal (residues 43- 309 and 589-746) regions that are critical for catalytic activity as well as a central tetratricopeptide repeat domain (residues 421-520) which likely plays a role in mediating protein-protein interactions, i addition, HAAH contains a hydrophobic region located between residues 54 and 75 and flanked by positively charged amino acid residues at its amino-terminal side and negatively charged residues at its carboxyl-terminal side that is a signal anchor of a type π transmembrane protein (Schutze et al., 1994, EMBO J. 13:1696- 1705). Thus, the predicted orientation of the HAAH polypeptide is that residues 1-45 of the NH2 terminus are in the cytoplasm and the remainder of the molecule, containing the catalytic domain, in the lumen of the endoplasmic reticulum and/or on the cell surface. A physiological function ofHAAH is the post-translational beta- hydroxylation of aspartic acid in vitamin K-dependent coagulation proteins. However, the abundant expression ofHAAH in several malignant neoplasms, and the low levels ofHAAH in many normal cells indicate a role for this enzyme in malignancy. The HAAH gene is also highly expressed in cytotrophoblasts, but not syncytiotrophoblasts of the placenta. Cytotrophoblasts are invasive cells that mediate placental implantation. The increased levels ofHAAH expression in human cholangiocarcinomas, hepatocellular carcinomas, colon cancers, and breast carcinomas were primarily associated with invasive or metastatic lesions. Moreover, overexpression ofHAAH does not strictly reflect increased DNA synthesis and cellular proliferation since high levels ofHAAH immunoreactivity were observed in 100 percent of cholangiocarcinomas, but not in human or experimental disease processes associated with regeneration or nonneoplastic proliferation of bile ducts. HAAH overexpression and attendant high levels of beta hydroxylase activity lead to invasive growth of transformed neoplastic cells. Detection of an increase in HAAH expression is useful for early and reliable diagnosis of the cancer types which have now been characterized as overexpressing this gene product. See WO 01/35102, incorporated by reference herein in its entirety. In addition to increased HAAH expression in neoplastic cells, the inventors have discovered the HAAH is strongly expressed on the surface of a mast cell. SUMMARY OF THE INVENTION The present invention provides novel compositions and methods of modulating mast cells by modulating HAAH activity. In one embodiment, the method of modulating a mast cell includes antibody mediated-cellular cytotoxicity (ADCC), or apoptosis by agonizing or antagonizing HAAH to induce mast cell death. In another embodiment the agent that modulates HAAH activity immunospecifically binds to HAAH on the surface of the mast cell. In a preferred embodiment the agent that modulates HAAH activity is an inhibitor of HAAH activity. HAAH inhibitors of the invention include, without limitation, antibodies, peptides, small molecules, antisense molecules, inhibitory RNA, and ribozymes, which are capable of inhibiting or preventing 1) the activity of a HAAH molecule of the invention on mast cell activity or development; or 2) the binding of a HAAH molecule with other binding partners; or 3) the biological activity of a HAAH molecule of the invention; or 4) the expression of a HAAH molecule of the invention by a cell. The present invention comprises methods for identifying compounds capable of depleting mast cells and/or preventing mast cell activation, wherein said compounds immunospecifically bind to HAAH on the surface of the mast cell. In a more preferred embodiment, the present invention comprises methods for identifying compounds capable of depleting mast cells and/or preventing mast cell activation, where in said compounds immunospecifically bind to HAAH on the surface of the mast cells and exhibit little or no binding to the surface of other hematopoeitic cells that are not mast cells or related cells or cell lines or derived cell lines thereof. The invention also encompasses a method for identifying compounds that immunospecifically binds to HAAH and are capable of depleting mast cells and/or preventing mast cell activation, wherein said compounds are non-toxic for other hematopoeitic cells that are not mast cells or related cells or cell lines or derived cell lines thereof. The present invention also provides for prophylactic and therapeutic protocols designed to prevent, manage, treat, or ameliorate respiratory conditions (e.g. asthma) or one or more symptoms thereof and to treat, manage or ameliorate an inflammatory or an autoimmune disorder or cardiac disease. In particular, the present invention provides methods for the prevention, management, treatment, or amelioration of respiratory conditions (e.g. asthma) or one or more symptoms thereof and/or to the treatment, management or amelioration of an inflammatory or an autoimmune disorder or cardiac disease, said methods comprising administering to a subject in need thereof an effective amount of one or more agents that modulate the activity of HAAH. In one embodiment, the respiratory condition or inflammatory disease being treated includes but is of not limited to asthma, chronic obstructive pulmonary disease, interstitial lung disease, chronic obstructive lung disease, chronic bronchitis, eosinophilic bronchitis, eosinophilic pneumonia, pneumonia, inflammatory bowel disease, atopic dermatitis, atopy, allergy, allergic rhinitis, idiopathic pulmonary fibrosis, scleroderma, rheumatoid arthritis and emphysema. , In another embodiment, the cardiac disease is a coronary heart disease (e.g. atherosclerosis). BRIEF DESCRIPTION OF THE FIGURES FIGURE 1 hrrmunohistochemistry of normal human tissues stained with an anti- HAAH specific antibody. A) Section of normal small intestine. B) Section of normal muscle tissue. C) Section of normal heart. Arrows indicate some of the intensely staining mast cells. FIGURE 2 is a Western blot of whole cell lysates. Molecular weight marker sizes are indicated and the arrow represents the position ofHAAH. FIGURE 3 is FACS analysis of UT-7 cells. The cells were labeled with A) no antibody. B) Secondary antibody alone. C) Anti-HAAH and secondary antibodies. FIGURE 4 is FACS analysis of KU812 cells. The cells were labeled with A) anti- HAAH and secondary antibodies. B) Secondary antibody alone. C) No antibody. DETAILED DESCRIPTION OF THE INVENTION The present invention is based, in part, on the inventors' discovery that human aspartyl (asparaginyl)-β hydroxylase (HAAH) is strongly expressed on the surface of mast cells. In particular, the invention provides prophylactic and therapeutic protocols for the prevention, treatment, management, or amelioration of respiratory conditions (e.g. asthma) or one or more symptoms thereof and/or to treat, manage or ameliorate an inflammatory or an autoimmune disorder or a cardiac disease, said protocol comprising admimstering to a subject in need thereof a prophylactically or therapeutically effective amount of one or more of HAAH antagonists alone or in combination with a prophylactically or therapeutically effective amount of at least one other therapy (e.g., at least one other prophylactic or therapeutic agent) other than HAAH antagonists. HAAH antagonists of the invention can include, but should not be construed as being limited to a chemical compound, a protein, a peptidomemetic, an antibody, a ribozyme, and an antisense nucleic acid molecule. HAAH antagonists of the invention includes, but not limited to, antibodies, antibody fragments, single chain antibodies, small molecules, fusion proteins, and BiTE molecules. The antagonists of the invention can modulate mast cell activity by preventing activation, degranulation or stimulation, or by inducing apoptosis, antibody mediated cell cytotoxicity (ADCC), complement mediated killing, a molecule conjugated to a toxin or drug (e.g. antibody conjugated to ricin), and/or direct killing of mast cells (e.g. small molecule). The above list is not meant to be an exhaustive list but just a representation of ways to modulate mast cell activity using a HAAH antagonist.
Antibodies or fragments thereof that immunospecifically bind to HAAH and modulate mast cells are specifically preferred embodiments of the invention. Preferably, antibodies or fragments thereof that immunospecifically bind to the HAAH polypeptides, or a fragment thereof, do not cross-react with other non-HAAH antigens. It is contemplated that an antibody or fragment thereof that immunospecifically binds to the human HAAH could also bind the ortholog of other species (i.e. mouse, rat, chimp). Preferred anti-HAAH antibodies of the invention recognize epitopes present on the extracellular portion of the protein. A more preferred anti-HAAH antibody of the invention will bind to the catalytic domain ofHAAH. A still more preferred anti-HAAH antibody of the invention will inhibit the activity ofHAAH. Another preferred anti-HAAH antibody will bind to the tetratricopeptide repeat domain ofHAAH. Yet another preferred anti-HAAH antibody will bind to HAAH and prevent interaction with HAAH binding partners. Antibodies or fragments that immunospecifically bind to HAAH polypeptides, or a fragment thereof can be identified, for example, by immunoassays or other techniques known to those of skill in the art. Antibodies of the invention include, but are not limited to, synthetic antibodies, monoclonal antibodies, recombinantly produced antibodies, intrabodies, multispecific antibodies, human antibodies, humanized antibodies, chimeric antibodies, synthetic antibodies, single-chain Fvs (scFv), Fab fragments, F (ab') fragments, disulfide- linked Fvs (sdFv), and anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above. In particular, antibodies of the present invention include immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen-binding site that immunospecifically binds to a HAAH poiypeptide (e.g., one or more complementarity determining regions (CDRs) of an anti-HAAH antibody). Preferably inhibitory or antagonistic antibodies or fragments that immunospecifically bind to a HAAH polypeptide, or a fragment thereof, preferentially modulate mast cells, and do not significantly modulate other cells types. "Modulate mast cells", as used herein, includes but is not limited to changes in, mast cell maturation, differentiation, proliferation, activation, stimulation, migration, degranulation, as well as mast cell inhibition and depletion. In a preferred embodiment, HAAH antagonists of the invention result in a depletion of mast cells or prevent mast cell activation. Furthermore, the invention provides prophylactic and therapeutic protocols for the prevention treatment, management, or amelioration of respiratory conditions (e.g. asthma) or one or more symptoms thereof and/or to treat, manage or ameliorate an inflammatory or an autoimmune disorder, said protocols comprising administering to a subject in need thereof a prophylactically or therapeutically effective amount of an HAAH antibody alone or in combination with a prophylactically or therapeutically effective amount of at least one other therapy (e.g., at least one other prophylactic or therapeutic agent) other than an HAAH antibody. The present invention also provides for pharmaceutical compositions, kits, and articles of manufacture comprising one or more HAAH antagonists for use in the prevention, treatment, management, or amelioration of respiratory conditions and/or an inflammatory or autoimmune disease or one or more symptoms thereof. The present invention also provides for pharmaceutical compositions, kits, and articles of manufacture comprising one or more HAAH antagonists (e.g. antibodies, small molecules, fusion proteins and/or BiTE molecule) and one or more prophylactic or therapeutic agents other than HAAH antagonists for use in prevention, treatment, management, or amelioration of respiratory conditions or one or more symptoms thereof.
Production of HAAH-specific Antibodies As discussed above, the invention encompasses administration of antibodies (preferably monoclonal antibodies) or fragments thereof that immunospecifically bind to HAAH polypeptides and modulate mast cells. Antibodies of the invention include, but are not limited to, monoclonal antibodies, synthetic antibodies, recombinantly produced antibodies, intrabodies, multispecific antibodies (including bi-specific antibodies), human antibodies, humanized antibodies, chimeric antibodies, single-chain Fvs (scFv) (including bi-specific scFvs), single chain antibodies, Fab fragments, F (ab') fragments, disulfide-linked Fvs (sdFv), and epitope-binding fragments of any of the above. In particular, antibodies used in the methods of the present invention include immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that immunospecifically binds to HAAH or a fragment thereof, and is a modulator of mast cells (e.g., proliferation, differentiation, activation, degranulation, migration). Furthermore, antibodies of the present invention preferentially bind a HAAH polypeptide epitope, and/or binds a HAAH polypeptide of the invention with a Koff of less than 3 X 10"3 s"1. The immunoglobulin molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGi, IgG2, IgG3, IgG4, IgAi and IgA2) or subclass of immunoglobulin molecule. The antibodies used in the methods of the invention maybe from any animal origin including birds and mammals (e.g., human, murine, donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken). Preferably, the antibodies are human or humanized monoclonal antibodies. As used herein, "human" antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from mice or other animals that express antibodies from human genes. The antibodies used in the methods of the present invention may be monospecific, bispecific, trispecific or of greater multispecificity. Multispecific antibodies may immunospecifically bind to different epitopes of a HAAH polypeptide or may immunospecifically bind to both a HAAH polypeptide as well a heterologous epitope, such as a heterologous polypeptide or solid support material. See, e.g., International Publication Nos. WO 93/17715, WO 92/08802, WO 91/00360, and WO 92/05793; Tutt, et al., 1991, J. Immunol. 147:60-69; U.S. Patent Nos. 4,474,893, 4,714,681, 4,925,648, 5,573,920, and 5,601,819; and Kostelny et al., 1992, J. Immunol. 148:1547-1553. The antibodies used in the methods of the invention include derivatives that are modified, i.e., by the covalent attachment of any type of molecule to the antibody. For example, but not by way of limitation, the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to, specific chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc. Additionally, the derivative may contain one or more non-classical amino acids. The present invention encompasses single domain antibodies, including camelized single domain antibodies (see e.g., Muyldermans et al., 2001, Trends Biochem. Sci. 26:230; Nuttall et al., 2000, Cur. Pharm. Biotech. 1:253; Reichmann and Muyldermans, 1999, J. Immunol. Meth. 231:25; International Publication Nos. WO 94/04678 and WO 94/25591; U.S. Patent No. 6,005,079; which are incorporated herein by reference in their entireties). In one embodiment, the present invention provides single domain antibodies comprising two NH domains having the amino acid sequence of any of the NH domains of a HAAH antibody with modifications such that single domain antibodies are formed. In another embodiment, the present invention also provides single domain antibodies comprising two NH domains comprising one or more of the NH CDRs of an HAAH antibody. The methods of the present invention also encompass the use of antibodies or fragments thereof that have half-lives (e.g., serum half-lives) in a mammal, preferably a human, of greater than 15 days, preferably greater than 20 days, greater than 25 days, greater than 30 days, greater than 35 days, greater than 40 days, greater than 45 days, greater than 2 months, greater than 3 months, greater than 4 months, or greater than 5 months. The increased half-lives of the antibodies of the present invention or fragments thereof in a mammal, preferably a human, result in a higher serum titer of said antibodies or antibody fragments in the mammal, and thus, reduce the frequency of the administration of said antibodies or antibody fragments and/or reduces the concentration of said antibodies or antibody fragments to be administered. Antibodies or fragments thereof having increased in vivo half-lives can be generated by techniques known to those of skill in the art. For example, antibodies or fragments thereof with increased in vivo half-lives can be generated by modifying (e.g., substituting, deleting or adding) amino acid residues identified as involved in the interaction between the Fc domain and the FcRn receptor (see, e.g., international Publication Νos. WO 97/34631 and WO 02/060919, which are incorporated herein by reference in their entireties). Antibodies or fragments thereof with increased in vivo half-lives can be generated by attaching to said antibodies or antibody fragments polymer molecules such as high molecular weight polyethyleneglycol (PEG). PEG can be attached to said antibodies or antibody fragments with or without a multifunctional linker either through site-specific conjugation of the PEG to the N- or C- terminus of said antibodies or antibody fragments or via epsilon- amino groups present on lysine residues. Linear or branched polymer derivatization that results in minimal loss of biological activity will be used. The degree of conjugation will be closely monitored by SDS-PAGE and mass spectrometry to ensure proper conjugation of PEG molecules to the antibodies. Unreacted PEG can be separated from antibody-PEG conjugates by, e.g., size exclusion or ion-exchange chromatography. Standard techniques known to those skilled in the art can be used to introduce mutations in the nucleotide sequence encoding an antibody, or fragment thereof, including, e.g., site-directed mutagenesis and PCR-mediated mutagenesis, which results in amino acid substitutions. Preferably, the derivatives include less than 15 amino acid substitutions, less than 10 amino acid substitutions, less than 5 amino acid substitutions, less than 4 amino acid substitutions, less than 3 amino acid substitutions, or less than 2 amino acid substitutions relative to the original antibody or fragment thereof. In a preferred embodiment, the derivatives have conservative amino acid substitutions made at one or more predicted non- essential amino acid residues. The present invention further encompasses antibodies or fragments thereof that immunospecifically bind to HAAH and modulated a mast cells (e.g. inhibit mast cell activity), said antibodies or antibody fragments comprising an amino acid sequence of one or more CDRs that is at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% identical to the amino acid sequence of one or more CDRs of a HAAH antibody. The determination of percent identity of two amino acid sequences can be determined by any method known to one skilled in the art, including BLAST protein searches.
