NZ744721B2 - Treatment for rheumatoid arthritis - Google Patents

Treatment for rheumatoid arthritis Download PDF

Info

Publication number
NZ744721B2
NZ744721B2 NZ744721A NZ74472113A NZ744721B2 NZ 744721 B2 NZ744721 B2 NZ 744721B2 NZ 744721 A NZ744721 A NZ 744721A NZ 74472113 A NZ74472113 A NZ 74472113A NZ 744721 B2 NZ744721 B2 NZ 744721B2
Authority
NZ
New Zealand
Prior art keywords
antibody
treatment
dose
use according
weeks
Prior art date
Application number
NZ744721A
Other versions
NZ744721A (en
Inventor
Stefan Haertle
Stephane Leclair
Amgad Shebl
Stefan Steidl
Original Assignee
Morphosys Ag
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 Morphosys Ag filed Critical Morphosys Ag
Publication of NZ744721A publication Critical patent/NZ744721A/en
Publication of NZ744721B2 publication Critical patent/NZ744721B2/en

Links

Abstract

The present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, comprising administering to said patient an anti-anti?GSM?CSF antibody intravenously weekly at a dose of about 1.0 mg/Kg, or at a subcutaneous dose which achieves a blood concentration equal to that dose. d concentration equal to that dose.

