MODIFIED GROWTH HORMONE POLYPEPTIDES

-

We describe modified growth hormone fusion proteins and dimers comprising said fusion proteins; nucleic acid molecules encoding said proteins and methods of treatment that use said proteins in the treatment of conditions that result from growth hormone excess.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS REFERENCE TO RELATED APPLICATIONS

This is a divisional of U.S. patent application Ser. No. 12/678,282, filed Mar. 15, 2012, which is incorporation by reference in its entirety, and which is the U.S. national stage of PCT Application No. PCT/GB2008/003056, filed on Sep. 10, 2008, which was published in English under PCT Article 21(2). PCT Application No. PCT/GB2008/003056 claims the benefit of U.S. Provisional Application No. 60/979,010, filed on Oct. 10, 2007 and claims priority to Great Britain Patent Application No. GB 0719818.7, filed on Oct. 11, 2007.

The invention relates to modified growth hormone fusion proteins and dimers comprising said fusion proteins; nucleic acid molecules encoding said proteins and methods of treatment that use said proteins.

Growth hormone (GH) is an anabolic cytokine hormone important for linear growth in childhood and normal body composition in adults. The regulation of GH activity is complex and involves a number of interacting polypeptide and peptide agonists and antagonists. GH can mediate its effects either directly by binding growth hormone receptor or indirectly by stimulating production of Insulin-like growth factor-1 (IGF-1). A major role of GH is therefore the stimulation of the liver to produce IGF-1. In addition the secretion of GH is controlled by two peptide hormones with opposing activities. Growth hormone releasing hormone (GHRH) is a 44 amino acid peptide produced by the arcuate nucleus of the hypothalamus. It functions to stimulate GH production by the anterior pituitary gland. Somatostatin is a peptide hormone that opposes the effects of GHRH and is processed from a larger pre-propeptide to a 14 and 28 amino acid form. Somatostatin is secreted by neuroendocrine cells of the periventricular nucleus of the hypothalamus into the hypothalamo-hypophysial portal system that connects with the anterior pituitary gland where it inhibits secretion of GH.

GH binds sequentially with two membrane bound growth hormone receptors (GHR) via two separate sites on GH referred as site 1 and site 2. Site 1 is a high affinity binding site and site 2 a low affinity site. A single GH molecule binds 1 GHR via site 1. A second GHR is then recruited via site 2 to form a GHR:GH:GHR complex. The complex is then internalised and activates a signal transduction cascade leading to changes in gene expression. The extracellular domain of GHR exists as two linked domains each of approximately 100 amino acids (SD-100), the C-terminal SD-100 domain (b) being closest to the cell surface and the N-terminal SD-100 domain (a) being furthest away. It is a conformational change in these two domains that occurs on hormone binding with the formation of the trimeric complex GHR-GH-GHR.

GH excess is associated with a number of disease conditions; for example acromegaly and pituitary gigantism. Most cases of GH excess result from a pituitary tumour in the somatotroph cells of the anterior pituitary gland. These tumours are benign and gradually increase the secretion of GH. The symptoms of growth hormone excess include thickening of the bones of the jaw, fingers and toes, pressure on the nerves/muscles and insulin resistance. The original treatment for tumour related GH excess is the surgical removal of the pituitary tumour. Latterly, the use of GH antagonists to inhibit GH signalling is becoming the preferred treatment due to its non-invasive nature. GH antagonists can either be recombinant forms of somatostatin or somatostatin analogues (e.g. octreotide, lanreotide) or modified GH.

A review of modified GH antagonists is provided in Kopchick (2003) European Journal of Endocrinology 148; S21-25 which describes a commercially available GH antagonist called pegvisomant which combines a modification to human GH at G120 with the addition of polyethylene glycol to increase the molecular weight of modified GH. A problem associated with the administration of growth hormone is its rapid clearance by renal filtration and/or proteolysis. The addition of polyethylene glycol reduces this loss. However, it is known that polyethylene glycol reduces the affinity of GH for GHR and therefore to compensate for this reduced affinity it is necessary to administer elevated amounts of modified GH. This can result in side effects. It would be desirable to provide a modified GH antagonist that can be administered at reduced dosage thereby avoiding the problems associated with pegvisomant. This can be a reduction in either to amount administered or a reduction in the frequency of administration.