Antibody Conjugates The present invention encompasses the use of antibodies or fragments thereof that bind to HAAH recombinantly fused or chemically conjugated (including both covalent and non-covalent conjugations) to a heterologous agent. The heterologous agent may be a polypeptide (or portion thereof, preferably to a polypeptide of at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90 or at least 100 amino acids), nucleic acid, small molecule (less than 1000 daltons), or inorganic or organic compound. The fusion does not necessarily need to be direct, but may occur through linker sequences. For example, antibodies may be used to target heterologous polypeptides to particular cell types, either in vitro or in vivo, by fusing or conjugating the antibodies to antibodies specific for particular cell surface receptors. Antibodies fused or conjugated to heterologous polypeptides may also be used in in vitro immunoassays and purification methods using methods known in the art. See e.g., International Publication WO 93/21232; EP 439,095; Naramura et al., 1994, Immunol. Lett. 39:91-99; U.S. Patent 5,474,981; Gillies et al., 1992, PNAS 89:1428-1432; and Fell et al, 1991, J. Immunol. 146:2446-2452, which are incorporated by reference in their entireties. The present invention further includes compositions comprising heterologous agents fused or conjugated to antibody fragments. For example, the heterologous polypeptides may be fused or conjugated to a Fab fragment, Fd fragment; Fv fragment, F (ab) 2 fragment, or portion thereof. Methods for fusing or conjugating polypeptides to antibody portions are known in the art. See, e.g., U.S. Patent Nos. 5,336,603, 5,622,929, 5,359,046, 5,349,053, 5,447,851, and 5,112,946; EP 307,434; EP 367,166; International Publication Nos. WO 96/04388 and WO 91/06570; Ashkenazi et al., 1991, PNAS 88: 10535-10539; Zheng et al., 1995, J. Immunol. 154:5590-5600; and Nil et al, 1992, PNAS 89:11337- 11341 (said references incorporated by reference in their entireties). Additional fusion proteins may be generated through the techniques of gene-shuffling, motif-shuffling, exon-shuffling, and/or codon-shuffling (collectively referred to as "DΝA shuffling"). DΝA shuffling may be employed to alter the activities of antibodies of the invention or fragments thereof (e.g., antibodies or fragments thereof with higher affinities and lower dissociation rates). See, generally, U.S. Patent os. 5,605,793; 5,811,238; 5,830,721; 5,834,252; and 5,837,458, and Patten et al, 1997, Curr. Opinion Biotechnol. 8:724-33; Harayama, 1998, Trends Biotechnol. 16:76; Hansson, et al., 1999, J. Mol. Biol. 287:265; and Lorenzo and Blasco, 1998, BioTechniques 24:308 (each of these patents and publications are hereby incorporated by reference in its entirety). Antibodies or fragments thereof, or the encoded antibodies or fragments thereof, may be altered by being subjected to random mutagenesis by error-prone PCR, random nucleotide insertion or other methods prior to recombination. One or more portions of a polynucleotide encoding an antibody or antibody fragment, which portions immunospecifically bind to HAAH may be recombined with one or more components, motifs, sections, parts, domains, fragments, etc. of one or more heterologous agents. In one embodiment, antibodies of the present invention or fragments or variants thereof are conjugated to a marker sequence, such as a peptide, to facilitate purification. In preferred embodiments, the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available. As described in Gentz et al., 1989, PNAS 86:821, for instance, hexa-histidine provides for convenient purification of the fusion protein. Other peptide tags useful for purification include, but are not limited to, the hemagglutinin "HA" tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al, 1984, Cell 37:767) and the "Flag" tag. In other embodiments, antibodies of the present invention or fragments or variants thereof are conjugated to a diagnostic or detectable agent. Such antibodies can be useful for monitoring or prognosing the development or progression of an asthma and/or autoimmune or inflammatory disease as part of a clinical testing procedure, such as determining the efficacy of a particular therapy. Such diagnosis and detection can accomplished by coupling the antibody to detectable substances including, but not limited to various enzymes, such as but not limited to horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; prosthetic groups, such as but not limited to streptavidin/biotin and avidin/biotin; fluorescent materials, such as but not limited to, umbelliferone, fluorescein, fluorescein isothiocynate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; luminescent materials, such as but not limited to, luminol; bioluminescent materials, such as but not limited to, luciferase, luciferin, and aequorin; radioactive materials, such as but not limited to, bismuth (213Bi), carbon (14C), chromium (51Cr), cobalt (57Co), fluorine (18F), gadolinium (153Gd, 159Gd), gallium (68Ga, 67Ga), germanium (68Ge), holmium (166Ho), indium (115In, 113In, 112ln, mIn), iodine (1311, 1251, 1231, 121I), lanthanium (140La), lutetium (177Lu), manganese (54Mn), molybdenum (99Mo), palladium (103Pd), phosphorous (32P), praseodymium (142Pr), promethium (149Pm), rhenium (186Re, 188Re), rhodium (105Rh), ruthemium (97Ru), samarium (153Sm), scandium (47Sc), selenium (75Se), strontium (85Sr), sulfur (35S), technetium (99Tc), thallium (201Ti), tin (113Sn, 117Sn), tritium (3H), xenon (133Xe), ytterbium (16 Yb, 175Yb), yttrium (90Y), zinc (65Zn); positron emitting metals using various positron emission tomographies, and nonradioactive paramagnetic metal ions. In other embodiments, antibodies of the present invention or fragments or variants thereof are conjugated to a therapeutic agent such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters. A cytotoxin or cytotoxic agent includes any agent that is detrimental to cells. Examples include paclitaxel, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1- dehydrotestosterone, glucocorticoids, procaine, tetracaine, onconase, lidocaine, propranolol, puromycin, epirubicin, and cyclophosphamide and analogs or homologs thereof. Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BCNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cisdichlorodiamine platinum (11) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine). In other embodiments, antibodies of the present invention or fragments or variants thereof are conjugated to a therapeutic agent or drug moiety that modifies a given biological response. Therapeutic agents or drug moieties are not to be construed as limited to classical chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, cholera toxin, or diphtheria toxin; a protein such as tumor necrosis factor, α-interferon, β-interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF-α, TNF-β, AIM I (see, International Publication No. WO 97/33899), AIM II (see, International Publication No. WO 97/34911), Fas Ligand (Takahashi et al, 1994, J. Iminunol., 6:1567), and NEGI (see, International Publication No. WO 99/23105), a thrombotic agent or an anti-angiogenic agent, e.g., angiostatin or endostatin; or, a biological response modifier such as, for example, a lymphokine (e.g., interleukin-1 ("IL-1"), interleukin-2 ("IL-2"), interleukin-6 ("IL-6"), granulocyte macrophage colony stimulating factor ("GM-CSF"), and granulocyte colony stimulating factor ("G-CSF")), or a growth factor (e.g., growth hormone ("GH")). hi other embodiments, antibodies of the present invention or fragments or variants thereof are conjugated to a therapeutic agent such as a radioactive materials or macrocyclic chelators useful for conjugating radiometal ions (see above for examples of radioactive materials). In certain embodiments, the macrocyclic chelator is 1,4,7,10- tetraazacyclododecane-N,N',N",N"-tetraacetic acid (DOTA) which can be attached to the antibody via a linker molecule. Such linker molecules are commonly known in the art and described in Denardo et al., 1998, Clin Cancer Res. 4:2483-90; Peterson et al, 1999, Bioconjug. Chem. 10:553; and Zimmerman et al., 1999, Nucl. Med. Biol. 26:943-50 each incorporated by reference in their entireties. Techniques for conjugating therapeutic moieties to antibodies are well known, see, e.g., Arnon et al., "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp. 243-56 (Alan R. Liss, Inc. 1985); Hellstrom et al., "Antibodies For Drug Delivery", in Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp. 623-53 (Marcel Dekker, Inc. 1987); Thorpe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review", in Monoclonal Antibodies '84: Biological And Clinical Applications, Pinchera et al. (eds.), pp. 475-506 (1985); "Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy", in Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thorpe et al., 1982, Immunol. Rev. 62:119-58. Methods for fusing or conjugating antibodies to polypeptide moieties are known in the art. See, e.g., U.S. Patent Nos. 5,336,603, 5,622,929, 5,359,046, 5,349,053, 5,447,851, and 5,112,946; EP 307,434; EP 367,166; International Publication Nos. WO 96/04388 and WO 91/06570; Ashkenazi et al, 1991, PNAS 88: 10535-10539; Zheng et al, 1995, J. Immunol. 154:5590-5600; and Nil et al., 1992, PNAS 89:11337- 11341. The fusion of an antibody to a moiety does not necessarily need to be direct, but may occur through linker sequences. Such linker molecules are commonly known in the art and described in Denardo et al., 1998, Clin Cancer Res. 4:2483-90; Peterson et al., 1999, Bioconjug. Chem. 10:553; Zimmerman et al., 1999, Nucl. Med. Biol. 26:943-50; Garnett, 2002, A dv. DrugDeliv. Rev. 53:171-216, each ofwhich is incorporated herein by reference in its entirety. Alternatively, an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described by Segal in U.S. Patent No. 4,676,980, which is incorporated herein by reference in its entirety. The method and antibodies of the present invention also encompasses antibodies that are bispecific. In a another embodiment, antibodies of the invention are bispecific T cell engagers (BiTE). Bispecific T cell engagers (BiTE) are bispecific antibodies that can redirect T cells for antigen-specific elimination of targets. A BiTE molecule has an antigen-binding domain that binds to a T cell antigen (e.g. CD3) at one end of the molecule and an antigen binding domain that will bind to an antigen on the target cell. A BiTE molecule was recently described in WO 99/54440, which is herein incorporated by reference. This publication describes a novel single-chain multifunctional polypeptide that comprises binding sites for the CD19 and CD3 antigens (CD19xCD3). This molecule was derived from two antibodies, one that binds to CD 19 on the B cell and an antibody that binds to CD3 on the T cells. The variable regions of these different antibodies are linked by a polypeptide sequence, thus creating a single molecule. Also described, is the linking of the variable heavy chain (NH) and light chain (NL) of a specific binding domain with a flexible linker to create a single chain, bispecific antibody. In an embodiment of this invention, an antibody or ligand that immunospecifically binds to HAAH will comprise a portion of the BiTE molecule. For example, the NH and/or NL (preferably a scFN) of an antibody that binds HAAH can be fused to an anti-CD3 binding portion such as that of the molecule described above, thus creating a BiTE molecule that targets HAAH on the mast cells. In addition to the variable heavy and or light chain of antibody against HAAH, other molecules that bind HAAH can comprise the BiTE molecule. In another embodiment, the BiTE molecule can comprise a molecule that binds to other T cell antigens (other than CD3). For example, ligands and/or antibodies that immunospecifically bind to T-cell antigens like CD2, CD4, CD8, CDl la, TCR, and CD28 are contemplated to be part of this invention. This list is not meant to be exhaustive but only to illustrate that other molecules that can immunospecifically bind to a T cell antigen can be used as part of a BiTE molecule. These molecules can include the NH and/or NL portions of the antibody or natural ligands (for example LFA3 whose natural ligand is CD3). The "binding domain" as used in accordance with the present invention denotes a domain comprising a three-dimensional structure capable of specifically binding to an epitope like native antibodies, free scFv fragments or one of their corresponding immunoglobulin chains, preferably the NH chain. Thus, said domain can comprise the NH and/or NL domain of an antibody or an immunoglobulin chain, preferably at least the NH domain or more preferably the NH and NL domain linked by a flexible polypeptide linker (scFv). On the other hand, said binding domain contained in the polypeptide of the invention may comprise at least one complementarity determining region (CDR) of an antibody or immunoglobulin chain recognizing an antigen on the T cell or a cellular antigen. In this respect, it is noted that the binding domain present in the polypeptide of the invention may not only be derived from antibodies but also from other T cell or cellular antigen binding protein, such as naturally occurring surface receptors or ligands. It is further contemplated that in an embodiment of the invention, said first and or second domain of the above- described polypeptide mimic or correspond to a NH and NL region from a natural antibody. The antibody providing the binding site for the polypeptide of the invention can be, e.g., a monoclonal antibody, polyclonal antibody, chimeric antibody, humanized antibody, bispecific antibody, synthetic antibody, antibody fragment, such as Fab, Fv or scFv fragments etc., or a chemically modified derivative of any of these. Antibodies may also be attached to solid supports, which are particularly useful for immunoassays or purification of the target antigen. Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
Methods Of Producing Antibodies The antibodies or fragments thereof can be produced by any method known in the art for the synthesis of antibodies, in particular, by chemical synthesis or preferably, by recombinant expression techniques. Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof. For example, monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling, et al., in: Monoclonal Antibodies and T-Cell Hybridomas 563-681 (Elsevier, Ν.Y., 1981); and Using Antibodies: A Laboratory Manual, E. Harlow and D. Lane, ed., Cold Spring Harbor Laboratory (Cold Spring Harbor, NY, 1999) (said references incorporated by reference in their entireties). The term "monoclonal antibody" as used herein is not limited to antibodies produced through hybridoma technology. The term "monoclonal antibody" refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced. Methods for producing and screening for specific antibodies using hybridoma technology are routine and well known in the art. Briefly, mice can be immunized with HAAH (either the full length protein or a domain thereof, e.g., the catalytic domain) and once an immune response is detected, e.g., antibodies specific for HAAH are detected in the mouse serum, the mouse spleen is harvested and splenocytes isolated. The splenocytes are then fused by well known techniques to any suitable myeloma cells, for example cells from cell line SP20 available from the ATCC. Hybridomas are selected and cloned by limited dilution. Hybridoma clones are then assayed by methods known in the art for cells that secrete antibodies capable of binding a polypeptide of the invention. Ascites fluid, which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones. Antigenic epitopes ofHAAH preferably contain a sequence of at least 4, at least 5, at least 6, at least 7, more preferably at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, and, most preferably, between about 15 to about 30 amino acids. Preferred polypeptides comprising immunogenic or antigenic epitopes are at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 amino acid residues in length. These antigenic epitopes can be used to raise antibodies; including monoclonal antibodies, that specifically bind the epitope. Preferred antigenic epitopes include the antigenic epitopes disclosed herein, as well as any combination of two, three, four, five or more of these antigenic epitopes. Antigenic epitopes can be used as the target molecules in immunoassays. (See, for instance, Wilson et al., Cell 37:767- 778 (1984); Sutcliffe et al., Science 219:660-666 (1983)). The polypeptides comprising one or more immunogenic epitopes may be presented for eliciting an antibody response together with a carrier protein, such as an albumin, to an animal system (such as rabbit or mouse), or, if the polypeptide is of sufficient length (at least about 25 amino acids), the polypeptide may be presented without a carrier. However, immunogenic epitopes comprising as few as 8 to 10 amino acids have been shown to be sufficient to raise antibodies capable of binding to, at the very least, linear epitopes in a denatured polypeptide (e.g., in Western blotting). Once the spleens are harvested and fused, monoclonal antibodies can be generated by culturing a hybridoma cell secreting an antibody of the invention wherein, preferably, the hybridoma is generated by fusing splenocytes isolated from a mouse immunized with HAAH or fragment thereof with myeloma cells and then screening the hybridomas resulting from the fusion for hybridoma clones that secrete an antibody able to bind HAAH. Antibody fragments which recognize specific HAAH epitopes may be generated by any technique known to those of skill in the art. For example, Fab and F(ab')2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments). F(ab')2 fragments contain the variable region, the light chain constant region and the CHI domain of the heavy chain. Further, the antibodies of the present invention can also be generated using various phage display methods known in the art. hi phage display methods, functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them. In particular, DNA sequences encoding VH and NL domains are amplified from animal cDΝA libraries (e.g., human or murine cDΝA libraries of lymphoid tissues). The DΝA encoding the NH and NL domains are recombined together with an scFv linker by PCR and cloned into a phagemid vector (e.g., p CANTAB 6 or pComb 3 HSS). The vector is electroporated in E. coli and the E. coli is infected with helper phage. Phage used in these methods are typically filamentous phage including fd and Ml 3 and the VH and VL domains are usually recombinantly fused to either the phage gene m or gene NHL Phage expressing an antigen binding domain that binds to the HAAH epitope of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead. Examples of phage display methods that can be used to make the antibodies of the present invention include those disclosed in Brinkman et al., 1995, J. Immunol. Methods 182:41-50; Ames et al., 1995, J. Immunol. Methods 184:177; Kettleborough et al., 1994, Eur. J. Immunol. 