CA2331880A1 - Bactericidal/permeability-increasing protein (bpi) deletion analogs - Google Patents

Bactericidal/permeability-increasing protein (bpi) deletion analogs Download PDF

Info

Publication number
CA2331880A1
CA2331880A1 CA002331880A CA2331880A CA2331880A1 CA 2331880 A1 CA2331880 A1 CA 2331880A1 CA 002331880 A CA002331880 A CA 002331880A CA 2331880 A CA2331880 A CA 2331880A CA 2331880 A1 CA2331880 A1 CA 2331880A1
Authority
CA
Canada
Prior art keywords
bpi
rbpi
leu
amino acid
val
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
CA002331880A
Other languages
French (fr)
Inventor
Arnold Horwitz
Stephen Fitzhugh Carroll
David Burke
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Xoma Technology Ltd USA
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2331880A1 publication Critical patent/CA2331880A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4742Bactericidal/Permeability-increasing protein [BPI]

Abstract

Novel BPI deletion analogs are provided that consist of amino acid residues (10 through 193) of mature human BPI wherein the cysteine residue at BPI amino acid position (132) is replaced by another amino acid. Fusion proteins comprising these analogs are also provided, as are polynucleotides encoding these products, materials and methods for their recombinant production, compositions and medicaments of these products, and therapeutic uses for these products.

Description

S
BACTERICIDAL/PERMEABILITY-INCREASING
PROTEIN (BPn DELETION ANALOGS
to BACKGROUND OF THE INVENTION
The present invention provides preparations of novel biologically active deletion analogs of bactericidal/permeability-increasing protein (BPI) characterized by improved stability and homogeneity as well as by enhanced in vivo 15 activity, and pharmaceutical compositions containing the same.
BPI is a protein isolated from the granules of mammalian polymorphonuclear leukocytes (PMNs or neutrophils), which are blood cells essential in the defense against invading microorganisms. BPI is known to bind to lipopolysaccharide, a major component of the outer membrane of gram-negative 20 bacteria that stimulates a potent inflammatory response which can lead to septic shock. Human BPI protein has been isolated from PMNs by acid extraction combined with either ion exchange chromatography [Elsbach, J. Biol. Chem. , 254:11000 (1979)] or E. coli affinity chromatography [Weiss, et al. , Blood, 69:652 (1987)]. BPI obtained in such a manner is referred to herein as natural BPI
and has 25 been shown to have potent bactericidal activity against a broad spectrum of gram-negative bacteria. The molecular weight of human BPI is approximately 55,000 daltons (55 kD). The amino acid sequence of the entire human BPI protein and the nucleic acid sequence of DNA encoding the protein have been reported in Figure 1 of Gray et al., J. Biol. Chem., 264:9505 (/989), incorporated herein by 30 reference. The Gray et al. amino acid sequence is set out in SEQ ID NO: 1 hereto.
U.S. Patent No. 5,198,541, the disclosure of which is incorporated herein by reference, discloses recombinant genes encoding, and methods for expression of, BPI proteins including recombinant BPI holoprotein, referred to as rBPI, and recombinant fragments of BPI.
A proteolytic N-terminal fragment of BPI having a molecular weight of about 25 kD has an amphipathic character, containing alternating hydrophobic and hydrophilic regions. This N-terminal fragment of human BPI possesses the anti-bacterial activity of the naturally-derived 55 kD human BPI holoprotein.
[Ooi et al., J. Bio. Chem., 262: 14891-14894 (1987)]. In contrast to the N-terminal portion, the C-terminal region of the isolated human BPI protein displays only slightly detectable anti-bacterial activity against gram-negative organisms.
[Ooi et al., J. Exp. Med., 174:649 (1991).] An N-terminal BPI fragment of approximately 23 kD, referred to as "rBPI~," has been produced by recombinant means and also retains anti-bacterial activity against gram-negative organisms. [Gazzano-Santoro et al., Infect. Immun. 60:4754-4761 (1992).] An N-terminal analog of BPI, rBPI2l, has been produced as described in Horwitz et al. , Protein Expression Purification, 8:28-40 (1996).
The bactericidal effect of BPI has been mpoited to be highly specific to gram-negative species, e.g., in Elsbach and Weiss, Inflaanmation: Basic Principles arid Clinical Correlates, eds. Gallin et al., Chapter 30, Raven Press, Ltd. (1992). This reported target cell specificity was believed to be the result of the strong attraction of BPI for lipopolysaccharide (LPS), which is unique to the outer membrane (or envelope) of gram-negative organisms. Although BPI was commonly th~ght to be non-tonic for other microorganisms, including yeast, and for higher eukaryotic cells, it has recently been discovered, as discussed infra, that BPI protein products, exhibit activity against gram-positive bacteria, mycoplasma, mycobacteria, fungi, protozoa, and chlamydia.
The precise mechanism by which BPI kills gram-negative bacteria is not yet completely elucidated, but it is believed that BPI must first bind to the surface of the bacteria through electrostatic and hydrophobic interactions between the cationic BPI protein and negatively charged sites on LPS . LPS has been referred to as "endotoxin" because of the potent inflammatory respanse that it stimulates, i. e. , the release of mediators by host inflammatory cells which may ultimately result in irreversible endotoxic shock. BPI binds to lipid A, reported to be the most toxic and most biologically active component of LPS.
In susceptible gram-negative bacteria, BPI binding is thought to disrupt LPS stmcture, leading to activation of bacterial enzymes that degrade phospholipids and peptidoglycans, altering the permeability of the cell's outer membrane, and initiating events that ultimately lead to cell death. [Elsbach and Weiss (1992), supra]. BPI is thought to act in two stages. The first is a sublethal stage that is characterized by immediate growth arrest, permeabilization of the outer membrane and selective activation of bacterial enzymes that hydrolyze phospholipids and peptidoglycans. Bacteria at this stage can be rescued by growth in serum albumin supplemented media [Mannion et al., J. Clip. Invest., 85:853-860 (1990)]. The second stage, defined by growth inhibition that cannot be reversed by senim albumin, occurs after prolonged exposure of the bacteria to BPI
and is characterized by extensive physiologic and structural changes, including apparent damage to the inner cytoplasmic membrane.
Initial binding of BPI to LPS leads to organizational changes that probably result from binding to the anionic groups of LPS, which normally stabilize the outer membrane through binding of Mg+'" and Ca++. Attachment of BPI to the outer membrane of gram-negative bacteria produces rapid permeabilization of the outer membrane to hydnaphobic agents such as actinomycin D. Binding of BPI and subsequent gram-negative bacterial killing depends, at least in part, upon the LPS polysaccharide chain length, with long O-chain bearing, "smooth" organisms being more resistant to BPI bactericidal effects than short O-chain bearing, "rough" organisms [Weiss et al., J. Clip. Invest. 65: 619-628 (1980)]. This first stage of BPI action, permeabilization of the gram-negative outer envelope, is reversible upon dissociation of the BPI, a process requiring high concentrations of divalent rations and synthesis of new LPS [Weiss et al., J.
Immunol. 132: 3109-3115 (1984)]. Loss of gram-negative bacterial viability, however, is not reversed by processes which restore the envelope integrity, suggesting that the bactericidal action is mediated by additional lesions induced in the target organism and which may be situated at the cytoplasmic membrane (Mannion et al., J. Clin. Invest. 86: 631-641 (1990)). Specific investigation of this possibility has shown that on a molar basis BPI is at least as inhibitory of cytoplasmic membrane vesicle function as polymyxin B (In't Veld et al., Infection and Immunity 56: 1203-1208 (1988)) but the exact mechanism as well as the relevance of such vesicles to studies of intact organisms has not yet been elucidated.
BPI protein products (which include naturally and m"combinantly produced BPI protein; natural, synthetic, and recombinant biologically active polypeptide fragments of BPI protein; biologically active polypeptide variants of BPI protein or fragments thereof, including hybrid fusion proteins and dimers;
biologically active polypeptide analogs of BPI protein or fragments or variants thereof, including cysteine-substituted analogs; and BPI-derived peptides) have been demonstrated to have a variety of beneficial activities. BPI protein products are known to be bactericidal for gram-negative bacteria, as described in U.S.
Patent Nos. 5,198,541 and 5,523,288, both of which are incorporated herein by reference.
BPI protein products are also known to enhance the effectiveness of antibiotic therapy in gram-negative bacterial infections, as described in U.S. Patent No.
5,523,288 and corresponding International Publication No. WO 95/08344 (PCT/US94/11225), which are incorporated herein by reference. BPI protein products are also known to be bactericidal for gram-positive bacteria and mycoplasma, and to enhance the effectiveness of antibiotics in gram-positive bacterial infections, as described in U.S. Patent No. 5,578,572 and corresponding International Publication No. WO 95/19180 (PCT/US95/00656), which are incorporated herein by reference. BPI protein products are further known to exhibit anti-fungal activity, and to enhance the activity of other anti-fungal agents, as described in U.S. Patent No. 5,627,153 and corresponding International Publication No. WO 95/19179 (PCT/US95/00498), and further as described for anti-fungal peptides in co-owned, co-pending U. S . Application Serial No.