In our co-pending application WO03/070765 we describe modified GH fusion proteins that include modifications to site 1 and site 2 in GH. These modified GH molecules are fused to an extracellular domain of GHR. We herein disclose modified GH fusion proteins that have vastly extended serum half life and form dimers which may be related to the improved pharmacokinetics of these fusion proteins either by reducing renal clearance or protecting modified GH from proteolysis. The improved pharmacokinetic profiles of these growth hormone fusion proteins will allow treatment regimes that do not require multiple administrations and reduce undesirable side effects.

SEQUENCE LISTING

The Sequence Listing is submitted as an ASCII text file 8617-84485-02_Sequence_Listing.txt, Aug. 22, 2012, 93.1 KB], which is incorporated by reference herein.

According to an aspect of the invention there is provided a nucleic acid molecule comprising a nucleic acid sequence selected from:

    • i) a nucleic acid sequence as represented in SEQ ID NO:1;
    • ii) a nucleic acid sequence as represented in SEQ ID NO:2;
    • iii) a nucleic acid sequence as represented in SEQ ID NO:4;
    • iv) a nucleic acid sequence as represented in SEQ ID NO:5;
    • v) a nucleic acid sequence as represented in SEQ ID NO:7;
    • vi) a nucleic acid sequence as represented in SEQ ID NO: 8;
    • vii) a nucleic acid sequence as represented in SEQ ID NO:10;
    • viii) a nucleic acid sequence as represented in SEQ ID NO:11;
    • ix) a nucleic acid sequence as represented in SEQ ID NO:13;
    • x) a nucleic acid sequence as represented in SEQ ID NO:14;
    • xi) a nucleic acid sequence as represented in SEQ ID NO:16;
    • xii) a nucleic acid sequence as represented in SEQ ID NO:17;
    • xiii) a nucleic acid sequence as represented in SEQ ID NO:19;
    • xiv) a nucleic acid sequence as represented in SEQ ID NO:20;
    • xv) a nucleic acid sequence as represented in SEQ ID NO:22;
    • xvi) a nucleic acid sequence as represented in SEQ ID NO:23;
    • xvii) a nucleic acid molecule comprising a nucleic sequence that hybridizes under stringent hybridization conditions to SEQ ID NO:1, 2, 4, 5, 7, 8, 10, 11, 13, 14, 16, 17, 19, 20, 22 or 23 and which encodes a polypeptide that has growth hormone receptor antagonist activity.

Hybridization of a nucleic acid molecule occurs when two complementary nucleic acid molecules undergo an amount of hydrogen bonding to each other. The stringency of hybridization can vary according to the environmental conditions surrounding the nucleic acids, the nature of the hybridization method, and the composition and length of the nucleic acid molecules used. Calculations regarding hybridization conditions required for attaining particular degrees of stringency are discussed in Sambrook et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 2001); and Tijssen, Laboratory Techniques in Biochemistry and Molecular Biology—Hybridization with Nucleic Acid Probes Part I, Chapter 2 (Elsevier, N.Y., 1993). The Tm is the temperature at which 50% of a given strand of a nucleic acid molecule is hybridized to its complementary strand. The following is an exemplary set of hybridization conditions and is not limiting:

Very High Stringency (Allows Sequences that Share at Least 90% Identity to Hybridize)

Hybridization: 5×SSC at 65° C. for 16 hours

Wash twice: 2×SSC at room temperature (RT) for 15 minutes each

Wash twice: 0.5×SSC at 65° C. for 20 minutes each

High Stringency (Allows Sequences that Share at Least 80% Identity to Hybridize)

Hybridization: 5×-6×SSC at 65° C.-70° C. for 16-20 hours

Wash twice: 2×SSC at RT for 5-20 minutes each

Wash twice: 1×SSC at 55° C.-70° C. for 30 minutes each

Low Stringency (Allows Sequences that Share at Least 50% Identity to Hybridize)

Hybridization: 6×SSC at RT to 55° C. for 16-20 hours

Wash at least twice: 2×-3×SSC at RT to 55° C. for 20-30 minutes each.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 1.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 2.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 4.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 5.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 7.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 8.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 10.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 11.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 13.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 14.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 16

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 17.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 19.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 20.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 22.

In a preferred embodiment of the invention said nucleic acid molecule comprises or consists of a nucleic acid sequence as represented in SEQ ID NO: 23.