24:952-958; Persic et al., 1997, Gene 187:9; Burton et, al., 1994, Advances in Immunology 57:191-280; International Application No. PCT/GB91/01134; International Publication Nos. WO 90/02809, WO 91/10737, WO 92/01047, WO 92/18619, WO 93/1 1236, WO 95/15982, WO 95/20401, and WO97/13844; and U.S. Patent Nos. 5,698,426, 5,223,409, 5,403,484, 5,580,717, 5,427,908, 5,750,753, 5,821,047, 5,571,698, 5,427,908, 5,516,637, 5,780,225, 5,658,727, 5,733,743 and 5,969,108; each ofwhich is incorporated herein by reference in its entirety. hi a preferred embodiment,, after phage selection, the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described below. Techniques to recombinantly produce Fab, Fab' and F(ab')2 fragments can also be employed using methods known in the art such as those disclosed in International Publication No. WO 92/22324; Mullinax et al., 1992, BioTechniques 12:864; Sawai et al., 1995, AJRI 34:26; and Better et al., 1988, Science 240:1041 (said references incorporated by reference in their entireties). The nucleotide sequence encoding an antibody of the invention can be obtained from sequencing hybridoma clone DNA. If a clone containing a nucleic acid encoding a particular antibody or an epitope-binding fragment thereof is not available, but the sequence of the antibody molecule or epitope-binding fragment thereof is known, a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA library, or a cDNA library generated from, or nucleic acid, preferably poly A+ RNA, isolated from any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody) by PCR amplification using synthetic primers that hybridize to the 3' and 5 'ends of the sequence or by cloning using an ohgonucleotide probe specific for the particular gene sequence to identify, e.g., a cDNA clone from a cDNA library that encodes the antibody. Amplified nucleic acids generated by PCR may then be cloned into replicable cloning vectors using any method well known in the art. Once the nucleotide sequence of the antibody is determined, the nucleotide sequence of the antibody may be manipulated using methods well known in the art for the manipulation of nucleotide sequences, e.g. recombinant DNA techniques, site directed mutagenesis, PCR, etc. (see, Or example, the techniques described in Current Protocols in Molecular Biology, F.M. Ausubel et al., ed., John Wiley & Sons (Chichester, England, 1998); Molecular Cloning: A Laboratory Manual, 3nd Edition, J. Sambrook et al., ed., Cold Spring Harbor Laboratory Press (Cold Spring Harbor, NY, 2001); Antibodies: A Laboratory Manual, E. Harlow and D. Lane, ed., Cold Spring Harbor Laboratory Press (Cold Spring Harbor, NY, 1988); and Using Antibodies: A Laboratory Manual, E. Harlow and D. Lane, ed., Cold Spring Harbor Laboratory (Cold Spring Harbor, NY, 1999) which are incorporated by reference herein in their entireties), to generate antibodies having a different amino acid sequence by, for example, introducing deletions, and/or insertions into desired regions of the antibodies. hi a more preferred embodiment, the NH and NL nucleotide sequences are cloned and used to generate whole antibodies. Utilizing cloning techniques known to those skilled in the art, the PCR primers including NH or N nucleotide sequences, a restriction site, and a flanking sequence to protect the restriction site are used to amplify the NH or NL sequences in scFv. The PCR amplified NH domains are cloned into vectors expressing a NH constant region, e.g., the human gamma 4 constant region, and the PCR amplified NL domains are cloned into vectors expressing a N constant region, e.g., human kappa or lambda constant regions. The NH and NL domains may also be cloned into one vector expressing the necessary constant regions. The heavy chain conversion vectors and light chain conversion vectors are then co-transfected into cell lines to generate stable or transient cell lines that express full-length antibodies, e.g., IgG, using techniques known to those of skill in the art. It is specifically contemplated that for some uses, including in vivo use of antibodies in humans and in vitro detection assays, it may be preferable to use human, chimeric or humanized antibodies. Completely human antibodies are particularly desirable for therapeutic treatment of human subjects. Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See also U.S. Patent Νos. 4,444,887 and 4,716,111; and International Publication Νos. WO 98/46645, WO 98/50433, WO 98/24893, WO 98/16654, WO 96/34096, WO 96/33735, and WO 91/10741; each of which is incorporated herein by reference in its entirety. Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes. For example, the human heavy and light chain immunoglobulin gene complexes may be introduced randomly or by homologous recombination into mouse embryonic stem cells. Alternatively, the human variable region, constant region, and diversity region may be introduced into mouse embryonic stem cells in addition to the human heavy and light chain genes. The mouse heavy and light chain immunoglobulin genes may be rendered non-functional separately or simultaneously with the introduction of human immunoglobulin loci by homologous recombination. In particular, homozygous deletion of the JH region prevents endogenous antibody production. The modified embryonic stem cells are expanded and microinjected into blastocysts to produce chimeric mice. The chimeric mice are then bred to produce homozygous offspring which express human antibodies. The transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a polypeptide of the invention. Monoclonal antibodies directed against the antigen can be obtained from the immunized, transgenic mice using conventional hybridoma technology. The human immunoglobulin transgenes harbored by the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation. Thus, using such a technique, it is possible to produce therapeutically useful IgG, IgA, IgM and IgE antibodies. For an overview of this technology for producing human antibodies, see Lonberg and Huszar (1995, Int. Rev. Immunol.. 13:65-93). For a detailed discussion of this technology for producing human antibodies and human monoclonal antibodies and protocols for producing such antibodies, see, e.g., International Publication Nos. WO 98/24893, WO 96/34096, and WO 96/33735; and U.S. Patent Nos. 5,413,923, 5,625,126, 5,633,425, 5,569,825, 5,661,016, 5,545,806, 5,814,318, and 5,939,598, which are incorporated by reference herein in their entirety. In addition, companies such as Abgenix, Inc. (Freemont, CA) and Medarex (Princeton, NJ) can be engaged to provide human antibodies directed against a selected antigen using technology similar to that described above. A chimeric antibody is a molecule in which different portions of the antibody are derived from different immunoglobulin molecules such as antibodies having a variable region derived from a non-human antibody and a human immunoglobulin constant region. Methods for producing chimeric antibodies are known in the art. See for example, Morrison, 1985, Science 229:1202; Oi et al., 1986, BioTechniques 4:214; Gillies et al., 1989, J. Immunol. Methods 125:191-202; and U.S. Patent Nos. 5,807,715, 4,816,567, and 4,816,397, CDR- grafting (EP 239,400; International Publication No. WO 91/09967; and U.S. Patent Nos. 5,225,539, 5,530,101, and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; Padlan, 1991, Molecular Immunology 28(4/5):489-498; Studnicka et al., 1994, Protein Engineering 7:805; and Roguska et al., 1994, PNAS 91:969), and chain shuffling (U.S. Patent No. 5,565,332). which are incorporated herein by reference in their entirety. Humanized are antibodies comprising one or more CDRs from a non-human species and framework regions from a human immunoglobulin molecule can be produced using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; International Publication No. WO 91/09967; and U.S. Patent Nos. 5,225,539, 5,530,101, and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; Padlan, 1991, Molecular Immunology 28(4/5):489-498; Srudnicka et al., 1994, Protein Engineering 7:805; and Roguska et al., 1994, PNAS 91 :969), and chain shuffling (U.S. Patent No. 5,565,332). A humanized antibody is an antibody or its variant or fragment thereof which is capable of binding to a predetermined antigen and which comprises a framework region having substantially the amino acid sequence of a human immunoglobulin and a CDR having substantially the amino acid sequence of a non-human immunoglobulin. A humanized antibody comprises substantially all of at least one, and typically two, variable domains in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin (i.e., donor antibody) and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence. Preferably, a humanized antibody also comprises at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. Ordinarily, the antibody will contain both the light chain as well as at least the variable domain of a heavy chain. The antibody also may include the CHI, hinge, CH2, CH3, and CH4 regions of the heavy chain. The humanized antibody can be selected from any class of immunoglobulins, including IgM, IgG, IgD, IgA and IgE, and any isotype, including IgGl5 IgG2, IgG3 and IgG4. Usually the constant domain is a complement fixing constant domain where it is desired that the humanized antibody exhibit cytotoxic activity, and the class is typically IgG Where such cytotoxic activity is not desirable, the constant domain may be of the IgG2 class. The humanized antibody may comprise sequences from more than one class or isotype, and selecting particular constant domains to optimize desired effector functions is within the ordinary skill in the art. The framework and CDR regions of a humanized antibody need not correspond precisely to the parental sequences, e.g., the donor CDR or the consensus framework maybe mutagenized by substitution, insertion or deletion of at least one residue so that the CDR or framework residue at that site does not correspond to either the consensus or the import antibody. Such mutations, however, will not be extensive. Usually, at least 75% of the humanized antibody residues will correspond to those of the parental framework region (FR) and CDR sequences, more often 90%, and most preferably greater than 95%. Humanized antibodies can be produced using variety of techniques known in the art, including but not limited to, CDR-grafting (European Patent No. EP 239,400; International Publication No. WO 91/09967; and U.S. Patent Nos. 5,225,539, 5,530,101, and 5,585,089), veneering or resurfacing (European Patent Nos. EP 592,106 and EP 519,596; Padlan, 1991, Molecular Immunology 28(4/5):489-498; Studnicka et al, 1994, Protein Engineering 7(6):805-814; and Roguska et al., 1994, PNAS 91:969-973), chain shuffling (U.S. Patent No. 5,565,332), and techniques disclosed in, e.g., U.S. Patent Nos. 6,407,213, 5,766,886, 5,585,089, International Publication No. WO 9317105, Tan et al., 2002, J. Immunol. 169:1119-25, Caldas et al., 2000, Protein Eng. 13:353-60, Morea et al, 2000, Methods 20:267-79, Baca et al., 1997, J. Biol. Chem. 272:10678-84, Roguska et al., 1996, Protein Eng. 9:895-904, Couto et al., 1995, Cancer Res. 55 (23 Supp):5973s-5977s, Couto et al., 1995, Cancer Res. 55:1717-22, Sandhu, 1994, Gene 150:409-10, Pedersen et al., 1994, j. Mol. Biol. 235:959-73, Jones et al., 1986, Nature 321:522-525, Riechmann et al., 1988, Nature 332:323, and Presta, 1992, Curr. Op. Struct. Biol. 2:593-596. Often, framework residues in the framework regions will be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding. These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Patent No. 5,585,089; and Riechmann et al., 1988, Nature 332:323, which are incorporated herein by reference in their entireties.) Further, the antibodies of the invention can, in turn, be utilized to generate anti- idiotype antibodies using techniques well known to those skilled in the art. (See, e.g., Greenspan & Bona, 1989, FASEB J. 7:437-444; and Nissinoff, 1991, J. Immunol. 147:2429- 2438). The invention provides methods employing the use of polynucleotides comprising a nucleotide sequence encoding an antibody of the invention or a fragment thereof.
Methods of Expressing Antibodies The antibodies of the invention can be produced by any method known in the art for the synthesis of antibodies, in particular, by chemical synthesis or preferably, by recombinant expression techniques. Recombinant expression of a HAAH specific antibody of the invention, or fragment, derivative or analog thereof, e.g., a heavy or light chain of an antibody of the invention, requires construction of an expression vector containing a polynucleotide that encodes the antibody. Once a polynucleotide encoding an antibody molecule or a heavy or light chain of an antibody, or portion thereof (preferably containing the heavy or light chain variable domain), of the invention has been obtained, the vector for the production of the antibody molecule may be produced by recombinant DNA technology using techniques well known in the art. Thus, methods for preparing a protein by expressing a polynucleotide containing an antibody encoding nucleotide sequence are described herein. Methods which are well known to those skilled in the art can be used to construct expression vectors containing antibody coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. The invention, thus, provides replicable vectors comprising a nucleotide sequence encoding an antibody molecule of the invention, or a heavy or light chain thereof, or a heavy or light chain variable domain, operably linked to a promoter. Such vectors may include the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., PCT Publication WO 86/05807; PCT Publication WO 89/01036; and U.S. Pat. No. 5,122,464) and the variable domain of the antibody may be cloned into such a vector for expression of the entire heavy or light chain. The expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce a HAAH specific antibody of the invention. Thus, the invention includes host cells containing a polynucleotide encoding an antibody of the invention, or a heavy or light chain thereof, operably linked to a heterologous promoter. In preferred embodiments for the expression of double-chained antibodies, vectors encoding both the heavy and light chains may be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below. The antibody expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce a substituted antibody have improved producibility. A variety of host-expression vector systems may be utilized to express the antibody molecules of the invention. Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences express an antibody molecule of the invention in situ. Thus, the invention includes host cells containing a polynucleotide encoding an antibody of the invention, or a heavy or light chain thereof, operably linked to a heterologous promoter. In preferred embodiments for the expression of double-chained antibodies, vectors encoding both the heavy and light chains may be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below. A variety of host-expression vector systems maybe utilized to express the antibody molecules of the invention. Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express an antibody molecule of the invention in situ. These include but are not limited to microorganisms such as bacteria (e.g., E. coli, B. subtilis ) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMN; tobacco mosaic virus, TMN) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter). Preferably, bacterial cells such as Escherichia coli , and more preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule. For example, mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., 1986, Gene 45:101; Cockett et al., 1990, Bio/Technology 8:2). In bacterial systems, a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed. For example, when a large quantity of such a protein is to be produced, for the generation of pharmaceutical compositions of an antibody molecule, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable. Such vectors include, but are not limited, to the E. coli expression vector pUR278 (Ruther et al., 1983, EMBO J . 