Description

TREATMENT FOR RHEUMATOID ARTHRITIS This application is a divisional ation of New Zealand application no. 705635 (which is the national phase entry of , published as WO2014/044768) dated 19 September 2013, and claims the benefit of and priority to EP 12185235.4 filed 20 September 2012 and U.S. Provisional ation No. 61/703,871, filed 21 September 2012, which are hereby incorporated by reference in their entirety.
SUMMARY OF THE INVENTION The ion the subject of the present invention particularly is set out in the following clauses: 1. The use of a pharmaceutical composition comprising an anti‐GM‐CSF antibody , in the cture of a medicament, for the treatment of a patient suffering from rheumatoid arthritis, the treatment comprising administering the anti‐GM‐CSF antibody enously weekly at a dose of 1.0 mg/Kg, or at a subcutaneous dose which achieves a blood concentration equal to that dose, wherein the anti‐GM‐CSF dy comprises a variable heavy chain of the sequence: QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMNWVRQAPGKGLEWVSGIENKYAGGATYYAASVKGR FTISRDNSKNTLYLQMNSLRAEDTAVYYCARGFGTDFWGQGTLVTVSS (SEQ ID NO.: 8) and a variable light chain of the sequence: DIELTQPPSVSVAPGQTARISCSGDSIGKKYAYWYQQKPGQAPVLVIYKKRPSGIPERFSGSNSGNTATLTISG TQAEDEADYYCSAWGDKGMVFGGGTKLTVLGQ (SEQ ID NO.: 9). 2. The use of 1, wherein the pharmaceutical composition comprises pharmaceutically acceptable carriers, excipients or stabilizers which are a buffer which is histidine, a sugar which is sorbitol and a non‐ionic surfactant which is polysorbate‐80. 3. The use of 2 n the composition consists of 30mM histidine, pH 6.0, 200mM sorbitol and 0.02% polysorbate‐80 as carriers, excipients or stabilizers. 4. The use of any one of 1 to 3, wherein the aneous dose is at least about 2mg/kg, about 3.0mg/kg or about 4.0 mg/kg.
. The use of any one of 1 to 3, wherein the subcutaneous dose is a fixed dose of between about 40 mg and 400 mg. 6. The use according to 5 wherein the fixed dose is 150 mg. 7. The use of any one of 1 to 6, wherein the subcutaneous dose is administered weekly, biweekly, monthly or hly. 8. The use according to any one of 1 to 7, wherein the treatment comprises stering the antibody to said patient in a manner to achieve to a serum tration of said antibody of at least 2 µg/ml in said patient over the duration of said treatment. (followed by page 2A) 9. The use according to any one of 1 to 8 wherein the treatment achieves a mean change in ACR20 score of at least 30.4 over four weeks of treatment.
. The use according to 9 wherein the treatment achieves a mean change in ACR20 score of at least 68.2 over four weeks of ent. 11. The use according to any one of 1 to 8, wherein the treatment achieves a mean change in ACR score of at least 26.1 over eight weeks of treatment. 12. The use according to 11, n the treatment achieves a mean change in ACR 20 score of at least 31.8 over eight weeks of treatment. 13. The use according to any one of 1 to 12, wherein the treatment ses administering said antibody in ation with a disease‐modifying anti‐rheumatic drug (DMARD). 14. The use according to 13, wherein the DMARD is methotrexate.
. A use according to 1, substantially as herein described and exemplified.
The present invention is further described below. In certain aspects the invention is described in broader terms which are heless included herein for completeness. (followed by page 2B) wed by page 3) to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
In another aspect, the present invention provides an anti-GM-CSF antibody, wherein said anti-GM-CSF antibody is an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7) for use in the treatment of a patient suffering from rheumatoid tis, wherein said antibody is administered intravenously at a dose of about 1.0 mg/kg or at a dose of about 1.5 mg/kg and wherein said antibody in administered weekly over at least four weeks.
In r aspect, the t invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, n said antibody is administered to said patient in a manner to e a therapeutically effective antibody level in the blood of said patient equal or higher compared to the enous administration of said dy at a dose of at least 1.0 mg/kg or at least 1.5mg/kg when administered weekly over at least four weeks, and n said anti-GM-CSF antibody is administered in combination with a DMARD, such as methotrexate.
In an embodiment, the administration of said antibody to achieve such a therapeutically effective amount comprises the administration of said antibody intravenously at a dose at least 0.6, at least 0.7, at least 0.8, at least 0.9 or at least 1.0 mg/kg. In other embodiments, the antibody of the present invention is administered intravenously at a dose of about 1.0 mg/kg or a dose of about 1.5 mg/kg. Administration may be monthly, biweekly (every two weeks) or weekly.
In another aspect, the present invention provides an anti-GM-CSF antibody for use in the ent of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient subcutaneously in a manner to achieve a therapeutically ive antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg or at least 1.5mg/kg when administered weekly over at least four weeks, and wherein said M- CSF antibody is stered in ation with a DMARD, such as methotrexate.
In an ment, the administration of said antibody to achieve such a therapeutically effective amount comprises the administration of said antibody subcutaneously at a dose of at least 1.0, at least 1.5, at least 2.0, at least 2.5, at least 3.0, at least 3.5 or at least 4.0 mg/kg. In other embodiments, the antibody of the present invention is administered subcutaneously at a dose of about 2.0 mg/kg, a dose of about 3.0 mg/kg or a dose of about 4.0 mg/kg. Administration may be monthly, biweekly (every two weeks) or weekly.