According to an aspect of the invention there is provided a polypeptide encoded by the nucleic acid according to the invention.

According to a further aspect of the invention there is provided a polypeptide comprising an amino acid sequence selected from:

    • i) an amino acid sequence as represented in SEQ ID NO:3;
    • ii) an amino acid sequence as represented in SEQ ID NO:6;
    • iii) an amino acid sequence as represented in SEQ ID NO:9;
    • iv) an amino acid sequence as represented in SEQ ID NO:12;
    • v) an amino acid sequence as represented in SEQ ID NO:15;
    • vi) an amino acid sequence as represented in SEQ ID NO:18;
    • vii) an amino acid sequence as represented in SEQ ID NO: 21;
    • viii) an amino acid sequence as represented in SEQ ID NO:24;
    • ix) an amino acid sequence as represented in SEQ ID NO:25;
    • x) an amino acid sequence as represented in SEQ ID NO:26;
    • xi) an amino acid sequence as represented in SEQ ID NO: 27;
    • xii) an amino acid sequence as represented in SEQ ID NO:28;
    • xiii) an amino acid sequence as represented in SEQ ID NO:29;
    • xiv) an amino acid sequence as represented in SEQ ID NO:30;
    • xv) an amino acid sequence as represented in SEQ ID NO:31;
    • xvi) an amino acid sequence as represented in SEQ ID NO:32; wherein said polypeptide has growth hormone receptor antagonist activity.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 3.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 6.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 9.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 12.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 15.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 18.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 21.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 24.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 25.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 26.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 27.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 28.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 29.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 30.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 31.

In a preferred embodiment of the invention said polypeptide comprises or consists of an amino acid sequence as represented in SEQ ID NO: 32.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 3.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 6.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 9.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 12.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 15.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 18.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 21.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 24.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 25.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 26.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 27.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 28.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 29.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 30.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 31.

According to a further aspect of the invention there is provided a homodimer comprising two polypeptides comprising or consisting of SEQ ID NO: 32.

According to a further aspect of the invention there is provided a vector comprising a nucleic acid molecule according to the invention.

In a preferred embodiment of the invention said vector is an expression vector adapted to express the nucleic acid molecule according to the invention.

A vector including nucleic acid (s) according to the invention need not include a promoter or other regulatory sequence, particularly if the vector is to be used to introduce the nucleic acid into cells for recombination into the genome for stable transfection. Preferably the nucleic acid in the vector is operably linked to an appropriate promoter or other regulatory elements for transcription in a host cell. The vector may be a bi-functional expression vector which functions in multiple hosts. By “promoter” is meant a nucleotide sequence upstream from the transcriptional initiation site and which contains all the regulatory regions required for transcription. Suitable promoters include constitutive, tissue-specific, inducible, developmental or other promoters for expression in eukaryotic or prokaryotic cells. “Operably linked” means joined as part of the same nucleic acid molecule, suitably positioned and oriented for transcription to be initiated from the promoter. DNA operably linked to a promoter is “under transcriptional initiation regulation” of the promoter.

In a preferred embodiment the promoter is a constitutive, an inducible or regulatable promoter.

According to a further aspect of the invention there is provided a cell transfected or transformed with a nucleic acid molecule or vector according to the invention.

Preferably said cell is a eukaryotic cell. Alternatively said cell is a prokaryotic cell.

In a preferred embodiment of the invention said cell is selected from the group consisting of; a fungal cell (e.g. Pichia spp, Saccharomyces spp, Neurospora spp); insect cell (e.g. Spodoptera spp); a mammalian cell (e.g. COS cell, CHO cell); a plant cell.

According to a further aspect of the invention there is provided a pharmaceutical composition comprising a polypeptide according to the invention including an excipient or carrier.

In a preferred embodiment of the invention said pharmaceutical composition is combined with a further therapeutic agent.

When administered the pharmaceutical composition of the present invention is administered in pharmaceutically acceptable preparations. Such preparations may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, and optionally other therapeutic agents.

The pharmaceutical compositions of the invention can be administered by any conventional route, including injection. The administration and application may, for example, be oral, intravenous, intraperitoneal, intramuscular, intracavity, intra-articuar, subcutaneous, topical (eyes), dermal (e.g a cream lipid soluble insert into skin or mucus membrane), transdermal, or intranasal.