2: 1791), in which the antibody coding sequence may be ligated individually into the vector in frame with the lacZ coding region so that a fusion protein is produced; pTΝ vectors (Inouye & friouye, 1985, Nucleic Acids Res. 13:3101-9; Nan Heeke & Schuster, 1989, J. Biol. Chem. 24:5503-9); and the like. pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to a matrix glutathione-agarose beads followed by elution in the presence of free glutathione. The pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety. In an insect system, Autographa californica nuclear polyhedrosis virus (AcΝPN) is used as a vector to express foreign genes. The virus grows in Spodoptera frugiperda cells. The antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcΝPN promoter (for example the polyhedrin promoter). In mammalian host cells, a number of viral-based expression systems maybe utilized. In cases where an adenovirus is used as an expression vector, the antibody coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non- essential region of the viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts, (e.g., see Logan & Shenk, 1984, Proc. Natl. Acad. Sci. USA 81:355-9). Specific initiation signals may also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bitter et al., 1987, Methods in Enzymol., 153:516-44). In addition, a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g. glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed. To this end, eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of-the gene product may be used. Such mammalian host cells include but are not limited to CHO, VERY, BHK, HeLa, COS, MDCK, 293, 3T3, W138, and in particular, breast cancer cell lines such as, for example, BT483, Hs578T, HTB2, BT20 and T47D, and normal mammary gland cell line such as, for example, CRL7030 andHs578Bst. For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines which stably express the antibody molecule may be engineered. Rather than using expression vectors which contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. Following the introduction of the foreign DNA, engineered cells maybe allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines which express the antibody molecule. Such engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the antibody molecule. A number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler et al., 1977, Cell 11 :223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, 1992, Proc. Natl. Acad. Sci. USA 48:202), and adenine phosphoribosyltransferase (Lowy et al., 1980, Cell 22:817) genes can be employed in tk-, hgprt- or aprt-cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al., 1980, Proc. Natl. Acad. Sci. USA 77:357; O'Hare et al., 1981, Proc. Natl. Acad. Sci. USA 78:1527); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, 1981, Proc. Natl. Acad. Sci. USA 78:2072); neo, which confers resistance to the aminoglycoside G-418 (Wu and Wu, 1991, Biotherapy 3:87-95; Tolstoshev, 1993, Ann. Rev. Pharmacol. Toxicol. 32:573-96; Mulligan, 1993, Science 260:926-32; and Morgan and Anderson, 1993, Ann. Rev. Biochem. 62:191-217; May, 1993, TIB TECH 11:155-215); and hygro, which confers resistance to hygromycin (Santerre et al, 1984, Gene 30: 147). Methods commonly known in the art of recombinant DNA technology which can be used are described in Current Protocols in Molecular Biology, F.M. Ausubel et al., ed., John Wiley & Sons (Chichester, England, 1998); Molecular Cloning: A Laboratory Manual, 3nd Edition, J. Sambrook et al., ed., Cold Spring Harbor Laboratory Press (Cold Spring Harbor, NY, 2001), which are incorporated by reference herein in their entireties. The expression levels of an antibody molecule can be increased by vector amplification (for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol. 3. (Academic Press, New York, 1987)). When a marker in the vector system expressing antibody is amplifiable, increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene. Since the amplified region is associated with the antibody gene, production of the antibody will also increase (Crouse et al., 1983, Mol. Cell. Biol. 3:257) The host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide. The two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides. Alternatively, a single vector may be used which encodes both heavy and light chain polypeptides. hi such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, 1986, Nature 322:52; Kohler, 1980, Proc. Natl. Acad. Sci. USA 77:2197). The coding sequences for the heavy and light chains may comprise cDNA or genomic DNA. Once an antibody molecule of the invention has been recombinantly expressed, it may be purified by any method known in the art for purification of an immunoglobulin molecule, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
Polypeptides And Polynucleotides Of The Invention Polypeptides of the invention include, but are not limited to, HAAH polypeptides exemplified by GenBank Ace. No. AAB50779, including full-length polypeptides, fragments, variants or derivatives and epitopes of the full-length HAAH. Specifically contemplated are polypeptides comprising the luminal/extracellular domains ofHAAH. HAAH luminal/extracellular domains include but are not limited to, the Aspartyl beta-hydroxylase N-terminal and Aspartyl/ Asparaginyl beta-hydroxylase C-terminal (residues 43-309 and 589- 746) regions that are critical for catalytic activity as well as a central tetratricopeptide repeat domain (residues 421-520). Polynucleotides of the invention include, but are not limited to, HAAH polynucleotides exemplified by GenBank Ace. No. NM_004318, including full-length polynucleotides, fragments, variants or derivatives and epitopes of the full-length HAAH. Alternative splicing of this gene results in five transcript variants (exemplified by GenBank Ace. Nos. NM_004318, NM_020164, NM_032466, NM B2467, NM_032468) which vary in protein translation, the coding of catalytic domains, and tissue expression. Variation among these transcripts impacts their functions which involve roles in the calcium storage and release process in the endoplasmic and sarcoplasmic reticulum as well as hydroxylation of aspartic acid and asparagine in epidermal growth factor-like domains of various proteins. It is specifically contemplated that polypeptides of the present invention may include full-length polypeptides, fragments, variants or derivatives and epitopes of HAAH splice variants. Polypeptides of the invention further include, but are not limited to, polypeptide antagonists ofHAAH polypeptides. The present invention further relates to polynucleotides which hybridize to the hereinabove-described sequences if there is at least 70%, preferably at least 90%, and more preferably at least 95% identity between the sequences. The present invention particularly relates to polynucleotides which hybridize under stringent conditions to the hereinabove- described polynucleotides.
Fusion Proteins The polypeptide of the present invention are could be fused to other heterologous sequences. These fusion proteins can be used for a variety of applications. For example, fusion of the present polypeptides to His-tag, HA-tag, protein A, IgG domains, and maltose binding protein facilitates purification. (See EP A 394,827; Traunecker, et al., 1988, Nature 331:84-86). Similarly, fusion to IgG-1, IgG-3, and albumin increases the half life time in vivo. Nuclear localization signals fused to the polypeptides of the present invention can target the protein to a specific subcellular localization, while covalent heterodimer or homodimers can increase or decrease the activity of a fusion protein. Fusion proteins can also create chimeric molecules having more than one function. Finally, fusion proteins can increase solubility and/or stability of the fused protein compared to the non-fused protein. All of the types of fusion proteins described above can be made by modifying the following protocol, which outlines the fusion of a polypeptide to an IgG molecule. Briefly, the human Fc portion of the IgG molecule can be PCR amplified. These primers also should have convenient restriction enzyme sites that will facilitate cloning into an expression vector, preferably a mammalian expression vector. For example, if pC4 (Accession No. 209646) is used, the human Fc portion can be ligated into the BamHI cloning site. Note that the 3' BamHI site should be destroyed. Next, the vector containing the human Fc portion is re-restricted with BamHI, linearizing the vector, and a polynucleotide of the present invention, isolated by the PCR, is ligated into this BamHI site. Note that the polynucleotide is cloned without a stop codon, otherwise a fusion protein will not be produced^ If the naturally occurring signal sequence is used to produce the secreted protein, pC4 does not need a second signal peptide. Alternatively, if the naturally occurring signal sequence is not used, the vector can be modified to include a heterologous signal sequence. (See, e.g., WO 96/34891.) As one of skill in the art will appreciate, and as discussed above, the polypeptides of the present invention comprising an immunogenic or antigenic epitope can be fused to other polypeptide sequences. For example, the polypeptides of the present invention maybe fused with the constant domain of inrmunoglobulins (IgA, IgE, IgG, IgM), or portions thereof (CHI, CH2, CH3, or any combination thereof and portions thereof) resulting in chimeric polypeptides. Such fusion proteins may facilitate purification and may increase half-life in vivo. This has been shown for chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian inrmunoglobulins. (See, e.g., EP 394,827; Traunecker et al, 1988, Nature, 331:84-86). Enhanced delivery of an antigen across the epithelial barrier to the immune system has been demonstrated for antigens (e.g., insulin) conjugated to an FcRn binding partner such as IgG or Fc fragments (see, e.g., PCT Publications WO 96/22024 and WO 99/04813). IgG Fusion proteins that have a disulfide-linked dimeric structure due to the IgG portion disulfide bonds have also been found to be more efficient in binding and neutralizing other molecules than monomeric polypeptides or fragments thereof alone. See, e.g., Fountoulakis et al., 1995, J. Biochem., 270:3958-3964. Measuring Mast Cell Modulation The present invention comprises methods for identifying compounds capable of depleting mast cells, wherein said compounds are immunospecifically binds to HAAH on the surface of the mast cell and not other hematopoeitic cells that are not mast cells or related cells or cell lines or derived cell lines thereof, comprising the steps consisting of: a) culturing mast cells in vitro in a suitable culture medium, b) adding to said culture medium at least one candidate compound to be tested and incubating said cells for a prolonged period of time, c) measuring the extent to which said compounds promote mast cells death or disrupt, interfere with, or inhibit mast cells growth, and selecting compounds for which mast cells depletion is observed, d) identifying a compound of a subset of a compound selected in step c) that promote significant death of a mast death or disrupt, interfere with, or inhibit mast cells growth, and selecting compounds for which mast cells depletion is observed. The invention also encompasses to a method for identifying compounds that immunospecifically binds to HAAH and are capable of depleting mast cells, wherein said compounds are non-toxic for other hematopoeitic cells that are not mast cells or related cells or cell lines or derived cell lines thereof, comprising the step consisting of: a) providing a culture of mast cells, wherein said mast cells are selected from wild type mast cells and cell lines derived thereof, activated mutant mast cell lines, and activated wild type mast cells and cell lines derived thereof, b) contacting the culture of said cells with at least one candidate compound under conditions allowing growth and/or survival of mast cells, measuring the level of cell death in the presence of the candidate compound; and comparing the level of cell death in the presence of the candidate compound to the level of cell death in the absence of the candidate compound, wherein an increase in the level of cell death in the presence of the candidate compound is indicative of the mast cells depletion ability of the candidate compound, c) providing a culture of at least one cell other than mast cells, wherein said cell is selected from hematopoeitic cells that are not mast cells or related cells or cell lines or derived cell lines thereof, such as SCF independent expanded human normal CD34+ cells, d) contacting the culture of said cells with at least one compound identified in step b) under conditions allowing growth and/or survival of a cell depicted in step c), measuring the level of cell death in the presence of said compound; and comparing the level of cell death in the presence of the compound to the level of cell death in the absence of the compound, wherein no significant increase in the level of cell death in the presence of said compound is indicative of mast cells depletion specificity of said compound versus at least another hematopoeitic cell. Method of modulating a mast cell includes antibody mediated cellular cytotoxicity (ADCC), apoptosis, agonizing or antagonizing HAAH to induce cells death, or recruiting a T cell as in a BiTE molecule (see supra). The cell death assay can further comprise a cell proliferation assay, a cell viability assay and/or an apoptosis assay. For example, the extent of cell death can be measured by 3H thymidine incorporation, the trypan blue exclusion method, using propidium iodide or by the 5"Cr-release assay. Alternatively, the extent of cell death can be determined by a test of intracellular esterase activity, and a test of plasma membrane integrity, preferably using fluorescent calcein and ethidium homodimer-1. These tests are described in J. Neurosci 15, 5389 (1995), in J. Cell Sci. 106, 685 (1993). Detailed protocols are given in the Molecular Probes Catalogue product number L-3224 (Live/Dead® Kits) incorporated herein by reference. Basically, calcein AM is the cell-permeant esterase substrate, which is nonfluorescent until converted by enzymatic activity to highly fluorescent calcein. It remains within living cells exhibiting an intense green fluorescence. Ethidium homodimer-1 fluorescence is enhanced upon binding nucleic acids. A bright red fluorescence is emitted. This dye cannot cross intact plasma membranes but it enters into dead cells. Thus, living cells are green, while dead cells emits a red fluorescence. This technique coupled with CDD camera and plate readers leads to high through put screening. In another embodiment, the extent of cell death is determined by discriminating between living and dead cells using DiOC18 and propidium iodide. Protocols are described in details in the Molecular Probes Catalogue product number L-7010 (Live/Dead® Kits) incorporated herein by reference. In still another embodiment, cell death can be determined using the Caspase activity test. Caspase is a key player in the activation of apoptosis. The Molecular probe kit E-13183 (EnzCheck Caspase-3 Assay kit®, Molecular Probe) is particularly useful for testing Jurkat cells. Phosphatidyl exposure can also be used in this regard. This method has been employed in Dan S, et al, 1998, Cell Death Differ. 5:710-5. In still another embodiment, cell death can be determined using the Mitochondrial membrane depolarization test using the JC-1 or JC-9 cationic dyes of Molecular Probe, which have been described as a useful indicator in HL-60 cells. For cell proliferation assays, it can be performed using MTS tetrazolium (Cell Titer96 Aqueous; Promega, Madison, Wis.). This test allows measuring the numbers of viable cells. In all the above-mentioned cell death tests, the invention encompasses fluorometric assays of cell viability and cytotoxicity using a fluorescence microscope, a fluorometer, a fluorescence microplate reader and/or a flow cytometer. The above methods are not meant to be an exhaustive list of all mast cell proliferation/death assays, but just an example assays that be utilized. The present invention comprises methods for identifying compounds capable of preventing mast cell activation, wherein said compounds are immunospecifically binds to HAAH on the surface of the mast cell and not other hematopoeitic cells that are not mast cells or related cells or cell lines or derived cell lines thereof, comprising the steps consisting of: a) culturing mast cells in vitro in a suitable culture medium, b) adding to said culture medium at least one candidate compound to be tested and incubating said cells for a prolonged period of time, c) adding to the culture medium at least one compound known to promote mast cell activation, d) measuring the extent to which said compounds prevent mast cell activation and selecting compounds for which inhibition of mast cell activation is observed, e) identifying a compound of a subset of a compound selected in step d) that prevent significant activation of a mast, and selecting compounds for which inhibition of mast cells activation is observed. Mast cell activation can be monitored by examining the amount of degranulation. Degranulation leads to the release of preformed mediators of inflammation including, histamine, JL-3, IL-5 and/or TNF. Thus, one can monitor the levels of one or more of these mediators released from a treated mast cell and compare it to a control treated mast cell.