In an embodiment, the administration of said antibody to achieve such a therapeutically effective amount ses the administration of said antibody subcutaneously at a fixed dose of about 40 mg, at a fixed dose of 75 mg, at a fixed dose of 100 mg, at a fixed dose of 140 mg, at a fixed dose of 150 mg, at a fixed dose of 180 mg, at a fixed dose of 200 mg, at a fixed dose of 280 mg, at a fixed dose of 300 mg or at a fixed dose of 400 mg.. Administration of fixed doses may be every week, every second week, every third week, every fourth week or every sixth week.
In another aspect, the present ion es a method of treating a patient suffering from rheumatoid arthritis, said method sing administering to said t an anti-GM- CSF antibody subcutaneously at (i) a dose of at least 1.0 mglkg, or (ii) a fixed dose of between 40 mg and 400 mg.
The anti-GM-CSF dy may be administered to said patient in a manner to achieve to a serum concentration of said antibody at at least 2 pg/ml in said patient over the duration of said treatment. The antibody may be stered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
In another aspect, the present invention provides an anti-GM-CSF antibody for inhibiting progression of structural joint damage in a rheumatoid arthritis patient comprising administering to said patient said antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
BRIEF PTION OF THE DRAWINGS Figure 1 shows the amino acid sequence and the DNA sequence of MORO4357.
Figure 2 shows the mean changes of the DA828 score after four weeks (left panel) and after eight weeks (right panel) of treatment ed. DA828 score changes are compared to baseline levels, i.e. disease status prior to treatment.
Figure 3 shows the average ACR20 score of all treatment arms after four weeks. An increase of the ACR20 scores ponds to an improvement of the severity of disease.
Figure 4 shows the average ACR20 score of all treatment arms after eight weeks. An increase of the ACR20 scores corresponds to an improvement of the severity of disease.
DESCRIPTION The terms “GM-CSF” and “GMCSF” refer to the protein known as GM-CSF or Granulocyte-macrophage colony-stimulating factor, having the following synonyms: Colonystimulating factor 2, CSF2, GMCSF, GM-CSF, Granulocyte—macrophage colony-stimulating factor, MGC131935, MGC138897, Molgramostin, Sargramostim. Human GM—CSF has the amino acid sequence of (UniProt P04141): MWLQSLLLLGTVACSISAPARSPSPSTQPWEHVNAIQEARRLLNLSRDTAAEMNET VEVISEMFDLQEPTCLQTRLELYKQGLRGSLTKLKGPLTMMASHYKQHCPPTPETS CATQIITFESFKENLKDFLLVIPFDCWEPVQE (SEQ ID NO.: 1) “MOR103” is an anti-GM-CSF antibody whose amino acid sequence and DNA sequence is provided in Figure 1. “MOR103” and “MOR04357” and 57” are used as synonyms to describe the antibody shown in Figure 1. MOR04357 comprises an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence GM (SEQ ID NO.: 7). MOR04357 comprises a variable heavy chain of the SGGGLVQPGGSLRLSCAASGFTFSSYWMNWVRQAPGKGLEWVSGIENKYAGGA TYYAASVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGFGTDFWGQGTLVTVSS (SEQ ID NO.: 8) and a variable light chain of the sequence DIELTQPPSVSVAPGQTARISCSGDSIGKKYAYWYQQKPGQAPVLVIYKKRPSGIPERFSGS NSGNTATLTISGTQAEDEADYYCSAWGDKGMVFGGGTKLTVLGQ (SEQ ID NO.: 9).
In certain embodiments, the antibody used in the present invention is an antibody ic for . In other embodiments, the antibody used in the present ion is an antibody specific for a polypeptide encoding an amino acid ce comprising SEQ ID NO.: 1.
As used herein, fically for" or "specifically binding to" refers to an antibody selectively or preferentially binding to GM—CSF. Preferably the binding affinity for antigen is of Kd value of 10'9 moI/I or lower (e.g. 10'10 mol/I), ably with a Kd value of 10'10 moI/I or lower (e.g. 10'12 moI/I). The binding affinity is determined with a standard binding assay, such as surface plasmon resonance technique (BIACORE®).
In certain embodiments, the antibody used in the present invention is MOR103. In other embodiments, the antibody used in the present invention is an antibody sing an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of ce GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7). In other embodiments, the antibody used in the present invention is an antibody comprising a variable heavy chain of the sequence QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMNWVRQAPGKGLEWVSGIENKYAGGA TYYAASVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGFGTDFWGQGTLVTVSS (SEQ ID NO.: 8) and a variable light chain of the sequence DIELTQPPSVSVAPGQTARISCSGDSIGKKYAYWYQQKPGQAPVLVIYKKRPSGIPERFSGS NSGNTATLTISGTQAEDEADYYCSAWGDKGMVFGGGTKLTVLGQ (SEQ ID NO.: 9). In other ments, the antibody used in the present invention is an antibody which cross- competes with an dy comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7). In other embodiments, the dy used in the present invention is an antibody which binds to the same epitope like an antibody specific for GM-CSF comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7).
The term ody" is used in the broadest sense and ically covers monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
"Antibody fragments" herein comprise a portion of an intact antibody which retains the ability to bind antigen. Examples of antibody nts include Fab, Fab', F(ab')2, and Fv nts; diabodies; linear antibodies; single—chain antibody molecules; and multispecific dies formed from antibody fragments.
The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous dies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variants that may arise during production of the monoclonal dy, such variants lly being t in minor amounts. In contrast to polyclonal antibody preparations that typically include different antibodies directed against different inants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the onal antibodies are advantageous in that they are uncontaminated by other immunoglobulins.
The monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or ss, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or ing to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological ty.
"Humanized" forms of non-human (e.g., murine) dies are chimeric antibodies that contain minimal ce derived from man globulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity. In some instances, framework region (FR) residues of the human immunoglobulin are replaced by ponding non- human residues. Furthermore, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to r refine antibody performance. In l, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable regions correspond to those of a non- human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence, except for FR substitution(s) as noted above. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region, typically that of a human globulin.
A "human antibody" herein is one comprising an amino acid sequence structure that corresponds with the amino acid sequence structure of an antibody able from a human B- cell, and includes antigen—binding fragments of human antibodies. Such antibodies can be identified or made by a variety of techniques, including, but not limited to: tion by transgenic animals (e.g., mice) that are capable, upon immunization, of producing human antibodies in the absence of endogenous immunoglobulin; selection from phage display libraries expressing human antibodies or human antibody; generation via in vitro activated B; and isolation from human antibody producing omas.
In certain embodiments, the antibody used in the present invention is a onal In other embodiments, the antibody used in the present invention is a chimeric, a humanized or a human antibody. In preferred embodiments, the antibody used in the present invention is a human dy.
In certain embodiments, the antibody used in the present invention is administered in combination with an additional drug that treats RA.
The additional drug may be one or more medicaments, and include, for example, suppressive agents, non—steroidal nflammatory drugs (NSAIDs), disease modifying anti-rheumatic drugs (DMARDs) such as methotrexate (MTX), anti-B-cell surface marker antibodies, such as anti—CD20 antibodies (e.g. rituximab), TNF-alpha-inhibitors, osteroids, and mulatory modifiers, or any combination thereof. Optionally, the second or additional drug is selected from the group consisting of non- biological DMARDs, NSAIDS, and corticosteroids.
These additional drugs are generally used in the same dosages and with stration routes as used hereinbefore and hereinafter. If such additional drugs are used at all, preferably, they are used in lower amounts than if the first medicament were not present, especially in subsequent dosings beyond the initial dosing with the first ment, so as to eliminate or reduce side effects caused thereby. The combined administration of an additional drug includes co—administration (concurrent stration), using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents (medicaments) simultaneously exert their biological activities.
The term “DMARD” refers to Disease-Modifying Anti-Rheumatic Qrugs" and includes among others hydroxycloroquine, sulfasalazine, methotrexate, leflunomide, oprine, D- penicillamine, gold salts (oral), gold salts (intramuscular), minocycline, cyclosporine including cyclosporine A and topical cyclosporine, and TNF-inhibitors, including salts, variants, and derivatives thereof. Exemplary DMARDs herein are non-biological, i.e. classic, DMARDs, including, azathioprine, chloroquine, hydroxychloroquine, leflunomide, methotrexate and alazine.
Methotrexate is an especially preferred DMARD of the present ion. Therefore, in certain embodiments, the antibody used in the present invention is administered in combination with a DMARD. In other embodiments, the antibody used in the present invention is administered in combination with methotrexate.
A "TNF-inhibitor" as used herein refers to an agent that inhibits, to some extent, a biological function of TNF-alpha, generally h binding to TNF-alpha and/or its or and neutralizing its ty. Examples of TNF inhibitors include etanercept (ENBREL®), infliximab (REMICADE®), adalimumab (HUMIRA®), certolizumab pegol A®), and golimumab (SIMPON|®).
"Treatment" of a patient or a subject refers to both therapeutic ent and prophylactic or preventative measures. The terms "effective amount" or “therapeutically effective” refer to an amount of the antibody that is effective for treating rheumatoid arthritis. Such effective amount can result in any one or more of ng the signs or symptoms of RA (e.g. achieving ACRZO), reducing disease activity (e.g. Disease Activity Score, DASZO), slowing the progression of structural joint damage or improving physical on. In one embodiment, such clinical response is comparable to that achieved with intravenously administered anti-GM-CSF antibody.
The antibody of the present invention may be administered in different suitable forms.
Potential forms of stration include systemic administration (subcutaneous, intravenous, intramuscular), oral administration, inhalation, transdermal administration, topical application (such as topical cream or ointment, etc.) or by other methods known in the art. The doses (in mg/kg) specified in the t invention refer to milligrams of antibody per kilogram of body weight of the patient. In vitro cell based assays showed that an M- CSF antibody (MOR103) is capable of inhibiting several GM-CSF mediated responses. ted responses include TF—1 cell eration, STAT5 phosphorylation, polymorphonuclear neutrophils (PMN) migration, PMN up-regulation of CD11b, monocyte up-regulation of MHC II, and eosinophil suwival. Complete tory effects were generally reached at concentrations of about 0.2 ug/ml anti-GM-CSF antibody. GM-CSF concentrations up to 1 ng/ml were applied in such s. As a reference, GM-CSF levels in the synovial fluid of RA ts were reported to be <500 pg/ml. It is reasonable to consider that similar GM-CSF concentrations as used in these in vitro studies are t in affected tissues of RA patients To effectively treat RA it may be important for an anti—GM-CSF antibody to penetrate the synovium. There is evidence to suggest that monoclonal antibodies can distribute into the synovium when dosed subcutaneously or intravenously. Based on a predicted penetration rate of 30%, continuous GM-CSF production and considering patient heterogeneity, the l or sub-optimal clinical effect level in RA patients is anticipated to be at a serum concentration of approximately 2 ug/ml antibody (thus, approximately 10-fold higher than the inhibitory tration derived from in vitro studies).
