AU4605401A - Multimeric erythropoietin with increased biological activity - Google Patents

Multimeric erythropoietin with increased biological activity Download PDF

Info

Publication number
AU4605401A
AU4605401A AU46054/01A AU4605401A AU4605401A AU 4605401 A AU4605401 A AU 4605401A AU 46054/01 A AU46054/01 A AU 46054/01A AU 4605401 A AU4605401 A AU 4605401A AU 4605401 A AU4605401 A AU 4605401A
Authority
AU
Australia
Prior art keywords
polypeptide
erythropoietin
succinimidyl
group
hydroxysuccinimide ester
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
AU46054/01A
Inventor
Arthur J Sytkowski
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.)
Beth Israel Deaconess Medical Center Inc
Original Assignee
Beth Israel Deaconess Medical Center Inc
Beth Israel Hospital Association
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 Beth Israel Deaconess Medical Center Inc, Beth Israel Hospital Association filed Critical Beth Israel Deaconess Medical Center Inc
Priority to AU46054/01A priority Critical patent/AU4605401A/en
Publication of AU4605401A publication Critical patent/AU4605401A/en
Abandoned legal-status Critical Current

Links

Landscapes

  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Description

1
AUSTRALIA
Patents Act 1990 DIVISIONAL APPLICATION Regulation 3.2 o o oooo o o o oooo oo o* o* oo Name of Applicant: Actual Inventor: Address for Service: Beth Israel Deaconess Medical Center SYTKOWSKI, Athur, J DAVIES COLLISON CAVE, Patent Attorneys, Level 3, 303 Coronation Drive, Milton, Queensland, 4064, Australia "Multimeric erythropoietin with increased biological activity" Invention Title: Details of Parent Application No: 53779/98 The following statement is a full description of this invention, including the best method of performing it known to me/us: Q:\Opcr\Vpa\May 2001\2415067 divisional Bcth Isracl.137.doc 17/5/01 -1A- MULTIMERIC ERYTHROPOIETIN WITH INCREASED BIOLOGICAL ACTIVITY Background Modification of naturally occurring polypeptides which have therapeutic value is often attempted in an effort to increase their biological activity. Several methods have been employed to increase the biological activity of therapeutic proteins. These methods often focus on increasing the size of the therapeutic agents. For example, 10 the size of a protein can be increased through chemical conjugation with a reagent such as polyethylene glycol (PEG) (Knusli, C. et al., Brit. J. Haematol. 82:654-663 (1992)).
This procedure, also known as "PEGylation", has been reported with several protein agents, first as a means to reduce antigenicity, but also as a way to increase biological activity.
~Another method of increasing a protein's size is through chemical cross-linking with another protein. For example, to increase the antigenicity of a protein, chemical 20 cross-linking agents are used to conjugate the immunogenic protein to a carrier molecule such as immunoglobulin or serum albumin.
However, the conjugation of chemical compounds or inert molecules to a polypeptide often results in a significant decrease of the overall biological activity, and of selected biological activity of the polypeptide, (Knusli, et al., Brit. J. Haematol., 82:654-663 (1992)). These conjugations must be designed such that the resulting modified polypeptide remains therapeutically efficacious and retains the desired biological properties of the unmodified, wild type naturally-occurring) polypeptide (Satake, et al., Biochem. Biophys. Acta. 1038:125-129 (1990)).
-2- Erythropoietin (EPO) is a glycoprotein hormone involved with the growth and development of mature red blood cells from erythrocyte precursor cells. It is a 166 amino acid polypeptide that exists naturally as a monomer. (Lin, F-K., et al. Proc. Natl. Acad. Sci. USA 82:7580-7584 (1985)).
Several forms of anemia, including those associated with renal failure, HIV infection, blood loss and chronic disease can be treated with this hematopoietic growth factor. Erythropoietin is typically administered by intravenous or subcutaneous injection three times weekly at a dose of approximately 25-100 U/kg. Though quite effective, this form of therapy is very expensive.
Estimates for the treatment of chronic dialysis patients have ranged from $8,000-10,000 per patient per year.
15 Another problem encountered in the practice of medicine when using injectable pharmaceuticals is the frequency at which those injections must be made in order to maintain a oo therapeutic level of the compound in the circulation. For example, erythropoietin has a relatively short plasma half-life (Spivak, and Hogans, Blood, 73:90 (1989); McMahon, et al., Blood, 76:1718(1990)), therefore, therapeutic plasma levels are rapidly lost, and repeated intravenous administrations must be made. An alternative route of administration is subcutaneous 25 injection. This route offers slower absorption from the site of administration, thus causing a sustained release effect. However, significantly lower plasma levels are achieved and, thus, a similar frequency of injection, as is required with intravenous administration, must be used to get a comparable therapeutic effect.
were not lodged with this application between the side chains of certain amino acids, between amino acids and carboxylic acid groups, or via carbohydrate moieties. In particular, the heterobifunctional crosslinking reagents used in the present invention contain either a cleavable disulfide bond group or a maleimido group.
The present invention also relates to multimeric erythropoietin comprising two, or more, erythropoietin molecules convalently linked together by one, or more, thioether bond(s). These erythropoietin multimers also exhibit increased biological activity. The present o. invention further relates to methods of producing the modified erythropoietin polypeptides with increased biological activity described herein, and to methods of their use.
The modification of wild type erythropoietin with a heterobifunctional cross-linking reagent containing a o cleavable disulfide bond group resulted in a modified erythropoietin with increased potency relative to unmodified 20 wild type erythropoietin. Importantly, the disulfide bond group can be reduced to a free sulfhydryl group. The availability of a free sulfhydryl group on the erythropoietin polypeptide permitted further modification of erythropoietin to produce multimeric erythropoietin with a prolonged circulating half-life relative to wild type erythropoietin. The production of multimeric erythropoietin was accomplished by a method of chemically cross-linking two, or more, modified erythropoietin polypeptides.
Briefly, the method is as follows.
A first erythropoietin derivative was produced by reacting wild type erythropoietin with a heterobifunctional cross-linking reagent containing a cleavable disulfide bond group. The disulfide bond was reduced to produce erythropoietin containing a free sulfhydryl group. A second erythropoietin derivative was produced by reacting wild type erythropoietin with a heterobifunctional cross-linking reagent containing a maleimido group. The first and second erythropoietin derivatives were reacted together, thereby forming at least one thioether bond between the sulfhydryl and maleimido groups, thus forming a homodimer or homotrimer of erythropoietin.
Surprisingly, these multimeric erythropoietin molecules exhibit biological activity comparable to wild type erythropoietin. More importantly, the erythropoietin dimers showed a significantly prolonged circulating half-life in vivo, relative to wild type erythropoietin.
Thus, as a result of the work presented herein, erythropoietin has now been modified to produce erythropoietin compositions which exhibit increased biological potency relative to wild type erythropoietin. Moreover, the modified erythropoietin of the present invention can be dimerized and trimerized with other modified erythropoietin molecules to produce multimeric erythropoietin molecules with prolonged in vivo circulating half-lives.
In a preferred embodiment, the biologically active erythropoietin homodimer comprises two erythropoietin polypeptides covalently linked by at least one thioether bond, wherein: a) the first erythropoietin polypeptide comprises an erythropoietin polypeptide with a heterobifunctional cross-linking reagent containing a free sulfhydryl group attached to the erythropoietin polypeptide; and b) the second erythropoietin polypeptide comprises an erythropoietin polypeptide with a heterobifunctional cross-linking reagent containing a maleimido group attached to the erythropoietin polypeptide, wherein said heterobifunctional cross-linking reagent is selected from the group consisting of: y-maleimidobutyric acid Nhyrdoxysuccinimide ester, m-maleimidobenzoyl-N-hydroxysuccinimide ester, Emaleimidocaproic acid N-hydroxysuccinimide ester, 4- (p-maleimidophenyl) butyric acid N-hydroxysuccinimide ester, and P-maleimidoproprionic acid Nhydroxysuccinimide ester, and at least one thioether bond forms between the free sulfhydryl group of the first erythropoietin polypeptide and the maleimido group of the second erythropoietin polypeptide.