Combination Treatment For Respiratory Conditions The present invention provides methods for preventing, managing, treating, or ameliorating respiratory conditions comprising administering to a subject in need thereof one or more HAAH antagonists alone or in combination with one or more therapies (e.g., one or more prophylactic or therapeutic agents) other than an HAAH antagonist. The present invention also provides compositions comprising one or more HAAH antagonists and one or more prophylactic or therapeutic agents other than HAAH antagonists and methods of preventing, managing, treating, or ameliorating respiratory conditions or one or more symptoms thereof utilizing said compositions. Therapeutic or prophylactic agents include, but are not limited to, small molecules, synthetic drugs, peptides, polypeptides, proteins, nucleic acids (e.g., DNA and RNA nucleotides including, but not limited to, antisense nucleotide sequences, triple helices, RNAi, and nucleotide sequences encoding biologically active proteins, polypeptides or peptides) antibodies, synthetic or natural inorganic molecules, mimetic agents, and synthetic or natural organic molecules. Any therapy which is known to be useful, or which has been used or is currently being used for the prevention, management, treatment, or amelioration of respiratory conditions or one or more symptoms thereof can be used in combination with an HAAH antagonist in accordance with the invention described herein. See, e.g., Gilman et al., Goodman and Gilman's: The Pharmacological Basis of Therapeutics, 10th ed., McGraw- Hill, New York, 2001; The Merck Manual of Diagnosis and Therapy, Berkow, M.D. et al. (eds.), 17th Ed., Merck Sharp & Dohme Research Laboratories, Rahway, NJ, 1999; Cecil Textbook of Medicine, 20th Ed., Bennett and Plum (eds.), W.B. Saunders, Philadelphia, 1996 for information regarding therapies (e.g., prophylactic or therapeutic agents) which have been or are currently being used for preventing, treating, managing, or ameliorating respiratory conditions or one or more symptoms thereof. Examples of such agents include, but are not limited to, immunomodulatory agents, anti-inflammatory agents (e.g., adrenocorticoids, corticosteroids (e.g., beclomethasone, budesonide, flunisolide, fluticasone, triamcinolone, methlyprednisolone, prednisolone, prednisone, hydrocortisone), glucocorticoids, steroids, non-steriodal anti-inflammatory drugs (e.g., aspirin, ibuprofen, diclofenac, and COX-2 inhibitors), leukotreine antagonists (e.g., montelukast, methyl xanthines, zafirlukast, and zileuton), beta2-agonists (e.g., albuterol, biterol, fenoterol, isoetharie, metaproterenol, pirbuterol, salbutamol, terbutalin formoterol, salmeterol, and salbutamol terbutaline), anticholinergic agents (e.g., ipratropium bromide and oxitropium bromide), sulphasalazine, penicillamine, dapsone, antihistamines, anti-malarial agents (e.g., hydroxychloroqume), antiviral agents, and antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, erythomycin, penicillin, mithramycin, and anthramycin (AMC)).
Immunomodulatory Agents Any immunomodulatory agent well-known to one of skill in the art may be used in the methods and compositions of the invention. Immunomodulatory agents can affect one or more or all aspects of the immune response in a subject. Aspects of the immune response include, but are not limited to, the inflammatory response, the complement cascade, leukocyte and lymphocyte differentiation, proliferation, and/or effector function, monocyte and/or basophil counts, and the cellular communication among cells of the immune system. In certain embodiments of the invention, an immunomodulatory agent modulates one aspect of the immune response. In other embodiments, an immunomodulatory agent modulates more than one aspect of the immune response, hi a preferred embodiment of the invention, the administration of an immunomodulatory agent to a subject inhibits or reduces one or more aspects of the subject's immune response capabilities. In an alternative embodiment of the invention, the immunomodulatory agent enhances one or more aspects of a subject's immune response. In accordance with the invention, an immunomodulatory agent is not an LL-9 antagonist. In certain embodiments, an immunomodulatory agent is not an anti- inflammatory agent. In other embodiments, an immunomodulatory agent is a chemotherapeutic agent. In yet other embodiments, an immunomodulatory agent an agent other than a chemotherapeutic agent. Examples of immunomodulatory agents include, but are not limited to, proteinaceous agents such as cytokines, peptide mimetics, and antibodies (e.g., human, humanized, chimeric, monoclonal, polyclonal, Fvs, ScFvs, Fab or F(ab)2 fragments or epitope binding fragments), nucleic acid molecules (e.g., antisense nucleic acid molecules and triple helices), small molecules, organic compounds, and inorganic compounds. In particular, immunomodulatory agents include, but are not limited to, methotrexate, leflunomide, cyclophosphamide, cytoxan, Immuran, cyclosporine A, minocycline, azathioprine, antibiotics (e.g., FK506 (tacrolimus)), methylprednisolone (MP), corticosteroids, steroids, mycophenolate mofetil, rapamycin (sirolimus), mizoribine, deoxyspergualin, brequinar, malononitriloamindes (e.g., leflunamide), T cell receptor modulators, cytokine receptor modulators, and modulators mast cell modulators. Examples of T cell receptor modulators include, but are not limited to, anti-T cell receptor antibodies (e.g., anti-CD4 antibodies (e.g., cM-T412 (Boeringer), IDEC-CE9.1® (IDEC and SKB), mAB 4162W94, Orthoclone and OKTcdr4a (Janssen-Cilag)), anti-CD3 antibodies (e.g., Nuvion (Product Design Labs), OKT3 (Johnson & Johnson), or Rituxan (IDEC)), anti-CD5 antibodies (e.g., an anti-CD5 ricin-linked immunoconjugate), anti-CD7 antibodies (e.g., CHH-380 (Novartis)), anti-CD8 antibodies, anti-CD40 ligand monoclonal antibodies (e.g., HOEC-131 (IDEC)), anti-CD52 antibodies (e.g., CAMPATH IH (Ilex)), anti- CD2 antibodies (e.g., MEDI-507 (Medlmmune, hie, International Publication Nos. WO 02/098370 and WO 02/069904), anti-CDl la antibodies (e.g., Xanelim (Genentech)), and anti-B7 antibodies (e.g., TDEC-114 (IDEC)), CTLA4-immunoglobulin, and LFA-3TTP (Biogen, International Publication No. WO 93/08656 and U.S. Patent No. 6,162,432). Examples of cytokine receptor modulators include, but are not limited to, soluble cytokine receptors (e.g., the extracellular domain of a TNF-α receptor or a fragment thereof, the extracellular domain of an LL-lβ receptor or a fragment thereof, and the extracellular domain of an LL-6 receptor or a fragment thereof), cytokines or fragments thereof (e.g., interleukin LL-2, IL-3, LL-4, IL-5, IL-6, LL-7, LL-8, IL-9, IL-10, LL-11, IL-12, IL-13, IL-15, IL-23, TNF-α, TNF-β, interferon (IFN)-α, IFN-β, IFN-γ, and GM-CSF), anti-cytokine receptor antibodies (e.g., anti-IFN receptor antibodies, anti-LL-2 receptor antibodies (e.g., Zenapax (Protein Design Labs)), anti-IL-3 receptor antibodies, anti-JL-4 receptor antibodies, anti-LL-6 receptor antibodies, anti-JL-lO receptor antibodies, anti-IL-12 receptor antibodies, anti-LL-13 receptor antibodies, anti-LL-15 receptor antibodies, and anti-LL-23 receptor antibodies), anti-cytokine antibodies (e.g., anti-IFN antibodies, anti-TNF-α antibodies, anti- IL-lβ antibodies, anti-IL-3 antibodies, anti-IL-6 antibodies, anti-IL-8 antibodies (e.g., ABX- IL-8 (Abgenix)), anti-IL-12 antibodies, anti-IL-13 antibodies, anti-IL-15 antibodies, and anti- IL-23 antibodies). In a specific embodiment, a cytokine receptor modulator is LL-3, LL-4, LL-10, or a fragment thereof. In another embodiment, a cytokine receptor modulator is an anti-LL-l β antibody, anti-IL-6 antibody, anti-IL-12 receptor antibody, or anti-TNF-α antibody. In another embodiment, a cytokine receptor modulator is the extracellular domain of a TNF-α receptor or a fragment thereof. In certain embodiments, a cytokine receptor modulator is not a TNF-α antagonist. In one embodiment, a cytokine receptor modulator is a mast cell modulator. In an alternative embodiment, a cytokine receptor modulator is not a mast cell modulator. Examples of mast cell modulators include, but are not limited to stem cell factor (c-kit receptor ligand) inhibitor (e.g., mAb 7H6, mAb 8H7a, pAb 1337, FK506, CsA, dexamthasone, and fluconcinonide), c-kit receptor inhibitor (e.g., STI 571 (formerly known as CGP 57148B)), mast cell protease inhibitor (e.g., GW-45, GW-58, wortmannin, LY 294002, calphostin C, cytochalasin D, genistein, KT5926, staurosproine, and lactoferrin), relaxin ("RLX"), IgE antagonist (e.g., antibodies rhuMAb-E25 omalizumab, HMK-12 and 6HD5, and mAB Hu-901), LL-3 antagonist, LL-4 antagonists, LL-10 antagonists, and TGF- beta. An immunomodulatory agent may be selected to interfere with the interactions between the T helper subsets (TH1 or TH2) and B cells to inhibit neutralizing antibody formation. Antibodies that interfere with or block the interactions necessary for the activation of B cells by TH (T helper) cells, and thus block the production of neutralizing antibodies, are useful as immunomodulatory agents in the methods of the invention. For example, B cell activation by T cells requires certain interactions to occur (Durie et al., 1994, Immunol. Today, 15:406-10), such as the binding of CD40 ligand on the T helper cell to the CD40 antigen on the B cell, and the binding of the CD28 and/or CTLA4 ligands on the T cell to the B7 antigen on the B cell. Without both interactions, the B cell cannot be activated to induce production of the neutralizing antibody. The CD40 ligand (CD40L)-CD40 interaction is a desirable point to block the immune response because of its broad activity in both T helper cell activation and function as well as the absence of redundancy in its signaling pathway. Thus, in a specific embodiment of the invention, the interaction of CD40L with CD40 is transiently blocked at the time of administration of one or more of the immunomodulatory agents. This can be accomplished by treating with an agent which blocks the CD40 ligand on the TH cell and interferes with the normal binding of CD40 ligand on the T helper cell with the CD40 antigen on the B cell. An antibody to CD40 ligand (anti-CD40L) (available from Bristol-Myers Squibb Co; see, e.g., European patent application 555,880) or a soluble CD40 molecule can be selected and used as an immunomodulatory agent in accordance with the methods of the invention. An immunomodulatory agent may be selected to inhibit the interaction between TH1 cells and cytotoxic T lymphocytes ("CTLs") to reduce the occurrence of CTL-mediated killing. An immunomodulatory agent may be selected to alter (e.g., inhibit or suppress) the proliferation, differentiation, activity and/or function of the CD4+ and/or CD8+ T cells. For example, antibodies specific for T cells can be used as immunomodulatory agents to deplete, or alter the proliferation, differentiation, activity and/or function of CD4+ and/or CD8+ T cells. In one embodiment of the invention, an immunomodulatory agent that reduces or depletes T cells, preferably memory T cells, is administered to a subject with respiratory conditions in accordance with the methods of the invention. See, e.g., U.S. Pat. No. 4,658,019. In another embodiment of the invention, an immunomodulatory agent that inactivates CD8+ T cells is administered to a subject with respiratory conditions in accordance with the methods of the invention. In a specific embodiment, anti-CD8 antibodies are used to reduce or deplete CD8+ T cells. In another embodiment, an immunomodulatory agent which reduces or inhibits one or more biological activities (e.g., the differentiation, proliferation, and/or effector functions) of TH0, TH1, and/or TH2 subsets of CD4+ T helper cells is administered to a subject with respiratory conditions in accordance with the methods of the invention. One example of such an immunomodulatory agent is LL-4. LL-4 enhances antigen-specific activity of TH2 cells at the expense of the TH1 cell function (see, e.g., Yokota et al, 1986 Proc. Natl. Acad. Sci., USA, 83:5894-5898; and U.S. Pat. No. 5,017,691). Other examples of immunomodulatory agents that affect the biological activity (e.g., proliferation, differentiation, and/or effector functions) of T-helper cells (in particular, TH1 and/or TH2 cells) include, but are not limited to, LL-2, LL-4, LL-5, LL-6, LL-10, IL-12, LL-13, LL-15, JL-23, and interferon (FFN)-γ. In another embodiment, an immunomodulatory agent administered to a subject with respiratory conditions in accordance with the methods of the invention is a cytokine that prevents antigen presentation. In a specific embodiment, an immunomodulatory agent used in the methods of the invention is LL-10. EL- 10 also reduces or inhibits macrophage action, which involves bacterial elimination. An immunomodulatory agent may be selected to reduce or inhibit the activation, degranulation, proliferation, and/or infiltration of mast cells, hi certain embodiments, the immunomodulatory agent interferes with the interactions between mast cells and mast cell activating agents, including, but not limited to stem cell factors (c-kit ligands), IgE, IL-4, environmental irritants, and infectious agents. In a specific embodiment, the immunomodulatory agent reduces or inhibits the response of mast cells to environmental irritants such as, but not limited to pollen, dust mites, tobacco smoke, and/or pet dander. In another specific embodiment, the immunomodulatory agent reduces or inhibits the response of mast cells to infectious agents, such as viruses, bacteria, and fungi. Examples of mast cell modulators that reduce or inhibit the activation, degranulation, proliferation, and/or infiltration of mast cells include, but are not limited to, stem cell factor (c-kit receptor ligand) inhibitors (e.g., mAb 7H6, mAb 8H7a, and pAb 1337 (see Mendiaz et al., 1996, Eur J Biochem 293:842-49), FK506 and CsA (Ito et al., 1999 Arch DermatolRes 291:275-83), dexamthasone and fluconcinonide (see Finooto et al. J Clin Invest 1997 99:1721-8), c-kit receptor inhibitors (e.g., STI 571 (formerly known as CGP 57148B) (see Heinrich et al., 2000, Blood 96:925-32)), mast cell protease inhibitors (e.g., GW-45 and GW-58 (see Temkin et al., 2002, J Immunol 169:2662-9), wortmannin, LY 294002, calphostin C, and cytochalasin D (see Nosseller et al., 1997, Mol Biol Cell 8:909-22), genistein, KT5926, and staurosproine (see Νagai et al., 1995, Biochem Biophys Res Commun 208:576-81), and lactoferrin (see He et al., 2003 Biochem Pharmacol 65:1007-15)), relaxin ("RLX") (see Bani et al, 2002, bit Immunopharmacol 2:1195-1294), ), IgE antagonists (e.g., antibodies rhuMAb-E25 omalizumab (see Finn et al., 2003, J Allergy Clin Immuno 111 :278-84; Corren et al, 2003, J Allergy Clin Immuno 111 : 87-90; B sse and Νeaville, 2001 , Curr Opin Allergy Clin Immuno 1:105-8; and Tang and Powell, 2001, EurJPediatr 160:696-704), HMK-12 and 6HD5 (see Miyajima et al, 2002, bit Arch Allergy Immuno 128:24-32), and mAB Hu-901 (see van Νeerven et al, 2001, bit Arch Allergy Immuno 124:400), LL-3 antagonist, LL-4 antagonists, LL-10 antagonists, and TGF-beta (see Metcalfe et al, 1995, Exp Dermatol 4:227-230). In a preferred embodiment, LL-9 antagonists that can be utilized in combination with the invention, but are not limited to, proteinaceous agents (e.g., proteins, polypeptides, peptides, fusion proteins, antibodies, and antibody fragments), nucleic acid molecules (e.g., DL-9 antisense nucleic acid molecules, triple helices, RNAi, or nucleic acid molecules encoding proteinaceous agents), organic molecules, inorganic molecules, small organic molecules, drugs, and small inorganic molecules that block, inhibit, reduce or neutralize a pathologic cellular or humoral phenotype associated with or resulting from LL-9 expression and/or activity (e.g., decreases the secretion of mucin, the differentiation of LL-9 expressing cells into a mucin-secreting cell, the secretion of inflammatory agents, the proliferation, migration, and increase in volume of cells (e.g., immune and smooth muscle cells), the secretion of extracellular matrix molecules or matrix metalloproteinases and/or the binding of LL-9 to the LL-9 receptor ("IL-9R")). In a specific embodiment, an LL-9 antagonist is an antibody or fragment thereof that immunospecifically binds to an LL-9 polypeptide. In another specific embodiment, an LL-9 antagonist is an antibody or fragment thereof that immunospecifically binds to an LL-9R or a subunit thereof. See, e.g. co-pending application 60/462,307, incorporated in its entirety by reference. In a preferred embodiment, proteins, polypeptides or peptides (including antibodies) that are utilized as immunomodulatory agents are derived from the same species as the recipient of the proteins, polypeptides or peptides so as to reduce the likelihood of an immune response to those proteins, polypeptides or peptides. In another preferred embodiment, when the subject is a human, the proteins, polypeptides, or peptides that are utilized as immunomodulatory agents are human or humanized. In a preferred embodiment, one or more immunomodulatory agents are administered in combination with an HAAH antagonist to a subject with respiratory conditions so as to transiently reduce or inhibit one or more aspects of the immune response. Such a transient inhibition or reduction of one or more aspects of the immune system can last for hours, days, weeks, or months. Preferably, the transient inhibition or reduction in one or more aspects of the immune response lasts for a few hours (e.g., 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, 14 hours, 16 hours, 18 hours, 24 hours, 36 hours, or 48 hours), a few days (e.g., 3 days, 4 days, 5 days, 6 days, 7 days, or 14 days), or a few weeks (e.g., 3 weeks, 4 weeks, 5 weeks or 6 weeks). The transient reduction or inhibition of one or more aspects of the immune response enhances the prophylactic and/or therapeutic effect(s) of an HAAH antagonist. Nucleic acid molecules encoding proteins, polypeptides, or peptides with immunomodulatory activity or proteins, polypeptides, or peptides with immunomodulatory activity can be administered to a subject with respiratory conditions in accordance with the methods of the invention. Further, nucleic acid molecules encoding derivatives, analogs, or fragments of proteins, polypeptides, or peptides with immunomodulatory activity, or derivatives, analogs, or fragments of proteins, polypeptides, or peptides with immunomodulatory activity can be administered to a subject with a respiratory infection in accordance with the methods of the invention. Preferably, such derivatives, analogs, and fragments retain the immunomodulatory activity of the full-length, wild type protein, polypeptide, or peptide. Preferably, agents that are commercially available and known to function as immunomodulatory agents are used in the methods of the invention. The immunomodulatory activity of an agent can be determined in vitro and/or in vivo by any technique well known to one skilled in the art, including, e.g., by CTL assays, proliferation assays, and immunoassays (e.g. ELISAs) for the expression of particular proteins such as co-stimulatory molecules and cytokines.