A specific anti-GM-CSF antibody (MOR103) has been administered to patients with active rheumatoid arthritis who ed 4 intravenous weekly doses of 0.3, 1, and 1.5 mg/kg. The anti-GM-CSF antibody showed significant clinical efficacy on DA828, EULAR, ACR20, ACR50, ACR70 and tender joint counts following once a week dosing with 1 and 1.5 mg/kg as compared to placebo.
In certain embodiments, the antibody of the present invention is administered intravenously. In other ments, the antibody of the present invention is administered subcutaneously.
From other eutic antibodies it is known that a concentration that leads to a certain level of the antibody in the blood when administered intravenously corresponds to about 50- 76% of the blood concentration achieved when the same antibody concentration is administered subcutaneously (Meibohm, B.: Pharmacokinetics and Pharmacodynamics of Biotech Drugs, Wiley-VCH, 2006). For MOR103 this ratio was determined to be 52%, Le. a given concentration administered subcutaneously leads to a blood concentration which is equivalent to about 52% of the blood concentration when the same given concentration is administered intravenously. ore, the concentration of a subcutaneous formulation needs to be about twice as high to achieve the same drug blood level as compared to an intravenous formulation.
In certain embodiments the blood level to be achieved in a patient is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
In ative embodiments said blood concentration to be achieved is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a doses of at least 0.4, 0.5, 0.6, 0.7, 0.8 or 0.9 mg/kg when stered weekly over at least four weeks. In alternative embodiments the blood level to be achieved in a patient is equal or higher ed to the blood concentration achieved with intravenous stration of the antibody of the present invention at a dose of at least 1.0 mg/kg when administered weekly over at least two weeks or at least three weeks.
In alternative embodiments the blood level to be achieved in a patient is equal or higher compared to the blood tration ed with intravenous administration of the antibody of the present invention at a dose of at least 1.0 mg/kg when administered biweekly over at least two weeks or at least four weeks.
In certain embodiments, the antibody of the present invention is administered intravenously. In other embodiments, the antibody of the t ion is administered intravenously at a dose at least 0.6, at least 0.7, at least 0.8, at least 0.9 or at least 1.0 mg/kg. In other embodiments, the antibody of the present ion is administered intravenously at a dose of about 1.0 mg/kg or a dose of about 1.5 mg/kg.
In certain embodiments, the antibody of the present invention is administered subcutaneously. Various dosing regimen have been simulated using the subcutaneous ry of MOR103 in order to e plasma concentrations that are similar those obtained after 1 mg/kg iv, a dose that was efficacious in RA. The majority of simulations produce trough concentration values greater than 2 ug/mL, a value that is believed to be the minimum blood concentration that is required to produce efficacy in the context of an anti- GM-CSF antibody. These studies indicate that subcutaneous doses of 1, 2, 3 and 4 mg/kg can produce plasma concentration similar to 1 mg/kg, lV depending on the dosing frequency.
In other ments, the antibody of the present invention is stered subcutaneously at a dose at least 1.0, at least 1.5, at least 2.0, at least 2.5, at least 3.0, at least 3.5 or at least 4.0 mg/kg. In other embodiments, the antibody of the present invention is administered subcutaneously at a dose of about 2.0 mg/kg, a dose of about 3.0 mg/kg or a dose of about 4.0 mg/kg. In certain embodiments, the antibody of the present ion is subcutaneously administered biweekly, monthly or bimonthly.
In other embodiments, the antibody of the present invention is administered subcutaneously at a fixed dose. In such “fixed dose” treatment the antibody is administered at a certain, fixed, concentration, i.e. without taking into account a patient’s body weight. In certain embodiments, the antibody of the present invention is stered at a fixed dose of between 40 mg and 400 mg, optionally at a fixed dose of 75 mg, at a fixed dose of 100 mg, at a fixed dose of 140 mg, at a fixed dose of 150 mg, at a fixed dose of 180 mg, at a fixed dose of 200 mg, at a fixed dose of 280 mg, at a fixed dose of 300 mg or at a fixed dose of 400 mg. Administration of fixed doses may be every week, every second week, every third week, every fourth week or every sixth week. Typically, the antibody will be administered weekly at a fixed dose.
In an embodiment, the antibody will be administered weekly, at a fixed aneous dose of 40, 56, 70, 75 100, 140, 150, 180, 200, 210, or 280 mg.
In an embodiment, the antibody will be stered biweekly, at a fixed subcutaneous dose of 70, 75, 100, 140, 150, 180, 200, 210, 280 or 300 mg.
In an embodiment, the antibody will be administered monthly, at a fixed subcutaneous dose of 100, 140, 150, 180, 200, 210, 280, 300, 320, 350. 360 or 400 mg.