Pharmaceutical compositions of the invention are administered in effective amounts. An “effective amount” is that amount of pharmaceuticals/compositions that alone, or together with further doses or synergistic drugs, produces the desired response. This may involve only slowing the progression of the disease temporarily, although more preferably, it involves halting the progression of the disease permanently. This can be monitored by routine methods or can be monitored according to diagnostic methods.

The doses of the pharmaceuticals compositions administered to a subject can be chosen in accordance with different parameters, in particular in accordance with the mode of administration used and the state of the subject (i.e. age, sex). When administered, the pharmaceutical compositions of the invention are applied in pharmaceutically-acceptable amounts and in pharmaceutically-acceptable compositions. When used in medicine salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically-acceptable salts thereof and are not excluded from the scope of the invention. Such pharmacologically and pharmaceutically-acceptable salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, salicylic, citric, formic, malonic, succinic, and the like. Also, pharmaceutically-acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts.

The pharmaceutical compositions may be combined, if desired, with a pharmaceutically-acceptable carrier. The term “pharmaceutically-acceptable carrier” as used herein means one or more compatible solid or liquid fillers, diluents or encapsulating substances that are suitable for administration into a human. The term “carrier” denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The components of the pharmaceutical compositions also are capable of being co-mingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction that would substantially impair the desired pharmaceutical efficacy.

The pharmaceutical compositions may contain suitable buffering agents, including: acetic acid in a salt; citric acid in a salt; boric acid in a salt; and phosphoric acid in a salt.

The pharmaceutical compositions also may contain, optionally, suitable preservatives, such as: benzalkonium chloride; chlorobutanol; parabens and thimerosal.

The pharmaceutical compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well-known in the art of pharmacy. All methods include the step of bringing the active agent into association with a carrier that constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product.

Compositions suitable for oral administration may be presented as discrete units, such as capsules, tablets, lozenges, each containing a predetermined amount of the active compound. Other compositions include suspensions in aqueous liquids or non-aqueous liquids such as syrup, elixir or an emulsion.

Compositions suitable for parenteral administration conveniently comprise a sterile aqueous or non-aqueous preparation that is preferably isotonic with the blood of the recipient. This preparation may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation also may be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1, 3-butane diol. Among the acceptable solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or di-glycerides. In addition, fatty acids such as oleic acid may be used in the preparation of injectables. Carrier formulation suitable for oral, subcutaneous, intravenous, intramuscular, etc. administrations can be found in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa.

According to a further aspect of the invention there is provided a method to treat a human subject suffering from growth hormone excess comprising administering an effective amount of at least one polypeptide according to the invention.

In a preferred method of the invention said polypeptide is administered intravenously.

In an alternative preferred method of the invention said polypeptide is administered subcutaneously.

In a further preferred method of the invention said polypeptide is administered daily or at two day intervals; preferably said polypeptide is administered at weekly, 2 weekly or monthly intervals.

In a preferred method of the invention said growth hormone excess results in acromegaly.

In a preferred method of the invention said growth hormone excess results in gigantism.

According to a further aspect of the invention there is provided a method to treat a human subject suffering from cancer comprising administering an effective amount of at least one polypeptide according to the invention.

As used herein, the term “cancer” refers to cells having the capacity for autonomous growth, i.e., an abnormal state or condition characterized by rapidly proliferating cell growth. The term is meant to include all types of cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness. The term “cancer” includes malignancies of the various organ systems, such as those affecting, for example, lung, breast, thyroid, lymphoid, gastrointestinal, and genito-urinary tract, as well as adenocarcinomas which include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer and/or testicular tumours, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus. The term “carcinoma” is art recognized and refers to malignancies of epithelial or endocrine tissues including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas. Exemplary carcinomas include those forming from tissue of the cervix, lung, prostate, breast, head and neck, colon and ovary. The term “carcinoma” also includes carcinosarcomas, e.g., which include malignant tumours composed of carcinomatous and sarcomatous tissues. An “adenocarcinoma” refers to a carcinoma derived from glandular tissue or in which the tumor cells form recognizable glandular structures.

The term “sarcoma” is art recognized and refers to malignant tumors of mesenchymal derivation.

In a preferred method of the invention said cancer is prostate cancer.

Throughout the description and claims of this specification, the words “comprise” and “contain” and variations of the words, for example “comprising” and “comprises”, means “including but not limited to”, and is not intended to (and does not) exclude other moieties, additives, components, integers or steps.