Anti-Inflammatory Agents Any anti-inflammatory agent, including agents useful in therapies for inflammatory disorders, well known to one of skill in the art can be used in the compositions and methods of the invention. Non-limiting examples of anti-inflammatory agents include non-steroidal anti-inflammatory drugs (NSALDs), steroidal anti-inflammatory drugs, anticholinergics (e.g., atropine sulfate, atropine methylnitrate, and ipratropium bromide (ATROVENT™)), beta2- agonists (e.g., abuterol (VENTOLIN™ and PROVENTIL™), bitolterol (TORNALATE™), levalbuterol (XOPONEX™), metaproterenol (ALUPENT™), pirbuterol (MAXAIR™), terbutlaine (BRETHAIRE™ and BRETHINE™), albuterol (PROVENTIL™, REPETABS™, and VOLMAX™), formoterol (FORADIL AEROLIZER™), and salmeterol (SEREVENT™ and SEREVENT DISKUS™)), and methylxanthines (e.g., theophylline (UNIPHYL™, THEO-DUR™, SLO-BID™, AND TEHO-42™)). Examples of NSALDs include, but are not limited to, aspirin, ibuprofen, celecoxib (CELEBREX™), diclofenac (VOLTAREN™), etodolac (LODLNE™), fenoprofen (NALFON™), indomethacin (INDOCIN™), ketoralac (TORADOL™), oxaprozin (DAYPRO™), nabumentone (RELAFEN™), sulindac (CLLNORIL™), tolmentin (TOLECTIN™), rofecoxib (NIOXX™), naproxen (ALEVE™, ΝAPROSYΝ™), ketoprofen (ACTROΝ™) and nabumetone (RELAFEΝ™). Such ΝSAIDs function by inhibiting a cyclooxgenase enzyme (e.g., COX-1 and/or COX-2). Examples of steroidal anti-inflammatory drugs include, but are not limited to, glucocorticoids, dexamethasone (DECADRON™), corticosteroids (e.g., methylprednisolone (MEDROL™)), cortisone, hydrocortisone, prednisone (PREDNISONE™ and DELTASONE™), prednisolone (PRELONE™ and PEDIAPRED™), triamcinolone, azulfidine, and inhibitors of eicosanoids (e.g., prostaglandins, thromboxanes, and leukotrienes (see Table 2, infra, for non-limiting examples of leukotriene and typical dosgages of such agents)). Examples of anti-inflammatory molecular antagonists include, but are not limited to, antagonists of HMGB1 and antagonists of one or more chitinase-like molecules. In certain embodiments, the anti-inflammatory agent is an agent useful in the prevention, management, treatment, and/or amelioration of asthma or one or more symptoms thereof. Non-limiting examples of such agents include adrenergic stimulants (e.g., catecholamines (e.g., epinephrine, isoproterenol, and isoetharine), resorcinols (e.g., metaproterenol, terbutaline, and fenoterol), and saligenins (e.g., salbutamol)), adrenocorticoids, blucocorticoids, corticosteroids (e.g., beclomethadonse, budesonide, flunisolide, fluticasone, triamcinolone, methylprednisolone, prednisolone, and prednisone), other steroids, beta2-agonists (e.g., albtuerol, bitolterol, fenoterol, isoetharine, metaproterenol, pirbuterol, salbutamol, terbutaline, formoterol, salmeterol, and albutamol terbutaline), anti-cholinergics (e.g., ipratropium bromide and oxitropium bromide), IgE antagonists, LL-4 antagonists (including antibodies), LL-5 antagonists (including antibodies), LL-13 antagonists (including antibodies), PDE4-inhibitor, NF-Kappa-B inhibitor, VLA-4 inhibitor, CpG, anti-CD23, selectin antagonists (TBC 1269), mast cell protease inhibitors (e.g., tryptase kinase inhibitors (e.g., GW-45, GW-58, and genisteine), phosphatidylinositide- 3' (PB)-kinase inhibitors (e.g., calphostin C), and other kinase inhibitors (e.g., staurosporine) (see Temkin et al, 2002, J Immunol 169:2662-9; Vosseller et al, 1997, Mol. Biol. Cell 8:909- 22; and Nagai et al, 1995, Biochem Biophys Res Commun 208:576-81)), C3 a receptor antagonists (including antibodies), immunosuppressant agents (e.g., methotrexate and gold salts), mast cell stabilizers (e.g., cromolyn sodium (INTAL™) and nedocromil sodium (TILADE™)), and mucolytic agents (e.g., acetylcysteine)). In a specific embodiment, the anti-inflammatory agent is a leukotriene modifier (e.g., montelukast (SINGULAIR™), zafirlukast (ACCOLATE™), pranlukast (ONON™), or zileuton (ZYFLO™) (see Table 1)).
Tab e 1. Leukotriene Modifiers for Asthma Therapy Leukotriene Modifier Usual Daily Dosage Montelukast (SLNGULAIR™) 4 mg for 2-5 years old 5 mg for 6 to 15 years old
Figure imgf000053_0001
In certain embodiments, the anti-inflammatory agent is an agent useful in preventing, treating, managing, or ameliorating allergies or one or more symptoms thereof. Non-limiting examples of such agents include antimediator drugs (e.g., antihistamine, see Table 2 for non- limiting examples of antihistamine and typical dosages of such agents), corticosteroids, decongestants, sympathomimetic drugs (e.g., α-adrenergic and β-adrenergic drugs), TNX901 (Leung et al, 2003, NEnglJMed 348:986-93), IgE antagonists (e.g., antibodies rhuMAb- E25 omalizumab (see Finn et al, 2003, J Allergy Clin Immuno 111:278-84; Corren et al, 2003, J Allergy Clin Immuno 11:87-90; Busse and Neaville, 2001, Curr Opin Allergy Clin Immuno 1:105-8; and Tang and Powell, 2001, EurJPediatr 160: 696-704), HMK-12 and 6HD5 (see Miyajima et al, 2002, Int Arch Allergy Immuno 128:24-32), and niAB Hu-901 (see van Neerven et al, 2001, Int Arch Allergy Immuno 124:400), theophylline and its derivatives, glucocorticoids, and immunotherapies (e.g., repeated long-term injection of allergen, short course desensitization, and venom immunotherapy).
Table 2. Hi Antihistamines Chemical class and representative drugs Usual daily dosage Ethanolamine Diphehydramine 25-50 mg every 4-6 hours Clemastine 0.34-2.68 mg every 12 hours Ethylenediamine Tripelennamine 25-50 mg every 4-6 hours Alkylamine Brompheniramine 4 mg every 4-6 hours; or 8-12 mg of SR form every 8-12 hour Chlorpheniramine 4 mg every 4-6 hours; or 8-12 mg ofSR form every 8-12 hour Triprolidine (1.25 mg/5ml) 2.5 mg every 4-6 hours Phenothiazine Promethazine 25 mg at bedtime Piperazine Hydroxyzine 25 mg every 6-8 hours Piperidines Astemizole (nonsedating) lO mg/day Azatadine 1-2 mg every 12 hours Cetirzine 10 mg/day Cyproheptadine 4 mg every 6-8 hour Fexofenadine (nonsedating) 60 mg every 12 hours Loratidme (nonsedating) 10 mg every 24 hours Anti-inflammatory therapies and their dosages, routes of administration, and recommended usage are known in the art and have been described in such literature as the Physician 's Desk Reference (57th ed., 2003) and The Merk Manual (17th ed., 1999).
Anti- Viral Agents Any anti- viral agent well-known to one of skill in the art for the treatment, prevention, management, or amelioration of respiratory conditions or a symptom thereof (e.g., viral respiratory infection) can be used in the compositions and methods of the invention. Non- limiting examples of anti-viral agents include proteins, polypeptides, peptides, fusion proteins antibodies, nucleic acid molecules, organic molecules, inorganic molecules, and small molecules that inhibit or reduce the attachment of a virus to its receptor, the internalization of a virus into a cell, the replication of a virus, or release of virus from a cell In particular, antiviral agents include, but are not limited to, nucleoside analogs (e.g., zidovudine, acyclovir, gangcyclovir, vidarabine, idoxuridine, trifluridine, and ribavirin), foscarnet, amantadine, rimantadine, saquinavir, indinavir, ritonavir, alpha-interferons and other interferons, and AZT. In specific embodiments, the anti- viral agent is an antibody agent that is immunospecific for a viral antigen. As used herein, the term "viral antigen" includes, but is not limited to, any viral peptide, polypeptide and protein (e.g., RSV F glycoprotein, RSV G glycoprotein, influenza virus neuraminidase, influenza virus hemagglutinin, and herpes simplex virus glycoprotein (e.g., gB, gC, gD, and gE)) that is capable of eliciting an immune response. Antibodies useful in this invention for prevention, management, treatment, and/or amelioration of a viral infectious disease include, but are not limited to, antibodies against antigens of pathogenic viruses, including as examples and not by limitation: adenovirdiae (e.g., mastadenovirus and aviadenovirus), herpesviridae (e.g., herpes simplex virus 1, herpes simplex virus 2, herpes simplex virus 5, and herpes simplex virus 6), leviviridae (e.g., levivirus, enterobacteria phase MS2, allolevirus), poxviridae (e.g., chordopoxvirinae, parapoxvims, avipoxvirus, capripoxvirus, leporiipoxvirus, suipoxvirus, moUuscipoxvirus, and entomopoxvirinae), papovaviridae (e.g., polyomavirus and papillomavirus), paramyxoviridae (e.g., paramyxovirus, parainfluenza virus 1, mobillivirus (e.g., measles virus), rubulavirus (e.g., mumps virus), pneumonovirinae (e.g., pneumovirus, human respiratory syncytial virus), and metapneumovirus (e.g., avian pneumovirus and human metapneumovirus)), picornaviridae (e.g., enterovirus, rhinovirus, hepato virus (e.g., human hepatits A virus), cardiovirus, and apthovirus), reoviridae (e.g., orthoreovirus, orbivirus, rotavirus, cypovirus, fijivirus, phytoreovirus, and oryzavirus), retro viridae (e.g., mammalian type B retroviruses, mammalian type C retroviruses, avian type C retroviruses, type D retrovirus group, BLV- HTLN retroviruses, lentivirus (e.g. human immunodeficiency virus 1 and human immunodeficiency virus 2), spumavirus), flaviviridae (e.g., hepatitis C virus), hepadnaviridae (e.g., hepatitis B virus), togaviridae (e.g., alphavirus (e.g., sindbis virus) and rubivirus (e.g., rubella virus)), rhabdo viridae (e.g., vesiculovirus, lyssavirus, ephemera virus, cytorhabdovirus, and necleorhabdovirus), arenaviridae (e.g., arenavirus, lymphocytic choriomeningitis virus, Ippy virus, and lassa virus), and coronaviridae (e.g., coronavirus and torovirus). Specific examples of antibodies available useful for the prevention, management, treatment, and/or amelioration of a viral infectious disease include, but are not limited to, PRO542 (Progenies) which is a CD4 fusion antibody useful for the treatment of HIN infection and SYΝAGIS® (Medlmmune, Inc.; International Publication No. WO 02/43660) which is a humanized antibody useful for treatment of RSV. In a specific embodiment, the anti- viral agent used in the compositions and methods of the invention inhibits or reduces a pulmonary or respiratory virus infection, inhibits or reduces the replication of a virus that causes a pulmonary or respiratory infection, or inhibits or reduces the spread of a virus that causes a pulmonary or respiratory infection to other cells or subjects. In another specific embodiment, the anti-viral agent used in the compositions and methods of the invention inhibits or reduces infection by RSV, hMPV, or PLV, inhibits or reduces the replication of RSV, hMPV, or PLV, or inhibits or reduces the spread of RSV, hMPV, or PIV to other cells or subjects. Examples of such agents include, but are not limited to, nucleoside analogs, such as zidovudine, acyclovir, gangcyclovir, vidarabine, idoxuridine, trifluridine, and ribavirin, as well as foscarnet, amantadine, rimantadine, saquinavir, indinavir, ritonavir, and the alpha-interferons. See U.S. Provisional Patent Application 60/398,475 filed July 25, 2002, entitled "Methods of Treating and Preventing RSV, HMPV, and PLV Using Anti-RSV, Anti-HMPV, and Anti-PLV Antibodies," which is incorporated herein by reference in its entirety. In preferred embodiments, the viral infection is RSV and the anti-viral antigen is an antibody that immunospecifically binds to an antigen of RSV. hi certain embodiments, the anti-RSV-antigen antibody immunospecifically binds to an RSV antigen of the Group A of RSV. In other embodiments, the anti-RSV-antigen antibody immunospecifically binds to an RSV antigen of the Group B of RSV. In other embodiments, the anti-RSV antigen antibody immunospecifically binds to an antigen of RSV of one Group and cross reacts with the analogous antigen of the other Group. In particular embodiments, the anti-RSV-antigen antibody immunospecifically binds to a RSV nucleoprotein, RSV phosphoprotein, RSV matrix protein, RSV small hydrophobic protein, RSV RNA-dependent RNA polymerase, RSV F protein, and/or RSV G protein, hi additional specific embodiments, the anti-RSV- antigen antibody binds to allelic variants of a RSV nucleoprotein, a RSV nucleocapsid protein, a RSV phosphoprotein, a RSV matrix protein, a RSV attachment glycoprotein, a RSV fusion glycoprotein, a RSV nucleocapsid protein, a RSV matrix protein, a RSV small hydrophobic protein, a RSV RNA-dependent RNA polymerase, a RSV F protein, a RSV L protein, a RSV P protein, and/or a RSV G protein. It should be recognized that antibodies that immunospecifically bind to a RSV antigen are known in the art. For example, SYNAGIS® (Palivizumab) is a humanized monoclonal antibody presently used for the prevention of RSV infection in pediatric patients. In a specific embodiment, an antibody to be used in accordance with the methods of the present invention is SYNAGIS® or an antibody-binding fragment thereof (e.g., a fragment containing one or more complementarity determining regions (CDRs) and preferably, the variable domain of SYNAGIS®). The amino acid sequence of SYNAGIS® is disclosed, e.g., in Johnson et al, 1997, J. Infectious Disease 176:1215-24, and U.S. Patent No. 5,824,307 and International Application Publication No.: WO 02/43660, entitled "Methods of Administering/Dosing Anti-RSV Antibodies for Prophylaxis and Treatment", by Young et al, which are incorporated herein by reference in their entireties. One or more antibodies or antigen-binding fragments thereof that immunospecifically bind to a RSV antigen comprise a Fc domain with a higher affinity for the FcRn receptor than the Fc domain of SYNAGIS® (Palivizumab) can also be used in accordance with the invention. Such antibodies are described in U.S. Patent Application No.: 10/020,354, filed December 12, 2001, which is incorporated herein by reference in its entireties. Further, the anti-RSV-antigen antibody A4B4; P12f2 P12f4; Plld4; Ale9; A12a6; A13c4; A17d4; A4B4; 1X-493L1; FR H3-3F4; M3H9; Y10H6; DG; AFFF; AFFF(l); 6H8; L1-7E5; L2-15B10; A13al 1; Alh5; A4B4(1);A4B4-F52S; or A4B4L1FR-S28R can be used in accordance with the invention. These antibodies are disclosed in International Application Publication No.: WO 02/43660, entitled "Methods of Administering/Dosing Anti-RSV Antibodies for Prophylaxis and Treatment", by Young et al, and US Provisional Patent Application 60/398,475 filed July 25, 2002, entitled "Methods of Treating and Preventing RSV, HMPV, and PLV Using Anti-RSV, Anti-HMPV, and Anti-PIN Antibodies" each ofwhich are incorporated herein by reference in their entireties. Ln certain embodiments, the anti-RSV-antigen antibodies are the anti-RSV-antigen antibodies of or are prepared by the methods of U.S. Application o: 09/724,531, filed November 28, 2000; 09/996,288, filed November 28, 2001; and 09/996,265, filed November 28, 2001, all entitled "Methods of Administering/Dosing Anti-RSV Antibodies for Prophylaxis and Treatment", by Young et al, which are incorporated by reference herein in their entireties. Methods and composition for stabilized antibody formulations that can be used in the methods of the present invention are disclosed in U.S. Provisional Application Nos.: 60/388,921, filed June 14, 2002, and 60/388,920, filed June 14, 2002, each ofwhich are incorporated by reference herein in their entireties. Anti- viral therapies and their dosages, routes of administration and recommended usage are known in the art and have been described in such literature as the Physician 's Desk Reference (57th ed., 2003). Additional information on respiratory viral infections is available in Cecil Textbook of Medicine (18th ed., 1988).
Anti-Bacterial Agents Anti-bacterial agents and therapies well-known to one of skill in the art for the prevention, treatment, management, or amelioration of respiratory conditions or one or more symptoms thereof (e.g., a bacterial respiratory infection) can be used in the compositions and methods of the invention. Non-limiting examples of anti-bacterial agents include proteins, polypeptides, peptides, fusion proteins, antibodies, nucleic acid molecules, organic molecules, inorganic molecules, and small molecules that inhibit or reduce a bacterial infection, inhibit or reduce the replication of bacteria, or inhibit or reduce the spread of bacteria to other cells or subjects. Specific examples of anti-bacterial agents include, but are not limited to, antibiotics such as penicillin, cephalosporin, imipenem, axtreonam, vancomycin, cycloserine, bacitracin, chloramphenicol, erythromycin, clindamycin, tetracycline, streptomycin, tobramycin, gentamicin, amikacin, kanamycin, neomycin, spectinomycin, trimethoprim, norfloxacin, rifampin, polymyxin, amphotericin B, nystatin, ketocanazole, isoniazid, metronidazole, and pentamidine. In certain embodiments, the anti-bacterial agent is an agent that inhibits or reduces a pulmonary or respiratory bacterial infection, inhibits or reduces the replication of a bacteria that causes a pulmonary or respiratory infection, or inhibits or reduces the spread of a bacteria that causes a pulmonary or respiratory infection to other cells or subjects. In cases in which the pulmonary or respiratory bacterial infection is a mycoplasma infection (e.g., pharyngitis, tracheobronchitis, and pneumonia), the anti-bacterial agent is preferably a tetracycline, erythromycin, or spectinomycin. In cases in which the pulmonary or respiratory bacterial infection is tuberculosis, the anti-bacterial agent is preferably rifampcin, isonaizid, pyranzinamide, ethambutol, and streptomycin. In cases in which the pulmonary or respiratory bacterial infection is pneumonia caused by an aerobic gram negative bacilli (GNB), the anti-bacterial agent is preferably penicillin, first, second, or third generation cephalosporin (e.g., cefaclor, cefadroxil, cephalexin, or cephazolin), erythomycin, clindamycin, an aminoglycoside (e.g., gentamicin, tobramycin, or amikacine), or a monolactam (e.g., aztreonam). In cases in which the respiratory infection is recurrent aspiration pneumonia, the anti-bacterial agent is preferably penicillin, an aminoglycoside, or a second or third generation cephalosporin. Anti-bacterial therapies and their dosages, routes of administration and recommended usage are known in the art and have been described in such literature as the Physician 's Desk Reference (57th ed., 2003), Cecil Textbook of Medicine (18th ed., 1988), and Tlie Merk Manual of Diagnosis and Tlierapy (17th ed. 1999).