08/621,259 filed March 21, 1996, which is in turn a continuation-in-part of U.S.
Application Serial No. 08/504, 841 filed July 20, 1994 and corresponding International Publication Nos. WO 96/08509 (PCT/US95/09262) and WO
97/04008 (PCT/US96/03845), all of which are incorporated herein by reference.
BPI protein products are further known to exhibit anti-protozoan activity, as described in U.S. Patent No. 5,646,114 and corresponding International Publication No. WO 96/01647 (PCT/US95/08624), all of which are incorporated herein by reference. BPI protein products are known to exhibit anti-chlamydial activity, as described in co-owned, co-pending U.S. Application Serial No.
08/694,843 filed August 9, 1996 and corresponding International Publication No.
WO 98/06415 (PCT/US97/13810), all of which are incorporated herein by reference. Finally, BPI protein products are known to exhibit anti-mycobacterial activity, as described in co-owned, co-pending U.S. Application Serial No.
08/626,646 filed April 1, 1996, which is in turn a continuation of U.S.
Application Serial No. 08/285,803 filed August 14, 1994, which is in turn a continuation-in-part of U.S. Application Serial No. 08/031,145 filed March 12, 1993 and corresponding International Publication No. W094/20129 (PCT/US94/02463), all of which are incorporated herein by reference.
The effects of BPI protein products in humans with endotoxin in circulation, including effects on TNF, IL-6 and endotoxin are described in U.S.
Patent Nos. 5,643,875 and 5,753,620 and corresponding International Publication No. WO 95/19784 (PCT/US95/01151), all of which are incorporated herein by reference.
BPI protein products are also known to be useful for treatment of specific disease conditions, such as meningococcemia in humans (as described in co-owned, co-pending U.S. Application Serial No. 081644,287 filed May 10, 1996 and corresponding International Publication No. 'WO 97/42966 (PCT/US97/08016), which are incorporated herein by reference), hemorrhagic trauma in humans, (as described in co-owned, co-pending U.S. Application Serial No. 08/862,785, a continuation-in-part of U.S. Serial No. 081652,292 filed May 23, 1996, now U.S. Patent No. 5,756,464, and corresponding International Publication No. WO 97/44056 (PCT/US97/08941), all of which are incorporated herein by reference), burn injury (as described in U.S. Patent No. 5,494,896 and corresponding International Publication No. WO 96/30037 (PCT/US96/02349), both of which are incorporated herein by reference), ischemia/repetfusion injury (as described in U.S. Patent No. 5,578,568, incorporated herein by reference), and liver resection (as described in co-owned, co-pending U.S. Application Serial No.
08/582,230 filed March 16, 1998 which is a continued prosecution application of the same serial no. filed January 3, 1996, which is in turn a continuation of U.S.
Application Serial No. 08/318,357 filed October 5, 1994, which is in turn a continuation-in-part of U.S. Application Serial No. 08/132,510 filed October 5, 1993, and corresponding International Publication No. WO 95/10297 (PCT/US94/11404), all of which are incorporated herein by reference).
BPI protein products are also known to neutralize the anti-coagulant activity of exogenous heparin, as described in U.S. Patent No. 5,348,942, incorporated herein by reference, as well as to be useful for treating chronic inflammatory diseases such as rheumatoid and reactive arthritis, as described in U.S. Patent No. 5,639,727, incorporated herein by reference, and for inhibiting angiogenesis and for treating angiogenesis-associated disorders including malignant tumors, ocular retinopathy and endometriosis, as described in co-owned, co-pending U.S. Applicaxion Serial Nos. 08/435,855, 08/466,624 and 08/466,826, and corresponding International Publication No. WO 94/20128 (PCT/US94/02401), all of which are incorporated herein by reference.
BPI protein products are also known for use in antithrombotic methods, as described in U.S. Patent No. 5,741,779 and corresponding International Publication No. W097/42967 (PCTlUS97/08017), which are incorporated herein by reference.

_7_ U.S. Patent Nos. 5,420,019 and 5,674,834 and corresponding International Publication No. W094/18323 (PCT/US94/01235), all of which are incorporated herein by reference, discloses that the replacement of the cysteine residue at amino acid position 132 or 135 with another amino acid renders the resulting BPI polypeptide resistant to dimerization and cysteine adduct formation.
It also discloses that terminating the N-terminal BPI fragment at BPI amino acid position 193 resulted in an expression product with reduced carboxy-terminal heterogeneity.
Of interest is the report in Capodici and Weiss, J. Immunol. , 156:4789-4796 (1996) that the in vitro transcription/translation products of DNA
encoding amino acid residues 1 through 193 (BPI,_193) and residues 13 through (BPI13-193) of mature BPI showed similar LPS-dependent binding to immobilized LPS.
There continues to be a need in the art for improved biologically active BPI protein product preparations, particularly those with enhanced stability, homogeneity and/or in vivo biological activity.
SUMMAII~Y OF THE INVENTION
The present invention provides novel biologically active BPI deletion analogs and preparations thereof characterized by enhanced stability and homogeneity, including for example, resistance to dimerization and cysteine adduct formation and reduced amino-terminal and carboxy-terminal heterogeneity of the recombinant product, as well as by enhanced in vivo biological activity, properties which render it highly suitable for therapeutic and diagnostic uses. Novel BPI
deletion analogs are the expression product of DNA encoding amino acid residues 10 through 193 of mature human BPI (SEQ ID NO: 2), in which the cysteine at position 132 has been replaced with a different amino acid, preferably a non-polar amino acid such as serine or alanine. In a preferred embodiment, designated _g_ "rBPI(10-193)C132A" or "rBPI(10-193)ala'32," the cysteine at position 132 is replaced with an alanine.
The invention further provides novel purified and isolated polynucleotide sequences (e. g. , DNA or RNA) encoding these BPI protein products; materials and methods for their recombinant production, including vectors and host cells comprising the DNA; improved stable pharmaceutical compositions comprising these BPI protein products; and improved treatment methods using these compositions, either alone or concurrently administered with other therapeutic agents. Also contemplated is the use of the BPI deletion analogs of the invention in manufacture of a medicament for treating a subject that would benefit from administration of BPI protein product.
Numerous additional aspects and advantages of the invention will become apparent to those skilled in the art upon considering the following detailed description of the invention, which describes the presently preferred embodiments thereof.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 depicts the elevation in blood pressure, measured as area under the curve (AUC) occurring after administration of either rBPI(10-193)C132A
or rBPI2, .
DETAILED DESCRIPTION OF THE 1N~VENTION
The present invention provides novel BPI deletion analogs consisting of amino acid residues 10 through 193 of mature human BPI (set forth in SEQ ID
NO: 2) wherein the cysteine residue at BPI amino acid position 132 is replaced by another amino acid, preferably a non-polar amino acid such as serine or alanine.
A preferred embodiment, in which the cysteine at position 132 is replaced with an alanine, has been designated rBPI(10-193)C132A or rBPI(10-193)ala'32_ WO 99!66044 PCT/US99/13860 The BPI protein product rBPI21 is the expression product of DNA
encoding amino acid residues 1 to 193 of mature human BPI wherein the cysteine at residue number 132 is substituted with alanine, described in U. S . Patent No.
5,420,019. Changes in the fermentation processes used to produce rBPIZI by recombinant methods that achieved higher cell densities and higher rBPI2, titers also resulted in an apparent increase in amino-terminal heterogeneity of the purified praluct. In some fermentation nms, up to about 20 ~ of the purified product was observed to be a species with amino acids 10-193 of BPI, rather than the encoded 1-193 amino acids. SDS-PAGE gels of 500-liter fermentor samples over the course of a fermentation nm showed that this 10-193 species appeared in the last 2-3 days of the run, with the greatest amount appearing on the day of harvest. Further investigation revealed that incubation of rBPI2, with a CHO-Kl cell homogenate yielded a digested product, suggesting that protease activity associated with the cells was involved. To simulate protease activity in a controlled manner, rBPI2, was incubated with aminopeptidase M and elastase. The rBPI21 was resistant to aminopeptidase M digestion, but elastase rapidly converted the rBPI2, into 40 BPI(8-193) and 60~ BPI(10-193).
As described herein, stable homogeneous preparations of rBPI(10-193)C132A were produced proteolytically and by recombinant methods. The protein was purified and was tested for biological activity. Experiments were performed to compare rBPI(10-193)C132A to rBPIzI in several in vitro biological assays, two different animal efficacy models and in pharmacoldnetic and toxicology studies. As described in Examples 5-7, rBPI(10-193)C132A and rBPI21 had similar in vitro activities when compared in radial diffusion and broth microdilution bactericidal assays with Escherichia coli J5, a radial diffusion assay with an L-form of Staphylococcus aureus, a competition binding assay with E. coli JS LPS, and in LPS neutralization assays with RAW and THPl cells. Additional experiments described in Example 5 showed that rBPI(10-193)C132A appeared to be approximately twice as potent as rBPIZ, in an LPS binding assay using rate nephelometry. As described in Example 8, purified rBPI(10-193)C132A and rBPI21 had similar toxicity profiles in a GLP toxicology study in rats at doses up to 120 mg/kg/day for three days and similar pharmacokinetics in rats at a dose of mg/kg. Experiments described in Example 8 also showed that in a mouse endotoxin challenge model, rBPI(10-193)C132A appeared to be at least two-fold more potent than rBPI21 in two studies whereas in a mouse model of lethal bacteremia, rBPI(10-193)C132A and rBPI2, were similarly potent. In additional in vivo experiments in conscious rats, doses of 40 and 50 mg/kg of infused rBPI2, caused significant transient decreases in blood pressure relative to the vehicle control, while the same doses of rBPI(10-193)C132A did not result in a statistically significant transient decrease in blood pressure relative to control. Thus, infusion of rBPI(10-193}C132A appears to provide a reduction in an adverse effect in blood pressure compared with infusion of rBPI2,.
The invention further contemplates fusion of rBPI(10-193)CI32A
with at least a portion of at least one other polypeptide. Examples of such hybrid fusion proteins are described in U.S. Patent No. 5,643,570 and corresponding International Publication No. WO 93/23434 (PCT/US93/04754), which are all incorporated herein by reference and include hybrid fusion proteins comprising, at the amino-terniinal end, a BPI protein or a biologically active fragment thereof and, at the carboxy-terminal end, at least one constant domain of an immunoglobulin heavy chain or allelic variant thereof.
The invention additionally contemplates purified and isolated polynucleotide sequences (e. g. , DNA or RNA) encoding the novel BPI deletion analogs or fusion proteins of the present invention; expression vectors containing such polynucleotides, preferably operatively linked to an endogenous or heterologous expression control sequence; prokaryotic or eukaryotic host cells stably transfected or transformed with a DNA or vector of the present invention;
and methods for the recombinant production of the novel deletion analog BPI
protein products of the present invention, e. g. , methods in which a host cell is grown in a suitable nutrient medium and the deletion analog BPI protein product is isolated from the cell or the medium. Such polynucleotide sequences or vectors may optionally encode the 27-amino acid BPI leader sequence and the mouse light chain polyadenylation signal.
The recombinantly produced novel BPI deletion analog of the present invention may be produced according to the methods described in U.S. Patent No.
5,439,807 and corresponding International Publication No. WO 93/23540 (PCT/US93/04752), which are all incorporated herein by reference. U.S. Patent No. 5,439,807 discloses methods for the purification of recombinant BPI
protein products expressed in and secreted from genetically transfected mammalian host cells in culture, and discloses how one may produce large quantities of recombinant BPI products suitable for incorporation into stable, homogeneous pharmaceutical preparations.
The present invention further provides improved stable pharmaceutical compositions comprising the novel BPI deletion analogs and improved treatrnent methods using these compositions, either alone or concurrently administered with other therapeutic agents. It is contemplated that such compositions may be utilized in any of the therapeutic uses known for BPI
protein products, including those discussed supra.
The administration of BPI protein products in general, including BPI
deletion analogs, is preferably accomplished with a pharmaceutical composition comprising a BPI protein product and a pharmaceutically acceptable diluent, adjuvant, or carrier. The BPI protein product may be administered without or in conjunction with known surfactants, other chemotherapeutic agents or additional known anti-chlamydial agents. A stable pharmaceutical composition containing BPI protein products (e.g., rBPI~) comprises the BPI protein product at a concentration of 1 mg/ml in citrate buffered saline (5 or 20 mM citrate, 150 mM
NaCI, pH 5.0) comprising 0.1 °& by weight of poloxamer 188 (Pluronic F-68, BASF, Parsippany, Nn and 0.002 ~ by weight of polysorbate 80 ('lween 80, ICI