Throughout the description and claims of this specification, the singular encompasses the plural unless the context otherwise requires. In particular, where the indefinite article is used, the specification is to be understood as contemplating plurality as well as singularity, unless the context requires otherwise.

Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein unless incompatible therewith.

An embodiment of the invention will now be described by example only and with reference to the following figures:

FIG. 1A-1B8v0: Consists of GH (contains site 1 mutation) linked to GHR extracellular (domains 1 and 2) via a G4Sx4 linker: this construct contains restriction enzyme sites around the linker region and at the 3′ end (SEQ ID NO: 1, SEQ ID NO: 2); FIG. 1B is the encoded amino acid sequence (SEQ ID NO: 3, SEQ ID NO: 25);

FIG. 2A 1B8v1: This molecule is derived from 1B8v0 but contains no extraneous sequence at the 5′ and 3′ termini and contains a G4Sx4 linker (SEQ ID NO: 4, SEQ ID NO: 5); FIG. 2B is the encoded amino acid sequence (SEQ ID NO: 6, SEQ ID NO: 26);

FIG. 3A 1B8v2: This molecule is derived from 1B8v0 but contains no extraneous sequence and contains a G4Sx5 linker (SEQ ID NO: 7, SEQ ID NO: 8);

FIG. 3C is the encoded amino acid sequence (SEQ ID NO: 9, SEQ ID NO: 27);

FIG. 4A 1B8v3: This molecule is derived from 1B8v0 but contains no extraneous sequence and contains no linker (SEQ ID NO: 10, SEQ ID NO: 11);

FIG. 4B is the encoded amino acid sequence (SEQ ID NO: 12, SEQ ID NO: 28);

FIG. 5A 1B9v0: Consists of GH (contains site 1 and site 2 mutations) linked to GHR (domains 1 and 2) via a G4Sx4 linker: this construct contains restriction enzyme sites around the linker region and at the 3′ end (SEQ ID NO:13, SEQ ID NO:14); FIG. 5B is the encoded amino acid sequence (SEQ ID NO: 15, SEQ ID NO: 29);

FIG. 6A 1B9v1: This molecule is derived from 1B9v0 but contains no extraneous sequence at the 5′ and 3′ termini and contains a G4Sx4 linker (SEQ ID NO:16, SEQ ID NO: 17); FIG. 6B is the encoded amino acid sequence (SEQ ID NO:18, SEQ ID NO: 30);

FIG. 7A 1B9v2: This molecule is derived from 1B9v0 but contains no extraneous sequence and contains a G4Sx5 linker (SEQ ID NO: 19, SEQ ID NO: 20);

FIG. 7B is the encoded amino acid sequence (SEQ ID NO: 21, SEQ ID NO: 31);

FIG. 8A 1B9v3: This molecule is derived from 1B9v0 but contains no extraneous sequence and contains no linker (SEQ ID NO:22, SEQ ID NO: 23); FIG. 8B is the encoded amino acid sequence (SEQ ID NO:24; SEQ ID NO: 32);

FIG. 9 illustrates the basic ligation strategy for subcloning the G120R molecule into a mammalian expression plasmid;

FIG. 10 illustrates the construction of IB9v0;

FIG. 11 illustrates 1B8 v2 fragment: Nar1-AvrII (524 bp). New linker region is shown in bold, with restriction enzyme site underlined. This fragment was ligated to the plasmid pGHsecTag-1B8v1 to produce the plasmid, pGHsecTag-1B8v2;

FIG. 12: illustrates 1B9 v2 fragment: Nar1-AvrII (524 bp). New linker region is shown in bold, with restriction enzyme site underlined. This fragment was ligated to the plasmid pGHsecTag-1B9v1 to produce the plasmid, pGHsecTag-1B9v2;

FIG. 13 shows a Western blot using a GH specific antibody to detect expression of both 1B8v2 (lanes 1 and 2) and 1B9v2 (lane 3) from cell culture media of a stable CHO Flp-In cell line. Samples are of correct size expected for each protein (˜75 kDa) and show no signs of degradation;

FIG. 14 illustrates that in the presence (+) of 0.5 nM rhGH, media samples from both 1B8v2 and 1B9v2 stable cell lines are able to antagonise the actions of rhGH. In the absence (−) of 0.5 nM GH both molecules show no bioactivity. The standard curve for GH is shown (0-5 nM);