In an embodiment, the antibody is administered in a dose sufficient to maintain trough concentration of antibody of at least 2 ug/mL. The trough concentration of antibody may be maintained at 2.0 ug/mL, 2.5 ug/mL, 3.0 ug/mL, 3.5 ug/mL, 4.0 ug/mL, 4.5 ug/mL or 5.0 ug/mL, during the course of y.
In alternative embodiments, the antibody will be administered weekly, at a fixed subcutaneous dose of 28 or 35 mg, In certain embodiments, the present invention provides an M-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mgikg when administered weekly over at least four weeks.
In certain embodiments, the t invention provides a method to treat a patient suffering from rheumatoid arthritis, said method comprising administering to said patient an anti-GM-CSF antibody in a manner to e a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
In certain embodiments, the present invention provides an anti-GM-CSF antibody for inhibiting progression of structural joint damage in a rheumatoid arthritis patient comprising stering to said t said antibody in a manner to achieve a therapeutically effective dy level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when stered weekly over at least four weeks.
The terms “drug” and “medicament" refer to an active drug to treat rheumatoid arthritis or joint damage or symptoms or side effects associated with RA. The term "pharmaceutical formulation" refers to a preparation which is in such form as to permit the ical activity of the active ingredient or ingredients, i.e. the antibody of the present invention, to be effective, and which ns no additional ents which are unacceptably toxic to a subject to which the formulation would be administered. Such formulations are sterile.
The antibody herein is preferably recombinantly produced in a host cell transformed with nucleic acid sequences encoding its heavy and light chains (e.g. where the host cell has been transformed by one or more s with the nucleic acid therein). The preferred host cell is a mammalian cell, most preferably a PER.CB cell.
Therapeutic formulations of the antibody of the present invention are ed for storage by mixing the antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers in the form of lyophilized formulations or aqueous solutions. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, histidine and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium de; , butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, n, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, ine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or ns; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt—forming counter—ions such as ; metal complexes {e.g. Zn- protein complexes); and/or non—ionic surfactants such as TWEENTM (such as Tween-80), PLURONICST'V' or polyethylene glycol (PEG).
In certain embodiments, the present invention es a pharmaceutical composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier and/or excipient for use in any of the methods provided in the present invention. In certain embodiments, the formulation for the antibody of the present invention ts of 30 mM histidine, pH 6.0, 200 mM sorbitol and 0.02% 80. In other embodiments, the formulation for the antibody of the present invention consists of PBS, pH 6.2 (0.2 g/l KCI, 0.96 g/l KH2PO4, 0.66 g/l Na2HPO4 x 7H20, 8 g/l NaCl).
EXAMPLES Example 1: Design and concept of a clinical Phase lb/Phase Ila trial A multi-center, randomized, double—blinded, placebo—controlled study to evaluate the safety, preliminary clinical activity and immunogenicity of multiple doses of MOR103 administered intravenously to patients with active rheumatoid arthritis was conducted.
Primary outcome measures were the adverse event rate and the safety e. Secondary outcome measures included DA828 , ACR scores and EULAR28 se criteria.
The al trial comprised three treatment arms. In each treatment arm patient ed either o or MOR103. The MOR103 doses were 0.3 mg/kg body weight for treatment arm 1, 1.0 mg/kg body weight for treatment arm 2 and 1.5 mg/kg body weight for ent arm 3. MOR103 and placebo were administered intravenously, weekly with 4 doses in total.
Summary of the treatment arms: Experimental: Group 1: Drug: MOR103 MOR103, experimental MOR103 0.3 mg/kg or placebo iv x 4 doses Biological: MOR103 0.3 mg/kg or placebo Experimental: Group 2: Drug: MOR103 MOR103, experimental MOR103 1.0 mg/kg or o iv x 4 doses Biological: MOR103 1.0 mg/kg or placebo Experimental: Group 3: Drug: MOR103 MOR103, experimental MOR103 1.5 mg/kg or placebo iv x 4 doses ical: MOR103 1.5 mg/kg or placebo Eligible for participation in the study were patients of 18 years and older and of either sex (male and female). Healthy volunteers were not accepted.
Inclusion ia were as s: . Rheumatoid tis (RA) per revised 1987 ACR criteria . Active RA: 23 swollen and 3 tender joints with at least 1 swollen joint in the hand, excluding the PIP joints . CRP > 5.0 mg/L (RF and anti-CCP seronegative); CRP >2 mg/l (RF and/or anti- CCP seropositive) o DA828 S 5.1 . Stable regimen of concomitant RA therapy (NSAIDs, steroids, non- biological DMARDs). 0 Negative PPD tuberculin skin test Exclusion criteria were as follows: . Previous therapy with B or T cell depleting agents other than Rituximab (e.g.
Campath). Prior treatment with Rituximab, TNF-inhibitors, other biologics (e.g. anti- |L-1 therapy) and systemic immunosuppressive agents is allowed with a washout period.