Americas Inc., Wilmington, DE or JT Baker, Phillipsburg, NJ). Another stable pharmaceutical composition containing BPI protein products (e.g., rBPI2,) comprises the BPI protein product at a concentration of 2 mg/ml in 5 mM
citrate, 150 mM NaCI, 0.2 9& poloxamer 188 and 0.002 % polysorbate 80. Such preferred combinations are described in U.S. Patent Nos. 5,488,034 and 5,696,090 and corresponding International Publication No. WO 94/17819 (PCT/US94/01239), the disclosures of all of which are incorporated herein by reference. As described in U.S. Application Serial No. 08/586,133 filed January 12, 1996, which is in turn a continuation-in-part of U.S. Application Serial No. 08/530,599 filed September 19, 1995, which is in turn a continuation-in-part of U.S. Application Serial No.
08/372,104 filed January 13, 1995, and corresponding International Publication No. W096/21436 (PCT/US96/01095), all of which are incorporated herein by reference, other poloxamer formulations of BPI protein products with enhanced activity may be utilized.
Therapeutic compositions comprising BPI protein product may be administered systemically or topically. Systemic routes of administration include oral and parenteral routes, including intravenous, intramuscular or subcutaneous injection (including into a depot for long-term release), intraocular and retrobulbar, intrathecal, intraperitoneal (e.g. by intraperitoneal lavage), intrapulmonary (using powdered drug, or an aerosolized or nebulized drug solution), or transdermal.
Improved aerosolized formulations are described in co-owned, co-pending U.S.
Application Serial No. 08/962,217 filed October 31, 1997 and corresponding International Publication No. WO 98/19694 (PCT/US97/19850), which are both incorporated herein by reference.
When given parenterally, BPI protein product compositions are generally injected in doses ranging from 1 ~cg/kg to 100 mg/kg per day, preferably at doses ranging from 0.1 mg/kg to 20 mg/kg per day, more preferably at doses ranging from 1 to 20 mg/kg/day and most preferably at doses ranging from 2 to mg/kg/day. The treatment may continue by continuous infusion or intermittent injection or infusion, at the same, reduced or increased dose per day for, e.g., 1 to 3 days, and additionally as determined by the treating physician. When administered intravenously, BPI pmtein products are preferably administered by an initial brief infusion followed by a continuous infusion. The preferred intravenous regimen is a 1 to 20 mg/kg brief intravenous infusion of BPI protein product followed by a continuous intravenous infusion at a dose of 1 to 20 mg/kg/day, continuing for up to one week. A particularly preferred intravenous dosing regimen is a 1 to 4 mg/kg initial brief intravenous infusion followed by a continuous intravenous infusion at a dose of 1 to 4 mg/kg/day, continuing for up to 72 hours.
Topical routes include administration in the form of salves, creams, jellies, ophthalmic drops or ointments (as described in co-owned, co-pending U.S.
Application Serial No. 08/557,289 filed November 14, 1995 and U.S. Patent No.
5,686,414 and corresponding International Publication Nos. WO 97/17990 {PCT/US96/18632) and WO 97/17989 (PCT/US96/I8416), all of which are incorporated herein by reference), ear drops, suppositories, irngation fluids (for, e. g. , irrigation of wounds) or medicated shampoos. For example, for topical administration in drop form, about 10 to 200 ~,L of a BPI protein product composition may be applied one or more times per day as determined by the treating physician.
Those skilled in the art can readily optimize effective dosages and administration regimens for therapeutic compositions comprising BPI protein product, as determined by good medical practice and the clinical condition of the individual patient.
Other aspects and advantages of the present invention will be understood upon consideration of the following illustrative examples. Example addn'sses the construction of an expression vector, pING1742, encoding rBPI(10-193)C132A. Example 2 addresses transformation of CHO cells with pING1742 and selection of the highest producing clones secreting rBPI(10-193)C132A.

WO 99/66044 PCTIpS99/13860 Example 3 addresses the production and purification of rBPI(10-193)C132A in 2-L
and 500-L fermenters. Example 4 addresses the biochemical characterization of rBPI(10-193)C132A and rBPI2,. Examples 5, 6 and 7 respectively address the in vitro LPS-binding activity in a competition binding assay and in an assay measuring rate of complex formation using rate nephelometry, bactericidal activity, and LPS
neutralization activity of rBPI(10-193)C132A as compared to rBPI2,. Example 8 addresses the in vivo activity of rBPI(10-193)C132A.

Construction of Expression Vector pING1742 The rBPI(10-193)C132A expression vector, pINGI742, was constructed as follows. The expression vector pING4155 was first constricted by ligating a BamHl BsaT fragment containing the neo gene from pING3174 with a BsaT Xhol fragment containing the CMV promoter and rBPlzl gene from pING4144 and an Xhol BamHl fragment containing the mouse (k ppa) light chain 3' untranslated region from pING4537 (pING3174, pING4144 and pING4537 are described in U.S. Patent No. 5,420,019, incorporated by reference). 'The resulting pING4155 vector contains the gene encoding rBPIzI fused to the human IgG
enhancer, the human CMV promoter and the mouse (kappa) light chain 3' untranslated region. It also contains the neo gene encoding neomycin phosphotransferase, for selection of transfectants resistant to the antibiotic Geneticin~ (G418).
The vector pING1732 was produced by deleting the 0.7 kbp Hindlll - HindTll fiagment of pING4155 containing the human Ig enhancer. Then, the 27 nucleotides encoding amino acids 1 through 9 of the mature portion of rBPI2, were deleted from pING1732 by overlap PCR mutagenesis using the following primers:
Primer 1: 5'-CTGCTCTAAAAGCTGCTGCAG-3' (SEQ ID NO: 3) Primer 2: 5'-CCAGGCCCTTCTGGGAGGCCGCTGTCACGGCGG-3' (SEQ ID
NO: 4) Primer 3: 5'-GCCGTGACAGCGGCCTCCCAGAAGGGCCTGGAC-3' (SEQ ID
NO: 5) Primer 4: 5'-CTGGGAACTGGGAAGCTG-3' (SEQ ID NO: 6) Overlapping complementary primers 2 and 3 incorporated the 27 by deletion of nucleotides encoding amino acids 1 through 9, while primers 1 and 4 encoded nucleotides immediately upstream and downstcuam, respectively, of unique Sall and EcoRl sites in pING1732. First, fragments were obtained by PCR amplification using the combination of oligonucleotide primers 1 and 3, and primers 2 and 4.
After these individual fiagments were obtained, they were annealed, extended and re-amplified using primers 1 and 4. This amplified fragment was then digested with SaU and EcoRl and cloned into SaU EcoRl digested pING 1732 to generate the plasmid pING1742.
To confirm that no mutations had occurred during PCR, the Sah EcoRl region from pING1742 was sequenced. No changes were observed in the mature coding region for BPI. However, a two base-pair change (ACC - > GCT) was found in DNA encoding the signal sequence, which resulted in the conversion of a Thr to an Ala at amino acid position -6 relative to the start of the mature protein sequence.

Transformation of CHO Cells with pING1742 CHO-K1 cells (American Type Culture Collection (ATCC) Accession No. CCL61) were adapted to growth in seivm-free Ex-Cell 301 medium as follows. CHO-Kl cells grown in Ham's F12 medium were trypsinized, centrifuged and resuspended in Ex-Cell 301 medium. Cells were grown in a 125-ml flask at 100 rpm and passaged every two to three days in either a 125-ml or 250-ml flask.
These Ex-Cell 301-adapted CHO cells were transfected by electroporation with pING1742. Prior to transfection, pING1742 was digested with Notl, which linearizes the plasmid. Following a 48-hour recovery, cells were plated at approximately 104 cells/well into 96-well plates containing Ex-Cell medium supplemented with 0.6 mg/mL 6418 (Life Technologies, Gaithersburg, MD). At approximately 2 weeks, supernatants from approximately 250 wells containing single colonies were screened by ELISA for the presence of BPI-reactive protein using an anti-BPI monoclonal antibody.
Fifteen clones having the highest expression levels were transferred to 24-well plates containing Ex-Cell 301 medium. To screen for productivity, the cells were grown in 24-well plates containing Ex-Cell medium supplemented with 2 ~ FBS and 40 ~cL sterile S-Sepharose beads for 10 days, after which the beads were removed, washed with low salt buffer (0.1 M NaCl in 10 mM Na acetate, pH
4.0) and the BPI eluted with 1.5 M NaCI in the same buffer. The levels of secreted rBPI(10-193)C132A were determined by ELISA. Western blot analysis of eluates run on a 12 % non-reducing SDS gel revealed a prominent band which migrated slightly faster than rBPIzI.
The top eight producers were transferred to sterile 125 mL
Erlenmeyer flasks and grown in Ex-Cell medium. These cells were evaluated again for productivity by growing them in flasks containing Ex-Cell 301 medium supplemented with 2~ FBS and 1 ~ (V/~ sterile S-sepharose beads. The rBPI(10-193)C132A was eluted from the S-Sepharose beads that had been incorporated in the culture medium and the levels of rBPI(10-193)C132A determined by HPLC.
Clone 139, which was among the highest producers, was chosen for further growth and product production.

Production and Purification of rBPI(10-193)C132A
Large quantities of rBPI(10-193)C132A were produced for characterization by growing Clone 139 cells in 2-liter research fermenters (Biolafitte, St. Germain en Laye, France) and then in a 500 liter ABEL
fermenter (ABEL, Allentown, PA). Protein product obtained from the 2-liter fermenters was used for the in vitro studies described below, while product obtained from the liter fermenter was used for animal toxicology and efficacy studies.
A. Growth in_ Two-Liter FermentPrs Clone 139 cells were passaged in spinner flasks of increasing volumes containing Ex-Cell medium supplemented with 1 °b FBS until sufficient volume and cell density was achieved to inoculate the 2 liter bioreactors at approximately 2 X 105 cells/mL. Cells were grown in three 2-liter fermenters in Ex-Cell medium supplemented with 1 °b FBS, at 37 °C, pH
7.2, 150 rpm with dissolved oxygen maintained at 5-10 °6 . Large sterile SP-Sepharose beads (Pharmacia and Upjohn, Piscataway, N~ were added at 1.5 °b (V/~. 'The initial glucose level was approximately 3.5 g/L and glucose was pulsed daily to 3 g/L
during the course of the run. The fermentation was terminated at 238 hours, at which time the cell viabilities were from 63 ~ , 80 ~ and 84 ~ .
Following fermentation, the beads from each fermenter were harvested, allowed to settle, and washed several times with 10 mM Na phosphate/O.15M NaCI, pH 7.0, to remove cellular components and weakly bound impurities firm the beads. The washed beads were packed into a column, washed with 10 mM Na phosphate, 0.25 M NaCI, pH 7.0, and eluted with the same buffer containing 0.8 M NaCI, 5 mM glycine. The eluate was then diluted with three volumes of sterile water for injection (VVFI), loaded onto a CM-spherodex column (Sepracor, Marlborough, MA) and washed with 10 mM Na phosphate, 0.25 M
NaCI, pH 7.0, followed by 20 mM Na acetate, 0.2 M NaCl, pH 4.0, followed by 20 mM Na acetate, 0.3 M NaCI, pH 4.0, and sample was eluted at 1.0 M NaCI in the same buffer. Following concentration on a Centricon membrane with a 10,000 MW cutoff (Amicon, Beverly, MA), the eluate from the CM column was loaded onto a Sephacryl S-100 column (Pharmacia and Upjohn) equilibrated with 5 mM
Na citrate, 0.15 M NaCI, pH 5Ø Fractions containing rBPI(10-193)C132A