In another preferred embodiment, the biologically active erythropoietin homotrimer comprises three erythropoietin polypeptides covalently linked by thioether bonds, wherein: a) the first and second erythropoietin polypeptides comprise erythropoietin polypeptides with a heterobifunctional cross-linking reagent containing a free sulfhydryl group attached to each erythropoietin polypeptide; and b) the third erythropoietin polypeptide comprises an erythropoietin polypeptide with a heterobifunctional cross-linking reagent containing two, or more, maleimido groups attached to the erythropoietin polypeptide, wherein said heterobifunctional crosslinking reagent is selected from the group consisting of: y-maleimidobutyric acid N-hyrdoxysuccinimide ester, m-maleimidobenzoyl-N-hydroxysuccinimide ester, Emaleimidocaproic acid N-hydroxysuccinimide ester, 4- (p-maleimidophenyl) butyric acid N-hydroxysuccinimide ester, and P-maleimidoproprionic acid Nhydroxysuccinimide ester, and the thioether bonds form between the free sulfhydryl group of the first and second erythropoietin polypeptides and the maleimido groups of the third erythropoietin polypeptide.
In yet another preferred embodiment, the biologically active erythropoietin homotrimer comprises three erythropoietin polypeptides covalently linked by thioether bonds, wherein: 20 a) the first erythropoietin polypeptide comprises an erythropoietin polypeptide with a heterobifunctional cross-linking reagent containing two, or more, free sulfhydryl groups attached to the polypeptide; and b) the second and third erythropoietin polypeptides comprise erythropoietin polypeptides with a heterobifunctional cross-linking reagent containing a maleimido group attached to each polypeptide, wherein said heterobifunctional cross-linking reagent is selected from the group consisting of: y-maleimidobutyric acid N-hyrdoxysuccinimide ester, m-maleimidobenzoyl-N-hydroxysuccinimide ester, E-maleimidocaproic acid N-hydroxysuccinimide ester, (pmaleimidophenyl) butyric acid N-hydroxysuccinimide ester, and Pmaleimidoproprionic acid N-hydroxysuccinimide ester, and the thioether bonds 5b form between the free sulfhydryl groups of the first erythropoietin polypeptide and the maleimido group of the second and third erythropoietin polypeptides.
Preferably, the heterobifunctional cross-linking reagents of a) and b) are attached to one, or more, primary amine or amines in the erythropoietin polypeptides.
Suitably, the erythropoietin is glycosylated erythropoietin and the heterobifunctional cross-linking reagents are attached to one, or more, carbohydrate moiety or moieties in the glycosylated erythropoietin polypeptides.
The above preferred erythropoietin homodimers are advantageous because the cross-linking groups used to link these homodimers together do not elicit immune responses as opposed to prior art coupling groups that do. Accordingly, the above preferred homodimers can be used to raise immune responses to the erythropoietin homodimers and not the coupling agent.
Brief Description of the Figures .Figure 1A shows the chemical structure of SPDP.
Figure 1B shows the chemical structure of LC-SPDP.
Figure C shows the chemical structure of sulfo-LC SPDP.
Figure 1C shows the chemical structure of sulfo-LC SPDP.
Figure 2 shows the chemical structure of SMCC.
Figure 3 is a histogram depicting the biological activity of the fractions containing homotrimers, homodimers and monomers of erythropoietin collected after high pressure liquid chromatography (HPLC).
Figure 4 is a graphic representation of the results of a bioassay demonstrating the increased in vivo half-life of the erythropoietin dimer and monomer.
Detailed Description of the Invention The present invention relates to modified polypeptides with increased biological activity, and methods of making and using these modified polypeptides. Polypeptides suitable for modification by the methods described herein are polypeptides, preferably monomeric polypeptides, which 10 do not contain any free sulfhydryl groups. Polypeptides of special interest are those polypeptides which interact with a cellular receptor to initiate cellular signaling events, o. for example, insulin and erythropoietin. Polypeptides encompassed by the present invention are typically used as injectable therapeutic agents. If polypeptides with increased biological activity are used as injectable therapeutic agents, the frequency of administration of these polypeptides can be reduced.
As described herein, these polypeptides can be modified 20 to increase their biological activity relative to the biological activity of the naturally occurring polypeptides.
Increased biological activity, is defined herein as a prolonged plasma half-life a longer circulating half-life than the naturally occurring polypeptide), or higher potency requiring a smaller quantity than the naturally occurring polypeptide to achieve a specified level of biological activity). Increased biological activity, as used herein, can also encompass a combination of the above described activities. For example, a modified polypeptide with higher potency can also have an increased circulating half-life. In any case, because the polypeptides described herein have increased biological activity, the frequency with which they must be administered can be reduced.
The polypeptides encompassed by the present invention are modified with a heterobifunctional cross-linking reagent. The heterobifunctional cross-linking reagent can be attached to one, or more primary amine or amines, within the polypeptide. For example, the heterobifunctional crosslinking reagent can be attached to the amino acid residue, lysine or to the alpha amino terminus of erythropoietin.
Alternatively, for glycoproteins, the heterobifunctional cross-linking reagent can be attached to one, or more carbohydrate moiety, or moieties, in an oligosaccharide chain on the polypeptide.
The heterobifunctional cross-linking reagent is 15 generally selected from a group of cross-linking reagents containing either a cleavable disulfide bond group or a maleimido group. The addition of a disulfide bond group to a polypeptide also permits the design of a cross-linking strategy to produce multimeric polypeptides. The disulfide bond can be cleaved by reaction with a known reducing agent, for example, dithiothreitol (DTT) which reduces the disulfide bond in the cross-linking reagent to produce a modified polypeptide derivative containing a free sulfhydryl (SH) group.
A second polypeptide derivative, capable of reacting with a free sulfhydryl group, is then produced by attaching a heterobifunc:ional cross-linking reagent containing a maleimido group to the naturally occurring polypeptide.
Again, the cross-linking reagent can be attached to primary amines or carbohydrate moieties in the polypeptide. The resulting polypeptide derivative containing a maleimido group is reacted with the polypeptide derivative containing a reactive sulfhydryl group resulting in a multimeric -8polypeptide molecule covalently linked together by at least one thioether bond formed between the SH group and the maleimido group.
Erythropoietin, a glycoprotein hormone involved with the growth and development of mature red blood cells from erythrocyte precursor cells, is a glycosylated polypeptide particularly suited for modification using the methods described herein. Erythropoietin is produced in the kidney in response to hypoxia red blood cell loss due to anemia) and regulates red blood cell growth and differentiation through interaction with its cognate cellular receptor. Wild type erythropoietin is defined herein to include recombinant human erythropoietin (Powell, et al., Proc. Natl. Acad. Sci. USA, 83:6465-6469 (1986)), or naturally occurring erythropoietin which has been isolated and purified from blood (Miyake, et al. J.
Biol. Chem., 252:5558-5564 (1977)) or sheep plasma (Goldwasser, et al. Proc. Natl. Acad. Sci. U.S.A., 68:697-698 (1971)), or chemically synthesized erythropoietin which can be produced using techniques well-known to those of skill in the art. Erythropoietin is a 166 amino acid polypeptide that exists naturally as a monomer. (Lin, F-K., et al. Proc. Natl. Acad. Sci. USA 82:7580-7584 (1985)).
The predicted secondary structure of erythropoietin has been reported (McDonald, et al., Mol. Cell. Biol., 6:842-848 (1986)).
It was noted from the structure of wild type erythropoietin that the polypeptide does not contain any free (reactive) sulfhydryl (SH) groups. (Boissel, et al., J. Biol. Chem. 268:15983-15993 (1993)). Free SH groups are useful for preparing conjugated proteins, such as radiolabeled antibodies Patent 4,659,839), or otherwise chemically modifying the polypeptide resulting in -9altered biological activity of a polypeptide. A free sulfhydryl group can also play a role in the binding of a polypeptide to its cellular receptor. For example, the polypeptide hormone, insulin, is covalently linked to its cellular receptor via a disulfide exchange mechanism.
(Clark, S. and Harrison, L. J. Biol. Chem., 258:11434- 11437 (1983); Clark, S. and Harrison, L. J. Biol. Chem., 257:12239-12344 (1982)). Thus, a free sulfhydryl group can be critical to the biological activity of a polypeptide.
Accordingly, a scheme was devised to modify wild type erythropoietin to attach a free sulfhydryl group.