FIG. 15A shows SDS-PAGE analysis of purified protein fractions by coomassie staining. Image shows that purified protein (IB8v2) is of correct size expected (˜75 kDa) and that no lower molecular weight degraded products are visible; FIG. 15B shows SDS-PAGE analysis of IB9v2;

FIG. 16A After SC administration 1B8 serum protein levels peak at 24 hrs post injection. 1B8 can still be detected 10 days post administration; FIG. 16B After IV administration 1B8 serum protein levels peak at 1 hr post injection and then decline sharply;

FIG. 17A Western blot of Native-PAGE samples: 1; 1B7v0 native GH fusion, 2: 1B7v1 native GH, 3: 1B7v2 native GH, 4:1 B7v3 native GH, 5: 1B8 modified GH fusion. All samples show a distinct double band, characteristic of a monomer and dimer formation; FIG. 17B the equivalent coomassie stained gel illustrating dimer formation;

FIG. 18 illustrates % weight gain in NZ white rabbits over 12 days comparing 5 doses of pegvisomant administered with single doses of IB8 and IB9; and

FIG. 19 illustrates PK of IB8 in NZ white rabbits over 250 hrs.

FIG. 20A is Table 1a and FIG. 20B is Table 1b. These two tables illustrate a Bradford Assay of 1B8v2 fractions.

MATERIALS AND METHODS Construction of 1B8 Antagonist Molecule

The molecule has been constructed to mutate amino acid glycine-120 to arginine in the site 2 (low affinity site) of the GH molecule (G120R). Binding of the GH molecule to the GH receptor via the high affinity site 1 is unaffected, however binding to the receptor via GH site 2 is inhibited by the bulky side group of the arginine molecule.

A PCR strategy was previously employed to generate a GH molecule containing the G120R mutation and by the use of suitable restriction sites allowed the cloning of this molecule into the pTrc-His expression plasmid to produce the clone pTrc-His-1A7 (G120R linked to the GHR extracellular B domain).

A 300 bp Bsu361-Not 1 fragment was then excised from this vector and ligated into the mammalian expression plasmid pGHsecTag-1B7 (GH linked to the GHR extracellular domains A and B) to produce pGHsecTag-1B8 (secreted expression is directed by the GH secretion signal). See FIG. 9

Construction of 1B9V0 Antagonist Molecule

The molecule has been constructed to mutate amino acids in both site 1 and site 2 of the GH molecule. Binding of the GH molecule to the GH receptor via the high affinity site 1 is enhanced by these mutations, whereas binding to the receptor via GH site 2 is inhibited by a single glycine to arginine change.

A single-strand DNA site directed mutation strategy was employed to generate a GH molecule containing both site 1 and site 2 mutations. The use of suitable restriction sites allowed the cloning of this molecule into the pTrc-His and pET21 a (+) expression plasmids. Using PCR, a clone was generated that contained the GH signal sequence (GHss) with flanking Nhe 1 and Not1 sites. This was ligated into the mammalian expression plasmid pGHsecTag-1B8v0 (GH linked to the GHR extracellular domains A and B) to produce pGHsecTag-1B9v0 (secreted expression is directed by the GH secretion signal). See FIG. 10.

Construction of the Variant Clones of Both 1B8V0 and 1B9V0

The plasmid pGHsecTag-1B7v3 was digested using the restriction enzymes HindIII-EcoRV and the fragment ligated into the plasmids pGHsecTag-1B8v0 and 1B9v0 to construct the plasmids pGHsecTag-1B8v1 and 1B9v1 (these molecules do not contain any erroneous sequence at the 3 prime end). The next stage was to remove restriction sites around the linker region to produce the plasmids pGHseTag-1B8v2 and 1B9v2. This was completed using gene synthesis and in which the original linker was replaced with a G4Sx5 linker.

The following fragments were constructed by Gene synthesis with flanking restriction sites, Nan and AvrII and ligated to either pGHsecTag-1B8v1 or 1B9v1; see FIGS. 11 and 12.

In Vitro Bioactivity of Antagonist Variant Molecules

The in vitro bioactivity of each chimera was tested using a GH-specific luciferase reporter assay. Essentially a human derived cell line was stably transfected with the human GH receptor and then transiently transfected with a luciferase signaling reporter. This assay detects physiological levels of GH, see FIG. 14.

Purification of Antagonist Molecules

CHO Flp-In cell lines expressing both 1B8v2 and 1B9v2 as a secreted product were grown in protein free media. Media was harvested, concentrated and clarified prior to affinity purification. For purification, a 20 ml NHS-activated Sepharose 4 Fast Flow resin coupled to 5E1 monoclonal antibody to hGH was prepared. Typically the media sample was concentrated ten fold and diluted 1:1 with Binding Buffer (25 mM Tris HC1/150 mM NaCl, pH 7.4) prior to purification.

Material was loaded onto the column at a flow rate of 2 ml/min. After washing, bound protein was eluted at 1 ml/min with 200 mM Glycine, pH 2.7 followed by neutralization with 1M Tris HC1, pH 9.0. Samples were analysed by SDS-PAGE (see FIGS. 15a and 15b). FIGS. 17a and 17b illustrate dimer formation of IB9 compared to native growth hormone chimeras.

Pharmacokinetic Studies of IB8

6 normal healthy rats were given a single dose injection of 1 nMol (75 ug) of protein, either subcutaneous (SC, FIG. 16a) or intravenous (IV, FIG. 16b). Control rats were given vehicle only. Samples were taken at time intervals over the course of a 10 day period and assayed for the presence of 1B8 using an in-house GH Elisa assay.

Claims

1. An isolated nucleic acid molecule comprising a nucleic acid sequence selected from the group consisting of:

the nucleic acid sequence set forth as SEQ ID NO:19; and the nucleic acid sequence set forth as SEQ ID NO:20.

2. An isolated polypeptide encoded by an isolated nucleic acid molecule comprising the nucleic acid sequence set forth as SEQ ID NO: 19 or SEQ ID NO: 20.

3. An isolated polypeptide comprising an amino acid sequence selected from the group consisting of:

the amino acid sequence set forth as SEQ ID NO:21; and
the amino acid sequence set forth as SEQ ID NO:31,
wherein said polypeptide has growth hormone receptor antagonist activity.

4. A homodimer comprising two identical polypeptides, wherein each of the two polypeptides comprises the amino acid sequence set forth as SEQ ID NO: 21; or SEQ ID NO: 31.

5. A vector comprising the nucleic acid molecule according to claim 1.

6. An isolated host cell transfected or transformed with a vector according to claim 5.

7. A pharmaceutical composition comprising a polypeptide according to claim 3, and an excipient or a carrier.

8. A composition according to claim 7, comprising a further therapeutic agent.

9. A method to treat a human subject suffering from growth hormone excess comprising

administering to the human subject an effective amount of the polypeptide of claim 3, thereby treating the growth hormone excess in the human subject.

10. The method according to claim 9, wherein said polypeptide is administered intravenously.

11. The method according to claim 10, wherein said polypeptide is administered subcutaneously.

12. The method according to claim 10, wherein said polypeptide is administered daily or at two day intervals.

13. The method according to claim 10, wherein said polypeptide is administered at weekly intervals.

14. The method according to claim 10, wherein said polypeptide is administered at bi-weekly intervals.

15. The method according to claim 10, wherein said polypeptide is administered at monthly intervals.

16. The method according to claim 10, wherein said growth hormone excess results in acromegaly.

17. The method according claim 10, wherein said growth hormone excess results in gigantism.

18. A method to treat a human subject suffering from cancer associated with excess growth hormone activity comprising administering to the human subject an effective amount of at least one polypeptide according to claim 3, thereby treating the cancer in the human subject.

19. The method according to claim 18, wherein the cancer is prostate cancer.

20. The isolated polypeptide of claim 3, comprising the amino acid sequence set forth as SEQ ID NO: 21.

21. An isolated polynucleotide encoding a polypeptide comprising the amino acid sequence set forth as SED ID NO: 21.

22. The isolated polynucleotide of claim 2, comprising the polynucleotide sequence set forth as SEQ ID NO: 19.

Patent History
Publication number: 20120322735
Type: Application
Filed: Aug 22, 2012
Publication Date: Dec 20, 2012
Applicant:
Inventors: Peter Artymiuk (Sheffield), Richard A. Ross (Sheffield), Jon Sayers (Chesterfield)
Application Number: 13/592,252