identified by absorbance at 280nm were pooled, concentrated on an Amicon filter to 1.9 mg/mL and formulated with 0.002 ~ polyso~ate 80 (JT Baker, Phillipsburg, NJ), 0.2 k poioxamer 188 (Pluronic F-68, BASF, Parsippany, NJ). The final preparation was filter sterilized using a 0.2 ~,m filter.
B. Growth i_n 500-Liter FermentPr Clone 139 cells were passaged in fetuin-free Ex-Cell medium with 1 °6 FBS in a series of spinner flasks of increasing volumes to provide inoculum for the 35L Bellco spinner flask (Bellco Glass, Vineland, NJ), which in turn provided the inoculum for the 500 liter ABEC fenmenter. Cells were grown in complete Ex-Cell medium without fetuin but supplemented with 1 % FBS, additional glucose (to 10 g/L) and glutamine (to 10 mM). The fermenter was operated in a fed-batch mode with one 0.5 ~ Primatone RL supplement pulse and one glucose/glutamine pulse added during the run. Five to six liters of large SP-Sepharose beads were added 24 hours after the 500 liter fermenter was inoculated. The pH was controlled manually with 10 % sodium bicarbonate to pH 7.0, oxygen was controlled at 5 ~ and temperature at 37°C. Agitation was maintained at 25 rpm with two three-blade paddle impellers. The fermentation run was terminated at hours, at which time the cell viability was 90 Rb .
As described above for the 2-liter fermentation, the beads were allowed to settle following fermentation and then washed several times with low salt (O.1M) phosphate buffer. The steps for this purification were similar to those described above for the 2-liter samples except that a pH 3.0 viral inactivation step was included after elution from the S-Sepharose beads and a second CM-spherodex column was included as a concentration step. For the second CM column, the eluate was diluted with three volumes of WFT, the pH adjusted to 5.0, the column was equilibrated and washed with 20 mM Na acetate, 0.3 M NaCI, pH 5.0 and the sample was eluted at 1.0 M NaCI in the same buffer. The rBPI(10-193)C132A was eluted from the Sephacryl S-100 column in 5 mM Na citrate, 0.15 M NaCI, pH

5.0, adjusted to 2 mg/mL, and filtered through a 0.2 ~cm filter. The rBPI(10-193)C 132A was then formulated with 0.002 ~ polysorbate 80, 0.2 ~ poloxamer 188, sterile filtered, and filled into 10 mL Type I glass serum vials.

Biochemical Characterization of rBPI(10-193)C132A
A. Protein_ from th_e 2-Liter Fermenta_tions The purified rBPI(10-193)C132A product from Example 3 was observed to be a single band that migrated slightly faster on SDS
polyacrylamide gel electrophoresis (SDS-PAGL~ than the rBPI2, band, consistent with the deletion of nine N-terminal amino acids from rBPI2l. Sequence analysis demonstrated that the rBPI(10-193)C132A contained the predicted N-terminal sequence of SQKGLDYASQQGTAALQKEL. On mass spectroscopy analysis (ESI-MS) two components were observed, one with a mass of 20,470 daltons, which was consistent with the predicted mass of 20,472 daltons for rBPI(10-193)C132A, and a second with a mass of 20,255 daltons, consistent with the predicted mass of 20,258 daltons for rBPI(10-191). The ion-exchange HPLC profiles (Hewlett-Packard, Model 1050, Palo Alto, CA) of rBPI(10-193)C132A and rBPI21 both exhibited single peaks with similar retention times.
B. Protein_ from the X500-Liter Fermentation On SDS-PAGE, the rBPI(IO-I93)C132A was a single band that migrated slightly faster than the rBPizl band. On mass spectroscopy, there was a major component with a mass of 20,471 daltons, which is consistent with the pn~dicted mass of 20,474 Da for rBPI(10-193)C132A), and two minor components with a mass of 20,668 daltons, which is consistent with addition of N-Acetylhexosamine (predicted mass 20,677 daltons) and a mass of 20,843 daltons, which is consistent with addition of N-Acetylhexosamine plus hexose (predicted mass 20,839 daltons). A similar component with added N-Acetylhexosamine is routinely observed during production of rBPI2, .
On reverse phase HPLC (Shimadzu, Kyoto, Japan) both the rBPI(10-193)C132A and rBPI2, eluted as one major peak and one minor peak. However, the rBPI(10-193)C132A peaks eluted slightly earlier than the corresponding rBPI2, peaks in the control. The minor peak in the rBPI(10-193)C132A profile most likely represents the glycosylated forms identified in the mass spectrum. The ion-exchange HPLC profiles of rBPI(10-193)C132A and rBPI2l both exhibited single peaks with similar retention times.
Tryptic mapping analysis was performed according to conventional methods. Acetone precipitated rBPiz, or rBPI(10-193)C 132A was first treated with dithiothreitol (DTT~ followed by iodoacetamide and then with trypsin. The trypsin-treated product was analyzed by HPLC (Beckman Model 126) with a C 18 column (Beckman Ulhasphere). In rBPiz,, there are two N-terminal tryptic fragments (Tl and Ala-Tl) which result from imprecise cleavage of the leader sequence. As pmdicted, the tryptic map of the rBPI(10-193)C132A was similar to rBPIz, except that the N-terminal fragments were missing.

In Vitm LPS-Binding Activity of rBPI(10-193)C132A
A. In a o rye i ion B'n ing A
The ability of purified rBPI(10-193)C132A produced according to Example 3A and rBPTzI to compete with labeled rBPIZ, for binding to LPS was evaluated in a competition binding assay. Briefly, a fixed concentration (0.5 nM) of'uI-labeled rBPIz, was mixed with unlabeled rBPI2, or rBPI(10-193)C132A at dilutions ranging from 5 ~M to 0.01 nM in DMEM containing HEPES buffer and bovine senim albumin (BSA) [U.S. Biochemicals, Cleveland, OHj and 100 ~,L of the mixture was added to Immulon-II plate wells pre-coated with 2.5 ~,g/mL E.
coli JS LPS (Calbiochem, San Diego, CA). The plates were incubated at 4°C for 5 hours and washed 3 times with the DMLM medium. 75 ~,L of 0.1 N NaOH was added and the bound "~I-rBPI2, was removed and counted. The results demonstrated that both proteins competed similarly with radiolabeled rBPI2,.
B.
The LPS binding activity of rBPI(10-193)C132A was compared to rBPiz, using rate nephelometry. This approach for evaluating rBPI2, binding to LPS
measures the rate of increase of light scattering as a result of LPS-BPI
protein product complex formation in solution. All of the experiments were performed with a Beckman Array 360 Rate Nephelometer which automatically mixes samples, measures light scattering and performs rate calculations.
Prior experiments using this approach examined optimal LPS species and concentration, assay specificity, assay reproducibility and correlation of assay results to bactericidal assays. It was observed that E. coli JS LPS and lipid A formed complexes with rBPI2, that could be measured in the nephelometer, but E. coli 0111:84 LPS did not form measurable complexes. Based on results of these studies, E. coli JS LPS was chosen for use at a concentration (in the flow cell) of 49.4 to 61.7 /.cg/ml, depending on the LPS lot, in combination with rBPI2, concentrations (in the flow cell) from 5 to 30 ~cg/ml. The optimal rBPI2, concentration range, which must be determined for each LPS lot, was from about 15 to 25 ,ug/ml which represented the most linear portion of the curve. The optimal range for the aggregation rate (RT) values was from 700 to 2000. Lower concentrations of rBPIz, were needed to achieve the same aggregation rate values when the formulation buffer was changed to include PLURONIC P103 or when the NaCI concentration was increased. The addition of either recombinant lipopolysaccharide binding protein (rLBPso) which binds to LPS, or heparin which binds to BPI protein products, inhibited the formation of rBPI2, LPS aggregates, demonstrating the specificity of the interaction. Assay reproducibility was confirmed by testing multiple lots of BPI and testing the same lot of rBPI2, multiple times. Nephelometric analysis of rBPI2, samples that had been partially inactivated by treatment at 45 °C for one week correlated well with those from broth microdilution bactericidal assays with E. coli JS cells.
Nephelometry experiments comparing rBPI(10-193)C132A and rBPhl were carried out as follows. Sonicated LPS [E. coli JS LPS Lot No. 30119B from List Biochemicals] and either rBPI(IO-193)C132A or rBPI21 [both of which were formulated in 0.2% PLURONIC F68 (poloxamer I88), 0.002% TWEEN 80 (polysorbate 80), 5 mM citrate, pH 5.0, 150 mM NaCI] were diluted directly into a PBS buffer (supplemented with PEG) supplied by Beckman. The LPS concentration was fixed while the BPI protein product concentration varied within each experiment.
Two concentrations of LPS were tested: 24.7 and 49.4 ~cg/ml LPS. Each reaction was initiated by addition of 600 ~1 of the PB S-PEG buffer to the flow cell followed by 42 ~.cl of the BPI protein product dilution. After a baseline was established, 42 ,ul of the E. coli JS LPS solution was added. After addition of the last component, the nephelometer measures the rate of complex formation based on the extent of light scatter. The data were analyzed by dividing the RT values for each test sample containing a given BPI protein product concentrations by the correspanding RT
values for the standard to generate a percent of control value. For each BPI
protein product concentration tested, the maximum aggregation rate was determined and a curve generated. Only points to the left of the maximum value (point of equivalence) were used for comparative analysis of various BPI protein product samples. The relative activity of samples can be measured by comparing the RT values for test and standard lots in the linear region of the curves. Either a point to point or curve fit approach can be used.
In addition to testing purified rBPI(10-193)C132A and purified rBPI2i [which contains about 7.8% rBPI(10-193)C132A], an equal mixture of these proteins as well as a rBPIz1 preparation with 16% rBPI(10-193}C132A was evaluated (at 49.4 ,ug/ml LPS only). The results demonstrated that at 49.4 ,ug/ml LPS, rBPI(10-193)C132A achieved aggregation rates similar to that of rBPI21 at an approximately 25% lower concentration. The rBPI(10-193)C132A also achieved a higher maximum aggregation rate than that of rBPIz1 at both 24.7 and 49.4 ~cg/ml LPS. An equal mix ofthe two molecules yielded a curve that ran between rBPI2t and rBPI(10-193) while the rBPI21 lots with 7.8% and 16% 10-193 behaved in an identical manner to each other. A point to point analysis of the results (LPS at 49.4 ~cg/ml) revealed that the rBPI(10-193) was approximately twice as potent as rBPIz, in this assay.