. Any history of ongoing, significant or recurring infections . Any active inflammatory diseases other than RA . Treatment with a ic investigational drug within 6 months prior to screening . Women of childbearing potential, unless receiving stable doses of methotrexate or leflunomide . Significant cardiac or pulmonary disease (including methotrexate- ated lung toxicity) 0 Hepatic or renal insufficiency Example 2: t recruitment and patient population Clinical sites for patient recruitment, screening and treatment were located in Bulgaria, Germany, the Netherlands, Poland and the Ukraine. 96 patients were included in the trial. 27 patients received placebo, 24 patients received MOR103 at a dose of 0.3 mg/kg, 22 patients ed MOR103 at a dose of 1.0 mg/kg and 23 patients received MOR103 at a dose of 1.5 mg/kg. The average age and the average Body Mass Index (BMI) was about the same for all treatment groups. Key teristics are summarized in the following Table: MOR 103 Active Treatment Groups Characteristic o 0.3 mglkg 1.0 mglkg 1.5 mglkg Total active N=27 N=24 N=22 N=23 N=69 Age £53.8 £514 £49 5£3 5£3.3 Em,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,263263 ..................................56}....................................25.7.. ..................................256 ,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,, White $27 $24 2$2 $23 $69 90% of all patients of the study were previously treated with DMARDs. The most commonly used DMARD was rexate (75% of all ts). The rate of previous ent with DMARDs was comparable in all treatment arms.
Prior to administration of MOR103 or the placebo the disease activity of all ts was measured according to accepted guidelines by calculating the DA828 score, a 28—joint Disease Activity Score (see e.g. Ann Rheum Dis (2009) 68, 954-60). DA828 score is a ted and commonly used tool to quantify the e status of RA patients. The average DA828 score was comparable for all treatment arms.
Example 3: Safety profile Based on the available observed safety data, MOR103 showed a favorable safety profile among all doses tested. The key observations are as s: - No deaths were observed during the conduct of the trial . No infusion related reactions were observed . Two serious adverse events (SAEs) were ed: — One patient in the placebo group developed paronychia — One patient in the 0.3 mg/kg treatment arm developed pleurisy . More treatment-emergent adverse effects (TEAEs) were observed in the placebo group (25.9%) than in the active groups (14.5%) - Most TEAEs were mild . No severe TEAEs were observed in the active groups In summary, it can be concluded that treatment with MOR103 at all doses tested is safe.
Two serious adverse events were observed, both none in the treatment arms that showed clinical efficacy (see below). Sub-cutaneous administration of MOR103 at a dose that leads to an antibody drug level in the blood of patients equivalent to the intravenous application of the present study is expected to show a similar safety profile.
Example 4: Efficacy — DA828 4 weeks and 8 weeks after the first administration of MOR103 (or placebo) the DA828 scores of all patients was determined. A decrease in DA828 scores correlates to diminished disease severity. Results are shown in Figure 2 as the mean changes ed to baseline, i.e. disease status prior to treatment.
The placebo group only shows marginal changes. Patients treated with MOR103 at 0.3 mg/kg showed a slight decrease in DA828 scores, ting slightly less severity of the disease. In contrast, patients treated with MOR103 at 1.0 mg/kg or with 1.5 mg/kg showed a significant decrease in DA828 , indicating the high efficacy of MOR103 at these doses.
Example 5: Efficacy — ACR20 As another measure of efficacy the ACR20 criteria were used. ACR ia measure improvement in tender or swollen joint counts and improvement in certain other parameters.
The procedure to measure ACR scores is highly standardized. The present clinical trial applied the respective applicable guidelines. Results are depicted in Figures 3 and 4. A higher score corresponds to an improvement in the severity of the disease.
In line with the results of the DA828 scores (see e 4), also the ACR scores show a strong clinical improvement of patients’ condition upon treatment with either 1.0 mg/kg MOR103 or 1.5 mg/kg MOR103. The improvement after 4 weeks is highly significant for the 1.0 mg/kg group (p<0.0001). Taken together, the ACR20 scores confirm the surprising finding that the efficacy of MOR103 can y be shown with a comparably low number or patients in each ent arm and a comparably short treatment period. e 6: Clinical trial with additional doses of MOR103 The clinical trial set out herein above is repeated with additional doses of MOR103.
MOR103 is administered to patients intravenously at a dose of 0.5 mg/kg (treatment arm 1) and 0.75 mg/kg (treatment arm 2). All other parameters are identical to e 1.
Both treatment arms show a favorable safety profile and demonstrate clinical efficacy as ed by DA828 scores and ACR20 scores.
Example 7: Clinical trial with a sub-cutaneous ation of MOR103 The clinical trial set out herein above is ed with a sub-cutaneous formulation of MOR103. In order to achieve similar levels of MOR103 in the blood of patients as observed for intravenous treatment, the sub—cutaneous dose of MOR103 is increased.
In different treatment arms MOR103 is administered to ts at 1.5 mg/kg, 2.0 mg/kg, 3.0 mg/kg and 4.0 mg/kg. The drug is administered sub-cutaneously, either biweekly, monthly or bimonthly. All other parameters are identical to Example 1.
All treatment arms show a ble safety profile and demonstrate clinical efficacy as measured by DA828 scores and ACRZO scores.
Example 8: al trial with a sub-cutaneous formulation of MOR103 at a fixed dose Example 7 is repeated with a fixed dose of MOR103. In different treatment arms MOR103 is administered to patients at fixed dose of 75 mg, of 100 mg, of 150 mg, of 200 mg, of 300 mg and of 400 mg. The drug is administered sub—cutaneously every week, every second week, every fourth week or every sixth week. All other parameters are identical to the Examples described herein above.
All treatment arms show a favorable safety e and demonstrate clinical efficacy as measured by DASZ8 scores and ACRZO scores.