In one embodiment of the present invention, wild type erythropoietin was chemically modified by the covalent :attachment of a heterobifunctional cross-linking reagent 15 containing a cleavable disulfide bond group. The crosslinking reagent was attached to a primary amine in the erythropoietin polypeptide. The attachment of a heterobifunctional cross-linking reagent to wild type erythropoietin resulted in erythropoietin with increased potency relative to unmodified erythropoietin.
Specifically, three different heterobifunctional cross-linking reagents were used to produce modified erythropoietin with increased biological activity. These cross-linking reagents were attached to one, or more, primary amine or amines in the wild type erythropoietin.
The cross-linking reagents were N-succinimidyl 3-(2-pyridyldithio) propionate (SPDP), "long chain" Nsuccinimidyl 3(2-pyridyldithio) propionate (LC-SPDP), wherein the length of the chain of SPDP is increased with additional methyl groups, and sulfonated "long-chain" Nsuccinimidyl 3(2-pyridyldithio) propionate (sulfo-LC-SPDP) wherein LC-SPDP is sulfonated. SPDP (Figure 1A), LC-SPDP (Figure 1B) and sulfo-LC-SPDP (Figure 1C) are commercially available cross-linking agents (Pierce Chemical Co., Rockford Ill). SPDP, LC-SPDP and sulfo-LC-SPDP all contain an N-hydroxysuccimmidyl group to react with free amino groups. In addition, these reagents all contain a disulfide bond group that can be further modified to form a reactive sulfhydryl group.
Another heterobifunctional cross-linking reagent that can be used to modify wild type erythropoietin is a carbohydrate specific reagent that attaches to carbohydrate moieties of glycosylated polypeptides. This cross-linking reagent, 3-(2-pyridyldithio) propionyl hydrazide (PDPH), contains an oxidized carbohydrate specific hydrazide and o oo also contains a cleavable disulfide bond group.
Wild type erythropoietin was modified with o. 15 heterobifunctional cross-linking reagents SPDP, LC-SPDP and sulfo-LC-SPDP as described in detail in Example 1. Briefly, erythropoietin was incubated in the presence of specified concentrations of the .chemical reagent N-succinimidyl 3-(2-pyridyldithio) propionate (SPDP) so as to achieve different molar ratios of SPDP:EPO in solution.
The unmodified wild type erythropoietin and SPDP modified erythropoietin (SPDP-EPO) were bioassayed according to the method of Krystal, (Krystal, Exp. Hematol., 11:649-660 (1983)), which measures the effect of erythropoietin on 25 erythropoiesis in intact mouse spleen cells. The results, shown in Table 1, demonstrate that SPDP-EPO exhibited an increased biological activity relative to the control wild type erythropoietin.
-11- TABLE 1 SPECIFIC ACTIVITY OF SPDP-MODIFIED ERYTHROPOIETIN Reaction Mixture, Specific Activity SPDP/EPO, mol/mol U/mcg 0:1 200 1:1 174 3:1 340 Erythropoietin modified with sulfo-LC-SPDP (sulfo LC- SPDP-EPO), which has the advantage of increased solubility 10 in aqueous solutions, was also prepared as described in Example 1. Incubation of erythropoietin in the presence of sulfo-LC-SPDP at different molar ratios, followed by dialysis and biological assay revealed that sulfo-LC-SPDP modification of erythropoietin resulted in a 530% increase 15 in potency over the activity of wild type erythropoietin, as shown in Table 2. Thus, the specific activity of the erythropoietin was increased from 170 U/mcg for the wild type erythropoietin to 900 U/mcg for the modified erythropoietin prepared in the presence of 10 fold molar excess of sulfo-LC-SPDP.
-12- TABLE 2 SPECIFIC ACTIVITY OF SULFO-LC-SPDP MODIFIED ERYTHROPOIETIN Reaction Mixture, Specific Activity SULFO-LC-SPDP/EPO, U/mcg mols/mol Experiment #1 0:1 170 5:1 220 10:1 900 30:1 600 50:1 250 100:1 350 Experiment #2 0:1 200 15 1:1 200 2:1 370 3:1 350 6:1 380 7:1 560 10:1 900 LC-SPDP EPO was also prepared as described in Example 1. Although the biological activity of this derivative was not evaluated, it is reasonable to believe that erythropoietin modified with LC-SPDP would also exhibit increased biological activity due to its close structural relationship to SPDP and sulfo-LC-SPDP.
The chemically modified erythropoietin derivatives described above, which contained a cleavable disulfide bond group, permitted the design of a strategy to cross-link erythropoietin to form EPO-EPO dimers and EPO-EPO-EPO -13trimers with increased biological activity. These homodimers (EPO-EPO) and homotrimers (EPO-EPO-EPO) are "long-acting" erythropoietin proteins (also referred to herein as LA-EPOs). That is, these multimeric erythropoietin derivatives exhibit a prolonged circulating half-life relative to unmodified, erythropoietin.
The methods of preparing multimeric erythropoietin with increased biological activity are described in detail in Examples 2 and 3. Although erythropoietin is used as the specific example, it is understood that the methods described herein can be used to produce multimers a polypeptide covalently cross-linked with one, or more, identical polypeptides) of any suitable polypeptide.
Briefly, a first derivative of erythropoietin was 15 prepared as described in Example 1, by reacting erythropoietin with the compound N-succinimidyl 3-(2-pyridyldithio) propionate (SPDP) to form SPDP-EPO.
This reaction introduced an external disulfide bond group into the erythropoietin molecule. To form a free (or reactive) sulfhydryl group, SPDP-EPO can be exposed to a reducing agent, known to those of skill in the art, to reduce the disulfide bond groups. As described in Example 2, SPDP-EPO was exposed to dithiothreitol (DTT), which reduces the disulfide bond in the SPDP moiety to produce an 25 erythropoietin molecule containing free SH groups, also referred to herein as SH-EPO.
A second erythropoietin derivative was produced by reacting ery:hropoietin with succinimidyl 4-(N-maleinidomethyl) cyclohexane-1-carboxylate, also known as SMCC (Figure 2) to form SMCC-EPO. This reagent has an Nhydroxy succinimidyl (NHS) group at one end and a maleimido group at the other. The NHS group of SMCC reacts with free amino groups in erythropoietin resulting in the formation of -14- SMCC-EPO. The maleimido group of SMCC, now pointing outward from the SMCC-EPO derivative, reacts with free sulfhydryl groups found on SH-EPO. Therefore, when SH-EPO and SMCC-EPO are mixed together in solution, the reactive groups combine resulting in the formation of an EPO-EPO dimer, one SH-EPO with one SMCC-EPO) or an EPO-EPO-EPO trimer one SMCC-EPO with two SH-EPOs, or two SMCC-EPOs with one SH- EPO) in which the modified erythropoietin polypeptides are covalently linked by at least one thioether bond one thioether bond in dimerized EPO and two thioether bonds in trimerized EPO). It is interesting to note that SMCC-EPO, when tested in the Krystal bioassay, did not exhibit any increased biological activity relative to unmodified erythropoietin.
15 Alternatively, a heterobifunctional cross-linking reagent which contains a maleimido group to attach to carbohydrate moieties such as 4-(4-N-maleimidophenyl) butyric acid hydrazide-HCl (MPBH) and 4-(N-maleimidomethyl) cyclohexane-1-carboxyl-hydrazide-HC1, can be used.
20 The first and second erythropoietin derivatives were reacted together as described in detail in Example 2. The reaction resulted in the formation of multimeric erythropoietin, as well as unreacted monomeric erythropoietin derivatives, which can be separated by high 25 pressure liquid chromatography (HPLC), as described in o* Example 2. The erythropoietin dimers comprised two erythropoietin polypeptides linked by one or more thioether bonds. The erythropoietin trimers comprised three erythropoietin polypeptides, also linked by thioether bonds.
The trimer can comprise two erythropoietin polypeptides, each containing a free sulfhydryl group which is linked with a third erythropoietin polypeptide containing two or more, maleimido groups. Alternatively, the erythropoietin trimer can comprise one erythropoietin polypeptide containing two, or more, free sulfhydryl groups which is linked with two erythropoietin polypeptides, each containing a maleimido group. The presence of EPO, EPO-EPO dimers and EPO-EPO-EPO trimers was confirmed by Western blot analysis using antibodies specific for erythropoietin as described in Sytkowski, and Fisher, J. Biol. Chem., 260:14727-14731 (1985).
Although the monomeric erythropoietin retained its biological activity, the erythropoietin dimers and trimers prepared under the conditions described in Example 2, with SH-EPO, did not exhibit biological activity when tested in the Krystal bioassay. Therefore, a second cross-linking protocol was designed in which a second type of SH-EPO 15 derivative was prepared using sulfo-LC-SPDP. This agent functions similarly to SPDP as outlined above, however, it contains a spacer arm of several angstroms in length wherein the number of CH 2 groups in the linear portion of the molecule is increased) resulting in increased physical separation of the species attached to its reactive ends. In particular, sulfo-LC-SPDP contains five methyl groups within the linear chain of the molecule, and is also sulfated to increase its aqueous solubility.
Multimeric erythropoietin produced using sulfo-LC-SPDP- 25 EPO (SH-LC-EPO) as the first erythropoietin derivative was prepared, and separated by HPLC as described in detail in Example 2. HPLC fractions containing the trimers, dimers and monomers were tested in the Krystal bioassay for biological activity. Importantly, all three of these species, monomers, dimers, and trimers exhibited biological activity in the Krystal assay. (See Figure 3) Multimeric erythropoietin was also produced using heterobifunctional cross-linking reagents containing a free sulfhydryl group attached to the erythropoietin polypeptide and various heterobifunctional cross-linking reagents containing a maleimido group, also referred to herein as "SMCC-like" reagents, as described in detail in Example 3.
As used herein, "SMCC-like" reagents are heterobifunctional cross-linking reagents characterized by a N-hydroxy succinimidyl (NHS) group at one end and a maleimido group at the other. As such they act in the same manner as SMCC in that the NHS group of the "SMCC-like" reagents reacts with free amino groups in erythropoietin and the maleimido group of the "SMCC-like" reagents reacts with free sulfhydryl groups. SMCC-like reagents include, the following: GMBS, y-maleimidobutyric acid N-hydroxysuccinimide ester; MMBS, m-maleimidobenzoyl-N-hydroxysuccinimide ester; EMCS, 15 E-maleimidocaproic acid N-hydroxysuccinimide ester; PMPBS, 4-(p-maleimidophenyl)butyric acid N-hydroxysuccinimide ester; and BMPS, 3 -maleimidoproprionic acid Nhydroxysuccinimide ester. Monomers, dimers and trimers produced with LC-SPDP and the SMCC-like reagents exhibited biological activity as measured in the Krystal assay.
The circulating half-life in vivo of erythropoietin homodimers was determined as described in detail in Example 4. Monomeric and dimeric erythropoietin was injected into rabbits, and blood samples were analyzed at 5 minutes and 2, 25 4, 6, 9, and 24 hours after injection. As shown in Figure 4, the biological activity of dimerized erythropoietin, as measured in the Krystal assay, was still evident 24 hours after the initial injection, whereas the biological activity of monomeric erythropoietin dropped off significantly earlier. Thus, the circulating half-life of dimerized erythropoietin was more than three times longer than wild type erythropoietin. The prolonged circulating half-life of the EPO dimer may be due to its increased size relative to -17monomeric erythropoietin, which would hinder its excretion from the body through the kidney. Although the erythropoietin trimers were not assayed at this time, it is reasonable to predict that an EPO homotrimer would exhibit similar, or even longer circulatory half-life as the homodimers because a trimer has even greater size than a dimer. These erythropoietin dimers and trimers are also referred to herein as long-acting erythropoietins (LA-EPOs).
Preferred isomers of erythropoietin dimers and trimers can also be prepared. Nine primary amino groups have been identified in the human erythropoietin molecule. At the amino terminus of erythropoietin is an alpha amino group of alanine 1. Additionally, there are eight epsilon amino groups found on lysine 20, 45, 52, 97, 116, 140, 152 and 154. When using LC-SPDP, SMCC, or SMCC-like reagents, one or more of these primary amino groups is/are modified by the *reagent.
Variations in the structure of the EPO/EPO dimer could S' alter the activity/potency of the isoform. Although the 20 three-dimensional structure of EPO is not known, certain regions are held to be important for receptor binding.
Since the side chain of lysine, including its epsilon amino group, is hydrophilic, it is expected to be accessible to solvent on the outside of the molecule and, therefore, could take part in EPO receptor binding.
Chemical modification of such a lysine, for example, could decrease activity of the EPO/EPO dimer. Therefore, within the mixture of all possible modifications, it is reasonable to expect that some molecules are less active than others due to such unfavorable linkages. To put it another way, some molecules are more active than others, that is, they are preferred isomers. Another possibility is that steric factors could position the receptor binding -18domains of the dimer subunits in more favorable steric or less favorable orientations. This could enhance or inhibit the likelihood that both binding domains of each dimer would bind simultaneously.
It is possible to modify amino groups preferentially so as to control isomer structure. Several methods to control (target) modifications of the primary amino groups are described in Example As a result of the work described herein, modified erythropoietin polypeptides are provided which exhibit increased biological activity. Erythropoietin modified with a heterobifunctional cross-linking reagent containing a cleavable disulfide bond group exhibited a 530% increase in biological activity relative to wild type erythropoietin.
15 This increase in biological activity indicates that an effective amount of modified erythropoietin is substantially less than a comparable effective amount of wild type erythropoietin. The effective amount of modified erythropoietin is defined herein as the amount of 20 erythropoietin required to elicit an erythropoietic response, as indicated by increased growth and/or differentiation of erythrocytic precursor cells. For example, if the typical effective dose of erythropoietin used therapeutically is 25 U/kg, then an effective dose of 25 modified erythropoietin can reasonably be as low as 5.0 U/kg to achieve the same effect.
Alternatively, the effective amount of multimeric erythropoietin described herein, with a prolonged circulating half-life, will require less frequent administration than an equivalent amount of wild type erythropoietin. For example, if an effective dose of erythropoietin is typically administered 3 times a week, multimeric erythropoietin with increased biological activity -19will only need to be administered once a week. In either case, a reduced quantity of erythropoietin modified with a heterobifunctional cross-linking reagent, or multimeric erythropoietin, will be required over the course of treatment than is necessary if wild type erythropoietin is used.
The modified erythropoietin with increased biological activity described herein can be used in place of wild type erythropoietin whenever treatment with erythropoietin is called for. For example, modified erythropoietin can be used for treatment in an individual experiencing anemia associated with renal failure, chronic disease, HIV infection, blood loss or cancer.
Erythropoietin is generally administered to humans.
Effective treatment with erythropoietin requires maintaining a therapeutic blood level. This can be done by continuous administration, that is, by continuous intravenous o. injections, by discreet intravenous injections, or by subcutaneous injection. The modified erythropoietin of this invention can be employed in admixture with conventional excipients, pharmaceutically acceptable organic or inorganic carrier substances suitable for parenteral administration that do not deleteriously react with the active derivatives.
Suitable pharmaceutically acceptable carriers include, but are not limited to, water, salt solutions, alcohols, gum arabic, vegetable oils, benzyl alcohols, polyethylene glycols, gelatine, carbohydrates such as lactose, amylose or starch, magnesium stearate, talc, silicic acid, viscous paraffin, perfume oil, fatty acid esters, hydroxymethycellulose, polyvinyl pyrrolidone, etc. For parenteral application, particularly suitable are injectable, sterile solutions, preferably oily or aqueous solutions, as well as suspensions, emulsions, or implants, including suppositories.
It will be appreciated that the actual preferred amounts of active compound in a specific case will vary according to the specific compound being utilized, the particular compositions formulated, the mode of application, the particular situs of application, and the organism being treated. Dosages for a given recipient will be determined on the basis of individual characteristics, such as body size, weight, age and the type and severity of the condition being treated.
In addition, the modified erythropoietin of the present S" invention, with increased biological activity, can be used in any in vitro application in place of wild type 15 erythropoietin. For example, modified erythropoietin can be used in studies of erythropoietin receptor activity. It will again be appreciated that the amount of modified •erythropoietin with increased biological activity needed to achieve desired results, increased hemoglobinization 20 of red blood cell precursor cells) will be substantially less than the amount of wild type erythropoietin required to achieve those desired results.
The present invention will now be illustrated by the following examples, which are not intended to be limiting in any way.
Example 1: SPDP-EPO Derivative with Higher Potency Three different heterobifunctional cross-linking reagents containing cleavable disulfide bond groups have been used to produce erythropoietin derivatives with increased biological activity. These agents are Nsuccinimidyl 3-(2-pyridyldithio) propionate (SPDP), "longchain" N-succinimidyl 3-(2-pyridyldithio) propionate, -21wherein the length of the chain of SPDP is increased with additional methyl groups (LC-SPDP), and sulfonated "long-chain" N-succinimidyl 3-(2-pyridyldithio) propionate (sulfo-LC-SPDP) Modified erythropoietin polypeptides were prepared as follows.
Recombinant human erythropoietin was produced by expression of the human erythropoietin gene in stably transfected BHK (baby hamster kidney) cells (Powell, J.S. et al., Proc. Nat. Sci. Acad. USA., 83:6465-6469 (1986) and purified using standard laboratory techniques. The purified protein was then incubated in the presence of specified concentrations of the chemical reagent N-succinimidyl 3-(2-pyridyldithio) propionate (SPDP), dissolved in dimethyl sulfoxide, so as to achieve molar ratios of 0:1, 1:1 and 3:1 15 (SPDP:EPO) in solution. After incubation overnight at room temperature, the solutions were dialyzed against phosphate buffered saline to remove unreacted SPDP.
~The wild type erythropoietin and modified erythropoietin (SPDP-EPO) samples were evaluated for biological activity according to the method of Krystal.
(Krystal, Exp. Hematol., 11:649-660 (1983)). Briefly, the bioassay of Krystal measures the effect of erythropoietin on intact mouse spleen cells. Mice are treated with phenylhydrazine to stimulate production of red 25 blood cell precursor cells in the spleen. After treatment, the spleens are removed, intact spleen cells are carefully isolated and incubated with various amounts of wild type erythropoietin or the modified erythropoietin described herein. After an overnight incubation, 3 H thymidine is added and its incorporation into cellular DNA is measured.
The amount of 3 H thymidine incorporation is indicative of erythropoietin-stimulated DNA synthesis in erythroid precursor cells via interaction of erythropoietin with its -22cellular receptor. The results demonstrate that SPDP-EPO exhibited an increased biological activity relative to the wild type erythropoietin, and that this increase in activity was proportional to the molar ratio of SPDP:EPO in the reaction mixture.
Additionally, wild type erythropoietin was modified using sulfo-LC-SPDP, a compound which has the advantage of increased solubility in aqueous solutions. Incubation of erythropoietin in the presence of sulfo-LC-SPDP at the previously described molar ratios followed by dialysis and biological assay revealed that sulfo-LC-SPDP modification of erythropoietin resulted in an increase in potency of approximately 530%. The specific activity of the erythropoietin was increased from 170 U/mcg for the 15 nonderivatized material to 900 U/mcg for the material derivatized in the presence of 10 fold molar excess of sulfo-LC-SPDP.
Example 2: Long-Acting Multimeric Erythropoietin Derivatives 20 To prepare the first SH-EPO derivative, 50 ug of human erythropoietin obtained as described in Example 1, was incubated in the presence of five-fold molar excess of N-succinimidyl 3-(2-pyridyldithio) propionate (SPDP) obtained from Pierce Chemical Company. After incubation at room temperature for sixteen hours, the solution was dialyzed against phosphate buffered saline. The modified erythropoietin was then exposed to 1mM DTT to reduce the disulfide bond in SPDP resulting in one, or more, free sulfhydryl group(s) on the erythropoietin molecule.
The second erythropoietin derivative, SMCC-EPO, was prepared as follows. A second 50 ug portion of human erythropoietin was incubated in the presence of five-fold -23molar excess of succinimidyl 4-(N-maleimidomethyl) cyclohexane-l-carboxylate (SMCC). After a sixteen hour incubation at room temperature, the solution was dialyzed against phosphate buffered saline.
The SH-EPO and SMCC-EPO were mixed together in phosphate buffered saline (20 mM sodium phosphate, 150 mM sodium chloride, pH 7.4) at room temperature for 90 minutes, and dialyzed against PBS. The mixture was then subjected to size exclusion HPLC chromatography on TSK 250, in PBS, room temperature, at 1 ml/min. The polypeptides were subjected to SDS polyacrylamide gel electrophoresis, electrophoretic transfer to nitrocellulose, and Western blotting using anti- S: erythropoietin antibodies according to Sytkowski, A. and Fisher, J. J. Biol. Chem., 260:14727-14731 (1985). The .15 results showed that the protocol succeeded in the formation of two higher molecular weight species of erythropoietin corresponding to erythropoietin dimers and trimers.
o* However, upon assay in the Krystal bioassay, the erythropoietin dimers and trimers produced with SPDP-SH-EPO did not exhibit any biological activity.
Thus, the protocol was revised to use LC-SPDP-EPO as the first derivative, 50 ug of recombinant human erythropoietin was incubated in the presence of three-fold :ee molar excess of LC-SPDP for sixteen hours at room 25 temperature. The material was then dialyzed and treated with 1 mM DTT resulting in SH-LC-EPO. SMCC-EPO was prepared as described above.
These two species were mixed together in solution and the mixture was subjected to size exclusion HPLC on TSK3000SW. Three erythropoietin protein species were detected with elution times of 10.2, 9.1, and 7.2 minutes respectively. The 10.2 minute elution time was known from previous experiments to be that of wild type erythropoietin -24monomer. Therefore, the more rapid elution times of 9.1 and 7.2 minutes corresponded to dimers and trimers, respectively. The fractions containing the erythropoietin dimers and trimers were collected and assayed in the Krystal bioassay. Importantly, as shown in Figure 3, upon testing in the Krystal bioassay, the erythropoietin homodimers and homotrimers exhibited biological activity.
Example 3: Crosslinkinq Erythropoietin Using LC-SPDP and SMCC-Like Reagents Multimers of erythropoietin were also produced using LC-SPDP-EPO derivatives and EPO derivatives produced by S* reaction with SMCC-like reagents. The five SMCC-like crosslinking reagents were: GMBS, 7-maleimidobutyric acid N-hydroxysuccinimide 15 ester; MMBS, m-maleimidobenzoyl-N-hydroxysuccinimide ester; ee* EMCS, E-maleimidocaproic acid N-hydroxysuccinimide ester; PMPBS, 4 -(p-maleimidophenyl)butyric acid N- 20 hydroxysuccinimide ester; and BMPS, -maleimidoproprionic acid N-hydroxysuccinimide ester.
All of these cross-linking reagents are commercially S* available, e.g. from Sigma Chemical Co., St. Louis, MO. The chemical structures of these cross-linkers are shown in Table 3.
TABLE 3 CHEMICAL STRUCTURES OF "SMCC-LIKE" CROSS-LINKING REAGENTS a. GMBS; 7-maleimidobutyric acid N-hydroxysuccinimide ester; a b. MMBS; m-maleimidobenzcy1-N-hydroxysuccinimide ester; C. EMCS; E-maleimidocaproi4c acid N-hydroxysuccinimide ester; I a a a.
a d. PMPBS; 4- (o-raleimidopheriyl)but-yric acid Nhydroxysuccin'imide ester; -0-
I
e. BMPS;- -maleimidoproprionic acid N-hydroxysuccinimide ester; -26f. SMCC; succinimidyl 4-(N-maleimidomethyl) cyclohexane-lcarboxylate; To prepare LC-SPDP, 20 ug of human erythropoietin oo.obtained as described in Example 1, was incubated in the presence of ten-fold molar excess of "longchain" N-succinimidyl 3-(2-pyridyldithio) propionate (LC-SPDP) -obtained from Pierce Chemical Company. The incubation occurred in sodium phosphate 20 mM, sodium chloride 100 mM, pH 7.0 (PBS) at 23 0 C for 30 min. To stop the reaction, excess PBS at 4 0 C was added to the mixture (final volume, 0.5 ml) and then dialyzed for at least 6 h at 4C against PBS (3X 1.0 L) Finally, DTT (final concentration, 1 mM) was added to the mixture for 10 min to reduce the disulfide bond in LC-SPDP, resulting in one, or more, free sulfhydryl group(s) on the erythropoietin molecule.
The second erythropoietin derivative, SMCC-like EPO, was prepared as follows. 20 ug portions of human erythropoietin was incubated with a ten-fold molar excess of each of the five SMCC-like reagents listed above and allowed -27to react. After a 30 min incubation at 30 0 C in PBS, the reaction was stopped by adding excess PBS at 4 0 C (final volume, 0.5 ml). The mixture was then dialyzed for at least 6 h at 4 0 C against PBS (3X 1.OL).
Equal molar amounts of LC-SPDP EPO and each of the five SMCC-like EPO were placed in five, separate dialysis bags and dialyzed against PBS overnight at 4 0 C (1 The mixture from each of the dialysis bags was then individually subjected to size exclusion HPLC chromatography. A size exclusion HPLC chromatography column, Progel TSK-3000 SWXL mm I.D. x 30 cm) and guard column, Progel TSK SWXL cm x 6.0 mm were equilibrated with 100 mM sodium phosphate, 150 mM sodium chloride, pH 7.0. 400 il (16 gg of total EPO) of monomer/dimer/trimer mixture EPO:LC-SPDP 15 EPO:GMBS) was separated on the equilibrated column at a flow rate of 1.0 ml/min and 0.2 ml fractions were collected.
The elution profile was monitored at 280 nm. Bovine serum albumin (final concentration, 2 mg/ml) was added to each fraction to stabilize the dimers/trimers and monomers.
20 Elution profiles of the cross-linked EPO multimers(e.g., 2. EPO:LC-SPDP EPO:GMBS; EPO:LC-SPDP EPO:MMBS; EPO:LC-SPDP EPO:EMCS; EPO:LC-SPDP EPO:PMPBS; EPO:LC-SPDP EPO:BMPS; and EPO:LC-SPDP EPO:SMCC) were similar to those shown in Figure 3 for EPO:LC-SPDP EPO:SMCC multimers.
10 pl of each HPLC fraction was diluted in 490 l of bioassay medium (78% a-MEM, 20% FBS, 0.1 mM 1mercaptoethanol, lX penicillin/streptomycin/ fungizone) and sterilly filtered through 0.2 .m filters. Further final dilutions of 100X, 500X and 5000X were made of the fractions in bioassay medium and assayed for activity using the Krystal in vitro assay, as previously described. Fractions containing monomeric EPO, dimerized EPO and trimerized EPO all exhibited biological activity in the Krystal assay.
-28- Western blot analysis was also performed on the HPLC fractions as follows. 10 il of each fraction was electrophoresed on a 10% SDS polyacrylamide gel and transferred to nitrocellulose at 25V for 18 h at 4 0 C in mM Tris, 192 mM glycine and 10% methanol. The membranes were blocked with 20 mM Tris-HCl, 500 mM NaC1, 0.1% (TBST) 10% Non-fat dry milk overnight with rocking at 4 0
C.
They were then rinsed 2X with TBST, washed 1X for 15 min, 2X for 5 min each, with TBST. The monoclonal EPO antibody AE- 7A5 (28 il Ab in 50 ml TBST/5% dry milk) was placed over the membranes and rocked at 23 0 C for 1 h. They were washed as above followed by incubation with goat anti-mouse IgG (Cappel, diluted 1000X in TBST/5% dry milk). Washing was .o carried out as above with additional 2X for 5 min each.
Bands were detected using the ECL detection reagents from Amersham. Equivolumes of solutions 1 and 2 were mixed and ml of the mixture placed over each membrane. After 1 min the membranes were wrapped in Saran Wrap brand plastic wrap and exposed to X-ray film. Fractions containing monomeric EPO, dimerized EPO and trimerized EPO all specifically reacted with the anti-EPO antibodies.
Example 4: In Vivo Testing of Multimeric Erythropoietin Derivatives A group of New Zealand white rabbits were injected intravenously either with wild type monomeric erythropoietin or with dimerized LA-EPO, as prepared in Example 2, at 0.4 mg/ml in PBS. Blood samples were obtained at 5 minutes and 2, 4, 6, 9, and 24 hours and measured the circulating erythropoietin by the Krystal in vitro biologic assay. The results shown in Figure 4 indicate that the in vivo half-life for monomeric wild type erythropoietin was approximately seven hours, as expected from previously -29published reports. The in vivo half-life of LA-EPO however, was prolonged beyond the twenty-four hour period of the experiment as shown in Figure 4.
Example 5: Methods of Preparing and Purifying Preferred Isomers of EPO Dimers Altering the pH of the reaction The pKa's of alpha amino groups and of the epsilon amino group are 9.69 and 10.53, respectively, but this is determined for free amino acids in solution. In contrast, when the amino acid is part of a polypeptide, these pKa's can vary greatly due to surrounding structures such as other amino acid side chains. This means that within a given protein such as erythropoietin, each of the epsilon amino groups of the eight lysines can have a different pKa.
Lowering the pH of the reaction causes ionization S(protonation) of the NH 2 group to form a NH3* group, thus reducing its reactivity with the succinimidyl moiety of LC- SPDP or SMCC.
Protecting (blocking) the amino group from the modifying reagent A number of means can be used to protect amino acid side chains from chemical modification. For example, site specific antibodies directed toward certain regions of the amino acid sequence could be used. Binding the antibody to the erythropoietin prior to chemical modification would greatly reduce or eliminate modification of those amino groups that form part of the antigenic determinant or are sterically restricted by the bulky immunoglobulin molecule.
A series of site specific antipeptide antibodies to erythropoietin covering numerous domains, some of which include lysine residues have been made, as described in Sytkowski, A.J. and Donahue, J.Biol. Chem, 262:1161 (1987).
In addition to antibody protection, reversible chemical modification of amino groups can be employed. Using this method, the protein is reacted with a reversible modifying reagent such as maleic anhydride. Certain amino groups can be modified, thus preventing subsequent modification when reacted with LC-SPDP, SMCC, or SMCC-like reagents.
Following the second modification, the protecting group is 10 removed with an additional chemical reaction at low pH.
This method can result in selective modification of unprotected amino groups.
A third means of protecting amino groups is s** specifically directed toward the alpha amino terminal alanine 1. Instead of expressing the mature EPO protein, the gene can be engineered so that additional amino acid sequence is expressed upstream of alanine 1. This can be engineered so as to include an enzymatic cleavage site immediately upstream of alanine 1. Then, following 20 modification with LC-SPDP or with SMCC, the upstream peptide sequence can be enzymatically cleaved, releasing the mature EPO protein with an unmodified alpha amino group at alanine i.
Side chain targeting due to physicochemical properties and/or physical characteristics of the modifying reagent The physicochemical properties of the modifying reagent can cause it to selectively interact'with certain amino groups of the protein. A classic example of this type of effect is seen in the modification of horse liver alcohol dehydrogenase with iodoacetic acid. Reacting the enzyme with iodoacetic acid results in the highly specific modification of cysteine 46, despite the fact that the -31enzyme contains numerous other free sulfhydryl groups. This specificity is due to the fact that negative charge interacts avidly with a positive charge on the arginyl residue adjacent to cysteine 46. This interaction directs the iodoacetate to this area of the enzyme resulting in a highly selective modification of cysteine 46.
With respect to the modifiers used to produce EPO dimers, the negative charge on sulfo-LC-SPDP or sulfo-SMCC can-reasonably similarly direct the modifying reagent to a positive charge. Additionally, nonpolar/hydrophobic moieties in the modifiers such as the cyclohexane portion of SMCC can target the reagent to lysine residues adjacent to hydrophobic nonpolar amino acids.
SH-EPO and maleimido EPO monomers, modified 15 preferentially on certain amino groups, can reasonably result in the production of site specific dimer isomers using the methods of producing dimers described herein. A list of these isomers is presented in Table 4.
•go *o o *ooo -32- Table 4 Production of Site Specific Dimer Isomers
V
V
V
SH-EO Moifie at Covalently Bonded to Maleimido SH-EO Moifie at Modified EPO at Alanine 1 Alanine 1, lys 20, lys 45, lys 52, lys 97, lys 116, lys 140, lys 152, lys 154 Lys 20 Alanine 1, lys 20, lys 45, lys 52, lys 97, lys 116, lys 140, lys 152, lys 154 Lys 45 Alanine 1, lys 20, lys 45, lys 52, lys 97, lys 116, lys 140, lys 152, lys 154 Lys 52 Alanine 1, lys 20, lys 45, lys 52, lys 97, lys 116, lys 140, lys 152, lys 154 Lys 97 Alanine 1, lys 20, lys 45, lys 52, lys 97, lys 116, lys 140, lys 152, lys 154 Lys 116 Alanine 1, lys 20, lys 45, lys 52, lys 97, lys 116, lys 140, lys 152, lys 154 10 Lys 140 Alanine 1, lys 20, lys 45, lys 52, lys 97, lys 116, lys 140, lys 152, lys 154 Lys 152 Alanine 1, lys 20, lys 45, lys 52, lys 97, lys 116, lys 140, lys 152, lys 154 Lys 154 Alanine 1, lys 20, lys 45, lys 52, lys 97, lys 116, lys 140, lys 152, lys 154 In addition. to these possible dimer isomers, it is reasonable to expect that favored trimer isomers also can be produced using these methods.
There are several methods that can be utilized to separate and purify the EPO monomers that had been modified -33selectively as described above. These methods include reverse phase HPLC (RP-HPLC), ion exchange chromatography DEAE or CM) and affinity chromatography on immobilized EPO receptor. Each of these are described in detail below.
Reverse phase HPLC (RP-HPLC) The combination of linker polarity plus that of the surrounding amino acid sidechains will determine the interaction of the modified EPO monomer with the RP matrix 10 and solvent. This will lead to chromatographically discrete behavior and specifically modified monomers can be isolated.
Ion exchange chromatography such as DEAE or CM Similarly, modification of specific amino groups will alter interaction of the charged EPO with both cation and 15 anion exchangers.
Affinity chromatography on immobilized EPO receptor EPO receptor protein can be expressed recombinantly, purified and linked covalently to a matrix such as agarose.
This affinity matrix can then be used to isolate monomers 20 with the highest affinity for the receptor, and simultaneously to exclude monomers with low or absent receptor binding.
The methods described above for isolation of modified monomers can be applied to dimer and trimer isomers as well.
Additionally, size exclusion chromatography is available for isolation of modified dimers and trimers. The different conformation of the dimers and trimers will lead to molecules exhibiting different average stokes radii resulting in differential behavior on high resolution size exclusion HPLC.
-34-
EQUIVALENTS
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to specific embodiments of the invention described specifically herein. Such equivalents are intended to be encompassed in the scope of the following claims.
Throughout this specification, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element or integer or group of elements or integers but not the exclusion of any other element or integer or group of elements or integers.
The reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggestion that that prior art forms part of the oooA common general knowledge in Australia.
o*o.
.oooe i

Claims (21)

1. A biologically active polypeptide homodimer comprising two polypeptides covalently linked by at least one thioether bond, wherein: a) the first polypeptide comprises a polypeptide with a heterobifunctional cross- linking reagent containing a free sulfhydryl group attached to the polypeptide; and b) the second polypeptide comprises a polypeptide with a heterobifunctional cross- linking reagent containing a maleimido group attached to the polypeptide and at least one thioether bond forms between the free sulfhydryl group of the first polypeptide and the maleimido group of the second polypeptide.
2. The polypeptide homodimer of Claim 1, wherein the polypeptide has altered biological activity.
3. The polypeptide homodimer of Claim 1 wherein the heterobifunctional cross-linking reagent is selected from the group consisting of: N-succinimidyl 3-(2-pyridyldithio) propionate, "long chain" N-succinimidyl 3-(2-pyridyldithio) propionate, sulfonated "long chain" N-succinimidyl 3-(2-pyridyldithio) propionate, succinimidyl 4-(N- maleimidomethyl) cyclohexane-1-carboxylate, "long chain" succinimidyl 4-(N- maleimidomethyl) cyclohexane-1-carboxylate, sulfonated "long chain" succinimidyl 4- (N-maleimidomethyl) cyclohexane- 1 -carboxylate, y-maleimidobutyric acid N- hydroxysuccinimide ester, m-maleimidobenzoyl-N-hydroxysuccinimide ester, E- maleimidocaproic acid N-hydroxysuccinimide ester, 4-(p-maleimidophenyl)butyric acid N-hydroxysuccinimide ester, and fl-maleimidoproprionic acid N-hydroxysuccinimide ester. -36-
4. A composition comprising a polypeptide homodimer according to Claim 1 and a pharmaceutically acceptable carrier.
A biologically active polypeptide homotrimer comprising three polypeptides covalently linked by thioether bonds, wherein: a) the first and second polypeptides comprise polypeptides with a heterobifunctional cross-linking reagent containing a free sulfhydryl group attached to each polypeptide; and b) the third polypeptide comprises a polypeptide with a heterobifunctional cross- linking reagent containing two, or more, maleimido groups attached to the Spolypeptide and the thioether bonds form between the free sulfhydryl group of the first and second polypeptides and the maleimido groups of the third polypeptide.
6. The polypeptide homotrimer of Claim 5, wherein the polypeptide has altered biological activity.
7. The polypeptide homotrimer of Claim 5 wherein the heterobifunctional cross-linking reagent is selected from the group consisting of: N-succinimidyl 3-(2-pyridyldithio) propionate, "long chain" N-succinimidyl 3-(2-pyridyldithio) propionate, sulfonated "long chain" N-succinimidyl 3-(2-pyridyldithio) propionate, succinimidyl 4-(N- maleimidomethyl) cyclohexane-1-carboxylate, "long chain" succinimidyl 4-(N- maleimidomethyl) cyclohexane-1-carboxylate, sulfonated "long chain" succinimidyl 4- (N-maleimidomethyl) cyclohexane-1-carboxylate, y-maleimidobutyric acid N- hydroxysuccinimide ester, m-maleimidobenzoyl-N-hydroxysuccinimide ester, E- maleimidocaproic acid N-hydroxysuccinimide ester, 4-(p-maleimidophenyl)butyric acid N-hydroxysuccinimide ester, and fl-maleimidoproprionic acid N-hydroxysuccinimide ester. -37-
8. A composition comprising a polypeptide homodimer according to Claim 5 and a pharmaceutically acceptable carrier.
9. A biologically active polypeptide homotrimer comprising three polypeptides covalently linked by thioether bonds, wherein: a) the first polypeptide comprises a polypeptide with a heterobifunctional cross- linking reagent containing two, or more, free sulfhydryl groups attached to the polypeptide; and b) the second and third polypeptides comprise polypeptides with a heterobifunctional cross-linking reagent containing a maleimido group attached to each polypeptide :and the thioether bonds form between the free sulfhydryl groups of the first polypeptide and the maleimido group of the second and third polypeptides.
10. The polypeptide homotrimer of Claim 9, wherein the polypeptide has altered biological activity.
11. The polypeptide homotrimer of Claim 9 wherein the heterobifunctional cross-linking reagent is selected from the group consisting of: N-succinimidyl 3-(2-pyridyldithio) S propionate, "long chain" N-succinimidyl 3-(2-pyridyldithio) propionate, sulfonated "long chain" N-succinimidyl 3-(2-pyridyldithio) propionate, succinimidyl 4-(N- maleimidomethyl) cyclohexane-1-carboxylate, "long chain" succinimidyl 4-(N- maleimidomethyl) cyclohexane-1-carboxylate, sulfonated "long chain" succinimidyl 4- (N-maleimidomethyl) cyclohexane- 1 -carboxylate, y-maleimidobutyric acid N- hydroxysuccinimide ester, m-maleimidobenzoyl-N-hydroxysuccinimide ester, E- maleimidocaproic acid N-hydroxysuccinimide ester, 4-(p-maleimidophenyl)butyric acid N-hydroxysuccinimide ester, and f-maleimidoproprionic acid N-hydroxysuccinimide ester. -38-
12. A composition comprising a polypeptide homodimer according to Claim 9 and a pharmaceutically acceptable carrier.
13. The polypeptide homodimer of Claim 1 wherein the heterobifunctional cross-linking reagents of a) and b) are attached to one, or more, primary amine or amines in the polypeptides.
14. The polypeptide homodimer of Claim 1 wherein the polypeptide is glycosylated and the heterobifunctional cross-linking reagents are attached to one, or more, carbohydrate moiety or moieties in the glycosylated polypeptides.
15. A method of preparing a biologically active polypeptide homodimer according to Claim 1, comprising the steps consisting of: a) preparing a first polypeptide derivative by reacting polypeptide with a heterobifunctional cross-linking reagent containing a cleavable disulfide bond S* group, under conditions sufficient to introduce the cleavable disulfide bond group into the polypeptide, thereby producing a first polypeptide derivative containing a cleavable disulfide bond; b) cleaving the disulfide bond group contained in the first polypeptide derivative, thereby producing a first polypeptide derivative containing a free sulfhydryl group; c) preparing a second polypeptide derivative by reacting polypeptide with a heterobifunctional cross-linking reagent containing a maleimido group, under conditions sufficient to introduce the maleimido group into the polypeptide, thereby producing a second polypeptide derivative containing a maleimido group; and d) reacting the first polypeptide derivative containing a free sulfhydryl group with the second polypeptide derivative containing a maleimide group, under conditions -39- sufficient to form a thioether bond between the free sulfhydryl group and the maleimido group resulting in the cross-linking of the first and second polypeptide derivatives, thereby producing a modified polypeptide comprising a first and second polypeptide derivative.
16. The polypeptide homodimer of Claim 15, wherein the polypeptide has altered biological activity.
17. The polypeptide homodimer of Claim 15 wherein the heterobifunctional cross-linking reagent is selected from the group consisting of: N-succinimidyl 3-(2-pyridyldithio) propionate, "long chain" N-succinimidyl 3-(2-pyridyldithio) propionate, sulfonated "long *chain" N-succinimidyl 3-(2-pyridyldithio) propionate, succinimidyl 4-(N- maleimidomethyl) cyclohexane-1-carboxylate, "long chain" succinimidyl 4-(N- maleimidomethyl) cyclohexane-1-carboxylate, sulfonated "long chain" succinimidyl 4- :i maleimidomethyl) cyclohexane-1 -carboxylate, sulfonated "long chain" succinimidyl 4- (N-maleimidomethyl) cyclohexane- 1 -carboxylate, y-maleimidobutyric acid N- i hydroxysuccinimide ester, m-maleimidobenzoyl-N-hydroxysuccinimide ester, E- maleimidocaproic acid N-hydroxysuccinimide ester, 4-(p-maleimidophenyl)butyric acid N-hydroxysuccinimide ester, and 0-maleimidoproprionic acid N-hydroxysuccinimide .ester.
18. The method of Claim 15 wherein the heterobifunctional cross-linking reagents of step a) and step c) react with one, or more, primary amine or amines in the polypeptide.
19. The method of Claim 15 wherein the polypeptide is a glycosylated polypeptide and the heterobifunctional cross-linking reagents of step a) and step c) react with one, or more, carbohydrate moiety or moieties in the glycosylated polypeptide.
A composition comprising a polypeptide homodimer according to Claim 15 and a pharmaceutically acceptable carrier.
21. A polypeptide homodimer according to any one of claims 1-3, or the composition according to claim 4, or the polypeptide homotrimer according to any one of claims 5-7, or the composition according to claim 8, or the polypeptide homotrimer according to any one of claims 9-11, or the composition according to claim 12, or the polypeptide homodimer according to claim 13 or claim 14, or the method according to claim 15, or the polypeptide homodimer according to claim 16 or claim 17, or the method according to claim 18 or claim 19, or the composition according to claim 20, substantially as herein before described with reference to the figures and/or examples. DATED this 17th day of May 2001 ""Beth Israel Deaconess Medical Center DAVIES COLLISON CAVE Patent Attorneys for the applicant *ooo *oo
AU46054/01A 1996-11-26 2001-05-17 Multimeric erythropoietin with increased biological activity Abandoned AU4605401A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU46054/01A AU4605401A (en) 1996-11-26 2001-05-17 Multimeric erythropoietin with increased biological activity

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/756134 1996-11-26
AU46054/01A AU4605401A (en) 1996-11-26 2001-05-17 Multimeric erythropoietin with increased biological activity

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU53779/98A Division AU730122B2 (en) 1996-11-26 1997-11-24 Multimeric erythropoietin with increased biological activity

Publications (1)

Publication Number Publication Date
AU4605401A true AU4605401A (en) 2001-07-26

Family

ID=3733141

Family Applications (1)

Application Number Title Priority Date Filing Date
AU46054/01A Abandoned AU4605401A (en) 1996-11-26 2001-05-17 Multimeric erythropoietin with increased biological activity

Country Status (1)

Country Link
AU (1) AU4605401A (en)

Similar Documents

Publication Publication Date Title
AU730122B2 (en) Multimeric erythropoietin with increased biological activity
US5919758A (en) Modified polypeptides with altered biological activity
US5580853A (en) Modified polypeptides with increased biological activity
ES2298402T3 (en) PEGILED AND DIGLICOSILED Erythropoietin.
US5711944A (en) Interferon polymer conjugates
JP5334347B2 (en) Chemically modified novel erythropoietin stimulating protein compositions and methods
HU221208B1 (en) Conjugates of a polypeptide and a biocompatible polymer
CA1309046C (en) Conjugates of superoxide dismutase coupled to high molecular weight polyalkylene glycols
JP5961730B2 (en) Modified erythropoietin
US6420340B2 (en) Chemical modification of proteins to improve biocompatibility and bioactivity
US5900404A (en) Chemical modification of proteins to improve biocompatibility and bioactivity
AU4605401A (en) Multimeric erythropoietin with increased biological activity
US20040235719A1 (en) Soluble protein-polymer systems for drug delivery
KR100534622B1 (en) Erythropoietin and Human Serum Albumin
EP0537222A1 (en) Immunoreactive compound
JPH01104164A (en) Superoxide dismutase derivative and production thereof
AU2002255172A1 (en) Soluble protein-polymer systems for drug delivery
AU8063591A (en) Immunoreactive compound

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application