Anti-Fungal Agents Anti-fungal agents and therapies well known to one of skill in the art for prevention, management, treatment, and/or amelioration of respiratory conditions or one or more symptoms thereof (e.g., a fungal respiratory infection) can be used in the compositions and methods of the invention. Non-limiting examples of anti-fungal agents include proteins, polypeptides, peptides, fusion proteins, antibodies, nucleic acid molecules, organic molecules, inorganic molecules, and small molecules that inhibit or reduce fungal infection, inhibit or reduce the replication of fungi, or inhibit or reduce the spread of fungi to other subjects. Specific examples of anti-fungal agents include, but are not limited to, azole drugs (e.g., miconazole, ketoconazole (NIZORAL®), caspofungin acetate (CANCIDAS®), imidazole, triazoles (e.g., fluconazole (DIFLUCAN®)), and itraconazole (SPORANOX®)), polyene (e.g., nystatin, amphotericin B (FUNGIZONE®), amphotericin B lipid complex ("ABLC")(ABELCET®), amphotericin B colloidal dispersion ("ABCD")(AMPHOTEC®), liposomal amphotericin B (AMBISONE®)), potassium iodide (KI), pyrimidine (e.g., flucytosine (ANCOBON®)), and voriconazole (VFEND®). See, e.g., Table 4 for a list of specific anti-fungal agents and their recommended dosages. In certain embodiments, the anti-fungal agent is an agent that inhibits or reduces a respiratory fungal infection, inhibits or reduces the replication of a fungus that causes a pulmonary or respiratory infection, or inhibits or reduces the spread of a fungus that causes a pulmonary or respiratory infection to other subjects. In cases in which the pulmonary or respiratory fungal infection is Blastomyces dermatitidis, the anti-fungal agent is preferably itraconazole, amphotericin B, fluconazole, or ketoconazole. In cases in which the pulmonary or respiratory fungal infection is pulmonary aspergilloma, the anti-fungal agent is preferably amphotericin B, liposomal amphotericin B, itraconazole, or fluconazole. In cases in which the pulmonary or respiratory fungal infection is histoplasmosis, the anti-fungal agent is preferably amphotericin B, itraconazole, fluconazole, or ketoconazole. In cases in which the pulmonary or respiratory fungal infection is coccidioidomycosis, the anti-fungal agent is preferably fluconazole or amphotericin B. In cases in which the pulmonary or respiratory fungal infection is cryptococcosis, the anti-fungal agent is preferably amphotericin B, fluconazole, or combination of the two agents. In cases in which the pulmonary or respiratory fungal infection is chromomycosis, the anti-fungal agent is preferably itraconazole, fluconazole, or flucytosine. hi cases in which the pulmonary or respiratory fungal infection is mucormycosis, the anti-fungal agent is preferably amphotericin B or liposomal amphotericin B. In cases in which the pulmonary or respiratory fungal infection is pseudoallescheriasis, the anti-fungal agent is preferably itraconazole ore miconazole. Anti-fungal therapies and their dosages, routes of administration, and recommended usage are known in the art and have been described in such literature as Dodds et al, 2000, Pharmacotherapy 20:1335-55, the Physician 's Desk Reference (57th ed., 2003) and the Merk Manual of Diagnosis and Therapy (17th ed., 1999).
Table 3. Anti-fun al A ents
Figure imgf000059_0001
Figure imgf000060_0001
Combination Treatment for Inflammatory or Autoimmune Disorders The present invention provides compositions for the treatment, prophylaxis, and amelioration of one or more symptoms associated with an autoimmune or inflammatory disorder. In a specific embodiment, a composition comprises one or more HAAH antagonists. In another embodiment, a composition comprises one or more nucleic acid molecules encoding one or more HAAH antagonists, h another embodiment, a composition comprises one or more HAAH antagonists and one or more prophylactic or therapeutic agents other than HAAH antagonists, said prophylactic or therapeutic agents known to be useful for, or having been or currently being used in the prevention, treatment or amelioration of one or more symptoms associated an autoimmune or inflammatory disorder. In another embodiment, a composition comprises one or more nucleic acid molecules encoding one or more HAAH antagonists and one or more prophylactic or therapeutic agents other than HAAH antagonists, said prophylactic or therapeutic agents known to be useful for, or having been or currently being used in the prevention, treatment or amelioration of one or more symptoms associated an autoimmune or inflammatory disorder. In another embodiment, a composition comprises one or more HAAH antagonists and one or more nucleic acid molecules encoding one or more prophylactic or therapeutic agents other than HAAH antagonists, said prophylactic or therapeutic agents known to be useful for, or having been or currently being used in the prevention, treatment or amelioration of one or more symptoms associated an autoimmune or inflammatory disorder. In yet another embodiment, a composition comprises one or more nucleic acid molecules encoding one or more HAAH antagonists and one or more nucleic acid molecules encoding one or more prophylactic or therapeutic agents other than HAAH antagonists, said prophylactic or therapeutic agents known to be useful for, or having been or currently being used in the prevention, treatment or amelioration of one or more symptoms associated an autoimmune or inflammatory disorder. In a specific embodiment, a composition comprises a one or more HAAH antagonists and one or more immunomodulatory agents. The immunomodulatory agents can be an αγβ antagonists (e.g. VITAXLN™), T cell antagonists (e.g. MedI-507, Amevive, or Xanelim) and TNF-α antagonists (Enbrel or Remicade). In another embodiment, a composition comprises small molecules the modulate the immune system, like methotrexate. In another embodiment, a composition comprises HAAH antagonist and one or more CD2 antagonists. In another embodiment, a composition comprises a HAAH agonist or more integrin αγβ3 antagonists and one or more CD2 binding molecules. In yet another embodiment, a composition comprises HAAH agonist or an antigen-binding fragment thereof and one or more CD2 binding molecules. In a preferred embodiment, a composition comprises a HAAH agonist, VITAXLN™ or an antigen-binding fragment thereof and MEDI-507 or an antigen- binding fragment thereof. See, e.g., co-pending application 10/091,236, incorporated here in its entirety. EXAMPLES The invention is now described with reference to the following examples. These examples are provided for the purpose of illustration only and the invention should in no way be construed as being limited to these examples but rather should be construed to encompass any and all variations which become evident as a result of the teachings provided herein.
Example 1.
Tissue Expression of HAAH It has been reported that HAAH is abundantly expressed in several malignant neoplasms, while low levels ofHAAH expression are seen in most normal tissues and cell types. Examination of the levels and distribution ofHAAH protein in tissues yielded the unexpected finding that HAAH is highly expressed in the tissue-resident mast cells of numerous normal human tissues. Normal human small intestine, skeletal muscle and heart tissue was stained with a monoclonal antibody specific for HAAH. In all tissue examined the tissue-resident mast cells stain intensely (see Figure 1). Some minimal nonspecific background staining is seen in the submucosal plexus of the small intestine and the cardiac myocytes.
Materials and Methods Immunohistochemistty To generate sections from fixed tissue, a microtome was used to cut tissue sections at 4-5 microns. The sections were floated on a water bath and picked up onto slides. Slides containing paraffin sections were deparaffinized through xylene and alcohol, rehydrated, and then subjected to the steam method of target retrieval (DAKO reagent #S1700). Antibody titration experiments were conducted with anti HAAH mAb 15C7 to establish dilutions that would result in minimal background and maximal detection of signal. Serial dilutions were performed at 1:50, 1:100, 1:250, 1:400, 1:500 and 1:600. The serial dilutions study demonstrated the highest signal-to-noise ratios at dilutions of 1 :400 and 1 :600 on paraffin-embedded, formalin fixed tissues after deparafrmization and rehydration. These dilutions were selected for the study. 15C7 mAb was used as the primary antibody, and the principal detection system consisted of a Vector anti-mouse secondary (BA-2000) and a Vector ABC-AP kit (AK-5000) with a Vector red substrate kit (SK-5100), which was used to produce a fuschia-colored deposit. Tissues were also stained with positive control antibodies (CD31 and vimentin) to ensure that the tissue antigens were preserved and accessible for immunohistochemical analysis. Only tissues that were positive for CD31 and vimentin staining were selected for the remainder of the study. The negative control consisted of performing the entire immunohistochemistry procedure on adjacent sections in the absence of primary antibody, or using the irrelevant isotype-matched control antibody 1A7. Slides were imaged with a DVC 130C digital camera coupled to a Nikon microscope.
Example 2.
HAAH Expression in Mast Cell Tissue Culture Cell Lines To expand the observation that tissue-resident mast cells stain intensely with an anti- HAAH specific antibody four cell lines (H460, a human lung cancer cell line known to express HAAH on the cell surface; UT-7, a human mast cell precursor cell line, KU812, another human mast cell precursor cell line, and RBL, a rat mast cell line) were probed with a monoclonal antibody directed against the catalytic domain ofHAAH (15C7). As shown in figure 2, all four cell lines stain positive, with the human lung cancer cell line and the rat mast cell line showing the strongest response (the band for intact HAAH runs at -lOOkDa, and lines up with the lOOkDa marker in the molecular weight standards shown on the left).
Interestingly, the antibody directed against HAAH (Human aspartyl/aspariginal β hydroxylase) cross reacts with rat AAH, probably due to the high degree of conservation between the human and rat catalytic domains (95% identity). The strength of the signal suggests that higher levels ofHAAH are expressed in H460 and RBL cells than in UT-7 and
KU812 cells, however, by FACS analysis the levels appear similar for all four cell lines (see below). In addition, the commonly used mast cell-precursor cell line (UT-7) was analyzed by real-time PCR to quantitate HAAH gene expression and by FACS to examine HAAH cell- surface protein expression. Real-time PCR analysis demonstrated that the level ofHAAH message was at least 100-fold more abundant than the level of message encoding LL-4, a cytokine known to be expressed by mast cells (see Table 4). FACS analysis demonstrated that a large number of UT-7 cells stain positive for HAAH when labeled with the HAAH specific monoclonal antibody 15C7 (see Figure 3). The 15C7-stained cells clearly show at least two populations, probably due to the fact that UT-7 cells are not a clonal cell line, but are composed of sub-populations of cells at different stages of differentiation. Several other cells lines including KU812, another human mast cell precursor cell line (Figure 3), Mel 9 and RBL-23H cells (mouse and rat mast cell-precursor cell lines respectively)(data not shown) were also examined by FACS analysis and are highly positive for cell-surface HAAH expression. The 15C7-stained KU812 cells are a more homogenous population reflective of the homogeneity of the cell line (Figure 4).
Figure imgf000063_0001
Figure imgf000064_0001
Materials and Methods Western Blot Analysis: Four cell lines were used for this analysis: H460, a human lung cancer cell line known to express HAAH; UT-7, a human mast cell-precursor cell line; KU812, another human mast cell-precursor cell line; and RBL, a rat mast cell line. Cells were harvested, counted, resuspended in 2X Laemmli buffer at 106 cells/ml, and stored at -80° C until analysis. For analysis, the cell lysates were thawed, and 20 μl aliquots were mixed with 2 μl of 0.2 M dithiothreitol and boiled for two minutes. The samples were then loaded on a 10% bis/tris gel and run for 40 minutes at 80 mA. The proteins were blotted onto to nitrocellulose for 1 hour at 100 V, and blocked with 5% low-fat milk in PBS. The blot was probed with 15C7 (1 μg/ml in block) for one hour, and then with goat-anti mouse HRP-conjugate. The blot was visualized by chemiluminescence. Real-time PCR Analysis: Total RNA was isolated from the UT7 cells using Trizol reagent. RNA was quantitated and 100 ngs were reverse transcribed using the First Strand cDNA kit from Roche. Analysis was performed using probes specific for HAAH messenger RNA (called ASPH in the NCBI database) and probes specific for EL-4. Each determination was performed in triplicate, two separate determinations were performed. For this analysis, the experimentally observed level of LL-4 message from one trial compared to the internal 18S RNA standard is set at 1, and the abundance of the other messenger RNA relative to that level is calculated. Real time PCR was performed using the GeneAmp7000 Sequence Detection System from ABI. The predeveloped Assay on Demand sequence detection reagents for ASPH and LL4 from ABI were used according to the manufacturer's recommendation. Relative message quantitation was determined using the ΔΔCt method using 18S RNA as the internal standard. FACS Analysis: Cells were harvested with trypsin-free dissociation buffer, spun down and resuspended in FACS buffer (PBS, 2%FCS, 0.2% NaN3). Viable cell concentration was determined by counting typan blue-excluding cells in a hemacytometer. Cells were transferred to eppendorf tubes at a concentration of 200,000 cells in 50 μl FACS buffer. 3 tubes were set up per cell line, labelled -1,-2; -1, +15C7. 1 μl of 1 mg/ml human IgG was added to each tube, to block non-specifc binding to Fc-receptor, and incubated at room temperature for 10 minutes. 1 μl of the anti-HAAH mAb 15C7 (1 mg/ml) was added to the +15C7 tube, and incubated at room temperature for 20 minutes. To wash, the tubes were spun in a microcentrifuge (~2 min at 8k), the supernatants by were removed by pipet, the pellets were resuspended in 0.5 ml FACS buffer, and spun as above. The supernatants were then removed, and each pellet was resuspended in 50 μl FACS buffer. 0.5 μl PE-labelled goat- anti mouse was added as the secondary antibody to all the tubes EXCEPT that labeled -1,-2, and incubated at room temperature for 20 minutes. The tubes were then washed as above, the pellets were resuspended in 0.5 ml PBS, 1%) formaldehyde, and stored at 4o C until FACS analysis was performed on a Becton Dickinson FACS calibur monitoring forward and side scatter.
Discussion Human Aspartyl (Asparaginyl) ?-Hydroxylase or HAAH, is a highly conserved enzyme which hydroxylates aspartic acid or asparagine residues in EGF-like domains of several proteins in the presence of ferrous iron. These EGF-like domains contain conserved motifs that form repetitive sequences in proteins such as clotting factors, extracellular matrix proteins, LDL receptor, NOTCH homologues or NOTCH ligand homologues. It is believed that EGF-like sequences play an important role in protein-protein interactions, as shown by mutations in EGF-like domains of fibrillin that cause Marfan's syndrome or factor LX, which produces hemophilia B (Downing et al, 1996, Cell, 85: 597-605; Nishimura et al, 1993, J. Biol. Chem., 268: 24041-24046). In addition, several of these EGF-like domain containing protein are also recognized as cancer transformation-associated proteins. Over-expression of HAAH is strongly associated with the progression and invasiveness of multiple cancers so it was unexpected that tissue-associated mast cells in normal tissues would show intense staining with the HAAH specific monoclonal antibody 15C7. Given the prominent role of mast cells in numerous immune responses the presence of HAAH on the surface of mast cells provides an excellent target for mast cell directed therapies. Little to no staining ofHAAH was seen on B- and T-cells and more limited staining was seen on other immune cells. Thus, targeting cell surface HAAH allows for highly directed therapies that more specifically target the mature mast cell. Such mast cell directed therapies would be useful for the treatment of a large number of disorders including inflammatory or autoimmune disorders or cardiovascular disease. The disclosures of each and every patent, patent application, and publication cited herein are hereby incorporated by reference in their entirety. While this invention has been disclosed with reference to specific embodiments, it is apparent that other embodiments and variations of this invention may be devised by others skilled in the art without departing from the true spirit and scope of the invention. The appended claims are intended to be construed to include all such embodiments and equivalent variations.