Claims (15)

What we claim is:
1. The use of a pharmaceutical composition comprising an anti‐GM‐CSF antibody , in the manufacture of a medicament, for the treatment of a patient suffering from rheumatoid arthritis, the treatment comprising administering the anti‐GM‐CSF antibody intravenously weekly at a dose of 1.0 mg/Kg, or at a aneous dose which achieves a blood concentration equal to that dose, wherein the anti‐GM‐CSF antibody comprises a le heavy chain of the sequence: QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMNWVRQAPGKGLEWVSGIENKYAGGATYYAASVKG RFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGFGTDFWGQGTLVTVSS (SEQ ID NO.: 8) and a le light chain of the sequence: DIELTQPPSVSVAPGQTARISCSGDSIGKKYAYWYQQKPGQAPVLVIYKKRPSGIPERFSGSNSGNTATLTIS GTQAEDEADYYCSAWGDKGMVFGGGTKLTVLGQ (SEQ ID NO.: 9).
2. The use of claim 1, wherein the pharmaceutical composition comprises ceutically acceptable carriers, excipients or stabilizers which are a buffer which is histidine, a sugar which is sorbitol and a non‐ionic surfactant which is polysorbate‐80.
3. The use of claim 2 wherein the composition consists of 30mM histidine, pH 6.0, 200mM sorbitol and 0.02% polysorbate‐80 as carriers, excipients or stabilizers.
4. The use of any one of claims 1 to 3, wherein the subcutaneous dose is at least about 2mg/kg, about 3.0mg/kg or about 4.0 mg/kg.
5. The use of any one of claims 1 to 3, wherein the subcutaneous dose is a fixed dose of between about 40 mg and 400 mg.
6. The use according to claim 5 wherein the fixed dose is 150 mg.
7. The use of any one of claims 1 to 6, wherein the subcutaneous dose is administered weekly, biweekly, y or bimonthly.
8. The use according to any one of claims 1 to 7, n the treatment comprises administering the antibody to said patient in a manner to achieve to a serum concentration of said antibody of at least 2 µg/ml in said patient over the on of said treatment.
9. The use according to any one of claims 1 to 8 wherein the treatment achieves a mean change in ACR20 score of at least 30.4 over four weeks of treatment.
10. The use according to claim 9 n the treatment es a mean change in ACR20 score of at least 68.2 over four weeks of treatment.
11. The use according to any one of claims 1 to 8, wherein the treatment achieves a mean change in ACR 20 score of at least 26.1 over eight weeks of treatment.
12. The use according to claim 11, wherein the treatment achieves a mean change in ACR 20 score of at least 31.8 over eight weeks of treatment.
13. The use according to any one of claims 1 to 12, wherein the treatment comprises administering said antibody in combination with a disease‐modifying anti‐rheumatic drug
14. The use according to claim 13, wherein the DMARD is methotrexate.
15. A use according to claim 1, substantially as herein described and exemplified.
NZ744721A 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis NZ744721B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP12185235 2012-09-20
EP12185235.4 2012-09-20
US201261703871P 2012-09-21 2012-09-21
US61/703,871 2012-09-21
NZ70563513 2013-09-19

Publications (2)

Publication Number Publication Date
NZ744721A NZ744721A (en) 2021-01-29
NZ744721B2 true NZ744721B2 (en) 2021-04-30

Family

ID=

Similar Documents

Publication Publication Date Title
US20210130451A1 (en) Treatment for rheumatoid arthritis
US20230303677A1 (en) Methods of treating new-onset plaque type psoriasis using il-17 antagonists
US20200231666A1 (en) Treatment paradigm
US20230009657A1 (en) Methods of treating lupus nephritis using interleukin-17 (il-17) antagonists
US12037387B2 (en) PAN-ELR+ CXC chemokine antibodies for the treatment of hidradenitis suppurativa
JP7132256B2 (en) Treatment of rheumatoid arthritis
NZ744721B2 (en) Treatment for rheumatoid arthritis
NZ744721A (en) Treatment for rheumatoid arthritis
WO2018158741A1 (en) Psoriasis disease modification following long-term treatment with an il-17 antagonist
CN113993543B (en) Combination therapy using anti-CD 38 antibodies
JP2023504679A (en) Methods of treating lichen planus using interleukin-17 (IL-17) antagonists
JP2024516019A (en) Treatment for systemic lupus erythematosus using anti-baffr antibodies - Patents.com