In Vitm Bactericidal Activity of rBPI(10-193)C132A
All of the assays in this example were conducted with rBPI(10-193)C132A produced in the 2-liter fermenters according to Example 3A.
A. Pffect on E. coli 'n a Ra ial ion Accav This radial diffusion assay compared the bactericidal effect of purified rBPI(10-193)C132A and rBPTz, on E. coli J5, which is a UDP-galactose-epimerase "rough" mutant of the smooth strain E. coli O11B4, and is relatively sensitive to rBPI2l. E. coli J5 cells (Mannion et al. , J. Clin. Invest. , 85:853-860 (1990); List Biological Laboratories, Campbell, CA) were grown to exponential phase, centrifuged and washed twice in 10 mM Na phosphate, pH 7.4, and added at a final concentration of approximately 1 X 106 CFU/ml to molten agarose supplemented with 3 ~ Trypticase Soy Broth (TSB, DIFCO Laboratories, Detroit, MI), 10 mM Na phosphate. Wells of 3 mm diameter were prepared in the hardened agarose and 5 ~uL of serially diluted rBPI21 or rBPI(10-193)C132A was added to the wells. The plates were incubated at 37°C for 3 hours to allow diffusion to occur, and then a molten agarose overlay containing 6 % TSB was added. The plates were incubated overnight at 37°C and the net area of inhibition was plotted vs. concentration. The results demonstrated that rBPI(10-193)C132A
and rBPI21 behaved in a similar manner in this assay.

B. Effect on S. Aureus L-Form in_ a Radial Diffusion Assav This radial diffusion assay compared the bactericidal effect of purified rBPI(10-193)C132A and rBPIzI on the gram-positive bacteria S. aureus grown as L-forms without their cell walls. As described in U.S. Patent No.
5,578,572, incorporated herein by reference, S. aureus L-form cells were grown to log phase in heart infusion (I~ broth supplemented with 3.5 % NaCI, 10 mM
CaCiz and 1000 U/mL penicillin G. The cells were diluted to approximately either 5 X 104 or 5 X 105 cells/mL in molten 0. 8 °.& agarose containing the NaCI-supplemented HI medium, and 8 ml of the cell-agarose suspension was poured into 10 cm plates. Wells of 3 mm diameter were prepared, and 5 wL of serially diluted rBPTzI or rBPI(10-193)C132A was added to the wells. The plates were incubated at 37°C for 24 hours and the net area of inhibition was plotted vs.
concentration.
The results demonstrated that both rBPTzI and rBPI(10-193)C132A inhibited growth of the S. aureus L-forms, at cell densities of about 5 X 104 and 5 X 105, in a similar fashion in this assay.
C. Effect on E. coli JS in a Broth Microdilution Assav This broth microdilution assay compared the bactericidal effect of purified rBPI(10-193)C132A and rBPlz~ on E. coli J5. E. coli JS cells were grown overnight in tryptone yeast extract (TYE) broth and then to logarithmic phase in TEA medium as previously described in Horwitz et al., Infect. Immun., 63:522-(1995). The cells were inoculated at approximately 1 X 104 and 1 X 105 cells/mL
in heart infusion (Hn broth, and 95 lcL was added to 96 well plates. Five ~cL
of various dilutions of rBPI(10-193)C132A or rBPIzI, prepared in formulation buffer, was added to each well and the plates were incubated at 37°C for 24 hours. The results demonstrated that rBPI(10-193)C132A and rBPI2, have similar activities in these assays.

In Vitro LPS Neutralization Activity of rBPI(10-193)C132A
The assay in section A of this example was conducted with rBPI(10-193)C132A produced in the 2-liter fenmenters according to Example 3A, while the assay in section B of this example was conducted with rBPI(10-193)C132A
produced in the 500-liter fermenter according to Example 3B.
A. Activit3r in_ a RA-W Cell Pro iferation a ~v, The RAW cell proliferation assay was used to compare the in vitro LPS neutralization activity of rBPI2, and rBPI(10-193)C132A. In this assay, the LPS inhibits the proliferation of RAW cells, and rBPiz~ neutralizes this effect of LPS.
Mouse RAW 264.7 cells (ATCC Accession No. T1B71), maintained in RPMI 1640 medium (GIBCO), supplemented with 10 mM HEPES buffer (pH
7.4), 2 mM L-glutamine, penicillin (100 U/mL), streptomycin (100 ~cg/mL), 0.075 ~ sodium bicarbonate, 0.15M 2-mercaptoethanol and 10 ~ fetal bovine serum (Hyclone, Inc., Logan, UT}, were first induced by incubation in the presence of 50 U/mL recombinant mouse y-interferon (Genzyme, Cambridge, MA) for 24 hours prior to assay. Induced cells were then mechanically collected and centrifuged at 500 x g at 4°C and then resuspended in 50 mL RPMI 1640 medium (without supplements), re-centrifuged and again resuspended in RPMI 1640 medium (without supplements). The cells were counted, their concentration adjusted to 2 x 105 cells/mL and 100 ~L aliquots were added to each well of a well plate.
The cells wem then incubated for about 15 hours with E. coli 0113 LPS (Control Standard, Assoc. of Cape Code, Woods Hole, MA), which was added in 100 ~,L/well aliquots at a concentration of 1 ng/mL in serum-free RPMI
1640 medium (this concentration being the result of titration experiments in which LPS concentration was varied between 50 pg/mL and 100 ng/mL). This incubation was performed in the absence or presence of rBPI2, or rBPI(10-193)C132A in varying concentrations between 25 ng/mL and 50 tcg/mL. Recombinant human rBPIzI, also designated rBPhlocys, which is rBPI 1-193 with alanine substituted at position 132 for cysteine [see co-owned U.S. Patent No. 5,420,019], was used as a positive control at a concentration of 1 ~.g/mL. Cell proliferation was quantitatively measured by the addition of 1 tcCi/well [3H]-thymidine 5 hours after the time of initiation of the assay. After the 15-hour incubation, labeled cells were harvested onto glass fiber filters with a cell harvester (Inotech Biosystems, 384, Sample Processing and Filter Counting System, Lansing, ML). The LPS-mediated inhibition of RAW 264.7 cell proliferation is dependent on the presence of LBP, as added to the reaction mixture either as a component of serum or as recombinant LBP (at a concentration of 1 tcglmL.
In these experiments, both rBPlzl and rBPI(10-193)C132A similarly inhibited the LPS-mediated inhibition of RAW cell proliferation.
B. Activi ~ in a TNF I hibi ion A cad A tumor necrosis factor ~ inhibition assay was used to compare the in vitro LPS neutralization activity of rBPI21 and the rBPI(10-193)C132A.
In this assay, the LPS, in combination with purified LBP (or serum containing LBP) stimulates synthesis of TNF by THP-1 cells (a human monocyte cell line), and rBPIz~ neutralizes this effect of LPS.
THP.1 cells (ATCC Accession No. TIB-202) were maintained in RPMI (GibcoBRL, Gaithersburg, MD) with 10 ~ FBS and were cultured in RPMI
with 10% FBS plus 50 ng/ml 1,25 dihydroxy vitamin D (BIOMOL Research Laboratories Inc. Plymouth Meeting, PA) for three days prior to treatment with LPS to induce CD14 expression. Before inducing with LPS, cells were washed three times with RPMI and suspended in either RPMI with 10 % FBS or in serum free medium [RPMI supplemented with 19& HB101 (Irvine Scientific, Santa Ana, CA)). Expression of TNF was induced with 1 ng/ml E. coli 0128 LPS (Sigma, St.
Louis, MO) in 96 well plates with approximately S x 104 cells per well. Plates were incubated for three hours at 37°C, 5 °b C02, then an aliquot of the supernatant liquid was removed and assayed for TNF by the WEHI 164 toxicity assay, using CellTiter 96T"" AQ (Promega Corp., Madison, WI) to monitor cell viability.
Recombinant human TNFa (Genzyme Diagnostics, Cambridge, MA) was used as a positive standard. Both rBPI21 and rBPI(10-193)C132A similarly inhibited LPS-induced stimulation of TNF synthesis.

In Vivo Biological Activity of rBPI(10-193)C132A
The in vivo assays described below were performed using the purified rBPI(10-193)C132A produced in the 500-liter fermenter according to Example 3B.
A. To i i r Stud) 'n a c Toxicity profiles of rBPI21 and rBPI(10-193)C132A were compared in rats. In this study, groups of six male and six female Sprague-Dawley rats received either vehicle control (formulation buffer), low (50 mg/kg/day) or high (120 mg/kg/day) doses of either rBPI21 or rBPI(10-193)C132A. Doses were administered by continuous intravenous infusion via an indwelling femoral catheter for three consecutive days at an infusion rate of 4.2 mLJkg/hour (100 mLkg/day).
Clinical observations were recorded at least twice daily and body weights were recorded daily. Blood and urine samples were collected near termination for hematology, clinical chemistry and urinalysis assessments. At termination, organs were weighed and tissues collected by histopathological examination. There were no deaths or significant test article-related effects. The data indicated similar toxicity profiles for rBPIz, and the rBPI(10-193)C132A when given by continuous infusion.

B. p~~~
The pharmacoldnetics of rBPIzI and rBPI(10-193)C132A at 2 mg/kg were investigated in rats. The plasma clearances of rBPlz1 and rBPI(10-193)C132A
were well described by a tri-exponential pharmacokinetic disposition function.
No statistical differences in the pharmacokinetic parameters among the rBPI
products were determined (non parametric Wicoxon rank test, p < 0.05). Most of the administered drug ( > 96 ~) was cleared with an alpha phase half life of 0.2-0.4 minutes and a beta half life of 3.9-4.3 minutes, while the remainder was cleared during the gamma phase with a half life of 27-33 minutes. The volume of distribution of the central compartment (Vc) was 41-45 mL/kg, and the clearance rate (CL) was 24-30 mL/minlkg. The steady state volume of distribution was 152-184 mL/kg.
C. Fix In Mouse ndoto~ in C allenee Two separate studies were conducted to examine relative potencies of rBPI21 and rBPI(10-193)C132A in a mouse model of lethal endotoxemia generally according to Ammons et al. , in "Novel Therapeutic Strategies in the Treatment of Sepsis," Morrison and Ryan, eds., Marvel Dekker, New York (1996), pages 55-69. In both studies, there were 14 mice in each treatment and control group. In the first study, CDl mice were challenged intravenously with 25 mg/kg of lipopolysaccharide (LPS) from E. coli 0111:B4. Immediately after the challenge, the mice were treated intravenously with rBPIzI or rBPI(10-193)C132A
at doses of 15, 20, 25 and 30 mg/kg, or with the control vehicle (formulation buffer only). Mortality was recorded twice daily for seven days.
The results from the first study, shown in Table 1 below, indicate that treatment with both rBPI(10-193)C132A and rBPI21 significantly increased SllrVlVal compared to the vehicle controls. In addition, rBPI(10-193)C132A was at least two-fold more potent than rBPI21 with a similar survival benefit seen with a two-fold lower dose of rBPI(10-193)C132A compared to rBPI2~-Table 1 No. of Survivors out of 15 Dose (mg/kg) Control rBPI21 rBPI(10-193)C132A

0(Vehicle) 0 NA' NA

0 15**.##

10 20 4 15**,#

11** 15**

13** 13**

1 NA, Not Applicable 15 **, p < 0.01 vs. control #, p < 0.05 vs. rBPI2, ##, p < 0.01 vs. control In the second study, a wider range of rBPI(10-193)C132A doses (5, 20 10, 15, 20, 25, 30 mg/kg) was studied. The results, shown in Table 2 below, confirm that while both rBPI2, and rBPI(10-193)C132A offered a significant survival benefit over the control, as in the first study, rBPI(10-193)C132A
was at least two-fold mom potent, achieving similar efficacy as rBPIZt with a 2-fold lower dose.

WO 99/66044 PCfIUS99/13860 Table 2 No. of Survivors out of Dose (mg/kg)Control rBPI21 rBPI(10-193)C132A

0(Vehicle) 2 NAl NA

5 NDl 3 10 ND 10*

15 ND 14**

20 7 14**,li 25 13** 15**

30 14** 15*

' NA, Not Applicable; ND, Not Done **, p < 0.05 vs. control **, p < 0.01 vs. control #, p < 0.05 vs. rBPI2, D.
Two separate studies were conducted to examine the relative potencies or rBPizl and rBPI(10-193)C132A in a mouse model of lethal bacteremia.
In both studies, there were 20 mice per treatment group. In the first study, CDl mice were challenged with 6.8 X 10' colony forming units (CFL~ of E. coli 07:K1 administemd intravenously. Immediately after the challenge, the mice were treated intravenously with rBPlzl or rBPI(10-193)C132A at doses of 10, 20 and 30 mg/kg, or with control vehicle (formulation buffer only). Mortality was recorded twice daily for seven days.
The results from the first study, shown in Table 3 below, demonstrate a significant increase in survival for the groups treated with 10 and 30 mg/kg of rBPI21 (p < 0.05 vs. control). While a similar significant increase in survival was not observed with the rBPI(10-193)C132A vs. control, there was not a significant difference in survival advantage between the rBPI21 and rBPI(10-193)C132A - treated groups in this study.
Table 3 No. of Survivors out of Dose (mg/kg) Control rBPIzI rBPI(10-193)C132A

0(Vehicle) 6 NA

10 14* 12 14* 10 15 *, p < 0.05 vs. control To more fully characterize the effects of rBPI2, and rBPI(10-193)C132A in this model, a second study was conducted in which a wider range of doses was studied. In this study, CD1 mice were challenged with 2.57 X 108 20 colony forming units (CFin of E. coli 07:K1 administered intravenously.
Immediately after the challenge, the mice were treated intravenously with 1.0, 3.0, 10 and 30 mg/kg rBPI,l, and 0.3, 1.0, 3.0, 10 and 30 mg/kg rBPI(10-193)C132A.
The results, shown in Table 4 below, indicate that both proteins provided protection, and that there was no significant difference in the protective effects of 25 the two variants at any dose.

Table 4 No. of Survivors out of 20 Dose (mg/kg) Control rBPI21 rBPI(10-193)C132A

0(Vehicle) 6 0.3 ND' 6 1.0 4 6 3.0 9 10*

10 13** 8 30 11* 14**

' ND, Not Done *, p < 0.05 vs. control **, p < 0.01 vs. control $,, ('ardiovaccular .ff .r in Con ciou Rat A series of experiments were conducted to determine the relative effects of rBPizl and rBPI(10-193)C132A on blood pressure in rats. Each rat was anesthetized with a mixture of ketamine (Fort Dodge Labs, Fort Dodge, LA) and Rompum {Bayer Coip., Shawnee Mission, KS). A catheter was then placed in the right carotid artery and connected to a pressure transducer to record blood pressure.
A second catheter was placed in the right jugular vein to inject rBPI or vehicle.
The rats were then allowed to recover before the experiments began.
Experiments were initiated when the rats were alert, mobile and when blood pressure was stable within the normal range. rBPI2l, rBPI(10-193)C132A or control vehicle (formulation buffer) were then injected as a bolus over 15 seconds and mean arterial blood pressure (mm Hg) was recorded over the next 60 minutes.
In preliminary experiments, it was determined that doses of 20 and 30 mgl kg of rBPI21 had no significant effect on blood pressure relative to the vehicle but that a dose of 40 mg/kg resulted in a significant decrease in blood pressure that was evident within 5 minutes. This hypotensive response was greatest 15 minutes after the injection when blood pressure had decreased by 48 X12 mm Hg (mean ASE; p > 0.05). After 60 minutes, the blood pressure of the rBPI21-treated animals recovered and was not significantly different from that of the vehicle - treated animals.
To compare effects of rBPI21 and rBPI(10-193)C132A, groups of 5 rats were given 40 mg/kg of each drug substance or vehicle control, and blood pressure responses were analyzed as area under the curve (AUC). Figure 1 shows that, as previously observed, rBPI2~ caused a significant drop in blood pressure indicated by the elevated AUC relative to the vehicle control. By comparison, rBPI(10-193)C132A had no significant effect on blood pressure compared with the vehicle control. A dose of 50 mg/kg rBPI2, (N = 4 rats) had an even greater hypotensive effect than that of the 40 mg/kg dose as indicated by a further increase in the AUC in Figure 1. At this higher dose, some reduction in blood pressure also occurred in rats administered rBPI(10-193)C132A (N=3), but this effect was not significant compared to the vehicle control.
Numerous modifications and variations of the above-described invention are expected to occur to those of skill in the art. Accordingly, only such limitations as appear in the appended claims should be placed thereon.

WO 99/66044 PC'TNS99/13860 SEQUENCE LISTING
<110> XOMA Limited Ltd.
Horwitz, Arnold (inventor) Carroll, Stephen F. (inventor) Burke, David (inventor) <120> Bactericidal/Permeability-Increasing Protein (BPI) Deletion Analogs <130> 27129/35765 PCT
<140>
<141>
<150> 09/099,725 <151> 1998-06-19 <160> 6 <170> PatentIn Ver. 2.0 <210> 1 <211> 1813 <212> DNA
<213> Homo Sapiens <220>
<221> CDS
<222> (31)..(149I) <220>
<221> mat peptide <222> (124)..(1491) <220>
<223> rBPI
<400> 1 caggccttga ggttttggca gctctggagg atg aga gag aac atg gcc agg ggc 54 Met Arg Glu Asn Met Ala Arg Gly cct tgc aac gcg ccg aga tgg gtg tcc ctg atg gtg ctc gtc gcc ata 102 Pro Cys Asn Ala Pro Arg Trp Val Ser Leu Met Val Leu Val Ala Ile ggc acc gcc gtg aca gcg gcc gtc aac cct ggc gtc gtg gtc agg atc 150 Gly Thr Ala Val Thr Ala Ala Val Asn Pro Gly Val Val Val Arg Ile tcc cag aag ggc ctg gac tac gcc agc cag cag ggg acg gcc get ctg 198 Ser Gln Lys Gly Leu Asp Tyr Ala Ser Gln Gln Gly Thr Ala Ala Leu cag aag gag ctg aag agg atc aag att cct gac tac tca gac agc ttt 246 Gln Lys Glu Leu Lys Arg Ile Lys Ile Pro Asp Tyr Ser Asp Ser Phe .CA 02331880 2000-12-18 aagatcaagcat.ctt gggaag gggcattatagc ttctacagc atggac 294 LysIleLysHis LeuGlyLys GlyHisTyrSer PheTyrSer MetAsp atccgtgaattc cagcttccc agttcccagata agcatggtg cccaat 342 IleArgGluPhe GlnLeuPro SerSerGlnIle SerMetVal ProAsn gtgggccttaag ttctccatc agcaacgccaat atcaagatc agcggg 390 ValGlyLeuLys PheSerIle SerAsnAlaAsn IleLysIle SerGly aaatggaaggca caaaagaga ttcttaaaaatg agcggcaat tttgac 438 LysTrpLysAla GlnLysArg PheLeuLysMet 5erGlyAsn PheAsp ctgagcatagaa ggcatgtcc atttcggetgat ctgaagctg ggcagt 486 LeuSerIleGlu GlyMetSer IleSerAlaAsp LeuLysLeu GlySer aaccccacgtca ggcaagccc accatcacctgc tccagctgc agcagc 534 AsnProThrSer GlyLysPro ThrIleThrCys SerSerCys SerSer cacatcaacagt gtccacgtg cacatctcaaag agcaaagtc gggtgg 582 HisIleAsnSer ValHisVal HisIleSerLys SerLysVal GlyTrp ctgatccaactc ttccacaaa aaaattgagtct gcgcttcga aacaag 630 LeuIleGlnLeu PheHisLys LysIIeGluSer AlaLeuArg AsnLys atgaacagccag gtctgcgag aaagtgaccaat tctgtatcc tccaag 678 MetAsnSerGln ValCysGlu LysValThrAsn SerValSer SerLys ctgcaaccttat ttccagact ctgccagtaatg accaaaata gattct 726 LeuGlnProTyr PheGlnThr LeuProValMet ThrLysIle AspSer gtggetggaatc aactatggt ctggtggcacct ccagcaacc acgget 774 ValAlaGlyIle AsnTyrGly LeuValAlaPro ProAlaThr ThrAla gagaccctggat gtacagatg aagggggagttt tacagtgag aaccac 822 GluThrLeuAsp ValGlnMet LysGlyGluPhe TyrSerGlu AsnHis cacaatccacct ccctttget ccaccagtgatg gagtttccc getgcc 870 HisAsnProPro ProPheAla ProProValMet GluPhePro AlaAla catgaccgcatg gtatacctg ggcctctcagac tacttcttc aacaca 918 HisAspArgMet ValTyrLeu GlyLeuSerAsp TyrPhePhe AsnThr gccgggcttgta taccaagag getggggtcttg aagatgacc cttaga 966 AlaGlyLeuVal TyrGlnGlu AlaGlyValLeu LysMetThr LeuArg gat gac atg att ~cca aag gag tcc aaa ttt cga ctg aca acc aag ttc 1014 Asp Asp Met Ile Pro Lys Glu Ser Lys Phe Arg Leu Thr Thr Lys Phe ttt gga acc ttc cta cct gag gtg gcc aag aag ttt ccc aac atg aag 1062 Phe Gly Thr Phe Leu Pro Glu Val Ala Lys Lys Phe Pro Asn Met Lys ata cag atc cat gtc tca gcc tcc acc ccg cca cac ctg tct gtg cag 1110 Ile Gln Ile His Val Ser Ala Ser Thr Pro Pro His Leu Ser Val Gln ccc acc ggc ctt acc ttc tac cct gcc gtg gat gtc cag gcc ttt gcc 1158 Pro Thr Gly Leu Thr Phe Tyr Pro Ala Val Asp Val Gln Ala Phe Ala gtc ctc ccc aac tcc tcc ctg get tcc ctc ttc ctg att ggc atg cac 1206 Val Leu Pro Asn Ser Ser Leu Ala Ser Leu Phe Leu Ile Gly Met His aca act ggt tcc atg gag gtc agc gcc gag tcc aac agg ctt gtt gga 1254 Thr Thr Gly Ser Met Glu Val Ser Ala Glu Ser Asn Arg Leu Val Gly gag ctc aag ctg gat agg ctg ctc ctg gaa ctg aag cac tca aat att 1302 Glu Leu Lys Leu Asp Arg Leu Leu Leu Glu Leu Lys His Ser Asn Ile ggc ccc ttc ccg gtt gaa ttg ctg cag gat atc atg aac tac att gta 1350 Gly Pro Phe Pro Val Glu Leu Leu Gln Asp Ile Met Asn Tyr Ile Val ccc att ctt gtg ctg ccc agg gtt aac gag aaa cta cag aaa ggc ttc 1398 Pro Ile Leu Val Leu Pro Arg Val Asn Glu Lys Leu Gln Lys Gly Phe cct ctc ccg acg ccg gcc aga gtc cag ctc tac aac gta gtg ctt cag 1446 Pro Leu Pro Thr Pro Ala Arg Val Gln Leu Tyr Asn Val Val Leu Gln cct cac cag aac ttc ctg ctg ttc ggt gca gac gtt gtc tat aaa 1491 Pro His Gln Asn Phe Leu Leu Phe Gly Ala Asp Val Val Tyr Lys tgaaggcacc aggggtgccg ggggctgtca gccgcacctg ttcctgatgg gctgtggggc 1551 accggctgcc tttccccagg gaatcctctc cagatcttaa ccaagagccc cttgcaaact 1611 tcttcgactc agattcagaa atgatctaaa cacgaggaaa cattattcat tggaaaagtg 1671 catggtgtgt attttaggga ttatgagctt ctttcaaggg ctaaggctgc agagatattt 1731 cctccaggaa tcgtgtttca attgtaacca agaaatttcc atttgtgctt catgaaaaaa 1791 aacttctggt ttttttcatg tg 1813 <210> 2 <211> 487 <212> PRT
<213> Homo Sapiens <400> 2 Met Arg Glu Asn Met Ala Arg Gly Pro Cys Asn Ala Pro Arg Trp Val Ser Leu Met Val Leu Val Ala Ile Gly Thr Ala Val Thr Ala Ala Val Asn Pro Gly Val Val Val Arg Ile Ser Gln Lys Gly Leu Asp Tyr Ala Ser Gln Gln Gly Thr Ala Ala Leu Gln Lys Glu Leu Lys Arg Ile Lys Ile Pro Asp Tyr Ser Asp Ser Phe Lys Ile Lys His Leu Gly Lys Gly His Tyr Ser Phe Tyr Ser Met Asp Ile Arg Glu Phe Gln Leu Pro Ser Ser Gln Ile Ser Met Val Pro Asn Val Gly Leu Lys Phe Ser Ile Ser Asn Ala Asn Ile Lys Ile Ser Gly Lys Trp Lys Ala Gln Lys Arg Phe Leu Lys Met Ser Gly Asn Phe Asp Leu Ser Ile Glu Gly Met Ser Ile Ser Ala Asp Leu Lys Leu Gly Ser Asn Pro Thr Ser Gly Lys Pro Thr Ile Thr Cys Ser Ser Cys Ser Ser His Ile Asn Ser Val His Val His Ile Ser Lys Ser Lys Val Gly Trp Leu Ile Gln Leu Phe His Lys Lys Ile Glu Ser Ala Leu Arg Asn Lys Met Asn Ser Gln Val Cys Glu Lys Val Thr Asn Ser Val Ser Ser Lys Leu Gln Pro Tyr Phe Gln Thr Leu Pro Val Met Thr Lys Ile Asp Ser Val Ala Gly Ile Asn Tyr Gly Leu Val Ala Pro Pro Ala Thr Thr Ala Glu Thr Leu Asp Val Gln Met Lys Gly Glu Phe Tyr Ser Glu Asn His His Asn Pro Pro Pro Phe Ala Pro Pro Val Met Glu Phe Pro Ala Ala His Asp Arg Met Val Tyr Leu Gly Leu Ser Asp Tyr Phe Phe Asn Thr Ala Gly Leu Val Tyr Gln Glu Ala Gly Val Leu Lys Met Thr Leu Arg Asp Asp Met Ile Pro Lys Glu Ser Lys Phe Arg Leu.Thr Thr Lys Phe Phe Gly Thr Phe Leu Pro Glu Val Ala Lys Lys Phe Pro Asn Met Lys Ile Gln Ile His Val Ser Ala Ser Thr Pro Pro His Leu Ser Val Gln Pro Thr Gly Leu Thr Phe Tyr Pro Ala Val Asp Val Gln Ala Phe Ala Val Leu Pro Asn Ser Ser Leu Ala Ser Leu Phe Leu Ile Gly Met His Thr Thr Gly Ser Met Glu Val Ser Ala Glu Ser Asn Arg Leu Val Gly Glu Leu Lys Leu Asp Arg Leu Leu Leu Glu Leu Lys His Ser Asn Ile Gly Pro Phe Pro Val Glu Leu Leu Gln Asp Ile Met Asn Tyr Ile Val Pro Ile Leu Val Leu Pro Arg Val Asn Glu Lys Leu Gln Lys Gly Phe Pro Leu Pro Thr Pro Ala Arg Val Gln Leu Tyr Asn Val Val Leu Gln Pro His Gln Asn Phe Leu Leu Phe Gly Ala Asp Val Val Tyr Lys <210> 3 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: primer <400> 3 ctgctctaaa agctgctgca g 21 <210> 4 <211> 33 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: primer <400> 9 ccaggccctt ctgggaggcc gctgtcacgg cgg 33 <210> 5 <211> 33 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: primer <400> 5 gccgtgacag cggcctccca gaagggcctg gac 33 <210> 6 <211> 18 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: primer <900> 6 ctgggaactg ggaagctg 18

Claims (19)

WHAT IS CLAIMED ARE:
1. A bactericidal/permeability-increasing protein (BPI) deletion analog consisting of amino acid residues 10 through 193 of mature human BPI, wherein a cysteine residue at position 132 is replaced by a different amino acid.
2. The BPI deletion analog of claim 1 wherein the amino acid replacing said cysteine residue is a non-polar amino acid selected from the group consisting of alanine and serine.
3. The BPI deletion analog of claim 1 wherein the cysteine residue at position 132 is replaced by alanine.
4. A polynucleotide encoding the BPI deletion analog of claim 1.
5. A polynucleotide encoding the BPI deletion analog of claim 3.
6. The polynucleotide of claim 4 further comprising the twenty-seven amino acid leader sequence of BPI.
7. The polynucleotide of claim 4 which is a DNA.
8. An expression vector comprising the DNA according to claim 7.
9. A host cell stably transformed or transfected with the DNA of claim 7 in a manner allowing expression in said host cell of said polypeptide deletion analog.
10. A eukaryotic host cell according to claim 9.
11. The host cell of claim 10 which is a CHO cell.
12. A method for producing a BPI deletion analog polypeptide comprising growing a host cell according to claim 9 in a suitable culture medium and isolating said polypeptide from said host cell or said culture medium.
13. The polypeptide product of the method of claim 12.
14. A composition comprising the BPI deletion analog of claim 1 and a pharmaceutically-acceptable diluent, adjuvant, or carrier.
15. A composition comprising the BPI deletion analog of claim 3 and a pharmaceutically-acceptable diluent, adjuvant, or carrier.
16. A composition comprising the BPI deletion analog of claim 13 and a pharmaceutically-acceptable diluent, adjuvant, or carrier.
17. An improved method of administering a BPI protein product to a subject comprising administering the composition of claim 14 to said subject.
18. An improved method of administering a BPI protein product to a subject comprising administering the composition of claim 15 to said subject.
19. An improved method of administering a BPI protein product to a subject comprising administering the composition of claim 16 to said subject.
CA002331880A 1998-06-19 1999-06-18 Bactericidal/permeability-increasing protein (bpi) deletion analogs Abandoned CA2331880A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/099,725 US6013631A (en) 1998-06-19 1998-06-19 Bactericidal/permeability-increasing protein (BPI) deletion analogs
US09/099,725 1998-06-19
PCT/US1999/013860 WO1999066044A1 (en) 1998-06-19 1999-06-18 Bactericidal/permeability-increasing protein (bpi) deletion analogs

Publications (1)

Publication Number Publication Date
CA2331880A1 true CA2331880A1 (en) 1999-12-23

Family

ID=22276330

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002331880A Abandoned CA2331880A1 (en) 1998-06-19 1999-06-18 Bactericidal/permeability-increasing protein (bpi) deletion analogs

Country Status (9)

Country Link
US (4) US6013631A (en)
EP (1) EP1086230A1 (en)
JP (1) JP2002518011A (en)
CN (1) CN1273598C (en)
AU (1) AU755624B2 (en)
CA (1) CA2331880A1 (en)
MX (1) MXPA00011398A (en)
NZ (1) NZ508816A (en)
WO (1) WO1999066044A1 (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5198541A (en) * 1987-08-11 1993-03-30 New York University Dna encoding bactericidal/permeability-increasing proteins
US6013631A (en) * 1998-06-19 2000-01-11 Xoma Corporation Bactericidal/permeability-increasing protein (BPI) deletion analogs
CA2373262A1 (en) * 1999-05-24 2000-11-30 Xoma (Us) Llc Therapeutic uses of bpi protein products in humans with otitis media with effusion
WO2002055099A2 (en) * 2000-12-01 2002-07-18 Xoma Technology Ltd Modulation of pericyte proliferation using bpi protein products or bpi inhibitors
CA2492008C (en) 2002-03-29 2012-06-26 Xoma Technology Ltd. Multigenic vectors plasmids and methods for increasing expression of recombinant polypeptides
AU2004216298B2 (en) * 2003-02-28 2009-04-23 Chugai Seiyaku Kabushiki Kaisha Stabilized protein-containing formulations
EP1891207A2 (en) * 2005-06-03 2008-02-27 Biovitrum AB Process for cultivating animal cells comprising the feeding of plant-derived peptones
EP1741440A1 (en) 2005-07-08 2007-01-10 Mellitus S.L. Use of BPI protein for the treatment of disorders of the metabolism and cardiovascular disorders
US8338376B2 (en) * 2006-10-20 2012-12-25 Biogen Idec Ma Inc. Compositions comprising variant LT-B-R-IG fusion proteins
CA2680792A1 (en) * 2007-03-15 2008-09-18 Biogen Idec Ma Inc. Treatment of autoimmune disorders
CN108042807B (en) 2011-04-05 2020-10-16 达纳-法伯癌症研究所有限公司 Use of BPI and its homologues as a radiation mitigant and radioprotectant
GB201319621D0 (en) 2013-11-06 2013-12-18 Norwegian University Of Science And Technology Antimicrobial agents and their use in therapy
GB201319620D0 (en) 2013-11-06 2013-12-18 Norwegian University Of Science And Technology Immunosuppressive agents and their use in therapy

Family Cites Families (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5198541A (en) * 1987-08-11 1993-03-30 New York University Dna encoding bactericidal/permeability-increasing proteins
US5576292A (en) * 1987-08-11 1996-11-19 New York University Biologically active bactericidal/permeability-increasing protein fragments
US5171739A (en) * 1989-02-14 1992-12-15 Incyte Pharmaceuticals, Inc. Treatment of endotoxin-associated shock and preventation thereof using a BPI protein
US5770694A (en) * 1990-08-13 1998-06-23 Incyte Pharmaceuticals, Inc. Genetically engineered BPI variant proteins
US5334584A (en) * 1989-02-14 1994-08-02 Incyte Pharamaceuticals, Inc. Recombinant, non-glycosylated bpi protein and uses thereof
US5308834A (en) * 1989-02-14 1994-05-03 Incyte Pharmaceuticals, Inc. Treatment of endotoxin-associated shock and prevention thereof using a BPI protein
US5234912A (en) * 1989-02-14 1993-08-10 Incyte Pharmaceuticals, Inc. Pharmaceutical compositions comprising recombinant BPI proteins and a lipid carrier and uses thereof
US5089274A (en) * 1989-02-14 1992-02-18 Incyte Pharmaceuticals, Inc. Use of bactericidal/permeability increasing protein or biologically active analogs thereof to treat endotoxin-related disorders
EP0563222B1 (en) * 1990-12-03 1998-02-25 New York University Biologically active bactericidal/permeability-increasing protein fragments
EP0605653A4 (en) * 1991-09-26 1995-03-15 Incyte Pharma Inc A new form of liposaccharide binding protein (lbp).
CA2136208C (en) * 1992-05-19 1999-08-10 Lynn S. Grinna Improved methods for the preparation of endotoxin-binding proteins
AU4382193A (en) * 1992-05-19 1993-12-13 Xoma Corporation BPI-immunoglobulin fusion proteins
ES2164098T3 (en) * 1993-02-02 2002-02-16 Xoma Technology Ltd PHARMACEUTICAL COMPOSITIONS CONTAINING A BACTERICIDE PROTEIN INCREASING PERMEABILITY AND A TENSIOACTIVE.
US5420019A (en) * 1993-02-02 1995-05-30 Xoma Corporation Stable bactericidal/permeability-increasing protein muteins
AU679207B2 (en) 1993-02-18 1997-06-26 Merck Sharp & Dohme Limited Azacyclic compounds, compositions containing them and their use as tachykinin antagonists
EP0690721B1 (en) * 1993-03-12 1998-05-13 Xoma Corporation Treatment of mycobacterial diseases by administration of bactericidal/permeability-increasing protein products
CN1105574C (en) * 1993-03-12 2003-04-16 爱克索马技术有限公司 Therapeutic uses of bactericidal/permeability increasing protein products
US5733872A (en) * 1993-03-12 1998-03-31 Xoma Corporation Biologically active peptides from functional domains of bactericidal/permeability-increasing protein and uses thereof
US5652332A (en) * 1993-03-12 1997-07-29 Xoma Biologically active peptides from functional domains of bactericidal/permeability-increasing protein and uses thereof
US5348942A (en) * 1993-03-12 1994-09-20 Xoma Corporation Therapeutic uses of bactericidal/permeability increasing protein products
ES2123134T3 (en) * 1993-03-12 1999-01-01 Xoma Corp BIOLOGICALLY ACTIVE PEPTIDES OF FUNCTIONAL DOMAINS OF PERMEABILITY-INCREASING BACTERICIDE PROTEIN AND USES THEREOF.
WO1994021280A1 (en) * 1993-03-22 1994-09-29 Incyte Pharmaceuticals, Inc. Use of bactericidal/permeability increasing protein and lipopolysaccharide binding protein levels and ratios thereof in diagnosis
CA2161971A1 (en) * 1993-04-30 1994-11-10 Randal W. Scott Recombinant bpi-based and lbp-based proteins, nucleic acid molecules encoding same, methods of producing same, and uses thereof
AU7175694A (en) * 1993-06-17 1995-01-17 Xoma Corporation Lipopolysaccharide binding protein derivatives
JPH09500269A (en) * 1993-07-02 1997-01-14 インサイト ファーマシューティカルズ,インク. Glycosylated and non-glycosylated proteins with increased bactericidal / permeability and methods of making the same
US5770561A (en) * 1993-07-14 1998-06-23 Xoma Corporation Method for potentiating BPI protein product bactericidal activity by administration of LBP protein products
AU7330994A (en) * 1993-07-14 1995-02-13 Xoma Corporation Method for potentiating bpi protein product bactericidal activity by administration of lbp protein products
US5466580A (en) * 1993-09-22 1995-11-14 Xoma Corporation Method for quantifying BPI in body fluids
CN1133634A (en) * 1993-09-22 1996-10-16 爱克斯欧玛公司 Method for quantifying BPI in body fluids
ES2178656T3 (en) * 1993-09-22 2003-01-01 Xoma Technology Ltd METHOD TO TREAT GRAM-NEGATIVE BACTERIAL INFECTION BY ADMINISTRATION OF BACTERICIDE PROTEIN PRODUCTS THAT INCREASES PERMEBILITY (BPI) AND ANTIBIOTICS.
DE69433848T2 (en) * 1993-10-05 2005-05-25 Xoma Technology Ltd., Berkeley BPI PROTEIN PRODUCTS FOR DEFINED FEATURES OF THE RETICULOENDOTHELIAL SYSTEM
CA2181164C (en) * 1994-01-14 2008-01-08 Arnold Horwitz Anti-gram-positive bacterial methods and materials
AU703211B2 (en) * 1994-01-14 1999-03-18 Xoma Corporation Anti-fungal methods and materials
US5484705A (en) * 1994-01-24 1996-01-16 Xoma Corporation Method for quantifying lipopolysaccharide binding protein
US5643875A (en) * 1994-01-24 1997-07-01 Friedmann; Nadav Human therapeutic uses of bactericidal/permeability increasing (BPI) protein products
US5447913A (en) * 1994-03-11 1995-09-05 Xoma Corporation Therapeutic uses of bactericidal/permeability-increasing protein dimer products
US5786324A (en) * 1994-03-24 1998-07-28 Regents Of The University Of Minnesota Synthetic peptides with bactericidal activity and endotoxin neutralizing activity for gram negative bacteria and methods for their use
US5578568A (en) * 1994-04-22 1996-11-26 Xoma Corporation Method of treating conditions associated with intestinal ischemia/reperfusion
US5532216A (en) * 1994-06-24 1996-07-02 Incyte Pharmaceuticals, Inc. Neutralization of non-lipopolysaccharide compounds by bactericidal/permeability-increasing protein
US5646114A (en) * 1994-07-11 1997-07-08 Xoma Corporation Anti-protozoan methods
DE69532302D1 (en) * 1994-09-15 2004-01-22 Xoma Technology Ltd PEPTIDES AGAINST MUSHROOMS
US5912228A (en) * 1995-01-13 1999-06-15 Xoma Corporation Therapeutic compositions comprising bactericidal/permeability-increasing (BPI) protein products
US5494896A (en) * 1995-03-31 1996-02-27 Xoma Corporation Method of treating conditions associated with burn injuries
JP2001520631A (en) * 1995-07-20 2001-10-30 ゾーマ コーポレイション Antifungal peptide
US5686414A (en) * 1995-11-14 1997-11-11 Xoma Corporation Methods of treating conditions associated with corneal transplantation
JP2000501386A (en) * 1995-11-14 2000-02-08 ゾーマ コーポレイション Bactericidal / permeability enhancing (BPI) protein for treating conditions associated with corneal injury
US5741779A (en) * 1996-05-10 1998-04-21 Xoma Corporation Antithrombotic materials and methods
PT914144E (en) * 1996-05-10 2001-05-31 Univ Texas THERAPEUTIC UTILIZATIONS OF BIP PROTEIN PRODUCTS AGAINST HUMAN MENINGOCOCCEMIA
EP0907372B1 (en) * 1996-05-23 2003-03-12 XOMA Technology Ltd. Therapeutic uses of bpi protein products in humans with hemorrhage due to trauma
US5888973A (en) * 1996-08-09 1999-03-30 Xoma Corporation Anti-chlamydial uses of BPI protein products
AU744918B2 (en) * 1996-11-01 2002-03-07 Xoma Corporation Therapeutic uses of BPI protein products in cystic fibrosis patients
US6013631A (en) * 1998-06-19 2000-01-11 Xoma Corporation Bactericidal/permeability-increasing protein (BPI) deletion analogs

Also Published As

Publication number Publication date
CN1312858A (en) 2001-09-12
EP1086230A1 (en) 2001-03-28
US20050130889A1 (en) 2005-06-16
US6599880B1 (en) 2003-07-29
US6087126A (en) 2000-07-11
NZ508816A (en) 2003-03-28
CN1273598C (en) 2006-09-06
AU4697699A (en) 2000-01-05
MXPA00011398A (en) 2003-04-22
AU755624B2 (en) 2002-12-19
US6013631A (en) 2000-01-11
WO1999066044A1 (en) 1999-12-23
JP2002518011A (en) 2002-06-25

Similar Documents

Publication Publication Date Title
US6652862B2 (en) Biologically active bactericidal/permeability-increasing protein fragments
JP3086685B2 (en) Biologically active bactericidal / permeability enhancing protein fragments
US6054431A (en) Anti-gram-positive bacterial methods and materials
AU703134B2 (en) Therapeutic uses of bactericidal/permeability-increasing protein dimer products
US6599880B1 (en) Bactericidal/permeability-increasing protein (BPI) deletion analogs
US6767893B2 (en) Method of treating conditions associated with intestinal ischemia/reperfusion
WO1995000641A1 (en) Lipopolysaccharide binding protein derivatives
US6274344B1 (en) Methods for recombinant microbial production of fusion proteins and BPI-derived peptides
WO1995002414A1 (en) Method for potentiating bpi protein product bactericidal activity by administration of lbp protein products
WO1996030037A1 (en) Method of treating conditions associated with burn injuries
DE69530689T2 (en) HEPARIN BINDING PROTEIN FOR TREATING SEPSIS
US6515104B1 (en) Therapeutic peptide-based constructs derived from domain II of bactericidal/permeability-increasing protein
US5770561A (en) Method for potentiating BPI protein product bactericidal activity by administration of LBP protein products
CA2367943A1 (en) Therapeutic uses of bpi protein products in bpi-deficient humans
US6482796B2 (en) Therapeutic uses of N-terminal BPI protein products in ANCA-positive patients
US6274348B1 (en) Methods for the preparation of positively charged proteins
US20020006638A1 (en) Methods for recombinant microbial production of fusion proteins and BPI-derived peptides

Legal Events

Date Code Title Description
EEER Examination request
FZDE Dead