Claims

CLAIMS:
1. A method of preventing, managing, treating or ameliorating bronchial hyper- responsiveness or one or more symptoms thereof, said method comprising administering to a human subject in need thereof an effective amount of a molecule that immunospecifically binds to HAAH.
2. A method of preventing, managing, treating or ameliorating acute or chronic asthma or an allergy or one or more symptoms thereof, said method comprising administering to a human subject in need thereof an effective amount of a molecule that immunospecifically binds to HAAH.
3. A method of preventing, managing, treating or ameliorating wheezing, said method comprising administering to a human subject in need thereof an effective amount of a molecule that immunospecifically binds to HAAH.
4. A method of preventing, managing, treating or ameliorating atherosclerosis, said method comprising administering to a human subject in need thereof an effective amount of a molecule that immunospecifically binds to HAAH.
5. A method of claim 1, 2, 3 or 4, wherein the molecule that immunospecifically binds to HAAH is an antibody, BiTE, or fusion protein.
6. The method of claim 5, wherein the molecule is an antagonist ofHAAH.
7. The method of claim 1, 2, 3, 4, 5 or 6 further comprising administering an effective amount of at least one other therapeutic agent.
8. The method of claim 7, wherein the therapeutic agent is an immunomodulatory agent, an anti-inflammatory agent, an anti- viral agent, an antibiotic, an antifungal agent or a mast cell modulator.
9. The method of claim 7 comprising administering to said subject an effective amount of an antibody that does not bind to HAAH.
10. The method of claim 7 or 8 further comprising administering to said subject a leukotriene modifier, an anti-histamine, an anti-IgE antibody, an anti-LL-4 antibody, an anti-LL9 antibody, or a mast cell protease inhibitor.
11. The method of claim 1 , 2, 3 or 4, wherein the molecule that immunospecifically binds to HAAH is administered parenterally, orally or intranasally.
12. A method of treating, managing or ameliorating an inflammatory disorder or an autoimmune disorder or one or more symptoms thereof, said method comprising administering to a subject in need thereof a prophylactically or therapeutically effective amount of a molecule that immunospecifically binds to HAAH.
13. A method of treating, managing, or ameliorating an inflammatory disorder or an autoimmune disorder or one or more symptoms thereof, said method comprising administering to a subject in need thereof a prophylactically or therapeutically effective amount of a molecule that immunospecifically binds to HAAH and one or more immunomodulatory agent.
14. The method of claim 12, wherein the molecule that immunospecifically binds to ' HAAH is an antibody, BiTE, or fusion protein.
15. The method of claim 13, wherein the immunomodulatory agent is a small organic molecule.
16. The method of claim 13, wherein at least one immunomodulatory agent is an antibody, BiTE, or fusion protein.
17. The method of claim 16, wherein the immunomodulatory agent is a T cell receptor modulator, cytokine receptor modulator, or a molecule that immunospecifically binds to a cytokine.
18. The method of claim 15, wherein the small organic molecule is methotrexate, leflunomide, cyclophosphamide, cyclosporine A, FK506, mycophenolate mofetil, rapamycin, mizoribine, deoxyspergualm, brequinar, a malononitriloamide, a steroid, or corticosteriod.
19. The method of claim 14, wherein the antibody is an anti-T cell antibody or an anti avb3 antibody.
20. . The method of claim 19, wherein the antibody is Vitaxin™.
21. The method of claim 20, wherein the antibody is Medi-507.
22. A method of inhibiting mast cell activity comprising contacting the mast cell with a compound that binds to HAAH.
23. The method of claim 22, wherein the compound that binds to HAAH is an antibody, BiTE, or fusion protein.
24. The method of claim 22, wherein the compound that bind to HAAH is a small organic molecule.
PCT/US2004/019400 2003-06-17 2004-06-17 Novel methods to modulate mast cells WO2005003302A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US47898603P 2003-06-17 2003-06-17
US60/478,986 2003-06-17

Publications (2)

Publication Number Publication Date
WO2005003302A2 true WO2005003302A2 (en) 2005-01-13
WO2005003302A3 WO2005003302A3 (en) 2005-06-09

Family

ID=33563783

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/019400 WO2005003302A2 (en) 2003-06-17 2004-06-17 Novel methods to modulate mast cells

Country Status (1)

Country Link
WO (1) WO2005003302A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1699485A2 (en) * 2003-11-14 2006-09-13 Massachusetts Institute Of Technology Anti-hydroxylase antibodies and uses thereof
CN101307302B (en) * 2007-05-17 2012-02-29 陕西北美基因股份有限公司 Process for preparing hybridoma cell for secreting anti-human asparagine hydroxylase monoclonal antibodies
CN113975386A (en) * 2014-05-23 2022-01-28 塞尔德克斯医疗公司 Treatment of eosinophil or mast cell related disorders

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6783758B2 (en) * 1999-11-08 2004-08-31 Rhode Island Hospital Diagnosis and treatment of malignant neoplasms

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6783758B2 (en) * 1999-11-08 2004-08-31 Rhode Island Hospital Diagnosis and treatment of malignant neoplasms

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1699485A2 (en) * 2003-11-14 2006-09-13 Massachusetts Institute Of Technology Anti-hydroxylase antibodies and uses thereof
EP1699485A4 (en) * 2003-11-14 2007-12-19 Massachusetts Inst Technology Anti-hydroxylase antibodies and uses thereof
US7413737B2 (en) 2003-11-14 2008-08-19 Massachusetts Institute Of Technology Anti-hydroxylase antibodies and uses thereof
CN101307302B (en) * 2007-05-17 2012-02-29 陕西北美基因股份有限公司 Process for preparing hybridoma cell for secreting anti-human asparagine hydroxylase monoclonal antibodies
CN113975386A (en) * 2014-05-23 2022-01-28 塞尔德克斯医疗公司 Treatment of eosinophil or mast cell related disorders

Also Published As

Publication number Publication date
WO2005003302A3 (en) 2005-06-09

Similar Documents

Publication Publication Date Title
EP2484696B1 (en) Antagonistic hLIGHT-specific human monoclonal antibodies
JP6289375B2 (en) Anti-IL-36R antibody
US20050059592A1 (en) EphA2 and hyperproliferative cell disorders
US20060099207A1 (en) High affinity antibodies against HMGB1 and methods of use thereof
US20060122138A1 (en) Modulators of EphA2 and EphrinA1 for the treatment of fibrosis-related disease
JP2010518873A (en) Antagonist anti-OX40 antibodies and their use in the treatment of inflammatory and autoimmune diseases
KR20060111464A (en) Antibodies that bind interleukin-4 receptor
CA2685222A1 (en) Methods of reducing eosinophil levels
US20160311914A1 (en) Use of b lymphocyte stimulator protein antagonists to promote transplantation tolerance
WO2005003302A2 (en) Novel methods to modulate mast cells
WO2009097262A2 (en) Chitotriosidase and methods of use thereof
AU2013203359B2 (en) Antagonistic human LIGHT-specific human monoclonal antibodies

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase